CC BY-NC-ND 4.0 · Thromb Haemost 2018; 118(08): 1491-1506
DOI: 10.1055/s-0038-1666861
Meeting Report
Georg Thieme Verlag KG Stuttgart · New York

Concepts and Controversies in Haemostasis and Thrombosis Associated with Liver Disease: Proceedings of the 7th International Coagulation in Liver Disease Conference

October 6th and 7th, 2017 Rome, Italy
N. M. Intagliata
1   Department of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia, United States
,
C. K. Argo
1   Department of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia, United States
,
J. G. Stine
2   Department of Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States
,
T. Lisman
3   Department of Surgery, University Medical Centre Groningen, Groningen, The Netherlands
,
S. H. Caldwell
1   Department of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia, United States
,
F. Violi
4   I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
,
On behalf of the faculty of the 7th International Coagulation in Liver Disease
› Author Affiliations
Funding Meeting Sponsors: Dova Pharmaceuticals; Diagnostica Stago; Bayer; AlfaSigma; Boehringer Ingelheim; Bristol-Myers-Squibb.
Further Information

Publication History

11 December 2017

17 May 2018

Publication Date:
30 July 2018 (online)

Introduction

Patients with acute and chronic liver disease frequently acquire unique changes in haemodynamic and haemostatic pathways that may result in life-threatening bleeding and thrombosis. Additionally, activation of haemostatic pathways may play a role in disease progression through parenchymal extinction, organ atrophy, recruitment of inflammatory cells and activation of stellate cells. In the setting of a previously entrenched set of clinical perceptions, the slowly evolving evidence-based paradigms of haemostasis in cirrhosis are now more carefully scrutinized.

The 7th International Conference on Coagulation in Liver Disease (held biennially since 2005) met in October of 2017 in Rome, Italy, to discuss and debate important topics in this field. This document provides a statement-based thematic summary of the meeting presented here amid a framework of the most current evidence. The review of the literature, preparation of the document and presentation of each summary statement below are based on systematic approaches used in published guidance documents.[1]

In particular, recognition of the potential disconnect between existing evidence and actual practice, we utilized a questionnaire and real-time poll of the conference attendees to compare actual clinical practice to the evidence-based support for each statement. The final section summarizes the results of the most salient results of the poll.

Laboratory Testing of the Coagulation System in Patients with Cirrhosis

1. Traditional coagulation measures, including pro-thrombin time (PT), activated partial thromboplastin time (aPTT), international normalized ratio (INR) and bleeding time (BT) do not measure bleeding risk in cirrhosis.

Conventional in vitro tests of the coagulation system are of limited value in estimating haemostatic competency in cirrhosis and current literature emphasizes these limitations when compared with global assays of coagulation.[2] [3] [4] [5] PT/INR, aPTT and BT are generally prolonged in patients with cirrhosis in direct relationship to the degree of hepatic decompensation. While useful for predicting prognosis in chronic liver disease, these tests do not predict bleeding or thrombosis in patients with chronic liver disease.

For the PT/INR and aPTT, a trigger reagent (tissue factor for extrinsic pathway or kaolin for the intrinsic pathway), phospholipids and calcium are added to patient plasma in a test tube and the time to the formation of fibrin is recorded. The PT/INR test performs poorly in assessing coagulation capacity in patients with liver disease[6] and suffers from high inter-laboratory variability.[7] [8] The PT/INR measures only pro-thrombotic pathways without measuring counter-balancing anticoagulation (AC) pathways including protein C. Numerous studies and clinical experience now clearly demonstrate that these tests do not measure in vivo activity of the coagulation system in cirrhosis.[9] [10] [11] [12] [13]

2. Platelet count alone provides an incomplete guide to bleeding risk in cirrhosis. However, values below 50,000/μL may be associated with higher risk of bleeding.

Thrombocytopenia is common in patients with advanced cirrhosis and indirectly correlates with the degree of portal hypertension and hepatic decompensation.[14] In vitro evidence indicates that platelet-dependent thrombin generation is preserved in patients with cirrhosis with platelet levels 56,000/μL or greater.[15] Notably, however, other important aspects of platelet function are not assessed by thrombin generation assay (TGA). Severe thrombocytopenia is associated with procedural-related bleeding events in some, but not all studies.[16] [17] While there is no evidence that prophylactic platelet transfusion improves haemostatic potential,[18] current guidelines and expert opinion recommend consideration of platelet transfusion prior to high-risk procedures or when active bleeding is encountered in patients with platelet counts below 50,000/μL.[4] [19] A notable exception is liver transplant surgery, in which platelet counts below 50,000/μl are accepted by many centres.

3. Global viscoelastic tests (VETs) provide a more physiologic assessment of coagulation, VETs are not standardized in patients with liver disease and do not appear to predict bleeding or thrombosis. Application of VETs in patients with liver disease remains controversial.

VETs measure clot strength and integrity in whole blood under low shear conditions, attempting to more closely recreate the environment in vivo during clot formation.[20] VETs are considered point-of-care tests and provide the clinician with information regarding the kinetics of clot formation and strength of the clot with the ability to distinguish contributions from fibrinogen, platelets and the fibrinolytic system. VETs are most extensively studied in patients with cirrhosis undergoing liver transplantation.[20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] [34] [35] [36] [37] [38] [39]

More recently VETs have been employed to examine the haemostatic system in a wide variety of patients with liver disease, including well-compensated patients,[40] [41] [42] patients with acute liver failure[43] and in acutely ill patients with liver disease.[44] [45] [46] A recent randomized control trial compared transfusion-directed prophylactic therapy in patients with cirrhosis undergoing procedures using VETs as a guide versus standard coagulation tests.[47] While significantly less blood product was used in the group undergoing VETs screening, the low rate of bleeding events, lack of control arm and inclusion of relatively low risk procedures significantly limits conclusions from these data.[48] Specific nuances of VETs such as test performance without stimulators (‘native’ VETs) and optimal duration of test remain uncertain in cirrhosis.

4. TGA provides a more comprehensive assessment of the haemostatic system in cirrhosis. However, the clinical utility of TGA in patients with cirrhosis is unexplored with current use confined mainly to research.

TGA is a valuable research tool that has led to important insights in liver disease and coagulation.[9] [10] [11] [12] [13] [15] [18] [49] [50] [51] [52] [53] [54] It measures the rate of production and decay of thrombin after introduction of a triggering agent (tissue factor and phospholipid).[55] [56] [57] [58] With the addition of thrombomodulin (required for thrombin-mediated activation of protein C), the integrity of the protein C anticoagulant system is measured. Tripodi et al demonstrated that thrombin generation is preserved in cirrhosis despite prolonged conventional coagulation assays due to decreased protein C.[10] [13] Automation and standardization of TGA is incomplete and clinical use is not currently available. Efforts at standardization and development of a point-of-care whole blood thrombin generation test are in progress with the hope for future clinical utility.[52] [59] [60]

5. Available laboratory tests of hyperfibrinolysis are of limited value in cirrhosis and clinical diagnosis is often necessary (see also anti-fibrinolytic therapy below).

Identifying the presence of hyperfibrinolysis in cirrhosis is challenging with limited clinically available and laboratory testing.[61] [62] [63] [64] [65] Reliance on clinical assessment and empiric therapy are currently recommend (see the “Pre-Procedure Prophylaxis and Treatment of Non-Portal Hypertension-Related Bleeding in Patients with Cirrhosis” section).

The fibrinolytic system regulates clot remodelling and breakdown and is likely rebalanced in cirrhosis. With exogenous disruption (e.g. infection), hyperfibrinolysis, a state of abnormal fibrin degradation mediated by plasmin, can lead to delayed bleeding and has been a recognized clinical feature of cirrhosis for over a century.[66] [67] [68] [69] [70] [71] [72] [73] [74] Joist, who coined the term ‘AICF’ or ‘accelerated intravascular coagulation and fibrinolysis’, recognized the entity of bleeding in cirrhosis similar to, but distinct, from disseminated intravascular coagulation (DIC).[75] This condition was seen in a clinical series where laboratory evidence of hyperfibrinolysis was detectable in a third of hospitalized cirrhosis patients, but clinically evident in only 6% of patients.[76] However, the divergent results of two key studies which examined the mechanistic role of liver synthesized thrombin activatable fibrinolysis inhibitor has resulted in on-going controversy regarding the clinical significance of AICF.[61] [62] These studies have failed to resolve the long known and clinically evident occurrence of diffuse mucosal bleeding or delayed post-procedure bleeding and efficacy of anti-fibrinolytic therapy.[71] [77] On the other hand, further laboratory-based studies have shown increased fibrinolytic capacity in cirrhosis patients[64] suggesting a precariously balanced system more prone to clot lysis. Both endotoxemia[78] and ascites fluid egress into the systemic circulation[79] have been postulated as mechanisms of increased clot lysis.


#

Pre-Procedure Prophylaxis and Treatment of Non-Portal Hypertension-Related Bleeding in Patients with Cirrhosis

Procedure-related bleeding is common in cirrhosis patients, but estimates of incidence vary widely.[16] For many years, the PT/INR served as a surrogate marker for estimating bleeding risk in cirrhosis. However, use of the INR and arbitrary ‘cut-offs’ as a clinical target is not recommended or supported by scientific evidence. Nonetheless, medical-legal concerns have dominated this field over scientific considerations for many years. This situation is exacerbated by the limited connection between the detached clinician performing the procedure and the ordering provider. This issue likely contributed to early termination of the well-conceived S.H.I.P. trial (Study of Hemostasis and Invasive Procedures NCT00233246) which did not enroll any patients. However, the field has substantially evolved since 2005.[80] This now allows for more refined recommendations based on recent data.

Assessment of individual patient characteristics is also essential as clinical factors, such as acute kidney injury[81] or infection[82] may alter bleeding risk in certain clinical scenarios. Global tests, such as VETs, have offered particular insight into potential pathophysiologic mechanism in patients with cirrhosis and active bacterial infection where release of endogenous heparinoids may contribute to an anticoagulant effect placing patients at higher risk to bleed.[83] [84] Large clinical studies examining bleeding outcomes are needed to provide clinicians with better data to assess risk.

6. Pre-procedure testing of plasma fibrinogen and platelet level is recommended for high-risk procedures (see [Table 1]) and pre-procedure correction is recommended for high risk procedures. Clinicians performing the procedure should define the level of risk.

Table 1

Examples of procedure risk[a]

Higher risk procedures

Intermediate risk procedures

Lower risk procedures

Brain or spinal surgery

Lumbar puncture

Paracentesis

All major surgery (cardiac, intra-abdominal and orthopaedic)

Percutaneous or transjugular liver biopsy

Thoracentesis

Intra-cranial pressure catheter insertion

Transjugular intrahepatic portosystemic shunt

Dental extraction

Endoscopy (large polypectomy with endoscopic mucosal or sub-mucosal resection, NOTES)

Endoscopy (e.g. percutaneous gastrostomy placement, cystgastrostomy, biliary sphincterotomy)

Endoscopy (e.g. diagnostic, variceal band ligation, uncomplicated polypectomy)

Percutaneous biopsy of extra-hepatic organ or lesions

Cardiac catheterization

Trans-arterial or percutaneous HCC therapies

Central line placement

Abbreviations: HCC, hepatocellular cancer; NOTES, natural orifice transluminal endoscopy.


a Risk is estimated here based on relative vascularity, degree of expected vascular breech and potential clinical consequences, but risk should always be defined by clinician preforming the procedure.


Single test assessments and even multi-parameter measures are unlikely to capture the nuances of a complicated physiology. Nonetheless, there are data suggesting that platelet level and the level of fibrinogen correlates to effective haemostasis, although it has not been established whether correction of low levels actually reduces bleeding risk.[85] Evidence is very limited regarding any measure as a guide to prophylactic measures and is mainly inferred from bleeding risk associative studies. Again, many centres do not correct low fibrinogen or platelets prior to liver transplantation, and a clinical trial has failed to demonstrate a beneficial effect of pre-emptive fibrinogen supplementation.[60] Despite the limitations and lack of firm data, with our current knowledge in the field, these parameters are currently considered to be useful in predicting bleeding in cirrhosis patients.

7. Routine prophylaxis for low or moderate risk procedures (see [Table 1]) is generally not recommended. However, individualized approaches are often necessary.

The most consistent finding with whole blood VETs (such as thromboelastography or rotational thromboelastometry) has been the demonstration of intact haemostatic mechanisms in cirrhosis with evidence that haemostatic support with blood products is not required despite abnormal PT or platelet count.[86] [87] Unfortunately, the majority of studies in this field have lacked a placebo control arm which limits our understanding of the overall baseline risk in patients without prophylaxis. To this end, we conducted a poll to assess actual current practice in potential contradistinction to rational-based therapy—see below. The most important point to remember is that prophylactic measures PT/INR can paradoxically increase bleeding risk by engorging the collateral bed without any haemostatic benefit. Greater recognition of the baseline bleeding risk and the limitations of prediction, by both clinicians that perform procedures and other clinicians, will improve patient outcomes.

Specific Therapeutic Agents

8. Platelet transfusion prior to high-risk procedures or with active bleeding has a rational in vitro basis, but lacks high level supportive data. Within the same limitations, thrombopoetin agonists may have a role in pre-planned procedural prophylaxis.

Conflicting data exists regarding bleeding risk and cirrhosis-related thrombocytopenia. The divergence in the literature likely reflects the complexity of haemostasis in cirrhosis and marked study design variation. Among patients undergoing liver transplant evaluation, procedure-related bleeding in one study was closely correlated to platelet levels < 75,000/µL.[16] Similarly, bleeding risk was higher in a retrospective study of cirrhosis patients undergoing dental extractions when platelets were < 40,000/µL.[88] Platelet levels < 30,000/µL were also an independent predictor of major bleeding among critically ill cirrhosis patients in the intensive care unit setting.[89] On the other hand, post-procedure bleeding in cirrhosis was not predicted by baseline platelet levels in another prospective study[90] and the platelet-related risk of post-banding ulcer bleeding has been variably reported.[91] [92] Compensatory mechanisms which may alter overall platelet performance, such as elevated von Willebrand factor (VWF),[93] increased circulating activated platelets[94] and platelet-derived microvesicles,[95] may also be significant factors which balance the bleeding risk related to thrombocytopenia. Additional factors may also contribute to platelet physiology.[96] [97] [98] Limited yield of platelet infusion in cirrhosis is well known and raises the question of whether platelet transfusion prior to high-risk procedures is more ‘cosmetic’ to cover procedure-related liability or an actual effective strategy—this is an understandable conundrum that remains unresolved. More definitive studies require placebo-controlled trials which present clinical investigators with almost insurmountable ethical hurdles at the current state of the art.

In elective and planned settings, such as planned dental extractions or other invasive procedures with moderate or high risk, oral thrombopoetin agonists are an alternative means to increase platelets prior to invasive procedures. Concern for a transient hypercoagulable state due to thrombocytosis with eltrombopag, with early reports of increased risk of portal vein thrombosis (PVT), have tempered enthusiasm for these agents.[99] [100] [101] However, new agents are in development, such as avatrombopag and lusutrombopag, which appear to predictably raise platelets to a range associated with improved thrombin production without apparent increased thrombosis risk.[102] [103]

9. The use of desmopressin in patients with cirrhosis lacks a sound physiological basis and does not appear to significantly alter risk of bleeding.

Desmopressin (1-deamino-8-arginine vasopressin or DDAVP) has been shown to be as effective as human blood products in cirrhosis patients undergoing dental extractions.[104] Notably, bleeding was very infrequent in both groups raising the question of actual efficacy and no control arm without prophylaxis was studied. Other studies show no benefit in variceal bleeding, liver biopsy in dialysis patients with chronic hepatitis C and with partial hepatectomy.[105] [106] [107] A recent report further demonstrated lack of significant pro-haemostatic effects in cirrhosis patients receiving a conventional dose of intravenous DDAVP compared with a control group of patients with haemophilia A.[108] These concerns raise the question again of the need for placebo controls to more definitively define the utility of various prophylactic strategies.

10. Fibrinogen replacement, pro-thrombin complex concentrates (PCC), and recombinant factor VIIa (rVIIa) may have a role in treatment of significant bleeding in patients with cirrhosis in specific clinical scenarios, but the utility of these agents is undefined.

Fibrinogen replacement: The clearest data regarding fibrinogen replacement in clinical bleeding is associated with non-cirrhosis conditions of severe trauma where fibrinogen levels > 200 mg/dL are associated with more effective haemostasis.[109] Although based on very limited data, levels below 120 mg/dL in active bleeding patients with cirrhosis have historically required correction. However, much remains to be learned about fibrinogen replacement in cirrhosis.[110] Replacement has conventionally involved cryoprecipitate, a plasma-derived product rich in VWF, fibrinogen and fibronectin, made by freezing and slow thawing human plasma and reconstituting the precipitate in 10 to 20 mL plasma. In contrast, fibrinogen concentrates are derived from a similar cryoprecipitation process of pooled plasma which then undergo isolation, viral exclusion and lyophilization.[111] The very low volume, standardization of fibrinogen content and lack of need for cross matching favour its use in cirrhosis. Recent in vitro data also suggests a clear improvement in haemostasis in cirrhosis.[112] However, further clinical studies are needed to justify the expense which currently limits wider application.

PCC: PCC have been studied in cirrhosis sporadically for over 40 years.[113] [114] However, recommended dosing is linked to AC therapy as the dose is based on factor IX content of the agent, INR (while on vitamin K antagonist [VKA]) and body weight. Data are emerging that lower doses may be effective in cirrhosis, but needs further study.[112] A typical dose is 25 to 30 units of factor IX per kilogram of body weight. The low volume needed for VKA-treated patients is attractive to avoid volume overload and collateral bed engorgement in cirrhosis, however concerns over increasing the risk of thrombosis remain.

rFVIIa: rFVIIa has mainly been studied in cirrhosis and variceal bleeding. No definitive survival benefit was evident in patients treated with rFVIIa as an adjunct in variceal bleeding.[115] [116] However, the use of this agent as a rescue agent in uncontrolled haemorrhage in cirrhosis remains unsettled by randomized trials.[117]

11. In delayed and diffuse mucosal bleeding in decompensated cirrhosis (hyperfibrinolysis physiology), consider use of anti-fibrinolytic therapy.

Optimal treatment of patients with suspected hyperfibrinolysis lacks well-controlled prospective trials to delineate risks and points of effective intervention. There are two available anti-fibrinolytics that block the lysine binding site on plasminogen preventing its attachment to fibrin and subsequent activation: trans-p-aminomethyl-cyclohexane carboxylic acid or tranexamic acid and epsilon-aminocaproic acid (EACA).[118] [119] Both agents can be given orally or intravenously. Neither are felt to be significantly pro-thrombotic based on studies in cardiac surgery of EACA versus placebo.[120] Several points of caution include the possibility of thrombotic issues if there is superimposed DIC, possible increased risk of central line related thrombosis[121] and the high concentration of the agent excreted in urine. A third agent, aprotinin, is a serine protease inhibitor that binds to plasmin to block its activity and also blocks thrombin-induced platelet aggregation. The agent decreased the need for blood transfusion in liver transplantation,[122] but remained controversial in that application[123] and was associated with increased mortality in cardiac surgery ultimately leading to marked restrictions in its clinical availability.[124]


#
#

Diagnosis and Screening for Non-Tumoural Portal Vein Thrombosis in Patients with Cirrhosis

12. Patients with advanced cirrhosis and low portal blood flow (< 15 cm/s) are at the highest risk to develop portal or splanchnic vein thrombosis.

The risk for PVT in cirrhosis parallels the severity of liver disease and hepatic decompensation. The prevalence of PVT is approximately 1% in compensated cirrhosis patients as opposed to 8 to 25% in decompensated, liver transplant candidates,[125] [126] and up to 40% in patients with hepatocellular carcinoma (HCC).[124] Of the three pathophysiologic factors predisposing to thromboembolism—slow blood flow, endothelial damage and hypercoagulability, portal flow is the most influential.[128] [129] Portal vein (PV) blood flow is inversely related to severity of liver disease as measured by the Child-Turcotte-Pugh (CTP) score,[130] and rates < 10 cm/s are associated with increased mortality.[131] Patients with underlying thrombophilia are likely at increased risk for PVT, but inherited deficiencies in natural anticoagulants can be difficult to detect in decompensated cirrhosis.[132] Emerging data suggest that cirrhosis aetiology may also have predictive power in determining which patients will develop future PVT. In particular, non-alcoholic steatohepatitis may be an independent risk factor for clinically significant thrombotic events, including PVT, in patients with decompensated cirrhosis.[133] [134] [135] The presence of a spontaneous portosystemic shunt is an under-appreciated risk for PVT as a result of a ‘steal’ syndrome and resulting PV stasis.[136]

13. Patients with cirrhosis and portal hypertension should be screened for development of PVT every 6 months with Doppler ultrasound (US).

In 2015, a systematic review concluded that PVT was associated with poorer survival in patients with cirrhosis.[137] Although hampered by heterogeneity, a recent meta-analysis similarly showed that PVT was associated with more frequent portal hypertension decompensation and higher mortality rates.[138] Furthermore, several studies indicate that PVT is associated with poorer outcomes related to variceal bleeding.[139] [140] [141] However, a large, multi-centre trial of over 1,200 subjects utilizing screening US in CTP class A and B cirrhosis demonstrated that PVT was not an independent predictor of mortality or hepatic decompensation.[142] Larger prospective studies are needed to better delineate the effect of PVT on patients with cirrhosis. As patients with cirrhosis should undergo screening US for HCC every 6 months, assessment of the PV can be performed simultaneously with Doppler US without significant additional cost and is recommended.

14. Patients with cirrhosis listed for liver transplantation should be screened at least every 6 months with Doppler US for PVT.

In general, the pre-operative presence of higher grade PVT leads to worse outcomes after liver transplantation.[126] [135] [143] [144] [145] [146] [147] However, some studies suggest that the presence of PVT may not necessarily alter the outcome for patient survival.[148] [149] These studies are challenging to compare due to the frequency of PVT in this population, selection bias and multiple confounding factors. The ability of a surgeon to establish a physiologic anastomosis for portal flow to the graft is the most important factor in predicting outcome and often related to the degree of thrombus burden.[126] [150] [151]

Detection of PVT prior to liver transplantation aids in surgical planning and allows for potential pre-operative therapy to re-canalize the PV. Detection of higher grade PVT may identify patients that are not appropriate candidates for transplantation. Re-canalization of the main PV is a high priority in transplant eligible patients.[143] [151] As treatment for PVT has a proportionally higher rate success early after diagnosis, timely recognition and prompt treatment is highly desirable (discussed below).

15. Patients with cirrhosis diagnosed with PVT by abdominal Doppler US should be assessed with cross-sectional imaging to confirm and stage the extent of thrombus.

While Doppler US is generally the initial screening modality to assess cirrhosis patients for PVT, it is limited by variation in sensitivity and specificity due in part to operator technique and experience.[152] [153] Despite this imprecision, Doppler US has a high negative predictive value (98%).[154] Contrast-enhanced computed tomography and magnetic resonance imaging have comparable sensitivity in verifying the presence of PVT seen on Doppler US, and both are equally effective in characterizing the extent of splanchnic involvement, evaluating for underlying malignancy and other sequelae.[154] [155] [156] There are several systems of grading PVT severity, yet none are widely adopted in clinical practice.[150] [157] [158] To date, the Yerdel system remains the most frequently utilized PVT classification system in clinical research.[150]

16. Testing for acquired and inherited thrombophilia in cirrhosis can be considered in patients with PVT on an individual basis, but universal screening is not currently recommended.

Inherited thrombophilia is reported in as many as 5.6% of patients with cirrhosis and PVT.[132] Several study cohorts have demonstrated more frequent occurrence of the pro-thrombin G20210A gene mutation in cirrhosis patients with PVT.[159] [160] Additional hypercoagulable risk factors may occur in patients with cirrhosis. The JAK2 V617F mutation is found in increased prevalence in patients with cirrhosis and PVT.[161] When a myeloproliferative neoplasm is suspected, consultation with a haematologist is recommended for further testing. Inherited protein C, protein S or anti-thrombin deficiencies may also predispose to PVT; however, these are difficult to detect due to co-existent liver synthetic dysfunction.[5] [11] [162]


#

Treatment of Portal Vein Thrombosis in Patients with Cirrhosis

17. Prior to AC, patients should be assessed for portal or systemic bleeding risk.

Assessment of bleeding risk and benefit of therapy is imperative when considering AC, but not yet well-defined in this population. Contraindications to AC include recent unexplained gastrointestinal (GI) bleeding, active alcohol abuse and poor functional status due to fall risk. Severe thrombocytopenia (< 50 × 109/L) may be predictive of bleeding events in cirrhosis patients treated with AC for PVT and thus special consideration should be given to this particular group.[163] Prior to AC, patients should ideally undergo upper endoscopy to assess for the presence of gastroesophageal varices.[164] If high-risk stigmata are found, endoscopic or pharmacologic therapy with non-selective β blockade should be utilized as per guideline recommendations.[165] [166]

18. AC appears to be a safe and effective treatment strategy in selected patients with compensated cirrhosis and splanchnic vein thrombosis.

Safety: AC in patients with cirrhosis and splanchnic vein thrombosis is an area of increasing interest in research.[165] [166] [167] Several series examining AC in PVT are currently published, the majority of which have been retrospective evaluating low molecular weight heparin (LMWH), VKA and direct-acting oral anti-coagulants (DOACs).[126] [163] [168] [169] [170] [171] [172] [173] [174] [175] [176] [177] [178] [179] Bleeding rates among treated and non-treated patients are similar in these highly selected patient populations.[180] [181] Rates of major bleeding range from 5 to 20% and minor bleeding rates range from 1 to 13%, but definitions of bleeding are not uniform. A recent meta-analysis examined AC for patients with cirrhosis and PVT and demonstrated a pooled bleeding rate of all bleeding events (both major and minor) of 3.3% for LMWH or VKA.[182] Remarkably, the administration of AC did not affect outcomes of upper GI bleeding events in patients with cirrhosis.[183] While overall bleeding rates appear acceptable, it is important to note the high rate of variability, the disparate outcome definitions and lack of prospective randomized studies. Caution must be maintained when considering patients with cirrhosis for treatment with AC, particularly in patients with more advanced cirrhosis (CTP B and C). Multi-disciplinary approaches on a case-by-case basis are needed.

Efficacy: Comparing studies examining the efficacy or safety of AC is challenging secondary to the heterogeneity of study design, patient population and variety of types of AC used. In general, response rates are variable and range from 42 to 82% in terms of either a partial or a complete re-canalization of the splanchnic veins.[126] [163] [168] [169] [170] [172] [173] [174] [176] Factors associated with successful therapy (re-canalization of the PV) include recent time of onset, early initiation of AC (< 6 months) and the degree of thrombus burden (partial vs. complete).[163] [184] [185] Recurrence after discontinuing therapy for PVT following clot resolution ranges from 27 to 38%.[163] [174]

19. VKAs, LMWH or direct oral anticoagulants can be considered for therapy in patients with compensated cirrhosis (CTP class A and B).

Patients with cirrhosis are no longer considered auto-anticoagulated and evidence clearly indicates risk of splanchnic and non-splanchnic thrombosis in this population. As patients with known liver disease have been excluded from clinical trials of these agents, current treatment paradigms are based on data and experience in smaller studies in cirrhosis populations.

Vitamin K antagonists: VKAs have been a cornerstone to AC therapy over many years. VKAs act through inhibition of vitamin K epoxide reductase which inhibits post-translational modification required for the function of factors II, VII, IX, X, protein C and protein S. Patients with liver dysfunction represent a particular challenge when using VKA as the INR is often elevated secondary to hepatic dysfunction. Therefore, determining the appropriate therapeutic target is problematic. Moreover, use of VKA in patients listed for transplant artificially inflates the MELD score and adversely affects organ allocation. Reversal agents for VKA include administration of vitamin K, FFP and PCC. Major advantages to VKA include familiarity through extensive clinical use over many years, low cost and oral formulation. Disadvantages include unclear therapeutic goal in liver disease patients, the requirement of routine monitoring with uncertain target levels and an apparent increase in bleeding risk.[186]

Low molecular weight heparin: LMWH is a short chain polysaccharide heparin which acts through anti-thrombin to inhibit factor Xa and thus the production of thrombin. LMWH is the most widely studied AC in patients with cirrhosis and appears safe and effective for treatment of PVT. However, studies in cirrhosis are generally small and retrospective with variation in dose and inclusion/exclusion characteristics. Expense and subcutaneous injections remain a significant disadvantage to long-term use. Monitoring therapy of LMWH to determine dosing efficacy is also controversial. Anti-Xa assays are routinely used for hospitalized patients on heparin-based AC; however, these assays can be misleading in liver disease.[181] [187] [188] TGA may offer a more reliable tool in cirrhosis to determine efficacy; however, these are not widely available.[50] In vitro studies using TGA suggest that the anticoagulant effect of LMWH may be altered in patients with cirrhosis.[50] [189] While no proven method for complete neutralization of the anticoagulant effect of LMWH is known, partial reversal of LMWH can be accomplished with protamine sulfate administration in severe hemorrhage.[190]

Direct oral anticoagulants: DOACs act to directly inhibit coagulation factors, factor IIa (dabigatran) and factor Xa (apixaban, edoxaban, rivaroxaban). DOACs are now routinely used in patients without liver function impairment with favourable safety and efficacy profiles. However, DOACs remain largely investigational in patients with cirrhosis with experience limited to in vitro studies and small case series.[177] [189] [191] [192] [193] [194] The largest series comparing DOAC to traditional AC shows similar bleeding risks.[195] In vitro data suggest that DOAC may have variable effects in patients with cirrhosis depending on the degree of hepatic decompensation present.[188] [189] [192] Notably, cirrhosis patients have been consistently excluded from DOAC clinical trials and data on pharmacodynamics is limited to in vitro studies with caution in CTP B and C cirrhosis advised. Currently, in the United States only dabigatran has an available direct reversal agent (idarucizumab).[196] [197] A recent report demonstrated successful reversal of dabigatran prior to liver transplantation without major bleeding or new thrombosis highlighting the practical use of this newly available reversal agent.[198] Clinical trials investigating andexanet α and ciraparantag as reversal agents of factor Xa inhibitors are awaiting potential approval.[196] [199] PCC may be considered for emergent reversal in the setting of DOAC therapy; however, the approach retains the possibility of inducing hypercoagulability compared with direct reversal agents.[200] [201] Use of DOAC in patients with cirrhosis should be restricted to well-compensated patients (CTP A or B) with platelet count > 50,000, and consultation with haematology is advised.

20. Liver transplant candidates with PVT should be considered for AC therapy. Non-transplant candidates may also benefit.

The indication for treatment of acute and chronic PVT in patients with cirrhosis remains controversial. The vast majority of literature focuses on the natural history, outcome and therapy of PVT in the context of liver transplantation.[143] [202] Studies outside of liver transplantation are limited and the risks and benefits of AC therapy are more difficult to estimate.

Transplant population: Significant PVT (Yerdel Grade 2 and higher) is associated with higher morbidity and mortality in patients undergoing liver transplantation.[126] [135] [144] [145] [146] [150] [203] Maintaining physiologic blood flow through the PV preferably via end-to-end anastomosis is imperative.[151] The ability to achieve physiologic inflow to the graft is generally dependent on the extent of the PVT prior to surgery. Patients with low grade PVT (Yerdel Grade 1) and non-occlusive branch PVT have similar outcomes to patients without PVT. With more advanced thrombus (Yerdel Grade 2 or higher) promoting re-canalization of the main PVT or superior mesenteric vein (SMV) thrombosis prior to transplantation is desirable. Liver transplant candidates with occlusive main PVT with or without proximal extension into the SMV should be prioritized for treatment with AC.

Non-transplant population: While prevention of PVT with AC may delay hepatic decompensation,[204] a recent study suggests PVT is not related to liver disease progression.[142] Patients with acute PVT can develop significant portal hypertension complications and progression of thrombosis may be a risk for intestinal ischemia or eventual cavernous transformation. Prospective trials are currently lacking; however, certain patients may benefit from AC therapy for acute PVT, particularly those with Yerdel Grade 2 PVT or higher where AC therapy should be considered on an individual basis after careful deliberation and multi-disciplinary review.

21. Trans-jugular intrahepatic portosystemic shunt (TIPS) is a potentially effective treatment of acute and chronic PVT in patients with cirrhosis requiring treatment for significant portal hypertension.

Historically, the presence of PVT in patients with cirrhosis was a contraindication for TIPS. However, more recent experience suggests that TIPS is safe and effective for the treatment of PVT when performed in the presence of significant portal hypertension or symptomatic complete occlusion of the main PV.[205] [206] [207] Rates of re-canalization are similar to reports from AC trials and range from 60 to 92% depending on the vascular access technique.[208] [209] [210] Recent reports suggest that TIPS alone may be adequate for re-canalization and no immediate chronic AC is required to maintain stent patency.[209] [210] [211] Patients with chronic PVT considered potential transplant candidates may also benefit from TIPS as reports are now emerging of the successful use of TIPS in this population to re-vascularize the portal venous system as a bridge to liver transplantation.[209] [210] [212]


#

Venous Thromboembolic Disease in Patients with Cirrhosis

22. Hospitalized medical patients with cirrhosis are at increased risk for VTE.

Acutely ill medical patients are at risk of developing venous thromboembolism (VTE) with an estimated rate of 4 to 15%.[213] However, large cohort studies examining the incidence of VTE and prophylactic/treatment trials with AC have generally excluded patients with cirrhosis. Recent studies indicate that patients with cirrhosis are at risk to develop VTE.[214] [215] [216] [217] [218] [219] [220] [221] [222] Hospitalized patients with cirrhosis are likely at an increased risk to develop VTE and studies indicate that thromboprophylaxis is under-utilized in this group.[223] [224] [225]

The Padua Prediction Score (PPS) was developed as a risk assessment model to predict the likelihood of developing VTE in hospitalized medical patients.[226] Current guidelines for prevention of VTE in non-surgical patients rely on the use of the PPS to distinguish between low risk of VTE (score < 4) and high risk of VTE (score > 4).[227] The PPS has been studied in a small retrospective cohort in patients with cirrhosis and was shown to be predictive of development of VTE.[223] The IMPROVE risk assessment model was developed in a large cohort of patients, which included patients with liver disease and can accurately risk stratify patients in terms of thrombosis and bleeding risk.[228]

23. In high-risk hospitalized cirrhosis patients without contraindication, medical thromboprophylaxis with LMWH or unfractionated heparin (UFH) should be considered, although efficacy remains uncertain.

Accurate risk assessment and use of VTE medical prophylaxis is sub-optimal in hospitalized medical patients.[229] Patients with cirrhosis are often perceived to be at higher risk of bleeding and likely do not receive appropriate medical VTE thromboprophylaxis.[230] One study examined 75 patients hospitalized with cirrhosis receiving LMWH for VTE prophylaxis and found relatively low rates of bleeding (five patients with reported bleeding events).[231] Two large retrospective studies show that use of UFH and LMWH appears safe with low in-hospital rates of bleeding.[224] [225] Bleeding rates were not different between patients with cirrhosis treated with prophylaxis (UFH or LMWH) (8.1%) compared with patients with cirrhosis not treated with prophylaxis (5.5%).[225] Furthermore, LMWH was safer than UFH, with decreased incidence of procedural-related bleeding.

While no current studies adequately assess efficacy for VTE prophylaxis in the hospitalized cirrhosis population, prophylactic LMWH was safe and prevented development of PVT in a small prospective cohort of cirrhosis patients.[204] Due to the relative infrequency of VTE, properly powered studies in hospitalized patients with cirrhosis will require large cohorts. Consequently, current recommendations are limited to extrapolation from studies in other populations without cirrhosis, small retrospective cohorts and expert opinion.

24. Cirrhosis patients with peripheral VTE should be treated with medical therapy with AC. LMWH, VKAs or direct oral anticoagulants can be considered.

The majority of studies examining safety and efficacy in therapeutic AC in patients with cirrhosis have been conducted in patients with PVT (see the “Treatment of Portal Vein Thrombosis in Patients with Cirrhosis” section). Very few retrospective studies have evaluated safety of therapeutic AC in patients with cirrhosis with peripheral VTE. One early, small study examined 17 patients with cirrhosis and VTE treated with LMWH and noted high bleeding rates on AC.[232] Six of the 17 patients had a major haemorrhage (defined as bleeding requiring blood transfusion) on AC. However, from this study the type of bleeding, the particular agent employed or the outcome of each patient was unclear. Numerous studies since have shown the relative safety of AC (LMWH, VKA and DOAC) at therapeutic dosing in patients with cirrhosis treated for PVT, VTE and atrial fibrillation.[126] [141] [193] [195] [231] [233] [234] [235] [236] In a recent study examining patients with cirrhosis treated with DOAC versus traditional AC (LMWH or VKA), 16 of the 39 patients were treated for VTE.[170] There was no difference between cohorts in bleeding events (4/20, 20% in the DOAC group vs. 3/19, 16% in the traditional AC group). A subsequent similar study compared DOAC and traditional AC for VTE in a total of 20 patients (8 in the traditional group and 12 in the DOAC group).[236] Bleeding rates were similar between groups (56% traditional vs. 30% DOAC). Efficacy was not assessed in either study.

The safety of AC and dosing strategy in peripheral VTE can likely be extrapolated from studies in PVT. However, the efficacy of AC therapy in VTE in cirrhosis is not well studied and the indications, timing of therapy and choice of agent are inferred from general guidelines for treatment of VTE.[237]

25. Prior to starting AC, the bleeding risk should be re-assessed with medical history and ideally an upper endoscopy. If high-risk varices are present, consideration of medical or endoscopic therapy is recommended prior to long-term AC.

See comments in the “Treatment of Portal Vein Thrombosis in Patients with Cirrhosis” section.


#

Survey of Concepts and Clinical Practice: Results of Polling of the 2017 7th International Coagulation in Liver Disease Conference Faculty and Participants

In recognition of the limitations of existing data and the variation between evidence-based and actual practice, we conducted both an interactive poll during the conference and a more deliberative on-line poll of specific scenarios related to the above statements. The polls were designed to address actual clinical practices as they related to evidenced-based concepts in this field. Among 150 attendees the overall poll response rate was 80 (53%): among responders, 65% were haepatologists, 10% were anesthesiologists and 7.5% were haematologists.

Table 2

Research goals: priorities and practicalities

Prophylaxis prior to procedures

Pre-procedural fibrinogen level and platelets have emerged as important markers of bleeding risk. However, prospective trials are now needed to confirm these markers and to establish other potential predictors of bleeding

Global coagulation tests

Future research is needed to better understand how to interpret and use global assays of haemostasis, such as thrombin generation tests (TGA) and VETs (ROTEM and TEG) in compensated and decompensated cirrhosis with a goal to establish standard references within this population

PVT in cirrhosis

In cirrhosis, PVT most clearly affects outcomes after liver transplantation. Studies are now needed to examine the efficacy and risk of anticoagulation for the various grades of PVT in all patients with cirrhosis (not just transplant eligible) with a focus on clinical outcomes. Key aspects to study include incidence, risk factors, natural history, relationships to symptoms and risks of therapy

PVT and disease progression

Intra-hepatic activation of clotting and small vessel thrombosis in parenchymal extinction may contribute to fibrosis and hepatic decompensation, but more robust studies (translational and clinical studies in humans) are necessary

VTE prophylaxis in cirrhosis

Prospective studies are needed to examine the utility of current risk models in cirrhosis patients with a goal of improving knowledge of the incidence and natural history of VTE in cirrhosis

Anticoagulants in cirrhosis

The pharmacokinetics and dosing of all anti-coagulants (low molecular weight heparin, vitamin K antagonists and direct oral anticoagulants) in patients with compensated and decompensated cirrhosis is poorly understood. Prospective studies are now needed to better guide therapy with these medications

Abbreviations: PVT, portal vein thrombosis; ROTEM, rotational thromboelasometry; TEG, thromboelastography; TGA, thrombin generation assay; VETs, viscoelastic tests; VTE, venous thromboembolism.


Regarding all laboratory testing of coagulation indices, agreement was modest for most statements. However, agreement was strongest when surveying perception surrounding the usefulness of traditional parameters of coagulation (PT/aPTT/INR) in poorly predicting haemostasis (77%), the hazard involved in rVIIa use (73%) and the lack of utility of DDAVP in preventing or treating bleeding (69%). One of the most interesting observations involved the necessity of pre-procedure plasma fibrinogen and platelet count assessment and correction: 68% of respondents agreed that these tests are useful in high-risk procedures, but 69% felt that these were not needed in low- or moderate-risk procedures.

VTE is a dynamic topic with numerous new developments. Most respondents agreed that hospitalized cirrhosis patients are at increased risk of VTE (84%), but less agreed regarding the use of medical prophylaxis with heparin agents (63%). There was strong agreement (91%) that a peripheral VTE diagnosis warrants therapeutic AC. There was strong agreement that endoscopic control of varices was recommended prior to initiating therapeutic AC (84%).

Regarding the diagnosis and treatment of splanchnic vein thrombosis, agreement was generally very high (greater than 75% for most statements). Most attendees felt that cirrhosis patients should be screened with US every 6 months for PVT (75%), and that patients listed for liver transplant should be assessed every 3 months (79%). Agreement with pursuing confirmation of US results with a cross-sectional imaging method prior to consideration of treatment was nearly unanimous (91%). Results were mixed, however, with regards to the utility of undertaking a hypercoagulability workup (56% agreement), as many felt it would not appreciably alter treatment decision-making in terms of both choice of AC and also duration of therapy. Attendees only had slightly more hesitation about treatment of PVT (76%) with AC compared with peripheral DVT (83%). Use of TIPS as a first-line modality to attempt to achieve re-canalization of PVT was less popular with only 63% agreement.

Compared with the prior 2005 survey of actual practice and opinion,[5] this 2017 survey demonstrated greater agreement on the limitations of conventional measures of bleeding risk assessment using PT/aPTT/INR, but fell well short of a consensus despite existing data. Health care providers are embracing the concept of hypercoagulability in cirrhosis and more willing to utilize therapeutic AC in the setting of peripheral VTE and PVT. Furthermore, there is emerging consensus as to the utility of fibrinogen and platelet measures as indicators of bleeding risk prior to high-risk procedures. However, the utility of more elaborate testing of hypercoagulable states, such as genetic testing remain uncertain in the setting of thrombotic complications in cirrhosis. Notably, in the general population, a hypercoagulable work-up is currently discouraged in the majority of patients with VTE and avoiding these tests in patients with VTE is on the ‘Choosing Wisely’ list endorsed by multiple specialty societies.[238]

While significant gains in our understanding of the complexities of the coagulation system in liver disease have been made over the last several years, future focused research will be essential to advance the field ([Table 2]).


#
 
  • References

  • 1 Atkins D, Best D, Briss PA. , et al; GRADE Working Group. Grading quality of evidence and strength of recommendations. BMJ 2004; 328 (7454): 1490
  • 2 Tripodi A, Mannucci PM. The coagulopathy of chronic liver disease. N Engl J Med 2011; 365 (02) 147-156
  • 3 Lisman T, Porte RJ. Rebalanced hemostasis in patients with liver disease: evidence and clinical consequences. Blood 2010; 116 (06) 878-885
  • 4 Northup PG, Caldwell SH. Coagulation in liver disease: a guide for the clinician. Clin Gastroenterol Hepatol 2013; 11 (09) 1064-1074
  • 5 Caldwell SH, Hoffman M, Lisman T. , et al; Coagulation in Liver Disease Group. Coagulation disorders and hemostasis in liver disease: pathophysiology and critical assessment of current management. Hepatology 2006; 44 (04) 1039-1046
  • 6 Ewe K. Bleeding after liver biopsy does not correlate with indices of peripheral coagulation. Dig Dis Sci 1981; 26 (05) 388-393
  • 7 Trotter JF, Olson J, Lefkowitz J, Smith AD, Arjal R, Kenison J. Changes in international normalized ratio (INR) and model for endstage liver disease (MELD) based on selection of clinical laboratory. Am J Transplant 2007; 7 (06) 1624-1628
  • 8 Tripodi A, Caldwell SH, Hoffman M, Trotter JF, Sanyal AJ. Review article: the prothrombin time test as a measure of bleeding risk and prognosis in liver disease. Aliment Pharmacol Ther 2007; 26 (02) 141-148
  • 9 Lisman T, Bakhtiari K, Pereboom IT, Hendriks HG, Meijers JC, Porte RJ. Normal to increased thrombin generation in patients undergoing liver transplantation despite prolonged conventional coagulation tests. J Hepatol 2010; 52 (03) 355-361
  • 10 Tripodi A, Salerno F, Chantarangkul V. , et al. Evidence of normal thrombin generation in cirrhosis despite abnormal conventional coagulation tests. Hepatology 2005; 41 (03) 553-558
  • 11 Tripodi A, Primignani M, Chantarangkul V. , et al. An imbalance of pro- vs anti-coagulation factors in plasma from patients with cirrhosis. Gastroenterology 2009; 137 (06) 2105-2111
  • 12 Tripodi A, Primignani M, Lemma L. , et al. Detection of the imbalance of procoagulant versus anticoagulant factors in cirrhosis by a simple laboratory method. Hepatology 2010; 52 (01) 249-255
  • 13 Tripodi A, Primignani M, Lemma L, Chantarangkul V, Mannucci PM. Evidence that low protein C contributes to the procoagulant imbalance in cirrhosis. J Hepatol 2013; 59 (02) 265-270
  • 14 Giannini EG, Savarino V. Thrombocytopenia in liver disease. Curr Opin Hematol 2008; 15 (05) 473-480
  • 15 Tripodi A, Primignani M, Chantarangkul V. , et al. Thrombin generation in patients with cirrhosis: the role of platelets. Hepatology 2006; 44 (02) 440-445
  • 16 Giannini EG, Greco A, Marenco S, Andorno E, Valente U, Savarino V. Incidence of bleeding following invasive procedures in patients with thrombocytopenia and advanced liver disease. Clin Gastroenterol Hepatol 2010; 8 (10) 899-902 , quiz e109
  • 17 Napolitano G, Iacobellis A, Merla A. , et al. Bleeding after invasive procedures is rare and unpredicted by platelet counts in cirrhotic patients with thrombocytopenia. Eur J Intern Med 2017; 38: 79-82
  • 18 Tripodi A, Primignani M, Chantarangkul V. , et al. Global hemostasis tests in patients with cirrhosis before and after prophylactic platelet transfusion. Liver Int 2013; 33 (03) 362-367
  • 19 Rockey DC, Caldwell SH, Goodman ZD, Nelson RC, Smith AD. ; American Association for the Study of Liver Diseases. Liver biopsy. Hepatology 2009; 49 (03) 1017-1044
  • 20 Mallett SV. Clinical utility of viscoelastic tests of coagulation (TEG/ROTEM) in patients with liver disease and during liver transplantation. Semin Thromb Hemost 2015; 41 (05) 527-537
  • 21 Zahr Eldeen F, Roll GR, Derosas C. , et al. Preoperative thromboelastography as a sensitive tool predicting those at risk of developing early hepatic artery thrombosis after adult liver transplantation. Transplantation 2016; 100 (11) 2382-2390
  • 22 Tafur LA, Taura P, Blasi A. , et al. Rotation thromboelastometry velocity curve predicts blood loss during liver transplantation. Br J Anaesth 2016; 117 (06) 741-748
  • 23 De Pietri L, Bianchini M, Rompianesi G, Bertellini E, Begliomini B. Thromboelastographic reference ranges for a cirrhotic patient population undergoing liver transplantation. World J Transplant 2016; 6 (03) 583-593
  • 24 Abuelkasem E, Mazzeffi MA, Lu SY, Planinsic RM, Sakai T, Tanaka KA. Ex vivo evaluation of 4 different viscoelastic assays for detecting moderate to severe coagulopathy during liver transplantation. Liver Transpl 2016; 22 (04) 468-475
  • 25 Abuelkasem E, Lu S, Tanaka K, Planinsic R, Sakai T. Comparison between thrombelastography and thromboelastometry in hyperfibrinolysis detection during adult liver transplantation. Br J Anaesth 2016; 116 (04) 507-512
  • 26 Seo H, Choi JH, Moon YJ, Jeong SM. FIBTEM of thromboelastometry does not accurately represent fibrinogen concentration in patients with severe hypofibrinogenemia during liver transplantation. Ann Transplant 2015; 20: 342-350
  • 27 Roullet S, Freyburger G, Cruc M. , et al. Management of bleeding and transfusion during liver transplantation before and after the introduction of a rotational thromboelastometry-based algorithm. Liver Transpl 2015; 21 (02) 169-179
  • 28 Jeong SM, Song JG, Seo H, Choi JH, Jang DM, Hwang GS. Quantification of both platelet count and fibrinogen concentration using maximal clot firmness of thromboelastometry during liver transplantation. Transplant Proc 2015; 47 (06) 1890-1895
  • 29 Yang Lu S, Tanaka KA, Abuelkasem E, Planinsic RM, Sakai T. Clinical applicability of rapid thrombelastography and functional fibrinogen thrombelastography to adult liver transplantation. Liver Transpl 2014; 20 (09) 1097-1105
  • 30 Song JG, Jeong SM, Jun IG, Lee HM, Hwang GS. Five-minute parameter of thromboelastometry is sufficient to detect thrombocytopenia and hypofibrinogenaemia in patients undergoing liver transplantation. Br J Anaesth 2014; 112 (02) 290-297
  • 31 Mohammed M, Fayed N, Hassanen A. , et al. Rotational thromboelastometry and standard coagulation tests for live liver donors. Clin Transplant 2013; 27 (02) E101-E108
  • 32 Krzanicki D, Sugavanam A, Mallett S. Intraoperative hypercoagulability during liver transplantation as demonstrated by thromboelastography. Liver Transpl 2013; 19 (08) 852-861
  • 33 Wang SC, Lin HT, Chang KY. , et al. Use of higher thromboelastogram transfusion values is not associated with greater blood loss in liver transplant surgery. Liver Transpl 2012; 18 (10) 1254-1258
  • 34 Blasi A, Beltran J, Pereira A. , et al. An assessment of thromboelastometry to monitor blood coagulation and guide transfusion support in liver transplantation. Transfusion 2012; 52 (09) 1989-1998
  • 35 Stancheva A, Spassov L, Tzatchev K. Correlation between rotation thrombelastometry ROTEM analysis and standard haemostatic parameters during liver transplantation. Clin Lab 2011; 57 (5-6): 407-413
  • 36 Wang SC, Shieh JF, Chang KY. , et al. Thromboelastography-guided transfusion decreases intraoperative blood transfusion during orthotopic liver transplantation: randomized clinical trial. Transplant Proc 2010; 42 (07) 2590-2593
  • 37 Trzebicki J, Flakiewicz E, Kosieradzki M. , et al. The use of thromboelastometry in the assessment of hemostasis during orthotopic liver transplantation reduces the demand for blood products. Ann Transplant 2010; 15 (03) 19-24
  • 38 Roullet S, Pillot J, Freyburger G. , et al. Rotation thromboelastometry detects thrombocytopenia and hypofibrinogenaemia during orthotopic liver transplantation. Br J Anaesth 2010; 104 (04) 422-428
  • 39 Hendriks HG, Meijer K, de Wolf JT. , et al. Effects of recombinant activated factor VII on coagulation measured by thromboelastography in liver transplantation. Blood Coagul Fibrinolysis 2002; 13 (04) 309-313
  • 40 Tripodi A, Primignani M, Chantarangkul V. , et al. The coagulopathy of cirrhosis assessed by thromboelastometry and its correlation with conventional coagulation parameters. Thromb Res 2009; 124 (01) 132-136
  • 41 Kleinegris MC, Bos MH, Roest M. , et al. Cirrhosis patients have a coagulopathy that is associated with decreased clot formation capacity. J Thromb Haemost 2014; 12 (10) 1647-1657
  • 42 Hugenholtz GCG, Lisman T, Stravitz RT. Thromboelastography does not predict outcome in different etiologies of cirrhosis. Research and Practice in Thrombosis and Haemostasis 2017; 1 (02) 275-285
  • 43 Stravitz RT, Lisman T, Luketic VA. , et al. Minimal effects of acute liver injury/acute liver failure on hemostasis as assessed by thromboelastography. J Hepatol 2012; 56 (01) 129-136
  • 44 Lloyd-Donald P, Vasudevan A, Angus P. , et al. Coagulation in acutely ill patients with severe chronic liver disease: Insights from thromboelastography. J Crit Care 2017; 38: 215-224
  • 45 Papatheodoridis GV, Patch D, Webster GJ, Brooker J, Barnes E, Burroughs AK. Infection and hemostasis in decompensated cirrhosis: a prospective study using thrombelastography. Hepatology 1999; 29 (04) 1085-1090
  • 46 Chau TN, Chan YW, Patch D, Tokunaga S, Greenslade L, Burroughs AK. Thrombelastographic changes and early rebleeding in cirrhotic patients with variceal bleeding. Gut 1998; 43 (02) 267-271
  • 47 De Pietri L, Bianchini M, Montalti R. , et al. Thrombelastography-guided blood product use before invasive procedures in cirrhosis with severe coagulopathy. A randomized controlled trial. Hepatology 2016; 63 (02) 566-573
  • 48 Intagliata NM, Caldwell SH, Porte RJ, Lisman T. Prediction of bleeding in cirrhosis patients: is the forecast any clearer?. Hepatology 2016; 64 (03) 989-990
  • 49 Gatt A, Riddell A, Calvaruso V, Tuddenham EG, Makris M, Burroughs AK. Enhanced thrombin generation in patients with cirrhosis-induced coagulopathy. J Thromb Haemost 2010; 8 (09) 1994-2000
  • 50 Senzolo M, Rodriguez-Castro KI, Rossetto V. , et al. Increased anticoagulant response to low-molecular-weight heparin in plasma from patients with advanced cirrhosis. J Thromb Haemost 2012; 10 (09) 1823-1829
  • 51 Tripodi A, Chantarangkul V, Primignani M. , et al. Thrombin generation in plasma from patients with cirrhosis supplemented with normal plasma: considerations on the efficacy of treatment with fresh-frozen plasma. Intern Emerg Med 2012; 7 (02) 139-144
  • 52 Groeneveld D, Porte RJ, Lisman T. Thrombomodulin-modified thrombin generation testing detects a hypercoagulable state in patients with cirrhosis regardless of the exact experimental conditions. Thromb Res 2014; 134 (03) 753-756
  • 53 Tripodi A, Fracanzani AL, Primignani M. , et al. Procoagulant imbalance in patients with non-alcoholic fatty liver disease. J Hepatol 2014; 61 (01) 148-154
  • 54 Magnusson M, Berndtsson M, Fischler B. , et al. Thrombin generation test in children and adolescents with chronic liver disease. Thromb Res 2015; 135 (02) 382-387
  • 55 Hemker HC, Willems GM, Béguin S. A computer assisted method to obtain the prothrombin activation velocity in whole plasma independent of thrombin decay processes. Thromb Haemost 1986; 56 (01) 9-17
  • 56 Hemker HC, Béguin S. Thrombin generation in plasma: its assessment via the endogenous thrombin potential. Thromb Haemost 1995; 74 (01) 134-138
  • 57 Hemker HC, Giesen P, AlDieri R. , et al. The calibrated automated thrombogram (CAT): a universal routine test for hyper- and hypocoagulability. Pathophysiol Haemost Thromb 2002; 32 (5-6): 249-253
  • 58 Tripodi A. Thrombin generation assay and its application in the clinical laboratory. Clin Chem 2016; 62 (05) 699-707
  • 59 Dargaud Y, Wolberg AS, Luddington R. , et al. Evaluation of a standardized protocol for thrombin generation measurement using the calibrated automated thrombogram: an international multicentre study. Thromb Res 2012; 130 (06) 929-934
  • 60 Ninivaggi M, Apitz-Castro R, Dargaud Y, de Laat B, Hemker HC, Lindhout T. Whole-blood thrombin generation monitored with a calibrated automated thrombogram-based assay. Clin Chem 2012; 58 (08) 1252-1259
  • 61 Lisman T, Leebeek FW, Mosnier LO. , et al. Thrombin-activatable fibrinolysis inhibitor deficiency in cirrhosis is not associated with increased plasma fibrinolysis. Gastroenterology 2001; 121 (01) 131-139
  • 62 Colucci M, Binetti BM, Branca MG. , et al. Deficiency of thrombin activatable fibrinolysis inhibitor in cirrhosis is associated with increased plasma fibrinolysis. Hepatology 2003; 38 (01) 230-237
  • 63 Stravitz RT. Potential applications of thromboelastography in patients with acute and chronic liver disease. Gastroenterol Hepatol (N Y) 2012; 8 (08) 513-520
  • 64 Rijken DC, Kock EL, Guimarães AH. , et al. Evidence for an enhanced fibrinolytic capacity in cirrhosis as measured with two different global fibrinolysis tests. J Thromb Haemost 2012; 10 (10) 2116-2122
  • 65 Peterson TJ, Webb AM, Vipler BS. Use of thromboelastography in the management of liver cirrhosis and accelerated intravascular coagulation and fibrinolysis (AICF). BMJ Case Rep 2016; 2016: xx
  • 66 Goodpasture EW. Fibrinolysis in chronic hepatic insufficiency. Bull Johns Hopkins Hosp 1914; 25: 330-336
  • 67 Palascak JE, Martinez J. Dysfibrinogenemia associated with liver disease. J Clin Invest 1977; 60 (01) 89-95
  • 68 Brodsky I, Dennis LH. Evaluation of fibrinolysis in hepatic cirrhosis. Relation of serial thrombin time and euglobulin lysis time. Am J Clin Pathol 1966; 45 (01) 61-69
  • 69 Violi F, Ferro D, Basili S. , et al. Hyperfibrinolysis increases the risk of gastrointestinal hemorrhage in patients with advanced cirrhosis. Hepatology 1992; 15 (04) 672-676
  • 70 Violi F, Ferro D, Basili S. , et al. Hyperfibrinolysis resulting from clotting activation in patients with different degrees of cirrhosis. The CALC Group. Coagulation abnormalities in liver cirrhosis. Hepatology 1993; 17 (01) 78-83
  • 71 Ferro D, Celestini A, Violi F. Hyperfibrinolysis in liver disease. Clin Liver Dis 2009; 13 (01) 21-31
  • 72 Ben-Ari Z, Osman E, Hutton RA, Burroughs AK. Disseminated intravascular coagulation in liver cirrhosis: fact or fiction?. Am J Gastroenterol 1999; 94 (10) 2977-2982
  • 73 Boks AL, Brommer EJ, Schalm SW, Van Vliet HH. Hemostasis and fibrinolysis in severe liver failure and their relation to hemorrhage. Hepatology 1986; 6 (01) 79-86
  • 74 Violl F, Basili S, Ferro D, Quintarelli C, Alessandril C, Cordova C. ; CALC Group. Association between high values of D-dimer and tissue-plasminogen activator activity and first gastrointestinal bleeding in cirrhotic patients. Thromb Haemost 1996; 76 (02) 177-183
  • 75 Joist JH. AICF and DIC in liver cirrhosis: expressions of a hypercoagulable state. Am J Gastroenterol 1999; 94 (10) 2801-2803
  • 76 Hu KQ, Yu AS, Tiyyagura L, Redeker AG, Reynolds TB. Hyperfibrinolytic activity in hospitalized cirrhotic patients in a referral liver unit. Am J Gastroenterol 2001; 96 (05) 1581-1586
  • 77 Gunawan B, Runyon B. The efficacy and safety of epsilon-aminocaproic acid treatment in patients with cirrhosis and hyperfibrinolysis. Aliment Pharmacol Ther 2006; 23 (01) 115-120
  • 78 Violi F, Ferro D, Basili S. , et al. Association between low-grade disseminated intravascular coagulation and endotoxemia in patients with liver cirrhosis. Gastroenterology 1995; 109 (02) 531-539
  • 79 Agarwal S, Joyner Jr KA, Swaim MW. Ascites fluid as a possible origin for hyperfibrinolysis in advanced liver disease. Am J Gastroenterol 2000; 95 (11) 3218-3224
  • 80 DeAngelis GA, Khot R, Haskal ZJ. , et al. Bleeding risk and management in interventional procedures in chronic liver disease. J Vasc Interv Radiol 2016; 27 (11) 1665-1674
  • 81 Hung A, Garcia-Tsao G. Acute kidney injury, but not sepsis, is associated with higher procedure-related bleeding in patients with decompensated cirrhosis. Liver Int 2018; DOI: 10.1111/liv.13712.
  • 82 Montalto P, Vlachogiannakos J, Cox DJ, Pastacaldi S, Patch D, Burroughs AK. Bacterial infection in cirrhosis impairs coagulation by a heparin effect: a prospective study. J Hepatol 2002; 37 (04) 463-470
  • 83 Senzolo M, Riddell A, Tuddenham E, Burroughs AK. Endogenous heparinoids contribute to coagulopathy in patients with liver disease. J Hepatol 2008; 48 (02) 371-372
  • 84 Senzolo M, Cholongitas E, Thalheimer U. , et al. Heparin-like effect in liver disease and liver transplantation. Clin Liver Dis 2009; 13 (01) 43-53
  • 85 Kalambokis GN, Oikonomou A, Christou L. , et al. von Willebrand factor and procoagulant imbalance predict outcome in patients with cirrhosis and thrombocytopenia. J Hepatol 2016; 65 (05) 921-928
  • 86 Bedreli S, Sowa JP, Gerken G, Saner FH, Canbay A. Management of acute-on-chronic liver failure: rotational thromboelastometry may reduce substitution of coagulation factors in liver cirrhosis. Gut 2016; 65 (02) 357-358
  • 87 De Pietri L, Bianchini M, Montalti R. , et al. Thrombelastography-guided blood product use before invasive procedures in cirrhosis with severe coagulopathy: A randomized, controlled trial. Hepatology 2016; 63 (02) 566-573
  • 88 Cocero N, Bezzi M, Martini S, Carossa S. Oral surgical treatment of patients with chronic liver disease: assessments of bleeding and its relationship with thrombocytopenia and blood coagulation parameters. J Oral Maxillofac Surg 2017; 75 (01) 28-34
  • 89 Drolz A, Horvatits T, Roedl K. , et al. Coagulation parameters and major bleeding in critically ill patients with cirrhosis. Hepatology 2016; 64 (02) 556-568
  • 90 Napolitano G, Iacobellis A, Merla A. , et al. Bleeding after invasive procedures is rare and unpredicted by platelet counts in cirrhotic patients with thrombocytopenia. Eur J Intern Med 2017; 38: 79-82
  • 91 Vieira da Rocha EC, D'Amico EA, Caldwell SH. , et al. A prospective study of conventional and expanded coagulation indices in predicting ulcer bleeding after variceal band ligation. Clin Gastroenterol Hepatol 2009; 7 (09) 988-993
  • 92 Vanbiervliet G, Giudicelli-Bornard S, Piche T. , et al. Predictive factors of bleeding related to post-banding ulcer following endoscopic variceal ligation in cirrhotic patients: a case-control study. Aliment Pharmacol Ther 2010; 32 (02) 225-232
  • 93 Lisman T, Bongers TN, Adelmeijer J. , et al. Elevated levels of von Willebrand Factor in cirrhosis support platelet adhesion despite reduced functional capacity. Hepatology 2006; 44 (01) 53-61
  • 94 Raparelli V, Basili S, Carnevale R. , et al. Low-grade endotoxemia and platelet activation in cirrhosis. Hepatology 2017; 65 (02) 571-581
  • 95 Rautou PE, Vion AC, Valla D, Boulanger CM. Circulating platelet derived microparticles are not increased in patients with cirrhosis. J Hepatol 2013; 59 (04) 912
  • 96 Mannucci PM, Tripodi A. Hemostatic defects in liver and renal dysfunction. Hematology (Am Soc Hematol Educ Program) 2012; 2012: 168-173
  • 97 Zwaginga JJ, Ijsseldijk MJ, Beeser-Visser N, de Groot PG, Vos J, Sixma JJ. High von Willebrand factor concentration compensates a relative adhesion defect in uremic blood. Blood 1990; 75 (07) 1498-1508
  • 98 Castillo R, Lozano T, Escolar G, Revert L, López J, Ordinas A. Defective platelet adhesion on vessel subendothelium in uremic patients. Blood 1986; 68 (02) 337-342
  • 99 Violi F, Pignatelli P. Eltrombopag before procedures in patients with cirrhosis and thrombocytopenia. N Engl J Med 2012; 367 (21) 2056
  • 100 Lisman T, Porte RJ. Eltrombopag before procedures in patients with cirrhosis and thrombocytopenia. N Engl J Med 2012; 367 (21) 2055-2056
  • 101 Afdhal NH, Giannini EG, Tayyab G. , et al; ELEVATE Study Group. Eltrombopag before procedures in patients with cirrhosis and thrombocytopenia. N Engl J Med 2012; 367 (08) 716-724
  • 102 Terrault N, Chen YC. , et al. Avatrombopag before procedures reduces need for platelet transfusion in patients with chronic liver disease and thrombocytopenia. Gastroenterology 2018; DOI: 10.1053/j.gastro.2018.05.025.
  • 103 Izumi NOY, Yamamoto K, Osaki Y, Imawari M. A phase 3, randomized, double-blind, placebo-controlled study of lusutrombopag for thrombocytopenia in patients with chronic liver disease undergoing elective invasive procedures in Japan (L-PLUS 1). [abstract no. LB-30] J Hepatol 2015; 62 (06) 1397A-1398A
  • 104 Stanca CM, Montazem AH, Lawal A, Zhang JX, Schiano TD. Intranasal desmopressin versus blood transfusion in cirrhotic patients with coagulopathy undergoing dental extraction: a randomized controlled trial. J Oral Maxillofac Surg 2010; 68 (01) 138-143
  • 105 de Franchis R, Arcidiacono PG, Carpinelli L. , et al. Randomized controlled trial of desmopressin plus terlipressin vs. terlipressin alone for the treatment of acute variceal hemorrhage in cirrhotic patients: a multicenter, double-blind study. New Italian Endoscopic Club. Hepatology 1993; 18 (05) 1102-1107
  • 106 Fraser IS, Porte RJ, Kouides PA, Lukes AS. A benefit-risk review of systemic haemostatic agents: part 1: in major surgery. Drug Saf 2008; 31 (03) 217-230
  • 107 Wong AY, Irwin MG, Hui TW, Fung SK, Fan ST, Ma ES. Desmopressin does not decrease blood loss and transfusion requirements in patients undergoing hepatectomy. Can J Anaesth 2003; 50 (01) 14-20
  • 108 Arshad F, Stoof SC, Leebeek FW. , et al. Infusion of DDAVP does not improve primary hemostasis in patients with cirrhosis. Liver Int 2015; 35 (07) 1809-1815
  • 109 Bolliger D, Szlam F, Molinaro RJ, Rahe-Meyer N, Levy JH, Tanaka KA. Finding the optimal concentration range for fibrinogen replacement after severe haemodilution: an in vitro model. Br J Anaesth 2009; 102 (06) 793-799
  • 110 Hugenholtz GC, Macrae F, Adelmeijer J. , et al. Procoagulant changes in fibrin clot structure in patients with cirrhosis are associated with oxidative modifications of fibrinogen. J Thromb Haemost 2016; 14 (05) 1054-1066
  • 111 Franchini M, Lippi G. Fibrinogen replacement therapy: a critical review of the literature. Blood Transfus 2012; 10 (01) 23-27
  • 112 Lisman T, Kleiss SF, Patel VC. , et al. In vitro efficacy of pro- and anticoagulant strategies in acutely ill patients with cirrhosis. Abstract 533 Hepatology 2017; 66 (S1): 292A
  • 113 Lorenz R, Kienast J, Otto U. , et al. Efficacy and safety of a prothrombin complex concentrate with two virus-inactivation steps in patients with severe liver damage. Eur J Gastroenterol Hepatol 2003; 15 (01) 15-20
  • 114 Mannucci PM, Franchi F, Dioguardi N. Correction of abnormal coagulation in chronic liver disease by combined use of fresh-frozen plasma and prothrombin complex concentrates. Lancet 1976; 2 (7985): 542-545
  • 115 Bosch J, Thabut D, Albillos A. , et al; International Study Group on rFVIIa in UGI Hemorrhage. Recombinant factor VIIa for variceal bleeding in patients with advanced cirrhosis: a randomized, controlled trial. Hepatology 2008; 47 (05) 1604-1614
  • 116 Bosch J, Thabut D, Bendtsen F. , et al; European Study Group on rFVIIa in UGI Haemorrhage. Recombinant factor VIIa for upper gastrointestinal bleeding in patients with cirrhosis: a randomized, double-blind trial. Gastroenterology 2004; 127 (04) 1123-1130
  • 117 Bendtsen F, D'Amico G, Rusch E. , et al. Effect of recombinant Factor VIIa on outcome of acute variceal bleeding: an individual patient based meta-analysis of two controlled trials. J Hepatol 2014; 61 (02) 252-259
  • 118 Franchini M, Mannucci PM. Adjunct agents for bleeding. Curr Opin Hematol 2014; 21 (06) 503-508
  • 119 Stewart DBMV. Therapy with Antifibrinolytic Agents. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. Philadelphia: Lippincott Williams, Wilkins, Wolters Kluwer; 2013
  • 120 Slaughter TF, Faghih F, Greenberg CS, Leslie JB, Sladen RN. The effects of epsilon-aminocaproic acid on fibrinolysis and thrombin generation during cardiac surgery. Anesth Analg 1997; 85 (06) 1221-1226
  • 121 Dentz ME, Slaughter TF, Mark JB. Early thrombus formation on heparin-bonded pulmonary artery catheters in patients receiving epsilon aminocaproic acid. Anesthesiology 1995; 82 (02) 583-586
  • 122 Porte RJ, Molenaar IQ, Begliomini B. , et al; EMSALT Study Group. Aprotinin and transfusion requirements in orthotopic liver transplantation: a multicentre randomised double-blind study. Lancet 2000; 355 (9212): 1303-1309
  • 123 Ramsay MA. Con: antifibrinolytics are not safe and effective in patients undergoing liver transplantation. J Cardiothorac Vasc Anesth 2006; 20 (06) 891-893
  • 124 Fergusson DA, Hébert PC, Mazer CD. , et al; BART Investigators. A comparison of aprotinin and lysine analogues in high-risk cardiac surgery. N Engl J Med 2008; 358 (22) 2319-2331
  • 125 Okuda K, Ohnishi K, Kimura K. , et al. Incidence of portal vein thrombosis in liver cirrhosis. An angiographic study in 708 patients. Gastroenterology 1985; 89 (02) 279-286
  • 126 Francoz C, Belghiti J, Vilgrain V. , et al. Splanchnic vein thrombosis in candidates for liver transplantation: usefulness of screening and anticoagulation. Gut 2005; 54 (05) 691-697
  • 127 Amitrano L, Guardascione MA, Brancaccio V. , et al. Risk factors and clinical presentation of portal vein thrombosis in patients with liver cirrhosis. J Hepatol 2004; 40 (05) 736-741
  • 128 Zocco MA, Di Stasio E, De Cristofaro 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 (04) 682-689
  • 129 Stine JG, Wang J, Shah PM. , et al. Decreased portal vein velocity is predictive of the development of portal vein thrombosis: a matched case-control study. Liver Int 2018; 38 (01) 94-101
  • 130 Zironi G, Gaiani S, Fenyves D, Rigamonti A, Bolondi L, Barbara L. Value of measurement of mean portal flow velocity by Doppler flowmetry in the diagnosis of portal hypertension. J Hepatol 1992; 16 (03) 298-303
  • 131 Zoli M, Iervese T, Merkel C. , et al. Prognostic significance of portal hemodynamics in patients with compensated cirrhosis. J Hepatol 1993; 17 (01) 56-61
  • 132 Qi X, De Stefano V, Wang J. , et al. Prevalence of inherited antithrombin, protein C, and protein S deficiencies in portal vein system thrombosis and Budd-Chiari syndrome: a systematic review and meta-analysis of observational studies. J Gastroenterol Hepatol 2013; 28 (03) 432-442
  • 133 Stine JG, Shah NL, Argo CK, Pelletier SJ, Caldwell SH, Northup PG. Increased risk of portal vein thrombosis in patients with cirrhosis due to nonalcoholic steatohepatitis. Liver Transpl 2015; 21 (08) 1016-1021
  • 134 Stine JG, Argo CK, Pelletier SJ, Maluf DG, Caldwell SH, Northup PG. Advanced non-alcoholic steatohepatitis cirrhosis: a high-risk population for pre-liver transplant portal vein thrombosis. World J Hepatol 2017; 9 (03) 139-146
  • 135 Ghabril M, Agarwal S, Lacerda M, Chalasani N, Kwo P, Tector AJ. 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 (01) 126-133
  • 136 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 (04) 568-574
  • 137 Qi X, De Stefano V, Senzolo M, Xu H, Mancuso A. Splanchnic vein thrombosis: etiology, diagnosis, and treatment. Gastroenterol Res Pract 2015; 2015: 506136
  • 138 Stine JG, Shah PM, Cornella SL. , et al. Portal vein thrombosis, mortality and hepatic decompensation in patients with cirrhosis: a meta-analysis. World J Hepatol 2015; 7 (27) 2774-2780
  • 139 D'Amico G, De Franchis R. ; Cooperative Study Group. Upper digestive bleeding in cirrhosis. Post-therapeutic outcome and prognostic indicators. Hepatology 2003; 38 (03) 599-612
  • 140 Augustin S, Altamirano J, González A. , et al. Effectiveness of combined pharmacologic and ligation therapy in high-risk patients with acute esophageal variceal bleeding. Am J Gastroenterol 2011; 106 (10) 1787-1795
  • 141 Amitrano L, Guardascione MA, Menchise A. , et al. 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 (06) 448-451
  • 142 Nery F, Chevret S, Condat B. , et al; Groupe d'Etude et de Traitement du Carcinome Hépatocellulaire. Causes and consequences of portal vein thrombosis in 1,243 patients with cirrhosis: results of a longitudinal study. Hepatology 2015; 61 (02) 660-667
  • 143 Francoz C, Valla D, Durand F. Portal vein thrombosis, cirrhosis, and liver transplantation. J Hepatol 2012; 57 (01) 203-212
  • 144 Stine JG, Pelletier SJ, Schmitt TM, Porte RJ, Northup PG. Pre-transplant portal vein thrombosis is an independent risk factor for graft loss due to hepatic artery thrombosis in liver transplant recipients. HPB 2016; 18 (03) 279-286
  • 145 Englesbe MJ, Kubus J, Muhammad W. , et al. Portal vein thrombosis and survival in patients with cirrhosis. Liver Transpl 2010; 16 (01) 83-90
  • 146 Englesbe MJ, Schaubel DE, Cai S, Guidinger MK, Merion RM. Portal vein thrombosis and liver transplant survival benefit. Liver Transpl 2010; 16 (08) 999-1005
  • 147 Rodríguez-Castro KI, Porte RJ, 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 (11) 1145-1153
  • 148 Ravaioli M, Zanello M, Grazi GL. , et al. Portal vein thrombosis and liver transplantation: evolution during 10 years of experience at the University of Bologna. Ann Surg 2011; 253 (02) 378-384
  • 149 Wu G, Liu YF, Liu SR, Zhang JL, Chen XC, Cheng DH. Liver transplantation in end-stage liver disease with portal vein thrombosis [in Chinese]. Zhonghua Wai Ke Za Zhi 2009; 47 (08) 590-593
  • 150 Yerdel MA, Gunson B, Mirza D. , et al. Portal vein thrombosis in adults undergoing liver transplantation: risk factors, screening, management, and outcome. Transplantation 2000; 69 (09) 1873-1881
  • 151 Hibi T, Nishida S, Levi DM. , et al. When and why portal vein thrombosis matters in liver transplantation: a critical audit of 174 cases. Ann Surg 2014; 259 (04) 760-766
  • 152 Ponziani FR, Zocco MA, Campanale C. , et al. Portal vein thrombosis: insight into physiopathology, diagnosis, and treatment. World J Gastroenterol 2010; 16 (02) 143-155
  • 153 Chawla Y, Duseja A, Dhiman RK. Review article: the modern management of portal vein thrombosis. Aliment Pharmacol Ther 2009; 30 (09) 881-894
  • 154 Parikh S, Shah R, Kapoor P. Portal vein thrombosis. Am J Med 2010; 123 (02) 111-119
  • 155 Bayraktar Y, Harmanci O. Etiology and consequences of thrombosis in abdominal vessels. World J Gastroenterol 2006; 12 (08) 1165-1174
  • 156 Margini C, Berzigotti A. Portal vein thrombosis: the role of imaging in the clinical setting. Dig Liver Dis 2017; 49 (02) 113-120
  • 157 Nonami T, Yokoyama I, Iwatsuki S, Starzl TE. The incidence of portal vein thrombosis at liver transplantation. Hepatology 1992; 16 (05) 1195-1198
  • 158 Sarin SK, Philips CA, Kamath PS. , et al. Toward a comprehensive new classification of portal vein thrombosis in patients with cirrhosis. Gastroenterology 2016; 151 (04) 574-577.e3
  • 159 Amitrano L, Brancaccio V, Guardascione MA. , et al. Portal vein thrombosis after variceal endoscopic sclerotherapy in cirrhotic patients: role of genetic thrombophilia. Endoscopy 2002; 34 (07) 535-538
  • 160 Erkan O, Bozdayi AM, Disibeyaz S. , et al. Thrombophilic gene mutations in cirrhotic patients with portal vein thrombosis. Eur J Gastroenterol Hepatol 2005; 17 (03) 339-343
  • 161 Saugel B, Lee M, Feichtinger S, Hapfelmeier A, Schmid RM, Siveke JT. Thrombophilic factor analysis in cirrhotic patients with portal vein thrombosis. J Thromb Thrombolysis 2015; 40 (01) 54-60
  • 162 Basit SA, Stone CD, Gish R. Portal vein thrombosis. Clin Liver Dis 2015; 19 (01) 199-221
  • 163 Delgado MG, Seijo S, Yepes I. , et al. Efficacy and safety of anticoagulation on patients with cirrhosis and portal vein thrombosis. Clin Gastroenterol Hepatol 2012; 10 (07) 776-783
  • 164 Dhar A, Mullish BH, Thursz MR. Anticoagulation in chronic liver disease. J Hepatol 2017; 66 (06) 1313-1326
  • 165 European Association for the Study of the Liver. Electronic address: easloffice@easloffice.eu. EASL clinical practice guidelines: vascular diseases of the liver. J Hepatol 2016; 64 (01) 179-202
  • 166 de Franchis R. . Baveno VI Faculty. Expanding consensus in portal hypertension: report of the Baveno VI Consensus Workshop: stratifying risk and individualizing care for portal hypertension. J Hepatol 2015; 63 (03) 743-752
  • 167 Stine JG, Northup PG. Coagulopathy before and after liver transplantation: from the hepatic to the systemic circulatory systems. Clin Liver Dis 2017; 21 (02) 253-274
  • 168 Ageno W, Riva N, Schulman S. , et al. Long-term clinical outcomes of splanchnic vein thrombosis: results of an international registry. JAMA Intern Med 2015; 175 (09) 1474-1480
  • 169 Cui SB, Shu RH, Yan SP. , et al. 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 (08) 914-919
  • 170 Naeshiro N, Aikata H, Hyogo H. , 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 (06) 656-662
  • 171 Dell'Era A, Iannuzzi F, Fabris FM. , et al. Impact of portal vein thrombosis on the efficacy of endoscopic variceal band ligation. Dig Liver Dis 2014; 46 (02) 152-156
  • 172 Werner KT, Sando S, Carey EJ. , et al. Portal vein thrombosis in patients with end stage liver disease awaiting liver transplantation: outcome of anticoagulation. Dig Dis Sci 2013; 58 (06) 1776-1780
  • 173 Senzolo M, , M Sartori T, Rossetto V. , et al. Prospective evaluation of anticoagulation and transjugular intrahepatic portosystemic shunt for the management of portal vein thrombosis in cirrhosis. Liver Int 2012; 32 (06) 919-927
  • 174 Amitrano L, Guardascione MA, Menchise A. , et al. 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 (06) 448-451
  • 175 Intagliata NM, Henry ZH, Maitland H. , et al. Direct oral anticoagulants in cirrhosis patients pose similar risks of bleeding when compared to traditional anticoagulation. Dig Dis Sci 2016; 61 (06) 1721-1727
  • 176 Chen H, Liu L, Qi X. , et al. Efficacy and safety of anticoagulation in more advanced portal vein thrombosis in patients with liver cirrhosis. Eur J Gastroenterol Hepatol 2016; 28 (01) 82-89
  • 177 De Gottardi A, Trebicka J, Klinger C. , et al; VALDIG Investigators. Antithrombotic treatment with direct-acting oral anticoagulants in patients with splanchnic vein thrombosis and cirrhosis. Liver Int 2017; 37 (05) 694-699
  • 178 Riva N, Ageno W, Schulman S. , et al; International Registry on Splanchnic Vein Thrombosis (IRSVT) study group. Clinical history and antithrombotic treatment of incidentally detected splanchnic vein thrombosis: a multicentre, international prospective registry. Lancet Haematol 2016; 3 (06) e267-e275
  • 179 Ageno W, Riva N, Schulman S. , et al. Long-term Clinical Outcomes of Splanchnic Vein Thrombosis: Results of an International Registry. JAMA Intern Med 2015; 175 (09) 1474-1480
  • 180 Samama MM, Cohen AT, Darmon JY. , et al; Prophylaxis in Medical Patients with Enoxaparin Study Group. A comparison of enoxaparin with placebo for the prevention of venous thromboembolism in acutely ill medical patients. N Engl J Med 1999; 341 (11) 793-800
  • 181 Bechmann LP, Sichau M, Wichert M, Gerken G, Kröger K, Hilgard P. Low-molecular-weight heparin in patients with advanced cirrhosis. Liver Int 2011; 31 (01) 75-82
  • 182 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 (01) 23-29
  • 183 Cerini F, Gonzalez JM, Torres F. , et al. Impact of anticoagulation on upper-gastrointestinal bleeding in cirrhosis. A retrospective multicenter study. Hepatology 2015; 62 (02) 575-583
  • 184 Francoz C, Valla D, Durand F. Portal vein thrombosis, cirrhosis, and liver transplantation. J Hepatol 2012; 57 (01) 203-212
  • 185 Kinjo N, Kawanaka H, Akahoshi T. , et al. Portal vein thrombosis in liver cirrhosis. World J Hepatol 2014; 6 (02) 64-71
  • 186 Levi M, Hovingh GK, Cannegieter SC, Vermeulen M, Büller HR, Rosendaal FR. Bleeding in patients receiving vitamin K antagonists who would have been excluded from trials on which the indication for anticoagulation was based. Blood 2008; 111 (09) 4471-4476
  • 187 Lisman T, Porte RJ. Towards a rational use of low-molecular-weight heparin in patients with cirrhosis. Liver Int 2011; 31 (07) 1063
  • 188 Potze W, Arshad F, Adelmeijer J. , et al. Routine coagulation assays underestimate levels of antithrombin-dependent drugs but not of direct anticoagulant drugs in plasma from patients with cirrhosis. Br J Haematol 2013; 163 (05) 666-673
  • 189 Potze W, Arshad F, Adelmeijer J. , et al. Differential in vitro inhibition of thrombin generation by anticoagulant drugs in plasma from patients with cirrhosis. PLoS One 2014; 9 (02) e88390
  • 190 Garcia DA, Baglin TP, Weitz JI, Samama MM. Parenteral anticoagulants: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141 (2 Suppl): e24S-e43S
  • 191 Intagliata NM, Maitland H, Caldwell SH. Direct oral anticoagulants in cirrhosis. Curr Treat Options Gastroenterol 2016; 14 (02) 247-256
  • 192 Potze W, Adelmeijer J, Lisman T. Decreased in vitro anticoagulant potency of rivaroxaban and apixaban in plasma from patients with cirrhosis. Hepatology 2015; 61 (04) 1435-1436
  • 193 Martinez M, Tandra A, Vuppalanchi R. Treatment of acute portal vein thrombosis by nontraditional anticoagulation. Hepatology 2014; 60 (01) 425-426
  • 194 Intagliata NM, Maitland H, Northup PG, Caldwell SH. Treating thrombosis in cirrhosis patients with new oral agents: ready or not?. Hepatology 2015; 61 (02) 738-739
  • 195 Intagliata NM, Henry ZH, Maitland H. , et al. Direct Oral Anticoagulants in Cirrhosis Patients Pose Similar Risks of Bleeding When Compared to Traditional Anticoagulation. Dig Dis Sci 2016; 61 (06) 1721-1727
  • 196 Rogers KC, Shelton MP, Finks SW. Reversal agents for direct oral anticoagulants: understanding new and upcoming options. Cardiol Rev 2016; 24 (06) 310-315
  • 197 Hu TY, Vaidya VR, Asirvatham SJ. Reversing anticoagulant effects of novel oral anticoagulants: role of ciraparantag, andexanet alfa, and idarucizumab. Vasc Health Risk Manag 2016; 12: 35-44
  • 198 Intagliata NM, Maitland H, Pellitier S, Caldwell SH. Reversal of direct oral anticoagulants for liver transplantation in cirrhosis: A step forward. Liver Transpl 2017; 23 (03) 396-397
  • 199 Siegal DM, Curnutte JT, Connolly SJ. , et al. Andexanet alfa for the reversal of factor Xa inhibitor activity. N Engl J Med 2015; 373 (25) 2413-2424
  • 200 Eerenberg ES, Kamphuisen PW, Sijpkens MK, Meijers JC, Buller HR, Levi M. Reversal of rivaroxaban and dabigatran by prothrombin complex concentrate: a randomized, placebo-controlled, crossover study in healthy subjects. Circulation 2011; 124 (14) 1573-1579
  • 201 Levi M, Moore KT, Castillejos CF. , et al. Comparison of three-factor and four-factor prothrombin complex concentrates regarding reversal of the anticoagulant effects of rivaroxaban in healthy volunteers. J Thromb Haemost 2014; 12 (09) 1428-1436
  • 202 Chen H, Turon F, Hernández-Gea V. , et al. Nontumoral portal vein thrombosis in patients awaiting liver transplantation. Liver Transpl 2016; 22 (03) 352-365
  • 203 Stine JG, Shah PM, Cornella SL. , et al. Portal vein thrombosis, mortality and hepatic decompensation in patients with cirrhosis: a meta-analysis. World J Hepatol 2015; 7 (27) 2774-2780
  • 204 Villa E, Cammà C, Marietta M. , et al. Enoxaparin prevents portal vein thrombosis and liver decompensation in patients with advanced cirrhosis. Gastroenterology 2012; 143 (05) 1253-60.e1 , 4
  • 205 Senzolo M, Tibbals J, Cholongitas E, Triantos CK, Burroughs AK, Patch D. Transjugular intrahepatic portosystemic shunt for portal vein thrombosis with and without cavernous transformation. Aliment Pharmacol Ther 2006; 23 (06) 767-775
  • 206 Van Ha TG, Hodge J, Funaki B. , et al. Transjugular intrahepatic portosystemic shunt placement in patients with cirrhosis and concomitant portal vein thrombosis. Cardiovasc Intervent Radiol 2006; 29 (05) 785-790
  • 207 Senzolo M, Patch D, Cholongitas E, Burroughs AK. Transjugular intrahepatic portosystemic shunt for portal vein thrombosis with and without underlying cirrhosis. Cardiovasc Intervent Radiol 2007; 30 (03) 545 , author reply 546
  • 208 Luca A, Miraglia R, Caruso S. , et al. Short- and long-term effects of the transjugular intrahepatic portosystemic shunt on portal vein thrombosis in patients with cirrhosis. Gut 2011; 60 (06) 846-852
  • 209 Salem R, Vouche M, Baker T. , et al. Pretransplant portal vein recanalization-transjugular intrahepatic portosystemic shunt in patients with complete obliterative portal vein thrombosis. Transplantation 2015; 99 (11) 2347-2355
  • 210 Thornburg B, Desai K, Hickey R. , et al. Portal vein recanalization and transjugular intrahepatic portosystemic shunt creation for chronic portal vein thrombosis: technical considerations. Tech Vasc Interv Radiol 2016; 19 (01) 52-60
  • 211 Wang Z, Jiang MS, Zhang HL. , et al. Is post-TIPS anticoagulation therapy necessary in patients with cirrhosis and portal vein thrombosis? A randomized controlled trial. Radiology 2016; 279 (03) 943-951
  • 212 Habib A, Desai K, Hickey R. , et al. 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 (04) 499-506
  • 213 Khoury H, Welner S, Kubin M, Folkerts K, Haas S. Disease burden and unmet needs for prevention of venous thromboembolism in medically ill patients in Europe show underutilisation of preventive therapies. Thromb Haemost 2011; 106 (04) 600-608
  • 214 Northup PG, McMahon MM, Ruhl AP. , et al. Coagulopathy does not fully protect hospitalized cirrhosis patients from peripheral venous thromboembolism. Am J Gastroenterol 2006; 101 (07) 1524-1528 , quiz 1680
  • 215 Gulley D, Teal E, Suvannasankha A, Chalasani N, Liangpunsakul S. Deep vein thrombosis and pulmonary embolism in cirrhosis patients. Dig Dis Sci 2008; 53 (11) 3012-3017
  • 216 Søgaard KK, Horváth-Puhó E, Grønbaek H, Jepsen P, Vilstrup H, Sørensen HT. Risk of venous thromboembolism in patients with liver disease: a nationwide population-based case-control study. Am J Gastroenterol 2009; 104 (01) 96-101
  • 217 Dabbagh O, Oza A, Prakash S, Sunna R, Saettele TM. Coagulopathy does not protect against venous thromboembolism in hospitalized patients with chronic liver disease. Chest 2010; 137 (05) 1145-1149
  • 218 Vivarelli M, Zanello M, Zanfi C. , et al. Prophylaxis for venous thromboembolism after resection of hepatocellular carcinoma on cirrhosis: is it necessary?. World J Gastroenterol 2010; 16 (17) 2146-2150
  • 219 Wu H, Nguyen GC. Liver cirrhosis is associated with venous thromboembolism among hospitalized patients in a nationwide US study. Clin Gastroenterol Hepatol 2010; 8 (09) 800-805
  • 220 Aldawood A, Arabi Y, Aljumah A. , et al. The incidence of venous thromboembolism and practice of deep venous thrombosis prophylaxis in hospitalized cirrhotic patients. Thromb J 2011; 9 (01) 1
  • 221 Ali M, Ananthakrishnan AN, McGinley EL, Saeian K. Deep vein thrombosis and pulmonary embolism in hospitalized patients with cirrhosis: a nationwide analysis. Dig Dis Sci 2011; 56 (07) 2152-2159
  • 222 Ambrosino P, Tarantino L, Di Minno G. , et al. The risk of venous thromboembolism in patients with cirrhosis. A systematic review and meta-analysis. Thromb Haemost 2017; 117 (01) 139-148
  • 223 Bogari H, Patanwala AE, Cosgrove R, Katz M. Risk-assessment and pharmacological prophylaxis of venous thromboembolism in hospitalized patients with chronic liver disease. Thromb Res 2014; 134 (06) 1220-1223
  • 224 Intagliata NM, Henry ZH, Shah N, Lisman T, Caldwell SH, Northup PG. Prophylactic anticoagulation for venous thromboembolism in hospitalized cirrhosis patients is not associated with high rates of gastrointestinal bleeding. Liver Int 2014; 34 (01) 26-32
  • 225 Shatzel J, Dulai PS, Harbin D. , et al. Safety and efficacy of pharmacological thromboprophylaxis for hospitalized patients with cirrhosis: a single-center retrospective cohort study. J Thromb Haemost 2015; 13 (07) 1245-1253
  • 226 Barbar S, Noventa F, Rossetto V. , et al. A risk assessment model for the identification of hospitalized medical patients at risk for venous thromboembolism: the Padua Prediction Score. J Thromb Haemost 2010; 8 (11) 2450-2457
  • 227 Kahn SR, Lim W, Dunn AS. , et al. Prevention of VTE in nonsurgical patients: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141 (2, Suppl): e195S-e226S
  • 228 Spyropoulos AC, Anderson Jr FA, FitzGerald G. , et al; IMPROVE Investigators. Predictive and associative models to identify hospitalized medical patients at risk for VTE. Chest 2011; 140 (03) 706-714
  • 229 Tapson VF, Decousus H, Pini M. , et al; IMPROVE Investigators. Venous thromboembolism prophylaxis in acutely ill hospitalized medical patients: findings from the International Medical Prevention Registry on Venous Thromboembolism. Chest 2007; 132 (03) 936-945
  • 230 Senzolo M, Sartori MT, Lisman T. Should we give thromboprophylaxis to patients with liver cirrhosis and coagulopathy?. HPB 2009; 11 (06) 459-464
  • 231 Bechmann LP, Sichau M, Wichert M, Gerken G, Kröger K, Hilgard P. Low-molecular-weight heparin in patients with advanced cirrhosis. Liver Int 2011; 31 (01) 75-82
  • 232 García-Fuster MJ, Abdilla N, Fabiá MJ, Fernández C, Oliver V, Forner MJ. Venous thromboembolism and liver cirrhosis. Rev Esp Enferm Dig 2008; 100 (05) 259-262
  • 233 Delgado MG, Seijo S, Yepes I. , et al. Efficacy and safety of anticoagulation on patients with cirrhosis and portal vein thrombosis. Clin Gastroenterol Hepatol 2012; 10 (07) 776-783
  • 234 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 (06) 686-691
  • 235 Senzolo M, , M Sartori T, Rossetto V. , et al. Prospective evaluation of anticoagulation and transjugular intrahepatic portosystemic shunt for the management of portal vein thrombosis in cirrhosis. Liver Int 2012; 32 (06) 919-927
  • 236 Hum J, Shatzel JJ, Jou JH, Deloughery TG. The efficacy and safety of direct oral anticoagulants vs traditional anticoagulants in cirrhosis. Eur J Haematol 2017; 98 (04) 393-397
  • 237 Kearon C, Akl EA, Ornelas J. , et al. Antithrombotic therapy for VTE disease: CHEST Guideline and Expert Panel Report. Chest 2016; 149 (02) 315-352
  • 238 Middeldorp S. Inherited thrombophilia: a double-edged sword. Hematology (Am Soc Hematol Educ Program) 2016; 2016 (01) 1-9