Review Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Sep 27, 2024; 16(9): 1229-1246
Published online Sep 27, 2024. doi: 10.4254/wjh.v16.i9.1229
Hematological abnormalities in liver cirrhosis
Oscar Manuel Fierro-Angulo, José Alberto González-Regueiro, Ariana Pereira-García, Astrid Ruiz-Margáin, Ricardo Ulises Macías-Rodríguez, Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico 14080, Mexico
Fernando Solis-Huerta, Department of Hematology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico 14080, Mexico
ORCID number: Oscar Manuel Fierro-Angulo (0000-0001-5462-647X); José Alberto González-Regueiro (0000-0001-5211-4710); Ariana Pereira-García (0000-0002-6524-7341); Astrid Ruiz-Margáin (0000-0003-2779-8641); Fernando Solis-Huerta (0000-0002-5096-6853); Ricardo Ulises Macías-Rodríguez (0000-0002-7637-4477).
Co-first authors: Oscar Manuel Fierro-Angulo and José Alberto González-Regueiro.
Author contributions: Fierro-Angulo OM and González-Regueiro JA contributed to conception and design of the review, they are co-first authors of this manuscript; Fierro-Angulo OM, González-Regueiro JA, Pereira-García A, Ruiz-Margáin A, Solis-Huerta F, and Macías-Rodríguez RU contributed to literature review, writing and analysis of data, drafting and critical revision; Ruiz-Margáin A and Macías-Rodríguez RU contributed to supervision, editing of the content of the manuscript and writing the final version of the manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ricardo Ulises Macías-Rodríguez, MD, MSc, PhD, Assistant Professor, Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Col. Belisario Domínguez Sección XVI, Mexico 14080, Mexico. ricardomacro@yahoo.com.mx
Received: June 6, 2024
Revised: August 9, 2024
Accepted: August 22, 2024
Published online: September 27, 2024
Processing time: 109 Days and 5.5 Hours

Abstract

Hematological abnormalities are common in cirrhosis and are associated with various pathophysiological mechanisms. Studies have documented a prevalence of thrombocytopenia, leukopenia, and anemia in patients with compensated cirrhosis of 77.9%, 23.5%, and 21.1%, respectively. These abnormalities carry significant clinical implications, including considerations for invasive procedures, infection risk, bleeding risk, and prognosis. Previously, cirrhosis was believed to predispose patients to bleeding due to alterations observed in classical coagulation tests such as prothrombin time, partial thromboplastin time, international normalized ratio, and thrombocytopenia. However, this understanding has evolved, and cirrhosis patients are now also acknowledged as being at a high risk for thrombotic events. Hemostasis in cirrhosis patients presents a complex phenotype, with procoagulant and anticoagulant abnormalities offsetting each other. This multifactorial phenomenon is inadequately reflected by routine laboratory tests. Thrombotic complications are more prevalent in decompensated cirrhosis and may correlate with disease severity. Bleeding is primarily associated with portal hypertension, endothelial dysfunction, mechanical vessel injury, disseminated intravascular coagulation, endotoxemia, and renal injury. This review comprehensively outlines hematologic index abnormalities, mechanisms of hemostasis, coagulation, and fibrinolysis abnormalities, limitations of laboratory testing, and clinical manifestations of bleeding and thrombosis in patients with liver cirrhosis.

Key Words: Cirrhosis; Anemia; Leukopenia; Thrombocytopenia; Coagulopathy; Bleeding; Thrombosis

Core Tip: Cirrhosis, characterized by progressive liver damage due to chronic injury, frequently induces changes in blood cell counts, functions, and coagulation abnormalities. Recognizing and understanding these alterations is pivotal as they often reflect the severity of liver disease. This knowledge enables healthcare providers to anticipate potential complications, initiate timely interventions, and tailor treatment strategies for patients with cirrhosis. This review aims to summarize available data on the prevalence of these abnormalities, delve into their pathophysiology, and elucidate their implications for patient management.



INTRODUCTION

Cirrhosis is the leading cause of liver-related mortality and ranks among the top ten leading causes of death[1]. Globally, it contributes to over 1.32 million deaths, comprising 2.4% of total mortality[2]. Patients living with cirrhosis develop complications stemming from two different mechanisms: Hepatocyte insufficiency (liver failure) and portal hypertension. These arise predominantly from progressive liver fibrosis, characterized by distorted hepatic architecture, and the loss of normal hepatocytes due to sustained inflammation. Understanding these mechanisms is crucial when addressing hematological abnormalities in patients with cirrhosis.

The hemostatic profile of individuals with cirrhosis typically manifests as thrombocytopenia, reduced levels of coagulation factors and inhibitors, diminished levels of fibrinolytic proteins, and elevated levels of coagulation factor VIII and von Willebrand factor (VWF). Hemostasis in cirrhosis adopts a complex phenotype, where procoagulant and anticoagulant dyscrasias counterbalance each other, resulting in a state of equilibrium[3].

This intricate interplay in cirrhosis is inadequately captured by routine laboratory tests utilized to evaluate coagulopathy. Consequently, emerging techniques such as viscoelastic testing show promise in comprehensively assessing the entire clot-forming process in individuals with liver cirrhosis. Importantly, hemostasis in these patients is precarious and susceptible to disruption by common insults. Hence, seemingly paradoxical occurrences, such as bleeding and thrombosis, can manifest in cirrhosis. In this review, we discuss abnormalities in hematologic indices, mechanisms of hemostasis, coagulation, and fibrinolysis, limitations of laboratory testing, as well as clinical manifestations of bleeding and thrombosis in patients with liver cirrhosis.

Epidemiology of hematological abnormalities in cirrhosis

Hematologic indices are frequently abnormal in patients with cirrhosis. Several studies have reported a prevalence of anemia, thrombocytopenia, and leukopenia between 6%-77%, although the assessment of the true occurrence of abnormal hematologic indices in cirrhosis is limited by the cross-sectional design of these studies[4-10]. A prospective cohort that included 213 patients with compensated cirrhosis followed for 9 years reported a baseline prevalence of thrombocytopenia, leukopenia, and anemia of 77.9%, 23.5%, and 21.1%, respectively[11]. Thrombocytopenia was the most common and earliest hematologic index abnormality to develop, followed by leukopenia and anemia. The occurrence of abnormal hematologic indices in patients with cirrhosis may have significant clinical implications, such as considerations regarding invasive procedures, risk of infections, and risk of bleeding. Therefore, knowing the frequency of these abnormalities helps the clinician to be prepared with the knowledge when facing this type of patients.

Pathophysiology of hematological abnormalities in cirrhosis

The pathophysiological basis of these abnormalities is multifactorial[12], and includes portal hypertension-induced splenic and splanchnic sequestration, alteration in bone marrow stimulating factors [hematopoietic stimulating factor, thrombopoietin (TPO), erythropoietin, granulocyte-colony stimulating factor and granulocyte macrophage-colony stimulating factor], bone marrow suppression mediated by alcohol, hepatitis B and C, alterations in hemostasis (elevated VWF), coagulopathy (increased factor VIII, low levels of all other procoagulation and anticoagulation factors), hyperfibrinolysis (increased tissue plasminogen activator and plasminogen activator inhibitor type 1, low levels of all other profibrinolytic and antifibrinolytic proteins) and increased blood loss[3,11,13-15]. Some of the previously described factors are simultaneously present in patients, contributing to specific hematological abnormalities. Therefore, understanding these factors can assist clinicians in identifying potential modifiable causes, ultimately improving patient outcomes.

Abnormalities in complete blood count in cirrhosis: These abnormalities are influenced by factors associated with the etiology of chronic liver damage (such as alcohol and viral hepatitis), the impact of liver failure (resulting in poor protein synthesis) and portal hypertension, as shown in Figure 1[16]. In addition to these factors, it is worth bearing in mind the role of extrahepatic factors, leading to hematological abnormalities in cirrhosis, such as hepatitis B or C, autoimmune hepatitis, drugs and nutritional deficiencies (iron, vitamin B12 and folate)[17]. Furthermore, beyond complete blood count, the peripheral blood smear is a cornerstone in evaluating certain hematologic disorders. Its importance is particularly evident in clinical settings where extrahepatic factors are involved and timely diagnosis is crucial for early medical decisions.

Figure 1
Figure 1  Liver-related causes of abnormal hematological parameters in patients with cirrhosis.

Erythrocytes: The main cause of anemia in liver cirrhosis is acute or chronic bleeding in the context of portal hypertension[18]. The prevalence of iron deficiency in these patients approaches 50% compared to 24.3% in the general population[19,20]. Causes of iron deficiency include bleeding[19], as well as deficiencies in vitamin B12 and folic acid, with some studies indicating a higher prevalence of folate deficiency than vitamin B12 deficiency[21] in patients with cirrhosis.

Macrocytosis (elevated mean corpuscular volume > 100 fL) is present in almost 50% of patients with liver disease, often associated with deficiencies in vitamin B12 and folate, thyroid diseases (more frequent in patients with autoimmune hepatitis-related cirrhosis) and liver disease itself. This abnormality is typically accompanied by a high red cell distribution width[22]. The main alteration in mean corpuscular hemoglobin is hypochromia (< 80 pg), typically linked to iron deficiency[19]. In addition to macrocytosis, another common finding in the peripheral blood smear of patients with cirrhosis is the presence of spur cells (acanthocytes). Proposed mechanisms for these abnormalities include hypersplenism and alterations in lipoprotein metabolism[23], with the reported prevalence of spur cells ranging from 5% to 19% in some series[24].

Leucocytes: Leukopenia is a frequent finding in patients with advanced liver disease. Additionally, up to 26% of patients with cirrhosis exhibit dysplastic changes in leukocytes during bone marrow examination without fulfilling the definition of myelodysplastic neoplasm[25]. These abnormalities have been linked to the proinflammatory state caused by liver cirrhosis and its complications.

Platelets: Thrombocytopenia, defined as a platelet count < 150000/μL, is the most common hematologic abnormality to develop (seen in as high as 64%-80%), and it typically occurs earliest, followed sequentially by leukopenia and anemia. While most patients exhibit mild thrombocytopenia, moderate thrombocytopenia occurs in 13%, and severe thrombocytopenia requiring platelet transfusions in 1%-2% of patients[10,11,26-28]. Thrombocytopenia combined with leukopenia (prevalence 15.5%) is the most common abnormality, followed by thrombocytopenia and anemia (prevalence 9.4%), and anemia with leukopenia (0.5%). Pancytopenia is reported in only 6.6% of these cases. Interestingly, individuals who develop thrombocytopenia and leukopenia have an increased risk of hepatic decompensation, hepatocellular carcinoma, mortality, or requiring transplant, even when controlling for other factors such as Child-Pugh score, hepatic vein pressure gradient, or alcohol use[11].

Prothrombin time/international normalized ratio: Prothrombin time (PT) evaluates the extrinsic and common pathways of coagulation, including the activity of tissue factor, factors II, V, VII, X, and even fibrinogen[29]. International normalized ratio (INR) is calculated as a ratio between the patient’s PT to the mean normal PT adjusted with a correction factor called the international sensitivity index[30]. The increase in PT and INR reflects worse liver function in cirrhosis, nevertheless, common tests such as PT/INR oversimplify the adaptations in the balance between procoagulant and anticoagulant factors in liver cirrhosis[31], it is more appropriate to use viscoelastic assays in specific clinical settings, where a more precise estimation of the coagulation status is required (for example, acute bleeding needing a high volume of blood products).

PT and INR are frequently used in patients with cirrhosis as indicators of liver dysfunction severity and are included in severity scores such as Child-Pugh, model for end-stage liver disease, and chronic liver failure. Although these routine laboratory tests have limited value in predicting bleeding or guiding clinical decisions related to the management of bleeding or thrombotic events, they are useful for reflecting the overall severity of liver disease. However, extrahepatic factors that can modify these results should be considered, including the use of vitamin K antagonists, disseminated intravascular coagulation, prolonged cholestasis and vitamin K deficiency. In these clinical contexts, alterations in PT/INR may falsely suggest more severe liver dysfunction than is present.

Viscoelastic hemostatic assays: Viscoelastic hemostatic assays (VHAs) are tests enabling a rapid and comprehensive assessment of coagulation, from clot formation to clot lysis in whole blood[32]. Currently, the most widely used VHAs include thromboelastography (TEG) and rotational thromboelastometry (ROTEM)[33], which use a variety of activators to examine different aspects of the hemostatic system such as clot formation with activation of the intrinsic and extrinsic pathways, contribution of fibrinogen, fibrinolysis and anticoagulant presence[34].

From a practical standpoint, VHAs allow the detection of specific hemostatic defects, potentially guiding tailored transfusion strategies in certain clinical scenarios such as patients undergoing invasive procedures, experiencing active bleeding and liver transplant surgery[32,35]. While both TEG and ROTEM traces look identical, their values are not interchangeable, necessitating separate interpretation of each test[36]. The main parameters reported in TEG and ROTEM, along with their equivalence, are detailed in Figure 2 and Table 1.

Figure 2
Figure 2 Clot formation and lysis sections in rotational thromboelastometry and thromboelastography tracings. Detailed explanation is in Table 1. CT: Clotting time; RT: Reaction time; ROTEM: Rotational thromboelastometry; TEG: Thromboelastography.
Table 1 Comparison between the definitions of the values reported in thromboelastography and rotational thromboelastometry[34,120].
Section
Parameter
Definition and function
Clot initiationROTEM: CTMeasures from test start until fibrin begins to be formed (clot reaches a 2 mm diameter) and reflects time to fibrin formation. Low clotting factors and/or low fibrinogen level, vitamin K antagonists, heparin and DOAC use prolong this measurement. Recommended therapy: Fresh frozen plasma/prothrombin complex concentrate may be considered
TEG: RT
Clot kinetics or fibrin polymerizationROTEM: CFTReflects the speed at which fibrin is formed and how well it binds to platelets. It is the time it takes until the clot reaches 20 mm (arbitrary size). α angle is defined as the slope between section 1 and 2 and is a measure of the rapidity of fibrin polymerization. This parameter is dependent on sufficient fibrinogen, fibrin cross-linking and platelet number and function
TEG: KT
Both: Α angle
Clot strength or stiffnessROTEM: MCFIt is the maximum clot diameter in millimeters and is a combined assessment of fibrinogen and platelet interactions. To differentiate their effects, the standard trace should be compared with the fibrinogen trace. For the clinician, this is the most useful parameter since it represents both primary and secondary hemostasis. Recommended therapy: Platelets (if normal fibrinogen assay), cryoprecipitate or fibrinogen concentrate (if low fibrinogen assay)
TEG: MA
Clot breakdown or fibrinolysisROTEM: CL-30As platelet retraction is a normal phenomenon, some clot strength diminution is expected. It is usually presented as a percentage reduction in the clot strength measure compared to maximal measurement at a given time (30 minutes after CT). Recommended therapy: Consider anti-fibrinolytic agent or reperfusion (if liver transplantation surgery)
TEG: LY30

While VHAs have utility across various clinical scenarios in patients with end-stage liver disease, their primary application lies in liver transplant surgery[37]. In this regard, a small randomized trial reported lower rates of fresh frozen plasma (FFP) transfusion in the TEG group compared to the control group (12.8 U vs 21.5 U, respectively, P < 0.05)[38]. Another randomized trial demonstrated a decrease in median intraoperative transfusion requirements in the thromboelastometry group compared to the control group (3 U vs 7 U, P = 0.005), with FFP and tranexamic acid being administered less frequently in the thromboelastometry group (15% vs 46.3% for FFP, P = 0.002; 27.5% vs 58.5% for tranexamic acid). Conversely, more fibrinogen was infused in the thromboelastometry group (72.5% vs 29.3%, P < 0.001)[39].

In patients with acute-on-chronic liver failure (ACLF), a syndrome associated with a high risk of short-term mortality in those with decompensated cirrhosis[39], hematologic failure has traditionally been defined solely by INR. However, this approach might oversimplify the complexity of coagulation in end-stage liver disease[40]. Therefore, several studies suggest that VHAs could provide more accurate guidance for the use of blood products in this population[41].

A study comparing TEG parameters with conventional coagulation parameters, including PT, INR, and platelet count, revealed several key findings. Notably, platelet count exhibited a strong correlation with maximum amplitude (r = 0.712) and coagulation index (r = 0.613), whereas PT and INR showed weak correlations with these parameters. Consequently, the authors concluded that while TEG might not correlate with the severity of liver disease, VHAs could provide better characterization of the type of coagulopathy in this patient population[40]. The main limitation related to VHAs as point-of-care tests is their poor standardization since apart from manufacturers’ reported reference ranges, there is no published consensus for normal ranges; thus, hospitals should determine their local reference ranges[34]. However, a significant advantage of VHAs is their application in guiding transfusion therapy in procedural settings. A 2015 Italian retrospective study demonstrated that the use of a VHA reduced platelet infusions by 64%, rendering this intervention cost-effective[41].

Guidelines recommendations and statements of VHAs in liver disease include the International Society on Thrombosis and Hemostasis which states that ROTEM/TEG may be an attractive means to reassure the proceduralist that hemostasis is “normal” as these assays are frequently normal despite prolonged coagulation times[17]. The British Society of Hematology recommends the use of a VHA in bleeding patients to guide fibrinogen replacement[34] and the Society of Critical Care Medicine suggest using viscoelastic testing (TEG/ROTEM) over measuring INR, platelets, and fibrinogen in critically ill patients with acute liver failure or ACLF[42].

DISORDERS OF HEMOSTASIS

Hemostasis is the process that maintains the integrity of a closed circulatory system by sealing breaches in vessel walls after vascular damage occurs[43]. Previously, liver cirrhosis was assumed to predispose individuals to bleeding due to abnormalities observed in traditional coagulation tests such as PT, partial thromboplastin time, and INR. However, this paradigm has gradually become obsolete with the recognition of a relative balance between the procoagulant and anticoagulant pathways in cirrhosis[14]. Moreover, accumulating evidence suggests that patients with cirrhosis are not inherently anticoagulated by the disease itself but, rather surprisingly, are at a higher risk of thrombotic events[44,45].

Primary hemostasis involves platelet and vessel wall interactions, during which platelets adhere to the subendothelium at the site of vessel injury, aggregate with one another, and form a platelet plug[28]. Platelets have two main functions: Adhering to damaged vessel walls by interacting with VWF in the endothelium and supporting thrombin generation on their surface. Primary hemostasis alterations involve thrombocytopenia, impaired platelet function, increased production of nitric oxide, and prostacyclin. However, compensatory effects also occur, including elevated levels of VWF[13] and low levels of the enzyme ADAMTS13[14,46]. Notably, ADAMTS13 concentrations have been observed to decrease with increasing severity of liver disease as assessed by the Child-Pugh score[46].

The pathogenesis of thrombocytopenia involves different mechanisms such as spleen sequestration (hypersplenism), increased turnover[47,48], decreased production due to the diminished TPO[49], bone marrow suppression[28], and increased destruction within the reticuloendothelial system. In addition, circulating antibodies against platelets may play a role, predominantly in chronic hepatitis C virus, although their isolated presence does not appear sufficient to induce thrombocytopenia, and their exact role remains uncertain[50,51]. Other potential mechanisms include alcohol-induced bone marrow suppression, deficiencies in vitamin B12 and folate, and adverse reactions to medications such as acetaminophen, amiodarone, heparin, phenytoin, trimethoprim-sulfamethoxazole, valproic acid and vancomycin[52].

TPO, primarily produced by the liver, serves as the primary regulator for platelet production from megakaryocytes in the bone marrow[28]. Its levels are inversely proportional to platelet count as it is predominantly cleared by platelets. However, it has been observed that patients with cirrhosis often exhibit inadequate TPO levels despite low platelet counts[48,53]. Proposed mechanisms for this discrepancy include decreased liver production, impaired TPO regulation, and increased degradation by pooled platelets in the spleen[52,54].

In addition to lower platelet counts, it remains uncertain whether patients with cirrhosis experience thrombocytopathy, manifested as impaired platelet aggregation. Some studies have demonstrated defective function[55,56], while recent data suggest that platelet function is not impaired under flow conditions and when platelet count and hematocrit are normalized[57,58]. However, patients with decompensated cirrhosis and acute kidney injury (AKI) do exhibit altered platelet function compared to those without AKI[58]. Furthermore, it is proposed that platelet location is crucial, as those in the portal circulation demonstrate higher activation compared to those in the peripheral blood, likely due to bacterial translocation and oxidative stress[59].

VWF, a multimeric protein produced in the endothelium that acts as an anchor for platelet adhesion at the site of vessel injury, is increased in liver cirrhosis, presumably due to endothelial damage, reduced liver and peripheral clearance. It has been considered to function as a compensatory mechanism[13]. ADAMTS13, produced mainly by hepatic stellate and endothelial cells, cleaves VWF in its antithrombotic role. In liver cirrhosis, ADAMTS13 activity is lowered leading to increased von Willebrand activity as a compensatory effect to thrombocytopenia. However, it is not well established whether these alterations are primary or secondary[52]. Secondary hemostasis comprises thrombin generation to form a clot consisting of fibrin and platelets. Apart from factor VIII (produced by the endothelium) and VWF, hepatocytes produce most procoagulant and anticoagulant mediators[60].

Due to synthetic liver dysfunction, both anticoagulant and procoagulant factors decrease as the disease progresses, leading to a rebalancing of blood coagulation mechanisms between prohemostatic and antihemostatic factors[3,60]. Prohemostatic abnormalities include increased levels of factors VIIa and IXa (due to elevated circulating tissue factor microparticles), high factor VIII, low levels of antithrombin, protein C, protein S, α2-macrogobulin and heparin cofactor II. Antihemostatic factors include decreased levels of fibrinogen, factors II, V, VII, IX, X, and XI, as well as vitamin K deficiency, hypofibrinogenemia, and dysfibrinogenemia[52,61-63]. Additionally, other elements may play a role, such as increased phosphatidylserine in the membranes of erythrocytes, leukocytes, and platelets, cell-free DNA and neutrophil extracellular traps. These elements can accelerate the coagulation cascade, activate the contact phase of coagulation, and increase thrombin generation[64-66]. However, their specific role in cirrhosis is yet to be determined.

Fibrinolysis is the process by which the fibrin produced in the coagulation cascade is degraded[14]. Its pivotal step involves the conversion of plasminogen to plasmin, a proteolytic enzyme responsible for this degradation. Tissue-plasminogen activator mediates the activation of plasmin when bound to fibrin, but its activity is rapidly countered by inhibitors such as plasminogen activator inhibitor 1, α2-antiplasmin and thrombin-activatable fibrinolysis inhibitor, all produced by the liver[67]. In liver cirrhosis, reduced levels of plasminogen and its activity likely stem from decreased synthesis and increased consumption due to coagulation activation[15]. In addition, levels and activity of tissue-plasminogen activator are increased, plasminogen activator inhibitor 1 activity is relatively diminished compared to tissue-plasminogen activator, and both thrombin-activatable fibrinolysis inhibitor and α2-antiplasmin are decreased. These alterations explain both the hyperactive and hypoactive fibrinolytic state reported in patients with chronic liver disease[68]. Consequently, although contradictory findings have been noted, the fibrinolytic imbalance appears to lean toward hyperfibrinolysis[15].

Despite this newly established equilibrium, it is evident that it is less stable compared to that in healthy individuals, due to noticeable hypo- and hypercoagulable features that can dynamically predominate over time[69]. Standard tests, such as the INR, primarily reflect variations in procoagulant factors and overlook the concurrent decline of anticoagulant factors[70]. A major limitation of these tests is the absence of thrombomodulin, crucial in in vivo coagulation, rendering them inadequate for assessing hemorrhage risk in patients.

Evidence of both procoagulant and anticoagulant alterations lies in thrombin generation tests in cirrhotic patients, which are sensitive to both types of mediators and yield thrombin levels similar to those in healthy patients. These tests incorporate soluble forms of thrombomodulin, the principal activator of protein C, thereby maintaining the balance between factors[71,72]. In addition, when thrombomodulin is added to the plasma of patients with cirrhosis, the thrombin generation ratio exceeds that of healthy controls, indicating partial resistance to thrombomodulin-mediated anticoagulation[14,62,72]. This contrasts with the previously held notion of an auto-anticoagulated state in chronic liver disease patients. This resistance may be attributed to elevated concentrations of factor VIII, a potent thrombin generator[73], and decreased levels of protein C, a potent thrombin generator inhibitor[74].

The concentration of factor VIII increases, likely due to reduced elimination caused by high von Willebrand concentrations, which protect factor VIII from degradation. This reduction in elimination is compounded by low expression of low-density lipoprotein receptor-related protein, a multifunctional endocytic receptor inadequately expressed in cirrhotic patients. This receptor collaborates in the cellular uptake and degradation of factor VIII, resulting in decreased clearance[75]. Moreover, this imbalance correlates proportionally with the severity of cirrhosis, as assessed by the Child-Pugh score[61,62]. Despite decreased fibrinogen concentrations, studies have demonstrated that fibrin clot quality remains unimpaired[76,77].

Many studies have highlighted the limited relevance of routine standard coagulation tests, particularly the INR, in accurately predicting bleeding risks after invasive procedures such as percutaneous[78,79], transjugular[80,81] and laparoscopic liver biopsy[82,83], therapeutic paracentesis[84], colonoscopy with polypectomy[85], percutaneous endoscopic gastrostomy[86], bronchoscopy, kidney biopsy, central venous catheter placement, and arteriography[81], as well as coronary artery catheterization[87].

The European Association for the Study of Liver (EASL) advises against using traditional hemostasis tests to predict procedural bleeding risk before low-risk procedures. They suggest their use solely to assess disease severity and establish an initial benchmark for management in the event of post-procedural bleeding. However, they caution against relying on these tests to predict post-procedural bleeding in high-risk patients, although they may provide a baseline status and aid physicians in managing bleeding events[88]. In contrast, the American Association for the Study of Liver Disease (AASLD) states that there is no data-driven specific INR or platelet cutoff indicating reliably increased procedural bleeding risk. They recommend against using the INR to gauge procedural bleeding risk in cirrhosis patients not taking vitamin K antagonists[89].

Despite this, certain circumstances have been associated with increased bleeding tendencies. AKI, a common complication in decompensated cirrhosis[90], has been linked to increased bleeding tendencies, particularly after invasive procedures[91]. Platelet function is altered in this context, yet coagulation tests show increased thrombin generation, indicating hypercoagulability[58]. Moreover, both hypofibrinolytic and hyperfibrinolytic changes have been observed to coexist[92], with the former primarily associated with diminished factor XIII concentration and fibrinolysis activation. Importantly, these abnormalities tend to improve upon resolution of AKI[58,93].

Furthermore, patients with cirrhosis and severe infection or sepsis exhibit dynamic coagulation profiles, leading to alterations that may result in hypocoagulable, hypercoagulable, or mixed phenotypes[94]. ACLF, characterized by systemic inflammation and organ system failure, is associated with significant systemic inflammation and an increased risk of death[95]. Such patients are more likely to exhibit hypocoagulable characteristics on TEG than those with acute decompensation, although no difference in bleeding events has been observed[96]. However, evidence also suggests that this “new” equilibrium is maintained in critically ill patients with ACLF[92]. The hemostatic balance in patients with cirrhosis is depicted in Figure 3.

Figure 3
Figure 3 Hemostatic balance in patients with chronic liver disease. Created with BioRender.com. TAFI: Thrombin-activatable fibrinolysis inhibitor; tPA: Tissue-plasminogen activator; PAI: Plasminogen activator inhibitor.
THROMBOSIS

The complex interplay of factors discussed previously contributes to a prothrombotic state in patients with cirrhosis. Venous thromboembolism (VTE), encompassing deep venous thrombosis (DVT) and pulmonary embolism (PE), stands as a common cause of mortality and morbidity. In the general population, the incidence rate ranges from 1 to 2 per 1000 person-years[97]. A Nationwide United States study compared the prevalence of VTE among patients with and without cirrhosis, assessing its impact on in-hospital mortality and length of stay. This study revealed that patients with cirrhosis under the age of 45 years were at higher risk for VTE compared to those without liver disease, warranting consideration for VTE prophylaxis. VTE was equally associated with increased mortality among patients with compensated cirrhosis [odds ratio (OR) = 2.16; 95% confidence interval (CI): 1.96-2.38] or decompensated cirrhosis (OR = 1.66, 95%CI: 1.47-1.87). Moreover, VTE was linked to prolonged hospitalization[98].

Additionally, a large population-based case-control study, including 99444 patients with VTE and a control group, demonstrated a substantially increased risk of VTE among patients with liver disease, approximately twice that of non-liver disease patients. Among cirrhosis cases, the adjusted relative risk was 2.02 (95%CI: 1.78-2.31) for DVT and 1.41 (95%CI: 1.20-1.65) for PE (P < 0.0001)[99] (Figure 4). Other retrospective studies have shown that patients with liver disease develop DVT and/or PE with an incidence ranging from 0.5% to 8.2%[100]. Portal vein thrombosis (PVT) is also a fairly common complication of liver cirrhosis; occlusive PVT can be associated with poor prognosis, especially in patients with a prior history of bleeding[101].

Figure 4
Figure 4 Relative risk of venous thromboembolism in patients with cirrhosis vs healthy individuals. Created with BioRender.com. VTE: Venous thromboembolism; PE: Pulmonary embolism; PVT: Portal vein thrombosis; DVT: Deep venous thrombosis; RR: Relative risk.

Thrombotic complications are more prevalent in decompensated cirrhosis and may correlate with disease severity. Patients with compensated cirrhosis have an estimated incidence of PVT of 1%, while those being evaluated for liver transplantation have an incidence ranging from approximately 8% to 25%[100]. Additionally, in post-transplant patients, PVT has been linked to a 30% increase in mortality[97]. In a prospective study conducted in France and Belgium, 1243 adults with cirrhosis without PVT were enrolled. A five-year cumulative incidence of PVT of 10.7% was observed, but the development of PVT was not associated with subsequent disease progression[102]. Furthermore, in a recent meta-analysis encompassing 74 studies, approximately one-seventh of cirrhotic patients were found to have PVT, with one-tenth developing it. The progression of liver cirrhosis and portal hypertension appears to parallel the risk of PVT. Based on cross-sectional data, factors such as Child-Pugh class B/C, higher D-dimer levels, ascites, and the use of non-selective beta-blockers were associated with the presence of PVT in liver cirrhosis[103].

Numerous investigators have explored additional risk factors linked to peripheral and portal thrombosis, identifying age, gender, comorbidities, nutritional status, and the presence of indwelling central lines as potential contributors[100]. One consistent finding across studies has been the association with low albumin levels. A retrospective case-control study conducted in the United States revealed a five-fold increase in the risk of VTE when albumin levels were below 1.9 mg/dL compared to patients with normal albumin levels[104].

MANAGEMENT OF THROMBOSIS

Failure to detect and treat thrombosis can result in mesenteric ischemia, chronic cavernous transformation, and complications of portal hypertension. In patients with cirrhosis, the development of PVT often progresses insidiously and may remain undetected until incidentally discovered[105]. The AASLD recommends that in patients with cirrhosis, it is mandatory to initially rule out malignant venous obstruction attributable to hepatocellular carcinoma with appropriate contrast-enhanced imaging studies[89].

There are no specific guidelines for the treatment of DVT or PE in patients with liver disease, so the safety of anticoagulation in these patients is extrapolated from the treatment of patients with PVT. Early anticoagulation for PVT represents a therapeutic consideration, supported by retrospective studies that found excellent recanalization rates in patients maintained on anticoagulation[100]. However, the possibility of spontaneous resolution of partial PVT raises questions about the necessity of anticoagulation for its treatment. Additionally, a relatively low recanalization rate of complete PVT after anticoagulation therapy suggests its limited usefulness in patients with complete PVT[101]. Due to the lack of well-designed prospective trials, the AASLD does not provide a recommendation for or against anticoagulation in patients with PVT and cirrhosis, emphasizing the need for individualized treatment decisions[89,100,106]. Moreover, there is insufficient data to support pretransplant treatment of PVT to improve post-transplant outcomes[89].

In a study of 55 patients receiving both low-molecular-weight heparin and vitamin K antagonists for PVT, there were 5 clinical bleeding events related to anticoagulation during 19 months of follow-up. An assessment of increased bleeding risk revealed a statistically significant association with platelet counts below 50 and a non-statistically significant association with the use of vitamin K antagonists[107]. Regarding PVT, previous meta-analyses have demonstrated the safety and efficacy of anticoagulation in recanalization in patients with cirrhosis. However, given the uncertainty regarding the role of anticoagulation in improving survival by recanalization of PVT in patients with cirrhosis, a recent meta-analysis (The IMPORTAL study) compared anticoagulation vs no treatment in patients with cirrhosis and PVT. Five studies were included, and the results indicated that anticoagulation reduced all-cause mortality in patients with cirrhosis and PVT (hazard ratio = 0.59; 95%CI: 0.49-0.70), independently of thrombosis severity and recanalization, but at the expense of increasing non-portal hypertension-related bleeding, with a significantly higher bleeding rate in the anticoagulation group[108].

Regarding VTE prophylaxis, a systematic review was conducted to analyze bleeding and VTE events in cirrhosis patients and controls receiving VTE prophylaxis. Ten studies were included, revealing bleeding and VTE events in 8.2% and 2.8% of cases, respectively. Surprisingly, the administration of VTE prophylaxis did not appear to reduce the incidence of VTE events. Consequently, the current evidence is deemed insufficient to either recommend or discourage the use of VTE prophylaxis, primarily due to the lack of quality and consistency in available data[109]. In this complex scenario, to date, there remains no clear consensus regarding the primary prevention of thrombosis in cirrhosis[60].

BLEEDING

Bleeding emerges as a multifaceted complication arising from diminished synthesis of clotting factors, platelet dysfunction, and compromised vascular integrity. Thrombocytopenia, a common manifestation, further heightens the risk of bleeding. The convergence of hematology and hepatology is extensive due to the liver’s central role in synthesizing pro- and anti-coagulation factors and clearing byproducts of hemostasis and coagulation[110]. Although thrombotic risk may be more severe, numerous hematologic aberrations in cirrhosis contribute to bleeding complications, making them prevalent[60]. In this context, variceal bleeding is the most common and concerning type of bleed, occurring in 25% to 35% of patients with cirrhosis. Nonvariceal bleeding is almost equally prevalent, noted in 20% of patients admitted with decompensated cirrhosis[100]. Other common presentations include bruising, purpura, epistaxis, menorrhagia, and procedural bleeding[60].

Variceal hemorrhage is associated with higher morbidity and mortality than other causes of gastrointestinal bleeding, as well as increased hospital costs[111]. However, variceal bleeding is primarily driven by portal pressure and is not attributable to failure of the hemostatic system[69]. The AASLD has defined 3 causes of bleeding in patients with cirrhosis[89]. First, portal hypertension-related bleeding complications, such as variceal bleeding, which appear to be unrelated to hemostatic failure, as mentioned above. Second, provoked bleeds may occur during procedures due to vessel mechanical injury. Third, spontaneous or unprovoked bleeds that may be related to hemostatic failure include bruising, mucosal bleeding, and oozing from puncture sites[69,89]. The presence of portal hypertension, infection, or renal dysfunction correlates with bleeding events in patients with cirrhosis[100]. The use of medication also plays a significant role. In a recent study, anti-platelet use was associated with increased bleeding and decompensation events among privately insured cirrhosis patients. Non-steroidal anti-inflammatory drugs use was associated with significant early bleeding but not with decompensations. Anticoagulants were not associated with bleeding or decompensation outcomes[112].

Procedural bleeding

Multiple factors such as patient-specific bleeding diatheses and the type of anesthesia, may contribute to the overall estimation of bleeding risk in procedural settings[113]. Current EASL and AASLD guidelines emphasize the importance that FFP, prothrombin complex concentrates, and fibrinogen sources should not be used to reduce periprocedural bleeding risk[88,89]. EASL also recommends a case-by-case consideration of platelet transfusion/TPO receptor agonist therapy for patients undergoing high-risk procedures with a platelet count of < 50 × 103/μL[88,114]. However, this requires time and planning, therefore, these interventions only apply to elective procedures. The risk lies predominantly on inherent procedural risk for which little preemptive action can be taken, mainly in the emergency setting[114]. Many non-surgical invasive procedures, including liver biopsy, paracentesis, thoracentesis, transesophageal echocardiography, central vein catheter insertion, endoscopic esophageal varices ligation, and transarterial chemoembolization, are safe and not associated with increased bleeding risk in cirrhosis[60].

Bleeding rates for paracentesis (0%-3.3%) and thoracentesis (2%) in cirrhosis patients are low and do not require routine preprocedural coagulation assessment, in decompensated cirrhosis or even in ACLF[115]. Common coagulation tests (PT/INR, partial thromboplastin time, platelet count, and fibrinogen) are often inaccurate predictors of bleeding risk as they do not reflect more clinically relevant hemodynamic risk factors[60]. In terms of hemoglobin, packed red blood cell transfusion before high-risk procedures or major surgery is recommended to maintain hematocrit > 25% in cirrhosis, as this is thought to improve platelet margination[116]. If packed red blood cell transfusion is required in the setting of active bleeding, a restrictive transfusion threshold of hemoglobin of 7 g/dL is known to significantly improve outcomes of upper gastrointestinal bleeding, particularly in patients with cirrhosis[117]. We must also take into consideration that blood product transfusions deliver substantial volumes of oncotically active fluid, which can exacerbate portal hypertension through circulatory overload as well as increase the risk of infection and transfusion reactions[60].

As for platelets, in the event of an upcoming high-risk procedure or surgery, targeting a platelet count of 50000-60000 either via TPO administration or platelet transfusion is suggested[60,116]. In the specific setting of liver biopsy, due to the conflicting data in the literature, current AASLD guideline recommendations do not specify a particular INR or platelet cutoff at which the risk of bleeding is substantially increased but suggest individualized approaches and assessment of other risk factors that may increase bleeding risk[89].

Esophageal variceal bleeding is a severe complication of portal hypertension in cirrhosis patients, with a high bleeding-related mortality rate of up to 20%. Treatment of patients with gastroesophageal varices includes prevention of the initial bleeding episode (primary prophylaxis), either with non-selective beta-blockers or/and endoscopic band ligation (EBL), the control of active hemorrhage, and the prevention of recurrent bleeding after a first episode (secondary prophylaxis)[111,118]. Bleeding from banding ulcers represents the main severe complication of EBL, occurring in 2.3% to 10% of patients. A retrospective multicenter study analyzing data from 1178 prophylactic EBL procedures across two large Vietnamese centers demonstrated that EBL is safe, with procedure-related bleeding rates being rare (2.6%), even in patients with thrombocytopenia < 50 × 103/μL or high INR ≥ 1.5. Only high model for end-stage liver disease scores, particularly high bilirubin levels, seemed to be associated with an increased risk of EBL-related bleeding[118]. However, further analysis of these data is needed, along with the development of prospective studies. Table 2 outlines the key interventions for managing bleeding in cirrhosis and includes the evidence supporting each treatment.

Table 2 Summary of blood products and/or drugs recommendations in bleeding in cirrhosis.
Choice of agent
Recommendations
Guidelines/evidence
Platelet transfusionProphylaxisCommon gastrointestinal procedures1AGA suggests against the routine use of blood products for bleeding prophylaxisAGA[116,121]
High risk proceduresDecisions about prophylactic blood transfusions should include discussions about potential benefits and risks in consultation with a hematologist. Threshold: > 50 × 109/L
Acute bleedingPlatelet transfusions should not be administered based on platelet count targets because there is no evidence of benefit of such transfusions in AVHAASLD[122]
In patients with cirrhosis and active bleeding (out of the setting of AVH), thrombocytopenia (if platelet count < 50 × 109/L)Clinical Gastroenterology and Hepatology[123], 2023
TPO-RAProphylaxisCommon gastrointestinal procedures1AGA suggests against the routine use of TPO-RAs for bleeding prophylaxisAGA[121]
High risk proceduresPatients who place a high value on the uncertain reduction of procedural bleeding events and a low value on the increased risk for PVT can reasonably select a TPO-RA
Acute bleedingNot appropriate for acute settingClinical Gastroenterology and Hepatology[123], 2023
FFPProphylaxisCommon gastrointestinal procedures1AGA suggests against the routine use of blood products for bleeding prophylaxisAGA[121]
High risk proceduresDecisions about prophylactic blood transfusions should include discussions about potential benefits and risks in consultation with a hematologist
Acute bleedingFresh frozen plasma should not be administered based on INR because there is no evidence of benefit of such transfusions in AVHAASLD[122]
Restricted to hemorrhagic shock to compensate blood lossClinical Gastroenterology and Hepatology[123], 2023
FibrinogenProphylaxisNo routine preprocedure correctionAASLD[89]
Acute bleedingThe following transfusion thresholds for management of active bleeding or high-risk procedures may optimize clot formation in advanced liver disease: Fibrinogen > 120 mg/dLAGA[116]
rFVIIaNot recommended for bleeding episodes in patients with Child-Pugh A cirrhosis. Efficacy of rFVIIa was considered uncertain in bleeding episodes in patients with Child-Pugh B and C cirrhosisEuropean Consensus Critical Care[124], 2006
PCCThe role of PCC is not yet defined. Limited data based on retrospective studiesAGA[116]
DesmopressinThe agent lacks a sound evidence-based foundation but may be useful in patients with concomitant renal failureAGA[116]
Antifibrinolytic agentsAnti-fibrinolytic therapy may be considered in patients with persistent bleeding from mucosal oozing or puncture wound bleeding consistent with impaired clot integrityAGA[125]
RCT shows tranexamic acid reduces failure to control bleeding and rebleeding in advanced cirrhosis with UGIB. However, further studies and robust evidence are needed to make a definitive recommendationHepatology[125], 2024
Specific interventions in ACLF

Despite intensive care, critically ill patients with cirrhosis are at a high risk of mortality within 1-3 months[115]. In the CANONIC study, kidneys were the most commonly affected organs (55.8%), followed by the liver (43.6%), coagulation (27.7%), the brain (24.1%), circulation (16.8%), and the lungs (9.2%)[119]. ACLF is characterized by systemic inflammation, and the acute phase reaction induced by this process alters blood concentrations of many of the components of hemostasis, including fibrinogen, platelets, and endothelial factors[114]. INR is dependent on procoagulant factors I, II, V, VII, and X and does not account for the rebalanced coagulation in patients with ACLF and decompensated cirrhosis due to anticoagulant deficiencies. In contrast, a VHA provides a functional evaluation of altered pro- and anti-coagulant pathways, measuring platelet function, hyperfibrinolysis, and premature clot dissolution in real-time. However, optimal cutoffs to guide platelet, cryoprecipitate, or four-factor prothrombin complex concentrate in ACLF have not been studied[115].

Navigating a delicate equilibrium involves addressing bleeding risks with cautious use of blood products and considering prophylaxis against thrombosis. Striking this balance is pivotal in optimizing outcomes for cirrhotic patients. Deeper comprehension of ACLF physiopathology is also necessary in the near future. Advances in understanding these complexities are crucial for refining therapeutic strategies in cirrhotic patients with hematologic challenges. Further research is crucial to refine therapeutic strategies for this intricate hematological landscape in liver cirrhosis.

CONCLUSION

Patients with cirrhosis show numerous hematological abnormalities with a multifactorial origin. Thrombocytopenia is the most frequent cytopenia, but these patients are not particularly prone to bleeding and when it occurs other critical factors such as portal hypertension, endothelial dysfunction, mechanical injury to vessels, disseminated intravascular coagulation, endotoxemia and renal injury should be considered. VTE also poses significant consequences. Finally, these disturbances become more critical with increasing cirrhosis severity, further narrowing the fragile new balance.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Corresponding Author’s Membership in Professional Societies: European Association for the Study of Liver; American Association for the Study of Liver Disease.

Specialty type: Gastroenterology and hepatology

Country of origin: Mexico

Peer-review report’s classification

Scientific Quality: Grade C

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Díaz J S-Editor: Wang JJ L-Editor: Webster JR P-Editor: Wang WB

References
1.  Devarbhavi H, Asrani SK, Arab JP, Nartey YA, Pose E, Kamath PS. Global burden of liver disease: 2023 update. J Hepatol. 2023;79:516-537.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 308]  [Reference Citation Analysis (1)]
2.  GBD 2017 Cirrhosis Collaborators. The global, regional, and national burden of cirrhosis by cause in 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol. 2020;5:245-266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 762]  [Cited by in F6Publishing: 829]  [Article Influence: 207.3]  [Reference Citation Analysis (2)]
3.  Lisman T, Porte RJ. Rebalanced hemostasis in patients with liver disease: evidence and clinical consequences. Blood. 2010;116:878-885.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 404]  [Cited by in F6Publishing: 417]  [Article Influence: 29.8]  [Reference Citation Analysis (0)]
4.  Qamar AA, Grace ND, Groszmann RJ, Garcia-Tsao G, Bosch J, Burroughs AK, Maurer R, Planas R, Escorsell A, Garcia-Pagan JC, Patch D, Matloff DS, Makuch R; Portal Hypertension Collaborative Group. Platelet count is not a predictor of the presence or development of gastroesophageal varices in cirrhosis. Hepatology. 2008;47:153-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 102]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
5.  Pursnani KG, Sillin LF, Kaplan DS. Effect of transjugular intrahepatic portosystemic shunt on secondary hypersplenism. Am J Surg. 1997;173:169-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
6.  Hutson DG, Zeppa R, Levi JU, Schiff ER, Livingstone AS, Fink P. The effect of the distal splenorenal shunt on hypersplenism. Ann Surg. 1977;185:605-612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 33]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
7.  Soper NJ, Rikkers LF. Effect of operations for variceal hemorrhage on hypersplenism. Am J Surg. 1982;144:700-703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 33]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
8.  SULLIVAN BH Jr, TUMEN HJ. The effect of portacaval shunt on thrombocytopenia associated with portal hypertension. Ann Intern Med. 1961;55:598-603.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 23]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
9.  Mutchnick MG, Lerner E, Conn HO. Effect of portacaval anastomosis on hypersplenism. Dig Dis Sci. 1980;25:929-938.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
10.  Bashour FN, Teran JC, Mullen KD. Prevalence of peripheral blood cytopenias (hypersplenism) in patients with nonalcoholic chronic liver disease. Am J Gastroenterol. 2000;95:2936-2939.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 159]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
11.  Qamar AA, Grace ND, Groszmann RJ, Garcia-Tsao G, Bosch J, Burroughs AK, Ripoll C, Maurer R, Planas R, Escorsell A, Garcia-Pagan JC, Patch D, Matloff DS, Makuch R, Rendon G; Portal Hypertension Collaborative Group. Incidence, prevalence, and clinical significance of abnormal hematologic indices in compensated cirrhosis. Clin Gastroenterol Hepatol. 2009;7:689-695.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 175]  [Article Influence: 11.7]  [Reference Citation Analysis (1)]
12.  Lingas EC. Hematological Abnormalities in Cirrhosis: A Narrative Review. Cureus. 2023;15:e39239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (1)]
13.  Lisman T, Bongers TN, Adelmeijer J, Janssen HL, de Maat MP, de Groot PG, Leebeek FW. Elevated levels of von Willebrand Factor in cirrhosis support platelet adhesion despite reduced functional capacity. Hepatology. 2006;44:53-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 417]  [Cited by in F6Publishing: 391]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
14.  Tripodi A, Mannucci PM. The coagulopathy of chronic liver disease. N Engl J Med. 2011;365:147-156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 963]  [Cited by in F6Publishing: 900]  [Article Influence: 69.2]  [Reference Citation Analysis (1)]
15.  Rijken DC, Kock EL, Guimarães AH, Talens S, Darwish Murad S, Janssen HL, Leebeek FW. Evidence for an enhanced fibrinolytic capacity in cirrhosis as measured with two different global fibrinolysis tests. J Thromb Haemost. 2012;10:2116-2122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 62]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
16.  Qamar AA, Grace ND. Abnormal hematological indices in cirrhosis. Can J Gastroenterol. 2009;23:441-445.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 42]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
17.  Roberts LN, Lisman T, Stanworth S, Hernandez-Gea V, Magnusson M, Tripodi A, Thachil J. Periprocedural management of abnormal coagulation parameters and thrombocytopenia in patients with cirrhosis: Guidance from the SSC of the ISTH. J Thromb Haemost. 2022;20:39-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 44]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
18.  Gonzalez-Casas R, Jones EA, Moreno-Otero R. Spectrum of anemia associated with chronic liver disease. World J Gastroenterol. 2009;15:4653-4658.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 113]  [Cited by in F6Publishing: 119]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
19.  Paternostro R, Kapzan L, Mandorfer M, Schwarzer R, Benedikt S, Viveiros A, Bauer D, Ferlitsch M, Zoller H, Trauner M, Ferlitsch A. Anemia and iron deficiency in compensated and decompensated cirrhosis: Prevalence and impact on clinical outcomes. J Gastroenterol Hepatol. 2020;35:1619-1627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
20.  GBD 2021 Anaemia Collaborators. Prevalence, years lived with disability, and trends in anaemia burden by severity and cause, 1990-2021: findings from the Global Burden of Disease Study 2021. Lancet Haematol. 2023;10:e713-e734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 35]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
21.  Muro N, Bujanda L, Sarasqueta C, Gil I, Hijona E, Cosme A, Arenas J Jr, Elosegui ME, Sarasola M, Calpasoro J, Arenas JI. [Plasma levels of folate and vitamin B(12) in patients with chronic liver disease]. Gastroenterol Hepatol. 2010;33:280-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 10]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
22.  Maruyama S, Hirayama C, Yamamoto S, Koda M, Udagawa A, Kadowaki Y, Inoue M, Sagayama A, Umeki K. Red blood cell status in alcoholic and non-alcoholic liver disease. J Lab Clin Med. 2001;138:332-337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 65]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
23.  Roy A, Rodge G, Goenka MK. Spur Cell Anaemia in Cirrhosis: A Narrative Review. J Clin Exp Hepatol. 2023;13:500-508.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
24.  Vassiliadis T, Mpoumponaris A, Vakalopoulou S, Giouleme O, Gkissakis D, Grammatikos N, Soufleris K, Kakafika A, Tziomalos K, Patsiaoura K, Papanikolaou V, Evgenidis N. Spur cells and spur cell anemia in hospitalized patients with advanced liver disease: Incidence and correlation with disease severity and survival. Hepatol Res. 2010;40:161-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 20]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
25.  Koschade SE, Moser LM, Sokolovskiy A, Michael FA, Serve H, Brandts CH, Finkelmeier F, Zeuzem S, Trebicka J, Ferstl P, Ballo O. Bone Marrow Assessment in Liver Cirrhosis Patients with Otherwise Unexplained Peripheral Blood Cytopenia. J Clin Med. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
26.  Giannini EG. Review article: thrombocytopenia in chronic liver disease and pharmacologic treatment options. Aliment Pharmacol Ther. 2006;23:1055-1065.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 131]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
27.  Kujovich JL. Coagulopathy in liver disease: a balancing act. Hematology Am Soc Hematol Educ Program. 2015;2015:243-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 68]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
28.  Afdhal N, McHutchison J, Brown R, Jacobson I, Manns M, Poordad F, Weksler B, Esteban R. Thrombocytopenia associated with chronic liver disease. J Hepatol. 2008;48:1000-1007.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 380]  [Cited by in F6Publishing: 384]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
29.  Daneshi M, Rashidpanah J, Narouei F.   An Overview of Hemostasis. In: Dorgalaleh A. Congenital Bleeding Disorders. Cham: Springer, 2023.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Dorgalaleh A, Favaloro EJ, Bahraini M, Rad F. Standardization of Prothrombin Time/International Normalized Ratio (PT/INR). Int J Lab Hematol. 2021;43:21-28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 41]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
31.  Turco L, de Raucourt E, Valla DC, Villa E. Anticoagulation in the cirrhotic patient. JHEP Rep. 2019;1:227-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 46]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
32.  Hartmann J, Hermelin D, Levy JH. Viscoelastic testing: an illustrated review of technology and clinical applications. Res Pract Thromb Haemost. 2023;7:100031.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 14]  [Reference Citation Analysis (0)]
33.  Faraoni D, DiNardo JA. Viscoelastic hemostatic assays: Update on technology and clinical applications. Am J Hematol. 2021;96:1331-1337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
34.  Curry NS, Davenport R, Pavord S, Mallett SV, Kitchen D, Klein AA, Maybury H, Collins PW, Laffan M. The use of viscoelastic haemostatic assays in the management of major bleeding: A British Society for Haematology Guideline. Br J Haematol. 2018;182:789-806.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 124]  [Article Influence: 20.7]  [Reference Citation Analysis (0)]
35.  Whiting D, DiNardo JA. TEG and ROTEM: technology and clinical applications. Am J Hematol. 2014;89:228-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 367]  [Cited by in F6Publishing: 411]  [Article Influence: 41.1]  [Reference Citation Analysis (0)]
36.  Nielsen VG. A comparison of the Thrombelastograph and the ROTEM. Blood Coagul Fibrinolysis. 2007;18:247-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 98]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
37.  Sakai T. Viscoelastic testing in liver transplantation. Transfusion. 2020;60 Suppl 6:S61-S69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
38.  Wang SC, Shieh JF, Chang KY, Chu YC, Liu CS, Loong CC, Chan KH, Mandell S, Tsou MY. Thromboelastography-guided transfusion decreases intraoperative blood transfusion during orthotopic liver transplantation: randomized clinical trial. Transplant Proc. 2010;42:2590-2593.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 185]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
39.  Bonnet A, Gilquin N, Steer N, Gazon M, Quattrone D, Pradat P, Maynard M, Mabrut JY, Aubrun F. The use of a thromboelastometry-based algorithm reduces the need for blood product transfusion during orthotopic liver transplantation: A randomised controlled study. Eur J Anaesthesiol. 2019;36:825-833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 27]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
40.  Shin KH, Kim IS, Lee HJ, Kim HH, Chang CL, Hong YM, Yoon KT, Cho M. Thromboelastographic Evaluation of Coagulation in Patients With Liver Disease. Ann Lab Med. 2017;37:204-212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 33]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
41.  Debernardi Venon W, Ponzo P, Sacco M, Mengozzi G, Raso S, Valpreda A, Rizzetto M, Marzano A. Usefulness of thromboelastometry in predicting the risk of bleeding in cirrhotics who undergo invasive procedures. Eur J Gastroenterol Hepatol. 2015;27:1313-1319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 28]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
42.  Nanchal R, Subramanian R, Karvellas CJ, Hollenberg SM, Peppard WJ, Singbartl K, Truwit J, Al-Khafaji AH, Killian AJ, Alquraini M, Alshammari K, Alshamsi F, Belley-Cote E, Cartin-Ceba R, Dionne JC, Galusca DM, Huang DT, Hyzy RC, Junek M, Kandiah P, Kumar G, Morgan RL, Morris PE, Olson JC, Sieracki R, Steadman R, Taylor B, Alhazzani W. Guidelines for the Management of Adult Acute and Acute-on-Chronic Liver Failure in the ICU: Cardiovascular, Endocrine, Hematologic, Pulmonary and Renal Considerations: Executive Summary. Crit Care Med. 2020;48:415-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
43.  Furie B, Furie BC. Mechanisms of thrombus formation. N Engl J Med. 2008;359:938-949.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1183]  [Cited by in F6Publishing: 1130]  [Article Influence: 70.6]  [Reference Citation Analysis (0)]
44.  Northup PG, McMahon MM, Ruhl AP, Altschuler SE, Volk-Bednarz A, Caldwell SH, Berg CL. Coagulopathy does not fully protect hospitalized cirrhosis patients from peripheral venous thromboembolism. Am J Gastroenterol. 2006;101:1524-8; quiz 1680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 274]  [Cited by in F6Publishing: 253]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
45.  Ambrosino P, Tarantino L, Di Minno G, Paternoster M, Graziano V, Petitto M, Nasto A, Di Minno MN. The risk of venous thromboembolism in patients with cirrhosis. A systematic review and meta-analysis. Thromb Haemost. 2017;117:139-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 131]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
46.  Uemura M, Fujimura Y, Matsumoto M, Ishizashi H, Kato S, Matsuyama T, Isonishi A, Ishikawa M, Yagita M, Morioka C, Yoshiji H, Tsujimoto T, Kurumatani N, Fukui H. Comprehensive analysis of ADAMTS13 in patients with liver cirrhosis. Thromb Haemost. 2008;99:1019-1029.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 87]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
47.  Rauber P, Lammert F, Grotemeyer K, Appenrodt B. Immature platelet fraction and thrombopoietin in patients with liver cirrhosis: A cohort study. PLoS One. 2018;13:e0192271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
48.  Rios R, Sangro B, Herrero I, Quiroga J, Prieto J. The role of thrombopoietin in the thrombocytopenia of patients with liver cirrhosis. Am J Gastroenterol. 2005;100:1311-1316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 92]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
49.  Miller JB, Figueroa EJ, Haug RM, Shah NL. Thrombocytopenia in Chronic Liver Disease and the Role of Thrombopoietin Agonists. Gastroenterol Hepatol (N Y). 2019;15:326-332.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Kajihara M, Okazaki Y, Kato S, Ishii H, Kawakami Y, Ikeda Y, Kuwana M. Evaluation of platelet kinetics in patients with liver cirrhosis: similarity to idiopathic thrombocytopenic purpura. J Gastroenterol Hepatol. 2007;22:112-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 44]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
51.  Panzer S, Seel E, Brunner M, Körmöczi GF, Schmid M, Ferenci P, Peck-Radosavljevic M. Platelet autoantibodies are common in hepatitis C infection, irrespective of the presence of thrombocytopenia. Eur J Haematol. 2006;77:513-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 46]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
52.  Zermatten MG, Fraga M, Moradpour D, Bertaggia Calderara D, Aliotta A, Stirnimann G, De Gottardi A, Alberio L. Hemostatic Alterations in Patients With Cirrhosis: From Primary Hemostasis to Fibrinolysis. Hepatology. 2020;71:2135-2148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 41]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
53.  Ishikawa T, Ichida T, Matsuda Y, Sugitani S, Sugiyama M, Kato T, Miyazaki H, Asakura H. Reduced expression of thrombopoietin is involved in thrombocytopenia in human and rat liver cirrhosis. J Gastroenterol Hepatol. 1998;13:907-913.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 44]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
54.  Violi F, Basili S, Raparelli V, Chowdary P, Gatt A, Burroughs AK. Patients with liver cirrhosis suffer from primary haemostatic defects? J Hepatol. 2011;55:1415-1427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 120]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
55.  Laffi G, Marra F, Gresele P, Romagnoli P, Palermo A, Bartolini O, Simoni A, Orlandi L, Selli ML, Nenci GG. Evidence for a storage pool defect in platelets from cirrhotic patients with defective aggregation. Gastroenterology. 1992;103:641-646.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 72]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
56.  Vinholt PJ, Hvas AM, Nielsen C, Söderström AC, Sprogøe U, Fialla AD, Nybo M. Reduced platelet activation and platelet aggregation in patients with alcoholic liver cirrhosis. Platelets. 2018;29:520-527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
57.  Lisman T, Adelmeijer J, de Groot PG, Janssen HL, Leebeek FW. No evidence for an intrinsic platelet defect in patients with liver cirrhosis--studies under flow conditions. J Thromb Haemost. 2006;4:2070-2072.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 41]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
58.  Zanetto A, Rinder HM, Campello E, Saggiorato G, Deng Y, Ciarleglio M, Wilson FP, Senzolo M, Gavasso S, Bulato C, Simioni P, Garcia-Tsao G. Acute Kidney Injury in Decompensated Cirrhosis Is Associated With Both Hypo-coagulable and Hyper-coagulable Features. Hepatology. 2020;72:1327-1340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 56]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
59.  Queck A, Carnevale R, Uschner FE, Schierwagen R, Klein S, Jansen C, Meyer C, Praktiknjo M, Thomas D, Strassburg C, Zeuzem S, Violi F, Trebicka J. Role of portal venous platelet activation in patients with decompensated cirrhosis and TIPS. Gut. 2020;69:1535-1536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 33]  [Article Influence: 8.3]  [Reference Citation Analysis (1)]
60.  McMurry HS, Jou J, Shatzel J. The hemostatic and thrombotic complications of liver disease. Eur J Haematol. 2021;107:383-392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
61.  Tripodi A, Primignani M, Lemma L, Chantarangkul V, Dell'Era A, Iannuzzi F, Aghemo A, Mannucci PM. Detection of the imbalance of procoagulant versus anticoagulant factors in cirrhosis by a simple laboratory method. Hepatology. 2010;52:249-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 102]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
62.  Tripodi A, Primignani M, Chantarangkul V, Dell'Era A, Clerici M, de Franchis R, Colombo M, Mannucci PM. An imbalance of pro- vs anti-coagulation factors in plasma from patients with cirrhosis. Gastroenterology. 2009;137:2105-2111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 386]  [Cited by in F6Publishing: 366]  [Article Influence: 24.4]  [Reference Citation Analysis (0)]
63.  Rautou PE, Vion AC, Luyendyk JP, Mackman N. Circulating microparticle tissue factor activity is increased in patients with cirrhosis. Hepatology. 2014;60:1793-1795.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
64.  Gould TJ, Vu TT, Swystun LL, Dwivedi DJ, Mai SH, Weitz JI, Liaw PC. Neutrophil extracellular traps promote thrombin generation through platelet-dependent and platelet-independent mechanisms. Arterioscler Thromb Vasc Biol. 2014;34:1977-1984.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 289]  [Cited by in F6Publishing: 299]  [Article Influence: 29.9]  [Reference Citation Analysis (0)]
65.  Liaw PC, Ito T, Iba T, Thachil J, Zeerleder S. DAMP and DIC: The role of extracellular DNA and DNA-binding proteins in the pathogenesis of DIC. Blood Rev. 2016;30:257-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 102]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
66.  Wu X, Yao Z, Zhao L, Zhang Y, Cao M, Li T, Ding W, Liu Y, Deng R, Dong Z, Chen H, Novakovic VA, Bi Y, Kou J, Tian Y, Zhou J, Shi J. Phosphatidylserine on blood cells and endothelial cells contributes to the hypercoagulable state in cirrhosis. Liver Int. 2016;36:1800-1810.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
67.  Chapin JC, Hajjar KA. Fibrinolysis and the control of blood coagulation. Blood Rev. 2015;29:17-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 411]  [Cited by in F6Publishing: 451]  [Article Influence: 50.1]  [Reference Citation Analysis (0)]
68.  Rossetto V, Spiezia L, Senzolo M, Rodriguez-Castro KI, Gavasso S, Woodhams B, Simioni P. Does decreased fibrinolysis have a role to play in the development of non-neoplastic portal vein thrombosis in patients with hepatic cirrhosis? Intern Emerg Med. 2014;9:397-403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 9]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
69.  Lisman T, Caldwell SH, Intagliata NM. Haemostatic alterations and management of haemostasis in patients with cirrhosis. J Hepatol. 2022;76:1291-1305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 24]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
70.  Hugenholtz GC, Northup PG, Porte RJ, Lisman T. Is there a rationale for treatment of chronic liver disease with antithrombotic therapy? Blood Rev. 2015;29:127-136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
71.  Tripodi A, Salerno F, Chantarangkul V, Clerici M, Cazzaniga M, Primignani M, Mannuccio Mannucci P. Evidence of normal thrombin generation in cirrhosis despite abnormal conventional coagulation tests. Hepatology. 2005;41:553-558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 488]  [Cited by in F6Publishing: 463]  [Article Influence: 24.4]  [Reference Citation Analysis (0)]
72.  Lebreton A, Sinegre T, Lecompte T, Talon L, Abergel A, Lisman T. Thrombin Generation and Cirrhosis: State of the Art and Perspectives. Semin Thromb Hemost. 2020;46:693-703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
73.  O'Donnell J, Mumford AD, Manning RA, Laffan MA. Marked elevation of thrombin generation in patients with elevated FVIII:C and venous thromboembolism. Br J Haematol. 2001;115:687-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 40]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
74.  Dahlbäck B. Progress in the understanding of the protein C anticoagulant pathway. Int J Hematol. 2004;79:109-116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 73]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
75.  Hollestelle MJ, Geertzen HG, Straatsburg IH, van Gulik TM, van Mourik JA. Factor VIII expression in liver disease. Thromb Haemost. 2004;91:267-275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 139]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
76.  Lisman T, Kleiss S, Patel VC, Fisher C, Adelmeijer J, Bos S, Singanayagam A, Stoy SH, Shawcross DL, Bernal W. In vitro efficacy of pro- and anticoagulant strategies in compensated and acutely ill patients with cirrhosis. Liver Int. 2018;38:1988-1996.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
77.  Hugenholtz GC, Macrae F, Adelmeijer J, Dulfer S, Porte RJ, Lisman T, Ariëns RA. Procoagulant changes in fibrin clot structure in patients with cirrhosis are associated with oxidative modifications of fibrinogen. J Thromb Haemost. 2016;14:1054-1066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 80]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
78.  Gilmore IT, Burroughs A, Murray-Lyon IM, Williams R, Jenkins D, Hopkins A. Indications, methods, and outcomes of percutaneous liver biopsy in England and Wales: an audit by the British Society of Gastroenterology and the Royal College of Physicians of London. Gut. 1995;36:437-441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 269]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
79.  Terjung B, Lemnitzer I, Dumoulin FL, Effenberger W, Brackmann HH, Sauerbruch T, Spengler U. Bleeding complications after percutaneous liver biopsy. An analysis of risk factors. Digestion. 2003;67:138-145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 152]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
80.  Bruzzi JF, O'Connell MJ, Thakore H, O'Keane C, Crowe J, Murray JG. Transjugular liver biopsy: assessment of safety and efficacy of the Quick-Core biopsy needle. Abdom Imaging. 2002;27:711-715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 38]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
81.  Segal JB, Dzik WH; Transfusion Medicine/Hemostasis Clinical Trials Network. Paucity of studies to support that abnormal coagulation test results predict bleeding in the setting of invasive procedures: an evidence-based review. Transfusion. 2005;45:1413-1425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 485]  [Cited by in F6Publishing: 478]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
82.  Denzer U, Helmreich-Becker I, Galle PR, Lohse AW. Liver assessment and biopsy in patients with marked coagulopathy: value of mini-laparoscopy and control of bleeding. Am J Gastroenterol. 2003;98:893-900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 41]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
83.  Ewe K. Bleeding after liver biopsy does not correlate with indices of peripheral coagulation. Dig Dis Sci. 1981;26:388-393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 230]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
84.  Grabau CM, Crago SF, Hoff LK, Simon JA, Melton CA, Ott BJ, Kamath PS. Performance standards for therapeutic abdominal paracentesis. Hepatology. 2004;40:484-488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 171]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
85.  Jeon JW, Shin HP, Lee JI, Joo KR, Pack KM, Cha JM, Park JJ, Lim JU, Lim K. The risk of postpolypectomy bleeding during colonoscopy in patients with early liver cirrhosis. Surg Endosc. 2012;26:3258-3263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
86.  Baltz JG, Argo CK, Al-Osaimi AM, Northup PG. Mortality after percutaneous endoscopic gastrostomy in patients with cirrhosis: a case series. Gastrointest Endosc. 2010;72:1072-1075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 39]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
87.  Townsend JC, Heard R, Powers ER, Reuben A. Usefulness of international normalized ratio to predict bleeding complications in patients with end-stage liver disease who undergo cardiac catheterization. Am J Cardiol. 2012;110:1062-1065.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
88.  European Association for the Study of the Liver. EASL Clinical Practice Guidelines on prevention and management of bleeding and thrombosis in patients with cirrhosis. J Hepatol. 2022;76:1151-1184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 128]  [Article Influence: 64.0]  [Reference Citation Analysis (0)]
89.  Northup PG, Garcia-Pagan JC, Garcia-Tsao G, Intagliata NM, Superina RA, Roberts LN, Lisman T, Valla DC. Vascular Liver Disorders, Portal Vein Thrombosis, and Procedural Bleeding in Patients With Liver Disease: 2020 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology. 2021;73:366-413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 283]  [Article Influence: 94.3]  [Reference Citation Analysis (1)]
90.  Tonon M, Rosi S, Gambino CG, Piano S, Calvino V, Romano A, Martini A, Pontisso P, Angeli P. Natural history of acute kidney disease in patients with cirrhosis. J Hepatol. 2021;74:578-583.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 29]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
91.  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;38:1437-1441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 49]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
92.  Blasi A, Patel VC, Adelmeijer J, Azarian S, Hernandez Tejero M, Calvo A, Fernández J, Bernal W, Lisman T. Mixed Fibrinolytic Phenotypes in Decompensated Cirrhosis and Acute-on-Chronic Liver Failure with Hypofibrinolysis in Those With Complications and Poor Survival. Hepatology. 2020;71:1381-1390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 64]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
93.  Intagliata NM, Davis JPE, Lafond J, Erdbruegger U, Greenberg CS, Northup PG, Caldwell SH. Acute kidney injury is associated with low factor XIII in decompensated cirrhosis. Dig Liver Dis. 2019;51:1409-1415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
94.  Premkumar M, Saxena P, Rangegowda D, Baweja S, Mirza R, Jain P, Bhatia P, Kumar G, Bihari C, Kalal C, Vyas T, Choudhury A, Sarin SK. Coagulation failure is associated with bleeding events and clinical outcome during systemic inflammatory response and sepsis in acute-on-chronic liver failure: An observational cohort study. Liver Int. 2019;39:694-704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 49]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
95.  Arroyo V, Moreau R, Jalan R. Acute-on-Chronic Liver Failure. N Engl J Med. 2020;382:2137-2145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 349]  [Article Influence: 87.3]  [Reference Citation Analysis (2)]
96.  Fisher C, Patel VC, Stoy SH, Singanayagam A, Adelmeijer J, Wendon J, Shawcross DL, Lisman T, Bernal W. Balanced haemostasis with both hypo- and hyper-coagulable features in critically ill patients with acute-on-chronic-liver failure. J Crit Care. 2018;43:54-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 75]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
97.  Tripodi A. Hemostasis abnormalities in cirrhosis. Curr Opin Hematol. 2015;22:406-412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 49]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
98.  Wu H, Nguyen GC. Liver cirrhosis is associated with venous thromboembolism among hospitalized patients in a nationwide US study. Clin Gastroenterol Hepatol. 2010;8:800-805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 102]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
99.  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;104:96-101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 301]  [Cited by in F6Publishing: 288]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
100.  Allison MG, Shanholtz CB, Sachdeva A. Hematological Issues in Liver Disease. Crit Care Clin. 2016;32:385-396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 10]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
101.  Qi X, Han G, Fan D. Management of portal vein thrombosis in liver cirrhosis. Nat Rev Gastroenterol Hepatol. 2014;11:435-446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 105]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
102.  Nery F, Chevret S, Condat B, de Raucourt E, Boudaoud L, Rautou PE, Plessier A, Roulot D, Chaffaut C, Bourcier V, Trinchet JC, Valla DC; 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;61:660-667.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 292]  [Cited by in F6Publishing: 297]  [Article Influence: 33.0]  [Reference Citation Analysis (0)]
103.  Pan J, Wang L, Gao F, An Y, Yin Y, Guo X, Nery FG, Yoshida EM, Qi X. Epidemiology of portal vein thrombosis in liver cirrhosis: A systematic review and meta-analysis. Eur J Intern Med. 2022;104:21-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 15]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
104.  Walsh KA, Lewis DA, Clifford TM, Hundley JC, Gokun Y, Angulo P, Davis GA. Risk factors for venous thromboembolism in patients with chronic liver disease. Ann Pharmacother. 2013;47:333-339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
105.  Intagliata NM, Caldwell SH, Tripodi A. Diagnosis, Development, and Treatment of Portal Vein Thrombosis in Patients With and Without Cirrhosis. Gastroenterology. 2019;156:1582-1599.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 199]  [Article Influence: 39.8]  [Reference Citation Analysis (0)]
106.  DeLeve LD, Valla DC, Garcia-Tsao G; American Association for the Study Liver Diseases. Vascular disorders of the liver. Hepatology. 2009;49:1729-1764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 739]  [Cited by in F6Publishing: 614]  [Article Influence: 40.9]  [Reference Citation Analysis (0)]
107.  Delgado MG, Seijo S, Yepes I, Achécar L, Catalina MV, García-Criado A, Abraldes JG, de la Peña J, Bañares R, Albillos A, Bosch J, García-Pagán JC. Efficacy and safety of anticoagulation on patients with cirrhosis and portal vein thrombosis. Clin Gastroenterol Hepatol. 2012;10:776-783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 269]  [Cited by in F6Publishing: 260]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
108.  Guerrero A, Campo LD, Piscaglia F, Scheiner B, Han G, Violi F, Ferreira CN, Téllez L, Reiberger T, Basili S, Zamora J, Albillos A; Baveno Cooperation: an EASL consortium. Anticoagulation improves survival in patients with cirrhosis and portal vein thrombosis: The IMPORTAL competing-risk meta-analysis. J Hepatol. 2023;79:69-78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 18]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
109.  Pasta A, Calabrese F, Labanca S, Marenco S, Pieri G, Plaz Torres MC, Intagliata NM, Caldwell SH, Giannini EG. Safety and efficacy of venous thromboembolism prophylaxis in patients with cirrhosis: A systematic review and meta-analysis. Liver Int. 2023;43:1399-1406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
110.  Shah NL, Northup PG, Caldwell SH. A clinical survey of bleeding, thrombosis, and blood product use in decompensated cirrhosis patients. Ann Hepatol. 2012;11:686-690.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Sharara AI, Rockey DC. Gastroesophageal variceal hemorrhage. N Engl J Med. 2001;345:669-681.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 298]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
112.  Lieber SR, Jiang Y, Moon A, Barritt AS. Antiplatelet Medications Are Associated With Bleeding and Decompensation Events Among Patients With Cirrhosis. J Clin Gastroenterol. 2022;56:627-634.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
113.  Spyropoulos AC, Brohi K, Caprini J, Samama CM, Siegal D, Tafur A, Verhamme P, Douketis JD; SSC Subcommittee on Perioperative and Critical Care Thrombosis and Haemostasis of the International Society on Thrombosis and Haemostasis. Scientific and Standardization Committee Communication: Guidance document on the periprocedural management of patients on chronic oral anticoagulant therapy: Recommendations for standardized reporting of procedural/surgical bleed risk and patient-specific thromboembolic risk. J Thromb Haemost. 2019;17:1966-1972.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 69]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
114.  Zanetto A, Northup P, Roberts L, Senzolo M. Haemostasis in cirrhosis: Understanding destabilising factors during acute decompensation. J Hepatol. 2023;78:1037-1047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 22]  [Reference Citation Analysis (0)]
115.  Karvellas CJ, Bajaj JS, Kamath PS, Napolitano L, O'Leary JG, Solà E, Subramanian R, Wong F, Asrani SK. AASLD Practice Guidance on Acute-on-chronic liver failure and the management of critically ill patients with cirrhosis. Hepatology. 2024;79:1463-1502.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 9]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
116.  O'Leary JG, Greenberg CS, Patton HM, Caldwell SH. AGA Clinical Practice Update: Coagulation in Cirrhosis. Gastroenterology. 2019;157:34-43.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 245]  [Article Influence: 49.0]  [Reference Citation Analysis (0)]
117.  Villanueva C, Colomo A, Bosch A, Concepción M, Hernandez-Gea V, Aracil C, Graupera I, Poca M, Alvarez-Urturi C, Gordillo J, Guarner-Argente C, Santaló M, Muñiz E, Guarner C. Transfusion strategies for acute upper gastrointestinal bleeding. N Engl J Med. 2013;368:11-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1186]  [Cited by in F6Publishing: 1007]  [Article Influence: 91.5]  [Reference Citation Analysis (0)]
118.  Pfisterer N, Schwarz M, Jachs M, Putre F, Ritt L, Mandorfer M, Madl C, Trauner M, Reiberger T. Endoscopic band ligation is safe despite low platelet count and high INR. Hepatol Int. 2023;17:1205-1214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
119.  Moreau R, Jalan R, Gines P, Pavesi M, Angeli P, Cordoba J, Durand F, Gustot T, Saliba F, Domenicali M, Gerbes A, Wendon J, Alessandria C, Laleman W, Zeuzem S, Trebicka J, Bernardi M, Arroyo V; CANONIC Study Investigators of the EASL–CLIF Consortium. Acute-on-chronic liver failure is a distinct syndrome that develops in patients with acute decompensation of cirrhosis. Gastroenterology. 2013;144:1426-1437, 1437.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1720]  [Cited by in F6Publishing: 1968]  [Article Influence: 178.9]  [Reference Citation Analysis (5)]
120.  Davis JPE, Northup PG, Caldwell SH, Intagliata NM. Viscoelastic Testing in Liver Disease. Ann Hepatol. 2018;17:205-213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 32]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
121.  O'Shea RS, Davitkov P, Ko CW, Rajasekhar A, Su GL, Sultan S, Allen AM, Falck-Ytter Y. AGA Clinical Practice Guideline on the Management of Coagulation Disorders in Patients With Cirrhosis. Gastroenterology. 2021;161:1615-1627.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 43]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
122.  Kaplan DE, Ripoll C, Thiele M, Fortune BE, Simonetto DA, Garcia-Tsao G, Bosch J. AASLD Practice Guidance on risk stratification and management of portal hypertension and varices in cirrhosis. Hepatology. 2024;79:1180-1211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 43]  [Article Influence: 43.0]  [Reference Citation Analysis (0)]
123.  Rautou PE, Caldwell SH, Villa E. Bleeding and Thrombotic Complications in Patients With Cirrhosis: A State-of-the-Art Appraisal. Clin Gastroenterol Hepatol. 2023;21:2110-2123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 11]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
124.  Vincent JL, Rossaint R, Riou B, Ozier Y, Zideman D, Spahn DR. Recommendations on the use of recombinant activated factor VII as an adjunctive treatment for massive bleeding--a European perspective. Crit Care. 2006;10:R120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 191]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
125.  Kumar M, Venishetty S, Jindal A, Bihari C, Maiwall R, Vijayaraghavan R, Saggere Muralikrishna S, Arora V, Kumar G, Sarin SK. Tranexamic acid in upper gastrointestinal bleed in patients with cirrhosis: A randomized controlled trial. Hepatology. 2024;80:376-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]