Minireviews Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. May 21, 2023; 29(19): 2916-2931
Published online May 21, 2023. doi: 10.3748/wjg.v29.i19.2916
Assessment of delayed bleeding after endoscopic submucosal dissection of early-stage gastrointestinal tumors in patients receiving direct oral anticoagulants
Mitsushige Sugimoto, Takashi Kawai, Department of Gastroenterological Endoscopy, Tokyo Medical University Hospital, Tokyo 160-0023, Japan
Masaki Murata, Department of Gastroenterology, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan
ORCID number: Mitsushige Sugimoto (0000-0002-9194-7392); Masaki Murata (0000-0002-4951-0584); Takashi Kawai (0000-0002-5320-8134).
Author contributions: Sugimoto M wrote the paper; Sugimoto M, Murata M and Kawai T collected the data.
Supported by the Grant-in-Aid for Scientific Research in Japan, No. 21K07949.
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: Mitsushige Sugimoto, AGAF, MD, PhD, Professor, Department of Gastroenterological Endoscopy, Tokyo Medical University Hospital, 6-7-1, Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan. sugimo@tokyo-med.ac.jp
Received: January 24, 2023
Peer-review started: January 24, 2023
First decision: March 21, 2023
Revised: April 3, 2023
Accepted: April 24, 2023
Article in press: April 24, 2023
Published online: May 21, 2023

Abstract

Delayed bleeding is a major and serious adverse event of endoscopic submucosal dissection (ESD) for early-stage gastrointestinal tumors. The rate of post-ESD bleeding for gastric cancer is higher (around 5%-8%) than that for esophagus, duodenum and colon cancer (around 2%-4%). Although investigations into the risk factors for post-ESD bleeding have identified several procedure-, lesion-, physician- and patient-related factors, use of antithrombotic drugs, especially anticoagulants [direct oral anticoagulants (DOACs) and warfarin], is thought to be the biggest risk factor for post-ESD bleeding. In fact, the post-ESD bleeding rate in patients receiving DOACs is 8.7%-20.8%, which is higher than that in patients not receiving anticoagulants. However, because clinical guidelines for management of ESD in patients receiving DOACs differ among countries, it is necessary for endoscopists to identify ways to prevent post-ESD delayed bleeding in clinical practice. Given that the pharmacokinetics (e.g., plasma DOAC level at both trough and Tmax) and pharmacodynamics (e.g., anti-factor Xa activity) of DOACs are related to risk of major bleeding, plasma DOAC level and anti-FXa activity may be useful parameters for monitoring the anti-coagulate effect and identifying DOAC patients at higher risk of post-ESD bleeding.

Key Words: Direct oral anticoagulants, Gastrointestinal tumors, Endoscopic submucosal dissection, Delayed bleeding, Adverse events, Anticoagulants

Core Tip: Recent international clinical guidelines for early-stage gastrointestinal tumors recommend endoscopic submucosal dissection (ESD) as the first-line treatment. Direct oral anticoagulants (DOACs) are a major risk factor for post-ESD bleeding and the pharmacokinetics and pharmacodynamics of DOACs may be related to risk of post-ESD bleeding. Therefore, one way to monitor the anticoagulant effect of DOACs in clinical practice may be to develop a system that effectively measures anti–FXa activity and plasma concentration. In the future, it may be useful to stratify risk of post-ESD delayed bleeding based on a scoring system that includes pharmacological parameters of DOACs.



INTRODUCTION

Endoscopic resection, a minimally invasive endoscopic non-surgical treatment, is now accepted as first-line management for most cases of early-stage esophageal cancer, gastric cancer and colorectal cancer or adenoma around the world[1]. Endoscopic resection mainly includes endoscopic mucosal resection (EMR) and endoscopic submucosal dissection (ESD). Indication for endoscopic resection for EMR and ESD is typically a local mucosal lesion with an extremely low risk of metastasis to lymph nodes (generally less than 1%), and lesions that can be resected en-bloc, irrespective of localization in the esophagus, stomach, duodenum or colon. Because ESD enables complete en-bloc resection and is associated with a lower recurrence rate than EMR[2,3], recent international clinical guidelines for early-stage gastrointestinal tumors recommend ESD as the first-line treatment over EMR and surgical resection[1,3-7]. To achieve good results and prognosis in ESD for early-stage gastrointestinal tumors, endoscopists and gastroenterologists require excellent skills and knowledge of the diagnosis, indications, actual procedures, and evaluation of curability, complications, long-term postoperative surveillance, and histopathology[3].

ESD often causes adverse events such as intra-operative and delayed bleeding from an artificial ulcer and perforation. The occurrence of such events has been linked to several procedure-, lesion-, physician- and patient-related factors[8-12]. Of the possible risk factors for post-ESD delayed bleeding, use of antithrombotic drugs, especially anticoagulants, is the biggest risk factor[13,14]. Although anticoagulants are mainly divided into warfarin and direct oral anticoagulants (DOACs), no parameters identified to date can be used to accurately monitor the anti-coagulate effect of DOACs in clinical practice or endoscopic/surgical procedures. Thus, it is important to clarify the association between post-ESD delayed bleeding for early-stage gastrointestinal tumors and DOACs, identify risk factors of post-ESD delayed bleeding in patients taking DOACs and methods to prevent bleeding in these patients.

Here, we review delayed bleeding after ESD for gastrointestinal tumors, risk factors for post-ESD bleeding, the pharmacological characteristics of DOACs, international clinical guidelines for endoscopic procedures in patients receiving DOACs, and post-ESD bleeding in patients receiving DOACs.

DELAYED BLEEDING AFTER ESD FOR GASTROINTESTINAL TUMORS

In Japan, upper and lower gastrointestinal tumors are detected in the early stages in many patients, mainly through the use of optimal screening methods, appropriate surveillance and the development of endoscopic diagnostic techniques for early detection and endoscopic equipment[15-17]. In general, evaluation of early-stage gastrointestinal tumors should be performed by expert endoscopists, using a high-definition endoscope by white-light imaging and advanced image-enhanced endoscopy[18]. The experience of the endoscopist may be related to the incidence of adverse events and effective prevention of procedure-related adverse events after ESD. Careful and appropriate coagulation for exposed blood vessels may reduce the gastrointestinal bleeding risk, especially in the stomach[7,19]. The rate of bleeding and its risk factors are known to differ among patients with esophagus, stomach, duodenum and colon cancer.

Esophageal squamous cell carcinoma

The recent development of high-vision endoscopes and techniques for endoscopic diagnosis, including narrow band imaging, has led to more frequent early detection of esophageal esophageal squamous cell carcinoma (SCC)[15,20]. Therefore, ESD is accepted as an effective procedure for detecting superficial esophageal SCC. In Japan, the Esophageal Cancer Practice Guidelines 2017 weakly recommends endoscopic resection as first-line treatment for preoperatively diagnosed cT1a-MM/T1b-SM1 non-circumferential SCC[6]. In 2014, a meta-analysis of 15 studies with 776 patients with ESD-treated SCC reported pooled estimates of complete resection and en bloc resection rates of 89.4% [95%CI: 86.2%–91.9%] and 95.1% (92.6%–96.8%), respectively[21]. In addition, pooled estimates of adverse events such as post-ESD bleeding, perforation, and stenosis were 2.1% (95%CI: 1.2%–3.8%), 5.0% (3.5%–7.2%), and 11.6% (8.2%–16.2%), respectively[21].

Barrett's neoplasia and esophageal adenocarcinoma

The incidence of esophageal adenocarcinoma (EAC) located in Barrett’s epithelium has been increasing, especially in Western countries, due to decreases in the Helicobacter pylori (H. pylori) infection rate and increases in reflux esophagitis[22]. The European Society of Gastrointestinal Endoscopy (ESGE) recommends using EMR for ≤ 20 mm visible lesions with low probability of submucosal invasion and for larger or multifocal benign dysplastic lesions. In Japan, ESD is strongly recommended over EMR for the radical treatment of superficial EAC with a low risk of metastasis[6]. A meta-analysis of 11 studies investigating the efficacy and safety of ESD for EAC (mean size: 27 mm) reported pooled estimates for en bloc resection and pooled R0 resection of 92.9% (95%CI: 90.3%-95.2%) and 74.5% (66.3%-81.9%), respectively[23]. Incidence of recurrence after curative resection was 0.17% (95%CI: 0%-0.3%) at a mean follow-up of 22.9 mo (17.5-28.3 mo)[23]. In adverse events, estimates for bleeding, perforation, and stricture were 1.7% (95%CI: 0.6%-3.4%), 1.5% (0.4%-3.0%) and 11.6% (0.9%-29.6%), respectively. Thus, the rate of post-ESD delayed bleeding in both esophageal SCC and EAC may not be very high (1.7%-2.1%).

Gastric cancer

In the ESGE, gastric cancers that are ≤ 30 mm, submucosal (sm1), and well-differentiated, or ≤ 20 mm, intramucosal, and poorly differentiated, and without ulcerative findings for both sets of criteria can be considered for ESD, although the decision should be individualized[4]. ESD for gastric cancer is associated with high rates of en bloc and R0 resection (> 90%), curative resection (75%–80%), low local recurrence (< 5%) and acceptable rates of adverse events (post-ESD bleeding 5%–10% and perforation < 3%)[24,25]. A recent meta-analysis of 22 studies in Western countries reported estimates for en bloc resection and R0 resection of 96% (95%CI: 93%-98%) and 84% (79%-89%), respectively[26]. Overall, adverse events occur in 9.5% of patients, including delayed bleeding (5.8%), perforation (3.4%), and stenosis (0.35%)[26]. The odds ratio (OR) indicates that there is no significant difference in risk of post-ESD bleeding between ESD and EMR (OR 1.26, 95%CI: 0.88-1.80)[27]. Another meta-analysis of 74 articles by Libânio et al[28] reported post-ESD bleeding rates ranging from 0.6% to 26.9% and a pooled bleeding rate of 5.1% (95%CI: 4.5%-5.7%), with significant heterogeneity across studies (I2: 84.46, P < 0.001). However, bleeding rates were not significantly different among different study designs (5.9% in randomized clinical trials, 6.1% in prospective studies, and 4.9% in retrospective studies). The elderly Japanese population aged ≥ 85 years has increased from 1.4 to 4.8 million over the last two decades[29] and our investigation of a cohort of 10,320 patients showed that the incidence of bleeding in elderly patients aged > 80 years was 5.7% (95%CI: 4.6%-6.9%), which was significantly higher than in patients aged < 80 years (4.5%, 4.1%-5.0%)[30].

Duodenum

Because ESD for duodenal tumors is associated with high rates of post-ESD bleeding and perforation at both the early and late phases, the ESGE suggests reserving its use for selected cases and tumors in expert centers[4]. In particular, perforation rates are high, with an incidence > 10% in studies involving expert centers[31]. Further, distal location to the ampulla of Vater is a risk factor for delayed perforation[32]. A meta-analysis reported pooled rates of en bloc resection, need for surgical intervention, delayed bleeding, intraoperative and delayed perforation of 87%, 4%, 2%, 15% and 2%, respectively[33]. Meanwhile, a recent large retrospective Japanese study reported that the rate of post-ESD adverse events was significantly reduced in cases with complete closure of the mucosal defect compared to partial closure and no closure (1.7%, 25.0% and 15.6%, respectively, P < 0.01)[34].

Colon

In Japan, indications for ESD for colorectal tumors are lesions for which endoscopic en bloc resection is required, as follows: (1) Lesions for which en bloc resection with snare EMR is difficult to apply; (2) Mucosal tumors with submucosal fibrosis; (3) Sporadic localized tumors in conditions of chronic inflammation such as ulcerative colitis; and (4) Post-EMR local residual or recurrent early-stage cancers[5]. A recent systematic review of 109 studies on 19484 colorectal lesions resected by ESD reported rates of en bloc resection of 91%, R0 resection of 82.9%, and local recurrence of 2%. The study also reported a rate of post-ESD bleeding of 2.7% and perforation of 5.2%, and that 1.1% of all patients needed surgical treatment by severe adverse events[35].

Because the rate of post-ESD bleeding may be higher for gastric cancer (5%-8%) than that at other sites (around 2%-4%) due to direct exposure of artificial ulcers to gastric acid and bile, it is necessary for endoscopists to be aware of and develop countermeasures for gastric ESD.

RISK FACTORS FOR POST-ESD GASTROINTESTINAL DELAYED BLEEDING

Risk factors for post-ESD bleeding are expected to differ among patients with esophagus, stomach, duodenum and colon cancer. Post-ESD bleeding has been shown to be associated with procedure-related factors (e.g., type of knife, coagulation machine and endoscope, and coagulation mode), lesion-related factors (e.g., gastrointestinal organ, large lesion size, location, presence within the ulcerated lesion, scarring, and fibrosis), physician-related factors (e.g., experience with ESD) and patient-related factors [hemodialysis, drugs (antiplatelet drugs, anticoagulants, steroids, and non-steroidal anti-inflammatory drugs), hemostasis ability, and platelet count] (Figure 1)[8-12].

Figure 1
Figure 1 Post-endoscopic submucosal dissection bleeding-related factors. DOACs: Direct oral anticoagulants; NSAIDs: Non-steroidal anti-inflammatory drugs; CHADS: Congestive heart failure, Hypertension, Age > 75 years, Diabetes mellitus, and Stroke.

Libânio et al[28] showed in a meta-analysis of 74 articles that male sex (OR 1.25), cardiopathy (OR 1.54), antithrombotic drugs (OR 1.63), cirrhosis (OR 1.76), chronic kidney disease (CKD) (OR 3.38), tumor size > 20 mm (OR 2.70), resected specimen size > 30 mm (OR 2.85), localization in the lesser curvature (OR 1.74), flat/depressed morphology (OR 1.43), carcinoma histology (OR 1.46), and ulceration (OR 1.64) were significant risk factors for post-ESD bleeding of gastric cancer, whereas age, hypertension, submucosal invasion, fibrosis, and location (upper, middle, or lower third of stomach) were not. In terms of procedural factors, procedure duration > 60 min (OR, 2.05) and use of histamine-2 receptor antagonists instead of proton pump inhibitors (PPIs) (OR, 2.13) were associated with post-ESD bleeding, whereas endoscopist experience was not. Recently, Hatta et al[36] conducted a nationwide multicenter retrospective study focusing on post-ESD bleeding for gastric cancer and used the data to develop a model that applies 10 factors [warfarin, DOAC, hemodialysis, P2Y12 receptor antagonist, aspirin, tumor size > 30 mm, tumor location in the lower third, presence of multiple tumors and interruption of each kind of antithrombotic agent] to predict post-ESD bleeding (BEST-J score). According to the BEST-J score, rates of bleeding in patients categorized as low-risk, intermediate-risk, high-risk, and very high-risk were 2.8%, 6.1%, 11.4%, and 29.7%, respectively[36]. A validation study of the BEST-J score showed that the area under the curve for the BEST-J score at multicenter trials was 0.713 (95%CI: 0.625–0.802), which suggests that the BEST-J score may be useful for predicting post-ESD bleeding in not only expert centers but also general hospitals[37]. In addition, because the healing speed of post-ESD artificial ulcers is related to the post-ESD bleeding rate, factors that affect the healing speed of ulcers, namely H. pylori infection status, type of acid inhibitory drug (e.g., PPIs and vonoprazan) and severity of gastric atrophy, may also be risk factors for post-ESD bleeding in gastric cancer[11,12,38]. In today’s aging society, the number of patients taking anti-platelet drugs and anticoagulants for the prevention of cardio- and cerebrovascular diseases has risen. That a multicenter study reported a high incidence of post-ESD bleeding in Japanese aged > 80 years, especially in patients receiving hemodialysis and taking warfarin[30], indicates that careful management of ESD is required to prevent bleeding in patients aged > 80 years compared to younger patients.

Major risk factors for post-ESD bleeding for colorectal tumors are generally larger tumor size, location in the rectum or cecum, long procedure time, number of tumors, and taking anti-thrombotic drugs. Recently, Li et al[39] reported that post-ESD bleeding for colorectal tumors is observed in 4.7% of patients, and that hypertension (OR 2.829, 95%CI: 1.101-7.265) and using hot biopsy forceps for wound management (OR 2.873, 95%CI: 1.013-8.147) remain significant risk factors for bleeding after multivariate analysis. In another study, Seo et al[40] developed a risk-scoring model to predict bleeding after colorectal ESD following identification of the tumor location in the rectosigmoid colon (OR 6.49; 95%CI: 1.96-21.42), large tumor (> 30 mm) (2.10, 1.01-4.40), and use of antiplatelet agents except for aspirin alone (4.04, 1.44-11.30) as risk factors for bleeding[40]. When use of antiplatelet agents except for aspirin alone was scored as 1 point, tumor size > 30 mm as 1 point, and location in the rectosigmoid area as 2 points, the incidence of bleeding in low-risk (score 0-2) and high-risk groups (score 3-4) was 1.5% and 6.0%, respectively.

Thus, current evidence indicates that the use of antithrombotic drugs, especially anticoagulants (DOACs and warfarin) is the biggest risk factor for post-ESD bleeding[13,14].

PHARMACOLOGICAL CHARACTERISTICS OF DOACS

DOACs are currently the first-line drug for the pharmacological prevention of systemic embolism or stroke in atrial fibrillation patients. They are categorized into two main classes: Direct thrombin inhibitors (i.e., dabigatran) and activated coagulation factor X (FXa) inhibitors (i.e., apixaban, edoxaban, rivaroxaban, and betrixaban). Compared with anticoagulation with vitamin K antagonists (i.e., warfarin) or low-molecular-weight heparins, DOACs are new agents that demonstrate superiority or noninferiority to prior standards of care in reducing the risk of thromboembolic complications and major and minor bleeding risk, have fewer monitoring requirements and less frequent follow-up need; and have more immediate drug onset and offset effects and fewer drug and food interactions[41-44]. However, an advantage of using vitamin K antagonist therapy is that a therapeutic international normalized ratio range of 2.0-3.0 has been established and is recommended to prevent embolic complications in non-valvular atrial fibrillation in the treatment of deep vein thrombosis and pulmonary embolism. In contrast, a disadvantage of DOACs is that their anticoagulant effects in patients are unclear because there are no Food and Drug Administration (FDA)-approved methods to measure correctly the anticoagulant effect of DOACs. Although qualitative coagulation assays (e.g., thrombin time, activated partial thromboplastin time, and prothrombin time) can be selected as first-line tests, they do not accurately measure the anticoagulant effect of DOACs (Table 1). Quantitative measures for direct assessment of anticoagulant effects do exist, including anti–FXa activity, plasma drug concentration (standard method in preclinical/clinical research and the most accurate method), dilute thrombin time, and ecarin thrombin time[45,46]. In fact, plotting anti–FXa activity against plasma levels of apixaban and rivaroxaban has been confirmed to show a direct linear relationship for both compounds[47].

Table 1 Possible methods for monitoring the anticoagulant ability of direct oral anticoagulants[49,71,87].

Qualitative methods
Quantitative methods
Other
aPTT
TT
PT
Anti-FIIa levels
Anti-Fxa levels
Plasma level
dTT
ECT/ECA
CBC
CMP
Dabigatran222112222
Apixaban21122
Edoxaban21122
Rivaroxaban21122

Pharmacokinetic characteristics such as oral bioavailability, plasma protein binding and relative involvement of renal and non-renal elimination differ substantially among DOACs (Table 2). Pharmacokinetics is influenced by the type of DOAC, dose, renal function, liver function, age, sex, body weight, drug metabolic enzyme gene polymorphisms and drug-drug interactions, but not ethnicity, geographic region, aspirin use, or clopidogrel use[48]. DOACs are relatively safe and effective in patients with moderate CKD [creatinine clearance (Ccr) 30–50 mL/min]. Rivaroxaban, dabigatran and edoxaban will require dose adjustment for renal impairment and patients with severe renal dysfunction (Ccr < 30 mL/min) are recommended to avoid their use[49]. Regulatory agencies such as the FDA and European Medicines Agency have provided guidelines for performing dose adjustments according to the DOAC dose based on Ccr and anticoagulant indications[50]. The International Society on Thrombosis and Hemostasis suggests that use of DOACs are safe in patients of body mass index ≤ 40 kg/m2 (body weight ≤ 120 kg) at standard doses but does not recommend them for patients of body weight > 120 kg[51]. Compared to warfarin, DOACs are more effective and safer in patients with low body weight (< 50 kg)[52].

Table 2 Pharmacological characteristics of direct oral anticoagulant[71,87].

Dabigatran
Apixaban
Edoxaban
Rivaroxaban
Target factorThrombin (Factor IIa)Factor XaFactor XaFactor Xa
Half-time (h)10.7-11.86.12-8.116.21-6.705.7-12.6
Time to peak effect (h)43.0-3.51-1.51.4-3.3
Distribution volume (L)50-702110750
Renal excretion (%)852735.4-5050
Fecal excretion (%)62562.250
Hepatic metabolismNoCYP3A4/5CYP3A4CYP3A4 and CYP2J2
TransporterP-gPP-gP/BCRPP-gPP-gP/BCRP
Protein binding (%)28.2-31.58740.0-58.992-95
DialyzableYesNoNoNo
ProdrugYesNoNoNo
Bioavailability (%)6.55061.866-112
Dose for AF (in Japan)150 mg5 mg60 mg15 mg
Dosing timeTwice dailyTwice dailyOnce dailyOnce daily
Reversal agentIdarucizumabAndexanet alfaAndexanet alfaAndexanet alfa
FDA-approved indicationsNonvalvular AF, VTE (T, SP, P) Nonvalvular AF, VTE (T, SP, P)Nonvalvular AF, VTE (T)Nonvalvular AF, VTE (T, SP, P)
Japanese insurance system-approved indicationsNonvalvular AF (P)Nonvalvular AF (P), VTE (T, SP)Nonvalvular AF (P), VTE (T, SP)Nonvalvular AF (P), VTE (T, SP)
Non-pharmacologic interactionsAge, reduced GFRAge, reduced body weight, reduced GFR, probable severe liver damageReduced GFR, probable severe liver damageAge, reduced GFR, probable severe liver damage
Drug interactionsDose reduction: Concomitant P-gp inhibitor, gastric acid inhibitory drugAvoid: Concomitant P-gp and CYP3A4 inhibitorsAvoid: Concomitant rifampinAvoid: Rivaroxaban with concomitant dual P-gp and CYP3A4 inhibitors
ContraindicationsCcr: < 30mL/minNonvalvular AF: Ccr: < 15mL/min, VTE:Nonvalvular AF: Ccr: < 15mL/min, VTE:Nonvalvular AF: Ccr: < 15mL/min, VTE:
Ccr: < 30mL/minCcr: < 30mL/minCcr: < 30mL/min

DOACs are metabolized by either cytochrome P450 (CYP) metabolic enzymes in the liver or permeability glycoprotein transporters; thus, agents that induce or inhibit CYP metabolic enzymes or glycoprotein transporters can lead to major drug-drug interactions and place the patient at undue risk for adverse events. In fact, concomitant use of apixaban, rivaroxaban, or dabigatran with clarithromycin, a potent inhibitor of CYP3A4, and ATP-binding cassette multidrug transporters increases serum levels of DOACs by 20% to 100% and prolongs coagulation time[53,54]. The pharmacokinetics of DOACs also depend on genetic variations, such as ABCB1 (ATP-binding cassette multidrug transporters, MDR1) (1236C>T, 2677G>T/A, and 3435C>T), ABCG2 (421C>A), and CYP3A5 polymorphisms (6986A>G)[55]. The plasma trough C/D ratio of apixaban is significantly higher in patients with the ABCG2 421A/A genotype and CYP3A5 6986 G allele carriers than in patients with the ABCG2 421C/C genotype and CYP3A5 6986 A/A genotype[55]. Given that CYP3A5 6986A>G and ABCG2 421C>A polymorphisms have allele frequencies of 65%-85%[56] and 29%-36%[57], respectively, in Asians, ABCB1, ABCG2, and CYP3A5 genotypes play pivotal roles in the interindividual variability of apixaban concentrations in Japanese patients.

Although the pharmacodynamic parameters of DOACs differ significantly among individuals and anticoagulant effects also vary widely among patients receiving DOACs, the lack of approved methods to monitor the anticoagulant effect of DOACs makes it unclear whether the effect is adequate in patients receiving DOAC. Compared with vitamin K antagonist therapy (therapeutic international normalized ratio range: 2.0-3.0), a disadvantage of DOACs is that the dosage of DOAC cannot be controlled according to anticoagulant effect. Therefore, the trough and time to reach maximum plasma concentration (Tmax) and anti-FXa activity of DOAC-metabolizing enzyme polymorphisms may be useful parameters for accurately monitoring the anti-coagulate effects of DOACs and selecting patients at higher risk of major bleeding. Developing a system that easily measures the anti–FXa activity and plasma level could be an important way to monitor the anticoagulant effect of DOACs in clinical practice.

PHARMACOKINETICS OF DOACS AND MAJOR BLEEDING

Clinical trials have investigated the efficacy of dabigatran (RE-LY trial[58]), apixaban (ARISTOTLE trial[41]), edoxaban (ENGAGE AF TIMI48 trial[59]) and rivaroxaban (ROCKET AF trial[60]) for the prevention of stroke and embolism. According to these trials, adverse events of major bleeding and gastrointestinal bleeding occur at rates of 3.11% and 1.51% for dabigatran 150 mg given twice daily (bid), 2.13% and 0.76% at apixaban 5 mg bid, 2.75% and 1.51% at edoxaban 60 mg given once daily (oid) and 3.6% and 3.2% at rivaroxaban 20 mg oid (Table 3), respectively.

Table 3 Comparison of clinical trials on patients receiving direct oral anticoagulant: Major bleeding.

Dabigatran
Apixaban
Edoxaban
Rivaroxaban
Trial nameRE-LY[4]ARISTOTLE[5]ENGAGE AF TIMI48[6]ROCKET AF[7]/J-ROCKET AF
Number of patients18113182012110514264
MethodPROBERCTRCTRCT
Primary endpointsStroke or systemic embolismIschemic or hemorrhagic stroke or systemic embolismStroke or systemic embolismStroke or systemic embolism
Period (years)2.01.82.81.9
CHADS2 score (mean)2.22.12.83.48 (J-ROCKET: 3.25)
Dosing dose150 mg/10 mg bid5 mg bid60 mg/30 mg qd20 mg od (J-ROCKET: 15 mg od)
Evaluation
Thrombus/embolism (vs warfarin)110 mg: Non-inferior, 150 mg: SuperiorSuperior60 mg: Similar, 30 mg: SimilarOn treatment: Superior, Intention-to-treat: Non-inferior
Outcomes: Stroke or systemic embolismWar: 1.69%/yr, D (110): 1.51%/yr, D (150): 1.11%/yrWar: 1.50%/yr, A: 1.27%/yrWar: 1.81%/yr, E (30): 2.06%/yr, E (60): 1.57%/yrWar: 2.2%/yr, R: 1.7%/yr
Major bleeding (vs warfarin)110 mg: Superior, 150 mg: SimilarSuperiorSuperiorSimilar
Bleeding rateWar: 3.36%/yr, D (110): 2.71%/yr, D (150): 3.11%/yrWar: 3.09%/yr, A: 2.13%/yrWar: 3.43%/yr, E (30): 1.61%/yr, E (60): 2.75%/yrWar: 3.4%/yr, R: 3.6%/yr
Intracranial bleedingWar: 0.74%/yr, D (110): 0.23%/yr, D (150): 0.30%/yrWar: 2.27%/yr, A: 1.79%/yrWar: 0.85%/yr, E (30): 0.26%/yr, E (60): 0.39%/yrWar: 0.7%/yr, R: 0.5%/yr
Gastrointestinal bleedingWar: 1.02%/yr, D (110): 1.12%/yr, D (150): 1.51%/yrWar: 0.86%/yr, A: 0.76%/yrWar: 1.23%/yr, E (30): 0.82%/yr, E (60): 1.51%/yrWar: 2.2%/yr, R: 3.2%/yr1
Minor bleeding (vs warfarin)War: 16.37%/yr, D (110): 13.16%/yr, D (150): 14.84%/yrWar: 6.01%/yr, A: 4.07%/yrWar: 4.89%/yr, E (30): 3.52%/yr, E (60): 4.12%/yrWar: 11.4%/yr, R: 11.8%/yr
Mortality rateWar: 4.13%/yr, D (110): 3.75%/yr, D (150): 3.64%/yrWar: 3.94%/yr, A: 3.52%/yrWar: 4.35%/yr, E (30): 3.80%/yr, E (60): 3.99%/yrWar: 2.2%/yr, R: 1.9%/yr

Because the anti-coagulate effects of DOACs are linked to plasma levels of DOAC and anti-FXa activity at trough and Tmax[48,61,62], these pharmacokinetic and pharmacodynamic parameters are also expected to be related to risk of major bleeding, such as intracerebral and gastrointestinal bleeding. Using multiple logistic regression, Reilly et al[48] showed that major bleeding risk increased with dabigatran exposure (P < 0.0001), age (P < 0.0001), aspirin use (P < 0.0003), and diabetes (P = 0.018) as significant covariates. Further, patients with major bleeding had higher trough levels (55%) and post-dose levels (36%) than non-bleeding patients[48]. Reilly et al[48] also reported a median trough level of 116 ng/mL in 323 major bleeding patients compared with 75.3 ng/mL in 5899 no bleeding patients[48]. Additionally, a Cox regression analysis of time to first major bleeding with trough level, age, and CHADS2 score as covariates showed that, compared with the median trough level of 88 ng/mL, the rate of major bleeding doubled at a level of 210 ng/mL after adjustment for age and CHADS2 score. Moreover, Sakaguchi et al[63] showed that, in rivaroxaban-treated patients with major bleeding in Japan, major bleeding is independently predicted to be higher peak rivaroxaban levels and higher anti-FXa activity. Additionally, Sin et al’s prospective study[64] of rivaroxaban-treated patients with atrial fibrillation with differing severity of CKD (Stage 1–3) showed that trough levels of rivaroxaban were higher in those with bleeding (59.9 ± 35.6 ng/mL) than in those without (41.1 ± 29.2 ng/mL; P < 0.05). Therefore, although plasma level and anti–FXa activity may be useful parameters for selecting patients receiving DOACs at higher risk of major bleeding, there is no evidence that patients with higher plasma levels and anti–FXa activity have higher risk of post-ESD bleeding.

A retrospective analysis of 5041 patients demonstrated that concomitant dabigatran–PPI treatment is linked to a significant reduction in bleeding risk compared with dabigatran alone without a PPI[65]. Although dabigatran is an orally administered prodrug, it is rapidly absorbed and converted to its active form, dabigatran. Thus, a potential mechanism for PPI–dabigatran interaction may be reduced dabigatran absorption and availability, which is most probably mediated via the effects of PPI on gastric pH, given the poor solubility of dabigatran at pH > 4[66]. Therefore, interactions between PPIs and dabigatran may lead to decreases in dabigatran levels[67].

In terms of gastrointestinal bleeding, unlike warfarin, DOACs remain in the gastrointestinal tract without being absorbed into the blood. Therefore, DOACs may directly inhibit the hemostatic mechanism in the gastrointestinal tract, thereby aggravating bleeding[68].

CLINICAL GUIDELINES: MANAGEMENT FOR PATIENTS TAKING DOACS IN ENDOSCOPIC PROCEDURES WITH HIGHER RISK FOR BLEEDING (ESD)

During gastrointestinal endoscopy and endoscopic treatment of patients receiving antithrombotic therapy, it is necessary to balance the risk of major and minor bleeding with the risk of thromboembolism resulting from withdrawal of antithrombotic therapy. Therefore, it is important to determine a management strategy (with withdrawal or not) that is optimized for individual patients based on consultation between the endoscopist and physician prescribing the antithrombotic drugs. The risk of thromboembolism is closely related to the underlying disease requiring anticoagulants, and the absolute risk of thromboembolism increases by more than 1% when anticoagulants are withdrawn for more than 4 d[69].

In 2012 the Japan Gastroenterological Endoscopy Society published “Guidelines for gastroenterological endoscopy in patients undergoing antithrombotic treatment” concerning thromboembolism associated with antithrombotic therapy withdrawal and bleeding[70]. The guidelines were updated with a 2017 appendix on anticoagulants in 2017[71]. Although the 2012 version recommends replacing DOACs with heparin during ESD in patients with high risk for post-procedure bleeding as an adverse event, the 2017 version recommends that patients receiving DOACs should continue to take the DOAC orally until the day before ESD and discontinue it on the morning of ESD (Table 4)[71]. Because the anticoagulant effects last 36–48 h for rivaroxaban and edoxaban given oid and 24–36 h for apixaban given bid, and the disappearance of anti-FXa activity and anticoagulant effects increase the risk of thrombosis, DOACs may be resumed the morning after ESD (up to 36 and 48 h when given bid and oid). In addition, when performing ESD, patients receiving concomitant DOAC and antiplatelet agents should be handled with care depending on the individual’s condition and needs, while ESD can be performed on patients receiving antiplatelet monotherapy with aspirin or cilostazol[71].

Table 4 Summary of international guidelines concerning withdrawal of direct oral anticoagulants during gastroenterological endoscopy.
Ref.
Country
Standard endoscopy
Biopsy
Low risk of bleeding
High risk of bleeding, including ESD
[71]Japan12Avoid peak plasma level2Avoid peak plasma level3(1) Withdraw on the day of treatment; and (2) Heparin replacement
[72]Europe13Withdraw on the day of treatment3Withdraw on the day of treatment3(1) Withdraw 3 d before treatment; (2) Withdraw 5 d before treatment for dabigatran patients at Ccr 30–50 mL/min; and (3) No heparin bridging
[73]United States1113(1) Withdraw; and (2) Bridge therapy required for patients at high risk for thromboembolic events
[74]Korea1113Withdraw 2 d before treatment
[75]Asia-Pacific1113Withdraw 2 d before treatment

In contrast to the Japanese guidelines, the updated guidelines by the British Society of Gastroenterology and ESGE suggest that patients receiving low-risk procedures for bleeding withdraw from the morning dose of DOAC on the morning of ESD, and recommend that the last dose of DOAC be taken 3 d before ESD for high-risk endoscopic procedures (strong recommendation and low-quality evidence) (Table 4)[72]. For patients receiving dabigatran with a Ccr 30–50 mL/min, this updated version recommends that the last dose be taken 5 d prior to the procedure (strong recommendation and low-quality evidence)[72].

The American Society for Gastrointestinal Endoscopy (ASGE) published a guideline on how to manage patients receiving antithrombotic agents for endoscopy in 2016[73]. This guideline suggests that patients undergoing low-risk procedures should continue taking DOACs (Table 4)[73] and that the resumption of DOACs after ESD be delayed until adequate hemostasis is ensured. If DOACs cannot be resumed within 12 to 24 h post-ESD, thromboprophylaxis should be considered to decrease thromboembolism risk in those with high risk for thromboembolism. However, this version of the ASGE guideline recommends that patients at high risk for thromboembolic events withdraw DOACs and receive bridge therapy to adequately manage patients taking DOACs when ESD is performed (Table 4).

In contrast, the Korean clinical practice guideline does not recommend withdrawal of DOACs before low-risk procedures (weak recommendation and low-quality evidence) but suggests withdrawing DOACs > 48 h before high-risk procedures (weak recommendation and low-quality evidence) (Table 4)[74]. The recommendation to withdraw DOACs > 48 h before ESD is based on the fact that the half-life of DOACs is about 12 h and predictions that DOAC levels and anti-FXa activity will be almost undetectable after 48 h[74].

The Asian Pacific Association of Gastroenterology and Asian Pacific Society for Digestive Endoscopy guidelines recommend withholding DOACs at least 48 h before the procedure in DOAC patients receiving gastrointestinal ESD (strong recommendation and low-quality evidence) and do not recommend bridging anticoagulation (strong recommendation and low-quality evidence) (Table 4)[75]. In addition, these guidelines provide recommendations related to the timing of DOAC discontinuation before high-risk procedures according to Ccr[76]. In these guidelines, EMR for large colon polyp (≥ 2 cm) and ESD procedures, which have a higher risk of gastrointestinal bleeding compared with other high-risk endoscopic procedures, are categorized as ultra-high-risk endoscopic procedures[74,75].

Thus, as summarized in Table 4, guidelines for the management of patients undergoing ESD for early-stage gastrointestinal tumors who receive DOACs differ by country. Although subtle differences in the management of DOACs with ESD are important in clinical practice, we consider that these differences are dependent on the year in which the guidelines were published, the different dosage of DOAC in each country, the different numbers of concomitant antithrombotic drugs, and differences in the rate of genetic variations (e.g., CYP3A4/5, ABCG2, and ABCB1 polymorphism).

GASTROINTESTINAL BLEEDING AFTER ESD IN PATIENTS RECEIVING DOACS

Post-ESD bleeding in patients receiving DOACs remains an unpreventable adverse event. Although the combination of heparin-bridging therapy and discontinuation of DOACs is one approach used to prevent thrombosis[70,71], heparin-bridging therapy with discontinuation of any anticoagulants causes an increased risk of delayed bleeding after surgical treatment, interventional procedures and ESD (gastric ESD: 10.8%-61.5%)[77-80] and does not reduce the risk of perioperative arterial thromboembolism[77-79]. In fact, a meta-analysis focused on heparin-bridging therapy with discontinuation of any anticoagulants for ESD found an increased risk of post-ESD bleeding without any benefit for thrombosis[81]. Another meta-analysis reported an increase in thrombosis in patients receiving heparin-bridging therapy with discontinuation of anticoagulants compared with those who discontinued anticoagulation without heparin bridging[82]. In Japan, although the updated guideline recommends discontinuing DOACs on the morning of ESD, most studies conducted on ESD for patients receiving DOACs have been retrospective and enrolled small numbers of patients (Table 5).

Table 5 Delayed bleeding after endoscopic submucosal dissection in patients receiving direct oral anticoagulants.
Ref.
Year
Country
Type
Organ
DOAC patients
Bleeding rate
Non-DOAC patients
Bleeding rate
Nagata et al[83]2018JapanRetrospectiveUpper GI27539.6%301 (warfarin)45.8%
Horie et al[85]2022JapanRetrospectiveEsophagus16113%86920.3%1
Yoshio et al[88]2017JapanRetrospectiveStomach2420.8%73 (warfarin)24.6%
Sanomura et al[89]2018JapanRetrospectiveStomach2119.0%40 (warfarin)17.5%
Saito et al[90]2020JapanRetrospectiveStomach7719.5%66 (warfarin)22.7%
Hatta et al[36]2021JapanRetrospectiveStomach25317.0%10,0674.4%1
Tomida et al[84]2021JapanRetrospectiveStomach26114%467 (warfarin)18%
Choi et al[91]2021KoreaRetrospectiveStomach238.7%14993.0%
Kagawa et al[37]2022JapanRetrospectiveStomach3915.4%7524.3%1
Nagata et al[83]2018JapanRetrospectiveLower GI12113.2%111 (warfarin)25.9%
Yamashita et al[92]2018JapanRetrospectiveColon922.0%19 (warfarin)26.3%
Ogiyama et al[93]2020JapanRetrospectiveColon4323.3%44 (warfarin)11.4%
Harada et al[94]2020JapanRetrospectiveColon2516.0%26 (warfarin)7.7%

However, one study examined a large national database including 16977 patients receiving anticoagulation therapy who underwent high-risk endoscopic procedures. It showed that although warfarin led to a significantly higher post-procedure bleeding rate than DOACs (12.0% vs 9.9%, P = 0.002), the post-procedure bleeding rate was not significantly different in either upper or lower gastrointestinal ESD between patients receiving DOACs and warfarin[83]. In sub-analyses of procedure types in propensity-matched patients, the gastrointestinal bleeding rate in the DOAC group was 39.6% in patients who received upper gastrointestinal ESD and 13.2% in those who received lower gastrointestinal ESD[83].

In a recent retrospective Japanese study of 261 patients with early-stage gastric cancer receiving DOACs, post-ESD bleeding occurred in 14% of patients, which is comparable to that in patients receiving warfarin (18%)[84]. Multivariate analysis demonstrated that age ≥ 65 (OR 2.96, 95%CI: 1.13-7.73), male sex (OR 2.12; 95%CI: 1.01–4.45), receiving multiple antithrombotic agents (OR 2.70, 95%CI: 1.74-4.21) and lesion size ≥ 20 mm (OR 1.67, 95%CI: 1.08-2.59) were independent risk factors for post-ESD bleeding in patients taking anticoagulants, and that cessation of anticoagulants without heparin-bridging therapy was associated with a low risk of bleeding (OR 0.32; 95%CI: 0.14–0.76). However, the multivariate analysis identified no significant independent increased risk factors for post-ESD bleeding and demonstrated that dabigatran was associated with a significantly lower risk of bleeding (OR 0.04, 95%CI: 0.16-0.97)[84]. Further, in a nationwide multicenter retrospective study of 10320 patients, multivariate analysis conducted by Hatta et al[36] showed that taking anticoagulants was an independent risk factor for post-ESD bleeding (OR 8.16; 95%CI: 4.74-14.04). Although the number of patients registered in each of these previous studies is relatively small (n = 21-261), both reported post-ESD bleeding in 8.7%-20.8% of gastric cancer patients receiving DOACs, considered to be equivalent to that in patients receiving warfarin (17.5%-22.7%) and higher than that in patients not receiving anticoagulants (Table 5).

Few studies have been conducted on esophageal, duodenal and colorectal ESD in patients receiving DOACs. Horie et al[85] reported that the post-endoscopic resection bleeding rate in esophageal cancer patients receiving DOACs (14 patients received ESD and 2 patients received EMR) was significantly higher than that in those not receiving antithrombotic drugs [13% (95%CI: 1.6%–38%) vs 0.3% (95%CI: 0.1%–1%), P = 0.003]. Moreover, the post-ESD bleeding rate in colorectal tumor patients receiving DOACs was 16.0%-23.3%, which is equivalent to that in those receiving warfarin (7.7%-26.3%) (Table 5).

Thus, despite the small number of DOAC patients who received ESD in previous studies, the post-ESD delayed bleeding rate appears to vary among different organs in patients not receiving DOACs, but not in patients receiving DOACs.

FUTURE OF ESD FOR PATIENTS RECEIVING DOACS

There is no doubt that patients receiving DOACs are at higher risk of post-ESD bleeding than patients not taking DOACs or receiving antithrombotic drugs. As mentioned above, although examining plasma DOAC level and anti-Xa activity in relation to CYP metabolic enzymes or glycoprotein transporter gene polymorphisms may serve as predictive markers for selecting DOAC patients with higher risk of post-ESD delayed bleeding, no studies have been conducted with such considerations. Although scoring systems such as the BEST-J score are being developed to assess the risk of post-ESD delayed bleeding by stratifying multiple possible risk factors in clinical practice, we propose the need for a new scoring system that considers pharmacological parameters of DOACs, namely plasma DOAC level and anti-Xa activity.

DOACs is current recommended for not only patients with nonvalvular atrial fibrillation and venous thrombosis (VTE), but also those with cancer due to prevention of VTE by clinical guidelines[86]. Treatment or prophylaxis of VTE for patients with cancer must always balance the risk of incidence or recurrent VTE with the increased risk of major bleeding and take into consideration the consequences of these outcomes (including mortality, financial cost, quality of life)[86]. Developing a system that easily measures the anti–FXa activity and plasma level could be an important way to monitor the anticoagulant effect of DOACs and may help physicians to treat DOAC patients receiving ESD, endoscopic treatment, and surgical treatment and with cancer in clinical practice.

CONCLUSION

Post-ESD delayed bleeding for gastrointestinal tumors is a major adverse event, with an incidence of around 5%-8% for gastric cancer and 2%-4% for esophageal, duodenum and colorectal tumors. Of the many risk factors for bleeding, taking anticoagulants, including DOACs, is currently the biggest. In fact, the post-ESD bleeding rate in DOAC patients is 13% in esophageal cancer, 8.7%-20.8% in gastric cancer and 7.7%-26.3% in colorectal cancer. Compared with warfarin, the anticoagulant effects of which can be monitored using prothrombin time and international normalized ratio tests, there is currently no established method for monitoring the effects of DOACs. Thus, it is important to develop simple and accurate methods to evaluate the pharmacokinetics (e.g., plasma DOAC level at trough and Tmax) and pharmacodynamics (e.g., anti-factor Xa activity) of DOACs. In the future, a scoring system that includes pharmacological parameters of DOACs may be useful for stratifying risk of post-ESD delayed bleeding in clinical practice.

Footnotes

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

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Japan

Peer-review report’s scientific quality classification

Grade A (Excellent): A

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Gu ZC, China; Tharavej C, Thailand S-Editor: Li L L-Editor: A P-Editor: Zhao S

References
1.  Gotoda T, Yamamoto H, Soetikno RM. Endoscopic submucosal dissection of early gastric cancer. J Gastroenterol. 2006;41:929-942.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 485]  [Cited by in F6Publishing: 489]  [Article Influence: 27.2]  [Reference Citation Analysis (0)]
2.  Tanabe S, Ishido K, Higuchi K, Sasaki T, Katada C, Azuma M, Naruke A, Kim M, Koizumi W. Long-term outcomes of endoscopic submucosal dissection for early gastric cancer: a retrospective comparison with conventional endoscopic resection in a single center. Gastric Cancer. 2014;17:130-136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 94]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
3.  Ono H, Yao K, Fujishiro M, Oda I, Nimura S, Yahagi N, Iishi H, Oka M, Ajioka Y, Ichinose M, Matsui T. Guidelines for endoscopic submucosal dissection and endoscopic mucosal resection for early gastric cancer. Dig Endosc. 2016;28:3-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 354]  [Cited by in F6Publishing: 370]  [Article Influence: 46.3]  [Reference Citation Analysis (0)]
4.  Pimentel-Nunes P, Libânio D, Bastiaansen BAJ, Bhandari P, Bisschops R, Bourke MJ, Esposito G, Lemmers A, Maselli R, Messmann H, Pech O, Pioche M, Vieth M, Weusten BLAM, van Hooft JE, Deprez PH, Dinis-Ribeiro M. Endoscopic submucosal dissection for superficial gastrointestinal lesions: European Society of Gastrointestinal Endoscopy (ESGE) Guideline - Update 2022. Endoscopy. 2022;54:591-622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 149]  [Article Influence: 74.5]  [Reference Citation Analysis (0)]
5.  Tanaka S, Kashida H, Saito Y, Yahagi N, Yamano H, Saito S, Hisabe T, Yao T, Watanabe M, Yoshida M, Kudo SE, Tsuruta O, Sugihara KI, Watanabe T, Saitoh Y, Igarashi M, Toyonaga T, Ajioka Y, Ichinose M, Matsui T, Sugita A, Sugano K, Fujimoto K, Tajiri H. JGES guidelines for colorectal endoscopic submucosal dissection/endoscopic mucosal resection. Dig Endosc. 2015;27:417-434.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 359]  [Cited by in F6Publishing: 384]  [Article Influence: 42.7]  [Reference Citation Analysis (0)]
6.  Ishihara R, Arima M, Iizuka T, Oyama T, Katada C, Kato M, Goda K, Goto O, Tanaka K, Yano T, Yoshinaga S, Muto M, Kawakubo H, Fujishiro M, Yoshida M, Fujimoto K, Tajiri H, Inoue H; Japan Gastroenterological Endoscopy Society Guidelines Committee of ESD/EMR for Esophageal Cancer. Endoscopic submucosal dissection/endoscopic mucosal resection guidelines for esophageal cancer. Dig Endosc. 2020;32:452-493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 169]  [Article Influence: 42.3]  [Reference Citation Analysis (0)]
7.  Draganov PV, Wang AY, Othman MO, Fukami N. AGA Institute Clinical Practice Update: Endoscopic Submucosal Dissection in the United States. Clin Gastroenterol Hepatol. 2019;17:16-25.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 244]  [Article Influence: 48.8]  [Reference Citation Analysis (0)]
8.  Fujishiro M, Yahagi N, Kakushima N, Kodashima S, Muraki Y, Ono S, Kobayashi K, Hashimoto T, Yamamichi N, Tateishi A, Shimizu Y, Oka M, Ogura K, Kawabe T, Ichinose M, Omata M. Successful nonsurgical management of perforation complicating endoscopic submucosal dissection of gastrointestinal epithelial neoplasms. Endoscopy. 2006;38:1001-1006.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 123]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
9.  Tanaka M, Ono H, Hasuike N, Takizawa K. Endoscopic submucosal dissection of early gastric cancer. Digestion. 2008;77 Suppl 1:23-28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 108]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
10.  Cao Y, Liao C, Tan A, Gao Y, Mo Z, Gao F. Meta-analysis of endoscopic submucosal dissection versus endoscopic mucosal resection for tumors of the gastrointestinal tract. Endoscopy. 2009;41:751-757.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 274]  [Cited by in F6Publishing: 298]  [Article Influence: 19.9]  [Reference Citation Analysis (1)]
11.  Yoshizawa Y, Sugimoto M, Sato Y, Sahara S, Ichikawa H, Kagami T, Hosoda Y, Kimata M, Tamura S, Kobayashi Y, Osawa S, Sugimoto K, Miyajima H, Furuta T. Factors associated with healing of artificial ulcer after endoscopic submucosal dissection with reference to Helicobacter pylori infection, CYP2C19 genotype, and tumor location: Multicenter randomized trial. Dig Endosc. 2016;28:162-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
12.  Otsuka T, Sugimoto M, Ban H, Nakata T, Murata M, Nishida A, Inatomi O, Bamba S, Andoh A. Severity of gastric mucosal atrophy affects the healing speed of post-endoscopic submucosal dissection ulcers. World J Gastrointest Endosc. 2018;10:83-92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 10]  [Cited by in F6Publishing: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
13.  Yano T, Tanabe S, Ishido K, Suzuki M, Kawanishi N, Yamane S, Watanabe A, Wada T, Azuma M, Katada C, Koizumi W. Different clinical characteristics associated with acute bleeding and delayed bleeding after endoscopic submucosal dissection in patients with early gastric cancer. Surg Endosc. 2017;31:4542-4550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 36]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
14.  Sato C, Hirasawa K, Koh R, Ikeda R, Fukuchi T, Kobayashi R, Kaneko H, Makazu M, Maeda S. Postoperative bleeding in patients on antithrombotic therapy after gastric endoscopic submucosal dissection. World J Gastroenterol. 2017;23:5557-5566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 42]  [Cited by in F6Publishing: 45]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
15.  Muto M, Minashi K, Yano T, Saito Y, Oda I, Nonaka S, Omori T, Sugiura H, Goda K, Kaise M, Inoue H, Ishikawa H, Ochiai A, Shimoda T, Watanabe H, Tajiri H, Saito D. Early detection of superficial squamous cell carcinoma in the head and neck region and esophagus by narrow band imaging: a multicenter randomized controlled trial. J Clin Oncol. 2010;28:1566-1572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 427]  [Cited by in F6Publishing: 476]  [Article Influence: 34.0]  [Reference Citation Analysis (0)]
16.  Dohi O, Yagi N, Naito Y, Fukui A, Gen Y, Iwai N, Ueda T, Yoshida N, Kamada K, Uchiyama K, Takagi T, Konishi H, Yanagisawa A, Itoh Y. Blue laser imaging-bright improves the real-time detection rate of early gastric cancer: a randomized controlled study. Gastrointest Endosc. 2019;89:47-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 65]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
17.  Ono S, Kawada K, Dohi O, Kitamura S, Koike T, Hori S, Kanzaki H, Murao T, Yagi N, Sasaki F, Hashiguchi K, Oka S, Katada K, Shimoda R, Mizukami K, Suehiro M, Takeuchi T, Katsuki S, Tsuda M, Naito Y, Kawano T, Haruma K, Ishikawa H, Mori K, Kato M; LCI-FIND Trial Group. Linked Color Imaging Focused on Neoplasm Detection in the Upper Gastrointestinal Tract : A Randomized Trial. Ann Intern Med. 2021;174:18-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 47]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
18.  Sugimoto M, Koyama Y, Itoi T, Kawai T. Using texture and colour enhancement imaging to evaluate gastrointestinal diseases in clinical practice: a review. Ann Med. 2022;54:3315-3332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
19.  Takizawa K, Oda I, Gotoda T, Yokoi C, Matsuda T, Saito Y, Saito D, Ono H. Routine coagulation of visible vessels may prevent delayed bleeding after endoscopic submucosal dissection--an analysis of risk factors. Endoscopy. 2008;40:179-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 235]  [Cited by in F6Publishing: 254]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
20.  Muto M, Katada C, Sano Y, Yoshida S. Narrow band imaging: a new diagnostic approach to visualize angiogenesis in superficial neoplasia. Clin Gastroenterol Hepatol. 2005;3:S16-S20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 139]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
21.  Kim JS, Kim BW, Shin IS. Efficacy and safety of endoscopic submucosal dissection for superficial squamous esophageal neoplasia: a meta-analysis. Dig Dis Sci. 2014;59:1862-1869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 64]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
22.  Dubecz A, Solymosi N, Stadlhuber RJ, Schweigert M, Stein HJ, Peters JH. Does the Incidence of Adenocarcinoma of the Esophagus and Gastric Cardia Continue to Rise in the Twenty-First Century? J Gastrointest Surg. 2013;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 82]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
23.  Yang D, Zou F, Xiong S, Forde JJ, Wang Y, Draganov PV. Endoscopic submucosal dissection for early Barrett's neoplasia: a meta-analysis. Gastrointest Endosc. 2018;87:1383-1393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 67]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
24.  Suzuki H, Takizawa K, Hirasawa T, Takeuchi Y, Ishido K, Hoteya S, Yano T, Tanaka S, Endo M, Nakagawa M, Toyonaga T, Doyama H, Hirasawa K, Matsuda M, Yamamoto H, Fujishiro M, Hashimoto S, Maeda Y, Oyama T, Takenaka R, Yamamoto Y, Naito Y, Michida T, Kobayashi N, Kawahara Y, Hirano M, Jin M, Hori S, Niwa Y, Hikichi T, Shimazu T, Ono H, Tanabe S, Kondo H, Iishi H, Ninomiya M; Ichiro Oda for J-WEB/EGC group. Short-term outcomes of multicenter prospective cohort study of gastric endoscopic resection: 'Real-world evidence' in Japan. Dig Endosc. 2019;31:30-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 99]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
25.  Tanabe S, Ishido K, Matsumoto T, Kosaka T, Oda I, Suzuki H, Fujisaki J, Ono H, Kawata N, Oyama T, Takahashi A, Doyama H, Kobayashi M, Uedo N, Hamada K, Toyonaga T, Kawara F, Tanaka S, Yoshifuku Y. Long-term outcomes of endoscopic submucosal dissection for early gastric cancer: a multicenter collaborative study. Gastric Cancer. 2017;20:45-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 66]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
26.  Zullo A, Manta R, De Francesco V, Manfredi G, Buscarini E, Fiorini G, Vaira D, Marmo R. Endoscopic submucosal dissection of gastric neoplastic lesions in Western countries: systematic review and meta-analysis. Eur J Gastroenterol Hepatol. 2021;33:e1-e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
27.  Tao M, Zhou X, Hu M, Pan J. Endoscopic submucosal dissection versus endoscopic mucosal resection for patients with early gastric cancer: a meta-analysis. BMJ Open. 2019;9:e025803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 32]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
28.  Libânio D, Costa MN, Pimentel-Nunes P, Dinis-Ribeiro M. Risk factors for bleeding after gastric endoscopic submucosal dissection: a systematic review and meta-analysis. Gastrointest Endosc. 2016;84:572-586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 88]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
29.  Bureau  MoIAaCS. Population estimates. [cited 3 April 2023]. Available from: http://wwwstatgojp/data/jinsui/.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Sugimoto M, Hatta W, Tsuji Y, Yoshio T, Yabuuchi Y, Hoteya S, Doyama H, Nagami Y, Hikichi T, Kobayashi M, Morita Y, Sumiyoshi T, Iguchi M, Tomida H, Inoue T, Mikami T, Hasatani K, Nishikawa J, Matsumura T, Nebiki H, Nakamatsu D, Ohnita K, Suzuki H, Ueyama H, Hayashi Y, Murata M, Yamaguchi S, Michida T, Yada T, Asahina Y, Narasaka T, Kuribayashi S, Kiyotoki S, Mabe K, Fujishiro M, Masamune A, Kawai T. Risk Factors for Bleeding After Endoscopic Submucosal Dissection for Gastric Cancer in Elderly Patients Older Than 80 Years in Japan. Clin Transl Gastroenterol. 2021;12:e00404.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
31.  Zou J, Chai N, Linghu E, Zhai Y, Li Z, Du C, Li L. Clinical outcomes of endoscopic resection for non-ampullary duodenal laterally spreading tumors. Surg Endosc. 2019;33:4048-4056.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
32.  Inoue T, Uedo N, Yamashina T, Yamamoto S, Hanaoka N, Takeuchi Y, Higashino K, Ishihara R, Iishi H, Tatsuta M, Takahashi H, Eguchi H, Ohigashi H. Delayed perforation: a hazardous complication of endoscopic resection for non-ampullary duodenal neoplasm. Dig Endosc. 2014;26:220-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 127]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
33.  Watanabe D, Hayashi H, Kataoka Y, Hashimoto T, Ichimasa K, Miyachi H, Tanaka S, Toyonaga T. Efficacy and safety of endoscopic submucosal dissection for non-ampullary duodenal polyps: A systematic review and meta-analysis. Dig Liver Dis. 2019;51:774-781.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
34.  Kato M, Ochiai Y, Fukuhara S, Maehata T, Sasaki M, Kiguchi Y, Akimoto T, Fujimoto A, Nakayama A, Kanai T, Yahagi N. Clinical impact of closure of the mucosal defect after duodenal endoscopic submucosal dissection. Gastrointest Endosc. 2019;89:87-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 69]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
35.  Fuccio L, Hassan C, Ponchon T, Mandolesi D, Farioli A, Cucchetti A, Frazzoni L, Bhandari P, Bellisario C, Bazzoli F, Repici A. Clinical outcomes after endoscopic submucosal dissection for colorectal neoplasia: a systematic review and meta-analysis. Gastrointest Endosc. 2017;86:74-86.e17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 163]  [Article Influence: 23.3]  [Reference Citation Analysis (0)]
36.  Hatta W, Tsuji Y, Yoshio T, Kakushima N, Hoteya S, Doyama H, Nagami Y, Hikichi T, Kobayashi M, Morita Y, Sumiyoshi T, Iguchi M, Tomida H, Inoue T, Koike T, Mikami T, Hasatani K, Nishikawa J, Matsumura T, Nebiki H, Nakamatsu D, Ohnita K, Suzuki H, Ueyama H, Hayashi Y, Sugimoto M, Yamaguchi S, Michida T, Yada T, Asahina Y, Narasaka T, Kuribasyashi S, Kiyotoki S, Mabe K, Nakamura T, Nakaya N, Fujishiro M, Masamune A. Prediction model of bleeding after endoscopic submucosal dissection for early gastric cancer: BEST-J score. Gut. 2021;70:476-484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 62]  [Article Influence: 20.7]  [Reference Citation Analysis (0)]
37.  Kagawa Y, Fukuzawa M, Sugimoto M, Nemoto D, Muramatsu T, Shinohara H, Matsumoto T, Madarame A, Yamaguchi H, Uchida K, Morise T, Koyama Y, Sugimoto A, Yamauchi Y, Kono S, Naito S, Yamamoto K, Kishimoto Y, Inuyama M, Kawai T, Itoi T. Validation of the BEST-J score, a prediction model for bleeding after endoscopic submucosal dissection for early gastric cancer: a multicenter retrospective observational study. Surg Endosc. 2022;36:7240-7249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
38.  Ban H, Sugimoto M, Otsuka T, Murata M, Nakata T, Hasegawa H, Fukuda M, Inatomi O, Bamba S, Kushima R, Andoh A. Letter: a potassium-competitive acid blocker vs a proton pump inhibitor for healing endoscopic submucosal dissection-induced artificial ulcers after treatment of gastric neoplasms. Aliment Pharmacol Ther. 2017;46:564-565.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
39.  Li R, Cai S, Sun D, Shi Q, Ren Z, Qi Z, Li B, Yao L, Xu M, Zhou P, Zhong Y. Risk factors for delayed bleeding after endoscopic submucosal dissection of colorectal tumors. Surg Endosc. 2021;35:6583-6590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
40.  Seo M, Song EM, Cho JW, Lee YJ, Lee BI, Kim JS, Jeon SW, Jang HJ, Yang DH, Ye BD, Byeon JS. A risk-scoring model for the prediction of delayed bleeding after colorectal endoscopic submucosal dissection. Gastrointest Endosc. 2019;89:990-998.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 28]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
41.  Granger CB, Alexander JH, McMurray JJ, Lopes RD, Hylek EM, Hanna M, Al-Khalidi HR, Ansell J, Atar D, Avezum A, Bahit MC, Diaz R, Easton JD, Ezekowitz JA, Flaker G, Garcia D, Geraldes M, Gersh BJ, Golitsyn S, Goto S, Hermosillo AG, Hohnloser SH, Horowitz J, Mohan P, Jansky P, Lewis BS, Lopez-Sendon JL, Pais P, Parkhomenko A, Verheugt FW, Zhu J, Wallentin L; ARISTOTLE Committees and Investigators. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2011;365:981-992.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6075]  [Cited by in F6Publishing: 6091]  [Article Influence: 468.5]  [Reference Citation Analysis (0)]
42.  Hu PT, Lopes RD, Stevens SR, Wallentin L, Thomas L, Alexander JH, Hanna M, Lewis BS, Verheugt FW, Granger CB, Jones WS. Efficacy and Safety of Apixaban Compared With Warfarin in Patients With Atrial Fibrillation and Peripheral Artery Disease: Insights From the ARISTOTLE Trial. J Am Heart Assoc. 2017;6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 29]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
43.  Easton JD, Lopes RD, Bahit MC, Wojdyla DM, Granger CB, Wallentin L, Alings M, Goto S, Lewis BS, Rosenqvist M, Hanna M, Mohan P, Alexander JH, Diener HC; ARISTOTLE Committees and Investigators. Apixaban compared with warfarin in patients with atrial fibrillation and previous stroke or transient ischaemic attack: a subgroup analysis of the ARISTOTLE trial. Lancet Neurol. 2012;11:503-511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 198]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
44.  Rose DK, Bar B. Direct Oral Anticoagulant Agents: Pharmacologic Profile, Indications, Coagulation Monitoring, and Reversal Agents. J Stroke Cerebrovasc Dis. 2018;27:2049-2058.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
45.  Conway SE, Hwang AY, Ponte CD, Gums JG. Laboratory and Clinical Monitoring of Direct Acting Oral Anticoagulants: What Clinicians Need to Know. Pharmacotherapy. 2017;37:236-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 74]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
46.  Cuker A, Siegal DM, Crowther MA, Garcia DA. Laboratory measurement of the anticoagulant activity of the non-vitamin K oral anticoagulants. J Am Coll Cardiol. 2014;64:1128-1139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 339]  [Cited by in F6Publishing: 324]  [Article Influence: 36.0]  [Reference Citation Analysis (0)]
47.  Frost C, Song Y, Barrett YC, Wang J, Pursley J, Boyd RA, LaCreta F. A randomized direct comparison of the pharmacokinetics and pharmacodynamics of apixaban and rivaroxaban. Clin Pharmacol. 2014;6:179-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 46]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
48.  Reilly PA, Lehr T, Haertter S, Connolly SJ, Yusuf S, Eikelboom JW, Ezekowitz MD, Nehmiz G, Wang S, Wallentin L; RE-LY Investigators. The effect of dabigatran plasma concentrations and patient characteristics on the frequency of ischemic stroke and major bleeding in atrial fibrillation patients: the RE-LY Trial (Randomized Evaluation of Long-Term Anticoagulation Therapy). J Am Coll Cardiol. 2014;63:321-328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 582]  [Cited by in F6Publishing: 630]  [Article Influence: 57.3]  [Reference Citation Analysis (0)]
49.  Chen A, Stecker E, A Warden B. Direct Oral Anticoagulant Use: A Practical Guide to Common Clinical Challenges. J Am Heart Assoc. 2020;9:e017559.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 245]  [Article Influence: 61.3]  [Reference Citation Analysis (0)]
50.  Derebail VK, Rheault MN, Kerlin BA. Role of direct oral anticoagulants in patients with kidney disease. Kidney Int. 2020;97:664-675.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
51.  Martin K, Beyer-Westendorf J, Davidson BL, Huisman MV, Sandset PM, Moll S. Use of the direct oral anticoagulants in obese patients: guidance from the SSC of the ISTH. J Thromb Haemost. 2016;14:1308-1313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 313]  [Cited by in F6Publishing: 318]  [Article Influence: 39.8]  [Reference Citation Analysis (0)]
52.  Lee SR, Choi EK, Park CS, Han KD, Jung JH, Oh S, Lip GYH. Direct Oral Anticoagulants in Patients With Nonvalvular Atrial Fibrillation and Low Body Weight. J Am Coll Cardiol. 2019;73:919-931.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 63]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
53.  Fralick M, Juurlink DN, Marras T. Bleeding associated with coadministration of rivaroxaban and clarithromycin. CMAJ. 2016;188:669-672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
54.  Mueck W, Kubitza D, Becka M. Co-administration of rivaroxaban with drugs that share its elimination pathways: pharmacokinetic effects in healthy subjects. Br J Clin Pharmacol. 2013;76:455-466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 261]  [Article Influence: 26.1]  [Reference Citation Analysis (0)]
55.  Ueshima S, Hira D, Fujii R, Kimura Y, Tomitsuka C, Yamane T, Tabuchi Y, Ozawa T, Itoh H, Horie M, Terada T, Katsura T. Impact of ABCB1, ABCG2, and CYP3A5 polymorphisms on plasma trough concentrations of apixaban in Japanese patients with atrial fibrillation. Pharmacogenet Genomics. 2017;27:329-336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
56.  Staatz CE, Goodman LK, Tett SE. Effect of CYP3A and ABCB1 single nucleotide polymorphisms on the pharmacokinetics and pharmacodynamics of calcineurin inhibitors: Part I. Clin Pharmacokinet. 2010;49:141-175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 262]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
57.  Ieiri I. Functional significance of genetic polymorphisms in P-glycoprotein (MDR1, ABCB1) and breast cancer resistance protein (BCRP, ABCG2). Drug Metab Pharmacokinet. 2012;27:85-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 116]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
58.  Connolly SJ, Ezekowitz MD, Yusuf S, Eikelboom J, Oldgren J, Parekh A, Pogue J, Reilly PA, Themeles E, Varrone J, Wang S, Alings M, Xavier D, Zhu J, Diaz R, Lewis BS, Darius H, Diener HC, Joyner CD, Wallentin L; RE-LY Steering Committee and Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med. 2009;361:1139-1151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7917]  [Cited by in F6Publishing: 7707]  [Article Influence: 513.8]  [Reference Citation Analysis (0)]
59.  Giugliano RP, Ruff CT, Braunwald E, Murphy SA, Wiviott SD, Halperin JL, Waldo AL, Ezekowitz MD, Weitz JI, Špinar J, Ruzyllo W, Ruda M, Koretsune Y, Betcher J, Shi M, Grip LT, Patel SP, Patel I, Hanyok JJ, Mercuri M, Antman EM; ENGAGE AF-TIMI 48 Investigators. Edoxaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2013;369:2093-2104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3447]  [Cited by in F6Publishing: 3477]  [Article Influence: 316.1]  [Reference Citation Analysis (0)]
60.  Patel MR, Mahaffey KW, Garg J, Pan G, Singer DE, Hacke W, Breithardt G, Halperin JL, Hankey GJ, Piccini JP, Becker RC, Nessel CC, Paolini JF, Berkowitz SD, Fox KA, Califf RM; ROCKET AF Investigators. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med. 2011;365:883-891.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6519]  [Cited by in F6Publishing: 6475]  [Article Influence: 498.1]  [Reference Citation Analysis (2)]
61.  Weitz JI, Connolly SJ, Patel I, Salazar D, Rohatagi S, Mendell J, Kastrissios H, Jin J, Kunitada S. Randomised, parallel-group, multicentre, multinational phase 2 study comparing edoxaban, an oral factor Xa inhibitor, with warfarin for stroke prevention in patients with atrial fibrillation. Thromb Haemost. 2010;104:633-641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 251]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
62.  Salazar DE, Mendell J, Kastrissios H, Green M, Carrothers TJ, Song S, Patel I, Bocanegra TS, Antman EM, Giugliano RP, Kunitada S, Dornseif B, Shi M, Tachibana M, Zhou S, Rohatagi S. Modelling and simulation of edoxaban exposure and response relationships in patients with atrial fibrillation. Thromb Haemost. 2012;107:925-936.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 115]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
63.  Sakaguchi T, Osanai H, Murase Y, Ishii H, Nakashima Y, Asano H, Suzuki S, Takefuji M, Inden Y, Sakai K, Murohara T, Ajioka M. Monitoring of anti-Xa activity and factors related to bleeding events: A study in Japanese patients with nonvalvular atrial fibrillation receiving rivaroxaban. J Cardiol. 2017;70:244-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
64.  Sin CF, Wong KP, Wong HM, Siu CW, Yap DYH. Plasma Rivaroxaban Level in Patients With Early Stages of Chronic Kidney Disease-Relationships With Renal Function and Clinical Events. Front Pharmacol. 2022;13:888660.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
65.  Chan EW, Lau WC, Leung WK, Mok MT, He Y, Tong TS, Wong IC. Prevention of Dabigatran-Related Gastrointestinal Bleeding With Gastroprotective Agents: A Population-Based Study. Gastroenterology. 2015;149:586-95.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 154]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
66.  Ollier E, Hodin S, Basset T, Accassat S, Bertoletti L, Mismetti P, Delavenne X. In vitro and in vivo evaluation of drug-drug interaction between dabigatran and proton pump inhibitors. Fundam Clin Pharmacol. 2015;29:604-614.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
67.  Bolek T, Samoš M, Škorňová I, Galajda P, Staško J, Kubisz P, Mokáň M. Proton Pump Inhibitors and Dabigatran Therapy: Impact on Gastric Bleeding and Dabigatran Plasma Levels. Semin Thromb Hemost. 2019;45:846-850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
68.  Desai J, Granger CB, Weitz JI, Aisenberg J. Novel oral anticoagulants in gastroenterology practice. Gastrointest Endosc. 2013;78:227-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 72]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
69.  Garcia DA, Regan S, Henault LE, Upadhyay A, Baker J, Othman M, Hylek EM. Risk of thromboembolism with short-term interruption of warfarin therapy. Arch Intern Med. 2008;168:63-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 245]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
70.  Fujimoto K, Fujishiro M, Kato M, Higuchi K, Iwakiri R, Sakamoto C, Uchiyama S, Kashiwagi A, Ogawa H, Murakami K, Mine T, Yoshino J, Kinoshita Y, Ichinose M, Matsui T; Japan Gastroenterological Endoscopy Society. Guidelines for gastroenterological endoscopy in patients undergoing antithrombotic treatment. Dig Endosc. 2014;26:1-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 304]  [Article Influence: 30.4]  [Reference Citation Analysis (0)]
71.  Kato M, Uedo N, Hokimoto S, Ieko M, Higuchi K, Murakami K, Fujimoto K. Guidelines for Gastroenterological Endoscopy in Patients Undergoing Antithrombotic Treatment: 2017 Appendix on Anticoagulants Including Direct Oral Anticoagulants. Dig Endosc. 2018;30:433-440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 127]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
72.  Veitch AM, Radaelli F, Alikhan R, Dumonceau JM, Eaton D, Jerrome J, Lester W, Nylander D, Thoufeeq M, Vanbiervliet G, Wilkinson JR, Van Hooft JE. Endoscopy in patients on antiplatelet or anticoagulant therapy: British Society of Gastroenterology (BSG) and European Society of Gastrointestinal Endoscopy (ESGE) guideline update. Gut. 2021;70:1611-1628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 49]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
73.  ASGE Standards of Practice Committee; Acosta RD, Abraham NS, Chandrasekhara V, Chathadi KV, Early DS, Eloubeidi MA, Evans JA, Faulx AL, Fisher DA, Fonkalsrud L, Hwang JH, Khashab MA, Lightdale JR, Muthusamy VR, Pasha SF, Saltzman JR, Shaukat A, Shergill AK, Wang A, Cash BD, DeWitt JM. The management of antithrombotic agents for patients undergoing GI endoscopy. Gastrointest Endosc. 2016;83:3-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 417]  [Cited by in F6Publishing: 388]  [Article Influence: 48.5]  [Reference Citation Analysis (1)]
74.  Lim H, Gong EJ, Min BH, Kang SJ, Shin CM, Byeon JS, Choi M, Park CG, Cho JY, Lee ST, Kim HG, Chun HJ. Clinical Practice Guideline for the Management of Antithrombotic Agents in Patients Undergoing Gastrointestinal Endoscopy. Clin Endosc. 2020;53:663-677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
75.  Chan FKL, Goh KL, Reddy N, Fujimoto K, Ho KY, Hokimoto S, Jeong YH, Kitazono T, Lee HS, Mahachai V, Tsoi KKF, Wu MS, Yan BP, Sugano K. Management of patients on antithrombotic agents undergoing emergency and elective endoscopy: joint Asian Pacific Association of Gastroenterology (APAGE) and Asian Pacific Society for Digestive Endoscopy (APSDE) practice guidelines. Gut. 2018;67:405-417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 99]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
76.  Dunn A. Perioperative management of oral anticoagulation: when and how to bridge. J Thromb Thrombolysis. 2006;21:85-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 23]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
77.  Siegal D, Yudin J, Kaatz S, Douketis JD, Lim W, Spyropoulos AC. Periprocedural heparin bridging in patients receiving vitamin K antagonists: systematic review and meta-analysis of bleeding and thromboembolic rates. Circulation. 2012;126:1630-1639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 307]  [Cited by in F6Publishing: 280]  [Article Influence: 23.3]  [Reference Citation Analysis (0)]
78.  Beyer-Westendorf J, Gelbricht V, Förster K, Ebertz F, Köhler C, Werth S, Kuhlisch E, Stange T, Thieme C, Daschkow K, Weiss N. Peri-interventional management of novel oral anticoagulants in daily care: results from the prospective Dresden NOAC registry. Eur Heart J. 2014;35:1888-1896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 243]  [Article Influence: 24.3]  [Reference Citation Analysis (0)]
79.  Shindo Y, Matsumoto S, Miyatani H, Yoshida Y, Mashima H. Risk factors for postoperative bleeding after gastric endoscopic submucosal dissection in patients under antithrombotics. World J Gastrointest Endosc. 2016;8:349-356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 27]  [Cited by in F6Publishing: 27]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
80.  Yamamoto Y, Kikuchi D, Nagami Y, Nonaka K, Tsuji Y, Fujimoto A, Sanomura Y, Tanaka K, Abe S, Zhang S, De Lusong MA, Uedo N. Management of adverse events related to endoscopic resection of upper gastrointestinal neoplasms: Review of the literature and recommendations from experts. Dig Endosc. 2019;31 Suppl 1:4-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 70]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
81.  Jaruvongvanich V, Sempokuya T, Wijarnpreecha K, Ungprasert P. Continued versus interrupted aspirin use and bleeding risk after endoscopic submucosal dissection of gastric neoplasms: a meta-analysis. Ann Gastroenterol. 2018;31:344-349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
82.  Dong J, Wei K, Deng J, Zhou X, Huang X, Deng M, Lü M. Effects of antithrombotic therapy on bleeding after endoscopic submucosal dissection. Gastrointest Endosc. 2017;86:807-816.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 38]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
83.  Nagata N, Yasunaga H, Matsui H, Fushimi K, Watanabe K, Akiyama J, Uemura N, Niikura R. Therapeutic endoscopy-related GI bleeding and thromboembolic events in patients using warfarin or direct oral anticoagulants: results from a large nationwide database analysis. Gut. 2018;67:1805-1812.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 56]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
84.  Tomida H, Yoshio T, Igarashi K, Morita Y, Oda I, Inoue T, Hikichi T, Sumiyoshi T, Doyama H, Tsuji Y, Nishikawa J, Hatta W, Mikami T, Iguchi M, Sumiyama K, Yamamoto K, Kitamura K, Kuribayashi S, Yanagitani A, Uraoka T, Yada T, Hasatani K, Kawaguchi K, Fujita T, Nishida T, Hiasa Y, Fujishiro M; FIGHT-Japan Study Group. Influence of anticoagulants on the risk of delayed bleeding after gastric endoscopic submucosal dissection: a multicenter retrospective study. Gastric Cancer. 2021;24:179-189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 19]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
85.  Horie Y, Horiuchi Y, Ishiyama A, Tsuchida T, Yoshimizu S, Hirasawa T, Fujisaki J, Maetani I, Yoshio T. The effect of antithrombotic drug use on delayed bleeding with esophageal endoscopic resection. J Gastroenterol Hepatol. 2022;37:1792-1800.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
86.  Lyman GH, Carrier M, Ay C, Di Nisio M, Hicks LK, Khorana AA, Leavitt AD, Lee AYY, Macbeth F, Morgan RL, Noble S, Sexton EA, Stenehjem D, Wiercioch W, Kahale LA, Alonso-Coello P. American Society of Hematology 2021 guidelines for management of venous thromboembolism: prevention and treatment in patients with cancer. Blood Adv. 2021;5:927-974.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 317]  [Cited by in F6Publishing: 363]  [Article Influence: 121.0]  [Reference Citation Analysis (0)]
87.  Samoš M, Bolek T, Stančiaková L, Péč MJ, Brisudová K, Škorňová I, Staško J, Mokáň M, Kubisz P. Tailored Direct Oral Anticoagulation in Patients with Atrial Fibrillation: The Future of Oral Anticoagulation? J Clin Med. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
88.  Yoshio T, Tomida H, Iwasaki R, Horiuchi Y, Omae M, Ishiyama A, Hirasawa T, Yamamoto Y, Tsuchida T, Fujisaki J, Yamada T, Mita E, Ninomiya T, Michitaka K, Igarashi M. Effect of direct oral anticoagulants on the risk of delayed bleeding after gastric endoscopic submucosal dissection. Dig Endosc. 2017;29:686-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
89.  Sanomura Y, Oka S, Tanaka S, Yorita N, Kuroki K, Kurihara M, Mizumoto T, Yoshifuku Y, Chayama K. Taking Warfarin with Heparin Replacement and Direct Oral Anticoagulant Is a Risk Factor for Bleeding after Endoscopic Submucosal Dissection for Early Gastric Cancer. Digestion. 2018;97:240-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
90.  Saito H, Igarashi K, Hirasawa D, Okuzono T, Suzuki K, Abe Y, Nawata Y, Tanaka Y, Tanaka I, Unno S, Nishikawa Y, Tsubokura M, Nakahori M, Chonan A, Matsuda T. The risks and characteristics of the delayed bleeding after endoscopic submucosal dissection for early gastric carcinoma in cases with anticoagulants. Scand J Gastroenterol. 2020;55:1253-1260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
91.  Choi J, Cho SJ, Na SH, Lee A, Kim JL, Chung H, Kim SG. Use of direct oral anticoagulants does not significantly increase delayed bleeding after endoscopic submucosal dissection for early gastric neoplasms. Sci Rep. 2021;11:9399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
92.  Yamashita K, Oka S, Tanaka S, Boda K, Hirano D, Sumimoto K, Mizumoto T, Ninomiya Y, Tamaru Y, Shigita K, Hayashi N, Sanomura Y, Chayama K. Use of anticoagulants increases risk of bleeding after colorectal endoscopic submucosal dissection. Endosc Int Open. 2018;6:E857-E864.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
93.  Ogiyama H, Inoue T, Maekawa A, Yoshii S, Yamaguchi S, Nagai K, Yamamoto M, Egawa S, Horimoto M, Ogawa H, Nishihara A, Komori M, Kizu T, Tsutsui S, Tsujii Y, Hayashi Y, Iijima H, Takehara T. Effect of anticoagulants on the risk of delayed bleeding after colorectal endoscopic submucosal dissection. Endosc Int Open. 2020;8:E1654-E1663.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
94.  Harada H, Nakahara R, Murakami D, Suehiro S, Nagasaka T, Ujihara T, Sagami R, Katsuyama Y, Hayasaka K, Tounou S, Amano Y. The effect of anticoagulants on delayed bleeding after colorectal endoscopic submucosal dissection. Surg Endosc. 2020;34:3330-3337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]