Endoscopy 2015; 47(10): a1-a46
DOI: 10.1055/s-0034-1393172
Guideline
© Georg Thieme Verlag KG Stuttgart · New York

Diagnosis and management of nonvariceal upper gastrointestinal hemorrhage: European Society of Gastrointestinal Endoscopy (ESGE) Guideline

Ian M. Gralnek
1   Institute of Gastroenterology and Liver Diseases, Ha'Emek Medical Center, Afula, Israel
2   Rappaport Faculty of Medicine Technion-Israel Institute of Technology, Haifa, Israel
,
Jean-Marc Dumonceau
3   Gedyt Endoscopy Center, Buenos Aires, Argentina
,
Ernst J. Kuipers
4   Departments of Internal Medicine and Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
,
Angel Lanas
5   University of Zaragoza, Aragon Health Research Institute (IIS Aragon), CIBERehd, Spain
,
David S. Sanders
6   Department of Gastroenterology, Sheffield Teaching Hospitals, United Kingdom
,
Matthew Kurien
6   Department of Gastroenterology, Sheffield Teaching Hospitals, United Kingdom
,
Gianluca Rotondano
7   Division of Gastroenterology and Digestive Endoscopy, Maresca Hospital, Torre del Greco, Italy
,
Tomas Hucl
8   Department of Gastroenterology and Hepatology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
,
Mario Dinis-Ribeiro
9   Department of Gastroenterology, IPO Porto, Portugal and CINTESIS, Porto Faculty of Medicine, Portgal
,
Riccardo Marmo
10   Division of Gastroenterology, Hospital Curto, Polla, Italy
,
Istvan Racz
11   First Department of Internal Medicine and Gastroenterology, Petz Aladar, Hospital, Gyor, Hungary
,
Alberto Arezzo
12   Department of Surgical Sciences, University of Torino, Torino, Italy
,
Ralf-Thorsten Hoffmann
13   Institute and Polyclinic for Diagnostic Radiology, University Hospital Dresden-TU, Dresden, Germany
,
Gilles Lesur
14   Department of Gastroenterology, Ambroise Paré Hospital, Boulogne, France
,
Roberto de Franchis
15   Department of Biomedical and Clinical Sciences, University of Milan, Gastroenterology Unit, Luigi Sacco University Hospital, Milan, Italy
,
Lars Aabakken
16   Department of Medical Gastroenterology, Rikshospitalet University Hospital, Oslo, Norway
,
Andrew Veitch
17   Department of Gastroenterology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, United Kingdom
,
Franco Radaelli
18   Department of Gastroenterology, Valduce Hospital, Como, Italy
,
Paulo Salgueiro
19   Department of Gastroenterology, Centro Hospitalar do Porto, Portugal
,
Ricardo Cardoso
20   Department of Gastroenterology, Centro Hospitalar e Universitário de Coimbra, Portugal
,
Luís Maia
19   Department of Gastroenterology, Centro Hospitalar do Porto, Portugal
,
Angelo Zullo
21   Endoscopy Unit, Nuovo Regina Margherita Hospital, Rome, Italy
,
Livio Cipolletta
22   Gastroenterology and Endoscopy Department, Antonio Cardarelli Hospital, Naples, Italy
,
Cesare Hassan
23   Digestive Endoscopy Unit, Catholic University, Rome, Italy
› Author Affiliations
Further Information

Corresponding author

Ian M. Gralnek, MD, MSHS
Institute of Gastroenterology and Liver Diseases, Ha'Emek Medical Center
Rappaport Faculty of Medicine, Technion-Israel Institute of Technology
Afula, Israel 18101
Fax: +972-4-6495314   

Publication History

Publication Date:
29 September 2015 (online)

 

This Guideline is an official statement of the European Society of Gastrointestinal Endoscopy (ESGE). It addresses the diagnosis and management of nonvariceal upper gastrointestinal hemorrhage (NVUGIH).

Main Recommendations

MR1. ESGE recommends immediate assessment of hemodynamic status in patients who present with acute upper gastrointestinal hemorrhage (UGIH), with prompt intravascular volume replacement initially using crystalloid fluids if hemodynamic instability exists (strong recommendation, moderate quality evidence).

MR2. ESGE recommends a restrictive red blood cell transfusion strategy that aims for a target hemoglobin between 7 g/dL and 9 g/dL. A higher target hemoglobin should be considered in patients with significant co-morbidity (e. g., ischemic cardiovascular disease) (strong recommendation, moderate quality evidence).

MR3. ESGE recommends the use of the Glasgow-Blatchford Score (GBS) for pre-endoscopy risk stratification. Outpatients determined to be at very low risk, based upon a GBS score of 0 – 1, do not require early endoscopy nor hospital admission. Discharged patients should be informed of the risk of recurrent bleeding and be advised to maintain contact with the discharging hospital (strong recommendation, moderate quality evidence).

MR4. ESGE recommends initiating high dose intravenous proton pump inhibitors (PPI), intravenous bolus followed by continuous infusion (80 mg then 8 mg/hour), in patients presenting with acute UGIH awaiting upper endoscopy. However, PPI infusion should not delay the performance of early endoscopy (strong recommendation, high quality evidence).

MR5. ESGE does not recommend the routine use of nasogastric or orogastric aspiration/lavage in patients presenting with acute UGIH (strong recommendation, moderate quality evidence).

MR6. ESGE recommends intravenous erythromycin (single dose, 250 mg given 30 – 120 minutes prior to upper gastrointestinal [GI] endoscopy) in patients with clinically severe or ongoing active UGIH. In selected patients, pre-endoscopic infusion of erythromycin significantly improves endoscopic visualization, reduces the need for second-look endoscopy, decreases the number of units of blood transfused, and reduces duration of hospital stay (strong recommendation, high quality evidence).

MR7. Following hemodynamic resuscitation, ESGE recommends early (≤ 24 hours) upper GI endoscopy. Very early (< 12 hours) upper GI endoscopy may be considered in patients with high risk clinical features, namely: hemodynamic instability (tachycardia, hypotension) that persists despite ongoing attempts at volume resuscitation; in-hospital bloody emesis/nasogastric aspirate; or contraindication to the interruption of anticoagulation (strong recommendation, moderate quality evidence).

MR8. ESGE recommends that peptic ulcers with spurting or oozing bleeding (Forrest classification Ia and Ib, respectively) or with a nonbleeding visible vessel (Forrest classification IIa) receive endoscopic hemostasis because these lesions are at high risk for persistent bleeding or rebleeding (strong recommendation, high quality evidence).

MR9. ESGE recommends that peptic ulcers with an adherent clot (Forrest classification IIb) be considered for endoscopic clot removal. Once the clot is removed, any identified underlying active bleeding (Forrest classification Ia or Ib) or nonbleeding visible vessel (Forrest classification IIa) should receive endoscopic hemostasis (weak recommendation, moderate quality evidence).

MR10. In patients with peptic ulcers having a flat pigmented spot (Forrest classification IIc) or clean base (Forrest classification III), ESGE does not recommend endoscopic hemostasis as these stigmata present a low risk of recurrent bleeding. In selected clinical settings, these patients may be discharged to home on standard PPI therapy, e. g., oral PPI once-daily (strong recommendation, moderate quality evidence).

MR11. ESGE recommends that epinephrine injection therapy not be used as endoscopic monotherapy. If used, it should be combined with a second endoscopic hemostasis modality (strong recommendation, high quality evidence).

MR12. ESGE recommends PPI therapy for patients who receive endoscopic hemostasis and for patients with adherent clot not receiving endoscopic hemostasis. PPI therapy should be high dose and administered as an intravenous bolus followed by continuous infusion (80 mg then 8 mg/hour) for 72 hours post endoscopy (strong recommendation, high quality evidence).

MR13. ESGE does not recommend routine second-look endoscopy as part of the management of nonvariceal upper gastrointestinal hemorrhage (NVUGIH). However, in patients with clinical evidence of rebleeding following successful initial endoscopic hemostasis, ESGE recommends repeat upper endoscopy with hemostasis if indicated. In the case of failure of this second attempt at hemostasis, transcatheter angiographic embolization (TAE) or surgery should be considered (strong recommendation, high quality evidence).

MR14. In patients with NVUGIH secondary to peptic ulcer, ESGE recommends investigating for the presence of Helicobacter pylori in the acute setting with initiation of appropriate antibiotic therapy when H. pylori is detected. Re-testing for H. pylori should be performed in those patients with a negative test in the acute setting. Documentation of successful H. pylori eradication is recommended (strong recommendation, high quality evidence).

MR15. In patients receiving low dose aspirin for secondary cardiovascular prophylaxis who develop peptic ulcer bleeding, ESGE recommends aspirin be resumed immediately following index endoscopy if the risk of rebleeding is low (e. g., FIIc, FIII). In patients with high risk peptic ulcer (FIa, FIb, FIIa, FIIb), early reintroduction of aspirin by day 3 after index endoscopy is recommended, provided that adequate hemostasis has been established (strong recommendation, moderate quality evidence).


#

Abbreviations

APC: argon plasma coagulation
ASA: American Society of Anesthesiologists
DAPT: dual antiplatelet therapy
CHADS2 : congestive heart failure, hypertension, age ≥ 75 years, diabetes mellitus, and previous stroke or transient ischemic attack [risk score]
CI: confidence interval
DOAC: direct oral anticoagulant
ESGE: European Society of Gastrointestinal Endoscopy
FFP: fresh frozen plasma
GBS: Glasgow-Blatchford Score
GI: gastrointestinal
GRADE: Grading of Recommendations Assessment, Development and Evaluation
HR: hazard ratio
INR: international normalized ratio
NBVV: nonbleeding visible vessel
NNT: number needed to treat
NOAC: non-VKA oral anticoagulant
NVUGIH: nonvariceal upper gastrointestinal hemorrhage
PAR: protease-activated receptor
PCC: prothrombin complex concentrate
PICO: patients, interventions, controls, outcomes
PPI: proton pump inhibitor
OR: odds ratio
PUB: peptic ulcer bleeding
RBC: red blood cell
RCT: randomized controlled trial
RR: relative risk or risk ratio
TAE: transcatheter angiographic embolization
UGIH: upper gastrointestinal hemorrhage
VCE: videocapsule endoscopy
VKA: vitamin K antagonist

Introduction

Acute upper gastrointestinal hemorrhage (UGIH) is a common condition worldwide that has an estimated annual incidence of 40−150 cases per 100 000 population [1] [2], frequently leads to hospital admission, and has significant associated morbidity and mortality, especially in the elderly. The most common causes of acute UGIH are nonvariceal [1] [2]. This includes peptic ulcers, 28 % – 59 % (duodenal ulcer 17 % – 37 % and gastric ulcer 11 % – 24 %); mucosal erosive disease of the esophagus/stomach/duodenum, 1 % – 47 %; Mallory – Weiss syndrome, 4 % – 7 %; upper GI tract malignancy, 2 % – 4 %; other diagnosis, 2 % – 7 %; or no exact cause identified, 7 % – 25 % [1] [2]. Moreover, in 16 % – 20 % of acute UGIH cases, more than one endoscopic diagnosis may be identified as the cause of bleeding. The aim of this evidence-based consensus guideline is to provide medical caregivers with a comprehensive review and recommendations on the clinical and endoscopic management of NVUGIH.


#

Methods

The ESGE commissioned this guideline on NVUGIH and appointed a guideline leader (I.M.G.) who in collaboration with the Chair of the ESGE Guidelines Committee (C.H.), invited the listed authors to participate in the guideline development and review. Key questions were prepared by the coordinating team (I.M.G. and C.H.) and reviewed and approved by all task force members. The coordinating team formed four task force subgroups, each with its own coordinator, and divided the key topics/questions amongst these four task force subgroups (see Appendix e1, online-only). Task force members included gastroenterologists/gastrointestinal endoscopists, an interventional radiologist, and a surgeon. Clinical questions were formulated using the PICO (patients, interventions, controls, outcomes) methodology.

Each task force subgroup performed a systematic literature search to identify the relevant literature that was subsequently used to prepare evidence-based, well-balanced statements on each of their assigned key questions. The Ovid MEDLINE, EMBASE, Google/Google Scholar, and the Cochrane Database of Systematic Reviews were searched for English-language articles including at a minimum the following key words: nonvariceal upper gastrointestinal (GI) hemorrhage/bleeding, peptic ulcer hemorrhage/bleeding, fluid resuscitation, fluid therapy, critical illness, crystalloid solutions, colloid solutions, plasma transfusions, red blood cell transfusion, platelet transfusion, hemoglobin, restrictive transfusion strategy, liberal transfusion strategy, risk stratification, mortality, rebleeding, anti-thrombotic agent, antiplatelet agent, aspirin, dual anti-platelet therapy (DAPT), anti-coagulation/anti-coagulant, direct/new oral anticoagulants (DOACs), coagulopathy, vitamin K inhibitor/antagonist, prokinetic agent, erythromycin, fresh frozen plasma, nasogastric tube, orogastric tube, proton pump inhibitor, prokinetic agent, erythromycin, endoscopic hemostasis, injection therapy, thermal therapy (contact, non-contact), mechanical therapy/endoscopic clipping, topical hemostasis therapy, second-look endoscopy, helicobacter pylori, H. pylori, transcatheter angiographic embolization (TAE), and surgery. The hierarchy of studies included as part of this evidence-based guideline was, in decreasing order of evidence level, published systematic reviews/meta-analyses, randomized controlled trials (RCTs), prospective and retrospective observational studies. All selected articles were graded using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) system [3] [4].

Each task force subgroup proposed statements for each of their assigned key questions which were discussed and voted on during the NVUGIH task force guideline meeting held in Berlin, Germany in November 2014. In August 2015, a manuscript draft prepared by I.M.G. was sent to all task force members. After agreement on a final version, the manuscript was reviewed by two members of the ESGE Governing Board and sent for further comments to the National Societies and ESGE individual members. After agreement on a final version, the manuscript was submitted to the journal Endoscopy for publication. All authors agreed on the final revised manuscript.

This NVUGIH guideline will be considered for review and updating in 2020, or sooner if new relevant evidence becomes available. Any updates to this guideline in the interim will be noted on the ESGE website: http://www.esge.com/esge-guidelines.html.


#

Statements and recommendations

See [Table 1].

Table 1

Summary of Guideline statements and recommendations. Diagnosis and management of nonvariceal upper gastrointestinal hemorrhage: European Society of Gastrointestinal Endoscopy (ESGE) Guideline.

Initial patient evaluation and hemodynamic resuscitation

1 ESGE recommends immediate assessment of hemodynamic status in patients who present with acute upper gastrointestinal hemorrhage (UGIH), with prompt intravascular volume replacement initially using crystalloid fluids if hemodynamic instability exists (strong recommendation, moderate quality evidence).

2 ESGE recommends a restrictive red blood cell transfusion strategy that aims for a target hemoglobin between 7 g/dL and 9 g/dL. A higher target hemoglobin should be considered in patients with significant co-morbidity (e. g., ischemic cardiovascular disease) (strong recommendation, moderate quality evidence).

Risk stratification

3 ESGE recommends the use of a validated risk stratification tool to stratify patients into high and low risk groups. Risk stratification can aid clinical decision making regarding timing of endoscopy and hospital discharge (strong recommendation, moderate quality evidence).

4 ESGE recommends the use of the Glasgow-Blatchford Score (GBS) for pre-endoscopy risk stratification. Outpatients determined to be at very low risk, based upon a GBS score of 0 – 1, do not require early endoscopy nor hospital admission. Discharged patients should be informed of the risk of recurrent bleeding and be advised to maintain contact with the discharging hospital (strong recommendation, moderate quality evidence).

Pre-endoscopy management

5 For patients taking vitamin K antagonists (VKAs), ESGE recommends withholding the VKA and correcting coagulopathy while taking into account the patient's cardiovascular risk in consultation with a cardiologist. In patients with hemodynamic instability, administration of vitamin K, supplemented with intravenous prothrombin complex concentrate (PCC) or fresh frozen plasma (FFP) if PCC is unavailable, is recommended (strong recommendation, low quality evidence).

6 If the clinical situation allows, ESGE suggests an international normalized ratio (INR) value < 2.5 before performing endoscopy with or without endoscopic hemostasis (weak recommendation, moderate quality evidence).

7 ESGE recommends temporarily withholding new direct oral anticoagulants (DOACs) in patients with suspected acute NVUGIH in coordination/consultation with the local hematologist/cardiologist (strong recommendation, very low quality evidence).

8 For patients using antiplatelet agents, ESGE recommends the management algorithm detailed in [Fig. 2] (strong recommendation, moderate quality evidence).

9 ESGE recommends initiating high dose intravenous proton pump inhibitors (PPI), intravenous bolus followed by continuous infusion (80 mg then 8 mg/hour), in patients presenting with acute UGIH awaiting upper endoscopy. However, PPI infusion should not delay the performance of early endoscopy (strong recommendation, high quality evidence).

10 ESGE does not recommend the use of tranexamic acid in patients with NVUGIH (strong recommendation, low quality evidence).

11 ESGE does not recommend the use of somatostatin, or its analogue octreotide, in patients with NVUGIH (strong recommendation, low quality evidence).

12 ESGE recommends intravenous erythromycin (single dose, 250 mg given 30 – 120 minutes prior to upper GI endoscopy) in patients with clinically severe or ongoing active UGIH. In selected patients, pre-endoscopic infusion of erythromycin significantly improves endoscopic visualization, reduces the need for second-look endoscopy, decreases the number of units of blood transfused, and reduces duration of hospital stay (strong recommendation, high quality evidence).

13 ESGE does not recommend the routine use of nasogastric or orogastric aspiration/lavage in patients presenting with acute UGIH (strong recommendation, moderate quality evidence).

14  In an effort to protect the patient's airway from potential aspiration of gastric contents, ESGE suggests endotracheal intubation prior to endoscopy in patients with ongoing active hematemesis, encephalopathy, or agitation (weak recommendation, low quality evidence).

15 ESGE recommends adopting the following definitions regarding the timing of upper GI endoscopy in acute overt UGIH relative to patient presentation: very early < 12 hours, early ≤ 24 hours, and delayed > 24 hours (strong recommendation, moderate quality evidence).

16 Following hemodynamic resuscitation, ESGE recommends early ( ≤ 24 hours) upper GI endoscopy. Very early ( < 12 hours) upper GI endoscopy may be considered in patients with high risk clinical features, namely: hemodynamic instability (tachycardia, hypotension) that persists despite ongoing attempts at volume resuscitation; in-hospital bloody emesis/nasogastric aspirate; or contraindication to the interruption of anticoagulation (strong recommendation, moderate quality evidence).

17 ESGE recommends the availability of both an on-call GI endoscopist proficient in endoscopic hemostasis and on-call nursing staff with technical expertise in the use of endoscopic devices to allow performance of endoscopy on a 24 /7 basis (strong recommendation, moderate quality evidence).

Endoscopic therapy (peptic ulcer bleeding)

18 ESGE recommends the Forrest (F) classification be used in all patients with peptic ulcer hemorrhage in order to differentiate low and high risk endoscopic stigmata (strong recommendation, high quality evidence).

19 ESGE recommends that peptic ulcers with spurting or oozing bleeding (Forrest classification Ia and Ib respectively), or with a nonbleeding visible vessel (Forrest classification IIa) receive endoscopic hemostasis because these lesions are at high risk for persistent bleeding or rebleeding (strong recommendation, high quality evidence).

20 ESGE recommends that peptic ulcers with an adherent clot (Forrest classification IIb) be considered for endoscopic clot removal. Once the clot is removed, any identified underlying active bleeding (Forrest classification Ia or Ib) or nonbleeding visible vessel (Forrest classification IIa) should receive endoscopic hemostasis (weak recommendation, moderate quality evidence).

21 In patients with peptic ulcers having a flat pigmented spot (Forrest classification IIc) or clean base (Forrest classification III), ESGE does not recommend endoscopic hemostasis as these stigmata present a low risk of recurrent bleeding. In selected clinical settings, these patients may be discharged to home on standard PPI therapy, e. g., oral PPI once-daily (strong recommendation, moderate quality evidence).

22 ESGE does not recommend the routine use of Doppler ultrasound or magnification endoscopy in the evaluation of endoscopic stigmata of peptic ulcer bleeding (strong recommendation, low quality evidence).

23 For patients with actively bleeding ulcers (FIa, FIb), ESGE recommends combining epinephrine injection with a second hemostasis modality (contact thermal, mechanical therapy, or injection of a sclerosing agent). ESGE recommends that epinephrine injection therapy not be used as endoscopic monotherapy (strong recommendation, high quality evidence).

24 For patients with nonbleeding visible vessel (FIIa), ESGE recommends mechanical therapy, thermal therapy, or injection of a sclerosing agent as monotherapy or in combination with epinephrine injection. ESGE recommends that epinephrine injection therapy not be used as endoscopic monotherapy (strong recommendation, high quality evidence).

25 For patients with active NVUGIH bleeding not controlled by standard endoscopic hemostasis therapies, ESGE suggests the use of a topical hemostatic spray or over-the-scope clip as salvage endoscopic therapy (weak recommendation, low quality evidence).

Endoscopic therapy (other causes of NVUGIH)

26 For patients with acid-related causes of NVUGIH different from peptic ulcers (e. g., erosive esophagitis, gastritis, duodenitis), ESGE recommends treatment with high dose PPI. Endoscopic hemostasis is usually not required and selected patients may be discharged early (strong recommendation, low quality evidence).

27 ESGE recommends that patients with a Mallory – Weiss lesion that is actively bleeding receive endoscopic hemostasis. There is currently inadequate evidence to recommend a specific endoscopic hemostasis modality. Patients with a Mallory – Weiss lesion and no active bleeding can receive high dose PPI therapy alone (strong recommendation, moderate quality evidence).

28 ESGE recommends that a Dieulafoy lesion receive endoscopic hemostasis using thermal, mechanical (hemoclip or band ligation), or combination therapy (dilute epinephrine injection combined with contact thermal or mechanical therapy) (strong recommendation, moderate quality evidence). Transcatheter angiographic embolization (TAE) or surgery should be considered if endoscopic treatment fails or is not technically feasible (strong recommendation, low quality evidence).

29 In patients bleeding from upper GI angioectasias, ESGE recommends endoscopic hemostasis therapy. However, there is currently inadequate evidence to recommend a specific endoscopic hemostasis modality (strong recommendation, low quality evidence).

30 In patients bleeding from upper GI neoplasia, ESGE recommends considering endoscopic hemostasis in order to avert urgent surgery and reduce blood transfusion requirements. However, no currently available endoscopic treatment appears to have long-term efficacy (weak recommendation, low quality evidence).

Post endoscopy/endoscopic hemostasis management

31 ESGE recommends PPI therapy for patients who receive endoscopic hemostasis and for patients with adherent clot not receiving endoscopic hemostasis. PPI therapy should be high dose and administered as an intravenous bolus followed by continuous infusion (80 mg then 8 mg /hour) for 72 hours post endoscopy (strong recommendation, high quality evidence).

32 ESGE suggests considering PPI therapy as intermittent intravenous bolus dosing (at least twice-daily) for 72 hours post endoscopy for patients who receive endoscopic hemostasis and for patients with adherent clot not receiving endoscopic hemostasis. If the patient’s condition permits, high dose oral PPI may also be an option in those able to tolerate oral medications (weak recommendation, moderate quality evidence).

33 In patients with clinical evidence of rebleeding following successful initial endoscopic hemostasis, ESGE recommends repeat upper endoscopy with hemostasis if indicated. In the case of failure of this second attempt at hemostasis, transcatheter angiographic embolization (TAE) or surgery should be considered (strong recommendation, high quality evidence).

34 ESGE does not recommend routine second-look endoscopy as part of the management of NVUGIH. However, second-look endoscopy may be considered in selected patients at high risk for rebleeding (strong recommendation, high quality evidence).

35 In patients with NVUGIH secondary to peptic ulcer, ESGE recommends investigating for the presence of Helicobacter pylori in the acute setting with initiation of appropriate antibiotic therapy when H. pylori is detected. Re-testing for H. pylori should be performed in those patients with a negative test in the acute setting. Documentation of successful H. pylori eradication is recommended (strong recommendation, high quality evidence).

36 ESGE recommends restarting anticoagulant therapy following NVUGIH in patients with an indication for long-term anticoagulation. The timing for resumption of anticoagulation should be assessed on a patient by patient basis. Resuming warfarin between 7 and 15 days following the bleeding event appears safe and effective in preventing thromboembolic complications for most patients. Earlier resumption, within the first 7 days, may be indicated for patients at high thrombotic risk (strong recommendation, moderate quality evidence).

37 In patients receiving low dose aspirin for primary cardiovascular prophylaxis who develop peptic ulcer bleeding, ESGE recommends withholding aspirin, re-evaluating the risks/benefits of ongoing aspirin use in consultation with a cardiologist, and resuming low dose aspirin following ulcer healing or earlier if clinically indicated (strong recommendation, low quality evidence).

38 In patients receiving low dose aspirin for secondary cardiovascular prophylaxis who develop peptic ulcer bleeding, ESGE recommends aspirin be resumed immediately following index endoscopy if the risk of rebleeding is low (e. g., FIIc, FIII). In patients with high risk peptic ulcer (FIa, FIb, FIIa, FIIb), early reintroduction of aspirin by day 3 after index endoscopy is recommended, provided that adequate hemostasis has been established (strong recommendation, moderate quality evidence).

39 In patients receiving dual antiplatelet therapy (DAPT) who develop peptic ulcer bleeding, ESGE recommends continuing low dose aspirin therapy. Early cardiology consultation should be obtained regarding the timing of resuming the second antiplatelet agent (strong recommendation, low quality evidence).

40 In patients requiring dual antiplatelet therapy (DAPT) and who have had NVUGIH, ESGE recommends the use of a PPI as co-therapy (strong recommendation, moderate quality evidence).

Initial patient evaluation and hemodynamic resuscitation

ESGE recommends immediate assessment of hemodynamic status in patients who present with acute upper gastrointestinal hemorrhage (UGIH), with prompt intravascular volume replacement initially using crystalloid fluids if hemodynamic instability exists (strong recommendation, moderate quality evidence).

The goals of hemodynamic resuscitation are to correct intravascular hypovolemia, restore adequate tissue perfusion, and prevent multi-organ failure. Early intensive hemodynamic resuscitation of patients with acute UGIH has been shown to significantly decrease mortality [5]. In an observational study of patients with acute UGIH and hemodynamic instability, patients who received intensive hemodynamic resuscitation had significantly fewer myocardial infarctions and lower mortality compared with those in the “observation group” (P = 0.04 for both comparisons). However, there is no evidence from randomized controlled trials (RCTs), for or against early or large-volume intravenous fluid administration in uncontrolled hemorrhage [6] [7]. Moreover, the selection of resuscitation fluid type in critically ill patients requires careful consideration based on safety, effects on patient outcomes, and costs. To date, there is ongoing uncertainty regarding the ideal fluid administration strategy in this clinical setting [8] [9].

ESGE recommends a restrictive red blood cell transfusion strategy that aims for a target hemoglobin between 7 g/dL and 9 g/dL. A higher target hemoglobin should be considered in patients with significant co-morbidity (e. g., ischemic cardiovascular disease) (strong recommendation, moderate quality evidence).

The use of red blood cell (RBC) transfusions may be lifesaving following massive UGIH. However, the role of RBC transfusion in less torrential GI bleeding remains controversial, with uncertainty existing regarding the hemoglobin level at which blood transfusion should be initiated. This uncertainty reflects concerns from both the critical care and gastroenterology literature suggesting poorer outcomes in patients managed with a liberal RBC transfusion strategy [2] [10] [11]. In a recent RCT that included 921 patients presenting with all causes of acute UGIH, a restrictive RBC transfusion strategy (target hemoglobin, 7 to 9 g/dL) was compared with a more liberal transfusion strategy (target hemoglobin, 9 to 11 g/dL) [12]. The restrictive RBC transfusion group had significantly improved 6-week survival (95 % vs. 91 %; hazard ratio [HR] 0.55, 95 % confidence interval [CI] 0.33 – 0.92) and reduced rebleeding (10 % vs.16 %; HR 0.68, 95 %CI 0.47 – 0.98) [12]. In the subgroup of patients with NVUGIH (n = 699), there was a statistical trend towards lower mortality in the restrictive vs. liberal RBC transfusion strategy (3.7 % vs. 6.9 %, P = 0.065). Because the study was not powered to specifically evaluate NVUGIH, these findings should be interpreted with caution. Other limitations of this study include the exclusion of patients with massive exsanguinating bleeding and defined co-morbidities. Furthermore, all patients underwent endoscopy within 6 hours of presentation, which may not be feasible in everyday clinical practice.

Coagulopathy at the time of NVUGIH presentation is another frequent and adverse prognostic factor [13]. Published data for the management of coagulopathy are limited and inconclusive. One small cohort study using an historical comparison group showed that aggressive volume resuscitation, including correction of coagulopathy (international normalized ratio [INR] < 1.8), led to an improvement in mortality outcomes [5]. In a systematic review that evaluated the relevance of initial INR before correction in patients with NVUGIH, INR did not appear to predict rebleeding, yet after adjusting for potential confounders, an initial INR > 1.5 predicted mortality (odds ratio [OR] 1.96, 95 %CI 1.13 – 3.41) [14]. This may in part reflect the presence of underlying liver disease. There is however no available evidence to help guide coagulopathy correction in critically ill patients and wide variation in management exists in this area, indicating clinical uncertainty regarding optimal practice [15]. Platelet count has not been shown to be a predictor of either rebleeding or mortality. Currently, there is no high quality evidence to guide platelet transfusion thresholds, although a platelet transfusion threshold of 50 × 109/L has been proposed for most patients, with a target of 10 × 109/L for patients in whom platelet dysfunction is suspected [16].


#

Risk stratification

ESGE recommends the use of a validated risk stratification tool to stratify patients into high and low risk groups. Risk stratification can aid clinical decision making regarding timing of endoscopy and hospital discharge (strong recommendation, moderate quality evidence).

ESGE recommends the use of the Glasgow-Blatchford Score (GBS) for pre-endoscopy risk stratification. Outpatients determined to be at very low risk, based upon a GBS score of 0 – 1, do not require early endoscopy nor hospital admission. Discharged patients should be informed of the risk of recurrent bleeding and be advised to maintain contact with the discharging hospital (strong recommendation, moderate quality evidence).

Risk stratification of patients presenting with acute UGIH can assist in identifying those who may require more urgent intervention and help triage patients to in-hospital vs. out-of-hospital management. A number of scoring tools have been created for predicting outcomes following acute UGIH, with the Glasgow-Blatchford Score (GBS) ([Table 2]) and Rockall score being the most widely evaluated and adopted [17] [18] [19]. However, no single scoring tool has been shown to excel at predicting all relevant outcomes in acute UGIH (e. g., rebleeding, need for intervention, mortality) [19]. This is not surprising as the most validated risk scores were derived to assess a specific UGIH outcome: that for the Rockall score being mortality and for the GBS being the need for intervention [17] [18].

Table 2

Glasgow-Blatchford Score (GBS).

Points

Systolic blood pressure, mmHg

100 – 109

1

90 – 99

2

 < 90

3

Blood urea nitrogen, mmol/L

6.5 – 7.9

2

8.0 – 9.9

3

10.0 – 24.9

4

 ≥ 25.0

6

Hemoglobin for men, g/dL

12.0 – 12.9

1

10.0 – 11.9

3

 < 10.0

6

Hemoglobin for women, g/dL

10.0 – 11.9

1

 < 10.0

6

Other risk variables

Pulse ≥ 100

1

Melena

1

Syncope

2

Hepatic disease

2

Cardiac failure

2

TOTAL GBS __________________
GBS restricted for use only in nonhospitalized, ambulatory patients
Risk variables measured at time of patient presentation
GBS = 0 – 1 denotes “low-risk”

A recent systematic review evaluating the accuracy of the available UGIH risk stratification tools demonstrated substantial heterogeneity in predicted outcomes and highlighted that methodological quality of the prediction scores was less than optimal [19]. Regarding the need for intervention, retrospective and prospective studies have assessed the prognostic value of the GBS vs. the Rockall score. These studies showed that the GBS correctly identified 98 % (95 %CI 89 % – 100 %) of those patients who did not require any subsequent intervention while 83 % (95 %CI 71 % – 91 %) of those patients were identified using the Rockall score. Randomized controlled trials and observational studies consistently indicate that clinical, endoscopic, and social factors may identify patients who may be safely discharged for outpatient management [20] [21] [22] [23] [24] [25] [26] [27] [28]. The most frequent adverse event reported is rebleeding ranging between 0.5 % and 4 %, with no deaths or hospital readmissions for surgery reported. Moreover, studies consistently indicate that outpatient management of appropriately selected patients with acute UGIH reduces resource utilization [20] [21] [27]. Emergency department discharge without inpatient endoscopy (i. e., outpatient management) should be considered for patients if: systolic blood pressure ≥ 110 mmHg, pulse < 100 beats/minute, hemoglobin ≥ 13.0 g/dL for men or ≥ 12.0 g/dL for women, blood urea nitrogen < 18.2 mg/dL, along with the absence of melena, syncope, hepatic disease, and cardiac failure [18]. (See Appendix e2, online-only.)


#

Pre-endoscopy management

Initial management of antithrombotic agents (anticoagulants and antiplatelet agents)

For patients taking vitamin K antagonists (VKAs), ESGE recommends withholding the VKA and correcting coagulopathy while taking into account the patient’s cardiovascular risk in consultation with a cardiologist. In patients with hemodynamic instability, administration of vitamin K, supplemented with intravenous prothrombin complex concentrate (PCC) or fresh frozen plasma (FFP) if PCC is unavailable, is recommended (strong recommendation, low quality evidence).

If the clinical situation allows, ESGE suggests an international normalized ratio (INR) value < 2.5 before performing endoscopy with or without endoscopic hemostasis (weak recommendation, moderate quality evidence).

GI bleeding represents a serious complication of VKA therapy, with an incidence of 1 % – 4 % per year [29] [30]. Discontinuation of anticoagulants and correction of coagulopathy before endoscopy is the “standard of practice” in patients with clinically significant GI bleeding [31] [32] [33]. Because data are limited, specific strategies to reverse VKAs in a patient with acute overt UGIH vary [34]. Practice guidelines recommend urgent reversal in all patients presenting with serious, life-threatening bleeding (i. e., hemodynamic instability or shock), either in the case of therapeutic or supratherapeutic INR elevations [32] [35]. For patients who are not actively bleeding and are hemodynamically stable, intravenous vitamin K administration may be an option. When more urgent reversal is required, administration of prothrombin complex concentrates (PCCs) or fresh frozen plasma (FFP) is necessary, with concomitant intravenous administration of 5 – 10 mg vitamin K to prevent “rebound coagulopathy” once the transfused factors have been cleared. Prothrombin complex concentrates contain clotting factors prepared from pooled and concentrated human plasma and are preferred over FFP because of several advantages, including no need to check the patient’s blood group, less risk for volume overload because of smaller transfusion volume, faster onset of action, similar thrombotic risk profile, and minimal risk of infectious transmission, albeit at a higher cost [36] [37] [38] [39] [40]. A recent prospective, nonrandomized, comparative study of 40 warfarin users who presented with UGIH and an INR > 2.1 reported that patients who received PCC had a near normalized INR at 2 hours following infusion (INR = 1.5) while those who received FFP had an INR of 2.4 at 6 hours following infusion [38]. No patient in the PCC group had active bleeding at endoscopy compared with 7 in the FFP group (0 vs. 35 %, P < 0.01). The risk of thrombosis following PCC administration approximates 1 %, and is similar to that reported with FFP [39] [40].

ESGE recommends temporarily withholding new direct oral anticoagulants (DOACs) in patients with suspected acute NVUGIH in coordination/consultation with the local hematologist/cardiologist (strong recommendation, very low quality evidence).

As an alternative to heparin and VKAs, the new non-VKA oral anticoagulants (NOACs; also referred to as direct oral anticoagulants [DOACs]) are being rapidly adopted worldwide, primarily for thromboembolic prevention in patients with nonvalvular atrial fibrillation and for prophylaxis or treatment of venous thromboembolism [41]. These pharmacological agents do however, present a risk of significant GI bleeding similar to or greater than that reported with warfarin [42] [43]. Moreover, DOACs differ in comparison with heparin and VKA. Specifically, in the absence of renal or hepatic failure, DOAC clearance and the subsequent loss of anticoagulation effect is rapid and predictable (occurring gradually over 12 – 24 hours), routine laboratory tests are not sensitive for the quantitative assessment of their anticoagulant activity, and there is currently no specific reversal agent/antidote for emergency use with any DOAC, although potential agents are in development and may be commercially available in the next 1 – 2 years [44] [45] [46]. As there are no published clinical trials addressing the management of GI bleeding in patients using DOAC, current recommendations are based on expert opinion or laboratory end-points [47] [48] [49].

At the time of patient presentation with acute UGIH, DOACs should be temporarily withheld. Given their relatively short half-life, time is the most important antidote against DOACs. Strategies to accelerate anticoagulation reversal are supported only by data collected from healthy human volunteers, animal models, and in vitro studies [50]. Based on those data, vitamin K or FFP have no place as reversal agents for DOACs. Prothrombin complex concentrates or activated PCC may be considered in patients with severe or life-threatening bleeding, and hemodialysis can be used to reduce the blood concentration of dabigatran, but not that of rivaroxaban and apixaban which are more tightly bound to plasma proteins [48] [49] [51]. Additional data on the clinical effectiveness of these strategies in acutely bleeding patients are urgently needed.

For patients using antiplatelet agents, ESGE recommends the management algorithm detailed in [Fig. 1] (strong recommendation, moderate quality evidence).

Zoom Image
Fig. 1 Algorithm for the management of patients with acute upper gastrointestinal hemorrhage who are using antiplatelet agent(s): European Society of Gastrointestinal Endoscopy (ESGE) Guideline.

Antiplatelet agents include low dose aspirin and thienopyridines (e. g., clopidogrel, prasugrel, ticlopidine) that irreversibly inhibit platelet aggregation, ticagrelor a reversible P2Y12 receptor antagonist, and vorapaxar, a protease-activated receptor (PAR-1) antagonist that inhibits thrombin. The minimum duration of antiplatelet agent discontinuation that allows for restoration of normal platelet aggregation is 5 – 7 days [52].

Studies have shown that in patients taking low dose aspirin for secondary cardiovascular prophylaxis, all-cause mortality was lower if aspirin was not discontinued following peptic ulcer bleeding [53] [54]. In an RCT, 156 recipients of low dose aspirin for secondary prophylaxis who had peptic ulcer bleeding were randomized to receive continuous aspirin or placebo [53]. At 8-week follow up, all-cause mortality was lower in the patients randomized to aspirin compared with placebo (1.3 % vs. 12.9 %, 95 %CI 3.7 % – 19.5 %; hazard ratio [HR] 0.20), with the difference being attributable to cardiovascular, cerebrovascular, or GI complications. The 30-day ulcer rebleeding rate was not significantly greater in the aspirin group. Patients who required dual antiplatelet therapy (DAPT) were excluded from this study. In a subsequent retrospective analysis that included 118 low dose aspirin recipients who had been treated for peptic ulcer bleeding and followed-up for a median of 2 years, 47 (40 %) patients stopped aspirin [54]. Patients who discontinued aspirin and those who continued aspirin had similar mortality rates (31 %). However, in a subgroup analysis limited to patients with cardiovascular co-morbidities, those patients who discontinued aspirin had an almost fourfold increase in the risk of death or acute cardiovascular event (P < 0.01) [54]. Randomized controlled trials have shown that neither aspirin nor clopidogrel use impede ulcer healing promoted by proton pump inhibitors (PPI) [55] [56].


#

Pharmacological therapy

ESGE recommends initiating high dose intravenous proton pump inhibitors (PPI), intravenous bolus followed by continuous infusion (80 mg then 8 mg/hour), in patients presenting with acute UGIH awaiting upper endoscopy. However, PPI infusion should not delay the performance of early endoscopy (strong recommendation, high quality evidence).

A Cochrane meta-analysis of 6 RCTs (n = 2223 patients) showed that administering PPIs before endoscopy significantly decreases the incidence of high risk stigmata of hemorrhage at the time of index endoscopy (37.2 % vs. 46.5 %; OR 0.67, 95 %CI 0.54 – 0.84) and the need for endoscopic hemostasis (8.6 % vs. 11.7 %; OR 0.68, 95 %CI 0.50 – 0.93), but has no effect on rebleeding, need for surgery, or mortality [57].

Cost – effectiveness studies suggest that high dose PPI infusion prior to endoscopy for patients with UGIH is more effective and less costly than placebo [58] [59]. (See Appendix e3, online-only.)

ESGE does not recommend the use of tranexamic acid in patients with NVUGIH (strong recommendation, low quality evidence).

Tranexamic acid reduces clot breakdown by inhibiting the fibrinolytic action of plasmin. A recent RCT demonstrated that tranexamic acid significantly reduces bleeding-related and all-cause mortality in trauma patients with significant hemorrhage [60]. A Cochrane meta-analysis evaluating the use of tranexamic acid in 1654 UGIH patients showed a beneficial effect of tranexamic acid on mortality when compared with placebo (relative risk [RR] 0.61, 95 %CI 0.42 – 0.89), but not on other patient outcomes including bleeding, surgery, or transfusion requirements [61]. However, the beneficial effect on mortality did not persist in subgroup analysis. The studies included in this meta-analysis have important limitations that affect their generalizability including their methodological quality and the fact that the majority were conducted before the widespread use of therapeutic endoscopy and PPIs. To date, no controlled trial assessing the role of alternative antifibrinolytic agents (e. g., aminocaproic acid, aprotinin) in patients with acute UGIH has been reported. (See Appendix e4, online-only.)

ESGE does not recommend the use of somatostatin, or its analogue octreotide, in patients with NVUGIH (strong recommendation, low quality evidence).

Somatostatin, and its analogue octreotide, inhibit both acid and pepsin secretion while also reducing gastroduodenal mucosal blood flow [62]. However, they are not routinely recommended in NVUGIH (e. g., peptic ulcer bleeding), either pre-endoscopy or as an adjunctive therapy post endoscopy, since published data show little or no benefit attributable to these pharmacological agents. (See Appendix e5, online-only.)

ESGE recommends intravenous erythromycin (single dose, 250 mg given 30 – 120 minutes prior to upper GI endoscopy) in patients with clinically severe or ongoing active UGIH. In selected patients, pre-endoscopic infusion of erythromycin significantly improves endoscopic visualization, reduces the need for second-look endoscopy, decreases the number of units of blood transfused, and reduces duration of hospital stay (strong recommendation, high quality evidence).

It has been reported that in 3 % to 19 % of UGIH cases, no obvious cause of bleeding is identified [63] [64]. This may in part be related to the presence of blood and clots impairing endoscopic visualization. There are four published meta-analyses evaluating the role of prokinetic agent infusion prior to upper GI endoscopy in patients presenting with acute UGIH [65] [66] [67] [68]. The most recently published meta-analysis (n = 558 patients) showed that erythromycin infusion prior to endoscopy significantly improved gastric mucosa visualization (OR 3.43, 95 %CI 1.81 – 6.50; P < 0.01), and decreased the need for second-look endoscopy (OR 0.47, 95 %CI 0.26−0.83, P = 0.01), RBC units transfused (weighted mean difference −0.41, 95 %CI −0.82 to −0.01, P = 0.04), and duration of hospital stay (weighted mean difference −1.51 days, 95 %CI −2.45 to −0.56, P < 0.01) [68].

A single intravenous dose of erythromycin is safe and generally well tolerated, with no adverse events reported in the meta-analyses. Studies that found a significant improvement in endoscopic visualization with pre-endoscopic erythromycin infusion included patients admitted to the intensive care unit because of UGIH with clinical evidence of active bleeding or hematemesis or blood seen on nasogastric lavage. These patients are most likely to benefit from erythromycin infusion prior to endoscopy. The dose of erythromycin most commonly used is 250 mg and is infused 30 to 120 minutes prior to upper GI endoscopy. A cost – effectiveness study found that pre-endoscopy erythromycin infusion in UGIH was cost-effective, primarily due to a reduction in the need for second-look endoscopies [69]. Contraindications to erythromycin administration include sensitivity to macrolide antibiotics and prolonged QT interval.

Metoclopramide has been less studied, it has been assigned a “black box warning” by the United States Food and Drug Administration because of the risk of neurologic side effects, and caution should therefore be advised with the use of this prokinetic agent.

(See Appendix e6, online-only.)


#

Role of gastric lavage and prophylactic endotracheal intubation

ESGE does not recommend the routine use of nasogastric or orogastric aspiration/lavage in patients presenting with acute UGIH (strong recommendation, moderate quality evidence).

A number of studies, including a meta-analysis, have evaluated the role of nasogastric aspiration/lavage in patients presenting with acute UGIH [70] [71] [72] [73]. In distinguishing upper from lower GI bleeding, nasogastric aspiration has low sensitivity 44 % (95 %CI 39 % – 48 %) yet high specificity 95 % (95 %CI 90 % – 98 %). In identifying severe UGIH, its sensitivity and specificity are 77 % (95 %CI 57 % – 90 %) and 76 % (95 %CI 32 % – 95 %), respectively [70]. This meta-analysis also found that as compared to nasogastric aspiration/lavage, clinical signs and laboratory findings (e. g., hemodynamic shock and hemoglobin < 8 g/dL) had similar ability to identify severe UGIH [70]. Others have reported that nasogastric aspiration/lavage failed to assist clinicians in correctly predicting the need for endoscopic hemostasis, did not improve visualization of the stomach at endoscopy, or improve clinically relevant outcomes such as rebleeding, need for second-look endoscopy, or blood transfusion requirements [71] [72] [73]. It also should be noted that nasogastric aspiration/lavage is a very uncomfortable procedure that is not well tolerated or desired by patients [74].

In an effort to protect the patient’s airway from potential aspiration of gastric contents, ESGE suggests endotracheal intubation prior to endoscopy in patients with ongoing active hematemesis, encephalopathy, or agitation (weak recommendation, low quality evidence).

It has been hypothesized that pre-endoscopic endotracheal intubation may prevent cardiorespiratory adverse events in patients with acute UGIH. However, between those patients who were prophylactically intubated prior to upper GI endoscopy as compared to those patients not intubated, published data show no significant difference in patient outcomes (e. g., pulmonary aspiration, in-hospital mortality) [75] [76] [77]. One study suggested that aspiration was actually more frequent in those patients who had undergone endotracheal intubation prior to upper GI endoscopy [75]. At this time, endotracheal intubation prior to upper GI endoscopy in patients with UGIH does not seem to make a difference in patient outcome but published data are limited with small numbers of subjects and low methodological quality.


#

Timing of endoscopy

ESGE recommends adopting the following definitions regarding the timing of upper GI endoscopy in acute overt UGIH relative to patient presentation: very early < 12 hours, early ≤ 24 hours, and delayed > 24 hours (strong recommendation, moderate quality evidence).

Following hemodynamic resuscitation, ESGE recommends early ( ≤ 24 hours) upper GI endoscopy. Very early ( < 12 hours) upper GI endoscopy may be considered in patients with high risk clinical features, namely: hemodynamic instability (tachycardia, hypotension) that persists despite ongoing attempts at volume resuscitation; in-hospital bloody emesis/nasogastric aspirate; or contraindication to the interruption of anticoagulation (strong recommendation, moderate quality evidence).

ESGE recommends the availability of both an on-call GI endoscopist proficient in endoscopic hemostasis and on-call nursing staff with technical expertise in the use of endoscopic devices to allow performance of endoscopy on a 24 /7 basis (strong recommendation, moderate quality evidence).

Performance of upper GI endoscopy within 24 hours of patient presentation with suspected NVUGIH and no contraindication to endoscopy has been proposed as a key quality indicator in the management of upper GI bleeding [78]. In a large European observational study that included 123 centers in 7 countries, there was wide variation in practice where anywhere from 70 % to 93 % of 2660 unselected patients with UGIH underwent upper endoscopy within 24 hours of hospital admission [79].

Two systematic reviews evaluating the timing of upper GI endoscopy demonstrated improved risk assessment and reduction in hospital length of stay if endoscopy was performed within 24 hours of patient presentation, yet the impact on need for surgery and in-hospital mortality was variable [80] [81]. More recently, a retrospective analysis of risk factors for mortality in more than 400 000 patients with NVUGIH found an increased mortality in patients who failed to receive upper endoscopy within 1 day of hospital admission (OR 1.32, 95 %CI 1.26 – 1.38) [82]. (See Appendix e7, online-only.)

With respect to very early upper GI endoscopy, an RCT that included 325 patients with peptic ulcer bleeding showed that upper GI endoscopy performed within 12 hours of admission (as compared with 12 – 24 hours) resulted in a significant reduction in transfusion requirements in patients with bloody nasogastric lavage (P < 0.001). No such reduction was observed in patients with “coffee grounds” or clear lavage [83]. A retrospective analysis that included 934 UGIH patients showed that in the subset of patients having a GBS ≥ 12 (n = 97, 10.4 %), the time lapse between presentation to endoscopy was the lone independent risk factor associated with all-cause in-hospital mortality [84]. In this study, a cutoff time of 13 hours in delay to endoscopy best discriminated between patient survival and nonsurvival.

In patients who are hemodynamically stable and without serious co-morbidities, RCTs have shown that performing endoscopy without hospital admission facilitates discharge in up to 46 % of patients and reduces costs/resource utilization [20] [85]. Discharging low risk suspected NVUGIH patients (GBS = 0) directly from the emergency department without undergoing upper GI endoscopy has been proposed as a safe and cost-saving option in multiple studies in various clinical settings [18] [86] [87] [88] [89]. Some investigators have suggested that using a GBS ≤ 1 (see [Table 2]) could double the number of patients eligible for ambulatory management while maintaining safety [89].

There are four published studies, one RCT and three prospective case series, that have evaluated the test characteristics and accuracy parameters of video capsule endoscopy (VCE) in risk stratification of patients presenting with acute UGIH [90] [91] [92] [93]. The overall sensitivity, specificity, positive predictive value, and negative predictive value of VCE for detecting blood in the upper GI tract in patients suspected of acute UGIH are 75 %, 76 %, 67 %, and 82 % respectively. Because the data are limited, at this time there is no role for VCE in the emergency department setting in evaluating acute upper GIH. However, additional studies are needed to further assess VCE in this patient population since, for low to moderate risk UGIH patients, VCE may be a cost-effective modality if post-VCE low risk patients are discharged directly home from the emergency department and hospital admission is avoided [94] [95].


#
#

Endoscopic management

Endoscopic diagnosis

ESGE recommends the Forrest (F) classification be used in all patients with peptic ulcer hemorrhage in order to differentiate low and high risk endoscopic stigmata (strong recommendation, high quality evidence).

ESGE recommends that peptic ulcers with spurting or oozing bleeding (Forrest classification Ia and Ib, respectively) or with a nonbleeding visible vessel (Forrest classification IIa) receive endoscopic hemostasis because these lesions are at high risk for persistent bleeding or rebleeding (strong recommendation, high quality evidence).

ESGE recommends that peptic ulcers with an adherent clot (Forrest classification IIb) be considered for endoscopic clot removal. Once the clot is removed, any identified underlying active bleeding (Forrest classification Ia or Ib) or nonbleeding visible vessel (Forrest classification IIa) should receive endoscopic hemostasis (weak recommendation, moderate quality evidence).

In patients with peptic ulcers having a flat pigmented spot (Forrest classification IIc) or clean base (Forrest classification III), ESGE does not recommend endoscopic hemostasis as these stigmata present a low risk of recurrent bleeding. In selected clinical settings, these patients may be discharged to home on standard PPI therapy, e. g., oral PPI once-daily (strong recommendation, moderate quality evidence).

The Forrest (F) classification was developed more than 40 years ago in an attempt to standardize the characterization of peptic ulcers [96]. The Forrest classification is defined as follows: FIa spurting hemorrhage, FIb oozing hemorrhage, FIIa nonbleeding visible vessel, FIIb an adherent clot, FIIc flat pigmented spot, and FIII clean base ulcer [97] [98] [99]. This classification has been used in numerous studies that aimed to identify patients at risk of persistent ulcer bleeding, rebleeding and mortality. Most of these studies have shown that the presence of an ulcer endoscopically classified as FIa or FIb is an independent risk factor for persistent bleeding or rebleeding [100] [101] [102] [103] [104] [105] [106] [107]. A potential limitation of the Forrest classification is that stigmata recognition and identification, as well as interobserver agreement, may be less than optimal, although the data are conflicting [108] [109].

In addition to the Forrest classification, there are other endoscopic features of peptic ulcers that can predict adverse outcomes and/or endoscopic treatment failure. These include large-size ulcer (> 2 cm), large-size nonbleeding visible vessel, presence of blood in the gastric lumen, and ulcer location on the posterior duodenal wall or the proximal lesser curvature of the stomach [100] [101] [103] [105] [110] [111].

A meta-analysis of RCTs that evaluated endoscopic hemostasis vs. no endoscopic hemostasis demonstrated that endoscopic hemostasis was effective in preventing persistent or recurrent bleeding in actively bleeding ulcers (FIa, FIb: RR 0.29, 95 %CI 0.20 – 0.43; number needed to treat [NNT] 2, 95 %CI 2 – 2) as well as in ulcers with a nonbleeding visible vessel (FIIa: RR 0.49, 95 %CI 0.40 – 0.59; NNT 5, 95 %CI 4 – 6) [112].

[Fig. 2] presents an algorithm for the endoscopic management of bleeding peptic ulcer, stratified by endoscopic stigmata.

Zoom Image
Fig. 2 Algorithm for the endoscopic management of patients with nonvariceal upper gastrointestinal hemorrhage (NVUGIH) secondary to peptic ulcer, stratified by endoscopic stigmata: European Society of Gastrointestinal Endoscopy (ESGE) Guideline. GI, gastrointestinal; PPI, proton pump inhibitor.
1 Use of a large single-channel or double-channel therapeutic upper GI endoscope is recommended.
2 The benefit of endoscopic hemostasis may be greater in patients at higher risk for rebleeding, e. g., older age, co-morbidities, in-hospital UGIH.
3 Large size 10-Fr probe recommended.
4 Absolute alcohol, polidocanol, or ethanolamine injected in limited volumes.
5 High dose oral PPI may be an option in those able to tolerate oral medications.

With respect to the incremental benefit of acid suppression in addition to endoscopic hemostasis, an RCT and a subsequent meta-analysis found a clear advantage for endoscopic hemostasis combined with PPI therapy over PPI therapy alone in preventing recurrent ulcer bleeding and need for surgery in patients with FIIa and FIIb ulcers [113] [114].

The indication for endoscopic treatment of FIIb ulcers (adherent clot) remains controversial because of conflicting data. In evaluation of the natural history of FIIb ulcers (that did not receive endoscopic hemostasis), it was found that 25 % of patients re-bled within 30 days of follow-up [115]. In patients with FIIb ulcers, RCTs and a meta-analysis comparing medical therapy alone with endoscopic hemostasis demonstrated a significant advantage for endoscopic hemostasis in reducing ulcer rebleeding (8.2 % vs. 24.7 %, P < 0.01, yet there was no difference in need for surgery or mortality [116] [117] [118]. In contrast, in a separate RCT, Sung and colleagues reported no ulcer rebleeding in those patients with adherent clots who received medical therapy alone; however the numbers of such patients in the trial were quite limited (n = 24) [113]. Moreover, a meta-analysis restricted only to RCTs showed no benefit for endoscopic hemostasis in patients with an adherent clot (RR 0.31, 95 %CI 0.06 – 1.77) [112].

In patients with peptic ulcers having a flat pigmented spot (FIIc) or clean base (FIII), rebleeding is rare and therefore endoscopic hemostasis does not provide a significant advantage [97] [98] [99].

ESGE does not recommend the routine use of Doppler ultrasound or magnification endoscopy in the evaluation of endoscopic stigmata of peptic ulcer bleeding (strong recommendation, low quality evidence).

The persistence of a positive Doppler signal following endoscopic hemostasis has been shown to predict recurrent bleeding [119]. The results of available studies have been disparate and limited by their methodology, older endoscopic treatments applied, and small numbers of subjects included; thus there is currently no consensus as to the advantage for the routine use of Doppler ultrasound in patents with NVUGIH [120] [121] [122] [123]. A cost-minimization analysis did however demonstrate per-patient cost savings with use of Doppler ultrasound in patients with peptic ulcer bleeding [124].

With respect to magnification endoscopy, one study suggested that FIIa ulcers can be classified as low risk or high risk and that some visible vessels classified as low risk using conventional endoscopy can be reclassified as high risk using magnification endoscopy [125]. However, the classification used has not been validated and no clinical benefit of this approach has been demonstrated.


#

Endoscopic therapy

For patients with actively bleeding ulcers (FIa, FIb), ESGE recommends combining epinephrine injection with a second hemostasis modality (contact thermal, mechanical therapy, or injection of a sclerosing agent). ESGE recommends that epinephrine injection therapy not be used as endoscopic monotherapy (strong recommendation, high quality evidence).

For patients with nonbleeding visible vessel (FIIa), ESGE recommends mechanical therapy, thermal therapy, or injection of a sclerosing agent as monotherapy or in combination with epinephrine injection. ESGE recommends that epinephrine injection therapy not be used as endoscopic monotherapy (strong recommendation, high quality evidence).

For patients with active NVUGIH bleeding not controlled by standard endoscopic hemostasis therapies, ESGE suggests the use of a topical hemostatic spray or over-the-scope clip as salvage endoscopic therapy (weak recommendation, low quality evidence).

Endoscopic hemostasis can be achieved using injection, thermal, and mechanical modalities (see Box 1), and any endoscopic therapy is superior to pharmacotherapy in patients with FIa, FIb and FIIa ulcers [112] [126]. Meta-analyses show that thermal devices (contact and noncontact), injectable agents other than epinephrine (i. e., sclerosing agents, thrombin/fibrin glue), and clips are all effective methods for achieving hemostasis, with no single modality being superior [112] [126] [137] [138] [139] [140] [141].

Box 1 Endoscopic hemostasis modalities: a primer

Injection therapy

The primary mechanism of action of injection therapy is local tamponade resulting from a volume effect. Diluted epinephrine (1:10 000 or 1:20 000 with normal saline injected in 0.5 – 2-ml aliquots in and around the ulcer base) may also have a secondary effect that produces local vasoconstriction [126]. Sclerosing agents such as absolute ethanol, ethanolamine, and polidocanol produce hemostasis by causing direct tissue injury and thrombosis. It should be noted that when using a sclerosing agent in nonvariceal upper gastrointestinal hemorrhage (NVUGIH), the volume injected should be limited because of concerns about tissue necrosis, perforation, or pancreatitis. Another class of injectable agents is tissue adhesives including thrombin, fibrin, and cyanoacrylate glues, which are used to create a primary seal at the site of bleeding.

Endoscopic injection is performed using needles which consist of an outer sheath and an inner hollow-core needle (19 – 25 gauge). The endoscopist or nursing assistant can retract the needle into the sheath for safe passage through the working channel of the endoscope. When the catheter is passed out of the working channel and placed near the site of bleeding, the needle is extended out of the sheath and the solution injected into the submucosa using a syringe attached to the catheter handle [126].

Thermal therapy

Thermal devices used in the treatment of upper gastrointestinal (UGI) bleeding are divided into contact and noncontact modalities. Contact thermal devices include heater probes which generate heat directly and bipolar electrocautery probes which generate heat indirectly by passage of an electrical current through the tissue. Noncontact thermal devices include argon plasma coagulation (APC) tools. Heat generated from these devices leads to edema, coagulation of tissue proteins, contraction of vessels, and indirect activation of the coagulation cascade, resulting in a hemostatic bond [126] [127].

Contact thermal probes use local tamponade (mechanical pressure of the probe tip directly onto the bleeding site) combined with heat or electrical current to coagulate blood vessels, a process known as “coaptive coagulation.” Heater probes (available in 7-Fr and 10-Fr sizes) consist of a Teflon-coated hollow aluminum cylinder with an inner heating coil combined with a thermocoupling device at the tip of the probe to maintain a constant energy output (measured in joules, commonly 15 – 30 joules of thermal energy are delivered). An endoscopist-controlled foot pedal activates the heater probe and provides waterjet irrigation. Multipolar/bipolar electrocautery contact probes (7-Fr and 10-Fr sizes) deliver thermal energy by completion of an electrical local circuit (no grounding pad required) between two electrodes on the tip of the probe as current flows through nondesiccated tissue. As the targeted tissue desiccates, there is a decrease in electrical conductivity, limiting the maximum temperature, depth, and area of tissue injury. An endoscopist-controlled foot pedal controls the delivery of the energy [127]. The standard setting for use in achieving hemostasis in peptic ulcer bleeding is 15 – 20 watts, which is delivered in 8 – 10-second applications (commonly referred to as tamponade stations) [96].

APC, a noncontact thermal modality, uses high frequency, monopolar alternating current conducted to the target tissue through a stream of ionized gas, without mechanical contact, resulting in coagulation of superficial tissue [128]. As the tissue surface loses its electrical conductivity, the argon plasma stream shifts to adjacent nondesiccated (conductive) tissue, which again limits the depth of tissue injury [126]. If the APC catheter is not near the target tissue, there is no ignition of the gas and depression of the foot pedal results only in flow of inert argon gas (flow rates of 0.5 – 0.7 L/min). Coagulation depth is dependent on the generator power setting, duration of application, and distance from the probe tip to the target tissue (optimal distance, 2 – 8 mm) [129] [130].

Mechanical therapy

Endoscopic mechanical therapies include clips (through-the-scope and over-the-scope) and band ligation devices. Endoscopic clips are deployed directly onto a bleeding site and typically slough off within days to weeks after placement [131]. Hemostasis is achieved by mechanical compression of the bleeding site.

Clips are available in a variety of jaw lengths and opening widths. The delivery catheter consists of a metal cable within a sheath enclosed within a Teflon catheter. After insertion of the catheter through the working channel of the endoscope, the clip is extended out of the sheath, positioned over the target area and opened with the plunger handle. A rotation mechanism on the handle is available on some commercially available clips and this allows the endoscopist to change the orientation of the clip at the site of bleeding. The jaws of the clip are applied with pressure and closed onto the target tissue by using the device handle. Some clips may be opened, closed, and repositioned, whereas others are permanently deployed and released upon clip closure. Some clips are provided with a reusable delivery sheath, greatly reducing costs. Similarly, some clips are automatically released on deployment, while others require repositioning of the plunger handle to release the deployed clip from the catheter [131].

The over-the-scope clip device includes an applicator cap, a nitinol clip, and a hand wheel [132] [133]. The applicator cap, with the mounted nitinol clip, is affixed to the tip of the endoscope in a manner similar to that of a variceal band ligation device. Caps are available in three sizes to accommodate various endoscope diameters: 11 mm, 12 mm, and 14 mm. Caps are also available in two lengths (3 mm and 6 mm) to allow variation in the amount of tissue grasped. Clips come in three different shapes of teeth: rounded, pointed and long-pointed. Clips with rounded teeth are used where the goal is tissue compression to achieve hemostasis. The applicator cap incorporates a clip release thread, which is pulled retrogradely through the working channel of the endoscope and fixed onto a hand wheel mounted on the working-channel access port of the endoscope. The clip is released by turning the hand wheel, in a manner similar to deploying a variceal ligation band [134].

Last, endoscopic band ligation devices, commonly used in esophageal variceal bleeding, have also been reported for treatment of NVUGIH (e. g., for Dieulafoy lesion) and involve the placement of elastic bands over tissue to produce mechanical compression and tamponade.

Topical therapy

Topical hemostatic sprays have been used in acute NVUGIH with promising results, but thus far in a limited number of patients and without any comparative data regarding standard endoscopic hemostasis therapies [135] [136]. Advantages of noncontact, spray catheter delivery of hemostatic agents include ease of use, lack of need for precise lesion targeting, access to lesions in difficult locations, and the ability to treat a large surface area.

Topical hemostatic sprays include TC-325, (Hemospray, Cook Medical Inc, Winston-Salem, North Carolina, USA), which is a proprietary, inorganic, absorbent powder that rapidly concentrates clotting factors at the bleeding site, forming a coagulum. Hemospray comes in a hand-held device consisting of a pressurized CO2 canister, a through-the-scope delivery catheter, and a reservoir for the powder cartridge. The powder is delivered via pushbutton in 1 – 2-second bursts until hemostasis is achieved. The maximum amount of TC-325 that can be safely administered during a single treatment session has not yet been established [135] [136]. The coagulum typically sloughs within 3 days and is naturally eliminated. Hemospray has received regulatory clearance in some countries.

Additional topical hemostatic sprays include EndoClot and the Ankaferd Blood Stopper [135] [136]. EndoClot (EndoClot Plus Inc, Santa Clara, California, USA) is a starch-derived compound that rapidly absorbs water from serum and concentrates platelets, red blood cells, and coagulation proteins at the bleeding site to accelerate the clotting cascade. Hemostatic sprays derived from plant products/extracts have also been evaluated. Clinical experience with these agents for endoscopic hemostasis is currently limited to the off-label use of the Ankaferd Blood Stopper (Ankaferd Health Products Ltd, Istanbul, Turkey). This topical agent promotes formation of a protein mesh that acts as an anchor for erythrocyte aggregation without significantly altering coagulation factors or platelets and is delivered onto the bleeding site via an endoscopic spray catheter until an adherent coagulum is formed. The particles are subsequently cleared from the bleeding site within hours to days later. The overall efficacy of these topical agents is unknown in brisk arterial bleeding and may be limited because of the rapid “wash-away” effect of the hemostatic agent by ongoing blood flow.

Epinephrine injection therapy is effective at achieving primary hemostasis, but inferior to other endoscopic hemostasis monotherapies or combination therapy in preventing ulcer rebleeding [112] [126] [139]. In the most recently published meta-analysis (19 RCTs, 2033 patients), epinephrine plus any second hemostasis modality significantly reduced rebleeding (OR 0.53, 95 %CI 0.35 – 0.81) and emergency surgery (OR 0.68, 95 %CI 0.50 – 0.93) but not mortality as compared with epinephrine injection monotherapy for high risk peptic ulcers [140]. Therefore, it is recommended that if epinephrine is used to treat peptic ulcer bleeding with high risk stigmata, it should only be used in combination with a second endoscopic hemostasis modality [97] [98] [99] [141].

With respect to contact thermal therapy (e. g., bipolar electrocoagulation, heater probe), a meta-analysis restricted only to RCTs found that contact thermal therapy was significantly more effective than no endoscopic hemostasis in achieving primary hemostasis (RR 11.7, 95 %CI 5.2 – 26.6), reducing recurrent bleeding (RR 0.44, 95 %CI 0.36 – 0.54; NNT = 4), need for urgent surgery (RR 0.39, 95 %CI 0.27 – 0.55; NNT = 8) and mortality (RR 0.58, 95 %CI 0.34 – 0.98) [112]. With respect to noncontact thermal therapy (e. g., argon plasma coagulation), limited data from three small RCTs suggest it is similar in efficacy to injection of a sclerosing agent (polidocanol) or contact thermal therapy (heater probe) [112].

Mechanical therapy using through-the-scope clips was found to be superior to injection monotherapy in four of five meta-analyses [112] [126] [137] [139] [142]. Mechanical therapy significantly reduced the risk of recurrent bleeding by 78 % (RR 0.22, 95 %CI 0.09 – 0.55) [112]. Compared with thermal coagulation, mechanical therapy provided no significant improvement in definitive hemostasis (RR 1.00, 95 %CI 0.77 – 1.31) [137]. However, a separate meta-analysis [126] found through-the-scope clips to be significantly more effective than thermal therapy in reducing the risk of recurrent bleeding (OR 0.24, 95 %CI 0.06 – 0.95). Two small studies from Japan compared the efficacy of clips versus hemostatic forceps [143] [144]. The first was an RCT conducted in 96 patients with high risk bleeding gastric ulcers and showed that use of monopolar, soft coagulation hemostatic forceps was as effective as clipping [143]. The second was an observational prospective cohort study on 50 patients in which use of bipolar hemostatic forceps was more effective than endoscopic clipping for both initial hemostasis (100 % vs. 78.2 %) and preventing recurrent bleeding (3.7 % vs. 22.2 %) [144]. Unlike thermal therapies and sclerosing agents, mechanical therapy using clips has the theoretical benefit of inducing only limited tissue injury, and therefore may be preferred in patients on antithrombotic therapy and those patients undergoing repeat endoscopic hemostasis for rebleeding. A multidisciplinary expert panel developed an explicit set of evidence-based quality indicators for NVUGIH [78]. Among them, it was felt that patients with ulcer-related bleeding with high risk stigmata and elevated INR (> 1.5 – 2.0), should receive endoscopic hemostasis using endoscopic clips or a combination of epinephrine injection plus clips.

Meta-analyses have shown that combination endoscopic hemostasis therapy (dilute epinephrine injection combined with a second hemostasis modality including injectable, thermal contact probe, or clips) is superior to injection therapy alone, but not to clips or contact thermal therapy alone [126] [139]. There may be practical reasons to pre-inject dilute epinephrine before other therapies for high risk endoscopic stigmata. Injection of epinephrine may slow or stop bleeding allowing improved visualization for application of subsequent therapy. Adverse events associated with combination endoscopic hemostasis are low and include induction of bleeding (1.7 %) and perforation (0.6 %) [139]. Recent international consensus guidelines endorse combination therapy (dilute epinephrine injection combined with contact thermal therapy, clips, or injection of a sclerosant [e. g., absolute ethanol]) as appropriate treatment in patients with peptic ulcer bleeding with high risk endoscopic stigmata [98] [99] [145].

New endoscopic hemostasis modalities (topical hemostatic sprays and over-the-scope clips) are emerging as possible alternative endotherapies for primary hemostasis when bleeding is refractory or not amenable to standard endoscopic hemostasis therapies [136] [146]. Moreover, several small retrospective studies have reported that an over-the-scope clip (OVESCO), may have a role as rescue hemostasis therapy for severe NVUGIH when conventional endoscopic treatment modalities fail [133] [134] [147]. An inert nanopowder (Hemospray) that causes immediate hemostasis when sprayed onto active bleeding [136] [148] has recently been used as a primary hemostasis agent or as a second-line salvage therapy. Several prospective uncontrolled studies, a large European registry [149] [150] [151] [152] [153] [154] and a systematic review of the current limited data suggests that Hemospray is safe and effective and may be best used in high risk cases as a temporizing measure or a bridge toward more definitive treatment [136]. Other topical agents, such as the starch-derived polysaccharide hemostatic system (EndoClot) and the Ankaferd blood stopper are also emerging [136]. However, RCTs directly comparing topical agents with traditional hemostasis methods are required to better define their optimal role and safety in the endoscopic management of NVUGIH.

For patients with acid-related causes of NVUGIH different from peptic ulcers (e. g., erosive esophagitis, gastritis, duodenitis), ESGE recommends treatment with high dose PPI. Endoscopic hemostasis is usually not required and selected patients may be discharged early (strong recommendation, low quality evidence).

ESGE recommends that patients with a Mallory – Weiss lesion that is actively bleeding receive endoscopic hemostasis. There is currently inadequate evidence to recommend a specific endoscopic hemostasis modality. Patients with a Mallory – Weiss lesion and no active bleeding can receive high dose PPI therapy alone (strong recommendation, moderate quality evidence).

ESGE recommends that a Dieulafoy lesion receive endoscopic hemostasis using thermal, mechanical (hemoclip or band ligation), or combination therapy (dilute epinephrine injection combined with contact thermal or mechanical therapy) (strong recommendation, moderate quality evidence). Transcatheter angiographic embolization (TAE) or surgery should be considered if endoscopic treatment fails or is not technically feasible (strong recommendation, low quality evidence).

In patients bleeding from upper GI angioectasias, ESGE recommends endoscopic hemostasis therapy. However, there is currently inadequate evidence to recommend a specific endoscopic hemostasis modality (strong recommendation, low quality evidence).

In patients bleeding from upper GI neoplasia, ESGE recommends considering endoscopic hemostasis in order to avert urgent surgery and reduce blood transfusion requirements. However, no currently available endoscopic treatment appears to have long-term efficacy (weak recommendation, low quality evidence).

Erosive esophagitis, gastritis and duodenitis are common causes of NVUGIH and generally have a benign course and excellent prognosis [2] [64] [155] [156] [157] [158]. Meta-analyses show that acid suppression therapy is effective, with high dose PPI therapy being significantly more effective than H2-receptor antagonists and no observed differences in effectiveness amongst PPIs [159] [160]. Endoscopic hemostasis is usually not required in this patient population and selected patients are candidates for early hospital discharge.

Although spontaneous resolution of bleeding is frequent, observational studies have demonstrated that acute UGIH secondary to Mallory – Weiss syndrome has a mortality similar to that of peptic ulcer bleeding [161] [162]. Risk factors for adverse outcomes include older age, medical co-morbidities, and active bleeding at the time of endoscopy. The latter supports early endoscopy to stratify risk and to perform endoscopic hemostasis if active bleeding is identified [162] [163] [164] [165] [166]. Despite suggestions that mechanical methods (clips and band ligation) are more effective than epinephrine injection, this has not been found in all studies [164] [167] [168]. Mechanical therapy appears to be safe, yet data are insufficient to make a clear recommendation of one hemostasis modality over another [164] [167] [169] [170].

The proximal stomach and duodenum are the most common locations for Dieulafoy lesions [171]. Endoscopic hemostasis is warranted if technically feasible. Observational studies have reported the superiority of combined, thermal and mechanical methods over injection monotherapy, in achieving primary hemostasis, preventing rebleeding, and in reducing the need for rescue therapy, yet with no proven mortality benefit [172] [173] [174] [175] [176] [177] [178] [179] [180]. All endoscopic hemostasis modalities (e. g., band ligation, through-the-scope clips, over-the-scope clips, contact thermal coagulation, and argon plasma coagulation) appear safe and have similar reported outcomes [171] [172] [173] [174] [175] [176] [177] [178] [179] [180]. Selective TAE has been described as an effective rescue therapy if endoscopic hemostasis fails or in patients who are poor surgical candidates [181] [182]. If both endoscopic and angiographic therapies fail, surgery should be considered.

Studies on endoscopic hemostasis therapy of angioectasias of the upper GI tract are observational and include only a limited number of subjects. In two recent meta-analyses, endoscopic hemostasis therapy (e. g., argon plasma coagulation, heater probe, bipolar coagulation, monopolar coagulation, band ligation, YAG laser) is reported to be initially effective and safe, yet bleeding recurrence rates are significant [183] [184]. Given the low quality of evidence and scarcity of comparative data, a recommendation on a specific endoscopic hemostasis treatment is not permitted at this time.

There are limited published data on the role of endoscopic hemostasis in bleeding due to upper GI tract neoplasia and evidence to support a specific modality is scarce [185] [186] [187] [188]. Numerous endoscopic hemostasis modalities (e. g., injection, thermal, mechanical, topical spray/powder) have been reported, generally with limited impact on primary hemostasis, prevention of rebleeding, or mortality. However, endoscopic treatment may avert urgent surgery, reduce transfusion requirements, and may provide a temporary bridge to oncologic therapy and/or selective embolization [185] [186] [187] [188].


#
#

Management following endoscopy/endoscopic hemostasis

ESGE recommends PPI therapy for patients who receive endoscopic hemostasis and for patients with adherent clot not receiving endoscopic hemostasis. PPI therapy should be high dose and administered as an intravenous bolus followed by continuous infusion (80 mg then 8 mg/hour) for 72 hours post endoscopy (strong recommendation, high quality evidence)

ESGE suggests considering PPI therapy as intermittent intravenous bolus dosing (at least twice-daily) for 72 hours post endoscopy for patients who receive endoscopic hemostasis and for patients with adherent clot not receiving endoscopic hemostasis. If the patient’s condition permits, high dose oral PPI may also be an option in those able to tolerate oral medications (weak recommendation, moderate quality evidence).

Based upon previously published meta-analytic data, evidence-based guidelines on NVUGIH have recommended that PPI therapy be given as an 80 mg intravenous bolus followed by 8 mg/hour continuous infusion to reduce rebleeding, surgery, and mortality in patients with high risk ulcers that had undergone successful endoscopic hemostasis [98] [99] [189] [190]. More recently however, a meta-analysis of RCTs of high risk bleeding ulcers treated with endoscopic hemostasis compared intermittent PPI dosing (oral or intravenous) with the currently recommended post hemostasis PPI regimen of 80 mg intravenous bolus followed by 8 mg/hour continuous infusion [191]. In that meta-analysis, Sachar et al reported that the risk ratio of recurrent ulcer bleeding within 7 days for intermittent infusion of PPI vs. bolus plus continuous infusion of PPI was 0.72 (upper boundary of one-sided 95 %CI 0.97), with an absolute risk difference of −2.64 %. Risk ratios for other outcomes, including radiologic/surgical intervention and mortality, showed no differences between infusion regimens. These meta-analytic data indicate that intermittent PPI therapy appears comparable to the currently recommended regimen of intravenous bolus plus continuous PPI infusion post endoscopic hemostasis. It should be noted however, that intermittent PPI bolus dosing is associated with a somewhat higher risk of rebleeding that in general can be managed endoscopically. Given the pharmacodynamic profile of PPIs, consideration should be given to use of high dose PPI infusion given at least twice-daily, and using high dose oral PPIs in patients able to tolerate oral medications [191]. The concept of high dose PPI varies between the different studies used in the meta-analysis conducted by Sachar et al. However, it appears that an 80 mg oral PPI dose followed by 40 – 80 mg orally every 12 hours for 72 hours yields an intragastric pH similar to that reported with continuous intravenous PPI infusion following successful endoscopic hemostasis of high risk peptic ulcers [192]. This is but one study, and therefore we need more data to confirm these findings before drawing firm practical conclusions for the post-endoscopy management of patients with NVUGIH. These data are in agreement with an RCT that randomized patients to high dose continuous infusion of esomeprazole vs. 40 mg of oral esomeprazole twice-daily for 72 hours (118 vs. 126 patients respectively) [193]. Recurrent bleeding at 30 days was reported in 7.7 % and 6.4 % of patients, respectively (difference −1.3 percentage points, 95 %CI −7.7 to 5.1 percentage points). However, this study was conducted in an Asian population (e. g., PPI slow metabolizers) and its findings may not be generalizable to Western NVUGIH populations. Moreover, this study was stopped prematurely since it was not designed as an equivalency trial, and based on the preliminary data, thousands of patients would have been required in order to complete the study. (See Appendix e8, online-only.)

In patients with clinical evidence of rebleeding following successful initial endoscopic hemostasis, ESGE recommends repeat upper endoscopy with hemostasis if indicated. In the case of failure of this second attempt at hemostasis, transcatheter angiographic embolization (TAE) or surgery should be considered (strong recommendation, high quality evidence).

An RCT comparing endoscopic therapy with surgery for recurrent peptic ulcer bleeding after successful initial endoscopic control of bleeding showed that 35/48 (73 %) of patients randomized to endoscopic re-treatment had long-term control of their peptic ulcer bleeding, avoided surgery, and had a lower rate of adverse events as compared to the surgery-treated patients [194]. The remaining 13 patients underwent salvage surgery because of failed repeat endoscopic hemostasis (n = 11) or perforation due to contact thermal therapy (n = 2).

If further bleeding occurs following a second endoscopic treatment, surgery for low risk patients or interventional radiology for high risk patients should be considered [195]. In recent systematic reviews and meta-analyses comparing TAE with surgery for peptic ulcer bleeding after failed endoscopic hemostasis, a higher rebleeding rate was observed following TAE. No significant difference in mortality or need for additional interventions was shown between treatments [196] [197]. Hemostatic powder and over-the-scope clips may also be considered as rescue/salvage therapy. Although limited, emerging data suggest that hemostatic powder may be successfully employed as salvage hemostasis therapy [154] [198]. The over-the-scope clip (OTSC) has also proven an effective and safe therapeutic option for severe acute GI bleeding when conventional endoscopic treatment modalities fail [134] [147].

(See Appendix e9, online-only.)

ESGE does not recommend routine second-look endoscopy as part of the management of NVUGIH. However, second-look endoscopy may be considered in selected patients at high risk for rebleeding (strong recommendation, high quality evidence).

Routine second-look endoscopy is defined as a scheduled repeat endoscopic assessment of the previously diagnosed bleeding lesion usually performed within 24 hours following the index endoscopy [98]. This strategy employs repeat endoscopy regardless of the type of bleeding lesion, perceived rebleeding risk, or clinical signs of rebleeding. A meta-analysis that evaluated the effectiveness of routine second-look endoscopy in NVUGIH reported a significant reduction in rebleeding (OR 0.55, 95 %CI 0.37 – 0.81) and need for emergency surgery (OR 0.43, 95 %CI 0.19 – 0.96), but not mortality (OR 0.65, 95 %CI 0.26 – 1.62) [199]. However, only one included study in that meta-analysis utilized high dose intravenous PPI, and in that study no benefit for second-look endoscopy was observed, while any protective effect was limited only to high risk patients (e. g., those with active bleeding at index endoscopy). Similarly, scheduled second-look endoscopy does not appear to be cost-effective outside the subgroup of patients thought to be at high risk for recurrent ulcer bleeding [200]. Thus, the clinical utility and cost – efficiency of routine second-look endoscopy in unselected patients remains to be proven.

In patients with NVUGIH secondary to peptic ulcer, ESGE recommends investigating for the presence of Helicobacter pylori in the acute setting with initiation of appropriate antibiotic therapy when H. pylori is detected. Re-testing for H. pylori should be performed in those patients with a negative test in the acute setting. Documentation of successful H. pylori eradication is recommended (strong recommendation, high quality evidence).

Peptic ulcer remains the most frequent cause of acute NVUGIH with H. pylori infection remaining the primary cause of peptic ulcer disease [201] [202]. Indeed, when H. pylori is eradicated, the risk of ulcer rebleeding is reported to be extremely low [203] [204]. However, the false-negative rate of H. pylori diagnostic testing is higher if the test is performed at the time of the acute bleeding episode as compared to later follow-up [205]. A meta-regression analysis including 8496 bleeding peptic ulcer patients found an H. pylori prevalence of 72 %, with the infection rate being significantly higher when diagnostic testing was delayed until at least 4 weeks following the bleeding event (OR 2.08, 95 %CI 1.10 – 3.93; P = 0.024) [206]. Therefore, it is advisable to re-test at a later time those patients who had a negative H. pylori test in the acute setting.

When H. pylori infection is found, eradication therapy should be initiated and guided by patient and local factors [98] [99]. Documentation of successful H. pylori eradication is strongly recommended given the high risk of recurrent ulcer bleeding in the presence of persistent H. pylori infection [98] [99]. (See Appendix e10, online-only.)

ESGE recommends restarting anticoagulant therapy following NVUGIH in patients with an indication for long-term anticoagulation. The timing for resumption of anticoagulation should be assessed on a patient by patient basis. Resuming warfarin between 7 and 15 days following the bleeding event appears safe and effective in preventing thromboembolic complications for most patients. Earlier resumption, within the first 7 days, may be indicated for patients at high thrombotic risk (strong recommendation, moderate quality evidence).

Retrospective, observational data have shown that resuming anticoagulation in patients with GI bleeding is associated with a lower risk of thrombosis and death [207] [208] [209]. Restarting warfarin therapy within 7 days of the index bleeding event was associated with an approximately twofold increased risk of rebleeding [207] [209]. Conversely, as compared with resuming warfarin beyond 30 days, resuming warfarin between 7 and 30 days did not increase the risk of rebleeding, but did significantly decrease the risk of thromboembolism and improved survival [209]. These data appear to support that resumption of anticoagulation after 7 days of interruption is safe and effective in preventing thromboembolic complications for most patients. However, in patients at high thrombotic risk (e. g., chronic atrial fibrillation with previous embolic event, CHADS2 score ≥ 3, mechanical prosthetic heart valve, recent [within past 3 months] deep venous thrombosis or pulmonary embolism, and patients with known severe hypercoagulable state), for whom early resumption of anticoagulation within the first week following an acute bleeding event might be appropriate, bridging therapy using unfractionated or low molecular weight heparin may be considered [210]. No data are currently available to guide the management of DOACs following NVUGIH. Yet caution in the early resumption of DOACs is required because of their rapid onset of action and the current lack of reversal agents. (See Appendix 11, online-only.)

In patients receiving low dose aspirin for primary cardiovascular prophylaxis who develop peptic ulcer bleeding, ESGE recommends withholding aspirin, re-evaluating the risks/benefits of ongoing aspirin use in consultation with a cardiologist, and resuming low dose aspirin following ulcer healing or earlier if clinically indicated (strong recommendation, low quality evidence). See [Fig. 1].

In patients receiving low dose aspirin for secondary cardiovascular prophylaxis who develop peptic ulcer bleeding, ESGE recommends aspirin be resumed immediately following index endoscopy if the risk of rebleeding is low (e. g., FIIc, FIII). In patients with high risk peptic ulcer (FIa, FIb, FIIa, FIIb), early reintroduction of aspirin by day 3 after index endoscopy is recommended, provided that adequate hemostasis has been established (strong recommendation, moderate quality evidence). See [Fig. 1].

In patients receiving dual antiplatelet therapy (DAPT) who develop peptic ulcer bleeding, ESGE recommends continuing low dose aspirin therapy. Early cardiology consultation should be obtained regarding the timing of resuming the second antiplatelet agent (strong recommendation, low quality evidence). See [Fig. 1].

Discontinuing low dose aspirin therapy in the setting of secondary cardiovascular prophylaxis significantly increases the risk of an adverse cardiovascular event, usually occurring within the first week of discontinuation [211] [212] [213] [214]. In a retrospective cohort study, patients with cardiovascular disease who discontinued low dose aspirin following peptic ulcer bleeding had an almost twofold increase in risk for death or an acute cardiovascular event in the first 6 months after hospital discharge, as compared with patients who continued aspirin therapy [54]. In an RCT evaluating continuous vs. interrupted aspirin treatment in patients with high risk peptic ulcers and at high cardiovascular risk, those receiving continuous aspirin had a twofold increased risk of early, nonfatal, recurrent bleeding (10.3 % vs. 5.4 % at 4 weeks; difference 4.9 percentage points, 95 %CI −3.6 to 13.4 percentage points; HR 1.9, 95 %CI 0.6 – 6.0), yet a 10-fold reduced risk of all-cause mortality at 8 weeks (1.3 % vs. 12.9 %; difference 11.6 percentage points, 95 %CI 3.7 – 19.5 percentage points; HR 0.2 95 %CI 0.06 – 0.60) and a lower mortality rate related to cardiovascular, cerebrovascular, or gastrointestinal events (1.3 % vs. 10.3 %; difference 9 percentage points, 95 %CI 1.7 – 16.3 percentage points; HR 0.2, 95 %CI 0.05 – 0.70), compared with those patients in whom aspirin was withheld [53]. Patients who required DAPT were excluded from this study. The antiplatelet effect of aspirin lasts for approximately 5 days (although new active platelets increase in number each day), and the risk of early recurrent bleeding is high in the first 3 days [53]. Therefore, restarting aspirin on day 3 in patients with high risk endoscopic stigmata is a reasonable trade-off between the risks of rebleeding and thrombosis. In patients with peptic ulcer bleeding with no high risk endoscopic stigmata, aspirin can be resumed immediately as RCTs have shown that neither aspirin nor clopidogrel use impede ulcer healing promoted by PPIs [53] [55] [56]. No high level evidence helps guide the timing for resumption of P2Y12 platelet receptor inhibitors (e. g., clopidogrel) following NVUGIH. However, in view of its similar antiplatelet activity, it seems reasonable to apply a similar management strategy. Moreover, there is no evidence in the literature to help guide the management of patients receiving DAPT in the setting of NVUGIH. The overriding principle of balancing bleeding and thrombotic event risks requires close collaboration between the gastroenterology and cardiology teams.

In patients requiring dual antiplatelet therapy (DAPT) and who have had NVUGIH, ESGE recommends the use of a PPI as co-therapy (strong recommendation, moderate quality evidence.

Dual antiplatelet therapy, combining low dose aspirin and a P2Y12 platelet receptor inhibitor (e. g., clopidogrel), is the cornerstone of management of patients with acute coronary syndromes and following coronary stent placement, but is associated with an increased risk of GI bleeding [215] [216] [217]. Proton pump inhibitors substantially reduce this risk and their use is recommended in patients with a previous GI bleeding event [218] [219] [220]. Pharmacodynamic studies have shown that the co-administration of PPIs with clopidogrel reduces platelet inhibition, but the clinical significance of this interaction has been extensively debated [221] [222] [223] [224] [225]. Previous meta-analyses suggest that concomitant clopidogrel and PPI use may be associated with increased adverse cardiovascular events and myocardial infarction, but no effect on mortality [226] [227]. However, the presence of significant heterogeneity in the included studies indicates that this evidence is at best, inconsistent, and at worst, potentially biased or confounded. A recent meta-analysis included a subanalysis limited to RCTs and propensity-matched studies evaluating the interaction between PPI and clopidogrel; the subanalysis showed no significant differences between patients using clopidogrel alone and patients receiving the combination of clopidogrel and a PPI (n = 11 770) for all-cause mortality (OR 0.91, 95 %CI 0.58 – 1.40; P = 0.66), acute coronary syndrome (OR 0.96, 95 %CI 0.88 – 1.05; P = 0.35), myocardial infarction (OR 1.05, 95 %CI 0.86 – 1.28; P = 0.65), and cerebrovascular accident (OR 1.47, 95 %CI 0.660 – 3.25; P = 0.34) [228]. The incidence of GI bleeding was significantly decreased in the group of patients who received a PPI (OR 0.24, 95 %CI 0.09 – 0.62; P = 0.003). Current evidence does not support a clinically relevant interaction between PPIs and clopidogrel. (See Appendices e12 and e13, online-only.)

ESGE guidelines represent a consensus of best practice based on the available evidence at the time of preparation. They may not apply in all situations and should be interpreted in the light of specific clinical situations and resource availability. Further controlled clinical studies may be needed to clarify aspects of these statements, and revision may be necessary as new data appear. Clinical consideration may justify a course of action at variance to these recommendations. ESGE guidelines are intended to be an educational device to provide information that may assist endoscopists in providing care to patients. They are not rules and should not be construed as establishing a legal standard of care or as encouraging, advocating, requiring, or discouraging any particular treatment.

Appendix e1

Nonvariceal upper gastrointestinal hemorrhage (NVUGIH): task forces and key questions.

Topics and key questions

Task forces
(leader in bold)

Task force 1: Initial patient evaluation/hemodynamic resuscitation/risk stratification

David S. Sanders
Jean-Marc Dumonceau
Matthew Kurien
Gilles Lesur
Riccardo Marmo

How should the patient be initially hemodynamically resuscitated?

Who should receive blood product transfusion? What target for hemoglobin?

How should patient risk stratification be used?

What risk stratification score(s) are reliable and valid? Pre-endoscopy risk score? Post-endoscopy risk score?

How should risk stratification tools be applied?

Task force 2: Pre-endoscopic management

Jean-Marc Dumonceau
Ian Gralnek
Cesare Hassan
Angel Lanas
Gilles Lesur
Istvan Racz
Franco Radaelli
Gianluca Rotondano

How to manage the patient using antiplatelet and anticoagulant drugs (known collectively as antithrombotic agents) at the time of acute upper gastrointestinal (UGI) bleeding?

Need to also consider the current data on potential adverse events related to antiplatelet/anticoagulant drug interruption (i. e. atrial fibrillation, cardiac stent thrombosis, cardiac ischemic event, neurovascular event)

What is the role of pre-endoscopy proton pump inhibitor (PPI) therapy?

What is the role of pre-endoscopy somatostatin therapy?

What is the role of naso-/orogastric tube aspiration/lavage?

What is the role of endotracheal intubation before upper endoscopy?

Is there a role for antifibrinolytic medications?

What is the role of prokinetic agents prior to upper endoscopy?

Is there a role for capsule endoscopy in the emergency department in evaluating acute UGI bleeding?

What is appropriate timing for upper endoscopy?

Task force 3: Endoscopic management

Ernst J. Kuipers
Ricardo Cardoso
Livio Cipolletta
Mário Dinis-Ribeiro
Luís Maia
Gianluca Rotondano
Paulo Salgueiro

Which endoscopic classification should be used for describing high and low risk endoscopic stigmata of recent hemorrhage in peptic ulcer bleeding? What are high risk vs. low risk endoscopic stigmata and their importance in risk stratification?

Is there a role for doppler ultrasonography, magnification endoscopy, chromoendoscopy in helping to better evaluate endoscopic stigmata of recent hemorrhage for peptic ulcer bleeding?

Which ulcer stigmata require endoscopic hemostasis? Which do not?

Which endoscopic hemostasis modality should be used (with focus on peptic ulcer bleeding)?

Injection therapy?

Thermal contact therapy?

Thermal noncontact therapy?

Mechanical therapy?

Combination therapy?

Topical spray/powder therapy

What to do in situations of nonvariceal, nonulcer bleeding lesions?

Task force 4: Post-endoscopic management

Angel Lanas
Lars Aabakken
Alberto Arezzo
Roberto de Franchis
Cesare Hassan
Ralf-Thorsten Hoffmann
Tomas Hucl
Gilles Lesur
Franco Radaelli
Andrew Veitch
Angelo Zullo

What is the medical management post endoscopic hemostasis?

What to do when rebleeding occurs? What is the role of repeat upper endoscopy?

Is there a role for scheduled second-look endoscopy?

Rebleeding/failed endoscopic hemostasis: When should the interventional radiologist be involved/when should the surgeon be involved?

Diagnosis and treatment of Helicobacter pylori? When? In whom? What if testing for H. pylori in the acute setting of bleeding is negative? Documentation of eradication?

How to manage the NVUGIH patient using antiplatelet and anticoagulant drugs (collectively known as antithrombotics agents) post endoscopy? How and when to reinstitute these medications?

When to discharge patients home?

Appendix e2

Criteria for outpatient management of patients with nonvariceal upper gastrointestinal hemorrhage (NVUGIH).

First author, year [ref.]

Study design, study objective

Participants

Patients, n (%)

Outcomes

Observed

Out-patients

Transfusion required

Re-bleeding

Need for surgery

Death

Longstreth, 1995 [229]

Prospective

Absolute criteria.
Absence of: high risk endoscopic findings (arterial bleeding, adherent clot, or visible vessel); varices; and portal hypertensive gastropathy

Nonabsolute criteria.
No debilitation; no orthostatic vital sign change; no severe liver disease; no serious concomitant disease; no anticoagulation therapy or coagulopathy; no fresh, voluminous hematemesis or multiple episodes of melena on the day of presentation; no severe anemia (hemoglobin < 8.0 g/dL); adequate support at home

141

34 (24 %)

1 (2.9 %)

0

0

Longstreth, 1998 [230]

Retrospective

As above

Not done

176

1 (0.5 %)

0

0

Cebollero-Santamaria, 1999 [231]

Prospective

Clinical criteria.
Absence of: severe cardiac disease/recent myocardial infarction; severe respiratory failure; decompensated cirrhosis; severe coagulopathy (international normalized ratio [INR] ≥ 1.5); poor social support; requirement for blood transfusion; recent cerebrovascular accident

Endoscopic criteria.
Clean base ulcer < 1.5 cm in diameter; erosive mucosal disease (esophagus, stomach, or duodenum); nonbleeding Mallory – Weiss tear; portal hypertensive gastropathy

84

24 (28 %)

1 (4.1 %)

0

0

Brullet, 2004 [232]

RCT

Clinical criteria.
Absence of signs of hypovolemia (defined as systolic blood pressure < 100 mmHg and pulse rate > 100 beats/minute with peripheral signs of circulatory failure);
Absence of severe associated diseases (defined as American Society of Anesthesiologists [ASA] classification III – V);
No anticoagulant drug therapy; and
Appropriate sociofamilial support.
The latter was defined as the following: (i) living within a family whose members were able to comprehend the clinical condition of the patient and capable of providing adequate care; (ii) having a telephone; and (iii) residing less than 20 minutes by car from the hospital.

Endoscopic criteria.
(1) peptic ulcer with an nonbleeding visible vessel;
(2) ulcer size smaller than 15 mm

297 endoscopically treated patients

40 (13 %)

7

2

0

0

Lai, 1997 [233] [1]

Prospective

Inclusion criteria were as follows:
(1) Presence of melena and/or hematemesis;
(2) Presence of duodenal ulcer and absence of other upper gastrointestinal pathology to account for gastrointestinal bleeding;
(3) Hemoglobin level greater than 10 gm/dL on admission;
(4) Systolic blood pressure above 100 mmHg and pulse less than 100 beats/minute on admission before any resuscitation with fluids, and orthostatic blood pressure drop of less than 10 mmHg (patient excluded if abnormal vital signs developed after admission);
(5) No concurrent serious medical illness (absence of heart failure, chronic obstructive airway disease, hepatic cirrhosis, hematologic malignancies, chronic renal failure, and strokes);
(6) no stigmata of recent hemorrhage on endoscopy, which was carried out within 24 h of admission;
(7) Age 60 years or younger; and
(8) Normal coagulation findings.

305

75 (24 %)

0

0

0

0

Lee, 1999 [234] [2]

RCT

Clinical criteria.
Absence of:
Co-morbid illness requiring intensive care (e. g., myocardial ischemia);
Hemodynamic instability after resuscitation by infusion of 2 L of fluid (heart rate greater than 115 beats/min, systolic blood pressure less than 90 mmHg, or diastolic blood pressure less than 60 mmHg);
Known or suspected variceal source, coagulopathy (use of any anticoagulant or thrombolytic agent within the preceding week, platelet count less than 50 000, INR < 1.5, or any other known coagulopathy);
Upper GI bleeding within the preceding 1 month; and
Age less than 18 years.
Endoscopic criteria.
No clot obscuring complete visualization;
Clean ulcer base;
Mallory-Weiss tear;
Esophagitis, gastritis, duodenitis, or other benign findings.
Hemodynamically stable, alert, wanted to go home

 56

26 (46 %)

1 (3.8 %)

0

0

Gralnek, 2004 [235]

Retrospective

Complete Rockall scores (after endoscopy) of ≤ 2

175

53 (30 %)

2 (3.7 %)

0

0

Cipolletta. 2002 [236] [2]

RCT

Clinical criteria.
Absence of hypovolemic shock, orthostatic change in vital signs, and need for blood transfusion;
Normal coagulation findings;
Absence of serious concurrent medical illness;
Easy accessibility to hospital; and
Adequate sociofamial support at home.

Endoscopic criteria.
Absence of varices and signs of portal hypertension, high risk stigmata of recent hemorrhage (active bleeding, visible vessel, or adherent clot);
Patients having a clean ulcer base or flat spot, gastritis or duodenitis, Mallory – Weiss tear, or other benign findings.

244

95 (39 %)

2 (2.%)

0

0

GI, gastrointestinal; INR, international normalized ratio; RCT, randomized controlled trial.

1 Only patients with peptic ulcer


2 The hospital stay and the costs of care were significantly less for early endoscopy


Appendix e3

Role of proton pump inhibitors (PPIs) prior to upper endoscopy in acute upper gastrointestinal hemorrhage.

First author, year [ref.]

Study type

Patient group

Key outcomes

Key results

Limitations

Conclusions

Sreedharan, 2012 [237]

Cochrane meta-analysis of 6 RCTs
Searches were re-run in Feb 2006 and Oct 2008

Total of 2223 patients were included with unselected upper GI bleeding.

Prior to endoscopy patients were given PPI (oral or intravenous) or H2RA or placebo

  • 30-day mortality

  • Rebleeding

  • Surgery

  • Stigmata of recent hemorrhage (SRH) at index endoscopy

  • Need for endoscopic therapy

  • Need for transfusion

No significant difference between PPI and control treatment for:

  • Mortality: 6.1 % vs. 5.5 %; OR 1.12, 95 %CI 0.72 – 1.73

  • Rebleeding rates: 13.9 % vs. 16.6 %; OR 0.81, 95 %CI 0.61 – 1.09

  • Surgery: 9.9 % – 10.2 %; OR 0.96, 95 %CI 0.68 – 1.35

Significant reduction:

  • Proportion with SRH: 37.2 % – 46.5 %; OR 0.67, 95 %CI 0.54 – 0.84

  • Need for endoscopic therapy: 8.6 % – 11.7 %; OR 0.68, 95 %CI 0.50 – 0.93

Oral and intravenous PPI studies are mixed

  • No data regarding the length of pre-emptive PPI and placebo treatment

  • Variceal and nonvariceal bleeding sources are not analyzed selectively

PPI treatment initiated before endoscopy for upper GI bleeding might reduce the proportion of patients with SRH at index endoscopy and significantly reduces requirement for endoscopic therapy during index endoscopy.
However, there is no evidence that PPI treatment affects clinically important outcomes, namely mortality or need for surgery.

Lau, 2007 [238]

Double-blind, placebo-controlled, randomized trial

Omeprazole group (n = 319): 80 mg omeprazole intravenous bolus + 8 mg/h control infusion until endoscopy
Placebo group (n = 319): placebo bolus + placebo infusion until endoscopy
Endoscopies were performed next morning.
Mean duration of infusion before endoscopy:

  • Omeprazole group 14.7 ± 6.3 h

  • Placebo group 15.2 ± 6.2 h

Primary:

  • Need for endoscopic therapy at the first endoscopic examination

Secondary:

  • Signs of bleeding

  • Need for urgent endoscopy

  • Emergency surgery

  • Recurrent bleeding

Among patients with peptic ulcer bleeding, endoscopic treatment required in:

  • Omeprazole group 22.5 %

  • Placebo group 36.8 %

  • OR 0.61, 95 %CI 0.44 – 0.84; P = 0.002

Active ulcer bleeding seen less frequently in omeprazole group than in placebo group: 6.4 % vs. 14.7 %; P = 0.001.
Rebleedings: 3.5 % in omeprazole group vs. 2.5 % in placebo group; P = 0.49

Long-term aspirin users were excluded

Pre-emptive omeprazole appears to accelerate the resolution of signs of bleeding.
Fewer cases of actively bleeding ulcers were seen among patients receiving omeprazole than among those who received placebo.
In patients awaiting endoscopy pre-emptive use of high dose intravenous omeprazole is recommended.

Liu, 2012 [239]

Randomized, single-center, prospective, double-blind

PPI parenteral (either omeprazole or esomeprazole) started prior to endoscopy (“early” endoscopy ≤ 24 h, “late” > 24 – 72 h)
Intensive regimen, 80 mg + 8 mg/h infusion: 410 patients
Standard regimen, 2 × 40 mg PPI/day: 456 patients

Rebleeding rate
Need for endoscopic therapy at first endoscopy

Rebleeding:
11 % in standard regimen group vs. 6.4 % in intensive regimen group (P = 0.03)

Early endoscopy therapy need:
16.7 % vs. 10.0 % (P = 0.05)

Rebleeding rates at late endoscopies:
7.5 % in standard vs. 4.0 % in intensive PPI group (P = 0.03)

Both omeprazole and esomeprazole were used
Endoscopic therapy was not standardized
No detailed data about the timing of endoscopy (how many hours after the initiation of pre-emptive PPI therapy)

High dose intravenous PPI initiated before endoscopy reduces rebleeding rates, blood transfusion volume and hospital stay, especially when endoscopy is delayed beyond 24 h of presentation.
No alteration on need for endoscopic treatment.

Barkun, 2010 [240]

International Consensus Guideline

NA
Patients receiving pre-emptive PPI before endoscopy.

Rebleeding
Need for surgery
Mortality
Need for intervention
Supportive cost-effective analysis

Pre-endoscopic PPI treatment significantly reduced the proportion of patients with high risk stigmata (OR 0.67, CI 0.54 – 0.84), and the need for pre-endoscopic therapy (OR 0.68, CI 0.50 – 0.93) compared with the control group (placebo or H2RA).
Economic dominance of pre-endoscopic high dose intravenous PPI therapy.

NA

Pre-endoscopic PPI therapy may be considered in order to downstage the endoscopic lesion and decrease the need for endoscopic intervention but should not delay endoscopy.
Comment: ESGE Board endorsed the consensus recommendation.

Tsoi, 2008 [241]

Decision analysis model
Cost-effectiveness comparison

Omeprazole group (80 mg bolus intravenous + 8 mg/h infusion):
n = 314 patients
187 peptic ulcer cases

  • initiated prior to endoscopy

Placebo group:
n = 317 patients
190 peptic ulcer cases

  • initiated prior to endoscopy

Number of patients who avoided endoscopic therapy within the follow-up period
Direct costs of medical treatment, diagnostic endoscopy, hemostasis, emergency surgery, hospitalization

248 patients in the PPI group and 227 patients in the placebo group avoided endoscopic therapy.
Overall direct costs: 2813 US dollars (USD) in the PPI group vs. 2948 USD in the placebo group
Costs were reduced by 7.4 % with pre-emptive PPI therapy

The estimated values were based on an assumed linear relationship

The use of high dose PPI before an endoscopy is shown to be an effective and cost-saving way to treat patients with upper GI bleeding.

Barkun, 2008 [242]

Editorial

NA

Key questions:
To contrast the pre-endoscopic with post-endoscopic hemostatic uses of PPI (in high dose intravenous administration) in patients with peptic ulcer bleeding
Almost no data about the time elapsed until endoscopy (i. e., the duration of intravenous PPI administration before gastroscopy)
What is the optimal duration of pre-emptive high dose intravenous PPI therapy regarding cost-effectiveness
Which subgroups of ulcer patients benefits most from pre-endoscopy PPI therapy

NA

NA

The earlier the endoscopy is performed, the less the cost-effectiveness of high dose intravenous PPI.
Pre-emptive PPI use should not replace the performance of early endoscopy.

Rácz, 2012 [243]

Single-center, retrospective, comparative cohort study

240 PUB patients, pantoprazole bolus 80 mg intravenous + 8 mg/h pantoprazole infusion before endoscopy
Control group: 93 PUB patients received saline infusion until endoscopy

Active ulcer bleeding at the first endoscopy
Need for endoscopic hemostasis at the first endoscopy
Need for urgent endoscopy
All outcomes were analyzed in subgroups of patients with different durations of intravenous pantoprazole before endoscopy.
Rebleeding rates
Separate analyses in duodenal ulcer and gastric ulcer patients

Mean duration of infusion before endoscopy:
5.4 ± 12.9 h in the saline group vs. 6.9 ± 13.2 h in the pantoprazole group (P = 0.29).
Active bleeding at first endoscopy:
19.2 % in the pantoprazole group vs. 24.7 % in the saline infusion group (P = 0.26).
Endoscopic treatment requirement at first endoscopy:
61.3 % in the pantoprazole group vs. 56.9 % in the saline group (P = 0.82)

Retrospective study
Aspirin, NSAIDs, and anticoagulants were discontinued at time of hospital admission.
Only data from patients admitted during evening hours (6pm to 8am) were analyzed

Pre-emptive infusion of high dose pantoprazole longer than 4 h before endoscopy decreased the ratio of active bleeding only in gastric and not in duodenal ulcer patients.

Lanas, 2013 [244]

Summary of main studies

NA

 – 

 – 

 – 

It has been reported that the administration of a PPI prior to endoscopy or the early performance of endoscopy within 6 h of admission in patients with UGIB does not improve the prognosis of the event.

Sung, 2011 [245]

Consensus report

NA

NA

 – 

 – 

A pre-endoscopy proton pump inhibitor (PPI) is recommended as a stopgap treatment when endoscopy within 24 h is not available.

Al-Sabah, 2008 [246]

Decision model compared high dose PPI initiated while awaiting endoscopy with administration on the basis of endoscopic findings

Cost-effectiveness

 – 

 – 

Intravenous PPIs given before endoscopy are slightly more effective than no administration.

Ghassemi. 2009 [247]

Overview

NA

NA

NA

 – 

Intravenous PPI therapy before endoscopy seems reasonable in patients presenting with severe upper gastrointestinal bleeding suspected from a peptic ulcer, if a delay is anticipated in urgent endoscopy.

Laursen, 2012 [248]

Guideline approved by the Society of Danish Society for Gastroenterology

NA

NA

NA

 – 

Treatment with PPI prior to endoscopy cannot be recommended and must not delay the timing of upper endoscopy.

Lin, 2010 [249]

Overview

NA

NA

NA

 – 

There was no evidence that PPI before endoscopy improves clinical outcomes.

ESGE, European Society of Gastrointestinal Endoscopy; H2RA, histamine-2 receptor antagonist; trial; NA, not available; NSAID, nonsteroidal anti-inflammatory drug; PPI, proton pump inhibitor; PUB, peptic ulcer bleeding; RCT, randomized controlled trial; UGIB, upper gastrointestinal bleeding.

Appendix e4

Role of tranexamic acid (TXA) in upper gastrointestinal hemorrhage.

First author, year [ref.]

Study type

Study population, Interventions

Key outcomes

Key results

Limitations

Conclusion

Gluud, 2012 [250]

Cochrane meta-analysis of 7 RCTs vs. placebo
1 RCT included also a treatment arm with cimetidine (Ref. 5)
1 RCT included also a treatment arm with lansoprazole and lansoprazole + TXA

Patients admitted with suspected upper gastrointestinal bleeding confirmed by gastric lavage, hematemesis or melena
Interventions:

  • Oral TXA administration (3 studies;

  • Intravenous TXA administration (4 studies.

Treatment duration (range): 2 – 7 days
Total daily TXA dose, range: 4 – 8 g (divided in four to six daily doses)
1 RCT offered endoscopic treatment to all randomized patients

  • Mortality (1654 patients, 7 studies)

  • Rebleeding or continued bleeding (1604 patients, 6 studies)

  • Surgery (1504 patients, 6 studies)

  • Need for transfusion (1504 patients, 6 studies)

  • Any thromboembolic event (1048 patients, 3 studies

TXA versus placebo:
Significant difference in mortality: 5 % vs. 8.2 %; RR 0.61 (95 %CI 0.42 – 0.89)
No significant difference in a subgroup analysis stratified for quality of bias control (trials with adequate allocation sequence generation or allocation concealment included): RR 0.78, 95 %CI 0.58 – 1.05)
No significant difference in:

  • Rebleeding: RR 0.73, 95 %CI 0.5 – 1.07

  • Surgery: RR 0.62, 95 %CI 0.35 – 1.09

  • Need for transfusion: 1.02, 95 %CI 0.93 – 1.11

  • Any thromboembolic event: RR 1.86, 95 %CI 0.66 – 5.24

TXA versus cimetidine or lansoprazole:
No significant difference in all outcome variables

Methodological quality of studies included (unclear or high risk of bias of selection bias in many of them)
Scarce applicability of evidence (most trials do not include the currently recommended intervention for management of NVUGIB)

TXA cannot be recommended for routine use.
Additional trials in which TXA is used in combination with the currently recommended interventions are required.

CI, confidence interval; NVUGIB, nonvariceal upper gastrointestinal bleeding; RCT, randomized controlled trial; RR, relative risk.

Appendix e5

Role of somatostatin in acute nonvariceal upper gastrointestinal hemorrhage (NVUGIH).

First author, year [ref.]

Study type

Patient group

Key outcomes

Key results

Limitation

Conclusion

Magnusson, 1985 [251]

Randomized, double‐blind

95 patients with acute nonvariceal GI bleeds with “massive” bleeding (clinical signs of shock or preshock)

  • 46 patients: 72 h somatostatin infusion

  • 49 patients: placebo

Need for surgical treatment (indication: > 6 units PRBC needed to keep hemodynamically stable
Evidence of active bleeding 1 day after treatment

Patients who needed surgery (P < 0.04):

  • 14 placebo

  • 5 somatostatin

P < 0.04

Rebleeding:

  • 5 placebo

  • 6 somatostatin

Only included patients with signs of shock
Endoscopic treatment was not standardized

Somatostatin infusion was superior to placebo regarding surgical need in patients with nonvariceal upper gastrointestinal bleeding.

Choi, 2011 [252]

Retrospective analysis of a prospective database

101 patients with acute ulcer bleeding, high risk stigmata (FIa,Ib, IIa)
Endoscopic therapy in all patients

  • 52 patients: pantoprazole 80-mg bolus + continuous infusion 8 mg/h for 72 h

  • 49 patients: pantoprazole as above, and also somatostatin as 250-µg bolus + continuous infusion 250 µg/h for 72 h

Clinically significant early rebleeding
Loss of endoscopic high risk stigmata at second-look endoscopy

Rebleeding rates:

  • PPI: 12.2 %

  • 14.3 % PPI + somatostatin

P = 0.766
Loss of stigmata:

  • 94.2 % PPI

  • 95.9 % PPI + somatostatin

P = 0.696

Not randomized study

Adjunctive somatostatin for management of nonvariceal upper gastrointestinal bleeding did not show an additive effect in reducing early rebleeding.

Avgerinos, 2005 [253]

Randomized, double‐blind

Adult patients admitted within 24 h of bleeding
Endoscopic stage IIc and III peptic ulcer bleeding
Not on PPI or H2 blockers in the previous week
No hypovolemia
Normal platelets, no coagulopathy
14 somatostatin
14 PPI
15 placebo

Gastric pH compared with patient baseline gastric pH during drug infusion
Intragastric pH > 4 in the fundus during the 24-h infusion period

Successful maintenance of pH > 4.0 in patients having somatostatin (P < 0.0001) and PPI (P < 0.0001)
During first half of treatment, time with above pH 4.0 and 5.4, respectively, was higher with somatostatin than PPI (P < 0.005) and (P < 0.02)

186 patients screened; 143 excluded
Patients with stigmata of recent hemorrhage were excluded
Evaluation of rebleeding was only a secondary end point

During the first 12 h of the infusion somatostatin was more effective than pantoprazole maintaining high intragastric pH.

Archimandritis, 2000 [254]

Prospective, randomized, open

84 patients with acute nonvariceal upper GI bleed

  • 44 ranitidine

  • 40 ranitidine + octreotide

Need for surgical intervention
Hospital stay length
Blood units transfused

No difference between groups for:

  • Hospital stay length (P = 0.25)

  • Amount of blood units (P = 0.16)

Need for emergency surgery did not differ (P = 1.0)

Small numbers
Not blinded

Lin, 1995 [255]

Randomized, open

Patients with active peptic ulcer bleeding, or Nonbleeding visible vessel at ulcer bases

  • 42 octreotide

  • 42 ranitidine

Hemostasis
Blood units transfused

Hemostasis achieved in 35/42 (83.3 %) octreotide group vs. 23/42 (54.8 %) in ranitidine group; (P < 0.01)
Octreotide group had:

  • Lower volume of blood (P < 0.05)

  • Fewer patients needing endoscopic hemostasis/surgery (P < 0.05)

  • Fewer days in hospital (P < 0.001)

Not blinded

Kim, 2008 [256]

Retrospective analysis

Patients with peptic ulcer bleeding, Forrest Ia, b, II a,b
Group A: 45 patients; 48 h pantoprazole infusion
Group B: 45 patients; 48 h pantoprazole + 250 µg/h infusion of somatostatin

Rebleeding in:

  • 72 h, and

  • 30 days

72 h rebleeding:

  • PPI alone: 11 %

  • PPI + somatostatin: 13 %

Not significant
30-day rebleeding:

  • 13 % vs. 16 %

Not significant

Not randomized
Endoscopic treatment was not standardized

Combined therapy with PPI and somatostatin did not result in better outcomes than PPI alone.

Antonioli, 1986 [257]

Multicenter, randomized, prospective, controlled trial

56 PUB patients
Group A: 250 µg/h infusion somatostatin for 48 h
Group B: 1600 µg/24 h cimetidine for 48 h after endoscopy

Secondary hemostasis
Transfusion requirement

Hemostasis:

  • Somatostatin group: 93.3 %

  • Cimetidine group: 61.5 %

P < 0.01

Blood requirement:

  • Somatostatin: 1.14 PRBC

  • Cimetidine: 2.46 PRBC

P < 0.05

Small patient numbers
Rebleeding rate was not defined

Somatostatin infusion for 48 h is superior to cimetidine infusion regarding definitive hemostasis.

Tisbouris, 2007 [258]

Randomized, double-blind, controlled trial

Patients with peptic ulcer bleeding, Forrest Ia, Ib, Iia
Group P: 82 patients, 8 mg/h pantoprazole infusion for 48 h
Group S: 82 patients, 250 µg somatostatin infusion for 48 h

Rebleeding 3 days after initial hemostasis
Need for endoscopic re-treatment

Rebleeding and need for endoscopic re-treatment:

  • Group P: 5 % vs.

  • Group S: 17 %

(P = 0.046)

Achieved pH > 6 for more than 85 % of the time:

  • Group P: 56.7 % vs.

  • Group S: 46.7 %

(P = 0.44)

Endoscopic treatment was not standardized
Not placebo-controlled

Infusion of pantoprazole after initial hemostasis was superior to somatostatin infusion to prevent bleeding recurrence
No difference in the need for surgery and in mortality.

Okan, 2000 [259]

Prospective, randomized, double-blind controlled study

Total of 48 patients with upper gastrointestinal bleeding
Group I: 15 Forrest Ib patients
Group II: 30 Forrest II patients
Randomly administered:

  • Somatostatin 250 µg bolus + continuous infusion 6 mg/day for 72 h, or

  • Ranitidine 300 mg/day continuous infusion for 72 h

Blood transfusion requirement
Time to bleeding cessation
Rebleeding rate

Transfusion requirement: no sign. difference
In group I (Forrest Ib patients), time to bleeding cessation was significantly shorter in those receiving somatostatin vs. those receiving ranitidine (3.24 vs. 11.25 h, P = 0.038)
No differences in rebleeding rate or mortality

Low patient numbers

Somatostatin is more effective than ranitidine in controlling acute nonvariceal upper gastrointestinal bleeding in patients with Forrest Ib bleeding activity.
No additional benefit with Forrest II bleeding activity.

Rutgeerts, 2006 [260]

Double-blind randomized controlled trial

369 PUB patients
Before endoscopy, randomized for intravenous treatment:

  • S group: 250 µg bolus + 12 mg/day somatostatin for 72 h

  • P group: placebo infusion for 72 h

Diagnostic endoscopy within 8 h of randomization

Failure rate (Forrest Ia or Ib at diagnostic endoscopy, clinical signs of active bleeding ≥ 1 h after the start of infusion
Use of rescue therapy (endoscopic treatment, surgery, vasoactive drugs) to stop bleeding

Failure rates in S and P groups:

  • No statistically significant difference, 34 % vs. 36 %

Stigmata of bleeding:

  • Statistically significant effect in favor of somatostatin

  • P = 0.034
    Rescue treatment:

  • Significantly less in S vs. P group

P = 0.012

Unusual presentation of results
“Failure rate” is not an established measure of results in studies

The results of this multicenter large trial were published only in abstract form. However, the results appear to support the use of intravenous somatostatin administered early and prior to diagnostic endoscopy and endotherapy to control bleeding in patients with suspected acute and severe PUB.

PPI, proton pump inhibitor; PRBC, packed red blood cells; PUB, peptic ulcer bleeding.

Appendix e6

Role of prokinetic agents in acute overt upper gastrointestinal hemorrhage.

First author, year [ref.]

Study type

Patient group

Key outcomes

Key results

Limitation

Conclusion

Carbonell, 2006, [261]

Prospective, randomized, double-blind

Upper gastrointestinal bleeding patients, all with nasogastric tube

  • 49 patients, 250 mg erythromycin prior to endoscopy

  • 50 patients, placebo prior to endoscopy

Improvement of gastric cleansing prior to endoscopy
Identification of bleeding point
Effectiveness of hemostasis treatment

  • Better visualization of entire gastric mucosa in erythromycin patients: 65 % vs. 44 %

P < 0.05

  • Clots in the stomach: 30 % erythromycin vs. 52 % placebo

P < 0.05

  • Hemostatic treatment: 67 % erythromycin vs. 62 % placebo

P = 0.67

Low patient numbers
Subjective criteria based on the endoscopist’s judgment

Intravenous erythromycin before endoscopy improves stomach cleansing and the quality of endoscopic examination.
Limited clinical benefit.

Coffin, 2002 [262]

Prospective, randomized, endoscopist-blinded, controlled trial

Acute upper gastrointestinal bleeding within 12 h requiring esophagogastro-duodenoscopy (EGD)
Before EGD patients were given:

  • 3 mg/kg erythromycin in 125 mL saline as a 30-minute infusion, or

  • Observation without motility agent

EGD within 60 – 120 min following erythromycin infusion
Erythromycin group: 19 patients
Control group: 22 patients

Quality of EGD examination evaluated using a score
Need for second EGD
Effectiveness of endoscopy therapy

Significantly better visualization during EGD in the erythromycin group (P = 0.02)
No interference with the hemostatic procedure
Need for second endoscopy: erythromycin:15.8 % vs. control:45.4 % (P = 0.089)

Low patient numbers
Qualitative evaluation of results (by scoring system)

Erythromycin infusion before endoscopy significantly improved the quality of EGD.
Tendency to reduce the need for second-look endoscopy.

Frossard, 2002 [263]

Prospective, randomized, double-blind, monocentric

  • Erythromycin intravenous 250 mg: 51 patients

  • Placebo: 54 patients

20 minutes before endoscopy
Within 12 h after hematemesis

Effect of intravenous erythromycin on endoscopic yield
Need for second-look endoscopy
Transfusion requirement

Clear stomach found more often in erythromycin group: 82 % vs. 33 %, P < 0.001
Erythromycin shortened the endoscopy duration and the need for second-look endoscopy

Low patient number with nonvariceal upper gastrointestinal bleeding

Erythromycin infusion before endoscopy in patients with recent hematemesis makes endoscopy shorter and easier, and reduces the need for second-look endoscopy.

Theivanayagam, 2013 [264]

Meta-analysis of 6 studies

558 patients with upper gastrointestinal bleeding (patients with variceal bleeds were also included)

Visualization of gastric mucosa
Need for second endoscopy

Erythromycin infusion before endoscopy significantly improved visualization vs. no erythromycin:
OR 3.43, 95 %CI 1.81 – 6.50
Significant decrease in the need for second endoscopy (P = 0.01)

Inclusion of variceal bleedings

Erythromycin infusion before endoscopy in patients with upper gastrointestinal bleeding significantly improves visualization of gastric mucosa and decreases the need for a second endoscopy.

Sussman, 2008 [265]

Single-center, prospective, randomized, controlled study
Intravenous metoclopramide bolus (10 mg intravenous)

Patients with overt hematemesis or melena
A total of 26 patients

Mucosal visualization during EGD (Avgerinos score)
Duration of EGD
Rebleeding rate

Better visualization after metoclopramide, P = not significant
No significant difference regarding blood transfusion, repeat EGD

Low patient numbers
Subjective quality score

Intravenous metoclopramide may increase visualization of the proximal stomach during EGD, but not significantly.
No translation into better clinical outcome.

Barkun, 2010 [266]

Meta-analysis
3 fully published articles, 2 abstracts

3 erythromycin studies
2 metoclopramide studies
316 patients

Primary:

  • Need for repeat EGD

Secondary:

  • Blood transfusion

  • Need for surgery

A prokinetic agent significantly reduced the need for repeat EGD:
OR 0.55, 95 %CI 0.32 – 0.94
No significant alterations:

  • Blood transfusion, and

  • Need for surgery:

Small numbers of patients

Intravenous erythromycin or metoclopramide immediately before EGD in patients with acute upper gastrointestinal bleeding decreases the need for repeat EGD, but does not improve clinical outcomes.

Habashi, 2007 [267]

Prospective, randomized, controlled trial

54 patients with hematemesis (42 % PUB patients)

  • Intravenous erythromycin: 15 patients

  • Intravenous metoclopramide: 15 patients

  • Placebo: 15 patients

Visualization of the mucosa
Quality of the EGD

Gastric mucosa entirely visualized:

  • erythromycin group: 86 %, vs.

  • metoclopramide group: 66 %

Quality of EGD significantly better in erythromycin group vs. metoclopramide and placebo groups

Small sample sizes
Subjective scores
Dosage of prokinetics not reported

No significant difference between placebo and either medication regarding key outcomes.

Winstead, 2007 [268]

Cost-effectiveness analysis

3 RCTs were analyzed with a total of 126 patients
(Frossard, Coffin and Carbonell studies)

Quality-adjusted life-years (QALYs)
Need for second-look endoscopy

Intravenous erythromycin before endoscopy was cost-effective when the rate of second-look endoscopy was less than 0.29
Intravenous erythromycin was cost-effective when the charges for uncomplicated peptic ulcer disease PUD were less than 8000 US dollars

The estimates need for repeat endoscopy.
The complex model may fail to capture all the slight variations in patterns of care among different hospitals.

Intravenous erythromycin before endoscopy in acute upper gastrointestinal hemorrhage (UGIH) is cost-saving and increases QALYs.
Erythromycin is recommended prior to EGD in UGIH.

CI, confidence interval; EGD, esophagogastroduodenoscopy; PUB, peptic ulcer bleeding; OR, odds ratio; RCT, randomized controlled trial.

Appendix e7

Summary of the evidence regarding impact of early endoscopy (≤ 24 h) on the outcome of patients with nonvariceal upper gastrointestinal hemorrhage (NVUGIH).

First author, year [ref.]

Country

Study type,
Study period

Patients, n

Major findings

Spiegel, 2001 [269]

Systematic review
1980 – 2000
23 studies

  • 6 controlled in low risk patients

  • 7 uncontrolled in low risk patients

  • 6 controlled in high risk patients

  • 4 comparing resource utilization

12 625

Early endoscopy ( ≤ 24 h) safe and effective in all risk groups.
Low risk: allows safe and prompt discharge.
High risk: significantly reduces recurrent bleeding, transfusion requirements, need for surgery and length of hospital stay.

Tsoi, 2009 [270]

Systematic review
1996 – 2007
8 studies

  • 3 RCT

  • 5 retrospective

5 677

Early endoscopy aids risk stratification and reduces the need for hospitalization; however it may increase the use of unnecessary therapeutic procedures.
Endoscopy performed ≤ 8 h of presentation has no advantage over endoscopy performed within 12 – 24 h of presentation in reducing recurrent bleeding or improving survival.

Sarin, 2009 [271]

Canada

Retrospective
2004 – 2006

502

No advantage for early endoscopy (< 6 h) compared with endoscopy within 24 h in terms of mortality, need for surgery, or transfusion requirements.

Lim, 2011 [272]

Singapore

Retrospective

837 low risk
97 high risk

Endoscopy within 13 h of presentation is associated with lower mortality in high risk but not low-risk patients with NVUGIH.

Marmo, 2011 [273]

Italy

Multicenter, prospective cohort studies (3 databases)
2004 – 2009

3 207

Significant increase of mortality in high risk patients when endoscopy is performed ≤ 12 h compared with endoscopy performed 13 – 24 h after presentation (14.3 % – 16.6 % vs. 5.2 %, P = 0.001).

Wysocki, 2012 [274]

US

Retrospective
Administrative data
NIS 2002 – 2007

435 765

Increased mortality risk in patients who do not receive endoscopy within 1 day of admission: OR 1.32, 95 %CI 1.26 – 1.38.

Jairath, 2012 [275]

UK

Multicenter, prospective cohort study
2007

4 478

Compared with later endoscopy (> 24 to 48 h), endoscopy performed ≤ 12 h did not affect mortality (OR 0.98, 95 %CI 0.88 – 1.09), but led to a decreased risk-adjusted length of hospital stay (1.7 days, 95 %CI 1.39 – 1.99).

CI, confidence interval; OR, odds ratio; RCT, randomized controlled trial.

Appendix e8

Medical management following endoscopic hemostasis

First author, year [ref.]

Study design

Intervention

Participants

Outcomes

Results

Level of evidence, conclusions

Barkun, 2010 [240]

International consensus guideline

After endoscopic therapy:

  • Proton pump inhibitor (PPI) vs.

  • Placebo or H2RA

Patients with endoscopic signs of high risk of rebleeding

Mortality
Rebleeding 3 – 7 days

Rebleeding: OR 0.45, CI 0.36 – 0.57
Surgery: OR 0.56, CI 0.45 – 0.70
Mortality: OR 0.90 CI 0.67 – 1.19

Intravenous PPI 80 mg bolus + 8 mg/h for 72 h recommended.
Evidence: High

Sachar, 2014 [276]

Systematic review

After endoscopic therapy:

  • PPI intravenous continuous infusion (80 mg + 8 mg/h for 72 h) vs.

  • Intermittent PPI

Patients with endoscopic signs of high risk of rebleeding

Rebleeding 7 days
Mortality

Rebleeding: risk ratio (RR) 0.72 (upper boundary of 1-sided 95 %CI 0.97).
RRs for rebleeding 3 – 30 days, mortality, urgent interventions were less than 1.

No differences between intermittent high dose PPI (all routes) vs. intravenous PPI 80 mg bolus + 8 mg/h for 72 h.
Evidence: High

Sung, 2014 [277]

Randomized, controlled trial, double blind

After endoscopic therapy:

  • Intravenous esomeprazole bolus 80 mg + 8 mg/h for 72 h vs.

  • Oral esomeprazole 40 mg/12 h

Patients with endoscopic signs of high risk of rebleeding

Rebleeding at 30 days

Intravenous esomeprazole group: 118 patients
Oral esomeprazole group: 126 patients
Recurrent bleeding:

  • Intravenous group: 7.7 %, vs.

  • Oral group: 6.4 %

High dose oral esomeprazole may be considered as a useful alternative to intravenous therapy.
Evidence: Moderate

Mostaghni, 2011 [278]

Randomized, open-label

  • Oral omeprazole (80 mg twice-daily for 3 days), or

  • Intravenous pantoprazole (80 mg bolus and 8 mg/hour infusion for 3 days)
    Followed in all patients by omeprazole (20 mg each day for 30 days).

Patients with endoscopic signs of high risk of rebleeding

Rebleeding (30 days?; timing not well-defined)

  • Oral omeprazole: 44 patients

  • Intravenous pantoprazole: 41 patients

Rebleeding:

  • Oral omeprazole: 11.4 %, vs.

  • Intravenous pantoprazole: 9.8 %

Mean hospital stay and blood transfusion were not different between the two groups.

Oral omeprazole and intravenous pantoprazole had equal effects on prevention of rebleeding after endoscopic therapy in patients with high risk bleeding peptic ulcers.
Evidence: Low

Yen, 2012 [279]

Randomized, open-label

Esomeprazole group: 40 mg continuous infusion of esomeprazole every 6 h for 3 days. Then oral esomeprazole 40 mg once-daily for 2 months.
Lansoprazole group: oral 30 mg four times daily for 3 days followed by once daily for 2 months.

Patients with endoscopic signs of high risk of rebleeding

Rebleeding rate within 14 days.
Secondary outcomes included:

  • Hospital stay

  • Volume of blood transfusion

  • Surgical intervention, and

  • Mortality within 1 month

100 patients enrolled.
Rebleeding rates:

  • esomeprazole intravenous group: 4 % (2 /50), and

  • Lansoprazole oral group: 4 % (2/50)

No difference between the two groups with regard to:

  • Hospital stay

  • Volume of blood transfusion

  • Surgery, or

  • Mortality rate

There is no evidence of a difference in clinical outcomes between oral and intravenous PPI treatment. (not powered to prove equivalence or noninferiority). Patients receiving oral PPI have a shorter hospital stay.
Evidence: Low

Tsai, 2008 [280]

Randomized, open-label

  • Oral rabeprazole (20 mg twice-daily for 3 days), or

  • Intravenous omeprazole (40 mg intravenous infusion every 12 h for 3 days).

Patients with endoscopic signs of high risk of rebleeding

Bleeding up to 14 days.
Also compared:

  • Hospital stay

  • Volume of blood transfusion

  • Surgery, and

  • Mortality within 14 days

156 patients with 78 in each group
Rebleeding:

  • Omeprazole group: 12 patients (15.4 %)

  • Rabeprazole group: 13 patients (16.7 %)

  • 95 %CI of difference: 12.82 – 10.22

No differences in the other evaluated outcomes

Oral rabeprazole and intravenous regular-dose omeprazole are equally effective in preventing rebleeding in patients with high risk bleeding peptic ulcers after successful endoscopic injection with epinephrine.
Evidence: Low

Wang, 2010 [281]

Systematic review

High dose PPIs (80 mg bolus + 8 mg/h for 3 days vs.
Non-high dose PPIs (40 mg160 mg/day for 3 days)

Patients with bleeding peptic ulcer

  • Rebleeding

  • Surgical intervention, and

  • Mortality

7 studies and 1157 patients

  • Rebleeding: OR 1.30, 95 %CI 0.88 – 1.91

  • Surgical intervention: OR 1.49, 0.66 – 3.37

  • Mortality: OR 0.89, 0.37 – 2.13

Compared with non – high dose PPIs, high dose PPIs do not further reduce the rates of rebleeding, surgical intervention, or mortality after endoscopic treatment.
Evidence: High

Masjedizadeh, 2014 [282]

Randomized, open-label

  • High dose pantoprazole (80 mg bolus, 8 mg per hour) infused for 3 days vs.

  • Low dose pantoprazole (40 mg bolus, 4 mg per hour) infused for 3 days

Patients with endoscopic signs of high risk of rebleeding

Rebleeding at 30 days

166 patients (83 patients per group)
Rebleeding observed in:

  • High dose group: 27 patients (32.53 %)

  • Low dose group: 21 patients (25.30 %)

P = 0.30

For controlling peptic ulcer bleeding, there is no difference between high dose and low dose pantoprazole infusion.
Evidence: Low

CI, confidence interval; H2RA, histamine-2 receptor antagonist; OR, odds ratio; PPI, proton pump inhibitor; RR, risk ratio.

Appendix e9

Salvage therapy in failed endoscopic hemostasis.

First author, year [ref.]

Study type

Patient group

Key outcomes

Key results

Limitations

Level of evidence, Conclusion

Lau, 1999 [283]

Prospective, randomized trial
Comparison between immediate endoscopic re-treatment vs. surgery

Endoscopic re-treatment with the same device (epinephrine injection + heater probe):

  • n = 48 patients

Surgery:

  • n = 44

Definitive hemostasis
Complications
Surgery
Mortality

Definitive hemostasis: 35 /48

Salvage surgery: 13

  • Persistent bleeding: 11

  • Perforation: 2

Complications:
7 patients in the endoscopy group (including 6 who underwent salvage surgery) had complications, as compared with 16 in the surgery group (P = 0.03)
7
Mortality: n.s.
Hypotension at randomization and ulcer > 2 cm were independent predictors of re-treatment failure

Very high expertise center
Not blinded
2 perforations related to re-treatment with heater probe

High quality
In patients with peptic ulcers and rebleeding after initial hemostatic success, endoscopic re-treatment reduces the need for surgery without increasing risk of death and is associated with fewer complications than surgery.

Wong, 2011 [284]

Retrospective

Transcatheterangiographic embolization (TAE): n = 23
Surgery: n = 56

Definitive hemostasis
Complications
Mortality

Bleeding recurrence:

  • TAE group 34.4 %

  • Surgery group 12.5 %

P = 0.1
More complications after surgery: 67.9 % vs. 40.6 %, P = 0.1
No difference in mortality: 25 % vs. 30.4 %

Results of initial failure and rebleeding are mixed

Moderate quality
In patients with ulcer bleeding after failed endoscopic hemostasis, TAE reduces the need for surgery without increasing the overall mortality and is associated with fewer complications.

Kyaw, 2014 [285]

Meta-analysis
6 retrospective studies that involved 423 patients

TAE patients were older, mean age:

  • TAE: 75 years vs.

  • Surgery: 68 years

Definitive hemostasis
Complications
Mortality

Risk of rebleeding was significantly higher in TAE vs. surgery:
RR 1.82, 95 %CI 1.23 – 2.67
After age exclusion, the high risk of rebleeding remained in the TAE group:
RR 2.64, 95 %CI 1.48 – 4.71
No difference in:

  • Mortality, or

  • Requirement for additional interventions

Retrospective studies

High quality
A higher rebleeding rate was observed after TAE, suggesting surgery more definitively secured hemostasis, with no significant differences in mortality rate or requirement for additional interventions.

Beggs, 2014 [286]

Systematic review (with homogeneity) of cohort studies

TAE: n = 347
Surgery: n = 364

Mortality
Rebleeding
Length of hospital stay
Adverse events

Deaths:

  • TAE: 61

  • Surgery: 101

Rebleeding:

  • TAE: 78

  • Surgery: 45

Length of hospital stay:

  • Mean difference between TAE and surgery: 0.75 days

Complications:

  • TAE: 92

  • Surgery: 81

Moderate quality
When compared with surgery, TAE had a significantly increased risk of rebleeding. However, there were no differences in mortality.

Smith, 2014 [287]

European register

Patients who otherwise might have required either surgery or TAE

n = 8

Definitive hemostasis

Definitive hemostasis:

  • n = 8 (100 %)

Small number of patients

Low quality
Hemospray may be useful as a rescue therapy.

Sulz, 2014 [288]

Prospective, case series

n = 16

Definitive hemostasis

Initial hemostasis: 15/16
Salvage therapy: 13/14
Monotherapy: 2/2
Rebleeding: 12.5 %

Small number of patients

Low quality
Hemospray may be useful as a salvage therapy.

Skinner, 2014 [289]

Retrospective

Over-the-scope clip after failure of conventional endoscopic treatment

n = 12
Cause of bleed:

  • Duodenal ulcer: 6

  • Gastric ulcer: 2

  • Dieulafoy lesion: 2

  • Anastomotic ulceration: 1

  • Mallory-Weiss: 1

  • Prior endoscopies

  • Mean of 2

  • Shock present:

  • n = 9 (75 %)

RBC:

  • Mean 5.1 units (2 – 12)

Primary hemostasis: 11/12
Rebleeding n = 2 (day1, day7)
No complication

Retrospective
Small number of patients
Expertise needed

Low quality
Over-the-scope clip effective and safe for severe acute gastrointestinal bleeding when conventional endoscopic therapy fails.

Manta, 2013 [290]

Retrospective

Over-the-scope clipping after failure of conventional endoscopic treatment

n = 23

Primary hemostasis: 22 (96 %)

Retrospective
Small number of patients
Expertise needed

Low quality
Over-the-scope clipping is effective and safe for severe acute gastrointestinal bleeding when conventional endoscopic therapy fails.

  • Duodenal ulcer: 12
    Forrest Ia n = 5
    Forrest Ib n = 4

  • Duodenal ulcer: 11 (1 rebleeding)

  • Gastric ulcer 6
    Forrest Ia: n = 2
    Forrest Ib: n = 2

 – Gastric ulcer: 6 (1 rebleeding)

  • Mallory-Weiss: n = 3

  • Mallory-Weiss: 2 (no rebleeding)

  • Dieulafoy: n = 2

  • Dieulafoy: 2 (no rebleeding)

  • Anastomosis: n = 1

  • Anastomosis1: (no rebleeding)

CI, confidence interval; NA, not available; NVUGIB, nonvariceal upper gastrointestinal bleeding; OR, odds ratio; RUT, rapid urease test; UBT, urea breath test.

Appendix e10

Helicobacter pylori and nonvariceal upper gastrointestinal hemorrhage (NVUGIH).

First author, year [ref.]

Study design, study objective

Participants

Outcomes

Results

Level of evidence, conclusions

Sánchez-Delgado, 2011 [291]

Meta-regression analysis

NVUGIB patients

H. pylori infection rate after the event

Delayed testing for H. pylori increases the detection rate (OR 2.08, 95 %CI 1.10 – 3.93)

Moderate

Gisbert, 2006 [292]

Meta-analysis

NVUGIB patients

H. pylori infection rate after the event

Low sensitivity of histology, RUT, culture, UBT, and serology

Moderate

Barkun, 2010 [240]

International consensus recommendations

NVUGIB patients

Re-bleeding

H. pylori therapy and eradication confirmation needed

High

Gisbert, 2012 [293]

Prospective study on 1000 patients

NVUGIB patients

Re-bleeding after H. pylori eradication

Rebleeding was 0.15 % per patient-year of follow-up and it was associated with either re-infection or NSAIDs use.

Moderate

Gisbert, 2004 [294]

Meta-analysis

NVUGIB patients

Re-bleeding after eradication

Rebleeding was significantly lower in the H. pylori eradication group than in antisecretory therapy group (1.6 % vs. 5.6 %)

High

Dixon, 1996 [295]

International workshop

Gastritis

NA

Presence of neutrophil histology strongly suggests H. pylori infection

Moderate

CI, confidence interval; NA, not available; NVUGIB, nonvariceal upper gastrointestinal bleeding; OR, odds ratio; RUT, rapid urease test; UBT, urea breath test.

Appendix e11

Risk of thromboembolism, recurrent gastrointestinal (GI) bleeding and death after warfarin therapy interruption for GI bleeding.

First author, year [ref.]

Study type

Study population
Intervention

Key outcomes

Key results

Limitations

Conclusion

Witt, (2012) [296]

Retrospective, cohort

442 patients with warfarin-associated GI bleeding
Intervention:
Withhold warfarin (no-warfarin group): 182 (41.2 %)
Resumed warfarin (warfarin group): 260 (58.8 %)

Thromboembolic events
Recurrent GI bleeding
Death

90-day thromboembolic event rate:

  • 0.4 % (1/260) in warfarin group

  • 5.5 % (10/182) in no-warfarin group

HR (95 %CI): 0.05 (0.001 – 0.58)

No thromboembolic events in patients who resumed therapy within 14 days

90-day recurrent GI bleeding rate:

  • Warfarin group: 10 % (26/260)

  • No-warfarin group: 5.5 % (10/182)

HR (95 %CI): 1.32 (0.50 – 3.57)

Higher risk of recurrent GI bleeding in patients who resumed warfarin within 7 days from index bleeding as compared with those who resumed warfarin later:

  • 12.4 % vs. 6.2 %

P = 0.03

90-day mortality rate:

  • Warfarin group: 5.8 % (15/260) in

  • No-warfarin group: 20.3 % (37/182)

HR (95 %CI): 0.31 (0.15 – 0.62)

Retrospective study
Data from administrative databases
Selection bias (greater co-morbidity burdens in no-warfarin group, which may have contributed to their worse outcomes)
Detection and survivorship biases

The decision to not resume warfarin therapy in the 90 days following a GI bleeding event is associated with increased risk for thrombosis and death.
Resuming warfarin within 7 days is associated with a twofold higher risk of rebleeding.

Qureshi, 2014 [297]

Retrospective, cohort study

1329 atrial fibrillation patients with warfarin-associated major GI bleeding
Intervention:
676 (50.9 %) withhold warfarin (no-warfarin group)
653 (49.1 %) resumed warfarin (warfarin group)
Time duration of interruption:

  • < 7 days: 62 patients

  • 7 – 30 days: 162 patients

  • 30 days: 429 patients

Thromboembolic events
Recurrent GI bleeding
Death

Warfarin group vs. no-warfarin group, adjusted HR (95 %CI):

  • Thromboembolism: 0.71 (054 – 0.93)

  • Recurrent GI bleeding; 1.20 (0.78 – 1.86)

  • Mortality: 0.72 (0.60 – 0.86)

Incidence of adverse outcomes per 100 person-years in the warfarin group, stratified by the duration of warfarin interruption:

  •  < 7 days (n = 62):
    Thromboembolism: 11.6 (8.3 – 16.2)
    Recurrent GI bleeding: 19.3 (14.6 – 25.5)

  • 7 – 15 days (n = 51):
    Thromboembolism: 12.0 (8.2 – 17.5)
    Recurrent GI bleeding: 10.8 (7.2 – 16.3)

  • 15 – 21 days (n = 58):
    Thromboembolism: 18.1 (13.4 – 24.5)
    Recurrent GI bleeding: 10.9 (7.2 – 16.4)

  • 21 – 30 days (n = 53):
    Thromboembolism 20.7 (15.5 – 27.7)
    Recurrent GI bleeding 9.9 (6.3 – 15.5)

  •  > 30 days (n = 429):
    Thromboembolism 20.4 (17.8 – 23.5)
    Recurrent GI bleeding 9.9 (8.0 – 12.3)

Retrospective study. Data from administrative database
Selection bias (greater co-morbidity burdens in no-warfarin group)
Detection and survivorship biases

The decision to not resume warfarin therapy after a GI bleeding event is associated with increased risk for thrombosis and death.
There is a trend toward reduced incidence of thromboembolic events the earlier the warfarin is introduced; this trend is more evident within the first 15 days.
Resuming warfarin within 7 days is associated with a twofold higher risk of rebleeding.
Decision to restart warfarin after 7 days of interruption is associated with improved survival and decreased thromboembolism without increased risk of recurrent GI bleeding.

Lee, 2011 [298]

Retrospective, case-control study

58 patients with NVUGIB on warfarin for native valvular heart disease
Controls (aspirin group):
41 age- and gender-matched patients with NVUGIB on aspirin for ischemic heart disease presenting
Intervention

Cases:

  • Discontinued warfarin (no-warfarin group): 36/58

  • Restarted warfarin (warfarin group): 22/58

Controls:

  • Discontinued aspirin: 41/41

Recurrent GI bleeding
Thromboembolic events
Mean follow-up: 255 + 14 days (range 182 – 330)

Warfarin vs. no-warfarin group:

  • Rebleeding rate: 7 % vs.0 %

  • Thromboembolism rate: 0 % vs. 17 % (6 thromboembolic events occurred, at days 21, 27, 28, 31, 58, 75 from admission)

Aspirin group:

  • Rebleeding rate: 0 %

  • Thromboembolic event rate: 2 % (1 myocardial infarction at day 95 from admission)

Retrospective design
Time of resuming anticoagulation not specified
Individual thromboembolic risk not specified

Anticoagulation is recommended to be resumed within 20 days from the cessation to prevent thromboembolic events.

CI, confidence interval; HR, hazard ratio; NVUGIB, nonvariceal upper gastrointestinal bleeding.

Appendix e12

Observational studies assessing the effect of proton pump inhibitors (PPIs) on clinical cardiovascular outcomes in patients prescribed clopidogrel.

First author, year [ref.]

Design

Population

Patients, n

End point

Results

PPI

No PPI

Studies with evidence of a clinically significant interaction

Goodman, 2012 [299]

Retrospective cohort within RCT (PLATO)

Acute coronary syndrome (ACS)

3 255

6 021

Cardiovascular death, myocardial infarction, cerebrovascular accident

Clopidogrel cohort: HR 1.20 (95 %CI 1.04 – 1.38)
Ticagrelor cohort: HR 1.24 (95 %CI, 1.07 – 1.45)

Stockl, 2010 [300]

Retrospective
Propensity matching

Post myocardial infarction or post percutaneous coronary intervention

1 033

1 033

Myocardial infarction
Myocardial infarction or revascularization

HR 1.93 (95 %CI, 1.05 – 3.54)
HR 1.91 (95 %CI 1.19 – 3.06)

Kreutz, 2010[301]

Retrospective cohort

Post percutaneous coronary intervention

6 828

9 862

Cardiovascular deaths, acute coronary syndrome, cerebrovascular accident revascularization

HR 1.51 (95 %CI, 1.39 – 1.64)

Ho, 2009 [302]

Retrospective cohort

Post myocardial infarction

5 244

2 961

Deaths, acute coronary syndrome

HR 1.25(95 %CI, 1.11 – 1.41)

Huang, 2010 [303]

Registry

Post percutaneous coronary intervention

572

2 706

Death

HR 1.65(95 % CI, 1.35 – 2.01)

Zou, 2014 [304]

Retrospective cohort

Post percutaneous coronary intervention

61 288

1 465

Myocardial infarction, stent thrombosis, cardiovascular deaths

HR 1.33 (95 % CI 1.12 – 1.57)

Van Boxel, 2010 [305]

Retrospective cohort

Clopidogrel users

5 734

12 405

Deaths, acute coronary syndrome, cerebrovascular accident

HR 1.75 (95 % CI, 1.58 – 1.94)

Munoz-Torrero, 2011 [306]

Registry

Vascular disease

519

703

Deaths, myocardial infarction, acute coronary syndrome, cerebrovascular accident, chronic limb ischemia

HR 1.8 (95 % CI, 1.1 – 2.7)

Studies without evidence of a clinically significant interaction

O’Donoghue, 2009 [307]

Retrospective cohort within RCT
(TRITON-TIMI 38)

Acute coronary syndrome and post percutaneous coronary intervention

2 257

4 538

Cardiovascular death, myocardial infarction, cerebrovascular accident

No effect

Hsiao, 2011 [308]

Retrospective
Propensity matching

Post myocardial infarction

622

9 131

Acute coronary syndrome

No effect

Banerjee, 2011 [309]

Retrospective
Propensity matching

Post percutaneous coronary intervention

867

3 678

Death, myocardial infarction, revascularization

No effect

Harjai, 2011 [310]

Registry
Propensity matching

Post percutaneous coronary intervention

751

1 900

Death, myocardial infarction, revascularization

No effect

Aihara, 2012 [311]

Registry
Propensity matching

Post percutaneous coronary intervention

819

1 068

Death, myocardial infarction

No effect

Tentzeris, 2010 [312]

Registry
Propensity matching

Post percutaneous coronary intervention

691

519

Death, acute coronary syndrome

No effect

Schmidt, 2012 [313]

Retrospective cohort

Post percutaneous coronary intervention

2 742

10 259

Cardiovascular deaths, acute coronary syndrome, cerebrovascular accident, revascularization

No effect

Rassen, 2009 [314]

Retrospective cohort

Post percutaneous coronary intervention or post acute coronary syndrome

3 996

14 569

Death, myocardial infarction, revascularization

No effect

Ray,
2010 [315]

Retrospective cohort

Post percutaneous coronary intervention or post acute coronary syndrome

7 593

13 003

Death, myocardial infarction, revascularization

No effect

CI, confidence interval; HR, hazard ratio; PLATO, Platelet Inhibition and Patient Outcomes; RCT, randomized controlled trial.

Appendix e13

Meta-analyses evaluating the effect of proton pump inhibitors (PPIs) on clinical outcomes in patients treated with clopidogrel.

First author, year [ref.]

Included studies

Patients, n

End point

Results

Kwok, 2010 [316]

1 nested case-control
20 retrospective
(3 studies used a propensity scoring method for the analysis)
3 post.hoc analyses of RCT
1 prospective RCT

93 278

Myocardial infarction or acute coronary syndrome (12 studies)

RR 1.43 (95 %CI 1.15 – 1.77)
RR 1.15 (0.89 – 1.48): analysis from propensity matched or trial participants

Overall mortality (13 studies)

RR 1.09 (95 %CI 0.94 – 1.53)
RR 1.00 (0.66 – 1.48): analysis from propensity matched or trial participants

Major adverse cardiovascular event (MACE)
(19 studies)

RR 1.25 (95 %CI 1.09 – 1.42)
RR 1.07 (0.90 – 1.48): analysis from propensity matched or trial participants

Siller-Matula, 2010 [317]

2 nested case-control
20 retrospective cohort
(3 studies used a propensity scoring method for the analysis)
2 post.hoc analyses of RCT
1 prospective RCT

159 138

Myocardial infarction
(13 studies)

RR 1.31 (95 %CI 1.12 – 1.53)

Death
(13 studies)

RR 1.04 (95 %CI 0.93 – 1.24)

MACE
(19 studies)

RR 1.29 (95 %CI 1.15 – 1.44)

CI, confidence interval; RCT, randomized controlled trial; RR, risk ratio


#
#
#

Competing interests: None

  • References

  • 1 van Leerdam ME. Epidemiology of acute upper gastrointestinal bleeding. Best Pract Res Clin Gastroenterol 2008; 22: 209-224
  • 2 Hearnshaw SA, Logan RF, Lowe D et al. Acute upper gastrointestinal bleeding in the UK: patient characteristics, diagnoses and outcomes in the 2007 UK audit. Gut 2011; 60: 1327-1335
  • 3 Guyatt GH, Oxman AD, Vist GE et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ 2008; 336: 924-926
  • 4 Dumonceau JM, Hassan C, Riphaus A et al. European Society of Gastroinestinal Endoscopy (ESGE) guideline development policy. Endoscopy 2012; 44: 626-629
  • 5 Baradarian R, Ramdhaney S, Chapalamadugu R et al. Early intensive resuscitation of patients with upper gastrointestinal bleeding decreases mortality. Am J Gastroenterol 2004; 99: 619-622
  • 6 Kwan I, Bunn F, Chinnock P et al. Timing and volume of fluid administration for patients with bleeding. Cochrane Database Syst Rev 2014; 3: CD002245
  • 7 Spahn DR, Bouillon B, Cerny V et al. Management of bleeding and coagulopathy following major trauma: an updated European guideline. Crit Care 2013; 17: R76
  • 8 Roberts I, Alderson P, Bunn F et al. Colloids versus crystalloids for fluid resuscitation in critically ill patients. Cochrane Database Syst Rev 2004; 4: CD000567
  • 9 Myburgh JA, Finfer S, Bellomo R et al. Hydroxyethyl starch or saline for fluid resuscitation in intensive care. N Engl J Med 2012; 367: 1901-1911
  • 10 Marik PE, Corwin HL. Efficacy of red blood cell transfusion in the critically ill: a systematic review of the literature. Crit Care Med 2008; 36: 2667-2674
  • 11 Restellini S, Kherad O, Jairath V et al. Red blood cell transfusion is associated with increased rebleeding in patients with nonvariceal upper gastrointestinal bleeding. Aliment Pharmacol Ther 2013; 37: 316-322
  • 12 Villanueva C, Colomo A, Bosch A et al. Transfusion strategies for acute upper gastrointestinal bleeding. N Engl J Med 2013; 368: 11-21
  • 13 Jairath V, Kahan BC, Stanworth SJ et al. Prevalence, management, and outcomes of patients with coagulopathy after acute nonvariceal upper gastrointestinal bleeding in the United Kingdom. Transfusion 2013; 53: 1069-1076
  • 14 Shingina A, Barkun AN, Razzaghi A et al. Systematic review: the presenting international normalized ratio (INR) as a predictor of outcome in patients with upper nonvariceal gastrointestinal bleeding. Aliment Pharmacol Ther 2011; 33: 1010-1018
  • 15 Karam O, Tucci M, Combescure C et al. Plasma transfusion strategies for critically ill patients. Cochrane Database Syst Rev 2013; 12: CD010654
  • 16 Razzaghi A, Barkun AN. Platelet transfusion threshold in patients with upper gastrointestinal bleeding: a systematic review. J Clin Gastroenterol 2012; 46: 482-486
  • 17 Rockall TA, Logan RF, Devlin HB et al. Risk assessment after acute upper gastrointestinal haemorrhage. Gut 1996; 38: 316-321
  • 18 Blatchford O, Murray WR, Blatchford M. A risk score to predict need for treatment for upper-gastrointestinal haemorrhage. Lancet 2000; 356: 1318-1321
  • 19 de Groot NL, Bosman JH, Siersema PD et al. Prediction scores in gastrointestinal bleeding: a systematic review and quantitative appraisal. Endoscopy 2012; 44: 731-739
  • 20 Lee JG, Turnipseed S, Romano PS et al. Endoscopy-based triage significantly reduces hospitalization rates and costs of treating upper GI bleeding: a randomized controlled trial. Gastrointest Endosc 1999; 50: 755-761
  • 21 Cipolletta L, Bianco MA, Rotondano G et al. Outpatient management for low-risk nonvariceal upper GI bleeding: a randomized controlled trial. Gastrointest Endosc 2002; 55: 1-5
  • 22 Brullet E, Campo R, Calvet X et al. A randomized study of the safety of outpatient care for patients with bleeding peptic ulcer treated by endoscopic injection. Gastrointest Endosc 2004; 60: 15-21
  • 23 Longstreth GF, Feitelberg SP. Outpatient care of selected patients with acute non-variceal upper gastrointestinal haemorrhage. Lancet 1995; 345: 108-111
  • 24 Longstreth GF, Feitelberg SP. Successful outpatient management of acute upper gastrointestinal hemorrhage: use of practice guidelines in a large patient series. Gastrointest Endosc 1998; 47: 219-222
  • 25 Rockall TA, Logan RF, Devlin HB et al. Selection of patients for early discharge or outpatient care after acute upper gastrointestinal haemorrhage. national audit of acute upper gastrointestinal haemorrhage. Lancet 1996; 347: 1138-1140
  • 26 Lai KC, Hui WM, Wong BC et al. A retrospective and prospective study on the safety of discharging selected patients with duodenal ulcer bleeding on the same day as endoscopy. Gastrointest Endosc 1997; 45: 26-30
  • 27 Cebollero-Santamaria F, Smith J, Gioe S et al. Selective outpatient management of upper gastrointestinal bleeding in the elderly. Am J Gastroenterol 1999; 94: 1242-1247
  • 28 Gralnek IM, Dulai GS. Incremental value of upper endoscopy for triage of patients with acute non-variceal upper GI hemorrhage. Gastrointest Endosc 2004; 60: 9-14
  • 29 Guerrouij M, Uppal CS, Alklabi A et al. The clinical impact of bleeding during oral anticoagulant therapy: assessment of morbidity, mortality and post-bleed anticoagulant management. J Thromb Thrombolysis 2011; 31: 419-423
  • 30 Holbrook A, Schulman S, Witt DM et al. Evidence-based management of anticoagulant therapy: Antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141: e152S-184S DOI: 10.1378/chest.11–2295.
  • 31 Irwin ST, Ferguson R, Weilert F et al. Supratherapeutic anticoagulation at presentation is associated with reduced mortality in nonvariceal upper gastrointestinal hemorrhage. Endosc Int Open 2014; 2: E148-E152 DOI: 10.1055/s-0034–1377287. [Epub 2014 Jul 10]
  • 32 Tran HA, Chunilal SD, Harper PL et al. An update of consensus guidelines for warfarin reversal. Med J Aust 2013; 198: 198-199
  • 33 Choudari CP, Rajgopal C, Palmer KR. Acute gastrointestinal hemorrhage in anticoagulated patients: diagnoses and response to endoscopic treatment. Gut 1994; 35: 464-466
  • 34 Radaelli F, Paggi S, Terruzzi V et al. Management of warfarin-associated coagulopathy in patients with acute gastrointestinal bleeding: a cross-sectional physician survey of current practice. Dig Liver Dis 2011; 43: 444-447
  • 35 Patriquin C, Crowther M. Treatment of warfarin-associated coagulopathy with vitamin K. Expert Rev Hematol 2011; 4: 657-665
  • 36 Baron TH, Kamath PS, McBane RD. New anticoagulant and antiplatelet agents: a primer for the gastroenterologist. Clin Gastroenterol Hepatol 2014; 12: 187-195
  • 37 Leissinger CA, Blatt PM, Hoots WK et al. Role of prothrombin complex concentrates in reversing warfarin anticoagulation: a review of the literature. Am J Hematol 2008; 83: 137-143
  • 38 Karaca MA, Erbil B, Ozmen MM. Use and effectiveness of prothrombin complex concentrates vs. fresh frozen plasma in gastrointestinal hemorrhage due to warfarin usage in the ED. Am J Emerg Med 2014; 32: 660-664
  • 39 Dentali F, Marchesi C, Pierfranceschi MG et al. Safety of prothrombin complex concentrates for rapid anticoagulation reversal of vitamin K antagonists. A meta-analysis. Thromb Haemost 2011; 106: 429-438
  • 40 Hickey M, Gatien M, Taljaard M et al. Outcomes of urgent warfarin reversal with frozen plasma versus prothrombin complex concentrate in the emergency department. Circulation 2013; 128: 360-364
  • 41 Chai-Adisaksopha C, Crowther M, Isayama T et al. The impact of bleeding complications in patients receiving target-specific oral anticoagulants: a systematic review and meta-analysis. Blood 2014; 124: 2450-2458
  • 42 Holster IL, Valkhoff VE, Kuipers EJ et al. New oral anticoagulants increase risk for gastrointestinal bleeding: a systematic review and meta-analysis. Gastroenterology 2013; 145: 105-112
  • 43 Ruff CT, Guigliano RP, Braunwald E et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: a meta-analysis of randomized trials. Lancet 2014; 383: 955-962
  • 44 Lu G, DeGuzman FR, Hollenbach SJ et al. A specific antidote for reversal of anticoagulation by direct and indirect inhibitors of coagulation factor Xa. Nat Med 2013; 19: 446-451
  • 45 Ansell JE, Bakhru SH, Laulicht BE et al. Use of PER977 to reverse the anticoagulant effect of edoxaban. N Engl J Med 2014; 371: 2141-2142
  • 46 Pollack Jr CV, Reilly PA, Eikelboob J et al. Idarucizumab for dabigatran reversal. N Engl J Med 2015; 373: 511-520
  • 47 Abraham NS, Castillo DL. Novel anticoagulants: bleeding risk and management strategies. Curr Opin Gastroenterol 2013; 29: 676-683
  • 48 Desai J, Kolb JM, Weitz JI et al. Gastrointestinal bleeding with the new oral anticoagulants – defining the issues and the management strategies. Thromb Haemost 2013; 110: 205-212
  • 49 Makris M, Van Veen JJ, Tait CR et al. British Committee for Standards in Haematology. Guideline on the management of bleeding in patients on antithrombotic agents. Br J Haematol 2013; 160: 35-46
  • 50 Siegal DM, Cuker A. Reversal of novel oral anticoagulants in patients with major bleeding. J Thromb Thrombolysis 2013; 35: 391-398
  • 51 Fawole A, Daw HA, Crowther MA. Practical management of bleeding due to the anticoagulants dabigatran, rivaroxaban, and apixaban. Clev Clin J Med 2013; 80: 443-451
  • 52 Boustiere C, Veitch A, Vanbiervliet G et al. Endoscopy and antiplatelet agents. European Society of Gastrointestinal Endoscopy (ESGE) guideline. Endoscopy 2011; 43: 445-461
  • 53 Sung JJY, Lau JYW, Ching JYL et al. Continuation of low dose aspirin therapy in peptic ulcer bleeding: a randomized trial. Ann Intern Med 2010; 152: 1-9
  • 54 Derogar M, Sandblom G, Lundell L et al. Discontinuation of low dose aspirin therapy after peptic ulcer bleeding increases risk of death and acute cardiovascular events. Clin Gastroenterol Hepatol 2013; 11: 38-42
  • 55 Liu C-P, Chen W-C, Lai K-H et al. Esomeprazole alone compared with esomeprazole plus aspirin for the treatment of aspirin-related peptic ulcers. Am J Gastroenterol 2012; 107: 1022-1029
  • 56 Ng FH, Wong BCY, Wong SY et al. Clopidogrel plus omeprazole compared with aspirin plus omeprazole for aspirin-induced symptomatic peptic ulcers/erosions with low to moderate bleeding/re-bleeding risk – a single-blind, randomized controlled study. Aliment Pharmacol Ther 2004; 19: 359-365
  • 57 Sreedharan A, Martin J, Leontiadis GI et al. Proton pump inhibitor treatment initiated prior to endoscopic diagnosis in upper gastrointestinal bleeding. Cochrane Database Syst Rev 2010; 7: CD005415
  • 58 Sabah AlS, Barkun AN, Herba K et al. Cost-effectiveness of proton-pump inhibition before endoscopy in upper gastrointestinal bleeding. Clin Gastroenterol Hepatol 2008; 6: 418-425
  • 59 Tsoi KKF, Lau JYW, Sung JJY. Cost-effectiveness analysis of high-dose omeprazole infusion before endoscopy for patients with upper-GI bleeding. Gastrointest Endosc 2008; 67: 1056-1063
  • 60 Shakur H, Roberts I. CRASH-2 trial collaborators et al. Effects of tranexamic acid on death, vascular occlusive events, and blood transfusion in trauma patients with significant haemorrhage (CRASH-2): a randomized, placebo-controlled trial. Lancet 2010; 376: 23-32
  • 61 Gluud LL, Klingenberg SL, Langholz E. Tranexamic acid for upper gastrointestinal bleeding. Cochrane Database Syst Rev 2012; 1: CD006640
  • 62 Raptis S, Dollinger HC, von Berger L et al. Effects of somatostatin on gastric secretion and gastrin release in man. Digestion 1975; 13: 15-26
  • 63 Hearnshaw SA, Logan RF, Lowe D et al. Use of endoscopy for management of acute upper gastrointestinal bleeding in the UK: results of a nationwide audit. Gut 2010; 59: 1022-1029
  • 64 Enestvedt BK, Gralnek IM, Mattek N et al. An evaluation of endoscopic indications and findings related to non-variceal upper-GI hemorrhage in a large multicenter consortium. Gastrointest Endosc 2008; 67: 422-429
  • 65 Barkun AN, Bardou M, Martel M et al. Prokinetics in acute upper GI bleeding: a meta-analysis. Gastrointest Endosc 2010; 72: 1138-1145
  • 66 Szary NM, Gupta R, Choudhary A et al. Erythromycin prior to endoscopy in acute upper gastrointestinal bleeding: a meta-analysis. Scand J Gastroenterol 2011; 46: 920-924
  • 67 Bai Y, Guo JF, Li ZS. Meta-analysis: erythromycin before endoscopy for acute upper gastrointestinal bleeding. Aliment Pharmacol Ther 2011; 34: 166-171
  • 68 Theivanayagam S, Lim RG, Cobell WJ et al. Administration of erythromycin before endoscopy in upper gastrointestinal bleeding: a meta-analysis of randomized controlled trials. Saudi J Gastroenterol 2013; 19: 205-210
  • 69 Winstead NS, Wilcox CM. Erythromycin prior to endoscopy for acute upper gastrointestinal haemorrhage: a cost–effectiveness analysis. Aliment Pharmacol Ther 2007; 26: 1371-1377
  • 70 Srygley FD, Gerardo CJ, Tran T et al. Does this patient have a severe upper gastrointestinal bleed?. JAMA 2012; 307: 1072-1079
  • 71 Aljebreen AM, Fallone CA, Barkun AN. Nasogastric aspirate predicts high-risk endoscopic lesions in patients with acute upper GI bleeding. Gastrointest Endosc 2004; 59: 172-178
  • 72 Pateron D, Vicaut E, Debuc E et al. Erythromycin infusion or gastric lavage for upper gastrointestinal bleeding: a multicenter randomized controlled trial. Ann Emerg Med 2011; 57: 582-589
  • 73 Huang ES, Karsan S, Kanwal F et al. Impact of nasogastric lavage on outcomes in acute GI bleeding. Gastrointest Endosc 2011; 74: 971-980
  • 74 Singer AJ, Richman PB, Kowalska A et al. Comparison of patient and practitioner assessments of pain from commonly performed emergency department procedures. Ann Emerg Med 1999; 33: 652-658
  • 75 Koch DG, Arguedas MR, Fallon MB. Risk of aspiration pneumonia in suspected variceal hemorrhage: the value of prophylactic endotracheal intubation prior to endoscopy. Dig Dis Sci 2007; 52: 2225-2228
  • 76 Rehman A, Iscimen R, Yilmaz M et al. Prophylactic endotracheal intubation in critically ill patients undergoing endoscopy for upper GI hemorrhage. Gastrointest Endosc 2009; 69: 55-59
  • 77 Rudolph SJ, Landsverk BK, Freeman ML. Endotracheal intubation for airway protection during endoscopy for severe upper GI hemorrhage. Gastrointest Endosc 2003; 57: 58-61
  • 78 Kanwal F, Barkun A, Gralnek IM et al. Measuring quality of care in patients with nonvariceal upper gastrointestinal hemorrhage: development of an explicit quality indicator set. Am J Gastroenterol 2010; 105: 1710-1718
  • 79 Lanas A, Aabakken L, Fonseca J et al. Variability in the management of nonvariceal upper gastrointestinal bleeding in Europe: an observational study. Adv Ther 2012; 29: 1026-1036
  • 80 Spiegel BM, Vakil NB, Ofman JJ. Endoscopy for acute nonvariceal upper gastrointestinal tract hemorrhage: is sooner better? A systematic review. Arch Intern Med 2001; 161: 1393-1404
  • 81 Tsoi KKF, Ma TKW, Sung JJY. Endoscopy for upper gastrointestinal bleeding: how urgent is it?. Nat Rev Gastroenterol Hepatol 2009; 6: 463-469
  • 82 Wysocki JD, Srivastav S, Winstead NS. A nationwide analysis of risk factors for mortality and time to endoscopy in upper gastrointestinal haemorrhage. Aliment Pharmacol Ther 2012; 36: 30-36
  • 83 Lin HJ, Wang K, Perng CL et al. Early or delayed endoscopy for patients with peptic ulcer bleeding. A prospective randomized study. J Clin Gastroenterol 1996; 22: 267-271
  • 84 Lim L, Ho K, Chan Y et al. Urgent endoscopy is associated with lower mortality in high-risk but not low-risk nonvariceal upper gastrointestinal bleeding. Endoscopy 2011; 43: 300-306
  • 85 Bjorkman DJ, Zaman A, Fennerty MB et al. Urgent vs. elective endoscopy for acute non-variceal upper-GI bleeding: an effectiveness study. Gastrointest Endosc 2004; 60: 1-8
  • 86 Stanley AJ, Ashley D, Dalton HR et al. Outpatient management of patients with low-risk upper-gastrointestinal haemorrhage: multicentre validation and prospective evaluation. Lancet 2009; 373: 42-47
  • 87 McLaughlin C, Vine L, Chapman L et al. The management of low-risk primary upper gastrointestinal haemorrhage in the community. Eur J Gastroenterol Hepatol 2012; 24: 288-293
  • 88 Girardin M, Bertolini D, Ditisheim S et al. Use of Glasgow-Blatchford bleeding score reduces hospital stay duration and costs for patients with low-risk upper GI bleeding. Endosc Int Open 2014; 2: E74-E79 DOI: 10.1055/s-0034–1365542. Epub 2014 May 7
  • 89 Laursen SB, Dalton HR, Murray IA et al. Performance of new thresholds of the Glasgow Blatchford score in managing patients with upper gastrointestinal bleeding. Clin Gastroenterol Hepatol 2015; 13: 115-121
  • 90 Rubin M, Hussain SA, Shalomov A et al. Live view video capsule endoscopy enables risk stratification of patients with acute upper GI bleeding in the emergency room: a pilot study. Dig Dis Sci 2011; 56: 786-791
  • 91 Chandran S, Testro A, Urquhart P et al. Risk stratification of upper GI bleeding with an esophageal capsule. Gastrointest Endosc 2013; 77: 891-898
  • 92 Gralnek IM, Ching JYL, Maza I et al. Capsule endoscopy in acute upper gastrointestinal hemorrhage: a prospective cohort study. Endoscopy 2013; 45: 12-19
  • 93 Meltzer AC, Ali MA, Kresiberg RB et al. Video capsule endoscopy in the emergency department: a prospective study of acute upper gastrointestinal hemorrhage. Ann Emerg Med 2013; 61: 438-443
  • 94 Meltzer AC, Pinchbeck C, Burnett S et al. Emergency physicians accurately interpret video capsule endoscopy findings in suspected upper gastrointestinal hemorrhage: a video survey. Acad Emerg Med 2013; 20: 711-715
  • 95 Meltzer AC, Ward MJ, Gralnek IM et al. The cost–effectiveness analysis of video capsule endoscopy compared to other strategies to manage acute upper gastrointestinal hemorrhage in the ED. Am J Emerg Med 2014; 32: 823-832
  • 96 Forrest JA, Finlayson ND, Shearman DJ. Endoscopy in gastrointestinal bleeding. Lancet 1974; 2: 394-397
  • 97 Gralnek IM, Barkun AN, Bardou M. Management of acute bleeding from a peptic ulcer. N Engl J Med 2008; 359: 928-937
  • 98 Barkun AN, Bardou M, Kuipers EJ et al. International consensus recommendations on the management of patients with nonvariceal upper gastrointestinal bleeding. Ann Intern Med 2010; 152: 101-113
  • 99 Laine L, Jensen DM. Management of patients with ulcer bleeding. Am J Gastroenterol 2012; 107: 345-360
  • 100 Chung IK, Kim EJ, Lee MS et al. Endoscopic factors predisposing to rebleeding following endoscopic hemostasis in bleeding peptic ulcers. Endoscopy 2001; 33: 969-975
  • 101 Guglielmi A, Ruzzenente A, Sandri M et al. Risk assessment and prediction of rebleeding in bleeding gastroduodenal ulcer. Endoscopy 2002; 34: 778-786
  • 102 Zaragoza AM, Tenías JM, Llorente MJ et al. Prognostic factors in gastrointestinal bleeding due to peptic ulcer: construction of a predictive model. J Clin Gastroenterol 2008; 42: 786-790
  • 103 Elmunzer BJ, Young SD, Inadomi JM et al. Systematic review of the predictors of recurrent hemorrhage after endoscopic hemostatic therapy for bleeding peptic ulcers. Am J Gastroenterol 2008; 103: 2625-2632
  • 104 Marmo R, Del Piano M, Rotondano G et al. Mortality from nonulcer bleeding is similar to that of ulcer bleeding in high-risk patients with nonvariceal hemorrhage: a prospective database study in Italy. Gastrointest Endosc 2012; 75: 263-272
  • 105 Bratanic A, Puljiz Z, Ljubicicz N et al. Predictive factors of rebleeding and mortality following endoscopic hemostasis in bleeding peptic ulcers. Hepatogastroenterology 2013; 60: 112-117
  • 106 Sung JJ, Barkun A, Kuipers EJ et al. Intravenous esomeprazole for prevention of recurrent peptic ulcer bleeding: a randomized trial. Ann Intern Med 2009; 50: 455-464
  • 107 de Groot NL, van Oijen MG, Kessels K et al. Reassessment of the predictive value of the Forrest classification for peptic ulcer rebleeding and mortality: can classification be simplified?. Endoscopy 2014; 46: 46-52
  • 108 Lau JY, Sung JJ, Chan AC et al. Stigmata of hemorrhage in bleeding peptic ulcers: an interobserver agreement study among international experts. Gastrointest Endosc 1997; 46: 33-36
  • 109 Mondardini A, Barletti C, Rocca G et al. Non-variceal upper gastrointestinal bleeding and Forrest’s classification: diagnostic agreement between endoscopists from the same area. Endoscopy 1998; 30: 508-512
  • 110 Lin HJ, Perng CL, Lee FY et al. Clinical courses and predictors for rebleeding in patients with peptic ulcers and non-bleeding visible vessels: a prospective study. Gut 1994; 35: 1389-1393
  • 111 Cheng CL, Lin CH, Kuo CJ et al. Predictors of rebleeding and mortality in patients with high-risk bleeding peptic ulcers. Dig Dis Sci 2010; 55: 2577-2583
  • 112 Laine L, McQuaid KR. Endoscopic therapy for bleeding ulcers: an evidence-based approach based on meta-analyses of randomized controlled trials. Clin Gastroenterol Hepatol 2009; 7: 33-47
  • 113 Sung J, Chan F, Lau J et al. The effect of endoscopic therapy in patients receiving omeprazole for bleeding ulcers with nonbleeding visible vessels or adherent clots: a randomized comparison. Ann Intern Med 2003; 139: 237-243
  • 114 Andriulli A, Annese V, Caruso N et al. Proton-pump inhibitors and outcome of endoscopic hemostasis in bleeding peptic ulcers: a series of meta-analyses. Am J Gastroenterol 2005; 100: 207-219
  • 115 Lin JH, Wang K, Perng CL et al. Natural history of bleeding peptic ulcers with a tightly adherent blood clot: a prospective observation. Gastrointest Endosc 1996; 43: 470-473
  • 116 Jensen DM, Kovacs TO, Jutabha R et al. Randomized trial of medical or endoscopic therapy to prevent recurrent ulcer hemorrhage in patients with adherent clots. Gastroenterology 2002; 123: 407-413
  • 117 Bleau BL, Gostout CJ, Sherman KE et al. Recurrent bleeding from peptic ulcer associated with adherent clot: a randomized study comparing endoscopic treatment with medical therapy. Gastrointest Endosc 2002; 56: 1-6
  • 118 Kahi CJ, Jensen DM, Sung JJY et al. Endoscopic therapy versus medical therapy for bleeding peptic ulcer with adherent clot: a meta-analysis. Gastroenterology 2005; 129: 855-862
  • 119 Wong RC, Chak A, Kobayashi K et al. Role of Doppler US in acute peptic ulcer hemorrhage: can it predict failure of endoscopic therapy?. Gastrointest Endosc 2000; 52: 315-121
  • 120 Kohler B, Maier M, Benz C et al. Acute ulcer bleeding. A prospective randomized trial to compare Doppler and Forrest classifications in endoscopic diagnosis and therapy. Dig Dis Sci 1997; 42: 1370-1374
  • 121 Fullarton GM, Murray WR. Prediction of rebleeding in peptic ulcers by visual stigmata and endoscopic Doppler ultrasound criteria. Endoscopy 1990; 22: 68-71
  • 122 Kohler B, Riemann JF. Endoscopic injection therapy of Forrest II and III gastroduodenal ulcers guided by endoscopic Doppler ultrasound. Endoscopy 1993; 25: 219-223
  • 123 van Leerdam ME, Rauws EA, Geraedts AA et al. The role of endoscopic Doppler US in patients with peptic ulcer bleeding. Gastrointest Endosc 2003; 58: 677-684
  • 124 Chen VK, Wong RC. Endoscopic doppler ultrasound versus endoscopic stigmata-directed management of acute peptic ulcer hemorrhage: a multimodel cost analysis. Dig Dis Sci 2007; 52: 149-160
  • 125 Cipolletta L, Bianco MA, Salerno R et al. Improved characterization of visible vessels in bleeding ulcers by using magnification endoscopy: results of a pilot study. Gastrointest Endosc 2010; 72: 413-418
  • 126 Barkun AN, Martel M, Toubouti Y et al. Endoscopic hemostasis in peptic ulcer bleeding for patients with high-risk lesions: a series of meta-analyses. Gastrointest Endosc 2009; 69: 786-799
  • 127 ASGE Technology Committee. Conway JD, Adler DG et al. Endoscopic hemostatic devices. Gastrointest Endosc 2009; 69: 987-996
  • 128 Laine L. Therapeutic endoscopy and bleeding ulcers. Bipolar/multipolar electrocoagulation. Gastrointest Endosc 1990; 36: S38-S41
  • 129 Ginsberg GG, Barkun AN, Bosco JJ et al. The argon plasma coagulator. Gastrointest Endosc 2002; 55: 807-810
  • 130 Watson JP, Bennett MK, Griffin SM et al. The tissue effect of argon plasma coagulation on esophageal and gastric mucosa. Gastrointest Endosc 2000; 52: 342-345
  • 131 Raju GS, Gajula L. Endoclips for GI endoscopy. Gastrointest Endosc 2004; 59: 267-279
  • 132 Chuttani R, Barkun A, Carpenter S et al. Endoscopic clip application devices. Gastrointest Endosc 2006; 63: 746-750
  • 133 Kirschniak A, Kratt T, Stüker D et al. A new endoscopic over-the-scope clip system for treatment of lesions and bleeding in the GI tract: first clinical experiences. Gastrointest Endosc 2007; 66: 162-167
  • 134 Kirschniak A, Subotova N, Zieker D et al. The over-the-scope clip (OTSC) for the treatment of gastrointestinal bleeding, perforations, and fistulas. Surg Endosc 2011; 25: 2901-2905
  • 135 Gottlieb KT, Banerjee S, Barth BA. ASGE Technology Committee et al. Endoscopic closure devices. Gastrointest Endosc 2012; 76: 244-251
  • 136 Barkun AN, Moosavi S, Martel M. Topical hemostatic agents: a systematic review with particular emphasis on endoscopic application in GI bleeding. Gastrointest Endosc 2013; 77: 692-700
  • 137 Sung JJ, Tsoi KK, Lai LH et al. Endoscopic clipping versus injection and thermo-coagulation in the treatment of non-variceal upper gastrointestinal bleeding: a meta-analysis. Gut 2007; 56: 1364-1373
  • 138 Calvet X, Vergara M, Brullet E et al. Addition of a second endoscopic treatment following epinephrine injection improves outcome in high-risk bleeding ulcers. Gastroenterology 2004; 126: 441-450
  • 139 Marmo R, Rotondano G, Piscopo R et al. Dual therapy versus monotherapy in the endoscopic treatment of high-risk bleeding ulcers: a meta-analysis of controlled trials. Am J Gastroenterol 2007; 102: 279-289
  • 140 Vergara M, Bennett C, Calvet X et al. Epinephrine injection versus epinephrine injection and a second endoscopic method in high risk bleeding ulcers. Cochrane Database Syst Rev 2014; 10: CD005584
  • 141 Hwang JH, Fisher DA, Ben-Menachem T et al. Standards of Practice Committee of the American Society for Gastrointestinal Endoscopy. The role of endoscopy in the management of acute non-variceal upper GI bleeding. Gastrointest Endosc 2012; 75: 1132-1138
  • 142 Yuan Y, Wang C, Hunt RH. Endoscopic clipping for acute nonvariceal upper-GI bleeding: a meta-analysis and critical appraisal of randomized controlled trials. Gastrointest Endosc 2008; 68: 339-351
  • 143 Arima S, Sakata Y, Ogata S et al. Evaluation of hemostasis with soft coagulation using endoscopic hemostatic forceps in comparison with metallic hemoclips for bleeding gastric ulcers: a prospective, randomized trial. J Gastroenterol 2010; 45: 501-505
  • 144 Kataoka M, Kawai T, Hayama Y et al. Comparison of hemostasis using bipolar hemostatic forceps with hemostasis by endoscopic hemoclipping for nonvariceal upper gastrointestinal bleeding in a prospective non-randomized trial. Surg Endosc 2013; 27: 3035-3038
  • 145 Sung JJ, Chan FK, Chen M et al. Asia-Pacific Working Group consensus on non-variceal upper gastrointestinal bleeding. Gut 2011; 60: 1170-1177
  • 146 Wong Kee Song LM, Banerjee S, Barth BA et al. Emerging technologies for endoscopic hemostasis. Gastrointest Endosc 2012; 75: 933-937
  • 147 Manta R, Galloro G, Mangiavillano B et al. Over-the-scope clip (OTSC) represents an effective endoscopic treatment for acute GI bleeding after failure of conventional techniques. Surg Endosc 2013; 27: 3162-3164
  • 148 Giday SA, Kim Y, Krishnamurty DM et al. Long-term randomized controlled trial of a novel nanopowder hemostatic agent (TC-325) for control of severe arterial upper gastrointestinal bleeding in a porcine model. Endoscopy 2011; 43: 296-269
  • 149 Chen YI, Barkun AN, Soulellis C et al. Use of the endoscopically applied hemostatic powder TC-325 in cancer-related upper GI hemorrhage: preliminary experience. Gastrointest Endosc 2012; 75: 1278-1281
  • 150 Leblanc S, Vienne A, Dhooge M et al. Early experience with a novel hemostatic powder used to treat upper GI bleeding related to malignancies or after therapeutic interventions. Gastrointest Endosc 2013; 78: 169-175
  • 151 Holster IL, Kuipers EJ, Tjwa ET. Hemospray in the treatment of upper gastrointestinal hemorrhage in patients on antithrombotic therapy. Endoscopy 2013; 45: 63-66
  • 152 Yau AH, Ou G, Galorport C et al. Safety and efficacy of Hemospray in upper gastrointestinal bleeding. Can J Gastroenterol Hepatol 2014; 28: 72-76
  • 153 Sung JJ, Luo D, Wu JC et al. Early clinical experience of the safety and effectiveness of Hemospray in achieving hemostasis in patients with acute peptic ulcer bleeding. Endoscopy 2011; 43: 291-295
  • 154 Smith LA, Stanley AJ, Bergman JJ et al. Hemospray application in nonvariceal upper gastrointestinal bleeding: results of the survey to evaluate the application of hemospray in the luminal tract. J Clin Gastroenterol 2014; 48: 89-92
  • 155 Barkun A, Sabbah S, Enns R et al. The Canadian Registry on Nonvariceal Upper Gastrointestinal Bleeding and Endoscopy (RUGBE): Endoscopic hemostasis and proton pump inhibition are associated with improved outcomes in a real-life setting. Am J Gastroenterol 2004; 99: 1238-1246
  • 156 Nahon S, Nouel O, Hagège H et al. Favorable prognosis of upper-gastrointestinal bleeding in 1041 older patients: results of a prospective multicenter study. Clin Gastroenterol Hepatol 2008; 6: 886-892
  • 157 Loperfido S, Baldo V, Piovesana E et al. Changing trends in acute upper-GI bleeding: a population-based study. Gastrointest Endosc 2009; 70: 212-224
  • 158 Guntipalli P, Chason R, Elliott A et al. Upper gastrointestinal bleeding caused by severe esophagitis: a unique clinical syndrome. Dig Dis Sci 2014; 59: 2997-3003
  • 159 Wang WH, Huang JQ, Zheng GF et al. Head-to-head comparison of H2-receptor antagonists and proton pump inhibitors in the treatment of erosive esophagitis: A meta-analysis. World J Gastroenterol 2005; 11: 4067-4077
  • 160 Gralnek IM, Dulai GS, Fennerty MB et al. Esomeprazole versus other proton pump inhibitors in erosive esophagitis: a meta-analysis of randomized clinical trials. Clin Gastroenterol Hepatol 2006; 4: 1452-1458
  • 161 Ljubičić N, Budimir I, Pavić T et al. Mortality in high-risk patients with bleeding Mallory–Weiss syndrome is similar to that of peptic ulcer bleeding. Results of a prospective database study. Scand J Gastroenterol 2014; 49: 458-464
  • 162 Bharucha AE, Gostout CJ, Balm RK. Clinical and endoscopic risk factors in the Mallory-Weiss syndrome. Am J Gastroenterol 1997; 92: 805-808
  • 163 Kortas DY, Haas LS, Simpson WG et al. Mallory–Weiss tear: predisposing factors and predictors of a complicated course. Am J Gastroenterol 2001; 96: 2863-2865
  • 164 Chung IK, Kim EJ, Hwang KY et al. Evaluation of endoscopic hemostasis in upper gastrointestinal bleeding related to Mallory–Weiss syndrome. Endoscopy 2002; 34: 474-479
  • 165 Kim JW, Kim HS, Byun JW et al. Predictive factors of recurrent bleeding in Mallory–Weiss syndrome. Korean J Gastroenterol 2005; 46: 447-454
  • 166 Fujisawa N, Inamori M, Sekino Y et al. Risk factors for mortality in patients with Mallory–Weiss syndrome. Hepatogastroenterology 2011; 58: 417-420
  • 167 Huang SP, Wang HP, Lee YC et al. Endoscopic hemoclip placement and epinephrine injection for Mallory-Weiss syndrome with active bleeding. Gastrointest Endosc 2002; 55: 842-846
  • 168 Park CH, Min SW, Sohn YH et al. A prospective, randomized trial of endoscopic band ligation vs. epinephrine injection for actively bleeding Mallory–Weiss syndrome. Gastrointest Endosc 2004; 60: 22-27
  • 169 Cho YS, Chae HS, Kim HK et al. Endoscopic band ligation and endoscopic hemoclip placement for patients with Mallory–Weiss syndrome and active bleeding. World J Gastroenterol 2008; 14: 2080-2084
  • 170 Lecleire S, Antonietti M, Iwanicki-Caron I et al. Endoscopic band ligation could decrease recurrent bleeding in Mallory–Weiss syndrome as compared to haemostasis by hemoclips plus epinephrine. Aliment Pharmacol Ther 2009; 30: 399-405
  • 171 Lara LF, Sreenarasimhaiah J, Tang SJ et al. Dieulafoy lesions of the GI tract: localization and therapeutic outcomes. Dig Dis Sci 2010; 55: 3436-3441
  • 172 Chung IK, Kim EJ, Lee MS et al. Bleeding Dieulafoy’s lesions and the choice of endoscopic method: comparing the hemostatic efficacy of mechanical and injection methods. Gastrointest Endosc 2000; 52: 721-724
  • 173 Kasapidis P, Georgopoulos P, Delis V et al. Endoscopic management and long-term follow-up of Dieulafoy’s lesions in the upper GI tract. Gastrointest Endosc 2002; 55: 527-531
  • 174 Cheng CL, Liu NJ, Lee CS et al. Endoscopic management of Dieulafoy lesions in acute nonvariceal upper gastrointestinal bleeding. Dig Dis Sci 2004; 49: 1139-1144
  • 175 Park CH, Sohn YH, Lee WS et al. The usefulness of endoscopic hemoclipping for bleeding Dieulafoy lesions. Endoscopy 2003; 35: 388-392
  • 176 Katsinelos P, Paroutoglou G, Mimidis K et al. Endoscopic treatment and follow-up of gastrointestinal Dieulafoy’s lesions. World J Gastroenterol 2005; 11: 6022-6026
  • 177 Iacopini F, Petruzziello L, Marchese M et al. Hemostasis of Dieulafoy’s lesions by argon plasma coagulation (with video). Gastrointest Endosc 2007; 66: 20-26
  • 178 Alis H, Oner OZ, Kalayci MU et al. Is endoscopic band ligation superior to injection therapy for Dieulafoy lesion?. Surg Endosc 2009; 23: 1465-1469
  • 179 Sone Y, Kumada T, Toyoda H et al. Endoscopic management and follow up of Dieulafoy’s lesion in the upper gastrointestinal tract. Endoscopy 2005; 37: 449-453
  • 180 Lim W, Kim TO, Park SB et al. Endoscopic treatment of Dieulafoy lesions and risk factors for rebleeding. Korean J Intern Med 2009; 24: 318-322
  • 181 Durham JD, Kumpe DA, Rothbarth LJ et al. Dieulafoy disease: arteriographic findings and treatment. Radiology 1990; 174: 937-941
  • 182 Alshumrani G, Almuaikeel M. Angiographic findings and endovascular embolization in Dieulafoy disease: a case report and literature review. Diagn Interv Radiol 2006; 12: 151-154
  • 183 Jackson CS, Gerson LB. Management of gastrointestinal angiodysplastic lesions (GIADs): a systematic review and meta-analysis. Am J Gastroenterol 2014; 109: 474-483
  • 184 Swanson E, Mahgoub A, MacDonald R et al. Medical and endoscopic therapies for angiodysplasia and gastric antral vascular ectasia: a systematic review. Clin Gastroenterol Hepatol 2014; 12: 571-582
  • 185 Heller SJ, Tokar JL, Nguyen MT et al. Management of bleeding GI tumors. Gastrointest Endosc 2010; 72: 817-824
  • 186 Sheibani S, Kim JJ, Chen B et al. Natural history of acute upper GI bleeding due to tumours: short-term success and long-term recurrence with or without endoscopic therapy. Aliment Pharmacol Ther 2013; 38: 144-150
  • 187 Kim YI, Choi IJ, Cho SJ et al. Outcome of endoscopic therapy for cancer bleeding in patients with unresectable gastric cancer. J Gastroenterol Hepatol 2013; 28: 1489-1495
  • 188 Koh KH, Kim K, Kwon DH et al. The successful endoscopic hemostasis factors in bleeding from advanced gastric cancer. Gastric Cancer 2013; 16: 397-403
  • 189 Leontiadis GI, Sharma VK, Howden CW. Proton pump inhibitor treatment for acute peptic ulcer bleeding. Cochrane Database Syst Rev 2006; 1: CD002094
  • 190 Leontiadis G, Martin J, Sharma V et al. T1942 Proton pump inhibitor (PPI) treatment for peptic ulcer (PU) bleeding: an updated Cochrane meta-analysis of randomized controlled trials (RCTs) [abstract]. Gastroenterology 2009; DOI: http://dx.doi.org/10.1016/S0016-5085(09)62789-X.
  • 191 Sachar H, Vaidya K, Laine L. Intermittent vs continuous proton pump inhibitor therapy for high-risk bleeding ulcers: a systematic review and meta-analysis. JAMA Intern Med 2014; 174: 1755-1762
  • 192 Javid G, Zargar SA, U-Saif R et al. Comparison of p. o. or i.v. proton pump inhibitors on 72-h intragastric pH in bleeding peptic ulcer. J Gastroenterol Hepatol 2009; 24: 1236-1243
  • 193 Sung JJ, Suen BY, Wu JC et al. Effects of intravenous and oral esomeprazole in the prevention of recurrent bleeding from peptic ulcers after endoscopic therapy. Am J Gastroenterol 2014; 109: 1005-1010
  • 194 Lau JYW, Sung JJY, Lam YH et al. Endoscopic retreatment compared with surgery in patients with recurrent bleeding after initial endoscopic control of bleeding ulcer. N Engl J Med 1999; 340: 751-756
  • 195 Wong TCF, Wong TT, Chiu PWY et al. A comparison of angiographic embolization with surgery after failed endoscopic hemostasis to bleeding peptic ulcers. Gastrointest Endosc 2011; 73: 900-908
  • 196 Kyaw M, Tse Y, Ang D et al. Embolization versus surgery for peptic ulcer bleeding after failed endoscopic hemostasis: a meta-analysis. Endos Int Open 2014; 2: E6-E14
  • 197 Beggs AD, Dilworth MP, Powell SL et al. A systematic review of transarterial embolization versus emergency surgery in treatment of major nonvariceal upper gastrointestinal bleeding. Clin Exp Gastroenterol 2014; 7: 93-104
  • 198 Sulz M, Frei R, Meyenberger C et al. Routine use of Hemospray for gastrointestinal bleeding: prospective two-center experience in Switzerland. Endoscopy 2014; 46: 619-624
  • 199 El OualiS, Barkun AN, Wyse J et al. Is routine second-look endoscopy effective after endoscopic hemostasis in acute peptic ulcer bleeding? A meta-analysis. Gastrointest Endosc 2012; 76: 283-292
  • 200 Imperiale TF, Kong N. Second look endoscopy for bleeding peptic ulcer disease: a decision and cost-effectiveness analysis. J Clin Gastroenterol 2012; 46: e71-e75
  • 201 Holster IL, Kuipers EJ. Management of acute nonvariceal upper gastrointestinal bleeding: current policies and future perspectives. World J Gastroenterol 2012; 18: 1202-1207
  • 202 Sbrozzi-Vanni A, Zullo A, Di Giulio E et al. Low prevalence of idiopathic peptic ulcer disease: an Italian endoscopic survey. Dig Liver Dis 2010; 42: 773-776
  • 203 Gisbert JP, Khorrami S, Carballo F et al. Meta-analysis: Helicobacter pylori eradication therapy vs. anti-secretory non-eradication therapy for the prevention of recurrent bleeding from peptic ulcer. Aliment Pharmacol Ther 2004; 19: 617-629
  • 204 Gisbert JP, Calvet X, Cosme A et al. Long-term follow-up of 1,000 patients cured of Helicobacter pylori infection following an episode of peptic ulcer bleeding. Am J Gastroenterol 2012; 107: 1197-1204
  • 205 Gisbert JP, Abraira V. Accuracy of Helicobacter pylori diagnostic tests in patients with bleeding peptic ulcer: a systematic review and meta-analysis. Am J Gastroenterol 2006; 101: 848-863
  • 206 Sánchez-Delgado J, Gené E, Suárez D et al. Has H. pylori prevalence in bleeding peptic ulcer been underestimated? A meta-regression. Am J Gastroenterol 2011; 106: 398-405
  • 207 Witt DM, Delate T, Garcia DA et al. Risk of thromboembolism, recurrent hemorrhage, and death after warfarin therapy interruption for gastrointestinal tract bleeding. Arch Intern Med 2012; 172: 1484-1491
  • 208 Lee JK, Kang HW, Kim SG et al. Risks related with withholding and resuming anticoagulation in patients with non-variceal upper gastrointestinal bleeding while on warfarin therapy. Int J Clin Pract 2012; 66: 64-68
  • 209 Qureshi W, Mittal C, Patsias I et al. Restarting anticoagulation and outcomes after major gastrointestinal bleeding in atrial fibrillation. Am J Cardiol 2014; 113: 662-668
  • 210 Douketis JD, Spyropoulos AC, Spencer FA et al. Perioperative management of antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest 2012; 141 Suppl e326S-e350S
  • 211 Sibon I, Orgogozo JM. Antiplatelet drug discontinuation is a risk factor for ischemic stroke. Neurology 2004; 62: 1187-1189
  • 212 Biondi-Zoccai GG, Lotrionte M, Agostoni P et al. A systematic review and meta-analysis on the hazards of discontinuing or not adhering to aspirin among 50,279 patients at risk for coronary artery disease. Eur Heart J 2006; 27: 2667-2674
  • 213 Garcia-Rodriguez LA, Cea-Soriano L, Martin-Merino E et al. Discontinuation of low dose aspirin and risk of myocardial infarction: case–control study in UK primary care. BMJ 2011; 343: d4094 DOI: 10.1136/bmj.d4094.
  • 214 Cea Soriano L, Bueno H, Lanas A et al. Cardiovascular and upper gastrointestinal bleeding consequences of low dose acetylsalicylic acid discontinuation. Thromb Haemost 2013; 110: 1298-1304
  • 215 King III SB, Smith Jr SC, Hirshfeld Jr JW et al. 2007 focused update of the ACC/AHA/SCAI 2005 guideline update for percutaneous coronary intervention: a report of the American College of Cardiology/American Heart Association Task Force on Practice guidelines. J Am Coll Cardiol 2008; 51: 172-209
  • 216 Anderson JL, Adams CD, Antman EM et al. 2012 ACCF/AHA focused update incorporated into the ACCF/AHA 2007 guidelines for the management of patients with unstable angina/non-ST-elevation myocardial infarction: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol 2013; 61: 179-347
  • 217 Garcia-Rodriguez LA, Lin KJ, Hernandez-Diaz S et al. Risk of upper gastrointestinal bleeding with low dose acetylsalicylic acid alone and in combination with clopidogrel and other medications. Circulation 2011; 123: 1108-1115
  • 218 Lanas A, Garcia-Rodriguez LA, Arroyo MT et al. Effect of anti-secretory drugs and nitrates on the risk of ulcer bleeding associated with non-steroidal anti-inflammatory drugs, antiplatelet agents, and anticoagulants. Am J Gastroenterol 2007; 102: 507-515
  • 219 Bhatt DL, Scheiman J, Abraham NS et al. ACCF/ACG/AHA 2008 expert consensus document on reducing the gastrointestinal risks of antiplatelet therapy and NSAID use: a report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus Documents. Circulation 2008; 118: 1894-1909
  • 220 Kwok CS, Nijar RS, Loke YK. Effects of proton pump inhibitors on adverse gastrointestinal events in patients receiving clopidogrel: a systematic review and meta-analysis. Drug Saf 2011; 34: 47-57
  • 221 Gilard M, Arnaud B, Cornily JC et al. Influence of omeprazole on the antiplatelet action of clopidogrel associated with aspirin: the randomized, double-blind OCLA (Omeprazole CLopidogrel Aspirin) study. J Am Coll Cardiol 2008; 51: 256-260
  • 222 Cuisset T, Frere C, Quilici J et al. Comparison of omeprazole and pantoprazole influence on a high 150-mg clopidogrel maintenance dose: the PACA (Proton Pump Inhibitors And Clopidogrel Association) prospective randomized study. J Am Coll Cardiol 2009; 54: 1149-1153
  • 223 Siller-Matula JM, Spiel AO, Lang IM et al. Effects of pantoprazole and esomeprazole on platelet inhibition by clopidogrel. Am Heart J 2009; 157: 148-145
  • 224 O’Donoghue ML, Braunwald E, Antman EM et al. Pharmacodynamic effect and clinical efficacy of clopidogrel and prasugrel with or without a proton-pump inhibitor: an analysis of two randomised trials. Lancet 2009; 374: 989-997
  • 225 Chen J, Chen SY, Lian JJ et al. Pharmacodynamic impacts of proton pump inhibitors on the efficacy of clopidogrel in vivo – a systematic review. Clin Cardiol 2013; 36: 184-189
  • 226 Kwok CS, Loke YK. Meta-analysis: the effects of proton pump inhibitors on cardiovascular events and mortality in patients receiving clopidogrel. Aliment Pharmacol Ther 2010; 31: 810-823
  • 227 Siller-Matula JM, Jilma B, Schror K et al. Effect of proton pump inhibitors on clinical outcome in patients treated with clopidogrel: a systematic review and meta-analysis. J Thromb Haemost 2010; 8: 2624-2641
  • 228 Cardoso RN, Benjo AM, DiNicolantonio JJ et al. Incidence of cardiovascular events and gastrointestinal bleeding in patients receiving clopidogrel with and without proton pump inhibitors: an updated meta-analysis. Open Heart 2015; 2: e000248 DOI: 10.1136/openhrt-2015-000248.
    • References for Appendices

    • 229 Longstreth GF, Feitelberg SP. Outpatient care of selected patients with acute non-variceal upper gastrointestinal haemorrhage. Lancet 1995; 345: 108-111
    • 230 Longstreth GF, Feitelberg SP. Successful outpatient management of acute upper gastrointestinal hemorrhage: use of practice guidelines in a large patient series. Gastrointest Endosc 1998; 47: 219-222
    • 231 Cebollero-Santamaria F, Smith J, Gioe S et al. Selective outpatient management of upper gastrointestinal bleeding in the elderly. Am J Gastroenterol 1999; 94: 1242-1247
    • 232 Brullet E, Campo R, Calvet X et al. A randomized study of the safety of outpatient care for patients with bleeding peptic ulcer treated by endoscopic injection. Gastrointest Endosc 2004; 60: 15-21
    • 233 Lai KC, Hui WM, Wong BC et al. A retrospective and prospective study on the safety of discharging selected patients with duodenal ulcer bleeding on the same day as endoscopy. Gastrointest Endosc 1997; 45: 26-30
    • 234 Lee JG, Turnipseed S, Romano PS et al. Endoscopy-based triage significantly reduces hospitalization rates and costs of treating upper GI bleeding: a randomized controlled trial. Gastrointest Endosc 1999; 50: 755-761
    • 235 Gralnek IM, Dulai GS. Incremental value of upper endoscopy for triage of patients with acute non-variceal upper GI hemorrhage. Gastrointest Endosc 2004; 60: 9-14
    • 236 Cipolletta L, Bianco MA, Rotondano G et al. Outpatient management for low-risk nonvariceal upper GI bleeding: a randomized controlled trial. Gastrointest Endosc 2002; 55: 1-5
    • 237 Sreedharan A, Martin J, Leontiadis GI et al. Proton pump inhibitor treatment initiated prior to endoscopic diagnosis in upper gastrointestinal bleeding. Cochrane Database Syst Rev 2010; 7: CD005415
    • 238 Lau JY, Leung WK, Wu JC et al. Omeprazole before endoscopy in patients with gastrointestinal bleeding. N Engl J Med 2007; 356: 1631-40
    • 239 Liu N, Liu L, Zhang HH et al. Effect of intravenous proton pump inhibitor requirements and timing of endoscopy of peptic ulcer bleeding. J Gastroenterol Hepatol 2012; 27: 1473-79
    • 240 Barkun AN, Bardou M, Kuipers EJ et al. International consensus recommendations on the management of patients with non-variceal upper gastrointestinal bleeding. Ann Intern Med 2010; 152: 101-113
    • 241 Tsoi KKF, Lau JYW, Sung JJY. Cost-effectiveness analysis of high-dose omeprazole infusion before endoscopy for patients with upper GI bleeding. Gastrointest Endosc 2008; 67: 1056-1063
    • 242 Barkun AN. Should every patient with suspected upper GI bleeding receive a proton pump inhibitor while awaiting endoscopy?. Gastrointest Endosc 2008; 67: 1064-1066
    • 243 Rácz I, Szalai M, Dancs N et al. Pantoprazole before endoscopy in patients with gastroduodenal ulcer bleeding: Does the duration of infusion and ulcer location influence the effects?. Gastroenterol Res Pract 2012; Article ID561207
    • 244 Lanas A. Update on non variceal gastrointestinal bleeding. Gastroenterol Hepatol 2013; 36: 57-65
    • 245 Sung JJ, Chan FK, Chen M et al. Asia-Pacific Working consensus on non-variceal upper gastrointestinal bleeding. Gut 2011; 60: 1170-1177
    • 246 Al-Sabah S, Barkun AN, Herba K et al. Cost-effectiveness of proton pump inhibition before endoscopy in upper gastrointestinal bleeding. Clin Gastroenterol Hepatol 2008; 6: 418-425
    • 247 Ghassemi KA, Kovacs TOG, Jensen DM. Gastric acid inhibition in the treatment of peptic ulcer haemorrhage. Curr Gastroenterol Rep 2009; 11: 462-469
    • 248 Laursen SB, Jorgensen HS, Schaffalitzky de Muckadell OB. Management of bleeding gastroduodenal ulcers. Dan Med J 2012; 59: C4473
    • 249 Lin HJ. Role of proton pump inhibitors in the management of peptic ulcer bleeding. World J Gastrointest Pharmacol Ther 2010; 1: 51-53
    • 250 Gluud LL, Klingenberg SL, Langholz E. Tranexamic acid for upper gastrointestinal bleeding. Cochrane Database Syst Rev 2012; 1: CD006640
    • 251 Magnusson I, Ihre T, Johansson C et al. Randomized double blind trial of somatostatin in the treatment of massive upper gastrointestinal hemorrhage. Gut 1985; 26: 221-226
    • 252 Choi CW, Kang DH, Kim HW et al. Somatostatin adjunctive therapy for non-variceal upper gastrointestinal rebleeding after endoscopic therapy. World J Gastroenterol 2011; 17: 3441-3447
    • 253 Avgerinos A, Sgouros S, Viazis N et al. Somatostatin inhibits gastric acid secretion more effectively than pantoprazole in patients with peptic ulcer bleeding: A prospective, randomized, placebo controlled trial. Scan J Gastroenterol 2005; 40: 515-522
    • 254 Archimandritis A, Tsirantonaki M, Tryphonos M et al. Ranitidine versus ranitidine plus octreotide in the treatment of acute nonvariceal upper gastrointestinal bleeding: a prospective randomized study. Curr Med Res Opin 2000; 16: 178-183
    • 255 Lin H, Perng C, Wang K et al. Octreotide for arrest of peptic ulcer hemorrhage – a prospective randomized controlled trial. Hepatogastroenterology 1995; 42: 856-860
    • 256 Kim I, Lee YS, Koh BS et al. Does adding somatostatin to proton pump inhibitor improve the outcome of peptic ulcer bleeding?. Korean J Crit Care Med 2008; 23: 75-78
    • 257 Antonioli A, Gandolfo M, Rigo GD et al. Somatostatin and cimetidine in the control of acute upper gastrointestinal bleeding. A controlled multicentre study. Hepatogastroenterology 1986; 33: 71-94
    • 258 Tisbouris P, Zintazas E, Lappas C et al. High-dose pantoprazole infusion is superior to somatostatin after endoscopic hemostasis in patients with peptic ulcer bleeding. Am J Gastroenterol 2007; 102: 1192-1199
    • 259 Okan A, Simsek I, Akpinar H et al. Somatostatin and ranitidine in the treatment of non-variceal upper gastrointestinal bleeding: a prospective randomized, double-blind, controlled study. Hepatogastroenterology 2000; 47: 1325-1327
    • 260 Rutgeerts P, Avgerinos A et al. Early administration of somatostatin before endoscopy to non-cirrhotic patients with suspected peptic ulcer bleeding: The PUB double-blind, randomized, placebo-controlled trial. Gut 2006; 55: A47
    • 261 Carbonell N, Pauwels A, Serfaty L et al. Erythromycin infusion prior to endoscopy for acute upper gastrointestinal bleeding: a randomized, controlled, double-blind trial. Am J Gastroenterol 2006; 101: 1211-1215
    • 262 Coffin B, Pocard M, Panis Y et al. Erythromycin improves the quality of EGD in patients with acute upper GI bleeding: a randomized controlled study. Gastrointest Endosc 2002; 56: 174-179
    • 263 Frossard JL, Spahr L, Queneau PE et al. Erythromycin intravenous bolus infusion in acute upper gastrointestinal bleeding: a randomized, controlled, double-blind trial. Gastroenterology 2002; 123: 17-23
    • 264 Theivanayagam S, Lim RG, Cobell WJ et al. Administration of erythromycin before endoscopy in upper gastrointestinal bleeding: a meta-analysis of randomized controlled trials. Saudi J Gastroenterol 2013; 19: 205-210
    • 265 Sussman DA, Deshpande AR, Parra JL et al. Intravenous metoclopramide to increase mucosal visualization during endoscopy in patients with acute upper gastrointestinal bleeding: a randomized, controlled study. Gastrontest Endosc 2008; 67: AB247
    • 266 Barkun AN, Bardou M, Martel M et al. Prokinetics in acute upper GI bleeding: a meta-analysis. Gastrointest Endosc 2010; 72: 1138-1145
    • 267 Habashi SL, Lambiase LR, Kottoor R. Prokinetics infusion prior endoscopy for acute upper gastrointestinal bleeding: A randomized, controlled, double-blind and placebo-controlled trial [abstract]. Am J Gastroenterol 2007; 102: S526
    • 268 Winstead NS, Wilcox CM. Erythromycin prior to endoscopy for acute upper gastrointestinal haemorrhage: a cost-effectiveness analysis. Aliment Pharmacol Ther 2007; 26: 1371-1377
    • 269 Spiegel BM, Vakil NB, Ofman JJ. Endoscopy for acute nonvariceal upper gastrointestinal tract hemorrhage: is sooner better? A systematic review. Arch Intern Med 2001; 161: 1393-1404
    • 270 Tsoi KKF, Ma TKW, Sung JJY. Endoscopy for upper gastrointestinal bleeding: how urgent is it?. Nat Rev Gastroenterol Hepatol 2009; 6: 463-469
    • 271 Sarin N, Monga N, Adams PC. Time to endoscopy and outcomes in upper gastrointestinal bleeding. Can J Gastroenterol 2009; 23: 489-493
    • 272 Lim L, Ho K, Chan Y et al. Urgent endoscopy is associated with lower mortality in high-risk but not low-risk nonvariceal upper gastrointestinal bleeding. Endoscopy 2011; 43: 300-306
    • 273 Marmo R, Del Piano M, Rotondano G et al. Mortality from nonvariceal upper gastrointestinal bleeding: is it time to differentiate the timing of endoscopy?. Gastrointest Endosc 2011; 73: AB224
    • 274 Wysocki JD, Srivastav S, Winstead NS. A nationwide analysis of risk factors for mortality and time to endoscopy in upper gastrointestinal haemorrhage. Aliment Pharmacol Ther 2012; 36: 30-36
    • 275 Jairath V, Kakan BC, Logan RF et al. Outcomes following acute nonvariceal upper gastrointestinal bleeding in relation to time to endoscopy: results from a nationwide study. Endoscopy 2012; 44: 723-730
    • 276 Sachar H, Vaidya K, Laine L. Intermittent vs. continuous proton pump inhibitor therapy for high-risk bleeding ulcers: a systematic review and meta-analysis. JAMA Intern Med 2014; 174: 1755-1762
    • 277 Sung JJ, Suen BY, Wu JC et al. Effects of intravenous and oral esomeprazole in the prevention of recurrent bleeding from peptic ulcers after endoscopic therapy. Am J Gastroenterol 2014; 109: 1005-1010
    • 278 Mostaghni AA, Hashemi SA, Heydari ST. Comparison of oral and intravenous proton pump inhibitor on patients with high risk bleeding peptic ulcers: a prospective, randomized, controlled clinical trial. Iran Red Crescent Med J 2011; 13: 458-463
    • 279 Yen HH, Yang CW, Su WW et al. Oral versus intravenous proton pump inhibitors in preventing re-bleeding for patients with peptic ulcer bleeding after successful endoscopic therapy. BMC Gastroenterology 2012; 12: 66
    • 280 Tsai JJ, Hsu YC, Perng CL et al. Oral or intravenous proton pump inhibitor in patients with peptic ulcer bleeding after successful endoscopic epinephrine injection. Br J Clin Pharmacol 2009; 67: 326-332
    • 281 Wang CH, Ma MH, Chou HC et al. High-dose vs non-high-dose proton pump inhibitors after endoscopic treatment in patients with bleeding peptic ulcer: a systematic review and meta-analysis of randomized controlled trials. Arch Intern Med 2010; 170: 751-758
    • 282 Mazjedizadeh AR, Hajiani E, Alavinejad P et al. High dose versus low dose intravenous pantoprazole in bleeding peptic ulcer: a randomized clinical trial. Middle East J Dig Dis 2014; 6: 137-143
    • 283 Lau JYW, Sung JJY, Lam YH et al. Endoscopic retreatment compared with surgery in patients with recurrent bleeding after initial endoscopic control of bleeding ulcer. N Engl J Med 1999; 340: 751-756
    • 284 Wong TCF, Wong TT, Chiu PWY et al. A comparison of angiographic embolization with surgery after failed endoscopic hemostasis to bleeding peptic ulcers. Gastrointest Endosc 2011; 73: 900-908
    • 285 Kyaw M, Tse Y, Ang D et al. Embolization versus surgery for peptic ulcer bleeding after failed endoscopic hemostasis: a meta-analysis. Endosc Int Open 2014; 2: E6-E14
    • 286 Beggs AD, Dilworth MP, Powell SL et al. A systematic review of transarterial embolization versus emergency surgery in treatment of major nonvariceal upper gastrointestinal bleeding. Clin Exp Gastroenterol 2014; 7: 93-104
    • 287 Smith LA, Stanley AJ, Bergman JJ et al. Hemospray application in nonvariceal upper gastrointestinal bleeding: results of the survey to evaluate the application of hemospray in the luminal tract. J Clin Gastroenterol 2014; 48: 89-92
    • 288 Sulz M, Frei R, Meyenberger C et al. Routine use of Hemospray for gastrointestinal bleeding: prospective two-center experience in Switzerland. Endoscopy 2014; 46: 619-624
    • 289 Skinner M, Gutteriez JP, Neumann H et al. Over-the-scope clip placement is effective rescue therapy for severe acute upper gastrointestinal bleeding. Endosc Int Open 2014; 02: E37-E40
    • 290 Manta R, Galloro G, Mangiavillano B et al. Over-the-scope clip (OTSC) represents an effective endoscopic treatment for acute GI bleeding after failure of conventional techniques. Surg Endosc 2013; 27: 3162-3164
    • 291 Sanchez-Delgado J, Gene E, Suarez D et al. Has H. pylori prevalence in bleeding peptic ulcer been underestimated? A meta-regression. Am J Gastroenterol 2011; 106: 398-405
    • 292 Gisbert JP, Abraira V. Accuracy of Helicobacter pylori diagnostic tests in patients with bleeding peptic ulcer: a systematic review and meta-analysis. Am J Gastroenterol 2006; 101: 848-863
    • 293 Gisbert JP, Calvet X, Cosme A et al. Long-term follow-up of 1,000 patients cured of Helicobacter pylori infection following an episode of peptic ulcer bleeding. Am J Gastroenterol 2012; 107: 1197-1204
    • 294 Gisbert JP, Khorrami S, Carballo F et al. Meta-analysis: Helicobacter pylori eradication therapy vs. anti-secretory non-eradication therapy for the prevention of recurrent bleeding from peptic ulcer. Aliment Pharmacol Ther 2004; 19: 617-629
    • 295 Dixon MF, Genta RM, Yardley JH et al. Classification and grading of gastritis. The updated Sydney System. Am J Surg Pathol 1996; 20: 1161-1181
    • 296 Witt DM, Delate T, Garcia DA et al. Risk of thromboembolism, recurrent hemorrhage, and death after warfarin therapy interruption for gastrointestinal tract bleeding. Arch Intern Med 2012; 172: 1484-1491
    • 297 Qureshi W, Mittal C, Patsias I et al. Restarting anticoagulation and outcomes after major gastrointestinal bleeding in atrial fibrillation. Am J Cardiol 2014; 113: 662-668
    • 298 Lee JK, Kang HW, Kim SG et al. Risks related with withholding and resuming anticoagulation in patients with non-variceal upper gastrointestinal bleeding while on warfarin therapy. Int J Clin Pract 2012; 66: 64-68
    • 299 Goodman SG, Clare R, Pieper KS et al. Association of proton pump inhibitor use on cardiovascular outcomes with clopidogrel and ticagrelor: insights from the platelet inhibition and patient outcomes trial. Circulation 2012; 125: 978-986
    • 300 Stockl KM, Le L, Zakharyan A et al. Risk of rehospitalization for patients using clopidogrel with a proton pump inhibitor. Arch Intern Med 2010; 170: 704-710
    • 301 Kreutz RP, Stanek EJ, Aubert R et al. Impact of proton pump inhibitors on the effectiveness of clopidogrel after coronary stent placement: the clopidogrel Medco outcomes study. Pharmacotherapy 2010; 30: 787-796
    • 302 Ho PM, Maddox TM, Wang L et al. Risk of adverse outcomes associated with concomitant use of clopidogrel and proton pump inhibitors following acute coronary syndrome. JAMA 2009; 301: 937-944
    • 303 Huang CC, Chen YC, Leu HB et al. Risk of adverse outcomes in Taiwan associated with concomitant use of clopidogrel and proton pump inhibitors in patients who received percutaneous coronary intervention. Am J Cardiol 2010; 105: 1705-1709
    • 304 Zou JJ, Chen SL, Tan J et al. Increased risk for developing major adverse cardiovascular events in stented Chinese patients treated with dual antiplatelet therapy after concomitant use of the proton pump inhibitor. PLoS One 2014; 9: e84985 DOI: 10.1371/journal.pone. 0084985.
    • 305 van Boxel OS, van Oijen MG, Hagenaars MP et al. Cardiovascular and gastrointestinal outcomes in clopidogrel users on proton pump inhibitors: results of a large Dutch cohort study. Am J Gastroenterol 2010; 105: 2430-2436
    • 306 Munoz-Torrero JF, Escudero D, Suarez C et al. Concomitant use of proton pump inhibitors and clopidogrel in patients with coronary, cerebrovascular, or peripheral artery disease in the factores de Riesgo y ENfermedad Arterial (FRENA) registry. J Cardiovasc Pharmacol 2011; 57: 13-19
    • 307 O’Donoghue ML, Braunwald E, Antman EM et al. Pharmacodynamic effect and clinical efficacy of clopidogrel and prasugrel with or without a proton-pump inhibitor: an analysis of two randomised trials. Lancet 2009; 374: 989-997
    • 308 Hsiao FY, Mullins CD, Wen YW et al. Relationship between cardiovascular outcomes and proton pump inhibitor use in patients receiving dual antiplatelet therapy after acute coronary syndrome. Pharmacoepidemiol Drug Saf 2011; 20: 1043-1049
    • 309 Banerjee S, Weideman RA, Weideman MW et al. Effect of concomitant use of clopidogrel and proton pump inhibitors after percutaneous coronary intervention. Am J Cardiol 2011; 107: 871-878
    • 310 Harjai KJ, Shenoy C, Orshaw P et al. Clinical outcomes in patients with the concomitant use of clopidogrel and proton pump inhibitors after percutaneous coronary intervention: an analysis from the Guthrie Health Off-Label Stent (GHOST) investigators. Circ Cardiovasc Interv 2011; 4: 162-170
    • 311 Aihara H, Sato A, Takeyasu N et al. Effect of individual proton pump inhibitors on cardiovascular events in patients treated with clopidogrel following coronary stenting: results from the Ibaraki Cardiac Assessment Study Registry. Catheter Cardiovasc Interv 2012; 80: 556-563
    • 312 Tentzeris I, Jarai R, Farhan S et al. Impact of concomitant treatment with proton pump inhibitors and clopidogrel on clinical outcome in patients after coronary stent implantation. Thromb Haemost 2010; 104: 1211-1218
    • 313 Schmidt M, Johansen MB, Robertson DJ et al. Concomitant use of clopidogrel and proton pump inhibitors is not associated with major adverse cardiovascular events following coronary stent implantation. Aliment Pharmacol Ther 2012; 35: 165-174
    • 314 Rassen JA, Choudhry NK, Avorn J et al. Cardiovascular outcomes and mortality in patients using clopidogrel with proton pump inhibitors after percutaneous coronary intervention or acute coronary syndrome. Circulation 2009; 120: 2322-2329
    • 315 Ray WA, Murray KT, Griffin MR et al. Outcomes with concurrent use of clopidogrel and proton-pump inhibitors: a cohort study. Ann Intern Med 2010; 152: 337-345
    • 316 Kwok CS, Loke YK. Meta-analysis: the effects of proton pump inhibitors on cardiovascular events and mortality in patients receiving clopidogrel. Aliment Pharmacol Ther 2010; 31: 810-823
    • 317 Siller-Matula JM, Jilma B, Schror K et al. Effect of proton pump inhibitors on clinical outcome in patients treated with clopidogrel: a systematic review and meta-analysis. J Thromb Haemost 2010; 8: 2624-2641

Corresponding author

Ian M. Gralnek, MD, MSHS
Institute of Gastroenterology and Liver Diseases, Ha'Emek Medical Center
Rappaport Faculty of Medicine, Technion-Israel Institute of Technology
Afula, Israel 18101
Fax: +972-4-6495314   

  • References

  • 1 van Leerdam ME. Epidemiology of acute upper gastrointestinal bleeding. Best Pract Res Clin Gastroenterol 2008; 22: 209-224
  • 2 Hearnshaw SA, Logan RF, Lowe D et al. Acute upper gastrointestinal bleeding in the UK: patient characteristics, diagnoses and outcomes in the 2007 UK audit. Gut 2011; 60: 1327-1335
  • 3 Guyatt GH, Oxman AD, Vist GE et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ 2008; 336: 924-926
  • 4 Dumonceau JM, Hassan C, Riphaus A et al. European Society of Gastroinestinal Endoscopy (ESGE) guideline development policy. Endoscopy 2012; 44: 626-629
  • 5 Baradarian R, Ramdhaney S, Chapalamadugu R et al. Early intensive resuscitation of patients with upper gastrointestinal bleeding decreases mortality. Am J Gastroenterol 2004; 99: 619-622
  • 6 Kwan I, Bunn F, Chinnock P et al. Timing and volume of fluid administration for patients with bleeding. Cochrane Database Syst Rev 2014; 3: CD002245
  • 7 Spahn DR, Bouillon B, Cerny V et al. Management of bleeding and coagulopathy following major trauma: an updated European guideline. Crit Care 2013; 17: R76
  • 8 Roberts I, Alderson P, Bunn F et al. Colloids versus crystalloids for fluid resuscitation in critically ill patients. Cochrane Database Syst Rev 2004; 4: CD000567
  • 9 Myburgh JA, Finfer S, Bellomo R et al. Hydroxyethyl starch or saline for fluid resuscitation in intensive care. N Engl J Med 2012; 367: 1901-1911
  • 10 Marik PE, Corwin HL. Efficacy of red blood cell transfusion in the critically ill: a systematic review of the literature. Crit Care Med 2008; 36: 2667-2674
  • 11 Restellini S, Kherad O, Jairath V et al. Red blood cell transfusion is associated with increased rebleeding in patients with nonvariceal upper gastrointestinal bleeding. Aliment Pharmacol Ther 2013; 37: 316-322
  • 12 Villanueva C, Colomo A, Bosch A et al. Transfusion strategies for acute upper gastrointestinal bleeding. N Engl J Med 2013; 368: 11-21
  • 13 Jairath V, Kahan BC, Stanworth SJ et al. Prevalence, management, and outcomes of patients with coagulopathy after acute nonvariceal upper gastrointestinal bleeding in the United Kingdom. Transfusion 2013; 53: 1069-1076
  • 14 Shingina A, Barkun AN, Razzaghi A et al. Systematic review: the presenting international normalized ratio (INR) as a predictor of outcome in patients with upper nonvariceal gastrointestinal bleeding. Aliment Pharmacol Ther 2011; 33: 1010-1018
  • 15 Karam O, Tucci M, Combescure C et al. Plasma transfusion strategies for critically ill patients. Cochrane Database Syst Rev 2013; 12: CD010654
  • 16 Razzaghi A, Barkun AN. Platelet transfusion threshold in patients with upper gastrointestinal bleeding: a systematic review. J Clin Gastroenterol 2012; 46: 482-486
  • 17 Rockall TA, Logan RF, Devlin HB et al. Risk assessment after acute upper gastrointestinal haemorrhage. Gut 1996; 38: 316-321
  • 18 Blatchford O, Murray WR, Blatchford M. A risk score to predict need for treatment for upper-gastrointestinal haemorrhage. Lancet 2000; 356: 1318-1321
  • 19 de Groot NL, Bosman JH, Siersema PD et al. Prediction scores in gastrointestinal bleeding: a systematic review and quantitative appraisal. Endoscopy 2012; 44: 731-739
  • 20 Lee JG, Turnipseed S, Romano PS et al. Endoscopy-based triage significantly reduces hospitalization rates and costs of treating upper GI bleeding: a randomized controlled trial. Gastrointest Endosc 1999; 50: 755-761
  • 21 Cipolletta L, Bianco MA, Rotondano G et al. Outpatient management for low-risk nonvariceal upper GI bleeding: a randomized controlled trial. Gastrointest Endosc 2002; 55: 1-5
  • 22 Brullet E, Campo R, Calvet X et al. A randomized study of the safety of outpatient care for patients with bleeding peptic ulcer treated by endoscopic injection. Gastrointest Endosc 2004; 60: 15-21
  • 23 Longstreth GF, Feitelberg SP. Outpatient care of selected patients with acute non-variceal upper gastrointestinal haemorrhage. Lancet 1995; 345: 108-111
  • 24 Longstreth GF, Feitelberg SP. Successful outpatient management of acute upper gastrointestinal hemorrhage: use of practice guidelines in a large patient series. Gastrointest Endosc 1998; 47: 219-222
  • 25 Rockall TA, Logan RF, Devlin HB et al. Selection of patients for early discharge or outpatient care after acute upper gastrointestinal haemorrhage. national audit of acute upper gastrointestinal haemorrhage. Lancet 1996; 347: 1138-1140
  • 26 Lai KC, Hui WM, Wong BC et al. A retrospective and prospective study on the safety of discharging selected patients with duodenal ulcer bleeding on the same day as endoscopy. Gastrointest Endosc 1997; 45: 26-30
  • 27 Cebollero-Santamaria F, Smith J, Gioe S et al. Selective outpatient management of upper gastrointestinal bleeding in the elderly. Am J Gastroenterol 1999; 94: 1242-1247
  • 28 Gralnek IM, Dulai GS. Incremental value of upper endoscopy for triage of patients with acute non-variceal upper GI hemorrhage. Gastrointest Endosc 2004; 60: 9-14
  • 29 Guerrouij M, Uppal CS, Alklabi A et al. The clinical impact of bleeding during oral anticoagulant therapy: assessment of morbidity, mortality and post-bleed anticoagulant management. J Thromb Thrombolysis 2011; 31: 419-423
  • 30 Holbrook A, Schulman S, Witt DM et al. Evidence-based management of anticoagulant therapy: Antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012; 141: e152S-184S DOI: 10.1378/chest.11–2295.
  • 31 Irwin ST, Ferguson R, Weilert F et al. Supratherapeutic anticoagulation at presentation is associated with reduced mortality in nonvariceal upper gastrointestinal hemorrhage. Endosc Int Open 2014; 2: E148-E152 DOI: 10.1055/s-0034–1377287. [Epub 2014 Jul 10]
  • 32 Tran HA, Chunilal SD, Harper PL et al. An update of consensus guidelines for warfarin reversal. Med J Aust 2013; 198: 198-199
  • 33 Choudari CP, Rajgopal C, Palmer KR. Acute gastrointestinal hemorrhage in anticoagulated patients: diagnoses and response to endoscopic treatment. Gut 1994; 35: 464-466
  • 34 Radaelli F, Paggi S, Terruzzi V et al. Management of warfarin-associated coagulopathy in patients with acute gastrointestinal bleeding: a cross-sectional physician survey of current practice. Dig Liver Dis 2011; 43: 444-447
  • 35 Patriquin C, Crowther M. Treatment of warfarin-associated coagulopathy with vitamin K. Expert Rev Hematol 2011; 4: 657-665
  • 36 Baron TH, Kamath PS, McBane RD. New anticoagulant and antiplatelet agents: a primer for the gastroenterologist. Clin Gastroenterol Hepatol 2014; 12: 187-195
  • 37 Leissinger CA, Blatt PM, Hoots WK et al. Role of prothrombin complex concentrates in reversing warfarin anticoagulation: a review of the literature. Am J Hematol 2008; 83: 137-143
  • 38 Karaca MA, Erbil B, Ozmen MM. Use and effectiveness of prothrombin complex concentrates vs. fresh frozen plasma in gastrointestinal hemorrhage due to warfarin usage in the ED. Am J Emerg Med 2014; 32: 660-664
  • 39 Dentali F, Marchesi C, Pierfranceschi MG et al. Safety of prothrombin complex concentrates for rapid anticoagulation reversal of vitamin K antagonists. A meta-analysis. Thromb Haemost 2011; 106: 429-438
  • 40 Hickey M, Gatien M, Taljaard M et al. Outcomes of urgent warfarin reversal with frozen plasma versus prothrombin complex concentrate in the emergency department. Circulation 2013; 128: 360-364
  • 41 Chai-Adisaksopha C, Crowther M, Isayama T et al. The impact of bleeding complications in patients receiving target-specific oral anticoagulants: a systematic review and meta-analysis. Blood 2014; 124: 2450-2458
  • 42 Holster IL, Valkhoff VE, Kuipers EJ et al. New oral anticoagulants increase risk for gastrointestinal bleeding: a systematic review and meta-analysis. Gastroenterology 2013; 145: 105-112
  • 43 Ruff CT, Guigliano RP, Braunwald E et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: a meta-analysis of randomized trials. Lancet 2014; 383: 955-962
  • 44 Lu G, DeGuzman FR, Hollenbach SJ et al. A specific antidote for reversal of anticoagulation by direct and indirect inhibitors of coagulation factor Xa. Nat Med 2013; 19: 446-451
  • 45 Ansell JE, Bakhru SH, Laulicht BE et al. Use of PER977 to reverse the anticoagulant effect of edoxaban. N Engl J Med 2014; 371: 2141-2142
  • 46 Pollack Jr CV, Reilly PA, Eikelboob J et al. Idarucizumab for dabigatran reversal. N Engl J Med 2015; 373: 511-520
  • 47 Abraham NS, Castillo DL. Novel anticoagulants: bleeding risk and management strategies. Curr Opin Gastroenterol 2013; 29: 676-683
  • 48 Desai J, Kolb JM, Weitz JI et al. Gastrointestinal bleeding with the new oral anticoagulants – defining the issues and the management strategies. Thromb Haemost 2013; 110: 205-212
  • 49 Makris M, Van Veen JJ, Tait CR et al. British Committee for Standards in Haematology. Guideline on the management of bleeding in patients on antithrombotic agents. Br J Haematol 2013; 160: 35-46
  • 50 Siegal DM, Cuker A. Reversal of novel oral anticoagulants in patients with major bleeding. J Thromb Thrombolysis 2013; 35: 391-398
  • 51 Fawole A, Daw HA, Crowther MA. Practical management of bleeding due to the anticoagulants dabigatran, rivaroxaban, and apixaban. Clev Clin J Med 2013; 80: 443-451
  • 52 Boustiere C, Veitch A, Vanbiervliet G et al. Endoscopy and antiplatelet agents. European Society of Gastrointestinal Endoscopy (ESGE) guideline. Endoscopy 2011; 43: 445-461
  • 53 Sung JJY, Lau JYW, Ching JYL et al. Continuation of low dose aspirin therapy in peptic ulcer bleeding: a randomized trial. Ann Intern Med 2010; 152: 1-9
  • 54 Derogar M, Sandblom G, Lundell L et al. Discontinuation of low dose aspirin therapy after peptic ulcer bleeding increases risk of death and acute cardiovascular events. Clin Gastroenterol Hepatol 2013; 11: 38-42
  • 55 Liu C-P, Chen W-C, Lai K-H et al. Esomeprazole alone compared with esomeprazole plus aspirin for the treatment of aspirin-related peptic ulcers. Am J Gastroenterol 2012; 107: 1022-1029
  • 56 Ng FH, Wong BCY, Wong SY et al. Clopidogrel plus omeprazole compared with aspirin plus omeprazole for aspirin-induced symptomatic peptic ulcers/erosions with low to moderate bleeding/re-bleeding risk – a single-blind, randomized controlled study. Aliment Pharmacol Ther 2004; 19: 359-365
  • 57 Sreedharan A, Martin J, Leontiadis GI et al. Proton pump inhibitor treatment initiated prior to endoscopic diagnosis in upper gastrointestinal bleeding. Cochrane Database Syst Rev 2010; 7: CD005415
  • 58 Sabah AlS, Barkun AN, Herba K et al. Cost-effectiveness of proton-pump inhibition before endoscopy in upper gastrointestinal bleeding. Clin Gastroenterol Hepatol 2008; 6: 418-425
  • 59 Tsoi KKF, Lau JYW, Sung JJY. Cost-effectiveness analysis of high-dose omeprazole infusion before endoscopy for patients with upper-GI bleeding. Gastrointest Endosc 2008; 67: 1056-1063
  • 60 Shakur H, Roberts I. CRASH-2 trial collaborators et al. Effects of tranexamic acid on death, vascular occlusive events, and blood transfusion in trauma patients with significant haemorrhage (CRASH-2): a randomized, placebo-controlled trial. Lancet 2010; 376: 23-32
  • 61 Gluud LL, Klingenberg SL, Langholz E. Tranexamic acid for upper gastrointestinal bleeding. Cochrane Database Syst Rev 2012; 1: CD006640
  • 62 Raptis S, Dollinger HC, von Berger L et al. Effects of somatostatin on gastric secretion and gastrin release in man. Digestion 1975; 13: 15-26
  • 63 Hearnshaw SA, Logan RF, Lowe D et al. Use of endoscopy for management of acute upper gastrointestinal bleeding in the UK: results of a nationwide audit. Gut 2010; 59: 1022-1029
  • 64 Enestvedt BK, Gralnek IM, Mattek N et al. An evaluation of endoscopic indications and findings related to non-variceal upper-GI hemorrhage in a large multicenter consortium. Gastrointest Endosc 2008; 67: 422-429
  • 65 Barkun AN, Bardou M, Martel M et al. Prokinetics in acute upper GI bleeding: a meta-analysis. Gastrointest Endosc 2010; 72: 1138-1145
  • 66 Szary NM, Gupta R, Choudhary A et al. Erythromycin prior to endoscopy in acute upper gastrointestinal bleeding: a meta-analysis. Scand J Gastroenterol 2011; 46: 920-924
  • 67 Bai Y, Guo JF, Li ZS. Meta-analysis: erythromycin before endoscopy for acute upper gastrointestinal bleeding. Aliment Pharmacol Ther 2011; 34: 166-171
  • 68 Theivanayagam S, Lim RG, Cobell WJ et al. Administration of erythromycin before endoscopy in upper gastrointestinal bleeding: a meta-analysis of randomized controlled trials. Saudi J Gastroenterol 2013; 19: 205-210
  • 69 Winstead NS, Wilcox CM. Erythromycin prior to endoscopy for acute upper gastrointestinal haemorrhage: a cost–effectiveness analysis. Aliment Pharmacol Ther 2007; 26: 1371-1377
  • 70 Srygley FD, Gerardo CJ, Tran T et al. Does this patient have a severe upper gastrointestinal bleed?. JAMA 2012; 307: 1072-1079
  • 71 Aljebreen AM, Fallone CA, Barkun AN. Nasogastric aspirate predicts high-risk endoscopic lesions in patients with acute upper GI bleeding. Gastrointest Endosc 2004; 59: 172-178
  • 72 Pateron D, Vicaut E, Debuc E et al. Erythromycin infusion or gastric lavage for upper gastrointestinal bleeding: a multicenter randomized controlled trial. Ann Emerg Med 2011; 57: 582-589
  • 73 Huang ES, Karsan S, Kanwal F et al. Impact of nasogastric lavage on outcomes in acute GI bleeding. Gastrointest Endosc 2011; 74: 971-980
  • 74 Singer AJ, Richman PB, Kowalska A et al. Comparison of patient and practitioner assessments of pain from commonly performed emergency department procedures. Ann Emerg Med 1999; 33: 652-658
  • 75 Koch DG, Arguedas MR, Fallon MB. Risk of aspiration pneumonia in suspected variceal hemorrhage: the value of prophylactic endotracheal intubation prior to endoscopy. Dig Dis Sci 2007; 52: 2225-2228
  • 76 Rehman A, Iscimen R, Yilmaz M et al. Prophylactic endotracheal intubation in critically ill patients undergoing endoscopy for upper GI hemorrhage. Gastrointest Endosc 2009; 69: 55-59
  • 77 Rudolph SJ, Landsverk BK, Freeman ML. Endotracheal intubation for airway protection during endoscopy for severe upper GI hemorrhage. Gastrointest Endosc 2003; 57: 58-61
  • 78 Kanwal F, Barkun A, Gralnek IM et al. Measuring quality of care in patients with nonvariceal upper gastrointestinal hemorrhage: development of an explicit quality indicator set. Am J Gastroenterol 2010; 105: 1710-1718
  • 79 Lanas A, Aabakken L, Fonseca J et al. Variability in the management of nonvariceal upper gastrointestinal bleeding in Europe: an observational study. Adv Ther 2012; 29: 1026-1036
  • 80 Spiegel BM, Vakil NB, Ofman JJ. Endoscopy for acute nonvariceal upper gastrointestinal tract hemorrhage: is sooner better? A systematic review. Arch Intern Med 2001; 161: 1393-1404
  • 81 Tsoi KKF, Ma TKW, Sung JJY. Endoscopy for upper gastrointestinal bleeding: how urgent is it?. Nat Rev Gastroenterol Hepatol 2009; 6: 463-469
  • 82 Wysocki JD, Srivastav S, Winstead NS. A nationwide analysis of risk factors for mortality and time to endoscopy in upper gastrointestinal haemorrhage. Aliment Pharmacol Ther 2012; 36: 30-36
  • 83 Lin HJ, Wang K, Perng CL et al. Early or delayed endoscopy for patients with peptic ulcer bleeding. A prospective randomized study. J Clin Gastroenterol 1996; 22: 267-271
  • 84 Lim L, Ho K, Chan Y et al. Urgent endoscopy is associated with lower mortality in high-risk but not low-risk nonvariceal upper gastrointestinal bleeding. Endoscopy 2011; 43: 300-306
  • 85 Bjorkman DJ, Zaman A, Fennerty MB et al. Urgent vs. elective endoscopy for acute non-variceal upper-GI bleeding: an effectiveness study. Gastrointest Endosc 2004; 60: 1-8
  • 86 Stanley AJ, Ashley D, Dalton HR et al. Outpatient management of patients with low-risk upper-gastrointestinal haemorrhage: multicentre validation and prospective evaluation. Lancet 2009; 373: 42-47
  • 87 McLaughlin C, Vine L, Chapman L et al. The management of low-risk primary upper gastrointestinal haemorrhage in the community. Eur J Gastroenterol Hepatol 2012; 24: 288-293
  • 88 Girardin M, Bertolini D, Ditisheim S et al. Use of Glasgow-Blatchford bleeding score reduces hospital stay duration and costs for patients with low-risk upper GI bleeding. Endosc Int Open 2014; 2: E74-E79 DOI: 10.1055/s-0034–1365542. Epub 2014 May 7
  • 89 Laursen SB, Dalton HR, Murray IA et al. Performance of new thresholds of the Glasgow Blatchford score in managing patients with upper gastrointestinal bleeding. Clin Gastroenterol Hepatol 2015; 13: 115-121
  • 90 Rubin M, Hussain SA, Shalomov A et al. Live view video capsule endoscopy enables risk stratification of patients with acute upper GI bleeding in the emergency room: a pilot study. Dig Dis Sci 2011; 56: 786-791
  • 91 Chandran S, Testro A, Urquhart P et al. Risk stratification of upper GI bleeding with an esophageal capsule. Gastrointest Endosc 2013; 77: 891-898
  • 92 Gralnek IM, Ching JYL, Maza I et al. Capsule endoscopy in acute upper gastrointestinal hemorrhage: a prospective cohort study. Endoscopy 2013; 45: 12-19
  • 93 Meltzer AC, Ali MA, Kresiberg RB et al. Video capsule endoscopy in the emergency department: a prospective study of acute upper gastrointestinal hemorrhage. Ann Emerg Med 2013; 61: 438-443
  • 94 Meltzer AC, Pinchbeck C, Burnett S et al. Emergency physicians accurately interpret video capsule endoscopy findings in suspected upper gastrointestinal hemorrhage: a video survey. Acad Emerg Med 2013; 20: 711-715
  • 95 Meltzer AC, Ward MJ, Gralnek IM et al. The cost–effectiveness analysis of video capsule endoscopy compared to other strategies to manage acute upper gastrointestinal hemorrhage in the ED. Am J Emerg Med 2014; 32: 823-832
  • 96 Forrest JA, Finlayson ND, Shearman DJ. Endoscopy in gastrointestinal bleeding. Lancet 1974; 2: 394-397
  • 97 Gralnek IM, Barkun AN, Bardou M. Management of acute bleeding from a peptic ulcer. N Engl J Med 2008; 359: 928-937
  • 98 Barkun AN, Bardou M, Kuipers EJ et al. International consensus recommendations on the management of patients with nonvariceal upper gastrointestinal bleeding. Ann Intern Med 2010; 152: 101-113
  • 99 Laine L, Jensen DM. Management of patients with ulcer bleeding. Am J Gastroenterol 2012; 107: 345-360
  • 100 Chung IK, Kim EJ, Lee MS et al. Endoscopic factors predisposing to rebleeding following endoscopic hemostasis in bleeding peptic ulcers. Endoscopy 2001; 33: 969-975
  • 101 Guglielmi A, Ruzzenente A, Sandri M et al. Risk assessment and prediction of rebleeding in bleeding gastroduodenal ulcer. Endoscopy 2002; 34: 778-786
  • 102 Zaragoza AM, Tenías JM, Llorente MJ et al. Prognostic factors in gastrointestinal bleeding due to peptic ulcer: construction of a predictive model. J Clin Gastroenterol 2008; 42: 786-790
  • 103 Elmunzer BJ, Young SD, Inadomi JM et al. Systematic review of the predictors of recurrent hemorrhage after endoscopic hemostatic therapy for bleeding peptic ulcers. Am J Gastroenterol 2008; 103: 2625-2632
  • 104 Marmo R, Del Piano M, Rotondano G et al. Mortality from nonulcer bleeding is similar to that of ulcer bleeding in high-risk patients with nonvariceal hemorrhage: a prospective database study in Italy. Gastrointest Endosc 2012; 75: 263-272
  • 105 Bratanic A, Puljiz Z, Ljubicicz N et al. Predictive factors of rebleeding and mortality following endoscopic hemostasis in bleeding peptic ulcers. Hepatogastroenterology 2013; 60: 112-117
  • 106 Sung JJ, Barkun A, Kuipers EJ et al. Intravenous esomeprazole for prevention of recurrent peptic ulcer bleeding: a randomized trial. Ann Intern Med 2009; 50: 455-464
  • 107 de Groot NL, van Oijen MG, Kessels K et al. Reassessment of the predictive value of the Forrest classification for peptic ulcer rebleeding and mortality: can classification be simplified?. Endoscopy 2014; 46: 46-52
  • 108 Lau JY, Sung JJ, Chan AC et al. Stigmata of hemorrhage in bleeding peptic ulcers: an interobserver agreement study among international experts. Gastrointest Endosc 1997; 46: 33-36
  • 109 Mondardini A, Barletti C, Rocca G et al. Non-variceal upper gastrointestinal bleeding and Forrest’s classification: diagnostic agreement between endoscopists from the same area. Endoscopy 1998; 30: 508-512
  • 110 Lin HJ, Perng CL, Lee FY et al. Clinical courses and predictors for rebleeding in patients with peptic ulcers and non-bleeding visible vessels: a prospective study. Gut 1994; 35: 1389-1393
  • 111 Cheng CL, Lin CH, Kuo CJ et al. Predictors of rebleeding and mortality in patients with high-risk bleeding peptic ulcers. Dig Dis Sci 2010; 55: 2577-2583
  • 112 Laine L, McQuaid KR. Endoscopic therapy for bleeding ulcers: an evidence-based approach based on meta-analyses of randomized controlled trials. Clin Gastroenterol Hepatol 2009; 7: 33-47
  • 113 Sung J, Chan F, Lau J et al. The effect of endoscopic therapy in patients receiving omeprazole for bleeding ulcers with nonbleeding visible vessels or adherent clots: a randomized comparison. Ann Intern Med 2003; 139: 237-243
  • 114 Andriulli A, Annese V, Caruso N et al. Proton-pump inhibitors and outcome of endoscopic hemostasis in bleeding peptic ulcers: a series of meta-analyses. Am J Gastroenterol 2005; 100: 207-219
  • 115 Lin JH, Wang K, Perng CL et al. Natural history of bleeding peptic ulcers with a tightly adherent blood clot: a prospective observation. Gastrointest Endosc 1996; 43: 470-473
  • 116 Jensen DM, Kovacs TO, Jutabha R et al. Randomized trial of medical or endoscopic therapy to prevent recurrent ulcer hemorrhage in patients with adherent clots. Gastroenterology 2002; 123: 407-413
  • 117 Bleau BL, Gostout CJ, Sherman KE et al. Recurrent bleeding from peptic ulcer associated with adherent clot: a randomized study comparing endoscopic treatment with medical therapy. Gastrointest Endosc 2002; 56: 1-6
  • 118 Kahi CJ, Jensen DM, Sung JJY et al. Endoscopic therapy versus medical therapy for bleeding peptic ulcer with adherent clot: a meta-analysis. Gastroenterology 2005; 129: 855-862
  • 119 Wong RC, Chak A, Kobayashi K et al. Role of Doppler US in acute peptic ulcer hemorrhage: can it predict failure of endoscopic therapy?. Gastrointest Endosc 2000; 52: 315-121
  • 120 Kohler B, Maier M, Benz C et al. Acute ulcer bleeding. A prospective randomized trial to compare Doppler and Forrest classifications in endoscopic diagnosis and therapy. Dig Dis Sci 1997; 42: 1370-1374
  • 121 Fullarton GM, Murray WR. Prediction of rebleeding in peptic ulcers by visual stigmata and endoscopic Doppler ultrasound criteria. Endoscopy 1990; 22: 68-71
  • 122 Kohler B, Riemann JF. Endoscopic injection therapy of Forrest II and III gastroduodenal ulcers guided by endoscopic Doppler ultrasound. Endoscopy 1993; 25: 219-223
  • 123 van Leerdam ME, Rauws EA, Geraedts AA et al. The role of endoscopic Doppler US in patients with peptic ulcer bleeding. Gastrointest Endosc 2003; 58: 677-684
  • 124 Chen VK, Wong RC. Endoscopic doppler ultrasound versus endoscopic stigmata-directed management of acute peptic ulcer hemorrhage: a multimodel cost analysis. Dig Dis Sci 2007; 52: 149-160
  • 125 Cipolletta L, Bianco MA, Salerno R et al. Improved characterization of visible vessels in bleeding ulcers by using magnification endoscopy: results of a pilot study. Gastrointest Endosc 2010; 72: 413-418
  • 126 Barkun AN, Martel M, Toubouti Y et al. Endoscopic hemostasis in peptic ulcer bleeding for patients with high-risk lesions: a series of meta-analyses. Gastrointest Endosc 2009; 69: 786-799
  • 127 ASGE Technology Committee. Conway JD, Adler DG et al. Endoscopic hemostatic devices. Gastrointest Endosc 2009; 69: 987-996
  • 128 Laine L. Therapeutic endoscopy and bleeding ulcers. Bipolar/multipolar electrocoagulation. Gastrointest Endosc 1990; 36: S38-S41
  • 129 Ginsberg GG, Barkun AN, Bosco JJ et al. The argon plasma coagulator. Gastrointest Endosc 2002; 55: 807-810
  • 130 Watson JP, Bennett MK, Griffin SM et al. The tissue effect of argon plasma coagulation on esophageal and gastric mucosa. Gastrointest Endosc 2000; 52: 342-345
  • 131 Raju GS, Gajula L. Endoclips for GI endoscopy. Gastrointest Endosc 2004; 59: 267-279
  • 132 Chuttani R, Barkun A, Carpenter S et al. Endoscopic clip application devices. Gastrointest Endosc 2006; 63: 746-750
  • 133 Kirschniak A, Kratt T, Stüker D et al. A new endoscopic over-the-scope clip system for treatment of lesions and bleeding in the GI tract: first clinical experiences. Gastrointest Endosc 2007; 66: 162-167
  • 134 Kirschniak A, Subotova N, Zieker D et al. The over-the-scope clip (OTSC) for the treatment of gastrointestinal bleeding, perforations, and fistulas. Surg Endosc 2011; 25: 2901-2905
  • 135 Gottlieb KT, Banerjee S, Barth BA. ASGE Technology Committee et al. Endoscopic closure devices. Gastrointest Endosc 2012; 76: 244-251
  • 136 Barkun AN, Moosavi S, Martel M. Topical hemostatic agents: a systematic review with particular emphasis on endoscopic application in GI bleeding. Gastrointest Endosc 2013; 77: 692-700
  • 137 Sung JJ, Tsoi KK, Lai LH et al. Endoscopic clipping versus injection and thermo-coagulation in the treatment of non-variceal upper gastrointestinal bleeding: a meta-analysis. Gut 2007; 56: 1364-1373
  • 138 Calvet X, Vergara M, Brullet E et al. Addition of a second endoscopic treatment following epinephrine injection improves outcome in high-risk bleeding ulcers. Gastroenterology 2004; 126: 441-450
  • 139 Marmo R, Rotondano G, Piscopo R et al. Dual therapy versus monotherapy in the endoscopic treatment of high-risk bleeding ulcers: a meta-analysis of controlled trials. Am J Gastroenterol 2007; 102: 279-289
  • 140 Vergara M, Bennett C, Calvet X et al. Epinephrine injection versus epinephrine injection and a second endoscopic method in high risk bleeding ulcers. Cochrane Database Syst Rev 2014; 10: CD005584
  • 141 Hwang JH, Fisher DA, Ben-Menachem T et al. Standards of Practice Committee of the American Society for Gastrointestinal Endoscopy. The role of endoscopy in the management of acute non-variceal upper GI bleeding. Gastrointest Endosc 2012; 75: 1132-1138
  • 142 Yuan Y, Wang C, Hunt RH. Endoscopic clipping for acute nonvariceal upper-GI bleeding: a meta-analysis and critical appraisal of randomized controlled trials. Gastrointest Endosc 2008; 68: 339-351
  • 143 Arima S, Sakata Y, Ogata S et al. Evaluation of hemostasis with soft coagulation using endoscopic hemostatic forceps in comparison with metallic hemoclips for bleeding gastric ulcers: a prospective, randomized trial. J Gastroenterol 2010; 45: 501-505
  • 144 Kataoka M, Kawai T, Hayama Y et al. Comparison of hemostasis using bipolar hemostatic forceps with hemostasis by endoscopic hemoclipping for nonvariceal upper gastrointestinal bleeding in a prospective non-randomized trial. Surg Endosc 2013; 27: 3035-3038
  • 145 Sung JJ, Chan FK, Chen M et al. Asia-Pacific Working Group consensus on non-variceal upper gastrointestinal bleeding. Gut 2011; 60: 1170-1177
  • 146 Wong Kee Song LM, Banerjee S, Barth BA et al. Emerging technologies for endoscopic hemostasis. Gastrointest Endosc 2012; 75: 933-937
  • 147 Manta R, Galloro G, Mangiavillano B et al. Over-the-scope clip (OTSC) represents an effective endoscopic treatment for acute GI bleeding after failure of conventional techniques. Surg Endosc 2013; 27: 3162-3164
  • 148 Giday SA, Kim Y, Krishnamurty DM et al. Long-term randomized controlled trial of a novel nanopowder hemostatic agent (TC-325) for control of severe arterial upper gastrointestinal bleeding in a porcine model. Endoscopy 2011; 43: 296-269
  • 149 Chen YI, Barkun AN, Soulellis C et al. Use of the endoscopically applied hemostatic powder TC-325 in cancer-related upper GI hemorrhage: preliminary experience. Gastrointest Endosc 2012; 75: 1278-1281
  • 150 Leblanc S, Vienne A, Dhooge M et al. Early experience with a novel hemostatic powder used to treat upper GI bleeding related to malignancies or after therapeutic interventions. Gastrointest Endosc 2013; 78: 169-175
  • 151 Holster IL, Kuipers EJ, Tjwa ET. Hemospray in the treatment of upper gastrointestinal hemorrhage in patients on antithrombotic therapy. Endoscopy 2013; 45: 63-66
  • 152 Yau AH, Ou G, Galorport C et al. Safety and efficacy of Hemospray in upper gastrointestinal bleeding. Can J Gastroenterol Hepatol 2014; 28: 72-76
  • 153 Sung JJ, Luo D, Wu JC et al. Early clinical experience of the safety and effectiveness of Hemospray in achieving hemostasis in patients with acute peptic ulcer bleeding. Endoscopy 2011; 43: 291-295
  • 154 Smith LA, Stanley AJ, Bergman JJ et al. Hemospray application in nonvariceal upper gastrointestinal bleeding: results of the survey to evaluate the application of hemospray in the luminal tract. J Clin Gastroenterol 2014; 48: 89-92
  • 155 Barkun A, Sabbah S, Enns R et al. The Canadian Registry on Nonvariceal Upper Gastrointestinal Bleeding and Endoscopy (RUGBE): Endoscopic hemostasis and proton pump inhibition are associated with improved outcomes in a real-life setting. Am J Gastroenterol 2004; 99: 1238-1246
  • 156 Nahon S, Nouel O, Hagège H et al. Favorable prognosis of upper-gastrointestinal bleeding in 1041 older patients: results of a prospective multicenter study. Clin Gastroenterol Hepatol 2008; 6: 886-892
  • 157 Loperfido S, Baldo V, Piovesana E et al. Changing trends in acute upper-GI bleeding: a population-based study. Gastrointest Endosc 2009; 70: 212-224
  • 158 Guntipalli P, Chason R, Elliott A et al. Upper gastrointestinal bleeding caused by severe esophagitis: a unique clinical syndrome. Dig Dis Sci 2014; 59: 2997-3003
  • 159 Wang WH, Huang JQ, Zheng GF et al. Head-to-head comparison of H2-receptor antagonists and proton pump inhibitors in the treatment of erosive esophagitis: A meta-analysis. World J Gastroenterol 2005; 11: 4067-4077
  • 160 Gralnek IM, Dulai GS, Fennerty MB et al. Esomeprazole versus other proton pump inhibitors in erosive esophagitis: a meta-analysis of randomized clinical trials. Clin Gastroenterol Hepatol 2006; 4: 1452-1458
  • 161 Ljubičić N, Budimir I, Pavić T et al. Mortality in high-risk patients with bleeding Mallory–Weiss syndrome is similar to that of peptic ulcer bleeding. Results of a prospective database study. Scand J Gastroenterol 2014; 49: 458-464
  • 162 Bharucha AE, Gostout CJ, Balm RK. Clinical and endoscopic risk factors in the Mallory-Weiss syndrome. Am J Gastroenterol 1997; 92: 805-808
  • 163 Kortas DY, Haas LS, Simpson WG et al. Mallory–Weiss tear: predisposing factors and predictors of a complicated course. Am J Gastroenterol 2001; 96: 2863-2865
  • 164 Chung IK, Kim EJ, Hwang KY et al. Evaluation of endoscopic hemostasis in upper gastrointestinal bleeding related to Mallory–Weiss syndrome. Endoscopy 2002; 34: 474-479
  • 165 Kim JW, Kim HS, Byun JW et al. Predictive factors of recurrent bleeding in Mallory–Weiss syndrome. Korean J Gastroenterol 2005; 46: 447-454
  • 166 Fujisawa N, Inamori M, Sekino Y et al. Risk factors for mortality in patients with Mallory–Weiss syndrome. Hepatogastroenterology 2011; 58: 417-420
  • 167 Huang SP, Wang HP, Lee YC et al. Endoscopic hemoclip placement and epinephrine injection for Mallory-Weiss syndrome with active bleeding. Gastrointest Endosc 2002; 55: 842-846
  • 168 Park CH, Min SW, Sohn YH et al. A prospective, randomized trial of endoscopic band ligation vs. epinephrine injection for actively bleeding Mallory–Weiss syndrome. Gastrointest Endosc 2004; 60: 22-27
  • 169 Cho YS, Chae HS, Kim HK et al. Endoscopic band ligation and endoscopic hemoclip placement for patients with Mallory–Weiss syndrome and active bleeding. World J Gastroenterol 2008; 14: 2080-2084
  • 170 Lecleire S, Antonietti M, Iwanicki-Caron I et al. Endoscopic band ligation could decrease recurrent bleeding in Mallory–Weiss syndrome as compared to haemostasis by hemoclips plus epinephrine. Aliment Pharmacol Ther 2009; 30: 399-405
  • 171 Lara LF, Sreenarasimhaiah J, Tang SJ et al. Dieulafoy lesions of the GI tract: localization and therapeutic outcomes. Dig Dis Sci 2010; 55: 3436-3441
  • 172 Chung IK, Kim EJ, Lee MS et al. Bleeding Dieulafoy’s lesions and the choice of endoscopic method: comparing the hemostatic efficacy of mechanical and injection methods. Gastrointest Endosc 2000; 52: 721-724
  • 173 Kasapidis P, Georgopoulos P, Delis V et al. Endoscopic management and long-term follow-up of Dieulafoy’s lesions in the upper GI tract. Gastrointest Endosc 2002; 55: 527-531
  • 174 Cheng CL, Liu NJ, Lee CS et al. Endoscopic management of Dieulafoy lesions in acute nonvariceal upper gastrointestinal bleeding. Dig Dis Sci 2004; 49: 1139-1144
  • 175 Park CH, Sohn YH, Lee WS et al. The usefulness of endoscopic hemoclipping for bleeding Dieulafoy lesions. Endoscopy 2003; 35: 388-392
  • 176 Katsinelos P, Paroutoglou G, Mimidis K et al. Endoscopic treatment and follow-up of gastrointestinal Dieulafoy’s lesions. World J Gastroenterol 2005; 11: 6022-6026
  • 177 Iacopini F, Petruzziello L, Marchese M et al. Hemostasis of Dieulafoy’s lesions by argon plasma coagulation (with video). Gastrointest Endosc 2007; 66: 20-26
  • 178 Alis H, Oner OZ, Kalayci MU et al. Is endoscopic band ligation superior to injection therapy for Dieulafoy lesion?. Surg Endosc 2009; 23: 1465-1469
  • 179 Sone Y, Kumada T, Toyoda H et al. Endoscopic management and follow up of Dieulafoy’s lesion in the upper gastrointestinal tract. Endoscopy 2005; 37: 449-453
  • 180 Lim W, Kim TO, Park SB et al. Endoscopic treatment of Dieulafoy lesions and risk factors for rebleeding. Korean J Intern Med 2009; 24: 318-322
  • 181 Durham JD, Kumpe DA, Rothbarth LJ et al. Dieulafoy disease: arteriographic findings and treatment. Radiology 1990; 174: 937-941
  • 182 Alshumrani G, Almuaikeel M. Angiographic findings and endovascular embolization in Dieulafoy disease: a case report and literature review. Diagn Interv Radiol 2006; 12: 151-154
  • 183 Jackson CS, Gerson LB. Management of gastrointestinal angiodysplastic lesions (GIADs): a systematic review and meta-analysis. Am J Gastroenterol 2014; 109: 474-483
  • 184 Swanson E, Mahgoub A, MacDonald R et al. Medical and endoscopic therapies for angiodysplasia and gastric antral vascular ectasia: a systematic review. Clin Gastroenterol Hepatol 2014; 12: 571-582
  • 185 Heller SJ, Tokar JL, Nguyen MT et al. Management of bleeding GI tumors. Gastrointest Endosc 2010; 72: 817-824
  • 186 Sheibani S, Kim JJ, Chen B et al. Natural history of acute upper GI bleeding due to tumours: short-term success and long-term recurrence with or without endoscopic therapy. Aliment Pharmacol Ther 2013; 38: 144-150
  • 187 Kim YI, Choi IJ, Cho SJ et al. Outcome of endoscopic therapy for cancer bleeding in patients with unresectable gastric cancer. J Gastroenterol Hepatol 2013; 28: 1489-1495
  • 188 Koh KH, Kim K, Kwon DH et al. The successful endoscopic hemostasis factors in bleeding from advanced gastric cancer. Gastric Cancer 2013; 16: 397-403
  • 189 Leontiadis GI, Sharma VK, Howden CW. Proton pump inhibitor treatment for acute peptic ulcer bleeding. Cochrane Database Syst Rev 2006; 1: CD002094
  • 190 Leontiadis G, Martin J, Sharma V et al. T1942 Proton pump inhibitor (PPI) treatment for peptic ulcer (PU) bleeding: an updated Cochrane meta-analysis of randomized controlled trials (RCTs) [abstract]. Gastroenterology 2009; DOI: http://dx.doi.org/10.1016/S0016-5085(09)62789-X.
  • 191 Sachar H, Vaidya K, Laine L. Intermittent vs continuous proton pump inhibitor therapy for high-risk bleeding ulcers: a systematic review and meta-analysis. JAMA Intern Med 2014; 174: 1755-1762
  • 192 Javid G, Zargar SA, U-Saif R et al. Comparison of p. o. or i.v. proton pump inhibitors on 72-h intragastric pH in bleeding peptic ulcer. J Gastroenterol Hepatol 2009; 24: 1236-1243
  • 193 Sung JJ, Suen BY, Wu JC et al. Effects of intravenous and oral esomeprazole in the prevention of recurrent bleeding from peptic ulcers after endoscopic therapy. Am J Gastroenterol 2014; 109: 1005-1010
  • 194 Lau JYW, Sung JJY, Lam YH et al. Endoscopic retreatment compared with surgery in patients with recurrent bleeding after initial endoscopic control of bleeding ulcer. N Engl J Med 1999; 340: 751-756
  • 195 Wong TCF, Wong TT, Chiu PWY et al. A comparison of angiographic embolization with surgery after failed endoscopic hemostasis to bleeding peptic ulcers. Gastrointest Endosc 2011; 73: 900-908
  • 196 Kyaw M, Tse Y, Ang D et al. Embolization versus surgery for peptic ulcer bleeding after failed endoscopic hemostasis: a meta-analysis. Endos Int Open 2014; 2: E6-E14
  • 197 Beggs AD, Dilworth MP, Powell SL et al. A systematic review of transarterial embolization versus emergency surgery in treatment of major nonvariceal upper gastrointestinal bleeding. Clin Exp Gastroenterol 2014; 7: 93-104
  • 198 Sulz M, Frei R, Meyenberger C et al. Routine use of Hemospray for gastrointestinal bleeding: prospective two-center experience in Switzerland. Endoscopy 2014; 46: 619-624
  • 199 El OualiS, Barkun AN, Wyse J et al. Is routine second-look endoscopy effective after endoscopic hemostasis in acute peptic ulcer bleeding? A meta-analysis. Gastrointest Endosc 2012; 76: 283-292
  • 200 Imperiale TF, Kong N. Second look endoscopy for bleeding peptic ulcer disease: a decision and cost-effectiveness analysis. J Clin Gastroenterol 2012; 46: e71-e75
  • 201 Holster IL, Kuipers EJ. Management of acute nonvariceal upper gastrointestinal bleeding: current policies and future perspectives. World J Gastroenterol 2012; 18: 1202-1207
  • 202 Sbrozzi-Vanni A, Zullo A, Di Giulio E et al. Low prevalence of idiopathic peptic ulcer disease: an Italian endoscopic survey. Dig Liver Dis 2010; 42: 773-776
  • 203 Gisbert JP, Khorrami S, Carballo F et al. Meta-analysis: Helicobacter pylori eradication therapy vs. anti-secretory non-eradication therapy for the prevention of recurrent bleeding from peptic ulcer. Aliment Pharmacol Ther 2004; 19: 617-629
  • 204 Gisbert JP, Calvet X, Cosme A et al. Long-term follow-up of 1,000 patients cured of Helicobacter pylori infection following an episode of peptic ulcer bleeding. Am J Gastroenterol 2012; 107: 1197-1204
  • 205 Gisbert JP, Abraira V. Accuracy of Helicobacter pylori diagnostic tests in patients with bleeding peptic ulcer: a systematic review and meta-analysis. Am J Gastroenterol 2006; 101: 848-863
  • 206 Sánchez-Delgado J, Gené E, Suárez D et al. Has H. pylori prevalence in bleeding peptic ulcer been underestimated? A meta-regression. Am J Gastroenterol 2011; 106: 398-405
  • 207 Witt DM, Delate T, Garcia DA et al. Risk of thromboembolism, recurrent hemorrhage, and death after warfarin therapy interruption for gastrointestinal tract bleeding. Arch Intern Med 2012; 172: 1484-1491
  • 208 Lee JK, Kang HW, Kim SG et al. Risks related with withholding and resuming anticoagulation in patients with non-variceal upper gastrointestinal bleeding while on warfarin therapy. Int J Clin Pract 2012; 66: 64-68
  • 209 Qureshi W, Mittal C, Patsias I et al. Restarting anticoagulation and outcomes after major gastrointestinal bleeding in atrial fibrillation. Am J Cardiol 2014; 113: 662-668
  • 210 Douketis JD, Spyropoulos AC, Spencer FA et al. Perioperative management of antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest 2012; 141 Suppl e326S-e350S
  • 211 Sibon I, Orgogozo JM. Antiplatelet drug discontinuation is a risk factor for ischemic stroke. Neurology 2004; 62: 1187-1189
  • 212 Biondi-Zoccai GG, Lotrionte M, Agostoni P et al. A systematic review and meta-analysis on the hazards of discontinuing or not adhering to aspirin among 50,279 patients at risk for coronary artery disease. Eur Heart J 2006; 27: 2667-2674
  • 213 Garcia-Rodriguez LA, Cea-Soriano L, Martin-Merino E et al. Discontinuation of low dose aspirin and risk of myocardial infarction: case–control study in UK primary care. BMJ 2011; 343: d4094 DOI: 10.1136/bmj.d4094.
  • 214 Cea Soriano L, Bueno H, Lanas A et al. Cardiovascular and upper gastrointestinal bleeding consequences of low dose acetylsalicylic acid discontinuation. Thromb Haemost 2013; 110: 1298-1304
  • 215 King III SB, Smith Jr SC, Hirshfeld Jr JW et al. 2007 focused update of the ACC/AHA/SCAI 2005 guideline update for percutaneous coronary intervention: a report of the American College of Cardiology/American Heart Association Task Force on Practice guidelines. J Am Coll Cardiol 2008; 51: 172-209
  • 216 Anderson JL, Adams CD, Antman EM et al. 2012 ACCF/AHA focused update incorporated into the ACCF/AHA 2007 guidelines for the management of patients with unstable angina/non-ST-elevation myocardial infarction: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol 2013; 61: 179-347
  • 217 Garcia-Rodriguez LA, Lin KJ, Hernandez-Diaz S et al. Risk of upper gastrointestinal bleeding with low dose acetylsalicylic acid alone and in combination with clopidogrel and other medications. Circulation 2011; 123: 1108-1115
  • 218 Lanas A, Garcia-Rodriguez LA, Arroyo MT et al. Effect of anti-secretory drugs and nitrates on the risk of ulcer bleeding associated with non-steroidal anti-inflammatory drugs, antiplatelet agents, and anticoagulants. Am J Gastroenterol 2007; 102: 507-515
  • 219 Bhatt DL, Scheiman J, Abraham NS et al. ACCF/ACG/AHA 2008 expert consensus document on reducing the gastrointestinal risks of antiplatelet therapy and NSAID use: a report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus Documents. Circulation 2008; 118: 1894-1909
  • 220 Kwok CS, Nijar RS, Loke YK. Effects of proton pump inhibitors on adverse gastrointestinal events in patients receiving clopidogrel: a systematic review and meta-analysis. Drug Saf 2011; 34: 47-57
  • 221 Gilard M, Arnaud B, Cornily JC et al. Influence of omeprazole on the antiplatelet action of clopidogrel associated with aspirin: the randomized, double-blind OCLA (Omeprazole CLopidogrel Aspirin) study. J Am Coll Cardiol 2008; 51: 256-260
  • 222 Cuisset T, Frere C, Quilici J et al. Comparison of omeprazole and pantoprazole influence on a high 150-mg clopidogrel maintenance dose: the PACA (Proton Pump Inhibitors And Clopidogrel Association) prospective randomized study. J Am Coll Cardiol 2009; 54: 1149-1153
  • 223 Siller-Matula JM, Spiel AO, Lang IM et al. Effects of pantoprazole and esomeprazole on platelet inhibition by clopidogrel. Am Heart J 2009; 157: 148-145
  • 224 O’Donoghue ML, Braunwald E, Antman EM et al. Pharmacodynamic effect and clinical efficacy of clopidogrel and prasugrel with or without a proton-pump inhibitor: an analysis of two randomised trials. Lancet 2009; 374: 989-997
  • 225 Chen J, Chen SY, Lian JJ et al. Pharmacodynamic impacts of proton pump inhibitors on the efficacy of clopidogrel in vivo – a systematic review. Clin Cardiol 2013; 36: 184-189
  • 226 Kwok CS, Loke YK. Meta-analysis: the effects of proton pump inhibitors on cardiovascular events and mortality in patients receiving clopidogrel. Aliment Pharmacol Ther 2010; 31: 810-823
  • 227 Siller-Matula JM, Jilma B, Schror K et al. Effect of proton pump inhibitors on clinical outcome in patients treated with clopidogrel: a systematic review and meta-analysis. J Thromb Haemost 2010; 8: 2624-2641
  • 228 Cardoso RN, Benjo AM, DiNicolantonio JJ et al. Incidence of cardiovascular events and gastrointestinal bleeding in patients receiving clopidogrel with and without proton pump inhibitors: an updated meta-analysis. Open Heart 2015; 2: e000248 DOI: 10.1136/openhrt-2015-000248.
    • References for Appendices

    • 229 Longstreth GF, Feitelberg SP. Outpatient care of selected patients with acute non-variceal upper gastrointestinal haemorrhage. Lancet 1995; 345: 108-111
    • 230 Longstreth GF, Feitelberg SP. Successful outpatient management of acute upper gastrointestinal hemorrhage: use of practice guidelines in a large patient series. Gastrointest Endosc 1998; 47: 219-222
    • 231 Cebollero-Santamaria F, Smith J, Gioe S et al. Selective outpatient management of upper gastrointestinal bleeding in the elderly. Am J Gastroenterol 1999; 94: 1242-1247
    • 232 Brullet E, Campo R, Calvet X et al. A randomized study of the safety of outpatient care for patients with bleeding peptic ulcer treated by endoscopic injection. Gastrointest Endosc 2004; 60: 15-21
    • 233 Lai KC, Hui WM, Wong BC et al. A retrospective and prospective study on the safety of discharging selected patients with duodenal ulcer bleeding on the same day as endoscopy. Gastrointest Endosc 1997; 45: 26-30
    • 234 Lee JG, Turnipseed S, Romano PS et al. Endoscopy-based triage significantly reduces hospitalization rates and costs of treating upper GI bleeding: a randomized controlled trial. Gastrointest Endosc 1999; 50: 755-761
    • 235 Gralnek IM, Dulai GS. Incremental value of upper endoscopy for triage of patients with acute non-variceal upper GI hemorrhage. Gastrointest Endosc 2004; 60: 9-14
    • 236 Cipolletta L, Bianco MA, Rotondano G et al. Outpatient management for low-risk nonvariceal upper GI bleeding: a randomized controlled trial. Gastrointest Endosc 2002; 55: 1-5
    • 237 Sreedharan A, Martin J, Leontiadis GI et al. Proton pump inhibitor treatment initiated prior to endoscopic diagnosis in upper gastrointestinal bleeding. Cochrane Database Syst Rev 2010; 7: CD005415
    • 238 Lau JY, Leung WK, Wu JC et al. Omeprazole before endoscopy in patients with gastrointestinal bleeding. N Engl J Med 2007; 356: 1631-40
    • 239 Liu N, Liu L, Zhang HH et al. Effect of intravenous proton pump inhibitor requirements and timing of endoscopy of peptic ulcer bleeding. J Gastroenterol Hepatol 2012; 27: 1473-79
    • 240 Barkun AN, Bardou M, Kuipers EJ et al. International consensus recommendations on the management of patients with non-variceal upper gastrointestinal bleeding. Ann Intern Med 2010; 152: 101-113
    • 241 Tsoi KKF, Lau JYW, Sung JJY. Cost-effectiveness analysis of high-dose omeprazole infusion before endoscopy for patients with upper GI bleeding. Gastrointest Endosc 2008; 67: 1056-1063
    • 242 Barkun AN. Should every patient with suspected upper GI bleeding receive a proton pump inhibitor while awaiting endoscopy?. Gastrointest Endosc 2008; 67: 1064-1066
    • 243 Rácz I, Szalai M, Dancs N et al. Pantoprazole before endoscopy in patients with gastroduodenal ulcer bleeding: Does the duration of infusion and ulcer location influence the effects?. Gastroenterol Res Pract 2012; Article ID561207
    • 244 Lanas A. Update on non variceal gastrointestinal bleeding. Gastroenterol Hepatol 2013; 36: 57-65
    • 245 Sung JJ, Chan FK, Chen M et al. Asia-Pacific Working consensus on non-variceal upper gastrointestinal bleeding. Gut 2011; 60: 1170-1177
    • 246 Al-Sabah S, Barkun AN, Herba K et al. Cost-effectiveness of proton pump inhibition before endoscopy in upper gastrointestinal bleeding. Clin Gastroenterol Hepatol 2008; 6: 418-425
    • 247 Ghassemi KA, Kovacs TOG, Jensen DM. Gastric acid inhibition in the treatment of peptic ulcer haemorrhage. Curr Gastroenterol Rep 2009; 11: 462-469
    • 248 Laursen SB, Jorgensen HS, Schaffalitzky de Muckadell OB. Management of bleeding gastroduodenal ulcers. Dan Med J 2012; 59: C4473
    • 249 Lin HJ. Role of proton pump inhibitors in the management of peptic ulcer bleeding. World J Gastrointest Pharmacol Ther 2010; 1: 51-53
    • 250 Gluud LL, Klingenberg SL, Langholz E. Tranexamic acid for upper gastrointestinal bleeding. Cochrane Database Syst Rev 2012; 1: CD006640
    • 251 Magnusson I, Ihre T, Johansson C et al. Randomized double blind trial of somatostatin in the treatment of massive upper gastrointestinal hemorrhage. Gut 1985; 26: 221-226
    • 252 Choi CW, Kang DH, Kim HW et al. Somatostatin adjunctive therapy for non-variceal upper gastrointestinal rebleeding after endoscopic therapy. World J Gastroenterol 2011; 17: 3441-3447
    • 253 Avgerinos A, Sgouros S, Viazis N et al. Somatostatin inhibits gastric acid secretion more effectively than pantoprazole in patients with peptic ulcer bleeding: A prospective, randomized, placebo controlled trial. Scan J Gastroenterol 2005; 40: 515-522
    • 254 Archimandritis A, Tsirantonaki M, Tryphonos M et al. Ranitidine versus ranitidine plus octreotide in the treatment of acute nonvariceal upper gastrointestinal bleeding: a prospective randomized study. Curr Med Res Opin 2000; 16: 178-183
    • 255 Lin H, Perng C, Wang K et al. Octreotide for arrest of peptic ulcer hemorrhage – a prospective randomized controlled trial. Hepatogastroenterology 1995; 42: 856-860
    • 256 Kim I, Lee YS, Koh BS et al. Does adding somatostatin to proton pump inhibitor improve the outcome of peptic ulcer bleeding?. Korean J Crit Care Med 2008; 23: 75-78
    • 257 Antonioli A, Gandolfo M, Rigo GD et al. Somatostatin and cimetidine in the control of acute upper gastrointestinal bleeding. A controlled multicentre study. Hepatogastroenterology 1986; 33: 71-94
    • 258 Tisbouris P, Zintazas E, Lappas C et al. High-dose pantoprazole infusion is superior to somatostatin after endoscopic hemostasis in patients with peptic ulcer bleeding. Am J Gastroenterol 2007; 102: 1192-1199
    • 259 Okan A, Simsek I, Akpinar H et al. Somatostatin and ranitidine in the treatment of non-variceal upper gastrointestinal bleeding: a prospective randomized, double-blind, controlled study. Hepatogastroenterology 2000; 47: 1325-1327
    • 260 Rutgeerts P, Avgerinos A et al. Early administration of somatostatin before endoscopy to non-cirrhotic patients with suspected peptic ulcer bleeding: The PUB double-blind, randomized, placebo-controlled trial. Gut 2006; 55: A47
    • 261 Carbonell N, Pauwels A, Serfaty L et al. Erythromycin infusion prior to endoscopy for acute upper gastrointestinal bleeding: a randomized, controlled, double-blind trial. Am J Gastroenterol 2006; 101: 1211-1215
    • 262 Coffin B, Pocard M, Panis Y et al. Erythromycin improves the quality of EGD in patients with acute upper GI bleeding: a randomized controlled study. Gastrointest Endosc 2002; 56: 174-179
    • 263 Frossard JL, Spahr L, Queneau PE et al. Erythromycin intravenous bolus infusion in acute upper gastrointestinal bleeding: a randomized, controlled, double-blind trial. Gastroenterology 2002; 123: 17-23
    • 264 Theivanayagam S, Lim RG, Cobell WJ et al. Administration of erythromycin before endoscopy in upper gastrointestinal bleeding: a meta-analysis of randomized controlled trials. Saudi J Gastroenterol 2013; 19: 205-210
    • 265 Sussman DA, Deshpande AR, Parra JL et al. Intravenous metoclopramide to increase mucosal visualization during endoscopy in patients with acute upper gastrointestinal bleeding: a randomized, controlled study. Gastrontest Endosc 2008; 67: AB247
    • 266 Barkun AN, Bardou M, Martel M et al. Prokinetics in acute upper GI bleeding: a meta-analysis. Gastrointest Endosc 2010; 72: 1138-1145
    • 267 Habashi SL, Lambiase LR, Kottoor R. Prokinetics infusion prior endoscopy for acute upper gastrointestinal bleeding: A randomized, controlled, double-blind and placebo-controlled trial [abstract]. Am J Gastroenterol 2007; 102: S526
    • 268 Winstead NS, Wilcox CM. Erythromycin prior to endoscopy for acute upper gastrointestinal haemorrhage: a cost-effectiveness analysis. Aliment Pharmacol Ther 2007; 26: 1371-1377
    • 269 Spiegel BM, Vakil NB, Ofman JJ. Endoscopy for acute nonvariceal upper gastrointestinal tract hemorrhage: is sooner better? A systematic review. Arch Intern Med 2001; 161: 1393-1404
    • 270 Tsoi KKF, Ma TKW, Sung JJY. Endoscopy for upper gastrointestinal bleeding: how urgent is it?. Nat Rev Gastroenterol Hepatol 2009; 6: 463-469
    • 271 Sarin N, Monga N, Adams PC. Time to endoscopy and outcomes in upper gastrointestinal bleeding. Can J Gastroenterol 2009; 23: 489-493
    • 272 Lim L, Ho K, Chan Y et al. Urgent endoscopy is associated with lower mortality in high-risk but not low-risk nonvariceal upper gastrointestinal bleeding. Endoscopy 2011; 43: 300-306
    • 273 Marmo R, Del Piano M, Rotondano G et al. Mortality from nonvariceal upper gastrointestinal bleeding: is it time to differentiate the timing of endoscopy?. Gastrointest Endosc 2011; 73: AB224
    • 274 Wysocki JD, Srivastav S, Winstead NS. A nationwide analysis of risk factors for mortality and time to endoscopy in upper gastrointestinal haemorrhage. Aliment Pharmacol Ther 2012; 36: 30-36
    • 275 Jairath V, Kakan BC, Logan RF et al. Outcomes following acute nonvariceal upper gastrointestinal bleeding in relation to time to endoscopy: results from a nationwide study. Endoscopy 2012; 44: 723-730
    • 276 Sachar H, Vaidya K, Laine L. Intermittent vs. continuous proton pump inhibitor therapy for high-risk bleeding ulcers: a systematic review and meta-analysis. JAMA Intern Med 2014; 174: 1755-1762
    • 277 Sung JJ, Suen BY, Wu JC et al. Effects of intravenous and oral esomeprazole in the prevention of recurrent bleeding from peptic ulcers after endoscopic therapy. Am J Gastroenterol 2014; 109: 1005-1010
    • 278 Mostaghni AA, Hashemi SA, Heydari ST. Comparison of oral and intravenous proton pump inhibitor on patients with high risk bleeding peptic ulcers: a prospective, randomized, controlled clinical trial. Iran Red Crescent Med J 2011; 13: 458-463
    • 279 Yen HH, Yang CW, Su WW et al. Oral versus intravenous proton pump inhibitors in preventing re-bleeding for patients with peptic ulcer bleeding after successful endoscopic therapy. BMC Gastroenterology 2012; 12: 66
    • 280 Tsai JJ, Hsu YC, Perng CL et al. Oral or intravenous proton pump inhibitor in patients with peptic ulcer bleeding after successful endoscopic epinephrine injection. Br J Clin Pharmacol 2009; 67: 326-332
    • 281 Wang CH, Ma MH, Chou HC et al. High-dose vs non-high-dose proton pump inhibitors after endoscopic treatment in patients with bleeding peptic ulcer: a systematic review and meta-analysis of randomized controlled trials. Arch Intern Med 2010; 170: 751-758
    • 282 Mazjedizadeh AR, Hajiani E, Alavinejad P et al. High dose versus low dose intravenous pantoprazole in bleeding peptic ulcer: a randomized clinical trial. Middle East J Dig Dis 2014; 6: 137-143
    • 283 Lau JYW, Sung JJY, Lam YH et al. Endoscopic retreatment compared with surgery in patients with recurrent bleeding after initial endoscopic control of bleeding ulcer. N Engl J Med 1999; 340: 751-756
    • 284 Wong TCF, Wong TT, Chiu PWY et al. A comparison of angiographic embolization with surgery after failed endoscopic hemostasis to bleeding peptic ulcers. Gastrointest Endosc 2011; 73: 900-908
    • 285 Kyaw M, Tse Y, Ang D et al. Embolization versus surgery for peptic ulcer bleeding after failed endoscopic hemostasis: a meta-analysis. Endosc Int Open 2014; 2: E6-E14
    • 286 Beggs AD, Dilworth MP, Powell SL et al. A systematic review of transarterial embolization versus emergency surgery in treatment of major nonvariceal upper gastrointestinal bleeding. Clin Exp Gastroenterol 2014; 7: 93-104
    • 287 Smith LA, Stanley AJ, Bergman JJ et al. Hemospray application in nonvariceal upper gastrointestinal bleeding: results of the survey to evaluate the application of hemospray in the luminal tract. J Clin Gastroenterol 2014; 48: 89-92
    • 288 Sulz M, Frei R, Meyenberger C et al. Routine use of Hemospray for gastrointestinal bleeding: prospective two-center experience in Switzerland. Endoscopy 2014; 46: 619-624
    • 289 Skinner M, Gutteriez JP, Neumann H et al. Over-the-scope clip placement is effective rescue therapy for severe acute upper gastrointestinal bleeding. Endosc Int Open 2014; 02: E37-E40
    • 290 Manta R, Galloro G, Mangiavillano B et al. Over-the-scope clip (OTSC) represents an effective endoscopic treatment for acute GI bleeding after failure of conventional techniques. Surg Endosc 2013; 27: 3162-3164
    • 291 Sanchez-Delgado J, Gene E, Suarez D et al. Has H. pylori prevalence in bleeding peptic ulcer been underestimated? A meta-regression. Am J Gastroenterol 2011; 106: 398-405
    • 292 Gisbert JP, Abraira V. Accuracy of Helicobacter pylori diagnostic tests in patients with bleeding peptic ulcer: a systematic review and meta-analysis. Am J Gastroenterol 2006; 101: 848-863
    • 293 Gisbert JP, Calvet X, Cosme A et al. Long-term follow-up of 1,000 patients cured of Helicobacter pylori infection following an episode of peptic ulcer bleeding. Am J Gastroenterol 2012; 107: 1197-1204
    • 294 Gisbert JP, Khorrami S, Carballo F et al. Meta-analysis: Helicobacter pylori eradication therapy vs. anti-secretory non-eradication therapy for the prevention of recurrent bleeding from peptic ulcer. Aliment Pharmacol Ther 2004; 19: 617-629
    • 295 Dixon MF, Genta RM, Yardley JH et al. Classification and grading of gastritis. The updated Sydney System. Am J Surg Pathol 1996; 20: 1161-1181
    • 296 Witt DM, Delate T, Garcia DA et al. Risk of thromboembolism, recurrent hemorrhage, and death after warfarin therapy interruption for gastrointestinal tract bleeding. Arch Intern Med 2012; 172: 1484-1491
    • 297 Qureshi W, Mittal C, Patsias I et al. Restarting anticoagulation and outcomes after major gastrointestinal bleeding in atrial fibrillation. Am J Cardiol 2014; 113: 662-668
    • 298 Lee JK, Kang HW, Kim SG et al. Risks related with withholding and resuming anticoagulation in patients with non-variceal upper gastrointestinal bleeding while on warfarin therapy. Int J Clin Pract 2012; 66: 64-68
    • 299 Goodman SG, Clare R, Pieper KS et al. Association of proton pump inhibitor use on cardiovascular outcomes with clopidogrel and ticagrelor: insights from the platelet inhibition and patient outcomes trial. Circulation 2012; 125: 978-986
    • 300 Stockl KM, Le L, Zakharyan A et al. Risk of rehospitalization for patients using clopidogrel with a proton pump inhibitor. Arch Intern Med 2010; 170: 704-710
    • 301 Kreutz RP, Stanek EJ, Aubert R et al. Impact of proton pump inhibitors on the effectiveness of clopidogrel after coronary stent placement: the clopidogrel Medco outcomes study. Pharmacotherapy 2010; 30: 787-796
    • 302 Ho PM, Maddox TM, Wang L et al. Risk of adverse outcomes associated with concomitant use of clopidogrel and proton pump inhibitors following acute coronary syndrome. JAMA 2009; 301: 937-944
    • 303 Huang CC, Chen YC, Leu HB et al. Risk of adverse outcomes in Taiwan associated with concomitant use of clopidogrel and proton pump inhibitors in patients who received percutaneous coronary intervention. Am J Cardiol 2010; 105: 1705-1709
    • 304 Zou JJ, Chen SL, Tan J et al. Increased risk for developing major adverse cardiovascular events in stented Chinese patients treated with dual antiplatelet therapy after concomitant use of the proton pump inhibitor. PLoS One 2014; 9: e84985 DOI: 10.1371/journal.pone. 0084985.
    • 305 van Boxel OS, van Oijen MG, Hagenaars MP et al. Cardiovascular and gastrointestinal outcomes in clopidogrel users on proton pump inhibitors: results of a large Dutch cohort study. Am J Gastroenterol 2010; 105: 2430-2436
    • 306 Munoz-Torrero JF, Escudero D, Suarez C et al. Concomitant use of proton pump inhibitors and clopidogrel in patients with coronary, cerebrovascular, or peripheral artery disease in the factores de Riesgo y ENfermedad Arterial (FRENA) registry. J Cardiovasc Pharmacol 2011; 57: 13-19
    • 307 O’Donoghue ML, Braunwald E, Antman EM et al. Pharmacodynamic effect and clinical efficacy of clopidogrel and prasugrel with or without a proton-pump inhibitor: an analysis of two randomised trials. Lancet 2009; 374: 989-997
    • 308 Hsiao FY, Mullins CD, Wen YW et al. Relationship between cardiovascular outcomes and proton pump inhibitor use in patients receiving dual antiplatelet therapy after acute coronary syndrome. Pharmacoepidemiol Drug Saf 2011; 20: 1043-1049
    • 309 Banerjee S, Weideman RA, Weideman MW et al. Effect of concomitant use of clopidogrel and proton pump inhibitors after percutaneous coronary intervention. Am J Cardiol 2011; 107: 871-878
    • 310 Harjai KJ, Shenoy C, Orshaw P et al. Clinical outcomes in patients with the concomitant use of clopidogrel and proton pump inhibitors after percutaneous coronary intervention: an analysis from the Guthrie Health Off-Label Stent (GHOST) investigators. Circ Cardiovasc Interv 2011; 4: 162-170
    • 311 Aihara H, Sato A, Takeyasu N et al. Effect of individual proton pump inhibitors on cardiovascular events in patients treated with clopidogrel following coronary stenting: results from the Ibaraki Cardiac Assessment Study Registry. Catheter Cardiovasc Interv 2012; 80: 556-563
    • 312 Tentzeris I, Jarai R, Farhan S et al. Impact of concomitant treatment with proton pump inhibitors and clopidogrel on clinical outcome in patients after coronary stent implantation. Thromb Haemost 2010; 104: 1211-1218
    • 313 Schmidt M, Johansen MB, Robertson DJ et al. Concomitant use of clopidogrel and proton pump inhibitors is not associated with major adverse cardiovascular events following coronary stent implantation. Aliment Pharmacol Ther 2012; 35: 165-174
    • 314 Rassen JA, Choudhry NK, Avorn J et al. Cardiovascular outcomes and mortality in patients using clopidogrel with proton pump inhibitors after percutaneous coronary intervention or acute coronary syndrome. Circulation 2009; 120: 2322-2329
    • 315 Ray WA, Murray KT, Griffin MR et al. Outcomes with concurrent use of clopidogrel and proton-pump inhibitors: a cohort study. Ann Intern Med 2010; 152: 337-345
    • 316 Kwok CS, Loke YK. Meta-analysis: the effects of proton pump inhibitors on cardiovascular events and mortality in patients receiving clopidogrel. Aliment Pharmacol Ther 2010; 31: 810-823
    • 317 Siller-Matula JM, Jilma B, Schror K et al. Effect of proton pump inhibitors on clinical outcome in patients treated with clopidogrel: a systematic review and meta-analysis. J Thromb Haemost 2010; 8: 2624-2641

Zoom Image
Fig. 1 Algorithm for the management of patients with acute upper gastrointestinal hemorrhage who are using antiplatelet agent(s): European Society of Gastrointestinal Endoscopy (ESGE) Guideline.
Zoom Image
Fig. 2 Algorithm for the endoscopic management of patients with nonvariceal upper gastrointestinal hemorrhage (NVUGIH) secondary to peptic ulcer, stratified by endoscopic stigmata: European Society of Gastrointestinal Endoscopy (ESGE) Guideline. GI, gastrointestinal; PPI, proton pump inhibitor.
1 Use of a large single-channel or double-channel therapeutic upper GI endoscope is recommended.
2 The benefit of endoscopic hemostasis may be greater in patients at higher risk for rebleeding, e. g., older age, co-morbidities, in-hospital UGIH.
3 Large size 10-Fr probe recommended.
4 Absolute alcohol, polidocanol, or ethanolamine injected in limited volumes.
5 High dose oral PPI may be an option in those able to tolerate oral medications.