Scolaris Content Display Scolaris Content Display

Methods to decrease blood loss during liver resection: a network meta‐analysis

This is not the most recent version

Collapse all Expand all

Abstract

available in

Background

Liver resection is a major surgery with significant mortality and morbidity. Various methods have been attempted to decrease blood loss and morbidity during elective liver resection. These methods include different methods of vascular occlusion, parenchymal transection, and management of the cut surface of the liver. A surgeon typically uses only one of the methods from each of these three categories. Together, one can consider this combination as a treatment strategy. The optimal treatment strategy for liver resection is unknown.

Objectives

To assess the comparative benefits and harms of different treatment strategies that aim to decrease blood loss during elective liver resection.

Search methods

We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, EMBASE, and Science Citation Index Expanded to July 2012 to identify randomised clinical trials. We also handsearched the references lists of identified trials.

Selection criteria

We included only randomised clinical trials (irrespective of language, blinding, or publication status) where the method of vascular occlusion, parenchymal transection, and management of the cut surface were clearly reported, and where people were randomly assigned to different treatment strategies based on different combinations of the three categories (vascular occlusion, parenchymal transection, cut surface).

Data collection and analysis

Two review authors identified trials and collected data independently. We assessed the risk of bias using The Cochrane Collaboration's methodology. We conducted a Bayesian network meta‐analysis using the Markov chain Monte Carlo method in WinBUGS 1.4 following the guidelines of the National Institute for Health and Care Excellence Decision Support Unit guidance documents. We calculated the odds ratios (OR) with 95% credible intervals (CrI) (which are similar to confidence intervals in the frequentist approach for meta‐analysis) for the binary outcomes and mean differences (MD) with 95% CrI for continuous outcomes using a fixed‐effect model or random‐effects model according to model‐fit.

Main results

We identified nine trials with 617 participants that met our inclusion criteria. Interventions in the trials included three different options for vascular occlusion, four for parenchymal transection, and two for management of the cut liver surface. These interventions were combined in different ways in the trials giving 11 different treatment strategies. However, we were only able to include 496 participants randomised to seven different treatment strategies from seven trials in our network meta‐analysis, because the treatment strategies from the trials that used fibrin sealant for management of the raw liver surface could not be connected to the network for any outcomes. Thus, the trials included in the network meta‐analysis varied only in their approaches to vascular exclusion and parenchymal transection and none used fibrin sealant. All the trials were of high risk of bias and the quality of evidence was very low for all the outcomes. The differences in mortality between the different strategies was imprecise (seven trials; seven treatment strategies; 496 participants). Five trials (six strategies; 406 participants) reported serious adverse events. There was an increase in the proportion of people with serious adverse events when surgery was performed using radiofrequency dissecting sealer compared with the standard clamp‐crush method in the absence of vascular occlusion and fibrin sealant. The OR for the difference in proportion was 7.13 (95% CrI 1.77 to 28.65; 15/49 (adjusted proportion 24.9%) in radiofrequency dissecting sealer group compared with 6/89 (6.7%) in the clamp‐crush method). The differences in serious adverse events between the other groups were imprecise. There was a high probability that 'no vascular occlusion with clamp‐crush method and no fibrin' and 'intermittent vascular occlusion with Cavitron ultrasonic surgical aspirator and no fibrin' are better than other treatments with regards to serious adverse events. Quality of life was not reported in any of the trials.

The differences in the proportion of people requiring blood transfusion was imprecise (six trials; seven treatments; 446 participants). Two trials (three treatments; 155 participants) provided data for quantity of blood transfused. People undergoing liver resection by intermittent vascular occlusion had higher amounts of blood transfused than people with continuous vascular occlusion when the parenchymal transection was carried out with the clamp‐crush method and no fibrin sealant was used for the cut surface (MD 1.2 units; 95% CrI 0.08 to 2.32). The differences in the other comparisons were imprecise (very low quality evidence). Three trials (four treatments; 281 participants) provided data for operative blood loss. People undergoing liver resection using continuous vascular occlusion had lower blood loss than people with no vascular occlusion when the parenchymal transection was carried out with clamp‐crush method and no fibrin sealant was used for the cut surface (MD ‐130.9 mL; 95% CrI ‐255.9 to ‐5.9). None of the trials reported the proportion of people with major blood loss.

The differences in the length of hospital stay (six trials; seven treatments; 446 participants) and intensive therapy unit stay (four trials; six treatments; 261 participants) were imprecise. Four trials (four treatments; 245 participants) provided data for operating time. Liver resection by intermittent vascular occlusion took longer than liver resection performed with no vascular occlusion when the parenchymal transection was carried out with Cavitron ultrasonic surgical aspirator and no fibrin sealant was used for the cut surface (MD 49.6 minutes; 95% CrI 29.8 to 69.4). The differences in the operating time between the other comparisons were imprecise. None of the trials reported the time needed to return to work.

Authors' conclusions

Very low quality evidence suggested that liver resection using a radiofrequency dissecting sealer without vascular occlusion or fibrin sealant may increase serious adverse events and this should be evaluated in further randomised clinical trials. The risk of serious adverse events with liver resection using no special equipment compared with more complex methods requiring special equipment was uncertain due to the very low quality of the evidence. The credible intervals were wide and considerable benefit or harm with a specific method of liver resection cannot be ruled out.

PICOs

Population
Intervention
Comparison
Outcome

The PICO model is widely used and taught in evidence-based health care as a strategy for formulating questions and search strategies and for characterizing clinical studies or meta-analyses. PICO stands for four different potential components of a clinical question: Patient, Population or Problem; Intervention; Comparison; Outcome.

See more on using PICO in the Cochrane Handbook.

Plain language summary

available in

Surgical methods to decrease blood loss during liver surgery

Background

Many cancerous and non‐cancerous growths that develop in the liver are treated by removing part of the liver (elective liver resection), which is major surgery with high risk of complications. Severe blood loss is a contributing factor to the complications since blood vessels are cut and blood entering the liver from the general circulation escapes during liver resection. Several methods have been proposed to decrease blood loss during liver resection. This includes blocking the blood supply to the liver during the operation, a process known as vascular occlusion, which could be performed continuously or intermittently. Special equipment using ultrasound waves and high‐frequency alternating current called radiofrequency waves have been proposed as alternatives to using simple surgical instruments in the division of the liver. Once the liver is divided, care is taken to ensure that there is no bleeding from the cut surface of the liver remaining in the body. This can be done by simply using the heat generated by standard alternating current (diathermy machine or electrocautery) or additional glue can be applied to the cut surface to prevent and control bleeding. However, the benefits that these special methods offer in liver resection are unknown. A surgeon typically uses one method from each of these three categories, that is, vascular occlusion (including no vascular occlusion), the use of special equipment to divide the liver, and the way that the cut surface is managed. Together, one can consider this combination of methods as a treatment strategy. The optimal treatment strategy for liver resection is unknown. We sought to clarify this information by performing a literature search to seek the differences in death, complications, quality of life, blood loss and blood transfusion requirements, length of hospital stay, intensive therapy unit stay, and time taken to return to work between the different treatment strategies. We used special statistical methods to compare the different treatments simultaneously as compared to the traditional Cochrane method of comparing two treatments at a time since there are multiple treatment strategies.

Study characteristics

We identified nine trials with 617 participants that met our inclusion criteria. However, we were only able to include 496 participants randomised to seven different treatment strategies from seven trials in our network meta‐analysis because of the complex statistical methodology that we used.

Key results

There was an increase in the proportion of people with major complications when the liver was divided using radiofrequency as compared with using standard surgical instruments. Major complications developed in approximately 25% of people undergoing liver resection using radiofrequency. In comparison, one in 15 people (7%) undergoing liver resection by standard method developed major complications. There was modest reduction in blood loss and number of units of blood transfused when continuous vascular occlusion was used compared with no vascular occlusion or intermittent vascular occlusion. Liver resection by intermittent vascular occlusion took about 50 minutes longer than liver resection performed with no vascular occlusion when the liver was divided using ultrasound. Quality of life and time needed to return to work were not reported in any of the trials. The differences in the other outcomes and other comparisons that were not mentioned above were imprecise.

Quality of evidence

All the trials were at high risk of bias, that is, there is possibility of arriving at wrong conclusions overestimating benefits or underestimating harms of one method or the other because of the way that the studies were conducted. The overall quality of evidence was very low.

Conclusions

We are unable to determine the comparative benefits and harms of different methods of liver resection. Liver resection using radiofrequency energy should be further assessed in randomised clinical trials in order to understand its safety profile better. The risk of serious adverse events following liver resection using no special equipment compared with more complex methods requiring special equipment is uncertain due to the very low quality of the evidence. However, there is considerable uncertainty about this and large benefits or harms with a specific method of liver resection cannot be ruled out.

Authors' conclusions

Implications for practice

Very low quality evidence suggests that liver resection using a radiofrequency dissecting sealer without vascular occlusion or fibrin sealant may increase serious adverse events and this should be evaluated in further randomised clinical trials. The risk of serious adverse events with liver resection using no special equipment compared to more complex methods requiring special equipment is uncertain due to the very low quality of the evidence. The credible intervals were wide and considerable benefit or harm with a specific method of liver resection cannot be ruled out.

Implications for research

Trials need to be conducted and reported according to the SPIRIT (Standard Protocol Items: Recommendations for Interventional Trials) statement (www.spirit‐statement.org/) and the CONSORT (Consolidated Standards for Reporting of Trials) statement (www.consort‐statement.org). Future randomised clinical trials ought to include people at higher anaesthetic risk eligible for liver resection and to employ blinded assessments of outcomes.

Summary of findings

Open in table viewer
Summary of findings for the main comparison. Methods to decrease blood loss during liver resection (mortality)

Methods to decrease blood loss during liver resection (mortality)

Patient or population: people undergoing open liver resection

Settings: secondary or tertiary

Intervention and control: various treatments

Number of trials (participants)

7 trials (496 participants)

Overall quality of evidence

Very low1,2

Groups

Illustrative risk

Treatment effect

Assumed risk in control group (NoVascClampNoFib)*

35 per 1000

Corresponding risk in NoVascCUSANoFib

239 per 1000
(4 to 1000)

OR 6.83 (95% CrI 0.1 to 459.49)

Corresponding risk in NoVascRFAblNoFib

29 per 1000
(1 to 1000)

OR 0.84 (95% CrI 0.02 to 46.5)

Corresponding risk in ContVascClampNoFib

64 per 1000
(1 to 1000)

OR 1.83 (95% CrI 0.04 to 89.57)

Corresponding risk in ContVascSharpNoFib

64 per 1000
(0 to 1000)

OR 1.83 (95% CrI 0 to 2660.48)

Corresponding risk in IntVascClampNoFib

38 per 1000
(2 to 839)

OR 1.1 (95% CrI 0.05 to 23.98)

Corresponding risk in IntVascCUSANoFib

10 per 1000
(0 to 688)

OR 0.29 (95% CrI 0 to 19.67)

*The basis for the assumed risk was from literature. The corresponding risk (and its 95% credible interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CrI).
ContVascClampNoFib: continuous vascular occlusion with clamp‐crush method and no fibrin; ContVascSharpNoFib: continuous vascular occlusion with sharp dissection and no fibrin; CrI: credible interval; IntVascClampNoFib: intermittent vascular occlusion with clamp‐crush method and no fibrin; IntVascCUSANoFib: intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin; NoVascCUSANoFib: no vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin; NoVascRFAblNoFib: no vascular occlusion with radiofrequency dissecting sealer and no fibrin; OR: odds ratio.

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1. The trials were at high risk of bias (2 points).

2. The number of events were fewer than 300 and the credible intervals overlapped 1 and 0.80 or 1.20 (2 points).

Open in table viewer
Summary of findings 2. Methods to decrease blood loss during liver resection (serious adverse events)

Methods to decrease blood loss during liver resection (serious adverse events)

Patient or population: people undergoing open liver resection

Settings: secondary or tertiary

Intervention and control: various treatments

Number of trials (participants)

5 trials (406 participants)

Overall quality of evidence

Very low1,2

Groups

Illustrative risk

Treatment effect

Assumed risk in control group (NoVascClampNoFib)*

67 per 1000

Corresponding risk in NoVascCUSANoFib

95 per 1000
(9 to 977)

OR 2.72 (95% CrI 0.27 to 27.92)

Corresponding risk in NoVascRFAblNoFib

249 per 1000
(62 to 1000)

OR 7.13 (95% CrI 1.77 to 28.65)

Corresponding risk in ContVascClampNoFib

164 per 1000
(26 to 1000)

OR 4.68 (95% CrI 0.74 to 29.47)

Corresponding risk in ContVascSharpNoFib

164 per 1000
(3 to 1000)

OR 4.68 (95% CrI 0.08 to 264.19)

Corresponding risk in IntVascClampNoFib

37 per 1000
(8 to 174)

OR 1.05 (95% CrI 0.22 to 4.96)

*The basis for the assumed risk was from the mean proportion with serious adverse events in control group. The corresponding risk (and its 95% credible interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CrI).
ContVascClampNoFib: continuous vascular occlusion with clamp‐crush method and no fibrin; ContVascSharpNoFib: continuous vascular occlusion with sharp dissection and no fibrin; CrI: credible interval; IntVascClampNoFib: intermittent vascular occlusion with clamp‐crush method and no fibrin; IntVascCUSANoFib: intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin; NoVascCUSANoFib: no vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin; NoVascRFAblNoFib: no vascular occlusion with radiofrequency dissecting sealer and no fibrin; OR: odds ratio.

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1. The trials were at high risk of bias (2 points).

2. The number of events were fewer than 300 and the credible intervals overlapped 1 and 0.80 or 1.20 (2 points).

Open in table viewer
Summary of findings 3. Methods to decrease blood loss during liver resection (blood transfusion proportion)

Methods to decrease blood loss during liver resection (blood transfusion proportion)

Patient or population: people undergoing open liver resection

Settings: secondary or tertiary

Intervention and control: various treatments

Number of trials (participants)

6 trials (446 participants)

Overall quality of evidence

Very low1,2

Groups

Illustrative risk

Treatment effect

Assumed risk in control group (NoVascClampNoFib)

157 per 1000

Corresponding risk in NoVascCUSANoFib

212 per 1000
(0 to 1000)

OR 6.06; 95% CrI 0 to 11,486.87

Corresponding risk in NoVascRFAblNoFib

57 per 1000
(0 to 1000)

OR 1.64; 95% CrI 0.01 to 373.55

Corresponding risk in ContVascClampNoFib

28 per 1000
(0 to 1000)

OR 0.79; 95% CrI 0 to 469.31

Corresponding risk in ContVascSharpNoFib

22 per 1000
(0 to 1000)

OR 0.63; 95% CrI 0 to 4494.19

Corresponding risk in IntVascClampNoFib

119 per 1000
(0 to 1000)

OR 3.4; 95% CrI 0.01 to 941.28

Corresponding risk in IntVascCUSANoFib

211 per 1000
(0 to 1000)

OR 6.03; 95% CrI 0 to 31,671.1

*The basis for the assumed risk was from the mean proportion with blood transfusion in control group. The corresponding risk (and its 95% credible interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CrI).
ContVascClampNoFib: continuous vascular occlusion with clamp‐crush method and no fibrin; ContVascSharpNoFib: continuous vascular occlusion with sharp dissection and no fibrin; CrI: credible interval; IntVascClampNoFib: intermittent vascular occlusion with clamp‐crush method and no fibrin; IntVascCUSANoFib: intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin; NoVascCUSANoFib: no vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin; NoVascRFAblNoFib: no vascular occlusion with radiofrequency dissecting sealer and no fibrin; OR: odds ratio.

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1. The trials were at high risk of bias (2 points).
2. The number of events were fewer than 300 and the credible intervals overlapped 1 and 0.80 or 1.20 (2 points).

Background

Description of the condition

Liver resection refers to removal of part of the liver. On average, 1800 liver resections are carried out in the UK (HES 2011), and 11,000 in the USA (Asiyanbola 2008), every year. In the western world, the main indication for liver resection is colorectal liver metastases. Colorectal cancer is the third most common cancer in the world. Approximately 1.2 million people develop colorectal cancer each year (IARC 2010), and 50% to 60% will have colorectal liver metastases (Garden 2006). Liver resection, the only curative option for people with colorectal liver metastases, is indicated in 20% to 30% of people in whom the metastasis is confined to the liver (Garden 2006). Five‐year survival for people with colorectal liver metastases who undergo liver resection is about 40% (Garden 2006).

The second most common reason for liver resection is hepatocellular carcinoma. Hepatocellular carcinoma is one of the most common cancers, with a worldwide annual incidence of 750,000 people (IARC 2010). Most hepatocellular carcinomas develop in cirrhotic livers (Llovet 2005). Liver resection and liver transplantation are the main curative treatments (Llovet 2005; Taefi 2013). Of people who present with hepatocellular carcinoma, about 5% are suitable for liver resection (Chen 2006). Survival after surgery depends on the stage of cancer and the severity of the underlying chronic liver disease. People with early‐stage disease (cancers smaller than 5 cm) have a five‐year survival of about 50%, whereas people with more advanced disease have a five‐year survival of about 30% (Chen 2006). Screening programmes in theory should lead to a diagnosis at an earlier stage, when surgery is feasible and is associated with better outcomes.

Liver resection may also be performed for benign liver tumours (Belghiti 1993). The liver is subdivided into eight Couinaud segments (Couinaud 1999), which can be removed individually or by right hemi‐hepatectomy (Couinaud segments 5 to 8), left hemi‐hepatectomy (segments 2 to 4), right trisectionectomy (segments 4 to 8), or left trisectionectomy (segments 2 to 5 and 8 ± 1) (Strasberg 2000). Although every liver resection is considered major surgery, only resection of three or more segments is considered a major liver resection (Belghiti 1993).

Blood loss during liver resection is an important factor affecting complications and mortality in people undergoing liver resection (Shimada 1998; Yoshimura 2004; Ibrahim 2006). Variable estimates of blood loss, ranging from 200 mL to 2 L, have been reported (Gurusamy 2009a). Major blood loss during surgery or in the immediate postoperative period may result in death of the patient. Major blood loss can be defined based on the Advanced Trauma Life Support (ATLS definition of class 3 or class 4 shock, where there is a loss of 30% or more of blood volume) (ATLS 2008). During liver resection, the liver parenchyma is transected at the plane of resection. The blood vessels and the bile duct branches in the plane of resection (cut surface) are then sealed by different methods to prevent blood or bile leakage.

Description of the intervention

Various interventions have been attempted to decrease blood loss during liver resection. These interventions include temporary occlusion of the blood vessels that supply the liver (Gurusamy 2009a; Table 1); different methods of liver transection (the way that the liver parenchyma is divided), such as the clamp‐crush method, the Cavitron ultrasonic surgical aspirator, or the radiofrequency dissecting sealer (Gurusamy 2009b; Table 2); and different methods of management of the cut surface of the liver (the way that the resection plane of the remnant liver is managed), such as use of fibrin sealant, argon beamer, or electrocautery and suture material (Frilling 2005; Table 3).

Open in table viewer
Table 1. Different methods of vascular occlusion

No vascular occlusion

Portal triad clamping (continuous) (occlusion of inflow alone)

Portal triad clamping (intermittent) (occlusion of inflow alone)

Hepatic vascular exclusion (occlusion of inflow and outflow)

Selective vascular occlusion (occlusion of inflow to the hemi‐liver that is being resected)

Selective hepatic artery occlusion (occlusion of hepatic artery supplying the hemi‐liver that is being resected)

Selective portal vein occlusion (occlusion of portal vein supplying the hemi‐liver that is being resected)

Selective hepatic vascular exclusion (occlusion of inflow to the hemi‐liver and outflow from the hemi‐liver that is being resected)

Open in table viewer
Table 2. Different methods of parenchymal transection

Finger‐fracture method

Clamp‐crush method

Cavitron ultrasonic surgical aspirator (CUSA)

Sharp dissection

Radiofrequency dissecting sealer

Ultrasonic shears

Open in table viewer
Table 3. Different methods of dealing with raw surface

Suturing for large and medium vessels and ducts and performing electrocauterisation of small vessels and ducts

Suturing for large vessels and performing ultrasonic shears for medium‐sized and small vessels and ducts

Suturing and argon beam coagulator

Suturing and fibrin sealant

Interventions selected to decrease blood loss can be used alone or in various combinations (Table 4). Usually surgeons at different centres follow their own protocol for decreasing blood loss. The finger‐fracture and clamp‐crush techniques do not involve specialist equipment. The minimum and standard method of managing the cut surface involves electrocautery and suture material. Altogether, the goal of these interventions is to decrease blood loss and the associated morbidity and mortality.

Open in table viewer
Table 4. Different categories of vascular occlusion, parenchymal transection, and methods of dealing with raw surface used in this review

Vascular occlusion

No vascular occlusion (NoVasc)

Continuous vascular occlusion (ConVasc)

Intermittent vascular occlusion (IntVasc)

Parenchymal transection

Finger‐fracture method (Finger)

Clamp‐crush method (Clamp)

Cavitron ultrasonic surgical aspirator (CUSA)

Sharp dissection (Sharp)

Radiofrequency dissecting sealer (RFAbl)

Ultrasonic shears (UShears)

Methods of dealing with raw surface

No fibrin sealant used (NoFib)

Fibrin sealant used (Fib)

How the intervention might work

Temporarily occluding the blood vessels that supply the liver may reduce the blood flow through the cut vessels. Different methods of liver transection are used to remove the liver parenchyma, so that damage to the blood vessels is minimized. This might result in clear visualisation of the blood vessels, which can be clamped and then divided. Different topical methods of managing the cut surface attempt to seal the blood vessels on the resection plane, preventing blood loss.

Why it is important to do this review

Liver resection is a major surgical procedure with significant mortality (estimated at 4%) and morbidity (estimated at 40%) (Reissfelder 2011). Interventions that decrease blood loss may improve outcomes of liver resection. Each category of interventions has been systematically reviewed previously (Gurusamy 2009a; Gurusamy 2009b). However, to our knowledge, no review has been conducted to assess and synthesise the comparative effectiveness of specific combinations of interventions when used together to decrease blood loss and associated morbidity and mortality. This systematic review is intended as a useful guide for patients and healthcare providers as they seek to understand the role of different combinations of interventions (treatment strategies) in decreasing blood loss and blood transfusion requirements in people undergoing elective liver resection.

Objectives

To assess the comparative benefits and harms of different treatment strategies that aim to decrease blood loss during elective liver resection.

Methods

Criteria for considering studies for this review

Types of studies

We considered only randomised clinical trials for this overview. We excluded studies of other design.

Types of participants

We included randomised clinical trials in which participants underwent elective liver resection using different types of vascular occlusion or no vascular occlusion, irrespective of the method of vascular occlusion or the nature of the background liver (i.e., normal or cirrhotic), different types of parenchymal transection, or different types of management of cut surface. We excluded randomised clinical trials in which participants underwent liver resection combined with other major surgical procedures (e.g., one‐stage liver and bowel resection for synchronous metastases from colorectal tumours).

Types of interventions

We included randomised clinical trials that assessed one or more of the following interventions in this review.

  1. Methods of vascular occlusion (including no vascular occlusion).

  2. Methods of liver parenchymal transection.

  3. Methods of management of the cut surface (resection plane) of the liver.

The surgeon (and hence the trialists) may use a particular combination of each of the above. For example, one surgeon may perform liver resection using intermittent vascular occlusion, clamp‐crush technique as the method of liver parenchymal transection, and a fibrin sealant on the cut surface; while another surgeon may perform liver resection without using any method of vascular occlusion, with the Cavitron ultrasonic surgical aspirator as the method of liver parenchymal transection, and without any fibrin sealant on the cut surface. Each combination was assessed as a treatment strategy, that is, a combination of several interventions. The purpose of this review was to identify the overall treatment effect of a treatment strategy rather than the contribution of each component intervention towards the overall effect.

Commonly used surgical techniques under each of the above categories are listed in Table 1, Table 2, and Table 3. In practice, any intervention in Table 1 can be used in combination with an intervention from Table 2 or Table 3. Any intervention in Table 2 can be used in combination with an intervention from Table 3. However, because of the few trials that could be included for network meta‐analysis (sparse data) in this review, we revised the categories of vascular occlusion, method of parenchymal transection, and dealing with the cut surface to those shown in Table 4.

Types of outcome measures

We assessed the comparative effectiveness of available treatment strategies that aimed to decrease blood loss during liver resection for the following outcomes.

Primary outcomes

  1. Mortality.

    1. Short‐term (30‐day mortality or in‐hospital mortality). We used in‐hospital mortality as defined in the included trials.

    2. Long‐term (at maximal follow‐up).

  2. Serious adverse events. An adverse event was defined as any untoward medical occurrence not necessarily having a causal relationship with the treatment but resulting in a dose reduction or discontinuation of treatment (ICH‐GCP 1997). A serious adverse event was defined as any event that would increase mortality; was life‐threatening; required inpatient hospitalisation; or resulted in persistent or significant disability; or any important medical event that might have jeopardised the person or required intervention to prevent it. Serious adverse events correspond approximately to Grade III or above of the Clavien‐Dindo classification ‐ the only validated system for classifying postoperative complications (Dindo 2004; Clavien 2009;Table 5). In cases where the authors did not classify the severity of adverse events, we followed the criteria provided in Table 5 to classify the severity.

    Open in table viewer
    Table 5. Clavien‐Dindo classification of postoperative complications

    Grades

    Definitions

    Examples

    I

    Any deviation from the normal postoperative course without the need for pharmacological treatment or surgical, endoscopic, and radiological interventions

    Drugs such as antiemetics, antipyretics, analgesics, diuretics, and electrolytes; physiotherapy; wound infections opened at the bedside

    II

    Requiring pharmacological treatment with drugs other than those allowed for grade I complications

    Blood transfusions, total parenteral nutrition

    III

    Requiring surgical, endoscopic or radiological intervention

    Bile leak requiring endoscopic stent; re‐operation for any cause; drainage of infected intra‐abdominal collection

    IV

    Life‐threatening complication requiring high dependency or intensive care management

    Dialysis

    V

    Death of patient

    Suffix d

    If the patient suffers from a complication at the time of discharge and needs further follow‐up to evaluate the complication fully

    Adapted from Dindo 2004; Clavien 2009.

  3. Quality of life as defined in the included trials.

    1. Short‐term (30 days, three months).

    2. Long‐term (maximal follow‐up).

Secondary outcomes

  1. Blood transfusion requirements.

    1. Number of participants who required red cell or whole blood heterologous blood transfusion.

    2. Quantity of blood transfusion (heterologous red cell or whole blood product, platelet, and fresh frozen plasma).

    3. Total operative blood loss.

    4. Number of participants who had major operative blood loss.

  2. Hospital stay.

    1. Length of total hospital stay (including re‐admissions).

    2. Intensive therapy unit stay.

  3. Operating time.

  4. Time needed to return to work.

Search methods for identification of studies

Electronic searches

We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, EMBASE, and Science Citation Index Expanded (Royle 2003) to 16 July 2012. We also searched the World Health Organization International Clinical Trials Registry Platform search portal, which searches various trial registers, including ISRCTN and ClinicalTrials.gov (apps.who.int/trialsearch/Default.aspx) to identify further trials. Because subsets of all available interventions on this topic have been reviewed comprehensively in existing Cochrane systematic reviews (Gurusamy 2009a; Gurusamy 2009b), we also used these reviews as a way to identify trials. Search strategies with time spans of the searches are available in Appendix 1.

Searching other resources

We searched the references of the identified trials to identify additional trials for inclusion.

Data collection and analysis

Selection of studies

Two review authors (CS and JV) independently identified the trials for inclusion by screening the titles and abstracts. We sought full text for any references that were identified for potential inclusion by at least one of the authors. We made further selection for inclusion based on the full text. We have listed the full texts of references that we excluded with reasons for the exclusion (Characteristics of excluded studies table). We planned to list any ongoing trials identified primarily through World Health Organization International Clinical Trials Registry Platform for further follow‐up. We resolved discrepancies through discussion.

Data extraction and management

Two review authors (CS and JV) independently extracted the following data.

  1. Year and language of publication.

  2. Country in which the participants were recruited.

  3. Year(s) in which the trial was conducted.

  4. Inclusion and exclusion criteria.

  5. Participant characteristics such as age, sex, underlying disease, comorbidity, number and proportion of participants with cirrhosis, and number and proportion of participants undergoing major versus minor liver resection.

  6. Details of the intervention and treatment strategy that aimed to decrease blood loss and blood transfusion requirements (e.g., surgical technique, procedure and co‐intervention, concurrent surgery, and medications).

  7. Outcomes (Primary outcomes; Secondary outcomes).

  8. Follow‐up time points.

  9. Risk of bias (Assessment of risk of bias in included studies).

We sought unclear or missing information by contacting the authors of the individual trials. If there was any doubt whether trials shared the same participants ‐ completely or partially (by identifying common authors and centres) ‐ we planned to contact the authors of the trials to clarify whether the trial report was duplicated. We resolved any differences in opinion through discussion.

Assessment of risk of bias in included studies

We followed the guidance given in the Cochrane Handbook for Systematic Reviews of Intervention (Higgins 2011), and those described in the Cochrane Hepato‐Biliary Group Module (Gluud 2013), to assess the risk of bias in included studies. Specifically, we assessed the risk of bias in included trials for the following domains (Schulz 1995; Moher 1998; Kjaergard 2001; Wood 2008; Lundh 2012; Savovic 2012a; Savovic 2012b).

Allocation sequence generation 

  • Low risk of bias: sequence generation was achieved using computer random number generation or a random number table. Drawing lots, tossing a coin, shuffling cards, and throwing dice were adequate if performed by an independent adjudicator.

  • Uncertain risk of bias: the trial was described as randomised, but the method of sequence generation was not specified.

  • High risk of bias: the sequence generation method was not, or may not have been, random. Quasi‐randomised studies (those using dates, names, or admittance numbers to allocate participants) were inadequate and were excluded for the assessment of benefits but not for assessing harms.

Allocation concealment

  • Low risk of bias: allocation was controlled by a central and independent randomisation unit, sequentially numbered, opaque and sealed envelopes, or something similar, so that intervention allocations could not have been foreseen in advance of, or during, enrolment.

  • Uncertain risk of bias: the trial was described as randomised, but the method used to conceal the allocation was not described, so that intervention allocations may have been foreseen in advance of, or during, enrolment.

  • High risk of bias: if the allocation sequence was known to the investigators who assigned participants, or the study was quasi‐randomised. Quasi‐randomised studies were excluded for assessment of benefits but not for assessment of harms.

Blinding of participants and personnel

  • Low risk of bias: blinding was performed adequately, or the outcome measurement was not likely to be influenced by lack of blinding.

  • Uncertain risk of bias: information was insufficient to allow assessment of whether the type of blinding used was likely to induce bias on the estimate of effect.

  • High risk of bias: no blinding or incomplete blinding and the outcome or the outcome measurements were likely to be influenced by lack of blinding.

Blinding of outcome assessors

  • Low risk of bias: blinding was performed adequately, or the outcome measurement was not likely to be influenced by lack of blinding.

  • Uncertain risk of bias: information was insufficient to allow assessment of whether the type of blinding used was likely to induce bias on the estimate of effect.

  • High risk of bias: no blinding or incomplete blinding and the outcome or the outcome measurements were likely to be influenced by lack of blinding.

Incomplete outcome data

  • Low risk of bias: the underlying reasons for missing data were unlikely to make treatment effects depart from plausible values, or proper methods have been employed to handle missing data.

  • Uncertain risk of bias: information was insufficient to allow assessment of whether the missing data mechanism in combination with the method used to handle missing data was likely to induce bias on the estimate of effect.

  • High risk of bias: the crude estimate of effects (e.g., complete case estimate) were clearly biased because of the underlying reasons for missing data, and the methods used to handle missing data were unsatisfactory.

Selective outcome reporting

  • Low risk of bias: pre‐defined or clinically relevant and reasonably expected outcomes (mortality and serious adverse events) were reported.

  • Uncertain risk of bias: not all pre‐defined or clinically relevant and reasonably expected outcomes were reported, or they were not reported fully, or it was unclear whether data on these outcomes were recorded.

  • High risk of bias: one or more clinically relevant and reasonably expected outcomes were not reported; data on these outcomes were likely to have been recorded.

Vested interest bias

  • Low risk of bias: the trial was conducted by a party with no vested interests (i.e., a party benefiting from the results of the trial) in the outcome of the trial.

  • Uncertain risk of bias: It was not clear if the trial was conducted by a party with vested interest in the outcome of the trial.

  • High risk of bias: the trial was conducted by a party with vested interests in the outcome of the trial (such as a drug manufacturer).

We considered a trial at low risk of bias if the trial was assessed as at low risk of bias for all domains. We considered a trial at low risk of bias for an outcome if the trial was assessed as at low risk of bias for all study level domains, as well as for outcome‐specific domains (e.g., blinding, incomplete outcome data). Otherwise, trials with uncertain risk of bias or with high risk of bias regarding one or more domains were considered trials with high risk of bias.

Measures of treatment effect

For dichotomous variables (short‐term mortality, serious adverse events, participants requiring blood transfusion), we calculated the odds ratio (OR) with 95% credible interval (CrI). For continuous variables, such as quantity of blood transfused, blood loss, hospital stay, and operating time, we calculated the mean difference (MD) with 95% CrI. We planned to use MD and 95% CrI for time needed to return to work but did not use this since none of the included trials reported this outcome. We planned to use standardised mean difference (SMD) with 95% CrI for quality of life if different scales were used (but did not plan to combine the quality of life at different time points) and for the quantity of blood transfused (some authors report this in litres transfused, while others report this as number of units transfused). For time‐to‐event data, such as long‐term survival, we planned to use hazard ratio (HR) with 95% CrI.

We have also presented the 'Summary of findings' tables using GRADEpro (ims.cochrane.org/revman/other‐resources/gradepro) for each outcome.

Unit of analysis issues

The unit of analysis were the people undergoing elective liver resection according to the intervention group to which they were randomly assigned.

Dealing with missing data

We performed an intention‐to‐treat analysis (Newell 1992), whenever possible. Otherwise, we used data that were available to us (e.g., a trial may have reported only per‐protocol analysis results). As such 'per protocol' analyses may be biased, we planned to conduct best‐worst case scenario and worst‐best case scenario analyses as sensitivity analyses.

For continuous outcomes, we imputed the standard deviation from P values according to guidance given in the Cochrane Handbook for Systematic Reviews of Intervention (Higgins 2011). If the data were likely to be normally distributed, we used the median for meta‐analysis when the mean was not available. If it was not possible to calculate the standard deviation from the P value or the confidence intervals, we imputed the standard deviation using the largest standard deviation in other trials for that outcome. This form of imputation may decrease the weight of the study for calculation of mean differences and may bias the effect estimate to no effect for calculation of SMDs (Higgins 2011).

Assessment of heterogeneity

We assessed clinical and methodological heterogeneity by carefully examining the characteristics and design of included trials. Major sources of clinical heterogeneity included cirrhotic compared to non‐cirrhotic livers and major compared to minor liver resections. In addition, we anticipated considerable heterogeneity in the way the intervention was performed. For example, intermittent portal triad clamping may be performed with different time periods of occlusion and non‐occlusion. In addition, different doses of fibrin sealant may be used. Different study design and risk of bias may contribute to methodological heterogeneity.

We used the residual deviance and Deviance Information Criteria (DIC) for assessing between study heterogeneity as per the guidance from the National Institute for Health and Care Excellence (NICE) Decision Support Unit (DSU) Technical Support Documents (Dias 2012b; Dias 2013). We also calculated the between‐trial standard deviation and have reported this if we used a random‐effects model. See Data synthesis for further details regarding residual deviance, DIC, and choice of model.

If substantial heterogeneity was identified ‐ clinical, methodological, or statistical ‐ we planned to explore and address heterogeneity in a subgroup analysis (see section on Subgroup analysis and investigation of heterogeneity).

Assessment of reporting biases

We planned to use visual asymmetry on a funnel plot to explore reporting bias in case at least 10 trials were included for direct comparison (Egger 1997; Macaskill 2001). In the presence of heterogeneity that could be explained by subgroup analysis, we planned to perform the funnel plot for each subgroup in the presence of the adequate number of trials. We planned to perform the linear regression approach described by Egger 1997 to determine the funnel plot asymmetry. However, these were not performed because of the lack of an adequate number of trials.

We also considered selective reporting as evidence of reporting bias.

Data synthesis

We planned to apply classifications described in Table 1, Table 2, and Table 3 to categorise different methods of vascular occlusion and parenchymal transection, as well as methods used to manage the cut surface of the liver. Each category in the table is broadly defined to encompass a relatively homogeneous group of interventions, although we anticipate that variations will be noted in the way each method is carried out. For example, intermittent portal triad clamping may be performed with different time periods of occlusion and non‐occlusion. We categorised them under intermittent portal triad clamping regardless of the time intervals. Likewise, we did not distinguish different maximum periods for continuous vascular occlusion (Clavien 1996), and did not determine whether the suprahepatic inferior vena cava or the hepatic veins were occluded for outflow obstruction. These practice variations might be a source of heterogeneity; however, evidence was insufficient to suggest that these variations may affect the outcome.

In liver resection, a surgeon typically uses one item from Table 1, one item from Table 2, and one item from Table 3. Together, one can consider this combination of one method from each table as a treatment strategy, or in terms of network meta‐analysis, each unique treatment strategy can be defined as a 'node'. Because of the large number of possible treatment strategies (eight methods of vascular occlusion × six methods of parenchymal transection × four methods of treatment of cut surface, i.e., 192 potential treatment strategies or nodes), we planned to construct a more sparse network graph based on treatment strategies used in the trials that we identified. We did not expect that all 192 nodes would be represented in the trials available in the literature. However, since the data were sparse, we categorised the treatments into fewer categories by having only three methods of vascular occlusion (no vascular occlusion, continuous vascular occlusion, or intermittent vascular occlusion) and by having only two methods of treatment of cut surface (fibrin sealant used or no fibrin sealant used) (Table 4) as indicated in the protocol. This reduced the categories to 36 treatment strategies or nodes (three methods of vascular occlusion × six methods of parenchymal transection × two methods of treatment of cut surface).

We did not anticipate that every node would be represented. Some methods are more commonly practiced than others. From Table 1, no vascular occlusion, intermittent portal triad clamping, and continuous portal triad clamping are used more often than other techniques (Gurusamy 2009a). From Table 2, clamp‐crush method and Cavitron ultrasonic surgical aspirator are more commonly applied (Gurusamy 2009b). The clamp‐crush method and the finger‐fracture method do not require any special equipment, but the remaining methods do require special equipment. From Table 3, common methods of managing cut surface include suturing for large and medium vessels and ducts and performing electrocauterisation of small vessels and ducts (Gurusamy 2009b).

Direct comparison

We planned to perform pair‐wise meta‐analyses using Review Manager 5 (RevMan 2012), in accordance with recommendations of The Cochrane Collaboration (Higgins 2011), and those described in the Cochrane Hepato‐Biliary Group Module (Gluud 2013). We planned to use both random‐effects models (DerSimonian 1986) and fixed‐effect models (DeMets 1987) for the meta‐analyses. In case of discrepancy between the two models, we planned to report the results of both; otherwise, we planned to report results of the random‐effects model. We planned to use the generic inverse method to combine the HRs for time‐to‐event outcome data.

We did not perform direct comparisons. This was because of the exclusion of many trials that might have been suitable for direct comparison but were unsuitable for the overview. Although these trials included comparisons of one aspect of different methods of vascular occlusion or parenchymal transection or management of cut surface, one or more aspects of methods of vascular occlusion or parenchymal transection or management of cut surface not being compared were either not stated or were chosen in a non‐random manner. Therefore, these trials had to be excluded for this review while such trials would be eligible for inclusion in a direct comparison involving only one aspect of methods of vascular occlusion or parenchymal transection or management of cut surface. For example, a trial comparing vascular occlusion versus no vascular occlusion provided details on the method of vascular occlusion (Table 1) but may not provide details of the parenchymal dissection (Table 2) or the method of dealing with cut surface (Table 3). Since the objective of this review was to assess the overall effect of the different components and not to assess the effect of each individual component, we excluded such trials. Performing and reporting the direct comparison after excluding such trials may not provide the same effect estimate as that obtained if such trials were included. Stakeholders interested in the effects of the individual components should refer to the reviews where the objectives were to assess the benefits and harms of individual components (Gurusamy 2009a; Gurusamy 2009b).

Network meta‐analysis

We conducted network meta‐analyses to compare multiple interventions simultaneously for each of the outcomes, primary outcomes and one secondary outcome on blood transfusion requirements. Network meta‐analysis combines direct evidence within trials and indirect evidence across trials (Mills 2012).

We obtained a network plot to ensure that the trials were connected by treatments using Stata/IC 11 (StataCorp LP). We excluded any trials that were not connected to the network. We conducted a Bayesian network meta‐analysis using the Markov chain Monte Carlo method in WinBUGS 1.4. We modelled the treatment contrast (e.g., log OR for binary outcomes, MD for continuous outcomes) for any two interventions ('functional parameters') as a function of comparisons between each individual intervention and an arbitrarily selected reference group ('basic parameters') (Lu 2004). The reference group was selected on the basis of the 'least intervention', for example, if a treatment group had no vascular occlusion, used finger‐fracture or clamp‐crush method for parenchymal transection, and no fibrin sealant for dealing with the cut surface, this treatment was used as the reference category. We performed the network analysis as per the guidance from The NICE DSU documents (Dias 2013). Further details of the codes used, the raw data, and the technical details of how we performed the analysis are shown in Appendix 2, Appendix 3, and Appendix 4. The codes allow handling of trials with multiple arms to be dealt in the same way as two‐arm trials, that is, one can enter the data from all the arms in a trial as number of events and the number of people exposed to the event for binary outcomes or the mean and standard error for continuous outcomes. The choice of the model between fixed‐effect model and random‐effects model was based on the model fit as per the guidelines of the NICE TSU (Dias 2013). We have reported the treatment contrasts (i.e., log ORs for binary outcomes and MDs for continuous outcomes) of the different treatments in relation to the reference treatment, the deviance residuals, number of effective parameters, and DIC for fixed‐effect model and random‐effects model for each outcome. We have also reported the parameters used to assess the model fit (i.e., deviance residuals, number of effective parameters, and DIC) for the inconsistency model for all the outcomes where there was evidence from direct and indirect comparisons. We have reported estimates of treatment effects (ORs for binary outcomes and MDs for continuous outcomes). The 95% CrIs are calculated in the Bayesian meta‐analysis, which is similar in use to the 95% confidence intervals in the frequentist meta‐analysis. We have calculated the 95% CrI from the mean and variance and have reported the effect estimates and associated 95% CrI for each pair‐wise comparison in a table. We have also estimated the probability that each intervention ranks at one of the possible positions. We have presented the probability that a treatment ranks as the best treatment in graphs. It should be noted that a less than 90% probability that the treatment is the best treatment is unreliable (Dias 2012a). We have also presented the cumulative probability of the treatment ranks (i.e., the probability that the treatment is within the top two, the probability that the treatment is within the top three, etc.) in graphs. We have also plotted the probability that each treatment is best for each of the different outcomes (rankogram), which are generally considered more informative (Salanti 2011; Dias 2012a).

Sample size calculations

To control for the risk of random errors, we interpreted the information with caution when the accrued sample size in the meta‐analysis was less than the required sample size (required information size). For calculation of the required information size, please see Appendix 5.

Subgroup analysis and investigation of heterogeneity

We planned to perform the following subgroup analyses when at least one trial was included in each subgroup.

  1. Trials with low risk of bias compared to trials with high risk of bias.

  2. Cirrhotic compared to non‐cirrhotic livers.

  3. Major liver resections compared to minor liver resections.

We planned to use the Chi2 test to identify subgroup differences. We planned to consider a P value < 0.05 as statistically significant. We also planned to use meta‐regression to assess the impact of cirrhotic versus non‐cirrhotic livers and major versus minor liver resections on effect estimates in the presence of at least 10 trials with this information.

We did not perform any of the above because of the few trials included in this network meta‐analysis.

Sensitivity analysis

Reporting of the severity of adverse events may be inadequate or incomplete. For example, minor bile leaks are considered mild adverse events, and major bile leaks are considered serious adverse events. In cases where the severity could not be determined, we planned to exclude those events from the main analysis. We planned to perform a sensitivity analysis to include those events and treat them as severe adverse events in the sensitivity analysis. We did not perform this since we were able to assess the severity of the reported complications.

Results

Description of studies

Results of the search

We identified 1347 references through electronic searches of CENTRAL (N =170), MEDLINE (N = 370), EMBASE (N = 442), Science Citation Index Expanded (N = 364), and randomised controlled trials registers (N = 1). We excluded 494 duplicates and 768 clearly irrelevant references through screening titles and reading abstracts. We retrieved 85 references for further assessment. No references were identified through scanning reference lists of the identified randomised trials. We excluded 76 references (73 studies) for the reasons listed under the table Characteristics of excluded studies. In total, nine references of nine completed randomised clinical trials met the inclusion criteria (Belghiti 1999; Capussotti 2003; Lesurtel 2005; Smyrniotis 2005; Capussotti 2006; Petrowsky 2006; Lupo 2007; Doklestic 2011; Park 2012). This is summarised in the study flow diagram (Figure 1).


Study flow diagram.

Study flow diagram.

Included studies

The treatments used in the nine randomised clinical trials have been summarised in Characteristics of included studies table and in Table 6. All the trials assessed different methods of open liver resection by using different combinations of vascular exclusion, parenchymal transection, and management of the liver cut surface, in order to decrease blood loss during liver resection. Seven trials were two‐arm trials (Belghiti 1999; Capussotti 2003; Smyrniotis 2005; Capussotti 2006; Petrowsky 2006; Lupo 2007; Park 2012). There was one three‐arm trial (Doklestic 2011), and one four‐arm trial (Lesurtel 2005). However, one arm in each of these trials was excluded since the method of parenchymal transections used in these trials (parenchymal transection using bipolar cautery and water jet) were not included in this review (Lesurtel 2005; Doklestic 2011). Eleven different treatments out of 36 possible treatments were included in the studies included in this review. A total of 617 participants were randomised to the 11 different treatments in these trials. However, four treatment strategies in two trials were not connected to the network in any of the outcomes (Belghiti 1999; Capussotti 2003). Thus, we included 496 participants randomised to seven different treatment strategies in the seven trials that contributed data for the network meta‐analysis (Lesurtel 2005; Smyrniotis 2005; Capussotti 2006; Petrowsky 2006; Lupo 2007; Doklestic 2011; Park 2012).

Open in table viewer
Table 6. Summary of treatments used and types of participants included

Study

Vascular occlusion

Parenchymal transection

Liver raw surface

Codes of the comparisons

Number of participants

Major liver resections

Cirrhosis

Belghiti 1999

Intermittent vascular occlusion

versus

continuous

vascular occlusion

Cavitron ultrasonic surgical aspirator (CUSA)

Fibrin sealant

IntVascCUSAFib versus ContVascCUSAFib

86

20 (23.3%)

Not stated

Capussotti 2003

Intermittent vascular occlusion versus

continuous vascular occlusion

Clamp‐crush

Fibrin sealant

IntVascClampFib versus ContVascClampFib

35

8 (22.9%)

35 (100%)

Capussotti 2006

Intermittent vascular occlusion versus

no vascular occlusion

Clamp‐crush

No fibrin sealant

IntVascClampNoFib versus NoVascClampNoFib

126

56 (44.4%)

19 (15.1%)

Doklestic 2011

Intermittent vascular occlusion versus

continuous vascular occlusion

Clamp‐crush

No fibrin sealant

IntVascCUSANoFib versus IntVascClampNoFib

40

10 (25.0%)

0 (0%)

Lesurtel 2005

No vascular occlusion versus

no vascular occlusion versus

continuous vascular occlusion

CUSA versus radiofrequency dissecting sealer versus clamp‐crush method

No fibrin sealant

NoVascCUSANoFib versus NoVascRFAblNoFib versus ContVascClampNoFib

75

42 (56.0%)

0 (0%)

Lupo 2007

No vascular occlusion

Radiofrequency dissecting sealer versus

clamp‐crush

No fibrin sealant

NoVascRFAblNoFib versus NoVascClampNoFib

50

Not stated

Not stated

Park 2012

Intermittent vascular occlusion versus no vascular occlusion

CUSA

No fibrin sealant

IntVascCUSANoFib versus NoVascCUSANoFib

50

Not stated

Not stated

Petrowsky 2006

Intermittent vascular occlusion versus continuous vascular occlusion

Clamp‐crush

No fibrin sealant

IntVascClampNoFib versus ContVascClampNoFib

73

37 (50.7%)

Not stated

Smyrniotis 2005

Continuous vascular occlusion

Sharp dissection versus clamp‐crush

No fibrin sealant

ContVascSharpNoFib versus ContVascClampNoFib

82

Not stated

6 (7%)

Excluded studies

Of the 73 studies excluded, 24 studies were excluded because they were not randomised clinical trials (Taniguchi 1992; Shimada 1994; Rau 1995; Johnson 1998; Cherqui 1999; Man 2002; Smyrniotis 2002; Smyrniotis 2003; Chau 2005; Nagano 2005; Noritomi 2005; Sugo 2005; Aldrighetti 2006; Felekouras 2006; Wu 2006; Chiappa 2007; Kim 2007; Xia 2008; Cresswell 2009; Fu 2010; Pietsch 2010; Wang 2011; Palibrk 2012; Yokoo 2012). One report was the protocol of a trial (Rahbari 2009). Seven trials did not compare different methods of vascular occlusion or parenchymal transection or method of management of cut surface (Lentschener 1997; Hasegawa 2002; Matot 2002; Yao 2006; Hashimoto 2007; Kato 2008; Guo 2010). One trial included participants undergoing liver resection along with other major procedures (Figueras 2007). Four trials compared variations of methods of vascular occlusion that would have been classified under the same treatment categories included in this review (Wu 2002; Chen 2006a; Fu 2011; Van Den Broek 2011). The remaining 36 trials included comparisons of one aspect of different methods of vascular occlusion or parenchymal transection or management of cut surface. However, one or more aspects of methods of vascular occlusion or parenchymal transection or management of cut surface not being compared were either not stated or were chosen in a non‐random manner. Therefore, we have excluded these trials (Kohno 1992; Belghiti 1996; Noun 1996; Man 1997; Chapman 2000; Takayama 2001; Figueras 2003; Man 2003; Wong 2003; Chouker 2004; El‐Kharboutly 2004; Schwartz 2004; Arita 2005; Figueras 2005; Frilling 2005; Koo 2005; Lodge 2005; Esaki 2006; Saiura 2006; Wang 2006; Campagnacci 2007; Chapman 2007; Izzo 2008; Kim 2008; Schmidt 2008; El‐Moghazy 2009; Ikeda 2009; Liang 2009; Richter 2009; Dello 2011; Fischer 2011; Gugenheim 2011; Mirza 2011; Rahbari 2011; Scatton 2011; Capussotti 2012).

Risk of bias in included studies

The risk of bias in the included trials is summarised in Figure 2 and Figure 3. All trials were at high risk of bias.


Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.


Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Allocation

Four trials (44%) had adequate sequence generation (Capussotti 2003; Capussotti 2006; Lupo 2007; Park 2012). Two trials (22%) had adequate allocation concealment (Petrowsky 2006; Doklestic 2011). Thus, no trials (0%) had low risk of bias due to allocation.

Blinding

None of the trials reported any blinding.

Incomplete outcome data

Eight of the nine trials (89%) were free from bias due to incomplete outcome data (Belghiti 1999; Lesurtel 2005; Smyrniotis 2005; Capussotti 2006; Petrowsky 2006; Lupo 2007; Doklestic 2011; Park 2012).

Selective reporting

Seven trials (78%) reported mortality and serious adverse events and hence were considered to be free from bias (Belghiti 1999; Capussotti 2003; Lesurtel 2005; Smyrniotis 2005; Capussotti 2006; Petrowsky 2006; Lupo 2007).

Other potential sources of bias

Only one trial reported the source of funding and we rated the vested interest bias to be low in this trial (Doklestic 2011). The remaining trials were at unclear risk of bias.

Effects of interventions

See: Summary of findings for the main comparison Methods to decrease blood loss during liver resection (mortality); Summary of findings 2 Methods to decrease blood loss during liver resection (serious adverse events); Summary of findings 3 Methods to decrease blood loss during liver resection (blood transfusion proportion)

All the data used for analysis are provided in Appendix 3. Analyses in this section were based on the 496 participants in the seven trials that contributed data for the network meta‐analysis (Lesurtel 2005; Smyrniotis 2005; Capussotti 2006; Petrowsky 2006; Lupo 2007; Doklestic 2011; Park 2012).

Mortality

All the seven trials (496 participants) provided data for the network meta‐analysis on short‐term mortality (Lesurtel 2005; Smyrniotis 2005; Capussotti 2006; Petrowsky 2006; Lupo 2007; Doklestic 2011; Park 2012). There were seven deaths in the included studies giving an overall mortality of 1.4%. The network plot is shown in Figure 4. Although there is no need to add an arbitrary constant of 0.5 to each of the cells for occasional zero‐event trials when the meta‐analysis is performed using the Bayesian methods (Dias 2013), we had to add this arbitrary constant in our meta‐analysis since many of the trials had no deaths reported.


Network plot of mortality Treatment codes are provided in .

Network plot of mortality

Treatment codes are provided in Table 6.

The results and model‐fit of the fixed‐effect model and random‐effects model along with the model‐fit of the inconsistency model is provided in Table 7. The between‐study standard deviation (tau) was 0.60. As indicated in Table 7, the fixed‐effect model was preferred based on the DIC statistics. There was no evidence of inconsistency in the network. The pair‐wise ORs for the different treatment comparisons are shown in Table 8. As shown in Table 8, there is no evidence of any significant difference in mortality between the different treatments. The absolute proportion of people with mortality based on an illustrative risk of 3.5% (Finch 2007) is shown in summary of findings Table for the main comparison. As shown in Figure 5, none of the treatments ranked best with more than 90% probability. As shown in Figure 6, there is substantial uncertainty about the treatment strategy with lowest mortality.


Mortality ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .

Mortality ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.


Mortality ‐ cumulative probability of ranks of different treatments There is more than 90% probability that IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) is within the five best treatments (of seven treatments). All the remaining treatments other than ContVascSharpNoFib (continuous vascular occlusion with sharp dissection and no fibrin) are within the six best treatments. This suggests that there is substantial uncertainty about the treatment with least mortality. Treatment codes are provided in .

Mortality ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) is within the five best treatments (of seven treatments). All the remaining treatments other than ContVascSharpNoFib (continuous vascular occlusion with sharp dissection and no fibrin) are within the six best treatments. This suggests that there is substantial uncertainty about the treatment with least mortality. Treatment codes are provided in Table 6.

Open in table viewer
Table 7. Mortality ‐ results and model fit

Fixed‐effect model

Random‐effects model

Inconsistency model (random‐effects)

d[2]

1.92 (95% CrI ‐2.29 to 6.13)

1.7 (95% CrI ‐5.33 to 8.73)

d[3]

‐0.18 (95% CrI ‐4.2 to 3.84)

‐0.21 (95% CrI ‐6.29 to 5.88)

d[4]

0.6 (95% CrI ‐3.29 to 4.5)

0.6 (95% CrI ‐6.08 to 7.27)

d[5]

0.6 (95% CrI ‐6.68 to 7.89)

0.62 (95% CrI ‐9.85 to 11.09)

d[6]

0.09 (95% CrI ‐2.99 to 3.18)

0.16 (95% CrI ‐5.25 to 5.58)

d[7]

‐1.23 (95% CrI ‐5.45 to 2.98)

‐0.89 (95% CrI ‐8.12 to 6.33)

Dbar

42.93

42.82

44.28

pD

11.07

11.68

12.06

DIC

54

54.49

56.35

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and so on.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. There is no evidence of inconsistency.

Open in table viewer
Table 8. Mortality ‐ pair‐wise comparisons

NoVascClampNoFib

NoVascCUSANoFib

NoVascRFAblNoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

IntVascCUSANoFib

NoVascClampNoFib

OR 6.83; 95% CrI 0.1 to 459.49

OR 0.84; 95% CrI 0.02 to 46.5

OR 1.83; 95% CrI 0.04 to 89.57

OR 1.83; 95% CrI 0 to 2660.48

OR 1.1; 95% CrI 0.05 to 23.98

OR 0.29; 95% CrI 0 to 19.67

NoVascCUSANoFib

OR 0.12; 95% CrI 0 to 41.17

OR 0.27; 95% CrI 0 to 82.55

OR 0.27; 95% CrI 0 to 1202.95

OR 0.16; 95% CrI 0 to 29.61

OR 0.04; 95% CrI 0 to 16.43

NoVascRFAblNoFib

OR 2.19; 95% CrI 0.01 to 587.54

OR 2.18; 95% CrI 0 to 8952.18

OR 1.31; 95% CrI 0.01 to 207.83

OR 0.35; 95% CrI 0 to 117.53

ConVascClampNoFib

OR 1; 95% CrI 0 to 3850.15

OR 0.6; 95% CrI 0 to 85.91

OR 0.16; 95% CrI 0 to 49.23

ConVascSharpNoFib

OR 0.6; 95% CrI 0 to 1634.74

OR 0.16; 95% CrI 0 to 718.88

IntVascClampNoFib

OR 0.27; 95% CrI 0 to 49.19

IntVascCUSANoFib

The treatment codes are provided in Table 6.

CrI; confidence interval; OR: odds ratio.

None of the trials reported long‐term mortality.

Serious adverse events

Five trials (406 participants) provided data for the network meta‐analysis on serious adverse events (Lesurtel 2005; Smyrniotis 2005; Capussotti 2006; Petrowsky 2006; Lupo 2007). There were 35 people with serious adverse events in the included studies (8.6%). The network plot is shown in Figure 7. The results and model‐fit of the fixed‐effect model and random‐effects model along with the model‐fit of the inconsistency model is provided in Table 9. The between‐study standard deviation (tau) was 0.03. As indicated in Table 9, the fixed‐effect model was preferred based on the DIC statistics. There was no evidence of inconsistency in the network. The pair‐wise ORs for the different treatment comparisons is shown in Table 8. As shown in Table 10, there was no evidence of any significant difference between the different treatments except for a significant increase in the proportion of people with serious adverse events in the NoVascRFAblNoFib (no vascular occlusion with radiofrequency dissecting sealer and no fibrin) compared with NoVascClampNoFib (no vascular occlusion with clamp‐crush and no fibrin) (OR 7.13; 95% CrI 1.77 to 28.65). The absolute proportion of people with serious adverse events based on an illustrative risk of 6.7% in the reference treatment is shown in summary of findings Table 2. As shown in Figure 8, none of the treatments ranked best with more than 90% probability. As shown in Figure 9, there is a high probability that NoVascClampNoFib (no vascular occlusion with clamp‐crush method and no fibrin) and IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasonic surgical aspirator (CUSA) and no fibrin) are better than other treatments with regards to serious adverse events.


Network plot of serious adverse events Treatment codes are provided in .

Network plot of serious adverse events

Treatment codes are provided in Table 6.


Serious adverse events ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .

Serious adverse events ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.


Serious adverse events ‐ cumulative probability of ranks of different treatments There is more than 90% probability that NoVascClampNoFib (no vascular occlusion with clamp‐crush method and no fibrin) and IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) are within the three best treatments (of seven treatments). This suggests that there is a high probability that these two treatments are better than other treatments with regards to serious adverse events. Treatment codes are provided in .

Serious adverse events ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that NoVascClampNoFib (no vascular occlusion with clamp‐crush method and no fibrin) and IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) are within the three best treatments (of seven treatments). This suggests that there is a high probability that these two treatments are better than other treatments with regards to serious adverse events. Treatment codes are provided in Table 6.

Open in table viewer
Table 9. Serious adverse events ‐ results and model fit

Fixed‐effect model

Random‐effects model

Inconsistency model (random‐effects)

d[2]

1 (95% CrI ‐1.33 to 3.33)

0.77 (95% CrI ‐6.22 to 7.76)

d[3]

1.96 (95% CrI 0.57 to 3.36)

1.78 (95% CrI ‐3.2 to 6.76)

d[4]

1.54 (95% CrI ‐0.3 to 3.38)

1.22 (95% CrI ‐4.57 to 7.01)

d[5]

1.54 (95% CrI ‐2.49 to 5.58)

1.2 (95% CrI ‐7.51 to 9.91)

d[6]

0.05 (95% CrI ‐1.51 to 1.6)

0.1 (95% CrI ‐4.91 to 5.11)

Dbar

44.12

41.43

41.4

pD

9.65

10.64

10.64

DIC

53.77

52.08

52.04

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and so on.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. There is no evidence of inconsistency.

Open in table viewer
Table 10. Serious adverse events ‐ pair‐wise comparisons

NoVascClampNoFib

NoVascCUSANoFib

NoVascRFAblNoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

NoVascClampNoFib

OR 2.72; 95% CrI 0.27 to 27.92

OR 7.13; 95% CrI 1.77 to 28.65

OR 4.68; 95% CrI 0.74 to 29.47

OR 4.68; 95% CrI 0.08 to 264.19

OR 1.05; 95% CrI 0.22 to 4.96

NoVascCUSANoFib

OR 2.62; 95% CrI 0.17 to 39.46

OR 1.72; 95% CrI 0.09 to 33.46

OR 1.72; 95% CrI 0.02 to 181.18

OR 0.39; 95% CrI 0.02 to 6.33

NoVascRFAblNoFib

OR 0.66; 95% CrI 0.07 to 6.59

OR 0.66; 95% CrI 0.01 to 46.8

OR 0.15; 95% CrI 0.02 to 1.18

ConVascClampNoFib

OR 1; 95% CrI 0.01 to 84.13

OR 0.22; 95% CrI 0.02 to 2.49

ConVascSharpNoFib

OR 0.22; 95% CrI 0 to 16.89

IntVascClampNoFib

The treatment codes are provided in Table 6. Results in bold text are significant.

CrI: credible interval; OR: odds ratio.

Quality of life

None of the trials reported quality of life.

Blood transfusion requirements

Proportion transfused

Six trials (446 participants) provided data for the network meta‐analysis on proportion of people transfused (Lesurtel 2005; Smyrniotis 2005; Capussotti 2006; Petrowsky 2006; Lupo 2007; Doklestic 2011). The network plot is shown in Figure 10. The results and model‐fit of the fixed‐effect model and random‐effects model along with the model‐fit of the inconsistency model is provided in Table 11. As indicated in Table 11, the random‐effects model was preferred based on the DIC statistics. The between‐study standard deviation (tau) was 0.61. There was no evidence of inconsistency in the network. The pair‐wise ORs for the different treatment comparisons are shown in Table 12. As shown in Table 12, there is no evidence of any significant difference in the proportion of people transfused between the different treatments. The absolute proportion of people requiring blood transfusion based on an illustrative risk of 15.7% in the reference treatment is shown in summary of findings Table 2. As shown in Figure 11, none of the treatments ranked best with more than 90% probability. As shown in Figure 12, there was substantial uncertainty about the treatment with lowest proportion of people transfused.


Network plot of blood transfusion proportion Treatment codes are provided in .

Network plot of blood transfusion proportion

Treatment codes are provided in Table 6.


Blood transfusion ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .

Blood transfusion ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.


Blood transfusion proportion ‐ cumulative probability of ranks of different treatments There is more than 90% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush and no fibrin) is within the five best treatments (of seven treatments) and that NoVascClampNoFib (no vascular occlusion with clamp‐crush and no fibrin) is within the six best treatments (of seven treatments). This suggests that there is substantial uncertainty about the treatment with proportion of people with blood transfusion. Treatment codes are provided in .

Blood transfusion proportion ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush and no fibrin) is within the five best treatments (of seven treatments) and that NoVascClampNoFib (no vascular occlusion with clamp‐crush and no fibrin) is within the six best treatments (of seven treatments). This suggests that there is substantial uncertainty about the treatment with proportion of people with blood transfusion. Treatment codes are provided in Table 6.

Open in table viewer
Table 11. Blood transfusion proportion ‐ results and model fit

Fixed‐effect model

Random‐effects model

Inconsistency model (random‐effects)

d[2]

1.29 (95% CrI ‐0.32 to 2.9)

1.8 (95% CrI ‐5.75 to 9.35)

d[3]

‐0.03 (95% CrI ‐1.08 to 1.01)

0.49 (95% CrI ‐4.94 to 5.92)

d[4]

‐0.04 (95% CrI ‐1.43 to 1.36)

‐0.24 (95% CrI ‐6.63 to 6.15)

d[5]

‐0.26 (95% CrI ‐1.93 to 1.41)

‐0.46 (95% CrI ‐9.32 to 8.41)

d[6]

0.8 (95% CrI ‐0.46 to 2.07)

1.22 (95% CrI ‐4.4 to 6.85)

d[7]

1.36 (95% CrI ‐1.1 to 3.82)

1.8 (95% CrI ‐6.77 to 10.36)

Dbar

63.46

56.44

56.61

pD

12.02

13.01

12.89

DIC

75.48

69.45

69.51

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and so on.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, random‐effects model is the preferred model. There is no evidence of inconsistency.

Open in table viewer
Table 12. Blood transfusion proportion ‐ pair‐wise comparisons

NoVascClampNoFib

NoVascCUSANoFib

NoVascRFAblNoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

IntVascCUSANoFib

NoVascClampNoFib

OR 6.06; 95% CrI 0 to 11,486.87

OR 1.64; 95% CrI 0.01 to 373.55

OR 0.79; 95% CrI 0 to 469.31

OR 0.63; 95% CrI 0 to 4494.19

OR 3.4; 95% CrI 0.01 to 941.28

OR 6.03; 95% CrI 0 to 31,671.1

NoVascCUSANoFib

OR 0.27; 95% CrI 0 to 2950.65

OR 0.13; 95% CrI 0 to 2563.91

OR 0.1; 95% CrI 0 to 11,933.13

OR 0.56; 95% CrI 0 to 6873.03

OR 1; 95% CrI 0 to 90,479.42

NoVascRFAblNoFib

OR 0.48; 95% CrI 0 to 2111.73

OR 0.39; 95% CrI 0 to 12,705.22

OR 2.08; 95% CrI 0 to 5166.35

OR 3.68; 95% CrI 0 to 93,690.74

ConVascClampNoFib

OR 0.81; 95% CrI 0 to 44,995.95

OR 4.32; 95% CrI 0 to 21,534.25

OR 7.66; 95% CrI 0 to 336,044.28

ConVascSharpNoFib

OR 5.37; 95% CrI 0 to 194,906

OR 9.51; 95% CrI 0 to 2,152,551.81

IntVascClampNoFib

OR 1.77; 95% CrI 0 to 50,000.24

IntVascCUSANoFib

The treatment codes are provided in Table 6.

CrI: credible interval; OR: odds ratio.

Quantity of blood transfused

Two trials (155 participants) provided data for the network meta‐analysis on quantity of blood transfused (Smyrniotis 2005; Petrowsky 2006). The network plot is shown in Figure 13. The results and model‐fit of the fixed‐effect model and random‐effects model is provided in Table 13. The between‐study standard deviation (tau) was 0. As indicated in Table 13, the fixed‐effect model was preferred based on the DIC statistics. We have not reported the model‐fit of the inconsistency model since there was no closed loop in the network. The pair‐wise MDs for the different treatment comparisons are shown in Table 14. As shown in Table 14, people undergoing liver resection by IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) had significantly higher amounts of blood transfused than people undergoing liver resection by ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin) (MD 1.2 units; 95% CrI 0.08 to 2.32). There were no significant differences in the other comparisons. As shown in Figure 14, none of the treatments ranked best with more than 90% probability. As shown in Figure 15, there was a high probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin) and ContVascSharpNoFib (continuous vascular occlusion with sharp dissection and no fibrin) are better than IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) with regards to quantity of blood transfused.


Network plot of quantity of blood transfused Treatment codes are provided in .

Network plot of quantity of blood transfused

Treatment codes are provided in Table 6.


Quantity of blood transfusion ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .

Quantity of blood transfusion ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.


Quantity of blood transfused ‐ cumulative probability of ranks of different treatments There is more than 90% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin) and ContVascSharpNoFib (continuous vascular occlusion with sharp dissection and no fibrin) are within the two best treatments. This suggests that these two treatments are better than IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) with regards to the quantity of blood transfused. Treatment codes are provided in .

Quantity of blood transfused ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin) and ContVascSharpNoFib (continuous vascular occlusion with sharp dissection and no fibrin) are within the two best treatments. This suggests that these two treatments are better than IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) with regards to the quantity of blood transfused. Treatment codes are provided in Table 6.

Open in table viewer
Table 13. Blood transfusion quantity ‐ results and model fit

Fixed‐effect model

Random‐effects model

d[2]

0 (95% CrI ‐1.36 to 1.36)

0 (95% CrI ‐5.83 to 5.83)

d[3]

1.2 (95% CrI 0.08 to 2.32)

1.2 (95% CrI ‐4.57 to 6.96)

Dbar

4.66

4.67

pD

4

4

DIC

8.65

8.67

d[2] indicates the log odds ratio between treatment 2 and treatment 1 and d[3] indicates the log odds ratio between treatment 3 and treatment 1.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. Model‐fit of the inconsistency model is not provided as there were no closed loops.

Open in table viewer
Table 14. Blood transfusion quantity ‐ pair‐wise comparisons

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

ConVascClampNoFib

MD 0; 95% CrI ‐1.36 to 1.36

MD 1.2; 95% CrI 0.08 to 2.32

ConVascSharpNoFib

MD 1.2; 95% CrI ‐0.56 to 2.96

IntVascClampNoFib

The treatment codes are provided in Table 6. Results in bold text are significant.

CrI: credible interval; MD: mean difference (units of blood transfused).

Operative blood loss

Three trials (281 participants) provided data for the network meta‐analysis on operative blood loss (Smyrniotis 2005; Capussotti 2006; Petrowsky 2006). The network plot is shown in Figure 16. The results and model‐fit of the fixed‐effect model and random‐effects model is provided in Table 15. The between‐study standard deviation (tau) was 0.02. As indicated in Table 15, the fixed‐effect model was preferred based on the DIC statistics. We have not reported the model‐fit of the inconsistency model since there was no closed loop in the network. The pair‐wise mean differences for the different treatment comparisons are shown in Table 16. As shown in Table 16, people undergoing liver resection by ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin) had significantly lower blood loss than those undergoing liver resection by NoVascClampNoFib (no vascular occlusion with clamp‐crush method and no fibrin) (MD ‐130.9 mL; 95% CrI ‐255.89 to ‐5.91). There were no significant differences in the other comparisons. As shown in Figure 17 and Figure 18, ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin) was ranked the best treatment with regards to operative blood loss with more than 90% probability.


Network plot of operative blood loss Treatment codes are provided in .

Network plot of operative blood loss

Treatment codes are provided in Table 6.


Operative blood loss ‐ best treatment There is a more than 90% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin) is the best treatment. Treatment codes are provided in .

Operative blood loss ‐ best treatment

There is a more than 90% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin) is the best treatment. Treatment codes are provided in Table 6.


Operative blood loss ‐ cumulative probability of ranks of different treatments There is more than 95% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush and no fibrin) is within the two best treatments. This suggests that there is little uncertainty about the treatment with least operative blood loss. Treatment codes are provided in .

Operative blood loss ‐ cumulative probability of ranks of different treatments

There is more than 95% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush and no fibrin) is within the two best treatments. This suggests that there is little uncertainty about the treatment with least operative blood loss. Treatment codes are provided in Table 6.

Open in table viewer
Table 15. Operative blood loss ‐ results and model fit

Fixed‐effect model

Random‐effects model

d[2]

‐130.9 (95% CrI ‐255.89 to ‐5.91)

‐127.9 (95% CrI ‐255.59 to ‐0.21)

d[3]

39.59 (95% CrI ‐111.37 to 190.55)

37.61 (95% CrI ‐113.21 to 188.43)

d[4]

3.62 (95% CrI ‐64.31 to 71.56)

3.45 (95% CrI ‐64.7 to 71.6)

Dbar

70.77

70.65

pD

4.24

4.24

DIC

75.01

74.89

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and d[4] indicates the log odds ratio between treatment 4 and treatment 1.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. Model‐fit of the inconsistency model is not provided as there were no closed loops.

Open in table viewer
Table 16. Operative blood loss ‐ pair‐wise comparisons

NoVascClampNoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

NoVascClampNoFib

MD ‐130.9; 95% CrI ‐255.89 to ‐5.91

MD 39.59; 95% CrI ‐111.37 to 190.55

MD 3.62; 95% CrI ‐64.31 to 71.56

ConVascClampNoFib

MD 170.49; 95% CrI ‐25.5 to 366.48

MD 134.52; 95% CrI ‐7.73 to 276.78

ConVascSharpNoFib

MD ‐35.97; 95% CrI ‐201.51 to 129.57

IntVascClampNoFib

The treatment codes are provided in Table 6. Results in bold text are significant.

CrI: credible interval; MD: mean difference (mL of blood loss).

Major blood loss

None of the trials included in the network meta‐analysis reported the proportion of people who developed major blood loss (class 3 or class 4 shock according to ATLS definition) (ATLS 2008). In one trial, two participants in the ContVascSharpNoFib (continuous vascular occlusion with sharp dissection and no fibrin sealant) were re‐operated due to significant post‐operative bleeding (Smyrniotis 2005). The authors stated that this was related to the sharp dissection method of parenchymal transection (Smyrniotis 2005). In another trial, one participant in NoVascCUSANoFib (no vascular occlusion with CUSA and no fibrin sealant) underwent re‐operation for significant post‐operative bleeding (Park 2012).

Hospital stay

Length of hospital stay

Six trials (446 participants) provided data for the network meta‐analysis on length of hospital stay (Lesurtel 2005; Smyrniotis 2005; Capussotti 2006; Petrowsky 2006; Doklestic 2011; Park 2012). The network plot is shown in Figure 19. The results and model‐fit of the fixed‐effect model and random‐effects model along with the model‐fit of the inconsistency model is provided in Table 17. The between study standard deviation (tau) was 0.01. As indicated in Table 17, the fixed‐effect model was preferred based on the DIC statistics. There was no evidence of inconsistency in the network. The pair wise mean differences for the different treatment comparisons is shown in Table 18. As shown in Table 18, there is no evidence of any significant difference in the length of hospital stay between the different treatments. As shown in Figure 20, none of the treatments ranked best with more than 90% probability. As shown in Figure 21, there is substantial uncertainty about the treatment with least length of hospital stay.


Network plot of length of hospital stay Treatment codes are provided in .

Network plot of length of hospital stay

Treatment codes are provided in Table 6.


Length of hospital stay ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .

Length of hospital stay ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.


Length of hospital stay ‐ cumulative probability of ranks of different treatments There is more than 90% probability that IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) is within the four best treatments (of seven treatments). NoVascClampNoFib (no vascular occlusion with clamp‐crush and no fibrin) and ContVascClampNoFib (continuous vascular occlusion with clamp‐crush and no fibrin) are within the five best treatments. This suggests that there is substantial uncertainty about the treatment with least length of hospital stay. Treatment codes are provided in .

Length of hospital stay ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) is within the four best treatments (of seven treatments). NoVascClampNoFib (no vascular occlusion with clamp‐crush and no fibrin) and ContVascClampNoFib (continuous vascular occlusion with clamp‐crush and no fibrin) are within the five best treatments. This suggests that there is substantial uncertainty about the treatment with least length of hospital stay. Treatment codes are provided in Table 6.

Open in table viewer
Table 17. Length of hospital stay ‐ results and model fit

Fixed‐effect model

Random‐effects model

Inconsistency model (random‐effects)

d[2]

2.29 (95% CrI ‐2.03 to 6.61)

2.28 (95% CrI ‐5.82 to 10.39)

d[3]

2.29 (95% CrI ‐3.01 to 7.58)

2.29 (95% CrI ‐6.94 to 11.51)

d[4]

2.29 (95% CrI ‐1.57 to 6.15)

2.29 (95% CrI ‐5.18 to 9.75)

d[5]

3.28 (95% CrI ‐2.59 to 9.15)

3.29 (95% CrI ‐6.64 to 13.22)

d[6]

0.3 (95% CrI ‐1.11 to 1.71)

0.29 (95% CrI ‐4.77 to 5.35)

d[7]

‐1.21 (95% CrI ‐5.19 to 2.78)

‐1.21 (95% CrI ‐8.77 to 6.34)

Dbar

41.68

42.11

42.7

pD

11.99

12.41

13.01

DIC

53.67

54.52

55.7

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and so on.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. There is no evidence of inconsistency.

Open in table viewer
Table 18. Length of hospital stay ‐ pair‐wise comparisons

NoVascClampNoFib

NoVascCUSANoFib

NoVascRFAblNoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

IntVascCUSANoFib

NoVascClampNoFib

MD 2.29; 95% CrI ‐2.03 to 6.61

MD 2.29; 95% CrI ‐3.01 to 7.58

MD 2.29; 95% CrI ‐1.57 to 6.15

MD 3.28; 95% CrI ‐2.59 to 9.15

MD 0.3; 95% CrI ‐1.11 to 1.71

MD ‐1.21; 95% CrI ‐5.19 to 2.78

NoVascCUSANoFib

MD 0; 95% CrI ‐6.84 to 6.83

MD 0; 95% CrI ‐5.79 to 5.79

MD 0.99; 95% CrI ‐6.3 to 8.28

MD ‐1.99; 95% CrI ‐6.54 to 2.55

MD ‐3.5; 95% CrI ‐9.37 to 2.38

NoVascRFAblNoFib

MD 0; 95% CrI ‐6.55 to 6.55

MD 0.99; 95% CrI ‐6.91 to 8.9

MD ‐1.99; 95% CrI ‐7.47 to 3.49

MD ‐3.5; 95% CrI ‐10.12 to 3.13

ConVascClampNoFib

MD 0.99; 95% CrI ‐6.03 to 8.02

MD ‐1.99; 95% CrI ‐6.1 to 2.12

MD ‐3.5; 95% CrI ‐9.04 to 2.05

ConVascSharpNoFib

MD ‐2.98; 95% CrI ‐9.02 to 3.06

MD ‐4.49; 95% CrI ‐11.58 to 2.61

IntVascClampNoFib

MD ‐1.51; 95% CrI ‐5.73 to 2.72

IntVascCUSANoFib

The treatment codes are provided in Table 6.

CrI: credible interval; MD: mean difference (days).

Intensive therapy unit stay

Four trials (261 participants) provided data for the network meta‐analysis on intensive therapy unit stay (Lesurtel 2005; Smyrniotis 2005; Petrowsky 2006; Doklestic 2011). The network plot is shown in Figure 22. The results and model‐fit of the fixed‐effect model and random‐effects model along with the model‐fit of the inconsistency model is provided in Table 19. The between‐study standard deviation (tau) was 0. As indicated in Table 19, the fixed‐effect model was preferred based on the DIC statistics. There was no evidence of inconsistency in the network. The pair‐wise MDs for the different treatment comparisons are shown in Table 20. As shown in Table 20, there is no evidence of any significant difference in intensive therapy unit stay between the different treatments. As shown in Figure 23, none of the treatments ranked best with more than 90% probability. As shown in Figure 24, IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) was within the three best treatments (of six treatments) and IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) may be better than other treatments with regards to intensive therapy unit stay with a high probability.


Network plot of intensive therapy unit stay Treatment codes are provided in .

Network plot of intensive therapy unit stay

Treatment codes are provided in Table 6.


Intensive therapy unit (ITU) stay ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .

Intensive therapy unit (ITU) stay ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.


Intensive therapy unit (ITU) stay ‐ cumulative probability of ranks of different treatments There is more than 90% probability that IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) is within the three best treatments (of six treatments). IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) is within the four best treatments with a more than 90% probability. This suggests that these two treatments may be better than other treatments with regards to ITU stay. Treatment codes are provided in .

Intensive therapy unit (ITU) stay ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) is within the three best treatments (of six treatments). IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) is within the four best treatments with a more than 90% probability. This suggests that these two treatments may be better than other treatments with regards to ITU stay. Treatment codes are provided in Table 6.

Open in table viewer
Table 19. Intensive therapy unit stay ‐ results and model fit

Fixed effect model

Random‐effects model

Inconsistency model (random‐effects)

d[2]

0 (95% CrI ‐3.32 to 3.33)

‐0.01 (95% CrI ‐6.56 to 6.55)

d[3]

0.01 (95% CrI ‐3.32 to 3.34)

‐0.01 (95% CrI ‐6.55 to 6.53)

d[4]

0.01 (95% CrI ‐4.69 to 4.71)

‐0.01 (95% CrI ‐9.27 to 9.24)

d[5]

‐2.18 (95% CrI ‐6.38 to 2.01)

‐2.2 (95% CrI ‐11.19 to 6.79)

d[6]

‐3.68 (95% CrI ‐9.04 to 1.68)

‐3.7 (95% CrI ‐14.81 to 7.41)

Dbar

27.07

27.07

27.07

pD

9

9

9

DIC

36.07

36.07

36.06

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and so on.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. There is no evidence of inconsistency.

Open in table viewer
Table 20. Intensive therapy unit stay ‐ pair‐wise comparisons

NoVascCUSANoFib

NoVascRFAblNoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

IntVascCUSANoFib

NoVascCUSANoFib

MD 0; 95% CrI ‐3.32 to 3.33

MD 0.01; 95% CrI ‐3.32 to 3.34

MD 0.01; 95% CrI ‐4.69 to 4.71

MD ‐2.18; 95% CrI ‐6.38 to 2.01

MD ‐3.68; 95% CrI ‐9.04 to 1.68

NoVascRFAblNoFib

MD 0.01; 95% CrI ‐4.7 to 4.71

MD 0.01; 95% CrI ‐5.75 to 5.76

MD ‐2.19; 95% CrI ‐7.54 to 3.17

MD ‐3.68; 95% CrI ‐9.99 to 2.62

ConVascClampNoFib

MD 0; 95% CrI ‐5.76 to 5.76

MD ‐2.19; 95% CrI ‐7.55 to 3.16

MD ‐3.69; 95% CrI ‐10 to 2.62

ConVascSharpNoFib

MD ‐2.19; 95% CrI ‐8.49 to 4.11

MD ‐3.69; 95% CrI ‐10.82 to 3.44

IntVascClampNoFib

MD ‐1.5; 95% CrI ‐8.3 to 5.31

IntVascCUSANoFib

The treatment codes are provided in Table 6.

CrI: credible interval; MD: mean difference (days).

Operating time

Four trials (245 participants) provided data for the network meta‐analysis on operating time (Lesurtel 2005; Smyrniotis 2005; Petrowsky 2006; Park 2012). The network plot is shown in Figure 25. The results and model‐fit of the fixed‐effect model and random‐effects model along with the model‐fit of the inconsistency model is provided in Table 21. The between‐study standard deviation (tau) was 0.01. As indicated in Table 21, the fixed‐effect model was preferred based on the DIC statistics. We have not reported the model‐fit of the inconsistency model since there was no closed loop in the network. The pair‐wise MDs for the different treatment comparisons are shown in Table 22. As shown in Table 18, people undergoing liver resection by IntVascCUSANoFib method (intermittent vascular occlusion with CUSA and no fibrin) had significantly longer operating time than people undergoing liver resection by NoVascCUSANoFib (no vascular occlusion with CUSA and no fibrin) (MD 49.61 minutes; 95% CrI 29.81 to 69.41). There is no evidence of any significant difference in the operating time between the other comparisons. As shown in Figure 26, none of the treatments ranked best with more than 90% probability. As shown in Figure 27, there is substantial uncertainty about the treatment with least operating time.


Network plot of operating time Treatment codes are provided in .

Network plot of operating time

Treatment codes are provided in Table 6.


Operating time ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .

Operating time ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.


Operating time ‐ cumulative probability of ranks of different treatments There is more than 90% probability that NoVascCUSANoFib (no vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin), ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin), and IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) are within the four best treatments (of five treatments). This suggests that there is substantial uncertainty about the treatment with least operating time. Treatment codes are provided in .

Operating time ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that NoVascCUSANoFib (no vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin), ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin), and IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) are within the four best treatments (of five treatments). This suggests that there is substantial uncertainty about the treatment with least operating time. Treatment codes are provided in Table 6.

Open in table viewer
Table 21. Operating time ‐ results and model fit

Fixed‐effect model

Random‐effects model

d[2]

8.41 (95% CrI ‐61.25 to 78.07)

7.94 (95% CrI ‐62.15 to 78.03)

d[3]

10.42 (95% CrI ‐74.02 to 94.86)

9.33 (95% CrI ‐75.48 to 94.14)

d[4]

7.25 (95% CrI ‐47.15 to 61.64)

6.83 (95% CrI ‐47.78 to 61.43)

d[5]

49.61 (95% CrI 29.81 to 69.41)

49.35 (95% CrI 28.83 to 69.87)

Dbar

67.38

67.4

pD

7.47

7.5

DIC

74.85

74.9

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and so on.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. There is no evidence of inconsistency.

Open in table viewer
Table 22. Operating time ‐ pairwise comparisons

NoVascCUSANoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

IntVascCUSANoFib

NoVascCUSANoFib

MD 8.41; 95% CrI ‐61.25 to 78.07

MD 10.42; 95% CrI ‐74.02 to 94.86

MD 7.25; 95% CrI ‐47.15 to 61.64

MD 49.61; 95% CrI 29.81 to 69.41

ConVascClampNoFib

MD 2.01; 95% CrI ‐107.45 to 111.47

MD ‐1.17; 95% CrI ‐89.55 to 87.21

MD 41.2; 95% CrI ‐31.22 to 113.61

ConVascSharpNoFib

MD ‐3.18; 95% CrI ‐103.61 to 97.26

MD 39.19; 95% CrI ‐47.54 to 125.92

IntVascClampNoFib

MD 42.37; 95% CrI ‐15.52 to 100.25

IntVascCUSANoFib

The treatment codes are provided in Table 6. Results in bold text are significant.

CrI: credible interval; MD: mean difference (minutes).

Time needed to return to work

None of the trials reported the time needed to return to work.

Overall results

As shown in Figure 28, none of the treatments appear clearly superior to others when all the outcomes are considered together. We did not give any specific weighting to the different outcomes. However, if serious adverse events are considered more important than all the outcomes other mortality, NoVascClampNoFib (no vascular occlusion with clamp‐crush method and no fibrin) and IntVascCUSANoFib (intermittent vascular occlusion with CUSA and no fibrin) are better than other treatments with regards to serious adverse events. NoVascRFAblNoFib (no vascular occlusion with radiofrequency dissecting sealer and no fibrin) appears to be the worst in terms of serious adverse events. There does not seem to be much correlation between a treatment being best in reducing blood transfusion and being best in reducing serious adverse events and mortality.


Rankogram This shows the probability that the treatment is best for each outcome. None of the treatments appear clearly superior to others when all the outcomes are considered together. There does not seem to be much correlation between a treatment being best in reducing blood transfusion and being best in reducing serious adverse events and mortality. Treatment codes are provided in .

Rankogram

This shows the probability that the treatment is best for each outcome. None of the treatments appear clearly superior to others when all the outcomes are considered together. There does not seem to be much correlation between a treatment being best in reducing blood transfusion and being best in reducing serious adverse events and mortality. Treatment codes are provided in Table 6.

Subgroup analysis

Subgroup analysis was not performed because of the paucity of data.

Discussion

Summary of main results

This is the first network meta‐analysis comparing different techniques aimed at decreasing blood loss during liver resection. Overall, there does not seem to be any major advantage of one combination of techniques over another. Mortality was generally low in all the groups compared to that reported in previous studies (Finch 2007). This may be because of the careful selection of participants included in randomised clinical trials compared to a consecutive case series where the results of all liver resections were reported. We have provided the sample size calculations based on a mortality of 3.5% observed in consecutive series (Finch 2007). To achieve a 20% relative reduction in mortality (20% relative risk reduction) from 3.5% to 2.8%, more than 20,000 participants are required for a single direct comparison to demonstrate a significant reduction in mortality with a specific intervention. As shown in the Methods section, the effective sample size in an indirect comparison involving just three treatments is only a fraction of the number of participants included in the trials. An example is shown in the Methods section where 10,000 participants included in the indirect comparisons is equivalent to fewer than 2000 participants in the absence of heterogeneity and fewer than 1000 participants in the presence of moderate heterogeneity. Even without these complicated calculations, one can easily observe from the credible intervals that the credible intervals were very wide (summary of findings Table for the main comparison; Table 7). This means that we cannot rule out a significant benefit or harm by using different treatments. Given the number of participants required to show a significant benefit of treatment with relation to mortality and serious adverse events, trials of this magnitude are unlikely to be funded. The serious adverse events were significantly higher with radiofrequency dissecting sealer compared with clamp‐crush method in the absence of vascular occlusion or use of fibrin. There was no significant difference between the other groups. There was a high probability that 'no vascular occlusion with clamp‐crush method and no fibrin' and 'intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin' were better than other treatments with regards to serious adverse events. However, based on wide credible intervals, there is considerable uncertainty about the benefit of these methods over the methods other than radiofrequency dissecting sealer. In addition, there is no corroborative evidence in the form of these treatments reducing intensive therapy unit stay or length of hospital stay, which would be anticipated if an intervention made a significant reduction in serious adverse events.

None of the trials reported quality of life, which is an important outcome used in assessing the cost‐effectiveness of a treatment in a state‐funded healthcare system. Given that the quality of life would depend upon various factors including peri‐operative complications, length of hospital stay, and time to return to work, it is likely to be easier to demonstrate a significant difference in quality of life if the treatment was effective than to demonstrate a difference in mortality or serious adverse events. Future randomised clinical trials should use a validated quality of life measure.

The major purpose of different methods of liver resection is to decrease the blood loss and blood transfusion requirements. As mentioned in the Background, various methods have been attempted to achieve this. Some methods do not require any additional equipment (e.g., vascular occlusion), while other methods require special equipment (e.g., Cavitron ultrasonic surgical aspirator or radiofrequency dissecting sealer). There was no significant difference in the proportion of people who underwent blood transfusion dependent on the technique utilised. Parenchymal transection by clamp‐crush method with continuous vascular occlusion resulted in significantly lower blood loss than parenchymal transection by clamp‐crush method with no vascular occlusion and significantly lower quantity of blood transfused than parenchymal transection by clamp‐crush method with intermittent vascular occlusion. However, the reduction in blood loss and quantity of blood transfused was modest. It should also be noted that ischaemic preconditioning (temporary occlusion of vessels supplying the liver to 'condition' the liver to blood flow occlusion before exposing the liver to a prolonged period of blood flow occlusion) was used prior to continuous vascular occlusion with a maximum continuous clamp period of 75 minutes in one trial (Petrowsky 2006), and in the other trial, ischaemic preconditioning was used in the second half of the trial (Smyrniotis 2005). Therefore, caution is needed in interpreting these results and in applying the results in people without ischaemic preconditioning. In addition, this must be put into context. Serious adverse events are likely to result in decreased quality of life for patients and increased costs to the healthcare provider and are, therefore, more important endpoints than a modest decrease in blood transfusion.

There was no significant difference in the hospital stay or intensive therapy unit stay. These are important to the patients, their carers, and the healthcare funders. None of the trials reported time taken to return to work, which is an important outcome for the patient and their carers in the absence of significant sickness benefit and is an important outcome for the healthcare provider in a state‐funded healthcare system with significant sickness benefits.

Thus overall, there is no current evidence to prefer one treatment over another. Simple methods such as clamp‐crush method do not appear to result in poorer outcomes than other methods that require special equipment. However, there is significant uncertainty on this topic.

Overall completeness and applicability of evidence

The participants included in this trial underwent elective open liver resection and were generally anaesthetically fit. The findings of this review are applicable only to such patients.

Quality of the evidence

The overall quality of evidence was very low. The risk of bias was high in all the trials. Using appropriate methods of randomisation and reporting the method of randomisation adequately will decrease selection bias. While healthcare providers (surgeons who performs the surgery) cannot be blinded to the treatments, it is possible to blind the surgeons who are involved in the day‐to‐day postoperative management of the patient. While it may be difficult to blind the anaesthetist to the treatment groups, using objective criteria for transfusion (NHS Blood and Transplant 2007), may overcome the problem of bias due to lack of blinding with regards to intra‐operative blood transfusion. The intensivist involved in the post‐operative care of the patient can be easily blinded. Objective criteria for detection of complications along with the postoperative management of the patient by a healthcare team not involved in the operation can decrease detection and performance bias. With regards to drop‐outs, randomising the participants after confirming that the tumour can be removed can avoid post‐randomisation drop‐outs due to metastatic spread identified at the time of laparotomy. This can decrease attrition bias. Reporting all the important clinical outcomes can decrease selective reporting bias. There was no significant heterogeneity in all the outcomes other than proportion of blood transfused as indicated by the good model‐fit achieved by fixed‐effect model as compared to the random‐effects model.

The effect estimates were wide with the credible intervals overlapping 1 and with either 20% reduction (0.80) or 20% increase (1.20) which can be considered a clinically significant effect. Future trials should be adequately powered to decrease the risk of random errors.

Potential biases in the review process

We selected a range of databases without any language restrictions and conducted the meta‐analysis according to the NICE TSU (Dias 2012a; Dias 2012b; Dias 2012c; Dias 2013). These are the strengths of the review process.

We have excluded studies because the methods of vascular occlusion, parenchymal transection, or method of management of the cut surface were not reported (Characteristics of excluded studies). Some of these studies may meet the inclusion criteria and may have contributed additional information. We imputed the standard deviation when they were not available from the trials. This may have resulted in a change in the effect estimates.

Agreements and disagreements with other studies or reviews

This is the first network meta‐analysis. Previously, we have compared individual components and concluded that intermittent vascular occlusion may decrease blood loss (Gurusamy 2009a), and that the clamp‐crush method may decrease blood loss (Gurusamy 2009b). In this review, we have concluded that there is no evidence for any significant advantage of different methods of liver resection. The differences in conclusion may be because of the exclusion of trials in which the methods were not reported or when the other aspects of liver resection other than the component being compared were chosen in a non‐random manner.

Study flow diagram.
Figures and Tables -
Figure 1

Study flow diagram.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figures and Tables -
Figure 2

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figures and Tables -
Figure 3

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Network plot of mortality Treatment codes are provided in .
Figures and Tables -
Figure 4

Network plot of mortality

Treatment codes are provided in Table 6.

Mortality ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .
Figures and Tables -
Figure 5

Mortality ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.

Mortality ‐ cumulative probability of ranks of different treatments There is more than 90% probability that IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) is within the five best treatments (of seven treatments). All the remaining treatments other than ContVascSharpNoFib (continuous vascular occlusion with sharp dissection and no fibrin) are within the six best treatments. This suggests that there is substantial uncertainty about the treatment with least mortality. Treatment codes are provided in .
Figures and Tables -
Figure 6

Mortality ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) is within the five best treatments (of seven treatments). All the remaining treatments other than ContVascSharpNoFib (continuous vascular occlusion with sharp dissection and no fibrin) are within the six best treatments. This suggests that there is substantial uncertainty about the treatment with least mortality. Treatment codes are provided in Table 6.

Network plot of serious adverse events Treatment codes are provided in .
Figures and Tables -
Figure 7

Network plot of serious adverse events

Treatment codes are provided in Table 6.

Serious adverse events ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .
Figures and Tables -
Figure 8

Serious adverse events ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.

Serious adverse events ‐ cumulative probability of ranks of different treatments There is more than 90% probability that NoVascClampNoFib (no vascular occlusion with clamp‐crush method and no fibrin) and IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) are within the three best treatments (of seven treatments). This suggests that there is a high probability that these two treatments are better than other treatments with regards to serious adverse events. Treatment codes are provided in .
Figures and Tables -
Figure 9

Serious adverse events ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that NoVascClampNoFib (no vascular occlusion with clamp‐crush method and no fibrin) and IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) are within the three best treatments (of seven treatments). This suggests that there is a high probability that these two treatments are better than other treatments with regards to serious adverse events. Treatment codes are provided in Table 6.

Network plot of blood transfusion proportion Treatment codes are provided in .
Figures and Tables -
Figure 10

Network plot of blood transfusion proportion

Treatment codes are provided in Table 6.

Blood transfusion ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .
Figures and Tables -
Figure 11

Blood transfusion ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.

Blood transfusion proportion ‐ cumulative probability of ranks of different treatments There is more than 90% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush and no fibrin) is within the five best treatments (of seven treatments) and that NoVascClampNoFib (no vascular occlusion with clamp‐crush and no fibrin) is within the six best treatments (of seven treatments). This suggests that there is substantial uncertainty about the treatment with proportion of people with blood transfusion. Treatment codes are provided in .
Figures and Tables -
Figure 12

Blood transfusion proportion ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush and no fibrin) is within the five best treatments (of seven treatments) and that NoVascClampNoFib (no vascular occlusion with clamp‐crush and no fibrin) is within the six best treatments (of seven treatments). This suggests that there is substantial uncertainty about the treatment with proportion of people with blood transfusion. Treatment codes are provided in Table 6.

Network plot of quantity of blood transfused Treatment codes are provided in .
Figures and Tables -
Figure 13

Network plot of quantity of blood transfused

Treatment codes are provided in Table 6.

Quantity of blood transfusion ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .
Figures and Tables -
Figure 14

Quantity of blood transfusion ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.

Quantity of blood transfused ‐ cumulative probability of ranks of different treatments There is more than 90% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin) and ContVascSharpNoFib (continuous vascular occlusion with sharp dissection and no fibrin) are within the two best treatments. This suggests that these two treatments are better than IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) with regards to the quantity of blood transfused. Treatment codes are provided in .
Figures and Tables -
Figure 15

Quantity of blood transfused ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin) and ContVascSharpNoFib (continuous vascular occlusion with sharp dissection and no fibrin) are within the two best treatments. This suggests that these two treatments are better than IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) with regards to the quantity of blood transfused. Treatment codes are provided in Table 6.

Network plot of operative blood loss Treatment codes are provided in .
Figures and Tables -
Figure 16

Network plot of operative blood loss

Treatment codes are provided in Table 6.

Operative blood loss ‐ best treatment There is a more than 90% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin) is the best treatment. Treatment codes are provided in .
Figures and Tables -
Figure 17

Operative blood loss ‐ best treatment

There is a more than 90% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin) is the best treatment. Treatment codes are provided in Table 6.

Operative blood loss ‐ cumulative probability of ranks of different treatments There is more than 95% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush and no fibrin) is within the two best treatments. This suggests that there is little uncertainty about the treatment with least operative blood loss. Treatment codes are provided in .
Figures and Tables -
Figure 18

Operative blood loss ‐ cumulative probability of ranks of different treatments

There is more than 95% probability that ContVascClampNoFib (continuous vascular occlusion with clamp‐crush and no fibrin) is within the two best treatments. This suggests that there is little uncertainty about the treatment with least operative blood loss. Treatment codes are provided in Table 6.

Network plot of length of hospital stay Treatment codes are provided in .
Figures and Tables -
Figure 19

Network plot of length of hospital stay

Treatment codes are provided in Table 6.

Length of hospital stay ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .
Figures and Tables -
Figure 20

Length of hospital stay ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.

Length of hospital stay ‐ cumulative probability of ranks of different treatments There is more than 90% probability that IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) is within the four best treatments (of seven treatments). NoVascClampNoFib (no vascular occlusion with clamp‐crush and no fibrin) and ContVascClampNoFib (continuous vascular occlusion with clamp‐crush and no fibrin) are within the five best treatments. This suggests that there is substantial uncertainty about the treatment with least length of hospital stay. Treatment codes are provided in .
Figures and Tables -
Figure 21

Length of hospital stay ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) is within the four best treatments (of seven treatments). NoVascClampNoFib (no vascular occlusion with clamp‐crush and no fibrin) and ContVascClampNoFib (continuous vascular occlusion with clamp‐crush and no fibrin) are within the five best treatments. This suggests that there is substantial uncertainty about the treatment with least length of hospital stay. Treatment codes are provided in Table 6.

Network plot of intensive therapy unit stay Treatment codes are provided in .
Figures and Tables -
Figure 22

Network plot of intensive therapy unit stay

Treatment codes are provided in Table 6.

Intensive therapy unit (ITU) stay ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .
Figures and Tables -
Figure 23

Intensive therapy unit (ITU) stay ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.

Intensive therapy unit (ITU) stay ‐ cumulative probability of ranks of different treatments There is more than 90% probability that IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) is within the three best treatments (of six treatments). IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) is within the four best treatments with a more than 90% probability. This suggests that these two treatments may be better than other treatments with regards to ITU stay. Treatment codes are provided in .
Figures and Tables -
Figure 24

Intensive therapy unit (ITU) stay ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that IntVascCUSANoFib (intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin) is within the three best treatments (of six treatments). IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) is within the four best treatments with a more than 90% probability. This suggests that these two treatments may be better than other treatments with regards to ITU stay. Treatment codes are provided in Table 6.

Network plot of operating time Treatment codes are provided in .
Figures and Tables -
Figure 25

Network plot of operating time

Treatment codes are provided in Table 6.

Operating time ‐ best treatment None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in .
Figures and Tables -
Figure 26

Operating time ‐ best treatment

None of the treatments are considered to be the best treatment since the probabilities did not reach 90% or above. Treatment codes are provided in Table 6.

Operating time ‐ cumulative probability of ranks of different treatments There is more than 90% probability that NoVascCUSANoFib (no vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin), ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin), and IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) are within the four best treatments (of five treatments). This suggests that there is substantial uncertainty about the treatment with least operating time. Treatment codes are provided in .
Figures and Tables -
Figure 27

Operating time ‐ cumulative probability of ranks of different treatments

There is more than 90% probability that NoVascCUSANoFib (no vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin), ContVascClampNoFib (continuous vascular occlusion with clamp‐crush method and no fibrin), and IntVascClampNoFib (intermittent vascular occlusion with clamp‐crush method and no fibrin) are within the four best treatments (of five treatments). This suggests that there is substantial uncertainty about the treatment with least operating time. Treatment codes are provided in Table 6.

Rankogram This shows the probability that the treatment is best for each outcome. None of the treatments appear clearly superior to others when all the outcomes are considered together. There does not seem to be much correlation between a treatment being best in reducing blood transfusion and being best in reducing serious adverse events and mortality. Treatment codes are provided in .
Figures and Tables -
Figure 28

Rankogram

This shows the probability that the treatment is best for each outcome. None of the treatments appear clearly superior to others when all the outcomes are considered together. There does not seem to be much correlation between a treatment being best in reducing blood transfusion and being best in reducing serious adverse events and mortality. Treatment codes are provided in Table 6.

Summary of findings for the main comparison. Methods to decrease blood loss during liver resection (mortality)

Methods to decrease blood loss during liver resection (mortality)

Patient or population: people undergoing open liver resection

Settings: secondary or tertiary

Intervention and control: various treatments

Number of trials (participants)

7 trials (496 participants)

Overall quality of evidence

Very low1,2

Groups

Illustrative risk

Treatment effect

Assumed risk in control group (NoVascClampNoFib)*

35 per 1000

Corresponding risk in NoVascCUSANoFib

239 per 1000
(4 to 1000)

OR 6.83 (95% CrI 0.1 to 459.49)

Corresponding risk in NoVascRFAblNoFib

29 per 1000
(1 to 1000)

OR 0.84 (95% CrI 0.02 to 46.5)

Corresponding risk in ContVascClampNoFib

64 per 1000
(1 to 1000)

OR 1.83 (95% CrI 0.04 to 89.57)

Corresponding risk in ContVascSharpNoFib

64 per 1000
(0 to 1000)

OR 1.83 (95% CrI 0 to 2660.48)

Corresponding risk in IntVascClampNoFib

38 per 1000
(2 to 839)

OR 1.1 (95% CrI 0.05 to 23.98)

Corresponding risk in IntVascCUSANoFib

10 per 1000
(0 to 688)

OR 0.29 (95% CrI 0 to 19.67)

*The basis for the assumed risk was from literature. The corresponding risk (and its 95% credible interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CrI).
ContVascClampNoFib: continuous vascular occlusion with clamp‐crush method and no fibrin; ContVascSharpNoFib: continuous vascular occlusion with sharp dissection and no fibrin; CrI: credible interval; IntVascClampNoFib: intermittent vascular occlusion with clamp‐crush method and no fibrin; IntVascCUSANoFib: intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin; NoVascCUSANoFib: no vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin; NoVascRFAblNoFib: no vascular occlusion with radiofrequency dissecting sealer and no fibrin; OR: odds ratio.

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1. The trials were at high risk of bias (2 points).

2. The number of events were fewer than 300 and the credible intervals overlapped 1 and 0.80 or 1.20 (2 points).

Figures and Tables -
Summary of findings for the main comparison. Methods to decrease blood loss during liver resection (mortality)
Summary of findings 2. Methods to decrease blood loss during liver resection (serious adverse events)

Methods to decrease blood loss during liver resection (serious adverse events)

Patient or population: people undergoing open liver resection

Settings: secondary or tertiary

Intervention and control: various treatments

Number of trials (participants)

5 trials (406 participants)

Overall quality of evidence

Very low1,2

Groups

Illustrative risk

Treatment effect

Assumed risk in control group (NoVascClampNoFib)*

67 per 1000

Corresponding risk in NoVascCUSANoFib

95 per 1000
(9 to 977)

OR 2.72 (95% CrI 0.27 to 27.92)

Corresponding risk in NoVascRFAblNoFib

249 per 1000
(62 to 1000)

OR 7.13 (95% CrI 1.77 to 28.65)

Corresponding risk in ContVascClampNoFib

164 per 1000
(26 to 1000)

OR 4.68 (95% CrI 0.74 to 29.47)

Corresponding risk in ContVascSharpNoFib

164 per 1000
(3 to 1000)

OR 4.68 (95% CrI 0.08 to 264.19)

Corresponding risk in IntVascClampNoFib

37 per 1000
(8 to 174)

OR 1.05 (95% CrI 0.22 to 4.96)

*The basis for the assumed risk was from the mean proportion with serious adverse events in control group. The corresponding risk (and its 95% credible interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CrI).
ContVascClampNoFib: continuous vascular occlusion with clamp‐crush method and no fibrin; ContVascSharpNoFib: continuous vascular occlusion with sharp dissection and no fibrin; CrI: credible interval; IntVascClampNoFib: intermittent vascular occlusion with clamp‐crush method and no fibrin; IntVascCUSANoFib: intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin; NoVascCUSANoFib: no vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin; NoVascRFAblNoFib: no vascular occlusion with radiofrequency dissecting sealer and no fibrin; OR: odds ratio.

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1. The trials were at high risk of bias (2 points).

2. The number of events were fewer than 300 and the credible intervals overlapped 1 and 0.80 or 1.20 (2 points).

Figures and Tables -
Summary of findings 2. Methods to decrease blood loss during liver resection (serious adverse events)
Summary of findings 3. Methods to decrease blood loss during liver resection (blood transfusion proportion)

Methods to decrease blood loss during liver resection (blood transfusion proportion)

Patient or population: people undergoing open liver resection

Settings: secondary or tertiary

Intervention and control: various treatments

Number of trials (participants)

6 trials (446 participants)

Overall quality of evidence

Very low1,2

Groups

Illustrative risk

Treatment effect

Assumed risk in control group (NoVascClampNoFib)

157 per 1000

Corresponding risk in NoVascCUSANoFib

212 per 1000
(0 to 1000)

OR 6.06; 95% CrI 0 to 11,486.87

Corresponding risk in NoVascRFAblNoFib

57 per 1000
(0 to 1000)

OR 1.64; 95% CrI 0.01 to 373.55

Corresponding risk in ContVascClampNoFib

28 per 1000
(0 to 1000)

OR 0.79; 95% CrI 0 to 469.31

Corresponding risk in ContVascSharpNoFib

22 per 1000
(0 to 1000)

OR 0.63; 95% CrI 0 to 4494.19

Corresponding risk in IntVascClampNoFib

119 per 1000
(0 to 1000)

OR 3.4; 95% CrI 0.01 to 941.28

Corresponding risk in IntVascCUSANoFib

211 per 1000
(0 to 1000)

OR 6.03; 95% CrI 0 to 31,671.1

*The basis for the assumed risk was from the mean proportion with blood transfusion in control group. The corresponding risk (and its 95% credible interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CrI).
ContVascClampNoFib: continuous vascular occlusion with clamp‐crush method and no fibrin; ContVascSharpNoFib: continuous vascular occlusion with sharp dissection and no fibrin; CrI: credible interval; IntVascClampNoFib: intermittent vascular occlusion with clamp‐crush method and no fibrin; IntVascCUSANoFib: intermittent vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin; NoVascCUSANoFib: no vascular occlusion with Cavitron ultrasound surgical aspirator and no fibrin; NoVascRFAblNoFib: no vascular occlusion with radiofrequency dissecting sealer and no fibrin; OR: odds ratio.

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1. The trials were at high risk of bias (2 points).
2. The number of events were fewer than 300 and the credible intervals overlapped 1 and 0.80 or 1.20 (2 points).

Figures and Tables -
Summary of findings 3. Methods to decrease blood loss during liver resection (blood transfusion proportion)
Table 1. Different methods of vascular occlusion

No vascular occlusion

Portal triad clamping (continuous) (occlusion of inflow alone)

Portal triad clamping (intermittent) (occlusion of inflow alone)

Hepatic vascular exclusion (occlusion of inflow and outflow)

Selective vascular occlusion (occlusion of inflow to the hemi‐liver that is being resected)

Selective hepatic artery occlusion (occlusion of hepatic artery supplying the hemi‐liver that is being resected)

Selective portal vein occlusion (occlusion of portal vein supplying the hemi‐liver that is being resected)

Selective hepatic vascular exclusion (occlusion of inflow to the hemi‐liver and outflow from the hemi‐liver that is being resected)

Figures and Tables -
Table 1. Different methods of vascular occlusion
Table 2. Different methods of parenchymal transection

Finger‐fracture method

Clamp‐crush method

Cavitron ultrasonic surgical aspirator (CUSA)

Sharp dissection

Radiofrequency dissecting sealer

Ultrasonic shears

Figures and Tables -
Table 2. Different methods of parenchymal transection
Table 3. Different methods of dealing with raw surface

Suturing for large and medium vessels and ducts and performing electrocauterisation of small vessels and ducts

Suturing for large vessels and performing ultrasonic shears for medium‐sized and small vessels and ducts

Suturing and argon beam coagulator

Suturing and fibrin sealant

Figures and Tables -
Table 3. Different methods of dealing with raw surface
Table 4. Different categories of vascular occlusion, parenchymal transection, and methods of dealing with raw surface used in this review

Vascular occlusion

No vascular occlusion (NoVasc)

Continuous vascular occlusion (ConVasc)

Intermittent vascular occlusion (IntVasc)

Parenchymal transection

Finger‐fracture method (Finger)

Clamp‐crush method (Clamp)

Cavitron ultrasonic surgical aspirator (CUSA)

Sharp dissection (Sharp)

Radiofrequency dissecting sealer (RFAbl)

Ultrasonic shears (UShears)

Methods of dealing with raw surface

No fibrin sealant used (NoFib)

Fibrin sealant used (Fib)

Figures and Tables -
Table 4. Different categories of vascular occlusion, parenchymal transection, and methods of dealing with raw surface used in this review
Table 5. Clavien‐Dindo classification of postoperative complications

Grades

Definitions

Examples

I

Any deviation from the normal postoperative course without the need for pharmacological treatment or surgical, endoscopic, and radiological interventions

Drugs such as antiemetics, antipyretics, analgesics, diuretics, and electrolytes; physiotherapy; wound infections opened at the bedside

II

Requiring pharmacological treatment with drugs other than those allowed for grade I complications

Blood transfusions, total parenteral nutrition

III

Requiring surgical, endoscopic or radiological intervention

Bile leak requiring endoscopic stent; re‐operation for any cause; drainage of infected intra‐abdominal collection

IV

Life‐threatening complication requiring high dependency or intensive care management

Dialysis

V

Death of patient

Suffix d

If the patient suffers from a complication at the time of discharge and needs further follow‐up to evaluate the complication fully

Adapted from Dindo 2004; Clavien 2009.

Figures and Tables -
Table 5. Clavien‐Dindo classification of postoperative complications
Table 6. Summary of treatments used and types of participants included

Study

Vascular occlusion

Parenchymal transection

Liver raw surface

Codes of the comparisons

Number of participants

Major liver resections

Cirrhosis

Belghiti 1999

Intermittent vascular occlusion

versus

continuous

vascular occlusion

Cavitron ultrasonic surgical aspirator (CUSA)

Fibrin sealant

IntVascCUSAFib versus ContVascCUSAFib

86

20 (23.3%)

Not stated

Capussotti 2003

Intermittent vascular occlusion versus

continuous vascular occlusion

Clamp‐crush

Fibrin sealant

IntVascClampFib versus ContVascClampFib

35

8 (22.9%)

35 (100%)

Capussotti 2006

Intermittent vascular occlusion versus

no vascular occlusion

Clamp‐crush

No fibrin sealant

IntVascClampNoFib versus NoVascClampNoFib

126

56 (44.4%)

19 (15.1%)

Doklestic 2011

Intermittent vascular occlusion versus

continuous vascular occlusion

Clamp‐crush

No fibrin sealant

IntVascCUSANoFib versus IntVascClampNoFib

40

10 (25.0%)

0 (0%)

Lesurtel 2005

No vascular occlusion versus

no vascular occlusion versus

continuous vascular occlusion

CUSA versus radiofrequency dissecting sealer versus clamp‐crush method

No fibrin sealant

NoVascCUSANoFib versus NoVascRFAblNoFib versus ContVascClampNoFib

75

42 (56.0%)

0 (0%)

Lupo 2007

No vascular occlusion

Radiofrequency dissecting sealer versus

clamp‐crush

No fibrin sealant

NoVascRFAblNoFib versus NoVascClampNoFib

50

Not stated

Not stated

Park 2012

Intermittent vascular occlusion versus no vascular occlusion

CUSA

No fibrin sealant

IntVascCUSANoFib versus NoVascCUSANoFib

50

Not stated

Not stated

Petrowsky 2006

Intermittent vascular occlusion versus continuous vascular occlusion

Clamp‐crush

No fibrin sealant

IntVascClampNoFib versus ContVascClampNoFib

73

37 (50.7%)

Not stated

Smyrniotis 2005

Continuous vascular occlusion

Sharp dissection versus clamp‐crush

No fibrin sealant

ContVascSharpNoFib versus ContVascClampNoFib

82

Not stated

6 (7%)

Figures and Tables -
Table 6. Summary of treatments used and types of participants included
Table 7. Mortality ‐ results and model fit

Fixed‐effect model

Random‐effects model

Inconsistency model (random‐effects)

d[2]

1.92 (95% CrI ‐2.29 to 6.13)

1.7 (95% CrI ‐5.33 to 8.73)

d[3]

‐0.18 (95% CrI ‐4.2 to 3.84)

‐0.21 (95% CrI ‐6.29 to 5.88)

d[4]

0.6 (95% CrI ‐3.29 to 4.5)

0.6 (95% CrI ‐6.08 to 7.27)

d[5]

0.6 (95% CrI ‐6.68 to 7.89)

0.62 (95% CrI ‐9.85 to 11.09)

d[6]

0.09 (95% CrI ‐2.99 to 3.18)

0.16 (95% CrI ‐5.25 to 5.58)

d[7]

‐1.23 (95% CrI ‐5.45 to 2.98)

‐0.89 (95% CrI ‐8.12 to 6.33)

Dbar

42.93

42.82

44.28

pD

11.07

11.68

12.06

DIC

54

54.49

56.35

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and so on.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. There is no evidence of inconsistency.

Figures and Tables -
Table 7. Mortality ‐ results and model fit
Table 8. Mortality ‐ pair‐wise comparisons

NoVascClampNoFib

NoVascCUSANoFib

NoVascRFAblNoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

IntVascCUSANoFib

NoVascClampNoFib

OR 6.83; 95% CrI 0.1 to 459.49

OR 0.84; 95% CrI 0.02 to 46.5

OR 1.83; 95% CrI 0.04 to 89.57

OR 1.83; 95% CrI 0 to 2660.48

OR 1.1; 95% CrI 0.05 to 23.98

OR 0.29; 95% CrI 0 to 19.67

NoVascCUSANoFib

OR 0.12; 95% CrI 0 to 41.17

OR 0.27; 95% CrI 0 to 82.55

OR 0.27; 95% CrI 0 to 1202.95

OR 0.16; 95% CrI 0 to 29.61

OR 0.04; 95% CrI 0 to 16.43

NoVascRFAblNoFib

OR 2.19; 95% CrI 0.01 to 587.54

OR 2.18; 95% CrI 0 to 8952.18

OR 1.31; 95% CrI 0.01 to 207.83

OR 0.35; 95% CrI 0 to 117.53

ConVascClampNoFib

OR 1; 95% CrI 0 to 3850.15

OR 0.6; 95% CrI 0 to 85.91

OR 0.16; 95% CrI 0 to 49.23

ConVascSharpNoFib

OR 0.6; 95% CrI 0 to 1634.74

OR 0.16; 95% CrI 0 to 718.88

IntVascClampNoFib

OR 0.27; 95% CrI 0 to 49.19

IntVascCUSANoFib

The treatment codes are provided in Table 6.

CrI; confidence interval; OR: odds ratio.

Figures and Tables -
Table 8. Mortality ‐ pair‐wise comparisons
Table 9. Serious adverse events ‐ results and model fit

Fixed‐effect model

Random‐effects model

Inconsistency model (random‐effects)

d[2]

1 (95% CrI ‐1.33 to 3.33)

0.77 (95% CrI ‐6.22 to 7.76)

d[3]

1.96 (95% CrI 0.57 to 3.36)

1.78 (95% CrI ‐3.2 to 6.76)

d[4]

1.54 (95% CrI ‐0.3 to 3.38)

1.22 (95% CrI ‐4.57 to 7.01)

d[5]

1.54 (95% CrI ‐2.49 to 5.58)

1.2 (95% CrI ‐7.51 to 9.91)

d[6]

0.05 (95% CrI ‐1.51 to 1.6)

0.1 (95% CrI ‐4.91 to 5.11)

Dbar

44.12

41.43

41.4

pD

9.65

10.64

10.64

DIC

53.77

52.08

52.04

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and so on.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. There is no evidence of inconsistency.

Figures and Tables -
Table 9. Serious adverse events ‐ results and model fit
Table 10. Serious adverse events ‐ pair‐wise comparisons

NoVascClampNoFib

NoVascCUSANoFib

NoVascRFAblNoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

NoVascClampNoFib

OR 2.72; 95% CrI 0.27 to 27.92

OR 7.13; 95% CrI 1.77 to 28.65

OR 4.68; 95% CrI 0.74 to 29.47

OR 4.68; 95% CrI 0.08 to 264.19

OR 1.05; 95% CrI 0.22 to 4.96

NoVascCUSANoFib

OR 2.62; 95% CrI 0.17 to 39.46

OR 1.72; 95% CrI 0.09 to 33.46

OR 1.72; 95% CrI 0.02 to 181.18

OR 0.39; 95% CrI 0.02 to 6.33

NoVascRFAblNoFib

OR 0.66; 95% CrI 0.07 to 6.59

OR 0.66; 95% CrI 0.01 to 46.8

OR 0.15; 95% CrI 0.02 to 1.18

ConVascClampNoFib

OR 1; 95% CrI 0.01 to 84.13

OR 0.22; 95% CrI 0.02 to 2.49

ConVascSharpNoFib

OR 0.22; 95% CrI 0 to 16.89

IntVascClampNoFib

The treatment codes are provided in Table 6. Results in bold text are significant.

CrI: credible interval; OR: odds ratio.

Figures and Tables -
Table 10. Serious adverse events ‐ pair‐wise comparisons
Table 11. Blood transfusion proportion ‐ results and model fit

Fixed‐effect model

Random‐effects model

Inconsistency model (random‐effects)

d[2]

1.29 (95% CrI ‐0.32 to 2.9)

1.8 (95% CrI ‐5.75 to 9.35)

d[3]

‐0.03 (95% CrI ‐1.08 to 1.01)

0.49 (95% CrI ‐4.94 to 5.92)

d[4]

‐0.04 (95% CrI ‐1.43 to 1.36)

‐0.24 (95% CrI ‐6.63 to 6.15)

d[5]

‐0.26 (95% CrI ‐1.93 to 1.41)

‐0.46 (95% CrI ‐9.32 to 8.41)

d[6]

0.8 (95% CrI ‐0.46 to 2.07)

1.22 (95% CrI ‐4.4 to 6.85)

d[7]

1.36 (95% CrI ‐1.1 to 3.82)

1.8 (95% CrI ‐6.77 to 10.36)

Dbar

63.46

56.44

56.61

pD

12.02

13.01

12.89

DIC

75.48

69.45

69.51

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and so on.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, random‐effects model is the preferred model. There is no evidence of inconsistency.

Figures and Tables -
Table 11. Blood transfusion proportion ‐ results and model fit
Table 12. Blood transfusion proportion ‐ pair‐wise comparisons

NoVascClampNoFib

NoVascCUSANoFib

NoVascRFAblNoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

IntVascCUSANoFib

NoVascClampNoFib

OR 6.06; 95% CrI 0 to 11,486.87

OR 1.64; 95% CrI 0.01 to 373.55

OR 0.79; 95% CrI 0 to 469.31

OR 0.63; 95% CrI 0 to 4494.19

OR 3.4; 95% CrI 0.01 to 941.28

OR 6.03; 95% CrI 0 to 31,671.1

NoVascCUSANoFib

OR 0.27; 95% CrI 0 to 2950.65

OR 0.13; 95% CrI 0 to 2563.91

OR 0.1; 95% CrI 0 to 11,933.13

OR 0.56; 95% CrI 0 to 6873.03

OR 1; 95% CrI 0 to 90,479.42

NoVascRFAblNoFib

OR 0.48; 95% CrI 0 to 2111.73

OR 0.39; 95% CrI 0 to 12,705.22

OR 2.08; 95% CrI 0 to 5166.35

OR 3.68; 95% CrI 0 to 93,690.74

ConVascClampNoFib

OR 0.81; 95% CrI 0 to 44,995.95

OR 4.32; 95% CrI 0 to 21,534.25

OR 7.66; 95% CrI 0 to 336,044.28

ConVascSharpNoFib

OR 5.37; 95% CrI 0 to 194,906

OR 9.51; 95% CrI 0 to 2,152,551.81

IntVascClampNoFib

OR 1.77; 95% CrI 0 to 50,000.24

IntVascCUSANoFib

The treatment codes are provided in Table 6.

CrI: credible interval; OR: odds ratio.

Figures and Tables -
Table 12. Blood transfusion proportion ‐ pair‐wise comparisons
Table 13. Blood transfusion quantity ‐ results and model fit

Fixed‐effect model

Random‐effects model

d[2]

0 (95% CrI ‐1.36 to 1.36)

0 (95% CrI ‐5.83 to 5.83)

d[3]

1.2 (95% CrI 0.08 to 2.32)

1.2 (95% CrI ‐4.57 to 6.96)

Dbar

4.66

4.67

pD

4

4

DIC

8.65

8.67

d[2] indicates the log odds ratio between treatment 2 and treatment 1 and d[3] indicates the log odds ratio between treatment 3 and treatment 1.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. Model‐fit of the inconsistency model is not provided as there were no closed loops.

Figures and Tables -
Table 13. Blood transfusion quantity ‐ results and model fit
Table 14. Blood transfusion quantity ‐ pair‐wise comparisons

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

ConVascClampNoFib

MD 0; 95% CrI ‐1.36 to 1.36

MD 1.2; 95% CrI 0.08 to 2.32

ConVascSharpNoFib

MD 1.2; 95% CrI ‐0.56 to 2.96

IntVascClampNoFib

The treatment codes are provided in Table 6. Results in bold text are significant.

CrI: credible interval; MD: mean difference (units of blood transfused).

Figures and Tables -
Table 14. Blood transfusion quantity ‐ pair‐wise comparisons
Table 15. Operative blood loss ‐ results and model fit

Fixed‐effect model

Random‐effects model

d[2]

‐130.9 (95% CrI ‐255.89 to ‐5.91)

‐127.9 (95% CrI ‐255.59 to ‐0.21)

d[3]

39.59 (95% CrI ‐111.37 to 190.55)

37.61 (95% CrI ‐113.21 to 188.43)

d[4]

3.62 (95% CrI ‐64.31 to 71.56)

3.45 (95% CrI ‐64.7 to 71.6)

Dbar

70.77

70.65

pD

4.24

4.24

DIC

75.01

74.89

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and d[4] indicates the log odds ratio between treatment 4 and treatment 1.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. Model‐fit of the inconsistency model is not provided as there were no closed loops.

Figures and Tables -
Table 15. Operative blood loss ‐ results and model fit
Table 16. Operative blood loss ‐ pair‐wise comparisons

NoVascClampNoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

NoVascClampNoFib

MD ‐130.9; 95% CrI ‐255.89 to ‐5.91

MD 39.59; 95% CrI ‐111.37 to 190.55

MD 3.62; 95% CrI ‐64.31 to 71.56

ConVascClampNoFib

MD 170.49; 95% CrI ‐25.5 to 366.48

MD 134.52; 95% CrI ‐7.73 to 276.78

ConVascSharpNoFib

MD ‐35.97; 95% CrI ‐201.51 to 129.57

IntVascClampNoFib

The treatment codes are provided in Table 6. Results in bold text are significant.

CrI: credible interval; MD: mean difference (mL of blood loss).

Figures and Tables -
Table 16. Operative blood loss ‐ pair‐wise comparisons
Table 17. Length of hospital stay ‐ results and model fit

Fixed‐effect model

Random‐effects model

Inconsistency model (random‐effects)

d[2]

2.29 (95% CrI ‐2.03 to 6.61)

2.28 (95% CrI ‐5.82 to 10.39)

d[3]

2.29 (95% CrI ‐3.01 to 7.58)

2.29 (95% CrI ‐6.94 to 11.51)

d[4]

2.29 (95% CrI ‐1.57 to 6.15)

2.29 (95% CrI ‐5.18 to 9.75)

d[5]

3.28 (95% CrI ‐2.59 to 9.15)

3.29 (95% CrI ‐6.64 to 13.22)

d[6]

0.3 (95% CrI ‐1.11 to 1.71)

0.29 (95% CrI ‐4.77 to 5.35)

d[7]

‐1.21 (95% CrI ‐5.19 to 2.78)

‐1.21 (95% CrI ‐8.77 to 6.34)

Dbar

41.68

42.11

42.7

pD

11.99

12.41

13.01

DIC

53.67

54.52

55.7

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and so on.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. There is no evidence of inconsistency.

Figures and Tables -
Table 17. Length of hospital stay ‐ results and model fit
Table 18. Length of hospital stay ‐ pair‐wise comparisons

NoVascClampNoFib

NoVascCUSANoFib

NoVascRFAblNoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

IntVascCUSANoFib

NoVascClampNoFib

MD 2.29; 95% CrI ‐2.03 to 6.61

MD 2.29; 95% CrI ‐3.01 to 7.58

MD 2.29; 95% CrI ‐1.57 to 6.15

MD 3.28; 95% CrI ‐2.59 to 9.15

MD 0.3; 95% CrI ‐1.11 to 1.71

MD ‐1.21; 95% CrI ‐5.19 to 2.78

NoVascCUSANoFib

MD 0; 95% CrI ‐6.84 to 6.83

MD 0; 95% CrI ‐5.79 to 5.79

MD 0.99; 95% CrI ‐6.3 to 8.28

MD ‐1.99; 95% CrI ‐6.54 to 2.55

MD ‐3.5; 95% CrI ‐9.37 to 2.38

NoVascRFAblNoFib

MD 0; 95% CrI ‐6.55 to 6.55

MD 0.99; 95% CrI ‐6.91 to 8.9

MD ‐1.99; 95% CrI ‐7.47 to 3.49

MD ‐3.5; 95% CrI ‐10.12 to 3.13

ConVascClampNoFib

MD 0.99; 95% CrI ‐6.03 to 8.02

MD ‐1.99; 95% CrI ‐6.1 to 2.12

MD ‐3.5; 95% CrI ‐9.04 to 2.05

ConVascSharpNoFib

MD ‐2.98; 95% CrI ‐9.02 to 3.06

MD ‐4.49; 95% CrI ‐11.58 to 2.61

IntVascClampNoFib

MD ‐1.51; 95% CrI ‐5.73 to 2.72

IntVascCUSANoFib

The treatment codes are provided in Table 6.

CrI: credible interval; MD: mean difference (days).

Figures and Tables -
Table 18. Length of hospital stay ‐ pair‐wise comparisons
Table 19. Intensive therapy unit stay ‐ results and model fit

Fixed effect model

Random‐effects model

Inconsistency model (random‐effects)

d[2]

0 (95% CrI ‐3.32 to 3.33)

‐0.01 (95% CrI ‐6.56 to 6.55)

d[3]

0.01 (95% CrI ‐3.32 to 3.34)

‐0.01 (95% CrI ‐6.55 to 6.53)

d[4]

0.01 (95% CrI ‐4.69 to 4.71)

‐0.01 (95% CrI ‐9.27 to 9.24)

d[5]

‐2.18 (95% CrI ‐6.38 to 2.01)

‐2.2 (95% CrI ‐11.19 to 6.79)

d[6]

‐3.68 (95% CrI ‐9.04 to 1.68)

‐3.7 (95% CrI ‐14.81 to 7.41)

Dbar

27.07

27.07

27.07

pD

9

9

9

DIC

36.07

36.07

36.06

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and so on.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. There is no evidence of inconsistency.

Figures and Tables -
Table 19. Intensive therapy unit stay ‐ results and model fit
Table 20. Intensive therapy unit stay ‐ pair‐wise comparisons

NoVascCUSANoFib

NoVascRFAblNoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

IntVascCUSANoFib

NoVascCUSANoFib

MD 0; 95% CrI ‐3.32 to 3.33

MD 0.01; 95% CrI ‐3.32 to 3.34

MD 0.01; 95% CrI ‐4.69 to 4.71

MD ‐2.18; 95% CrI ‐6.38 to 2.01

MD ‐3.68; 95% CrI ‐9.04 to 1.68

NoVascRFAblNoFib

MD 0.01; 95% CrI ‐4.7 to 4.71

MD 0.01; 95% CrI ‐5.75 to 5.76

MD ‐2.19; 95% CrI ‐7.54 to 3.17

MD ‐3.68; 95% CrI ‐9.99 to 2.62

ConVascClampNoFib

MD 0; 95% CrI ‐5.76 to 5.76

MD ‐2.19; 95% CrI ‐7.55 to 3.16

MD ‐3.69; 95% CrI ‐10 to 2.62

ConVascSharpNoFib

MD ‐2.19; 95% CrI ‐8.49 to 4.11

MD ‐3.69; 95% CrI ‐10.82 to 3.44

IntVascClampNoFib

MD ‐1.5; 95% CrI ‐8.3 to 5.31

IntVascCUSANoFib

The treatment codes are provided in Table 6.

CrI: credible interval; MD: mean difference (days).

Figures and Tables -
Table 20. Intensive therapy unit stay ‐ pair‐wise comparisons
Table 21. Operating time ‐ results and model fit

Fixed‐effect model

Random‐effects model

d[2]

8.41 (95% CrI ‐61.25 to 78.07)

7.94 (95% CrI ‐62.15 to 78.03)

d[3]

10.42 (95% CrI ‐74.02 to 94.86)

9.33 (95% CrI ‐75.48 to 94.14)

d[4]

7.25 (95% CrI ‐47.15 to 61.64)

6.83 (95% CrI ‐47.78 to 61.43)

d[5]

49.61 (95% CrI 29.81 to 69.41)

49.35 (95% CrI 28.83 to 69.87)

Dbar

67.38

67.4

pD

7.47

7.5

DIC

74.85

74.9

d[2] indicates the log odds ratio between treatment 2 and treatment 1; d[3] indicates the log odds ratio between treatment 3 and treatment 1; and so on.

Dbar indicates the posterior mean of the residual deviance.

pD indicates the effective number of parameters (leverage).

DIC indicates the 'Deviance Information Criterion'.

A lower Dbar indicates a better model fit. However, a model with lower DIC is generally chosen to aid better interpretation as it takes the model complexity into account. A lower DIC indicates a better model fit. Differences of less than 3 to 5 between the models are not considered important.

Based on the above information, fixed‐effect model is the preferred model. There is no evidence of inconsistency.

Figures and Tables -
Table 21. Operating time ‐ results and model fit
Table 22. Operating time ‐ pairwise comparisons

NoVascCUSANoFib

ConVascClampNoFib

ConVascSharpNoFib

IntVascClampNoFib

IntVascCUSANoFib

NoVascCUSANoFib

MD 8.41; 95% CrI ‐61.25 to 78.07

MD 10.42; 95% CrI ‐74.02 to 94.86

MD 7.25; 95% CrI ‐47.15 to 61.64

MD 49.61; 95% CrI 29.81 to 69.41

ConVascClampNoFib

MD 2.01; 95% CrI ‐107.45 to 111.47

MD ‐1.17; 95% CrI ‐89.55 to 87.21

MD 41.2; 95% CrI ‐31.22 to 113.61

ConVascSharpNoFib

MD ‐3.18; 95% CrI ‐103.61 to 97.26

MD 39.19; 95% CrI ‐47.54 to 125.92

IntVascClampNoFib

MD 42.37; 95% CrI ‐15.52 to 100.25

IntVascCUSANoFib

The treatment codes are provided in Table 6. Results in bold text are significant.

CrI: credible interval; MD: mean difference (minutes).

Figures and Tables -
Table 22. Operating time ‐ pairwise comparisons