Loco-regional therapies competing with radiofrequency ablation in potential indications for hepatocellular carcinoma: a network meta-analysis

Article information

Clin Mol Hepatol. 2023;29(4):1013-1028
Publication date (electronic) : 2023 July 5
doi : https://doi.org/10.3350/cmh.2023.0131
1Gastroenterology and Hepatology, Hanyang University College of Medicine, Guri, Korea
2Biostatistics, College of Medicine, Korea University, Seoul, Korea
3Asan Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
4Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
Corresponding author : Ju Hyun Shim Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea Tel: +82-2-3010-3190, Fax: +82-2-485-5782, E-mail: s5854@amc.seoul.kr
Seungbong Han Department of Biostatistics, College of Medicine, Korea University, 73, Goryeodae-ro, Seongbuk-gu, Seoul 02841, Korea Tel: +82-2-2286-1425, Fax: +82-2-2286-1438, E-mail: hanseungbong@gmail.com
*HI Kim and J An contributed equally to this work.
Editor: Ju Dong Yang, Cedars-Sinai Medical Center, USA
Received 2023 March 29; Revised 2023 July 3; Accepted 2023 July 4.

Abstract

Background/Aims

There is no clear consensus on the relative ranking of interventional and radiation techniques with indications similar to those of radiofrequency ablation (RFA) for the treatment of early hepatocellular carcinoma (HCC). We used a network meta-analysis to compare the efficacy of non-surgical treatments for early HCC.

Methods

We searched databases for randomized trials assessing the efficacy of loco-regional treatments for HCCs ≤5 cm with no extrahepatic spread or portal invasion. The primary outcome was the pooled hazard ratio (HR) for overall survival (OS), and secondary outcomes included overall and local progression-free survival (PFS). A frequentist network meta-analysis was performed, and the relative ranking of therapies was assessed with P-scores.

Results

Nineteen studies comparing 11 different strategies in 2,793 patients were included. Chemoembolization plus RFA improved OS better than RFA alone (HR 0.52, 95% confidence interval [CI] 0.33–0.82; P-score=0.951). Cryoablation, microwave ablation, laser ablation, and proton beam therapy had similar effects on OS compared with RFA. For overall PFS, but not local PFS, only chemoembolization plus RFA performed significantly better than RFA (HR 0.61, 95% CI 0.42–0.88; P-score=0.964). Injection of percutaneous ethanol or acetic acid was significantly less effective than RFA for all measured outcomes, while no differences in progression outcomes were identified for other therapies included in the network.

Conclusions

Our results suggest that chemoembolization combined with RFA is the best option for local treatment of early HCC. Cases with potential contraindications for RFA may benefit from a tailored approach using thermal or radiation modalities.

Graphical Abstract

INTRODUCTION

Surgical resection and liver transplantation are the mainstays of curative treatment for early hepatocellular carcinoma (HCC) within the Milan criteria for tumor size and number [1,2]. Radiofrequency ablation (RFA) has achieved comparable outcomes to surgery, with fewer complications, in several randomized controlled trials (RCTs) and remains the standard of care for patients with small HCCs that are unresectable or medically inoperable, or for whom a donor is not available [3,4]. However, despite advances in imaging guidance and efforts to improve procedural safety [5], poor accessibility to intrahepatic lesions as well as proximity to blood vessels or biliary ductslimit the broad application of RFA [6,7].

Since the initial clinical implementation of RFA, prospective and retrospective studies on other interventional and radiation techniques have reported favorable outcomes in terms of local tumor progression and life expectancy. Several milestone trials of loco-regional methods in early HCC have focused on alternative primary strategies such as microwave ablation (MWA), cryoablation (CA), stereotactic body radiation therapy (SBRT), and proton beam therapy (PBT). These approaches, alone or combined with transarterial chemoembolization (TACE), are considered the next standard option for tumors within the Milan criteria according to international guidelines [1,8-12].

There is no consensus on the relative effectiveness of these local treatments because there is a lack of high-level direct evidence from head-to-head pairwise comparisons. Moreover, previous meta-analyses did not consider all relevant interventions, including radiotherapies, that had comparable efficacies, as demonstrated in randomized or matched-pair studies, and that were useful alternativesto standard therapy in HCC indicated for RFA [13,14]. Before the results of pairwise treatments become available, there is an urgent need for evidence-based indicators of the most appropriate procedure for early unresectable tumors. To address this critical real-life question, we conducted a systemic literature review of prospective pairwise comparisons of different types of loco-regional HCC treatments. Based on our findings, we performed a network meta-analysis (NMA) comparing treatment efficacy in terms of controlling target lesions and improving patient survival and then estimated the relative benefits of individual approaches by combining direct and indirect evidence.

MATERIALS AND METHODS

This study adhered to the standard guidelines of Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension statement for network meta-analysis (PRISMANMA) [15], and the study was registered in PROSPERO (protocol no. CRD42021278742). The institutional review board of Asan Medical Center approved this trial-level NMA, and the requirement for informed consent of individual patients was waived (IRB No. 2021-0823). Data generated or analyzed during the study are available from the corresponding authors by request.

Eligibility criteria

RCTs published as full-text articles in peer-reviewed journals that prospectively examined the efficacy of loco-regional therapies for primary or recurrent HCCs ≤5 cm without extrahepatic spread or portal invasion, which are generally accepted as standard indications for RFA, were eligible for this NMA [11,16]. We included studies comparing single or combined modalities with alternative intervention(s) that reported extractable data for at least one measure of overall survival (OS) and local or overall recurrence/progression. Only full text articles published in English in peer-reviewed journals were included.

Studies that had any arm of liver resection or transplantation or those that included any HCC >5 cm in diameter, which can be difficult to ablate, were excluded from the analysis. Duplicates, letters, conference proceedings, meeting abstracts, and prospective or retrospective studies of non-randomized design were also excluded.

Search strategy and selection criteria

Two separate literature searches were conducted to identify studies relevant to this NMA. The primary search was conducted on March 17, 2021, using the following databases: PubMed, EMBASE, Cochrane Library, CINAHL, and Web of Science. The search was limited to articles published in English between January 1, 2000, and March 17, 2021. The updated search was conducted on February 17, 2023, and used the same search strategy and databases to identify any newly published studies that met the inclusion criteria. Furthermore, we checked references cited in the relevant systematic reviews and meta-analyses. The detailed search strategy with query terms is presented in Supplementary Table 1. Two reviewers (JA and HIK, with 12 and 6 years of experience in the field of HCC treatment, respectively) independently screened all titles and abstracts identified by the searches and then scrutinized all the full manuscripts containing potentially relevant studies selected by either reviewer using the predefined criteria. Any discrepancies were resolved by discussion and consensus, or with the participation of an additional reviewer (JHS, with 19 years of experience in the field of HCC treatment).

Outcome definition and data extraction

The primary outcome was OS defined as the time from the date of enrollment to death from any cause. Secondary outcomes were overall progression-free survival (PFS) and local PFS. Based on the definition of endpoints used in all the included trials, overall progression included the following local and distant tumor events: (1) local progression defined as intrahepatic tumor recurrence or progression onto or along the peripheral margin of the treated lesion; and (2) development of any new HCC remote from the treated site, in any location, defined as distant progression. Overall PFS was measured by the interval from enrollment to either local/distant progression or death, whichever was first. For studies in which PFS was not reported as an endpoint, time elapsed to objective tumor progression or recurrence (TTP) was substituted asthe secondary outcome in the NMA [17]. The principal data extracted or derived from the included studies were the log hazard ratio (HR) and standard error (SE) or relevant information allowing estimation of HR and SE (e.g., an HR and confidence interval [CI] or P-value for survival, recurrence, and/or progression). All data were publicly available or computable from the individual studies. Additional summary data including details ofstudy design, treatment methods, and numbers of patients and their demographics were also extracted (Table 1).

Baseline characteristics of the study participants in 19 randomized controlled studies included in the network meta-analysis

Risk of bias assessment

Risk of bias was assessed for each study independently by two of the authors (JA and HIK) using the revised Cochrane risk-of-bias tool for RCTs [18].

Statistical analysis

To conduct the NMA, we required the log HRs for the survival outcomes and the corresponding SEs. However, for studies reporting only the P-value for the log-rank test along with the total number of events (e.g., deaths or progressions) for comparison groups, we derived the log HRs and SEs indirectly from the log-rank test results. For studies with no available log-rank test or log HRs, we calculated the log HRs and SEs indirectly from the Kaplan–Meier curves after constructing >15 time intervals [19]. After obtaining the summary statistics for the survival outcomes, we derived the therapeutic hierarchy for several therapies from an NMA in which we treated RFA as the control group.

We initially fitted both the fixed effects and random effects models simultaneously and evaluated study heterogeneity across the included trials. Statistical heterogeneity was calculated using the Higgins and Thomson I2 statistic, which describes the percentage of total variation across studies due to heterogeneity rather than chance [20]. We defined the substantial heterogeneity range as comprising values >50%. We also considered Cochran’s Q test for heterogeneity. Since there was no substantial evidence for heterogeneity across the studies with respect to any endpoint, we reported the results for the fixed effect model only. For direct and indirect comparisons between interventional and radiation techniques, we used a frequentist NMA approach employing weighted least squares regression [21]. In order to rank the treatments in terms of superiority, we computed the P-score for each treatment, which measures the degree of certainty that the treatment is better than another treatment, averaged over all the competing treatments [22]. We also performed subgroup analyses for the sub-networks involving studies based on HCC nodules ≤3 cm. Details of the statistical methods are provided in the Supplementary methods. All data analyses were performed using R packages (version 4.0.4.) netmeta and gemtc to conduct the main NMA and network-meta regression.

RESULTS

Study selection and characteristics

Figure 1 shows a flowchart of the study selection process. A total of 4,209 titles and abstracts of potentially relevant studies were screened. Of these, 48 fulfilled the eligibility criteria for full-text assessment. We examined the references in the relevant systematic reviews but identified no new records, as all references were already included in our database search results. Of the 48 studies, 29 were excluded after applying the exclusion criteria (Supplementary Table 2). Finally, 19 RCTs investigating 11 interventions in 2,793 patients were used for the NMA; these included the following treatment arms: RFA (1,124 patients in 15 trials) [6,8,10,23-34], TACE+RFA (115 in 2) [31,32], MWA (276 in 4) [23,25,34,35], CA (180 in 1) [10], laser ablation (LA; 70 in 1) [26], PBT (72 in 1) [8], TACE (84 in 1) [35], TACE+MWA (89 in 1) [35], percutaneous ethanol (PEI; 585 in 9) [6,24,27-29,33,36-38] and acetic acid injections (PAI; 159 in 3) [29,30,38], and TACE+PEI (39 in 2) [36,37].

Figure 1.

PRISMA flow diagram of the process of screening and selecting studies. PRISMA, Preferred Reporting Items for Systematic Reviews and Meta-Analyses; RCT, randomized controlled trial; HCC, hepatocellular carcinoma.

A network map of the treatment relationships and comparisons is presented in Figure 2. Node size and line thickness were proportional to the number of included patients and number of trials, respectively. Characteristics of the 19 studies included in the network are summarized in Table 1. The percentage of patients with solitary HCC ranged from 56.9% to 100%, while the percentage of patients with multifocal HCCs ranged from 0% to 43.1%. The number of tumors per patient in each study ranged from 1 to 4, and only eight patients in two trials, Shiina et al. [33], 2005 and Paul et al. [30], 2020, had >3 index lesions. Overall, 37.9% to 97.2% of patients were classified as Child-Turcotte-Pugh (CTP) class A, and 2.8% to 55.9% of patients were classified as CTP class B. All included trials reported OS as a study outcome; 11 included overall PFS (or TTP); and 11 included local PFS (or TTP). Local or overall PFS results were substituted for the corresponding TTP in seven and one studies, respectively.

Figure 2.

Network plots for (A) overallsurvival, (B) overall progression-free survival, and (C) local progression-free survival for direct comparisons of 19, 10, and 9 selected RCTs, respectively. Circle sizes reflect numbers of participants, while line widths reflect numbers of direct comparisons. The absence of a connecting line between two treatments indicates that there was no direct comparison. RCT, randomized controlled trial; RFA, radiofrequency ablation; TACE, transarterial chemoembolization; PEI, percutaneous ethanol injection; MWA, microwave ablation; PBT, proton beam therapy; PAI, percutaneous acetic acid injection; LA, laser ablation; CA, cryoablation.

Risk of bias assessments

Most studies were graded as low risk for the domains including randomization process, missing outcome data, measurement of outcome data, and selection of the reported result. Three studies reported the results of a modified intention-to-treat analysis, which were considered to be at high risk for domain deviations from intended interventions. Overall, the trials were considered to be at low risk for bias. Details of these assessments are presented in Figure 3 and Supplementary Figure 1.

Figure 3.

Evaluation of risk of biasin the randomized controlled trials.

OS analysis

The NMA included all RCTs in the OS analysis (Fig. 2A). Only TACE+RFA had significantly better OS than RFA when used for HCCs ≤5 cm (HR, 0.52; 95% CI, 0.33–0.82; Fig. 4A). Conversely, PEI (1.51, 1.16–1.96) and PAI (1.99, 1.30–3.06) resulted in poorer OS than RFA. No significant differences in OS were observed between RFA and the following treatments: TACE+MWA, CA, PBT, MWA, and LA. Probabilistic ranking metrics based on P-scores indicated that TACE+RFA (0.951) also ranked highest of all 11 treatment classes with respect to magnitude of treatment effect on OS, with a 61.4% probability of being the most effective option on the rankogram (Fig. 4A and Supplementary Fig. 2A). TACE+MWA and CA were the second-best and third-best treatments, respectively, and PAI had the highest probability (48.3%) of being the lowest ranked. Compared to RFA, no significant difference in OS was found with TACE (HR, 1.53; 95% CI, 0.74–3.16) despite having the second lowest P-score (0.279). The same trend in the ranking of treatments according to OS was observed for the extrapolated parametric survival NMA model for time-to-event (Fig. 5).

Figure 4.

Forest plots of the fixed effects network meta-analysis modelsfor (A) overallsurvival (OS), (B) overall progression-free survival (PFS), and (C) local PFS, with treatments ranked by the probability of being the best therapy based on P-scores in the trials finally selected. (A) The OS model. Data used were from all 19 trialsinvestigating 11 treatment modalitiesinvolving the 2,793 patients that were finally selected for this study. (B) The overall PFS model. Data used were from 10 trials investigating eight treatment modalities in 1,477 patients. (C) The local PFS model. Data used were from nine trials investigating eight treatment modalities in 1,394 patients. HR, hazard ratio; CI, confidence interval; TACE, transarterial chemoembolization; RFA, radiofrequency ablation; MWA, microwave ablation; CA, cryoablation; PBT, proton beam therapy; LA, laser ablation; PEI, percutaneous ethanol injection; PAI, percutaneous acetic acid injection.

Figure 5.

Overall survival probabilities with time for each treatment, based on estimates of the relative treatment effects. TACE, transarterial chemoembolization; RFA, radiofrequency ablation; MWA, microwave ablation; CA, cryoablation; PBT, proton beam therapy; LA, laser ablation; PEI, percutaneous ethanol injection; PAI, percutaneous acetic acid injection.

In pairwise comparisons within the complete network of 19 trials, TACE+RFA was significantly superior with respect to OS to every other treatment except TACE+MWA, CA, and PBT (Fig. 6). In addition, the four thermal or radiation therapies (i.e., CA, RFA, PBT, and MWA) were associated with significantly greater OS than PEI or PAI.

Figure 6.

League table showing hazard ratios(HRs) for pairwise comparisons of overallsurvival (OS) and overall progression-free survival (PFS) between treatments. Comparisons should be read from left to right. HRs(95% confidence intervals[CI]) for comparisons are in the cells shared by the column-defining and row-defining interventions. Numbers written in bold are statistically significant. For OS, an HR of <1 favors the row-defining treatment. For overall PFS, an HR of <1 favors the column-defining treatment. NA, not applicable; TACE, transarterial chemoembolization; RFA, radiofrequency ablation; MWA, microwave ablation; CA, cryoablation; PBT, proton beam therapy; LA, laser ablation; PEI, percutaneous ethanol injection; PAI, percutaneous acetic acid injection.

Overall PFS analysis

To evaluate overall PFS, we analyzed the relevant data from 10 studies involving eight different therapies in 1,477 patients (Fig. 2B). Compared to RFA alone, significant HRs for overall PFS were found for TACE+RFA (HR, 0.61; 95% CI, 0.42–0.88) and PAI (3.85; 1.25–11.79) (Fig. 4B). These two treatments were ranked in first and last place, respectively, based on P-score (0.964 and 0.030, respectively). The ranking profile was generally consistent with that of OS (Supplementary Fig. 2B). In addition, the seven pairwise treatment comparisons indicated a significant advantage of TACE+RFA over RFA, MWA, PEI, or PAI in improving overall PFS (Fig. 6).

Local PFS analysis

Local PFS analysis comprised nine studies investigating eight therapeutic options in 1,394 patients (Fig. 2C). No treatment modalities detected a local PFS benefit when compared with RFA (Fig. 4C). When ordered by P-score, CA and TACE+RFA were ranked in first and second place, respectively. The rankogram also indicated that CA was the most effective treatment in prolonging local PFS duration (Supplementary Fig. 2C). In terms of pairwise comparisons, all treatment pairs other than PAI and PEI yielded comparable rates of local PFS (Supplementary Fig. 3).

Adverse events

A total of 16 studies reported major adverse events among 2,304 patients undergoing 11 distinct therapies [6,8,10,23-31,33-38]. Due to the lack of a consistent definition for major adverse events across the studies, a quantitative assessment could not be made. The observed prevalence of major complications ranged from 0% to 7.7%,depending on the specific criteria used in each study.

Regarding treatment modalities, RFA was associated with a major complication rate that varied from 0% to 6.9% across 12 studies. Post-RFA complications included lung-related problems such as hemothorax, pneumothorax, and pleural effusion, in addition to hemorrhage, hepatic decompensation (manifesting as ascites or jaundice),malignant cell seeding, infection, and hepatic infarct/necrosis.

The major complication rates for other treatment modalities were as follows: PEI, 0–2.9% (eight studies); PAI, 0–7.7% (three studies); MWA, 2.2–2.8% (two studies); and TACE+RFA, 2.2–2.9% (two studies).Notable adverse events associated by modality were as follows: PEI (one death due to bowel infarction, neoplastic seeding, and liver abscess), PAI (two deaths resulting from hepatic necrosis and tumor rupture, tumor seeding, and gross hematuria), MWA (hemorrhage and tumor seeding), and TACE+RFA (hepatic decompensation and segmental hepatic infarction). The remaining six treatment modalities were each investigated in a single study. A comprehensive description of adverse events is presented in Supplementary Table 3.

Subgroup analysis

Among the 19 RCTs, seven studies included a total of 1,128 patients with HCC nodules ≤3 cm in diameter undergoing six distinct therapies. These studies were included in a network created to analyze HRs for OS. No significant differences in OS (HR [95% CI], 0.67 [0.21–2.20]; Supplementary Fig. 4A) were found between the combined TACE+RFA and RFA alone groups, although survival was slightly better in the TACE+RFA group. Similar results were obtained for both overall PFS in four studies with 652 patients (0.84 [0.51–1.41]) and local PFS in three studies with 420 patients (0.63 [0.25–1.59]), as shown in Supplementary Figure 4B and 4C.

Assessment of transitivity and inconsistency

Overall, the transitivity assumption was not challenged, as there were no significant differences in the baseline parameters examined to evaluate its plausibility (Supplementary Fig. 5). There were also no significant differences for either OS or local PFS in terms of inconsistencies between direct and indirect estimates in the node-splitting analysis within the closed loop in the evidence network (PEI-PAI-RFA) (Supplementary Table 4).

DISCUSSION

The above analysis based on the published outcomes of RCTs of loco-regional therapies revealed that combined TACE and RFA occupies the first position in a ranking of non-surgical treatments of early HCCs meeting the Milan criteria. The analysis based on the integrated scores for OS and PFS showed that combined TACE and RFA is superior to all the other mono- or dual-therapeutic options, including TACE and RFA individually. In addition, all single interventions, other than PEI and PAI, the oldest techniques for the treatment of HCC, had comparable survival- and progression-related efficacy compared to RFA, the standard strategy according to the current evidence-based management of this disease stage.

The RCTs and meta-analyses had limitations in terms of sample size,study quality, and statistical power. Nonetheless, the cumulative evidence from these studies indicated that TACE+RFA is superior to RFA alone in terms of survival and/or recurrence, particularly for studies including HCCs >3 cm, and that it was no associated with any significant major complications [39,40]. It is likely that the benefits of TACE followed as soon as possible by RFA derive from (1) improved control of microsatellite lesions due to the larger treated area [41]; (2) reduced vascular cooling effect that can lead to incomplete ablation; (3) prevention of portal invasion of the tumor by hepatic arterial and portal venous flows occluded by embolic materials; (4) boosting of the thermal effect by chemotherapeutic drugs and ischemic edema; and (5) optimization of heat diffusion by the disruption of intratumoral septa [42,43]. Our investigation of nodules up to 5 cm in diameter also concluded that this combination was more effective than RFA, MWA, or LA therapy alone. Furthermore, given the discrepancy between the overall (positive) and local (negative) PFS results of TACE+RFA, our results suggest that the addition of TACE may prolong OS, compared with RFA alone, by targeting microsatellites together around the target lesion. Surprisingly, our analysis revealed no beneficial effect on OS when adding TACE to MWA, although this combination was comparable to TACE+RFA based on pairwise comparisons. A retrospective study in the U.S. comparing 38 patients treated with TACE+RFA and 51 treated with TACE+MWA for HCCs of varying sizes yielded similar safety and efficacy outcomes [44]; the ranking of these combination therapies in term of safety and efficacy needs to be confirmed by further relevant RCTs.

Our findings indicate that TACE+RFA deserves primary consideration for curative control of early HCCs, apart from ultrasound-invisible HCCs requiring radiographic guidance for curative RFA treatment by TACE-induced iodized oil retention [45]. Subgroup analyses based on sub-networks of RCTs explicitly reporting data restricted to patients with HCC nodules ≤3 cm in diameter reported no significant differences in local PFS, overall PFS, or OS between combined TACE+RFA and RFA alone (Supplementary Fig. 4). The study from Japan of Shibata et al. [32] included in our network illustratesthis point. Nevertheless, these findings support the idea that RFA monotherapy is adequate for treating small (≤3 cm) HCCs, when patient convenience, hospital stay, and medical costs are taken into account [46]. Therefore, combination therapy may be more useful for medium-sized (3–5 cm) HCCs, as complete necrosis is difficult to achieve with RFA alone, particularly in infiltrating tumor types [16]. This conclusion is consistent with the current recommendations stipulated in the recently updated Korean guidelines [2].

For imaging-guided percutaneous ablation techniques, such as RFA, MWA, CA, and LA, our direct and indirect comparison data revealed no significant differences in both survival and progression endpoints among the mono-modalities; this is consistent with reports of individual paired comparative studies based on various design types [13,23,25,26]. Each technique has its own advantages and limitations compared with the traditional RFA reference: for instance, MWA can produce larger coagulation areas than RFA with shorter ablation times; it is also less sensitive to the heat-sink effect and hence achieves adequate ablation of nodules close to large vessels; however, it is contraindicated for treating nodules at high-risk locations as well as subcapsular nodules [47,48]. In spite of technical difficulties, LA is safer for treating nodules at difficult locations, as well as multiple lesions in one session, as it uses multiple fibers and spares the uninvolved hepatic parenchyma [49]. Lastly, CA, which is much less painful than RFA, has the advantage of protecting better against vascular and biliary injuries by monitoring the extent of ablation during the procedure, despite producing larger ablation volumes with multiple probes [50]. Our findings provide prognostic evidence justifying a tailored approach in which other liver-directed thermal ablation procedures are placed alongside RFA as standard-of-care for maximizing treatment efficacy and minimizing procedure-related complications in patients considered to be candidates for HCC ablation.

Several HCC studies have reported good outcomes with hypo-fractionated PBT having a finite range of energy deposition and thus low rates of hepatic and gastrointestinal toxicity relative to photon beam therapy [8,51]. A recent U.S. registry study of 918 patients with T1 or T2 HCC suggested that PBT may have superior survival outcomes compared to SBRT [52]. The latter is another conformal external technique, which was not included in our NMA because of the absence of relevant RCTs. However, a meta-analysis of 70 non-comparative observational studies suggesting that the two radiotherapies have similar efficacy with respect to OS and overall and local PFSs warrants prospective corroboration [53]. Based on our NMA findings and the existing evidence, PBT (or SBRT if PBT is not feasible) may be considered alternative treatments for percutaneous options including RFA, particularly for nodules considered to be at high risk, with limited accessibility, or invisible under ultrasound guidance [54,55]. Unlike injection therapies with ethanol or acetic acid, which have few clinical applications and have been found to be inferior to RFA in numerous comparisons [6,24,33], TACE, with a broader spectrum of indications, remains the first choice in early-stage cases that are neither suitable nor appropriate for local ablation when based on the stage migration strategy [1]. Pairwise direct and indirect evidence from our NMA may justify a second-best role of TACE monotherapy as an alternative next-line option for HCCs below the intermediate stage [35]. It should also not be overlooked that several matched and non-matched studies of within-Milan HCCs have found that TACE has comparable effects to therapeutic ablation and radiation on tumors and patients [56,57].

Apart from the inherent limitations of collecting aggregate patient data from completed studies, some limitations of this work should be acknowledged. First, we could not include an estimate of safety profile as an outcome of interest in the present NMA because of the heterogeneous criteria for, and definitions of, procedure-related adverse events across the studies. However, almost all the intervention arms in all the RCTs had <5% serious adverse events(Table 1). Second, a few patients with CTP class C liver function or >3 target lesions who are not usually good candidates for loco-regional therapies but were included in our analysis, albeit rarely, may have affected the interpretation of survival and efficacy outcomes (Table 1) [25,28,30,36]. Lastly, in most (n=12) of the included RCTs, the inclusion criteria specified HCCs up to 5 cm, which prevented us from enrolling only ideal candidates with tumors ≤ 3cm. However, we were able to determine the outcomes for HCC lesions ≤3 cm through a subgroup analysis of seven studies.

In conclusion, in this loco-regional therapy-based NMA, we found that TACE+RFA was ranked highest with regard to both survival and progression outcomes in the treatment of patients with early HCCs ≤5 cm. Equivalent outcomes are likely for HCCs within the Milan criteria that can be optimally treated by thermal ablation using a variety of possible energy sources or PBT. Further evidence concerning specific indications for the individual modalities is urgently needed to develop precisely tailored strategies.

Notes

Authors’ contribution

HI Kim, J An, and JH Shim contributed to the study concept and design, acquisition, analysis and interpretation of data, verification of the underlying data, drafting of the manuscript, and critical revision of the manuscript for important intellectual content. S Han contributed to the statistical analysis, verification of the underlying data, drafting of the manuscript, and critical revision of the manuscript for important intellectual content. All authors confirm that they had full access to all the data in the study and accept responsibility for submission of this manuscript.

Conflicts of Interest

The authors have no conflictsto disclose.

Acknowledgements

This study was supported by grants from the National Research Foundation of Korea funded by the Ministry of Science and ICT (NRF-2022R1A2C3008956 and RS-2022-00166674), the Research Supporting Program of The Korean Association for the Study of the Liver and The Korean Liver Foundation (KASLKLF2018-05) and Asan Institute for Life Sciences, Asan Medical Center (2022IP0046).

Supplementary materials

Supplementary material is available at Clinical and Molecular Hepatology website (http://www.e-cmh.org).

Supplementary Figure 1.

Detailed profiles of risk of biasin the randomized controlled trials.

cmh-2023-0131-Supplementary-Fig-1.pdf
Supplementary Figure 2.

Ranking curves displaying the probabilities per rank of (A) overall survival, (B) overall progression-free survival (PFS), and (C) local PFS for each treatment. PAI, percutaneous acetic acid injections; CA, cryoablation; RFA, radiofrequency ablation; LA, laser ablation; PBT, proton beam therapy; TACE, trans-arterial chemoembolization; MWA, microwave ablation; PEI, percutaneous ethanol injection.

cmh-2023-0131-Supplementary-Fig-2.pdf
Supplementary Figure 3.

League table showing hazard ratios of local progression-free survival for pairwise comparisons between treatments. Comparisons should be read from left to right. HRs (95% confidence intervals [CI]) for comparisons are in the shared cells. PAI, percutaneous acetic acid injections; CA, cryoablation; RFA, radiofrequency ablation; LA, laser ablation; PBT, proton beam therapy; TACE, trans-arterial chemoembolization; MWA, microwave ablation; PEI, percutaneous ethanol injection; HRs, hazard ratios.

cmh-2023-0131-Supplementary-Fig-3.pdf
Supplementary Figure 4.

Treatment effects for (A) overall survival and (B) overall and (C) local progression-free survivals in the sensitivity analysis based on randomized controlled trials(RCTs) limited for HCC nodules ≤3 cm in diameter. Six, five, and five treatmentsfrom seven, four, and three of the 19 RCTs, respectively, were included in networks created to analyze hazard ratios (HRs) for overall survival and overall and local progression-free survivals, respectively. PAI, percutaneous acetic acid injections; CA, cryoablation; RFA, radiofrequency ablation; LA, laser ablation; PBT, proton beam therapy; TACE, trans-arterial chemoembolization; MWA, microwave ablation; PEI, percutaneous ethanol injection; HCC, hepatocellular carcinoma; CI, confidence interval.

cmh-2023-0131-Supplementary-Fig-4.pdf
Supplementary Figure 5.

Checking the transitivity assumption. The study and population baselines evaluable in all trials were included in testing transitivity. The patients in all of the comparisons were similar in age (left) and other main parameters with a P-value>0.05 (right). On the left, the blue bar indicates mean age with 3 standard deviations of the mean, while the red bar indicates the median age with range. *Median ages with interquartile range were provided in Vietti Violi et al. [34], 2018; Shiina et al. [33], 2005 was excluded from the assessment because of the absence of data on the mean or median age of the patients. PAI, percutaneous acetic acid injections; CA, cryoablation; RFA, radiofrequency ablation; LA, laser ablation; PBT, proton beam therapy; TACE, trans-arterial chemoembolization; MWA, microwave ablation; PEI, percutaneous ethanol injection; CTP, Child-Turcotte-Pugh.

cmh-2023-0131-Supplementary-Fig-5.pdf
Supplementary Table 1.

Search strategies with query terms

cmh-2023-0131-Supplementary-Table-1.pdf
Supplementary Table 2.

Exclusion criteria and characteristics of excluded studies(n=29)

cmh-2023-0131-Supplementary-Table-2.pdf
Supplementary Table 3.

Major adverse eventsin 16 randomized controlled studies

cmh-2023-0131-Supplementary-Table-3.pdf
Supplementary Table 4.

Node-splitting analysis of inconsistency between direct and indirect effectsin a closed loop network (PEI-PAI-RFA) in terms of overallsurvival and local progression-free survival

cmh-2023-0131-Supplementary-Table-4.pdf
REFERENCES FOR SUPPLEMENTARY MATERIALScmh-2023-0131-Supplementary-Reference.pdf

Abbreviations

CA

cryoablation

CI

confidence interval

CTP

Child-Turcotte-Pugh

HCC

hepatocellular carcinoma

HR

hazard ratio

LA

laser ablation

MWA

microwave ablation

NMA

network meta-analysis

OS

overall survival

PAI

percutaneous acetic acid injection

PBT

proton beam therapy

PEI

percutaneous ethanol injection

PFS

progression-free survival

PRISMA

Preferred Reporting Items for Systematic Reviews and Meta-Analyses

RCT

randomized controlled trial

RFA

radiofrequency ablation

SBRT

stereotactic body radiation therapy

SE

standard error

TACE

transarterial chemoembolization

TTP

time-to-progression

References

1. European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. J Hepatol 2018;69:182–236.
2. Korean Liver Cancer Association (KLCA) and National Cancer Center (NCC) Korea. 2022 KLCA-NCC Korea practice guidelines for the management of hepatocellular carcinoma. Clin Mol Hepatol 2022;28:583–705.
3. Xu XL, Liu XD, Liang M, Luo BM. Radiofrequency ablation versus hepatic resection for small hepatocellular carcinoma: Systematic review of randomized controlled trials with meta-analysis and trialsequential analysis. Radiology 2018;287:461–472.
4. Takayama T, Hasegawa K, Izumi N, Kudo M, Shimada M, Yamanaka N, et al. Surgery versus radiofrequency ablation for small hepatocellular carcinoma: A randomized controlled trial (SURF trial). Liver Cancer 2021;11:209–218.
5. Lee DH, Lee JM. Recent advances in the image-guided tumor ablation of liver malignancies: Radiofrequency ablation with multiple electrodes, real-time multimodality fusion imaging, and new energy sources. Korean J Radiol 2018;19:545–559.
6. Lencioni RA, Allgaier HP, Cioni D, Olschewski M, Deibert P, Crocetti L, et al. Small hepatocellular carcinoma in cirrhosis: randomized comparison of radio-frequency thermal ablation versus percutaneous ethanol injection. Radiology 2003;228:235–240.
7. Rhim H, Yoon KH, Lee JM, Cho Y, Cho JS, Kim SH, et al. Major complications after radio-frequency thermal ablation of hepatic tumors: spectrum of imaging findings. Radiographics 2003;23:123–134. discussion 134-136.
8. Kim TH, Koh YH, Kim BH, Kim MJ, Lee JH, Park B, et al. Proton beam radiotherapy vs. radiofrequency ablation for recurrent hepatocellular carcinoma: A randomized phase III trial. J Hepatol 2021;74:603–612.
9. Yu J, Yu XL, Han ZY, Cheng ZG, Liu FY, Zhai HY, et al. Percutaneous cooled-probe microwave versus radiofrequency ablation in early-stage hepatocellular carcinoma: a phase III randomised controlled trial. Gut 2017;66:1172–1173.
10. Wang C, Wang H, Yang W, Hu K, Xie H, Hu KQ, et al. Multicenter randomized controlled trial of percutaneous cryoablation versus radiofrequency ablation in hepatocellular carcinoma. Hepatology 2015;61:1579–1590.
11. Rajyaguru DJ, Borgert AJ, Smith AL, Thomes RM, Conway PD, Halfdanarson TR, et al. Radiofrequency ablation versus stereotactic body radiotherapy for localized hepatocellular carcinoma in nonsurgically managed patients: Analysis of the national cancer database. J Clin Oncol 2018;36:600–608.
12. Yoon SM, Kim SY, Lim YS, Kim KM, Shim JH, Lee D, et al. Stereotactic body radiation therapy for small (≤5 cm) hepatocellular carcinoma not amenable to curative treatment: Results of a single-arm, phase II clinical trial. Clin Mol Hepatol 2020;26:506–515.
13. Gupta P, Maralakunte M, Kumar-M P, Chandel K, Chaluvashetty SB, Bhujade H, et al. Overall survival and local recurrence following RFA, MWA, and cryoablation of very early and early HCC: a systematic review and Bayesian network meta-analysis. Eur Radiol 2021;31:5400–5408.
14. Lin Y, Wen Q, Guo L, Wang H, Sui G, Sun Z. A network metaanalysis on the efficacy and prognosis of different interventional therapies for early-stage hepatocellular carcinoma. Int J Hyperthermia 2018;35:450–462.
15. Hutton B, Salanti G, Caldwell DM, Chaimani A, Schmid CH, Cameron C, et al. The PRISMA extension statement for reporting of systematic reviews incorporating network meta-analyses of health care interventions: checklist and explanations. Ann Intern Med 2015;162:777–784.
16. Livraghi T, Goldberg SN, Lazzaroni S, Meloni F, Ierace T, Solbiati L, et al. Hepatocellular carcinoma: radio-frequency ablation of medium and large lesions. Radiology 2000;214:761–768.
17. Giuliano M, Schettini F, Rognoni C, Milani M, Jerusalem G, Bachelot T, et al. Endocrine treatment versus chemotherapy in postmenopausal women with hormone receptor-positive, HER2-negative, metastatic breast cancer: a systematic review and network meta-analysis. Lancet Oncol 2019;20:1360–1369.
18. Sterne JAC, Savović J, Page MJ, Elbers RG, Blencowe NS, Boutron I, et al. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ 2019;366:l4898.
19. Parmar MK, Torri V, Stewart L. Extracting summary statistics to perform meta-analyses of the published literature for survival endpoints. Stat Med 1998;17:2815–2834.
20. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ 2003;327:557–560.
21. Rücker G. Network meta-analysis, electrical networks and graph theory. Res Synth Methods 2012;3:312–324.
22. Rücker G, Schwarzer G. Ranking treatments in frequentist network meta-analysis works without resampling methods. BMC Med Res Methodol 2015;15:58.
23. Abdelaziz A, Elbaz T, Shousha HI, Mahmoud S, Ibrahim M, Abdelmaksoud A, et al. Efficacy and survival analysis of percutaneous radiofrequency versus microwave ablation for hepatocellular carcinoma: an Egyptian multidisciplinary clinic experience. Surg Endosc 2014;28:3429–3434.
24. Brunello F, Veltri A, Carucci P, Pagano E, Ciccone G, Moretto P, et al. Radiofrequency ablation versus ethanol injection for early hepatocellular carcinoma: A randomized controlled trial. Scand J Gastroenterol 2008;43:727–735.
25. Chong CCN, Lee KF, Cheung SYS, Chu CCM, Fong AKW, Wong J, et al. Prospective double-blinded randomized controlled trial of Microwave versus RadioFrequency Ablation for hepatocellular carcinoma (McRFA trial). HPB (Oxford) 2020;22:1121–1127.
26. Di Costanzo GG, Tortora R, D’Adamo G, De Luca M, Lampasi F, Addario L, et al. Radiofrequency ablation versus laser ablation for the treatment of small hepatocellular carcinoma in cirrhosis: a randomized trial. J Gastroenterol Hepatol 2015;30:559–565.
27. Giorgio A, Di Sarno A, De Stefano G, Scognamiglio U, Farella N, Mariniello A, et al. Percutaneous radiofrequency ablation of hepatocellular carcinoma compared to percutaneous ethanol injection in treatment of cirrhotic patients: an Italian randomized controlled trial. Anticancer Res 2011;31:2291–2295.
28. Lin SM, Lin CJ, Lin CC, Hsu CW, Chen YC. Radiofrequency ablation improves prognosis compared with ethanol injection for hepatocellular carcinoma < or =4 cm. Gastroenterology 2004;127:1714–1723.
29. Lin SM, Lin CJ, Lin CC, Hsu CW, Chen YC. Randomised controlled trial comparing percutaneous radiofrequency thermal ablation, percutaneous ethanol injection, and percutaneous acetic acid injection to treat hepatocellular carcinoma of 3 cm or less. Gut 2005;54:1151–1156.
30. Paul SB, Acharya SK, Gamanagatti SR, Sreenivas V, Shalimar S, Gulati MS. Acetic acid versus radiofrequency ablation for the treatment of hepatocellular carcinoma: A randomized controlled trial. Diagn Interv Imaging 2020;101:101–110.
31. Peng ZW, Zhang YJ, Liang HH, Lin XJ, Guo RP, Chen MS. Recurrent hepatocellular carcinoma treated with sequential transcatheter arterial chemoembolization and RF ablation versus RF ablation alone: a prospective randomized trial. Radiology 2012;262:689–700.
32. Shibata T, Isoda H, Hirokawa Y, Arizono S, Shimada K, Togashi K. Small hepatocellular carcinoma: is radiofrequency ablation combined with transcatheter arterial chemoembolization more effective than radiofrequency ablation alone for treatment? Radiology 2009;252:905–913.
33. Shiina S, Teratani T, Obi S, Sato S, Tateishi R, Fujishima T, et al. A randomized controlled trial of radiofrequency ablation with ethanol injection for small hepatocellular carcinoma. Gastroenterology 2005;129:122–130.
34. Vietti Violi N, Duran R, Guiu B, Cercueil JP, Aubé C, Digklia A, et al. Efficacy of microwave ablation versus radiofrequency ablation for the treatment of hepatocellular carcinoma in patients with chronic liver disease: a randomised controlled phase 2 trial. Lancet Gastroenterol Hepatol 2018;3:317–325.
35. Zaitoun MMA, Elsayed SB, Zaitoun NA, Soliman RK, Elmokadem AH, Farag AA, et al. Combined therapy with conventional transarterial chemoembolization (cTACE) and microwave ablation (MWA) for hepatocellular carcinoma >3-<5cm. Int J Hyperthermia 2021;38:248–256.
36. Koda M, Murawaki Y, Mitsuda A, Oyama K, Okamoto K, Idobe Y, et al. Combination therapy with transcatheter arterial chemoembolization and percutaneous ethanol injection compared with percutaneous ethanol injection alone for patients with small hepatocellular carcinoma: a randomized control study. Cancer 2001;92:1516–1524.
37. Mizuki A, Tatemichi M, Tsukada N, Nagamatsu R, Kawaguchi M, Itoshima T, et al. Addition of transcatheter arterial chemoembolization decreased local recurrence but had no survival benefit to percutaneous ethanol injection therapy for patients with small hepatocellular carcinoma: A multicenter randomized controlstudy. Oncol Lett 2010;1:855–859.
38. Tsai WL, Cheng JS, Lai KH, Lin CP, Lo GH, Hsu PI, et al. Clinical trial: percutaneous acetic acid injection vs. percutaneous ethanol injection for small hepatocellular carcinoma--a long-term follow-up study. Aliment Pharmacol Ther 2008;28:304–311.
39. Lan T, Chang L, Mn R, Wu L, Yuan YF. Comparative efficacy of interventional therapies for early-stage hepatocellular carcinoma: A PRISMA-compliant systematic review and network meta-analysis. Medicine (Baltimore) 2016;95e3185.
40. Peng ZW, Zhang YJ, Chen MS, Xu L, Liang HH, Lin XJ, et al. Radiofrequency ablation with or without transcatheter arterial chemoembolization in the treatment of hepatocellular carcinoma: a prospective randomized trial. J Clin Oncol 2013;31:426–432.
41. Izzo F, Granata V, Grassi R, Fusco R, Palaia R, Delrio P, et al. Radiofrequency ablation and microwave ablation in liver tumors: An update. Oncologist 2019;24:e990–e1005.
42. Seki T, Tamai T, Nakagawa T, Imamura M, Nishimura A, Yamashiki N, et al. Combination therapy with transcatheter arterial chemoembolization and percutaneous microwave coagulation therapy for hepatocellular carcinoma. Cancer 2000;89:1245–1251.
43. Goldberg SN, Girnan GD, Lukyanov AN, Ahmed M, Monsky WL, Gazelle GS, et al. Percutaneous tumor ablation: increased necrosis with combined radio-frequency ablation and intravenous liposomal doxorubicin in a rat breast tumor model. Radiology 2002;222:797–804.
44. Ginsburg M, Zivin SP, Wroblewski K, Doshi T, Vasnani RJ, Van Ha TG. Comparison of combination therapies in the management of hepatocellular carcinoma: transarterial chemoembolization with radiofrequency ablation versus microwave ablation. J Vasc Interv Radiol 2015;26:330–341.
45. Park BJ, Byun JH, Jin YH, Won HJ, Shin YM, Kim KW, et al. CT-guided radiofrequency ablation for hepatocellular carcinomas that were undetectable at US: therapeutic effectiveness and safety. J Vasc Interv Radiol 2009;20:490–499.
46. Naugler WE, Sonnenberg A. Survival and cost-effectiveness analysis of competing strategies in the management of small hepatocellular carcinoma. Liver Transpl 2010;16:1186–1194.
47. Yu J, Cheng ZG, Han ZY, Liu FY, Zheng RQ, Cheng W, et al. Period-dependent survival benefit of percutaneous microwave ablation for hepatocellular carcinoma: A 12-year real-world, multicentric experience. Liver Cancer 2022;11:341–353.
48. Tamai H, Okamura J. New next-generation microwave thermosphere ablation for small hepatocellular carcinoma. Clin Mol Hepatol 2021;27:564–574.
49. Sartori S, Di Vece F, Ermili F, Tombesi P. Laser ablation of liver tumors: An ancillary technique, or an alternative to radiofrequency and microwave? World J Radiol 2017;9:91–96.
50. Song KD. Percutaneous cryoablation for hepatocellular carcinoma. Clin Mol Hepatol 2016;22:509–515.
51. Hong TS, Wo JY, Yeap BY, Ben-Josef E, McDonnell EI, Blaszkowsky LS, et al. Multi-institutional phase II study of high-dose hypofractionated proton beam therapy in patients with localized, unresectable hepatocellular carcinoma and intrahepatic cholangiocarcinoma. J Clin Oncol 2016;34:460–468.
52. Hasan S, Abel S, Verma V, Webster P, Arscott WT, Wegner RE, et al. Proton beam therapy versus stereotactic body radiotherapy for hepatocellular carcinoma: practice patterns, outcomes, and the effect of biologically effective dose escalation. J Gastrointest Oncol 2019;10:999–1009.
53. Qi WX, Fu S, Zhang Q, Guo XM. Charged particle therapy versus photon therapy for patients with hepatocellular carcinoma: a systematic review and meta-analysis. Radiother Oncol 2015;114:289–295.
54. Yoon SM. External beam radiotherapy for hepatocellular carcinoma: A review of the current guidelines in the east and the west. J Liver Cancer 2021;21:25–33.
55. Jang JY, Jung J, Lee D, Shim JH, Kim KM, Lim YS, et al. Stereotactic body radiation therapy for elderly patients with small hepatocellular carcinoma: a retrospective observational study. J Liver Cancer 2022;22:136–145.
56. Hsu CY, Huang YH, Chiou YY, Su CW, Lin HC, Lee RC, et al. Comparison of radiofrequency ablation and transarterial chemoembolization for hepatocellular carcinoma within the Milan criteria: a propensity score analysis. Liver Transpl 2011;17:556–566.
57. Kim JW, Kim JH, Sung KB, Ko HK, Shin JH, Kim PN, et al. Transarterial chemoembolization vs. radiofrequency ablation for the treatment of single hepatocellular carcinoma 2 cm or smaller. Am J Gastroenterol 2014;109:1234–1240.

Article information Continued

Notes

Study Highlights

• In a network meta-analysis of 19 randomized trials exploring 11 loco-regional therapies, only the combination of TACE and RFA showed a significant improvement in the overall survival of patients with HCC measuring ≤5 cm, compared to RFA alone (HR, 0.52; 95% CI, 0.33–0.82). This combination treatment also ranked first based on P-score (0.964).

• An analysis of overall progression-free survival involving eight treatments in ten trials revealed that the HR for the combination therapy was significantly better than that for RFA alone (HR, 0.58; 95% CI, 0.38–0.89), again having the highest P-score (0.999).

• No modalities outperformed RFA alone in terms of local progression-free survival rates.

Figure 1.

PRISMA flow diagram of the process of screening and selecting studies. PRISMA, Preferred Reporting Items for Systematic Reviews and Meta-Analyses; RCT, randomized controlled trial; HCC, hepatocellular carcinoma.

Figure 2.

Network plots for (A) overallsurvival, (B) overall progression-free survival, and (C) local progression-free survival for direct comparisons of 19, 10, and 9 selected RCTs, respectively. Circle sizes reflect numbers of participants, while line widths reflect numbers of direct comparisons. The absence of a connecting line between two treatments indicates that there was no direct comparison. RCT, randomized controlled trial; RFA, radiofrequency ablation; TACE, transarterial chemoembolization; PEI, percutaneous ethanol injection; MWA, microwave ablation; PBT, proton beam therapy; PAI, percutaneous acetic acid injection; LA, laser ablation; CA, cryoablation.

Figure 3.

Evaluation of risk of biasin the randomized controlled trials.

Figure 4.

Forest plots of the fixed effects network meta-analysis modelsfor (A) overallsurvival (OS), (B) overall progression-free survival (PFS), and (C) local PFS, with treatments ranked by the probability of being the best therapy based on P-scores in the trials finally selected. (A) The OS model. Data used were from all 19 trialsinvestigating 11 treatment modalitiesinvolving the 2,793 patients that were finally selected for this study. (B) The overall PFS model. Data used were from 10 trials investigating eight treatment modalities in 1,477 patients. (C) The local PFS model. Data used were from nine trials investigating eight treatment modalities in 1,394 patients. HR, hazard ratio; CI, confidence interval; TACE, transarterial chemoembolization; RFA, radiofrequency ablation; MWA, microwave ablation; CA, cryoablation; PBT, proton beam therapy; LA, laser ablation; PEI, percutaneous ethanol injection; PAI, percutaneous acetic acid injection.

Figure 5.

Overall survival probabilities with time for each treatment, based on estimates of the relative treatment effects. TACE, transarterial chemoembolization; RFA, radiofrequency ablation; MWA, microwave ablation; CA, cryoablation; PBT, proton beam therapy; LA, laser ablation; PEI, percutaneous ethanol injection; PAI, percutaneous acetic acid injection.

Figure 6.

League table showing hazard ratios(HRs) for pairwise comparisons of overallsurvival (OS) and overall progression-free survival (PFS) between treatments. Comparisons should be read from left to right. HRs(95% confidence intervals[CI]) for comparisons are in the cells shared by the column-defining and row-defining interventions. Numbers written in bold are statistically significant. For OS, an HR of <1 favors the row-defining treatment. For overall PFS, an HR of <1 favors the column-defining treatment. NA, not applicable; TACE, transarterial chemoembolization; RFA, radiofrequency ablation; MWA, microwave ablation; CA, cryoablation; PBT, proton beam therapy; LA, laser ablation; PEI, percutaneous ethanol injection; PAI, percutaneous acetic acid injection.

Table 1.

Baseline characteristics of the study participants in 19 randomized controlled studies included in the network meta-analysis

Study name (Country) Arm No. of patients Inclusion criteria for tumor Main tumor size* (cm) Single HCC (%) HCC ≤3 cm (%) Male sex (%) Age* (years) Follow-up duration (months) CTP class A/B/C (%) Outcome Major complications (%)
Abdelaziz et al. [23] 2014 (Egypt) RFA 45 All <5 cm, number ≤3 2.95 86.7 61.5 68.9 56.8 27 53.3/46.7/- OS ND
MWA 66 2.90 86.4 72.4 72.7 53.6 27 37.9/62.1/- ND
Brunello et al. [24] 2008 (Italy) PEI 69 All ≤3 cm, number ≤3 2.25 78.7 100 71.0 70.3 26 56.5/43.5/- OS 2.9
RFA 70 2.42 77.1 100 61.4 69 25 54.3/45.7/- 2.9
Chong et al. [25] 2020 (Hong Kong) MWA 47 All ≤5 cm, number ≤3 Median, 3.1 91.5 51.1 63.8 Median, 63 38.3 83.0/14.9/2.1 OS, overall PFS ND
RFA 46 Median, 2.8 84.8 32.6 82.6 Median, 64.5 34 87.0/13.0/- ND
Di Costanzo et al. [26] 2015 (Italy) RFA 70 Within Milan criteria 2.55 90.0 90.0 75.7 Median, 70 Mean, 42.8 90.0/10.0/- OS, local PFS 1.4
LA 70 2.62 87.1 78.6 67.1 Median, 70 Mean, 42.2 95.7/4.3/- 1.4
Giorgio et al. [27] 2011 (Italy) PEI 143 Single ≤3 cm 2.27 100 100 71.3 72 37 52.4/47.6/- OS ND
RFA 142 2.34 100 100 73.9 70 37 49.3/50.7/- ND
Kim et al. [8] 2021 (South Korea) PBT 72 All <3 cm, number ≤2 Median, 1.2 93.1 100 84.7 Median, 60 51.6 97.2/2.8/- OS, overall PFS, local PFS 0
RFA 72 Median, 1.2 91.7 100 81.9 Median, 61.5 50.8 97.2/2.8/- 1.4
Koda et al. [36] 2001 (Japan) TACE+PEI 26 All <3 cm, number ≤3 2.0 61.5 100 53.8 66.2 29 73.1/19.2/7.7 OS 7.7
PEI 26 1.9 57.7 100 69.2 66.4 31.2 53.8/30.8/15.4 0
Lencioni et al. [6] 2003 (Italy) RFA 52 Within Milan criteria 2.8 76.9 88.5 69.2 67 22.9 86.5/13.5/- OS, overall PFS, local PFS 0
PEI 50 2.8 62.0 84.0 60.0 69 22.4 70.0/30.0/- 0
Lin et al. [28] 2004 (Taiwan) RFA 52 All 1-4 cm, number ≤3 2.9 73.1 71.2 67.3 62 24.5 78.8/21.2/- OS, overall PFS, local PFS 0
PEI 52 2.8 76.9 73.1 65.4 59 23.8 75.0/25.0/- 0
Lin et al. [29] 2005 (Taiwan) RFA 62 All ≤3 cm, number ≤3 2.5 79.0 100 64.5 61 28 74.2/25.8/- OS, overall PFS, local PFS 4.8
PEI 62 2.3 79.0 100 62.9 60 26 75.8/24.2/- 0
PAI 63 2.3 76.2 100 66.7 63 26 71.4/28.6/- 0
Mizuki et al. [37] 2010 (Japan) PEI 14 All 2-4 cm, number ≤3 2.64 78.6 ND 50.0 63.6 33 ND OS, overall PFS 0
TACE+PEI 13 2.64 61.5 ND 69.2 65.8 39.7 ND 0
Paul et al. [30] 2020 (India) PAI 26 All ≤5 cm, number ≤ 5 2.7 80.8 66.7 76.9 53.5 13 73.1/26.9/- OS 7.7
RFA 29 2.7 89.7 57.6 69.0 54.5 15 62.1/37.9/- 6.9
Peng et al. [31] 2012 (China) TACE+RFA 69 Within Milan criteria 2.1 94.2 59.4 85.5 57.5 39.2 87.0/13.0/- OS, overall PFS 2.9
RFA 70 2.1 92.9 65.7 78.6 55.1 33.6 84.3/15.7/- 2.9
Shibata et al. [32] 2009 (Japan) TACE+RFA 46 All ≤3 cm, number ≤3 1.7 ND 100 67.4 67.2 30.4 69.6/30.4/- OS, overall PFS, local PFS 2.2
RFA 43 1.6 ND 100 76.7 69.8 30.4 76.7/23.3/- 2.3
Shiina et al. [33] 2005 (Japan) RFA 118 All ≤3 cm, number ≤3 ND 61.0 100 66.9 ≤65/>65, n=44/74 37.2 72.0/28.0/- OS, overall PFS 3.4
PEI 114 ND 52.6 100 76.3 ≤65/>65, n=41/73 34.8 74.6/25.4/- 2.6
Tsai et al. [38] 2008 (Taiwan) PAI 70 All ≤4 cm, number ≤3 2.2 84.3 ND 67.1 65 Mean, 43 80.0/20.0/- OS, local PFS 1.4
PEI 55 2.2 85.5 ND 74.5 66 Mean, 43 70.9/29.1/- 0
Vietti Violi et al. [34] 2018 (France and Switzerland) MWA 70 All ≤4 cm, number ≤3 1.8 62.0 ND 83.1 Median, 68 26 80.3/19.7/- OS, local PFS 2.8
RFA 73 1.8 63.0 ND 84.9 Median, 65 25 72.6/27.4/- 4.1
Wang et al. [10] 2015 (China) CA 180 All ≤4 cm, number ≤2 ND 89.4 ND 77.8 53.9 25 67.2/32.8/- OS, overall PFS, local PFS 3.9
RFA 180 ND 95.0 ND 83.3 53.3 25 60.6/39.4/- 3.3
Zaitoun et al. [35] 2021 (Egypt) TACE 84 Single 3-5 cm 3.6 100 ND 61.9 51.3 Mean, 19 84.5/15.5/- OS 3.6
MWA 92 3.9 100 ND 54.3 53.8 Mean, 21 84.8/15.2/- 2.2
TACE+MWA 89 3.7 100 ND 58.4 52.1 Mean, 24 89.9/10.1/- 1.1

HCC, hepatocellular carcinoma; CTP, Child-Turcotte-Pugh; RFA, radiofrequency ablation; MWA, microwave ablation; PEI, percutaneous ethanol injection; LA, laser ablation; TACE, transarterial chemoembolization; PBT, proton beam therapy; PAI, percutaneous acetic acid injections; CA, cryoablation; OS, overall survival; PFS, progression-free survival; ND, not described in the text.

*

Main tumor size and age are presented as mean values, unless otherwise stated.

Follow-up durations are presented as median values, unless otherwise stated.

Results for major complications were based on the criteria defined and set by the individual studies: (for example) bowel perforation, hemorrhage, infection, third space fluid collection needing intervention, pneumothorax needing chest tube, needle-track tumor seeding, hepatic failure, and procedure-related death.