Back to Journals » Journal of Hepatocellular Carcinoma » Volume 9

Trial Designs for Integrating Novel Therapeutics into the Management of Intermediate-Stage Hepatocellular Carcinoma

Authors Su YY , Liu YS, Hsiao CF, Hsu C, Chen LT

Received 1 March 2022

Accepted for publication 23 April 2022

Published 2 June 2022 Volume 2022:9 Pages 517—536

DOI https://doi.org/10.2147/JHC.S220978

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Ahmed Kaseb



Yung-Yeh Su,1– 3,* Yi-Sheng Liu,4,* Chin-Fu Hsiao,5 Chiun Hsu,6– 8 Li-Tzong Chen1,2,9,10

1National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; 2Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; 3Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; 4Department of Medical Imaging, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan, Taiwan; 5Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan; 6Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan; 7Graduate Institute of Oncology, National Taiwan University, Taipei, Taiwan; 8National Taiwan University Cancer Center, Taipei, Taiwan; 9Department of Internal Medicine, Kaohsiung Medical University Hospital, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; 10Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan

*These authors contributed equally to this work

Correspondence: Li-Tzong Chen, Department of Internal Medicine, Kaohsiung Medical University Hospital, 100 Tzyou 1 st Road, Kaohsiung, 80756, Taiwan, Tel +886-7-3121101 ext 7451, Fax +886-7-3135612, Email [email protected] Chiun Hsu, Department of Oncology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, 10002, Taiwan, Tel +886 2 23123456 ext. 62859, Fax +886 2 23711174, Email [email protected]

Abstract: Intermediate-stage hepatocellular carcinoma (HCC) consists of heterogeneous groups of patients in terms of tumor burden and organ function reserves. Although liver-directed therapy (LDT), including trans-catheter arterial chemoembolization, radiofrequency ablation or even surgical resection, is the recommended frontline treatment modality, intrahepatic and distant failures are common. The recent advances in systemic treatment, notably the introduction of immune checkpoint inhibitor (ICI)-based therapy, have significantly improved the objective tumor response rate, quality of response and overall survival in patients with recurrent and advanced HCC. Whether the combination of systemic treatment and LDT can further improve the outcome of patients with intermediate-stage HCC is currently being extensively evaluated. In this article, the recent clinical trials incorporating different ICI-based combinations with different LDT for intermediate-stage HCC were reviewed focusing on trial design issues, including patient selection, endpoint definition, and biomarker development. The strength and caveats of different combination strategies and novel biomarker development were discussed.

Keywords: immunotherapy, liver-directed therapy, biomarker

Introduction

In the past few years we have witnessed major advances in systemic therapy for hepatocellular carcinoma (HCC) in the following aspects. First, immune checkpoint inhibitor (ICI)-based combination therapy has demonstrated a survival benefit, compared with single-agent sorafenib therapy, as first-line systemic therapy for unresectable HCC, thus becoming the new standard therapy for unresectable HCC.1–4 Anti-PD1/ anti-PDL1 ICI therapy is currently the backbone for development of new therapeutic regimens for HCC. Second, with more than a decade of struggle over failed clinical trials,5 the platforms of clinical and translational research of drug development for HCC became more mature to accommodate new treatment options in different clinical settings.6–9 Third, the heterogeneity of HCC biology, clinical presentation and practice patterns among different geographic regions was increasingly recognized and incorporated in clinical trials for advanced HCC as stratification factors. The difference in practice patterns was also recognized by international practice guidelines to refine treatment recommendations based on randomized controlled trials, cohort studies, as well as expert consensus10–14

The advances in systemic therapy for HCC ushered in a burgeoning of clinical trials of systemic therapy in earlier stage HCC, mostly in combination with liver-directed therapy (LDT). However, initial attempts at combining the multi-kinase inhibitor (MKI) sorafenib with trans-catheter arterial chemo-embolization (TACE) did not demonstrate improved overall survival for patients who received combination therapy (Supplementary Table S1).15–20 Design and conduct of this type of clinical trials have to tackle several challenges. TACE may not be suitable for all patients with intermediate-stage HCC, and the criteria of patient selection and determination of TACE refractoriness are still evolving.11,21 Determination of tumor progression based on imaging criteria was subjected to evaluation bias and led to inconsistent adherence to study treatment. Furthermore, interpretation of survival benefit in these trials was confounded by heterogeneous post-progression therapy and treatment cross-over.22,23

To realize the therapeutic potential of the new systemic therapy in intermediate-stage HCC, the above-mentioned challenges have to be overcome, and novel approaches of biomarker development for patient enrichment and efficacy prediction have to be developed.24 In this article, the study design of different treatment strategies for intermediate stage HCC were reviewed, in the context of the most updated practice guidelines from the Barcelona Clinic Liver Cancer (BCLC),13 the American Association for the Study of Liver Diseases (AASLD),25 and the European Society for Medical Oncology (ESMO).14 Adaptation based on variations of practice patterns around the world was addressed. Novel approaches of biomarker development and future perspectives were discussed.

The Evolving Concept of Heterogeneity in Intermediate-Stage HCC

Intermediate-stage (BCLC stage B) HCC was originally defined as HCC patients with preserved liver function, good performance status, and multifocal liver tumors (>3 nodules or >3 cm in diameter) without vascular invasion or extrahepatic spread. Discussion of heterogeneity issues initially focused on the definition of tumor burden because of its direct impact on treatment choice, particularly the use of TACE. The up-to-seven criteria were the most commonly used criteria in sub-classification of BCLC stage B patients.21,26,27 Although the criteria were developed to identify candidates for liver transplantation,28 patients within the up-to-seven criteria were also more likely to benefit from TACE, and selected patients may even be considered for surgery or ablation therapy with curative intent.29,30 At the other end of the spectrum, patients with diffuse, bilobar tumors or infiltrative tumors were unlikely to benefit from TACE, and systemic therapy was increasingly recommended as the first-line therapy for this subgroup of patients.11,13,14,31 Therefore, a key issue of defining the subgroups based on liver tumor burden is to facilitate selection between liver-directed therapy and systemic therapy.11,32

Another key issue of sub-grouping is to stratify patients for better prognostic prediction, which is vital for clinical trial design. In previous randomized trials of TACE plus targeted therapy for intermediate-stage HCC, various criteria of tumor burden were used to stratify subjects (Supplementary Table S1). Variations of the up-to-seven criteria were also proposed, based on retrospective analysis of large patient cohorts, for predicting response to TACE and survival in intermediate-stage HCC.33–35 These variations partly reflected the different practice patterns of liver-directed therapy in different geographic regions.36,37 Moreover, the prognosis of HCC patients is closely related to the patients’ liver function reserves, which may be impaired by the underlying liver diseases or previous liver-directed therapy. The Child–Pugh classification is at best a crude estimate of the patients’ liver function status, and new scoring systems, such as the ALBI grade, the HAP score, and the ART score, may provide more detailed prognostic information.38–40 Harmonization of the stratification criteria in international clinical trials can be a challenging task.

The multitude of liver-directed therapies added yet another aspect in the heterogeneity of management for intermediate-stage HCC. TACE was recommended as standard of care for intermediate-stage HCC primarily based on meta-analysis of early randomized trials.41,42 However, the type and dosage of chemotherapeutic agents, the schedule and techniques of intervention, and the criteria to determine progression and re-treatment, differed from country to country and even from institution to institution.43–45 Other liver-directed therapies, including yttrium-90 selective internal radiation therapy (Y-90 SIRT, also known as trans-arterial radioembolization, TARE), external beam radiation therapy (EBRT), and hepatic arterial infusional chemotherapy (HAIC), were recommended by regional practice guidelines, based primarily on retrospective or prospective cohort studies as well as expert opinions (Table 1).13,46–52 Of note, HAIC was recently shown by a randomized trial from China to achieve superior overall and progression-free survival, compared with TACE, for large unresectable HCC.53 HAIC and EBRT may also improve survival in HCC patients with portal vein thrombosis.54,55 These liver-directed therapies may serve as “post-progression” therapy in clinical trials for intermediate-stage HCC, and the survival analysis will be confounded by the availability and technical expertise of these treatment options.

Table 1 Variations of Treatment Recommendation for Intermediate-Stage HCC by Different Practice Guidelines

Challenges of Designing Clinical Trials for Intermediate-Stage HCC

Although overall survival (OS) is the traditional endpoint for clinical trials pursuing regulatory approval of new treatments, using OS to evaluate the efficacy of new treatment strategies for intermediate-stage HCC has the following caveats. First, the median OS in intermediate-stage HCC usually exceeds two years but varies widely because of the different tumor characteristics mentioned earlier. Estimation of the survival outcome for the control group is thus difficult. Second, as the treatment stage migration becomes more and more common, the survival outcome of both the treatment group and the control group in new clinical trials for intermediate-stage HCC is likely to improve.56,57 Third, the potential survival benefit of the new treatment tended to be negated by the use of post-progression therapy (liver-directed or systemic). Therefore, the value of surrogate efficacy endpoints was increasingly recognized.6

Using surrogate endpoints to evaluate the efficacy of new anti-cancer therapy continues to be a trade-off between efficiency of clinical trials and risk of inadequate scientific and clinical validity.58,59 The correlation between OS and surrogate efficacy endpoints, including PFS, time to progression (TTP), and objective response rate (ORR), was explored in 21 Phase III trials of advanced HCC.60 A moderate correlation (R = 0.83–0.84) between OS and PFS or TTP was found, and the investigators suggested that a PFS hazard ratio of ≤0.6 may predict OS benefit of new treatment for advanced HCC.60 However, this conclusion was derived primarily from the positive trials of targeted therapy (for advanced stage HCC) in the second-line setting, in which the post-progression survival was relatively short. The confounding effect of post-progression survival will be more problematic in clinical trials for earlier stages of HCC.61

The most challenging issue for the design of clinical trials for intermediate-stage HCC is to overcome the confounding effects of heterogeneity of patient characteristics and treatment patterns. Stratified (block) randomization is the most commonly used method to balance important covariates among treatment groups in randomized trials. Other covariate-adaptive randomization techniques to improve the marginal balance (i.e., balance at individual covariate levels) were recommended, such as minimization or dynamic hierarchical randomization, which allowed different levels of imbalance among the covariates.62 However, balancing on the margins of the stratification variables will increase the variance of the treatment effect estimates more than stratified permuted blocks, thus impairing the efficiency of the primary endpoint analysis.63 A hybrid approach to both achieving marginal balance and improving the inefficiency caused by minimization or dynamic hierarchical randomization was thus proposed.64 In reality, although these new randomization methods may accommodate more stratification factors than conventional block randomization, some factors, such as the participating sites in international trials, are still difficult to handle, thus harmonization of practice patterns is crucial for the efficiency of clinical trials for intermediate-stage HCC.

PFS is increasingly being adopted in HCC trials as a primary endpoint. Therefore, the potential bias associated with the criteria to determine progression and re-treatment for HCC patients who received liver-directed therapy must be carefully controlled.65 Tumor response measurement by imaging should be done at regular intervals with the same frequency among treatment arms, and the results should be centrally reviewed to avoid evaluation bias. Definition of progression and feasibility of further liver-directed therapy should be clarified to avoid variation of drop-out among treatment arms (attrition bias). In clinical trials of TACE plus targeted therapy for intermediate-stage HCC, drop-out may be due to either tumor progression or deterioration of organ function, and the drop-out patterns of different treatment arms should be analyzed in detail. Moreover, in real-world practice patients may receive repeated TACE for intra-hepatic recurrence unless major vascular invasion or extra-hepatic metastases occurred. Therefore, the criteria of “time to untreatable/unTACEable progression (TTUP)” based on response to prior TACE, occurrence of major vascular invasion or extra-hepatic metastases, and organ function reserves, were developed.11 Three studies (BRISK-TA, ORIENTAL and TACTICS) incorporated this concept to define tumor progression, and all of the 3 studies demonstrated improved TTP for the study drug arm compared with the control arm. However, no OS benefit was observed in these studies to date.

Ongoing Trials of Immunotherapy in Intermediate Stage HCC

To obtain a comprehensive view of development of immune checkpoint inhibitors-based therapy, anti-PD1/ anti-PDL1 with and without anti-CTLA4 for intermediate stage HCC, we searched the clinical trials registered at ClinicalTrials.gov and World Health Organization International Clinical Trials Registry Platform (WHO ICTRP), using combinations of the following keywords: (hepatocellular carcinoma) AND (intermediate stage OR locoregional OR downstage) AND (nivolumab OR pembrolizumab OR tislelizumab OR camrelizumab OR sintilimab OR cemiplimab OR toripalimab OR atezolizumab OR durvalumab OR avelumab OR ipilimumab OR tremelimumab). A total of 102 studies were screened and 42 studies, including 8 phase III and 34 phase I/II studies, were included in this review (Figure 1). The following aspects of trial design were further analyzed: types and timing of liver-directed intervention, definition of potential confounding factors, and the prospect of systemic therapy alone for intermediate stage HCC.

Figure 1 PRISMA flow chart for the selection of studies. Number of studies identified by search terms of (hepatocellular carcinoma) AND (intermediate stage OR locoregional OR downstage) AND (nivolumab OR pembrolizumab OR tislelizumab OR camrelizumab OR sintilimab OR cemiplimab OR toripalimab OR atezolizumab OR durvalumab OR avelumab OR ipilimumab OR tremelimumab). Last accessed on November 21, 2021.

Type of Liver-Directed Intervention

Liver-directed therapy may induce immunogenic cell death and therefore enhance the efficacy of immunotherapy.66 Among 42 ongoing trials as of November 21, 2021, TACE was the most commonly used local reginal therapy, followed by Y-90 SIRT. Although cell damage caused by TACE may increase exposure of tumor antigens, TACE-induced hypoxia may increase hypoxia inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) production in tumor microenvironment that resulted in immune suppression.67,68 On the other hand, Y-90 SIRT may induce more tumor infiltrating lymphocytes (TILs) than TACE or direct surgical resection.69 In a randomized phase II study, Y-90 SIRT demonstrated significantly longer TTP and numerically better OS.70 Non-randomized studies also suggested better tolerability, shorter hospitalization, and comparable survival compared with historical results of standard treatments.71,72 The efficacy and safety of ICI therapy plus Y-90 SIRT was demonstrated in a phase II study by Lee et al.73 Of 36 treated subjects, 11 subjects showed objective response as per RECIST 1.1, while ≥ grade 3 treatment-related adverse events occurred in only 5 of 36 subjects. Comparison between ICI therapy plus Y-90 SIRT and ICI therapy plus TACE is ongoing (NCT04522544).

EBRT was generally reserved for patients with localized HCC not suitable for resection or ablation,74,75 and was only listed as a treatment option by the KLCSG and TLCA practice guideline (Table 1). A recent randomized trial demonstrated EBRT plus TACE was superior to sorafenib in HCC patients with vascular invasion.54 Besides, 2 non-randomized studies using propensity score-matched analysis demonstrated comparable OS and better local control for EBRT, compared with TACE.76,77 Induction of immunogenic cell death by EBRT has been extensively studied, which has been shown to improve antitumor efficacy of ICI therapy in other cancer types.78–81 It is thus reasonable to explore potential synergistic antitumor immunity between EBRT and ICI therapy in HCC.

The use of HAIC was also heterogeneous in terms of types and dosage of chemotherapeutic agents and criteria of patient selection. HAIC has been widely adopted by Japanese physicians, and retrospective studies disclosed comparable efficacy between HAIC and TACE.82–86 However, it has not been recommended by most international and regional practice guidelines.14,31,46,47,49,52 One study group in China reported 2 positive randomized trials of HAIC using oxaliplatin, leucovorin, and fluorouracil in HCC. The first trial reported superior OS of HAIC plus sorafenib, compared with sorafenib monotherapy, in HCC patients with portal vein invasion.55 The second trial reported superior OS, PFS, and response rate for patients with unresectable HCC, compared with sorafenib, in patients with unresectable HCC.87 Reproducibility of these promising results should be confirmed before incorporating HAIC into clinical practice guidelines. Nevertheless, combination of HAIC and ICI therapy is reasonable because of the potential of immunogenic cell death induction by cytotoxic agents.

Timing of Liver-Directed and ICI Intervention

Most of the earlier studies combining TACE and targeted therapy adopted the post-LDT approach (Figure 2). Theoretically, HIF-1α, VEGF, and other cytokines induced by TACE may promote tumor angiogenesis, recurrence and metastasis after TACE.88–90 Therefore, immediate inhibition of angiogenesis after TACE may provide the best therapeutic effect. In the Japan-Korea Post-TACE study, central image review of response to TACE was required before randomization. This requirement resulted in delay of sorafenib treatment for subjects randomized to the sorafenib treatment arm (about 60% of subjects started treatment more than 9 weeks after TACE), and the delay of treatment start may be associated with worse clinical outcome.15 All subsequent trials randomized the subjects before TACE and 3 studies (SPACE, TACE-2 and TACTICS) even started study drug treatment before TACE. Starting drug treatment before TACE allowed the physicians to adjust the optimal tolerated dosage of study drugs before TACE and therefore drug treatment after TACE may be less likely to be interrupted by treatment-related toxicities.

Figure 2 Ongoing trials of ICI-based therapy in intermediate stage HCC. The pie chart (left) demonstrates trial numbers of liver-directed therapy in combination with ICIs. The stacked bars (right) elucidate the composition of trial design with different liver-directed therapy. The timing of TACE are not clearly described in three trials including two randomized phase III trials.

Post-LDT therapy is also the most common design of randomized trials combining TACE and ICI-based therapy (Table 2 and Figure 3).91–94 The complexity of study design issues has been extensively reviewed elsewhere to get insight from previous trials.22,95 Most trials incorporated more detailed definition of patient characteristics (e.g., ALBI grade,96,97 HAP score,6,98,99 AFP level100) and treatment patterns (e.g., geographic regions or study sites, drug-eluting beads versus conventional TACE) in the hope of better stratification for this very heterogeneous patient population. Several trials incorporated TACE-specific endpoints, such as time to TACE progression (TTTP), to avoid confounding by post-progression therapy. OS was not used by all trials as a primary endpoint, most likely due to concerns of confounding by post-progression therapy.

Table 2 Randomized Phase III Trials of Combining TACE and ICI-Based Therapy in Intermediate Stage HCC

Figure 3 Eligibility criteria of tumor burden in ongoing randomized trials. Most trials do not preclude patients with low tumor burden (within Milan’s criteria) except 3 phase III trials (CheckMate-74W, RENOTACE and ABC-HCC). The RENOTACE trial, which directly compares systemic therapy with TACE, only includes patients beyond up-to-seven but without vascular invasion. Invasion of portal vein branch (Vp1-2) are acceptable in 3 phase III and 1 phase II trials. One phase III trial (NCT05056337) includes patients with main portal vein invasion (Vp3-4).

Neoadjuvant therapy became an attractive approach recently. Neoadjuvant ICI therapy may stimulate more diverse T cell responses to overcome the potential immune suppression induced by liver-directed therapy.101 The higher response rates of ICI-based therapy may increase the opportunity of tumor downstaging, but there is a risk of delay of primary treatment and disease progression, or even hyper-progression, during neoadjuvant therapy (see below).102–104

Early-phase clinical trials testing ICI-based systemic therapy for intermediate-stage HCC were more flexible in endpoint selection, timing of systemic therapy, and the choice of liver-directed therapy (Tables 35).73,105–112 The vast majority of these trials were single-arm studies, and PFS and ORR were commonly used as the primary endpoints. Both modified RECIST and RECIST 1.1 were used for response evaluation. Immune-related RECIST113 was not commonly used. Systemic therapy was given before (neoadjuvant), concurrent, after (adjuvant), or both before and after liver-directed therapy (peri-LDT). In a phase I study, Harding et al reported nivolumab could be safely delivered with TACE either as adjuvant (cohort 1), as peri-LDT (cohort 2) or concurrently (cohort 3) (Table 3).110 No new safety concern was observed; only one subject in cohort 3 experienced grade 3 transaminitis and this resolved spontaneously without recurrence after rechallenge of nivolumab. The efficacy of TACE followed by nivolumab was further demonstrated by Vogel et al in a phase II study, the IMMUTACE study.111 Of the 49 enrolled intermediate stage subjects, the ORR according to mRECIST was 71% and the median PFS and OS were 6.1 months and 28.3 months, respectively. The most common ≥ grade 3 adverse events were that aspartate aminotransferase increased (14%) and GGT increased (10%).

Table 3 Phase I/II Trials of Combining TACE and ICI-Based Therapy in Intermediate Stage HCC

Table 4 Other Liver-Directed Therapy Combined Systemic Therapy in Intermediate Stage HCC

Table 5 Systemic Therapy Without TACE

As mentioned earlier, neoadjuvant therapy is increasingly applied to intermediate stage HCC. Zhang et al reported a preliminary result of 38 subjects who underwent neoadjuvant lenvatinib plus camrelizumab/sintilimab and TACE, followed by resection if feasible.112 The ORR based on mRECIST was 84%. Of the 19 subjects (50%) who were able to undergo conversion resection, 6 had major pathological responses. Of note, the use of systemic therapy alone for intermediate-stage HCC is now entering the stage. About 15–20% of patients enrolled into previous clinical trials of systemic therapy for unresectable HCC had intermediate-stage disease, and these patients demonstrated similar efficacy/safety outcome compared with patients with advanced-stage disease.1,115 Retrospective studies suggested that systemic therapy may achieve better ORR and PFS compared with TACE in intermediate-stage HCC patients with high tumor burden.116,117 Currently clinical practice guidelines suggest upfront systemic therapy for intermediate-stage HCC with huge bilobar tumors or infiltrative tumors.11,13,14,31 Clinical trials of upfront systemic therapy, on the other hand, generally enrolled patients whose tumors exceeded the Milan criteria or the up-to-seven criteria (Table 5).6,11

Future Perspective: Neoadjuvant ICI-Based Therapy as “Window of Opportunity”

The increased efficacy, in terms of survival and ORR, and favorable safety profile of ICI-based combination therapy for HCC provides several “windows of opportunities” for patients with intermediate-stage disease. Surgery and other curative therapy were reasonable treatment options for selected patients with intermediate-stage HCC (Table 1),29,118,119 and response achieved by neoadjuvant systemic therapy may increase the proportion of patients for curative therapy and improve treatment outcome.120,121 Tumor or blood samples obtained at diagnosis may help develop predictive biomarkers of treatment efficacy, and samples obtained after treatment may help explore pharmacodynamic endpoints and mechanisms of antitumor immunity.

Kaseb et al reported the first neoadjuvant ICI-based therapy for HCC (Table 5).122 Patients with resectable HCC were randomized to pre-operative nivolumab monotherapy (240 mg every 2 weeks for up to 3 doses) or the same dosage of nivolumab plus ipilimumab 1 mg/kg concurrent with the first dose of nivolumab. Subjects in the nivolumab monotherapy group received adjuvant nivolumab, 480 mg every 4 weeks for up to 2 years, and subjects in the nivolumab plus ipilimumab group received adjuvant nivolumab, 480 mg every 4 weeks for up to 2 years plus ipilimumab 1 mg/kg every 6 weeks for up to 4 doses. Objective response to pre-operative therapy, according to RECIST 1.1, was achieved in 3 subjects in nivolumab group and 0 in nivolumab + ipilimumab group. Twenty of the 27 randomized subjects (13 in nivolumab group and 14 in nivolumab + ipilimumab group) received surgery, and major pathological response was achieved in 3 subjects in nivolumab group and 3 in nivolumab + ipilimumab group. Exploratory biomarker research using CyTOF and Nanostring transcriptomic analysis suggested that (1) increased immune cell infiltration in baseline tumor samples was associated with response to nivolumab therapy but less so with nivolumab + ipilimumab therapy; (2) major pathological response was associated with increased infiltration of activated T cells in the post-treatment samples; and (3) myeloid cells expressing the V-domain Ig suppressor of T-cell activation (VISTA) in the tumor microenvironment may contribute to resistance to ICI therapy.

This study highlighted several vital design issues for neoadjuvant or peri-operative systemic therapy for HCC. The regimen approved by the US FDA for advanced HCC (nivolumab 1 mg/kg and ipilimumab 3 mg/kg every 3 weeks, with a total of 4 doses of ipilimumab)123 was not adopted in this study, probably due to the concern of treatment-related toxicity of a higher dose of ipilimumab. Longer duration of pre-operative systemic therapy may increase the objective response rate and chance of downstaging, but again may increase the odds of treatment-related toxicity. The contribution of adjuvant therapy in long-term outcome, especially OS, is difficult to measure, considering the enormous variation of LDT and systemic therapy if the tumors recur.

Ho et al reported another pilot study of neoadjuvant cabozantinib (40 mg per day for 8 weeks) plus nivolumab (240 mg every 2 weeks at weeks 3, 5, 7 and 9) in patients with potentially resectable HCC (Table 5).124 Objective response per RECIST 1.1 was observed in only one of 15 subjects. Of the 12 subjects who successfully underwent surgical resection, 5 had major pathological responses. NanoString and CyTOF studies of the post-treatment tissue samples showed that (1) the number of tertiary lymphoid structures (TLS), effector CD8+ T cells, CD20+ B cells and CD 138+ plasma cells were significantly higher in responders than non-responders; (2) the existence of Arg-1/PD-1 expressing myeloid-derived suppressor cells (MDSCs) near T cells and B cells was associated with resistance to ICIs combination.

Transcriptomic analysis and CyTOF were two major approaches of high-dimensional analysis of the immune microenvironment.125 Nanostring technology pioneered using formalin-fixed, paraffin-embedded (FFPE) tumor samples to perform RNA analysis and measurement of immune cell contexture, using pre-defined gene signatures representing individual classes of immune cells. Now RNA-seq can be performed using FFPE samples, thus providing opportunities of more comprehensive exploration of immune-related gene expression patterns and regulatory mechanisms. CyTOF, on the other hand, provides phenotypic characterization of immune cells based on protein expression and may thus complement RNA-based analysis. Fresh tissue samples were required for CyTOF analysis. Newer technology, such as multiplexed ion beam imaging (MIBI), may use FFPE samples and may analyze the spatial relationship of immune cells in the tumor microenvironment. The most important opportunity provided by neoadjuvant therapy may be biomarker development. For biomarkers to predict prognosis or treatment efficacy, conventional predictive markers such as PD-L1 expression and tumor mutation burden (TMB) in HCC was limited because of the low incidence of high PD-L1 expression or high TMB in HCC. Rapid technological progress in multi-omics analysis enabled more comprehensive characterization of interaction of different immune cells in the tumor microenvironment and key signaling pathways of immune regulation.124,126 However, a common challenge was external validation of biomarkers identified by these approaches. In addition, recent studies suggested that enrichment of particular species in the gut microbiome of HCC patients was associated with response to ICI therapy.127,128 On the other hand, more specific pharmacodynamic assessments of drugs with different mechanisms still rely on analysis of post-treatment tumor samples, whose availability was seriously limited. Development of biomarkers using peripheral blood samples (circulating immune cells/DNA, cytokines, etc.) will greatly enhance our ability to detect direct pharmacodynamic effects of different drugs and their combinations as well as to facilitate mechanistic exploration.129

Conclusion

The HCC practice guidelines have evolved rapidly in the past 2–3 years to accommodate the advances in systemic therapy, to recognize the variations of practice patterns around the world, and to facilitate multi-disciplinary management, particularly in patients with intermediate-stage HCC. The advances of systemic therapy have led to a bloom of studies evaluating combination of different LDT and ICI-based systemic therapy in intermediate-stage HCC. Critical appraisal of the multiple confounding factors inherent in this patient population is vital for successful design and conduct of clinical trials as well as interpretation of trial results, which may be available in the next few years. A neoadjuvant approach provides a chance of cure and unique opportunities to collect bio-samples for mechanistic research and biomarker development. Extensive analytic and clinical validation of the novel biomarkers must be done to realize their potential in clinical use.

Abbreviations

AASLD, American Association for the Study of Liver Diseases; APASL, Asian Pacific Association for the Study of the Liver; BCLC, Barcelona Clinic Liver Cancer; EASL, European Association for the Study of the Liver; EBRT, external beam radiation therapy; ESMO, European Society for Medical Oncology; HAIC, hepatic arterial infusion chemotherapy; HCC, hepatocellular carcinoma; HIF-1α, hypoxia inducible factor-1α; HKLCS, Hong Kong Liver Cancer staging; ICI, immune checkpoint inhibitor; JSH, Japan Society of Hepatology; KLCSG, Korean Liver Cancer Study Group; LDT, liver-directed therapy; LT, liver transplantation; ORR, objective response rate; OS, overall survival; PFS, progression-free survival; Q2W, every 2 weeks; Q3W, every 3 weeks; Q4W, every 4 weeks; Q6W, every 6 weeks; TACE, transarterial chemoembolization; TARE, transarterial radioembolization; TILs, tumor-infiltrating lymphocytes; TLCA, Taiwan Liver Cancer Association; TMB, tumor mutation burden; TTP, time to progression; TTTP, time to TACE progression; US FDA, US Food and Drug Administration; VEGF, vascular endothelial growth factor; Y-90 SIRT, yttrium-90 selective internal radiation therapy.

Funding

This study is funded by Kaohsiung Medical University (grant number: KMU-TC109A04-0) and Ministry of Health and Welfare, Taiwan (grant number: MOHW111-TDU-B-221-014007).

Disclosure

Dr Yung-Yeh Su reports grants from National Health Research Institutes, during the conduct of the study; Dr Chiun Hsu reports grants, honorarium from Bristol-Myers Squibb/ONO, grants, honorarium from IPSEN, grants, honorarium from Roche, honorarium from Astra-Zeneca, honorarium from Bayer, honorarium from Eisai, honorarium from MSD, grants from Kintor, during the conduct of the study. Dr Li-Tzong Chen reports grants from Kaohsiung Medical University, during the conduct of the study. The authors report no other conflicts of interest in this work.

References

1. Finn RS, Qin S, Ikeda M, et al. Atezolizumab plus Bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med. 2020;382(20):1894–1905. doi:10.1056/NEJMoa1915745

2. Ren Z, Xu J, Bai Y, et al. Sintilimab plus a bevacizumab biosimilar (IBI305) versus sorafenib in unresectable hepatocellular carcinoma (ORIENT-32): a randomised, open-label, phase 2–3 study. Lancet Oncol. 2021;22(7):977–990. doi:10.1016/S1470-2045(21)00252-7

3. Abou-Alfa GK, Chan SL, Kudo M, et al. Phase 3 randomized, open-label, multicenter study of tremelimumab (T) and durvalumab (D) as first-line therapy in patients (pts) with unresectable hepatocellular carcinoma (uHCC): HIMALAYA. J Clin Oncol. 2022;40(suppl4):379. doi:10.1200/JCO.2022.40.4_suppl.379

4. Kelley RK, Yau T, Cheng AL, et al. VP10-2021: cabozantinib (C) plus atezolizumab (A) versus sorafenib (S) as first-line systemic treatment for advanced hepatocellular carcinoma (aHCC): results from the randomized phase III COSMIC-312 trial. Ann Oncol. 2022;33(1):114–116. doi:10.1016/j.annonc.2021.10.008

5. Bruix J, da Fonseca LG, Reig M. Insights into the success and failure of systemic therapy for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2019;16(10):617–630. doi:10.1038/s41575-019-0179-x

6. Llovet JM, Villanueva A, Marrero JA, et al. Trial design and endpoints in hepatocellular carcinoma: AASLD consensus conference. Hepatology. 2021;73(suppl 1):158–191. doi:10.1002/hep.31327

7. Singal AG, Hoshida Y, Pinato DJ, et al. International Liver Cancer Association (ILCA) white paper on biomarker development for hepatocellular carcinoma. Gastroenterology. 2021;160(7):2572–2584. doi:10.1053/j.gastro.2021.01.233

8. Foerster F, Galle PR. The current landscape of clinical trials for systemic treatment of HCC. Cancers. 2021;13(8). doi:10.3390/cancers13081962

9. Dong Y, Liu TH, Yau T, Hsu C. Novel systemic therapy for hepatocellular carcinoma. Hepatol Int. 2020;14(5):638–651. doi:10.1007/s12072-020-10073-7

10. Chen LT, Martinelli E, Cheng AL, et al. Pan-Asian adapted ESMO clinical practice guidelines for the management of patients with intermediate and advanced/relapsed hepatocellular carcinoma: a TOS-ESMO initiative endorsed by CSCO, ISMPO, JSMO, KSMO, MOS and SSO. Ann Oncol. 2020;31(3):334–351. doi:10.1016/j.annonc.2019.12.001

11. Kudo M, Han KH, Ye SL, et al. A changing paradigm for the treatment of intermediate-stage hepatocellular carcinoma: Asia-Pacific primary liver cancer expert consensus statements. Liver Cancer. 2020;9(3):245–260. doi:10.1159/000507370

12. Greten TF, Abou-Alfa GK, Cheng AL, et al. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of hepatocellular carcinoma. J Immunother Cancer. 2021;9(9):e002794. doi:10.1136/jitc-2021-002794

13. Reig M, Forner A, Rimola J, et al. BCLC strategy for prognosis prediction and treatment recommendation: the 2022 update. J Hepatol. 2022;76(3):681–693. doi:10.1016/j.jhep.2021.11.018

14. Vogel A, Martinelli E; ESMO Guidelines Committee. Updated treatment recommendations for hepatocellular carcinoma (HCC) from the ESMO clinical practice guidelines. Ann Oncol. 2021;32(6):801–805. doi:10.1016/j.annonc.2021.02.014

15. Kudo M, Imanaka K, Chida N, et al. Phase III study of sorafenib after transarterial chemoembolisation in Japanese and Korean patients with unresectable hepatocellular carcinoma. Eur J Cancer. 2011;47(14):2117–2127. doi:10.1016/j.ejca.2011.05.007

16. Kudo M, Han G, Finn RS, et al. Brivanib as adjuvant therapy to transarterial chemoembolization in patients with hepatocellular carcinoma: a randomized phase III trial. Hepatology. 2014;60(5):1697–1707. doi:10.1002/hep.27290

17. Lencioni R, Llovet JM, Han G, et al. Sorafenib or placebo plus TACE with doxorubicin-eluting beads for intermediate stage HCC: the SPACE trial. J Hepatol. 2016;64(5):1090–1098. doi:10.1016/j.jhep.2016.01.012

18. Kudo M, Cheng AL, Park JW, et al. Orantinib versus placebo combined with transcatheter arterial chemoembolisation in patients with unresectable hepatocellular carcinoma (ORIENTAL): a randomised, double-blind, placebo-controlled, multicentre, phase 3 study. Lancet Gastroenterol Hepatol. 2018;3(1):37–46. doi:10.1016/S2468-1253(17)30290-X

19. Meyer T, Fox R, Ma YT, et al. Sorafenib in combination with transarterial chemoembolisation in patients with unresectable hepatocellular carcinoma (TACE 2): a randomised placebo-controlled, double-blind, phase 3 trial. Lancet Gastroenterol Hepatol. 2017;2(8):565–575. doi:10.1016/S2468-1253(17)30156-5

20. Kudo M, Ueshima K, Ikeda M, et al. Randomised, multicentre prospective trial of transarterial chemoembolisation (TACE) plus sorafenib as compared with TACE alone in patients with hepatocellular carcinoma: TACTICS trial. Gut. 2020;69(8):1492–1501. doi:10.1136/gutjnl-2019-318934

21. Bolondi L, Burroughs A, Dufour JF, et al. Heterogeneity of patients with intermediate (BCLC B) hepatocellular carcinoma: proposal for a subclassification to facilitate treatment decisions. Semin Liver Dis. 2012;32(4):348–359. doi:10.1055/s-0032-1329906

22. Kudo M, Arizumi T. Transarterial chemoembolization in combination with a molecular targeted agent: lessons learned from negative trials (Post-TACE, BRISK-TA, SPACE, ORIENTAL, and TACE-2). Oncology. 2017;93(Suppl 1):127–134. doi:10.1159/000481243

23. Bruix J. Endpoints in clinical trials for liver cancer and their value in evidence-based clinical decision making: an unresolved Gordian knot. J Hepatol. 2021;74(6):1483–1488. doi:10.1016/j.jhep.2021.01.033

24. Cheng AL, Hsu C, Chan SL, Choo SP, Kudo M. Challenges of combination therapy with immune checkpoint inhibitors for hepatocellular carcinoma. J Hepatol. 2020;72(2):307–319. doi:10.1016/j.jhep.2019.09.025

25. Marrero JA, Kulik LM, Sirlin CB, et al. Diagnosis, staging, and management of hepatocellular carcinoma: 2018 practice guidance by the American Association for the Study of Liver Diseases. Hepatology. 2018;68(2):723–750. doi:10.1002/hep.29913

26. Kudo M, Arizumi T, Ueshima K, Sakurai T, Kitano M, Nishida N. Subclassification of BCLC B stage hepatocellular carcinoma and treatment strategies: proposal of modified Bolondi’s subclassification (Kinki Criteria). Dig Dis. 2015;33(6):751–758. doi:10.1159/000439290

27. Hucke F, Pinter M, Graziadei I, et al. How to STATE suitability and START transarterial chemoembolization in patients with intermediate stage hepatocellular carcinoma. J Hepatol. 2014;61(6):1287–1296. doi:10.1016/j.jhep.2014.07.002

28. Mazzaferro V, Llovet JM, Miceli R, et al. Predicting survival after liver transplantation in patients with hepatocellular carcinoma beyond the Milan criteria: a retrospective, exploratory analysis. Lancet Oncol. 2009;10(1):35–43. doi:10.1016/S1470-2045(08)70284-5

29. Ho MC, Hasegawa K, Chen XP, et al. Surgery for intermediate and advanced hepatocellular carcinoma: a consensus report from the 5th Asia-Pacific Primary Liver Cancer Expert Meeting (APPLE 2014). Liver Cancer. 2016;5(4):245–256.

30. Nouso K, Kariyama K, Nakamura S, et al. Application of radiofrequency ablation for the treatment of intermediate-stage hepatocellular carcinoma. J Gastroenterol Hepatol. 2017;32(3):695–700. doi:10.1111/jgh.13586

31. European Association for the Study of the Liver. EASL clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2018;69(1):182–236. doi:10.1016/j.jhep.2018.03.019

32. Galle PR, Tovoli F, Foerster F, Worns MA, Cucchetti A, Bolondi L. The treatment of intermediate stage tumours beyond TACE: from surgery to systemic therapy. J Hepatol. 2017;67(1):173–183. doi:10.1016/j.jhep.2017.03.007

33. Kim JH, Shim JH, Lee HC, et al. New intermediate-stage subclassification for patients with hepatocellular carcinoma treated with transarterial chemoembolization. Liver Int. 2017;37(12):1861–1868. doi:10.1111/liv.13487

34. Koroki K, Ogasawara S, Ooka Y, et al. Analyses of intermediate-stage hepatocellular carcinoma patients receiving transarterial chemoembolization prior to designing clinical trials. Liver Cancer. 2020;9(5):596–612. doi:10.1159/000508809

35. Hung YW, Lee IC, Chi CT, et al. Redefining tumor burden in patients with intermediate-stage hepatocellular carcinoma: the seven-eleven criteria. Liver Cancer. 2021;10(6):629–640. doi:10.1159/000517393

36. Lin YJ, Lin CN, Sedghi T, et al. Treatment patterns and survival in hepatocellular carcinoma in the United States and Taiwan. PLoS One. 2020;15(10):e0240542. doi:10.1371/journal.pone.0240542

37. Park JW, Chen M, Colombo M, et al. Global patterns of hepatocellular carcinoma management from diagnosis to death: the BRIDGE Study. Liver Int. 2015;35(9):2155–2166. doi:10.1111/liv.12818

38. Demirtas CO, D’Alessio A, Rimassa L, Sharma R, Pinato DJ. ALBI grade: evidence for an improved model for liver functional estimation in patients with hepatocellular carcinoma. JHEP Rep. 2021;3(5):100347. doi:10.1016/j.jhepr.2021.100347

39. Hucke F, Sieghart W, Pinter M, et al. The ART-strategy: sequential assessment of the ART score predicts outcome of patients with hepatocellular carcinoma re-treated with TACE. J Hepatol. 2014;60(1):118–126. doi:10.1016/j.jhep.2013.08.022

40. Kadalayil L, Benini R, Pallan L, et al. A simple prognostic scoring system for patients receiving transarterial embolisation for hepatocellular cancer. Ann Oncol. 2013;24(10):2565–2570. doi:10.1093/annonc/mdt247

41. Llovet JM, Bruix J. Systematic review of randomized trials for unresectable hepatocellular carcinoma: chemoembolization improves survival. Hepatology. 2003;37(2):429–442. doi:10.1053/jhep.2003.50047

42. Camma C, Schepis F, Orlando A, et al. Transarterial chemoembolization for unresectable hepatocellular carcinoma: meta-analysis of randomized controlled trials. Radiology. 2002;224(1):47–54. doi:10.1148/radiol.2241011262

43. Sieghart W, Hucke F, Peck-Radosavljevic M. Transarterial chemoembolization: modalities, indication, and patient selection. J Hepatol. 2015;62(5):1187–1195. doi:10.1016/j.jhep.2015.02.010

44. Muller L, Stoehr F, Mahringer-Kunz A, Hahn F, Weinmann A, Kloeckner R. Current strategies to identify patients that will benefit from TACE treatment and future directions a practical step-by-step guide. J Hepatocell Carcinoma. 2021;8:403–419. doi:10.2147/JHC.S285735

45. Horikawa M, Miyayama S, Irie T, Kaji T, Arai Y. Development of conventional transarterial chemoembolization for hepatocellular carcinomas in Japan: historical, strategic, and technical review. AJR Am J Roentgenol. 2015;205(4):764–773. doi:10.2214/AJR.15.14825

46. Zhou J, Sun H, Wang Z, et al. Guidelines for the diagnosis and treatment of hepatocellular carcinoma (2019 edition). Liver Cancer. 2020;9(6):682–720. doi:10.1159/000509424

47. Korean Liver Cancer Association, National Cancer Center. 2018 Korean Liver Cancer Association-National Cancer Center Korea practice guidelines for the management of hepatocellular carcinoma. Gut Liver. 2019;13(3):227–299. doi:10.5009/gnl19024

48. Kudo M, Matsui O, Izumi N, et al. JSH Consensus-based clinical practice guidelines for the management of hepatocellular carcinoma: 2014 update by the liver cancer study group of Japan. Liver Cancer. 2014;3(3–4):458–468. doi:10.1159/000343875

49. Shao YY, Wang SY, Lin SM; Diagnosis Group, Systemic Therapy Group. Management consensus guideline for hepatocellular carcinoma: 2020 update on surveillance, diagnosis, and systemic treatment by the Taiwan Liver Cancer Association and the Gastroenterological Society of Taiwan. J Formos Med Assoc. 2021;120(4):1051–1060. doi:10.1016/j.jfma.2020.10.031

50. Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet. 2012;379(9822):1245–1255. doi:10.1016/S0140-6736(11)61347-0

51. Yau T, Tang VY, Yao TJ, Fan ST, Lo CM, Poon RT. Development of Hong Kong Liver Cancer staging system with treatment stratification for patients with hepatocellular carcinoma. Gastroenterology. 2014;146(7):1691–1700 e1693. doi:10.1053/j.gastro.2014.02.032

52. Omata M, Cheng AL, Kokudo N, et al. Asia-Pacific clinical practice guidelines on the management of hepatocellular carcinoma: a 2017 update. Hepatol Int. 2017;11(4):317–370. doi:10.1007/s12072-017-9799-9

53. Li QJ, He MK, Chen HW, et al. Hepatic arterial infusion of oxaliplatin, fluorouracil, and leucovorin versus transarterial chemoembolization for large hepatocellular carcinoma: a randomized phase III trial. J Clin Oncol. 2022;40(2):150–160. doi:10.1200/JCO.21.00608

54. Yoon SM, Ryoo BY, Lee SJ, et al. Efficacy and safety of transarterial chemoembolization plus external beam radiotherapy vs sorafenib in hepatocellular carcinoma with macroscopic vascular invasion: a randomized clinical trial. JAMA Oncol. 2018;4(5):661–669. doi:10.1001/jamaoncol.2017.5847

55. He M, Li Q, Zou R, et al. Sorafenib plus hepatic arterial infusion of oxaliplatin, fluorouracil, and leucovorin vs sorafenib alone for hepatocellular carcinoma with portal vein invasion: a randomized clinical trial. JAMA Oncol. 2019;5(7):953–960. doi:10.1001/jamaoncol.2019.0250

56. Reig M, Darnell A, Forner A, Rimola J, Ayuso C, Bruix J. Systemic therapy for hepatocellular carcinoma: the issue of treatment stage migration and registration of progression using the BCLC-refined RECIST. Semin Liver Dis. 2014;34(4):444–455. doi:10.1055/s-0034-1394143

57. Yen C, Sharma R, Rimassa L, et al. Treatment stage migration maximizes survival outcomes in patients with hepatocellular carcinoma treated with sorafenib: an observational study. Liver Cancer. 2017;6(4):313–324. doi:10.1159/000480441

58. Chen EY, Haslam A, Prasad V. FDA acceptance of surrogate end points for cancer drug approval: 1992–2019. JAMA Intern Med. 2020;180(6):912–914. doi:10.1001/jamainternmed.2020.1097

59. Schnog JB, Samson MJ, Gans ROB, Duits AJ. An urgent call to raise the bar in oncology. Br J Cancer. 2021;125(11):1477–1485. doi:10.1038/s41416-021-01495-7

60. Llovet JM, Montal R, Villanueva A. Randomized trials and endpoints in advanced HCC: role of PFS as a surrogate of survival. J Hepatol. 2019;70(6):1262–1277. doi:10.1016/j.jhep.2019.01.028

61. Finn RS. Progression-free survival: starting point or endpoint in advanced HCC trial design? J Hepatol. 2019;70(6):1062–1064. doi:10.1016/j.jhep.2019.03.002

62. Lin Y, Zhu M, Su Z. The pursuit of balance: an overview of covariate-adaptive randomization techniques in clinical trials. Contemp Clin Trials. 2015;45(Pt A):21–25. doi:10.1016/j.cct.2015.07.011

63. Kaiser LD. Inefficiency of randomization methods that balance on stratum margins and improvements with permuted blocks and a sequential method. Stat Med. 2012;31(16):1699–1706. doi:10.1002/sim.5345

64. Lin Y, Su Z. A hybrid approach to achieving both marginal and conditional balances for stratification variables in sequential clinical trials. Pharm Stat. 2013;12(5):275–281. doi:10.1002/pst.1587

65. Dancey JE, Dodd LE, Ford R, et al. Recommendations for the assessment of progression in randomised cancer treatment trials. Eur J Cancer. 2009;45(2):281–289. doi:10.1016/j.ejca.2008.10.042

66. Singh P, Toom S, Avula A, Kumar V, Rahma OE. The immune modulation effect of locoregional therapies and its potential synergy with immunotherapy in hepatocellular carcinoma. J Hepatocell Carcinoma. 2020;7:11–17. doi:10.2147/JHC.S187121

67. Chiu DK, Tse AP, Xu IM, et al. Hypoxia inducible factor HIF-1 promotes myeloid-derived suppressor cells accumulation through ENTPD2/CD39L1 in hepatocellular carcinoma. Nat Commun. 2017;8(1):517. doi:10.1038/s41467-017-00530-7

68. Yuen VW, Wong CC. Hypoxia-inducible factors and innate immunity in liver cancer. J Clin Invest. 2020;130(10):5052–5062. doi:10.1172/JCI137553

69. Craciun L, de Wind R, Demetter P, et al. Retrospective analysis of the immunogenic effects of intra-arterial locoregional therapies in hepatocellular carcinoma: a rationale for combining selective internal radiation therapy (SIRT) and immunotherapy. BMC Cancer. 2020;20(1):135. doi:10.1186/s12885-020-6613-1

70. Salem R, Gordon AC, Mouli S, et al. Y90 radioembolization significantly prolongs time to progression compared with chemoembolization in patients with hepatocellular carcinoma. Gastroenterology. 2016;151(6):1155–1163 e1152. doi:10.1053/j.gastro.2016.08.029

71. El Fouly A, Ertle J, El Dorry A, et al. In intermediate stage hepatocellular carcinoma: radioembolization with yttrium 90 or chemoembolization? Liver Int. 2015;35(2):627–635. doi:10.1111/liv.12637

72. Salem R, Gabr A, Riaz A, et al. Institutional decision to adopt Y90 as primary treatment for hepatocellular carcinoma informed by a 1000-patient 15-year experience. Hepatology. 2018;68(4):1429–1440. doi:10.1002/hep.29691

73. Lee JJX, Tan SH, Hennedige TP, et al. 947P Updated survival and secondary safety and efficacy analyses from CA 209-678: a phase II open-label single-centre study of Y90-radioembolisation (Y90) in combination with nivolumab in Asian patients (pts) with advanced hepatocellular carcinoma (aHCC). Ann Oncol. 2021;32:S825. doi:10.1016/j.annonc.2021.08.167

74. Takeda A, Sanuki N, Tsurugai Y, et al. Phase 2 study of stereotactic body radiotherapy and optional transarterial chemoembolization for solitary hepatocellular carcinoma not amenable to resection and radiofrequency ablation. Cancer. 2016;122(13):2041–2049. doi:10.1002/cncr.30008

75. Kim JW, Kim DY, Han KH, Seong J. Phase I/II trial of helical IMRT-based stereotactic body radiotherapy for hepatocellular carcinoma. Dig Liver Dis. 2019;51(3):445–451. doi:10.1016/j.dld.2018.11.004

76. Sapir E, Tao Y, Schipper MJ, et al. Stereotactic body radiation therapy as an alternative to transarterial chemoembolization for hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2018;100(1):122–130. doi:10.1016/j.ijrobp.2017.09.001

77. Shen PC, Chang WC, Lo CH, et al. Comparison of stereotactic body radiation therapy and transarterial chemoembolization for unresectable medium-sized hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2019;105(2):307–318. doi:10.1016/j.ijrobp.2019.05.066

78. Wu M, Liu J, Wu S, et al. Systemic immune activation and responses of irradiation to different metastatic sites combined with immunotherapy in advanced non-small cell lung cancer. Front Immunol. 2021;12:803247. doi:10.3389/fimmu.2021.803247

79. Chiang CL, Chiu KW, Lee FA, Kong FS, Chan AC. Combined stereotactic body radiotherapy and immunotherapy versus transarterial chemoembolization in locally advanced hepatocellular carcinoma: a propensity score matching analysis. Front Oncol. 2021;11:798832. doi:10.3389/fonc.2021.798832

80. Hecht M, Eckstein M, Rutzner S, et al. Induction chemoimmunotherapy followed by CD8+ immune cell-based patient selection for chemotherapy-free radioimmunotherapy in locally advanced head and neck cancer. J Immunother Cancer. 2022;10(1):e003747. doi:10.1136/jitc-2021-003747

81. Perez-Romasanta LA, Gonzalez-Del Portillo E, Rodriguez-Gutierrez A, Matias-Perez A. Stereotactic radiotherapy for hepatocellular carcinoma, radiosensitization strategies and radiation-immunotherapy combination. Cancers. 2021;13(2):192. doi:10.3390/cancers13020192

82. Kudo M. Treatment of advanced hepatocellular carcinoma with emphasis on hepatic arterial infusion chemotherapy and molecular targeted therapy. Liver Cancer. 2012;1(2):62–70. doi:10.1159/000342402

83. Nouso K, Miyahara K, Uchida D, et al. Effect of hepatic arterial infusion chemotherapy of 5-fluorouracil and cisplatin for advanced hepatocellular carcinoma in the Nationwide survey of primary liver cancer in Japan. Br J Cancer. 2013;109(7):1904–1907. doi:10.1038/bjc.2013.542

84. Sumie S, Yamashita F, Ando E, et al. Interventional radiology for advanced hepatocellular carcinoma: comparison of hepatic artery infusion chemotherapy and transcatheter arterial lipiodol chemoembolization. AJR Am J Roentgenol. 2003;181(5):1327–1334. doi:10.2214/ajr.181.5.1811327

85. Guo W, Gao J, Zhuang W, Wu Z, Li B, Chen S. Efficacy and safety of hepatic arterial infusion chemotherapy combined with transarterial embolization for unresectable hepatocellular carcinoma: a propensity score-matching cohort study. JGH Open. 2020;4(3):477–483. doi:10.1002/jgh3.12285

86. Hu J, Bao Q, Cao G, et al. Hepatic arterial infusion chemotherapy using oxaliplatin plus 5-fluorouracil versus transarterial chemoembolization/embolization for the treatment of advanced hepatocellular carcinoma with major portal vein tumor thrombosis. Cardiovasc Intervent Radiol. 2020;43(7):996–1005. doi:10.1007/s00270-019-02406-3

87. Lyu N, Wang X, Li JB, et al. Arterial chemotherapy of oxaliplatin plus fluorouracil versus sorafenib in advanced hepatocellular carcinoma: a biomolecular exploratory, randomized, Phase III trial (FOHAIC-1). J Clin Oncol. 2022;40(5):468–480. doi:10.1200/JCO.21.01963

88. Schmitt M, Horbach A, Kubitz R, Frilling A, Haussinger D. Disruption of hepatocellular tight junctions by vascular endothelial growth factor (VEGF): a novel mechanism for tumor invasion. J Hepatol. 2004;41(2):274–283. doi:10.1016/j.jhep.2004.04.035

89. Hsieh MY, Lin ZY, Chuang WL. Serial serum VEGF-A, angiopoietin-2, and endostatin measurements in cirrhotic patients with hepatocellular carcinoma treated by transcatheter arterial chemoembolization. Kaohsiung J Med Sci. 2011;27(8):314–322. doi:10.1016/j.kjms.2011.03.008

90. Jia ZZ, Jiang GM, Feng YL. Serum HIF-1α and VEGF levels pre- and post-TACE in patients with primary liver cancer. Chin Med Sci J. 2011;26(3):158–162. doi:10.1016/S1001-9294(11)60041-2

91. Sangro B, Kudo M, Qin S, et al. P-347 A phase 3, randomized, double-blind, placebo-controlled study of transarterial chemoembolization combined with durvalumab or durvalumab plus bevacizumab therapy in patients with locoregional hepatocellular carcinoma: EMERALD-1. Ann Oncol. 2020;31:S202–S203. doi:10.1016/j.annonc.2020.04.429

92. Ogasawara S, Llovet J, El-Khoueiry A, et al. P-107 LEAP-012: a randomized, double-blind, phase 3 study of pembrolizumab plus lenvatinib in combination with transarterial chemoembolization (TACE) in patients with intermediate-stage hepatocellular carcinoma not amenable to curative treatment. Ann Oncol. 2020;31:S124–S125. doi:10.1016/j.annonc.2020.04.189

93. Sangro B, Harding JJ, Johnson M, et al. A Phase III, double-blind, randomized study of nivolumab (NIVO) and ipilimumab (IPI), nivo monotherapy or placebo plus transarterial chemoembolization (TACE) in patients with intermediate-stage hepatocellular carcinoma (HCC). J Clinl Oncol. 2021;39(suppl 3):TPS349. doi:10.1200/JCO.2021.39.3_suppl.TPS349

94. Kudo M, Guo Y, Hua Y, et al. TALENTACE: a phase III, open-label, randomized study of on-demand transarterial chemoembolization combined with atezolizumab + bevacizumab or on-demand transarterial chemoembolization alone in patients with untreated hepatocellular carcinoma. J Clinl Oncol. 2022;40(suppl 4):TPS487. doi:10.1200/JCO.2022.40.4_suppl.TPS487

95. Kudo M, Ueshima K, Ikeda M, et al. Final Results of TACTICS: a randomized, prospective trial comparing transarterial chemoembolization plus sorafenib to transarterial chemoembolization alone in patients with unresectable hepatocellular carcinoma. Liver Cancer. 2022. doi:10.1159/000522547

96. Johnson PJ, Berhane S, Kagebayashi C, et al. Assessment of liver function in patients with hepatocellular carcinoma: a new evidence-based approach-the ALBI grade. J Clin Oncol. 2015;33(6):550–558. doi:10.1200/JCO.2014.57.9151

97. Chi CT, Lee IC, Lee RC, et al. Effect of transarterial chemoembolization on ALBI grade in intermediate-stage hepatocellular carcinoma: criteria for unsuitable cases selection. Cancers. 2021;13(17):4325. doi:10.3390/cancers13174325

98. Pinato DJ, Arizumi T, Allara E, et al. Validation of the hepatoma arterial embolization prognostic score in European and Asian populations and proposed modification. Clin Gastroenterol Hepatol. 2015;13(6):1204–1208 e1202. doi:10.1016/j.cgh.2014.11.037

99. Waked I, Berhane S, Toyoda H, et al. Transarterial chemo-embolisation of hepatocellular carcinoma: impact of liver function and vascular invasion. Br J Cancer. 2017;116(4):448–454. doi:10.1038/bjc.2016.423

100. Hiraoka A, Michitaka K, Kumada T, et al. Prediction of prognosis of intermediate-stage HCC patients: validation of the tumor marker score in a nationwide database in Japan. Liver Cancer. 2019;8(5):403–411. doi:10.1159/000495944

101. Bakos O, Lawson C, Rouleau S, Tai LH. Combining surgery and immunotherapy: turning an immunosuppressive effect into a therapeutic opportunity. J Immunother Cancer. 2018;6(1):86. doi:10.1186/s40425-018-0398-7

102. Pinato DJ, Fessas P, Sapisochin G, Marron TU. Perspectives on the neoadjuvant use of immunotherapy in hepatocellular carcinoma. Hepatology. 2021;74(1):483–490. doi:10.1002/hep.31697

103. Su YY, Li CC, Lin YJ, Hsu C. Adjuvant versus neoadjuvant immunotherapy for hepatocellular carcinoma: clinical and immunologic perspectives. Semin Liver Dis. 2021;41(3):263–276. doi:10.1055/s-0041-1730949

104. Kim CG, Kim C, Yoon SE, et al. Hyperprogressive disease during PD-1 blockade in patients with advanced hepatocellular carcinoma. J Hepatol. 2021;74(2):350–359. doi:10.1016/j.jhep.2020.08.010

105. De Toni EN. Immune checkpoint inhibitors: use them early, combined and instead of TACE? Gut. 2020;69(10):1887–1888. doi:10.1136/gutjnl-2019-319658

106. Ueshima K, Kudo M, Yamanaka T, et al. REPLACEMENT trial in progress: combination therapy with atezolizumab plus bevacizumab for TACE unsuitable patients with beyond up-to-seven criteria in intermediate stage hepatocellular carcinoma: a phase II study. J Clinl Oncol. 2021;39(suppl 15):TPS4162. doi:10.1200/JCO.2021.39.15_suppl.TPS4162

107. Kaseb AO, Cao HST, Mohamed YI, et al. Final results of a randomized, open label, perioperative phase II study evaluating nivolumab alone or nivolumab plus ipilimumab in patients with resectable HCC. J Clinl Oncol. 2020;38(suppl 15):4599. doi:10.1200/JCO.2020.38.15_suppl.4599

108. Su Y, Lin Y, Hsiao C, et al. P-124 Nivolumab plus ipilimumab as neoadjuvant therapy for potentially resectable hepatocellular carcinoma. Ann Oncol. 2021;32:S141. doi:10.1016/j.annonc.2021.05.179

109. Foerster F, Kloeckner R, Reig M, et al. ABC-HCC: a phase IIIb, randomized, multicenter, open-label trial of atezolizumab plus bevacizumab versus transarterial chemoembolization (TACE) in intermediate-stage hepatocellular carcinoma. J Clinl Oncol. 2022;40(suppl 4):TPS498. doi:10.1200/JCO.2022.40.4_suppl.TPS498

110. Harding JJ, Yarmohammadi H, Reiss KA, et al. Nivolumab (NIVO) and drug eluting bead transarterial chemoembolization (deb-TACE): updated results from an ongoing Phase 1 study of patients (pts) with liver limited hepatocellular carcinoma (HCC). J Clinl Oncol. 2022;40(suppl 4):437. doi:10.1200/JCO.2022.40.4_suppl.437

111. Vogel A, Saborowski A, Hinrichs J, et al. LBA37 - IMMUTACE: a biomarker-orientated, multi center phase II AIO study of transarterial chemoembolization (TACE) in combination with nivolumab performed for intermediate stage hepatocellular carcinoma (HCC). Ann Oncol. 2021;32:S1283–S1346. doi:10.1016/j.annonc.2021.08.2114

112. Zhang X, Zhu X, Liu C, et al. The safety and efficacy of transarterial chemoembolization (TACE) + lenvatinib + programmed cell death protein 1 (PD-1) antibody of advanced unresectable hepatocellular carcinoma. J Clinl Oncol. 2022;40(suppl 4):453. doi:10.1200/JCO.2022.40.4_suppl.453

113. Seymour L, Bogaerts J, Perrone A, et al. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017;18(3):e143–e152. doi:10.1016/S1470-2045(17)30074-8

115. Zhu AX, Kang YK, Yen CJ, et al. Ramucirumab after sorafenib in patients with advanced hepatocellular carcinoma and increased alpha-fetoprotein concentrations (REACH-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2019;20(2):282–296. doi:10.1016/S1470-2045(18)30937-9

116. Kudo M, Ueshima K, Chan S, et al. Lenvatinib as an initial treatment in patients with intermediate-stage hepatocellular carcinoma beyond up-to-seven criteria and child-pugh a liver function: a proof-of-concept study. Cancers. 2019;11(8):1084. doi:10.3390/cancers11081084

117. Kudo M, Finn RS, Morimoto M, et al. Ramucirumab for patients with intermediate-stage hepatocellular carcinoma and elevated alpha-fetoprotein: pooled results from two phase 3 studies (REACH and REACH-2). Liver Cancer. 2021;10(5):451–460. doi:10.1159/000516605

118. Hsu CY, Hsia CY, Huang YH, et al. Comparison of surgical resection and transarterial chemoembolization for hepatocellular carcinoma beyond the Milan criteria: a propensity score analysis. Ann Surg Oncol. 2012;19(3):842–849. doi:10.1245/s10434-011-2060-1

119. Hyun MH, Lee YS, Kim JH, et al. Hepatic resection compared to chemoembolization in intermediate- to advanced-stage hepatocellular carcinoma: a meta-analysis of high-quality studies. Hepatology. 2018;68(3):977–993. doi:10.1002/hep.29883

120. Chen X, Lai L, Ye J, Li L. Downstaging therapies for unresectable hepatocellular carcinoma prior to hepatic resection: a systematic review and meta-analysis. Front Oncol. 2021;11:740762. doi:10.3389/fonc.2021.740762

121. Kudo M. A novel treatment strategy for patients with intermediate-stage HCC who are not suitable for TACE: upfront systemic therapy followed by curative conversion. Liver Cancer. 2021;10(6):539–544. doi:10.1159/000519749

122. Kaseb AO, Hasanov E, Cao HST, et al. Perioperative nivolumab monotherapy versus nivolumab plus ipilimumab in resectable hepatocellular carcinoma: a randomised, open-label, phase 2 trial. Lancet Gastroenterol Hepatol. 2022;7(3):208–218. doi:10.1016/S2468-1253(21)00427-1

123. Yau T, Kang YK, Kim TY, et al. Efficacy and safety of nivolumab plus ipilimumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib: the checkmate 040 randomized clinical trial. JAMA Oncol. 2020;6(11):e204564. doi:10.1001/jamaoncol.2020.4564

124. Ho WJ, Zhu Q, Durham J, et al. Neoadjuvant cabozantinib and nivolumab converts locally advanced hcc into resectable disease with enhanced antitumor immunity. Nat Cancer. 2021;2(9):891–903. doi:10.1038/s43018-021-00234-4

125. Thommen DS, Schumacher TN. T cell dysfunction in cancer. Cancer Cell. 2018;33(4):547–562. doi:10.1016/j.ccell.2018.03.012

126. Zhu AX, Guan Y, Abbas AR, et al. Abstract CT044: genomic correlates of clinical benefits from atezolizumab combined with bevacizumab vs. atezolizumab alone in patients with advanced hepatocellular carcinoma (HCC). Cancer Res. 2020;80(suppl16):CT044.

127. Zheng Y, Wang T, Tu X, et al. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J Immunother Cancer. 2019;7(1):193. doi:10.1186/s40425-019-0650-9

128. Su YY, Lee WH, Wang JH, et al. Potential roles of gut microbiome in patients with hepatocellular carcinoma treated with immune checkpoint inhibitors. Paper presented at: Annual Meeting of Japanese Society of Medical Oncology (JSMO). Kyoto; 2022.

129. An HJ, Chon HJ, Kim C. Peripheral blood-based biomarkers for immune checkpoint inhibitors. Int J Mol Sci. 2021;22(17):9414. doi:10.3390/ijms22179414

Creative Commons License © 2022 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.