Distinct Characteristics and Changes in Liver Function of Patients with Hepatocellular Carcinoma Treated with Atezolizumab Plus Bevacizumab for More Than 1 Year

Article information

Cancer Res Treat. 2024;.crt.2024.237
Publication date (electronic) : 2024 May 27
doi : https://doi.org/10.4143/crt.2024.237
1CHA University School of Medicine, Seongnam, Korea
2Division of Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
3Division of Oncology, Department of Internal Medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
4Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
5Department of Radiology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
Correspondence: Hong Jae Chon, Division of Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam 13496, Korea Tel: 82-31-780-7590 Fax: 82-31-780-3929 E-mail: minidoctor@cha.ac.kr
Co-correspondence: Chansik An, Department of Radiology, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam 13496, Korea Tel: 82-31-780-5386 Fax: 82-31-780-5381 E-mail: chansik79@chamc.co.kr
*Youngun Kim and Jung Sun Kim contributed equally to this work.
Received 2024 March 4; Accepted 2024 May 24.

Abstract

Purpose

Since 2020, atezolizumab plus bevacizumab (Ate/Bev) has been the standard first-line therapy for unresectable hepatocellular carcinoma (HCC), but long-term treatment studies are limited. This study evaluated the clinical characteristics and effects of Ate/Bev for over 1 year.

Materials and Methods

This study included patients with unresectable HCC treated with first-line Ate/Bev between May 2020 and April 2022. Those receiving Ate/Bev for 1 year or more were classified as the long-term treatment group.

Results

Of 246 patients, 69 (28.0%) were in the long-term treatment group, which comprised more proportions of intrahepatic tumor burden < 25%, Eastern Cooperative Oncology Group 0, and a lower proportion of portal vein tumor thrombosis than the short-term treatment group. The long-term treatment group had a higher incidence of atezolizumab-related thyroid dysfunction (31.9% vs. 10.7%, p < 0.001; median time to onset [mTTO], 2.8 months), dermatologic toxicity (29.0% vs. 14.7%, p=0.017; mTTO, 3.3 months), bevacizumab-related hypertension (44.9% vs. 22.0%, p=0.001; mTTO, 4.2 months), and proteinuria (69.6% vs. 38.4%, p < 0.001; mTTO, 6.8 months), compared to the short-term treatment group. Regarding liver function in the long-term treatment group, patients initially classified as Child-Pugh class A decreased from 87.0% to 75.4%, and albumin-bilirubin grade 1 decreased from 68.1% to 50.7% after 1 year of treatment.

Conclusion

The Ate/Bev long-term treatment group had a lower intrahepatic tumor burden, less portal vein tumor thrombosis, and better performance status and liver function at baseline. Atezolizumab-related immunological adverse events emerged relatively early in treatment compared to the bevacizumab-related. Additionally, some patients demonstrated liver function deterioration during long-term Ate/Bev treatment.

Introduction

The systemic treatment of unresectable hepatocellular carcinoma (HCC) has evolved with an understanding of the molecular pathogenesis of the disease and the development of various therapeutic agents [1-6]. The recent emergence of immune checkpoint inhibitors (ICIs) has dramatically changed the treatment landscape for unresectable HCC and opened up the possibility of long-term survival in certain patients [7-10]. In particular, atezolizumab plus bevacizumab (Ate/Bev), a combination of an ICI with a monoclonal antibody against vascular endothelial growth factor, has demonstrated significantly improved survival outcomes compared to those of sorafenib [11]. The Food and Drug Administration in May 2020 approved the use of Ate/Bev for patients with unresectable HCC who have not received prior systemic therapy, making Ate/Bev the standard of care in the first-line setting [12].

Since the approval of Ate/Bev, numerous studies have been conducted to identify real-world data and optimal biomarkers or clinical characteristics for predicting the treatment response [13-18]. However, studies on patients who receive long-term Ate/Bev treatment are lacking. Therefore, we aimed to evaluate the clinical characteristics of patients treated with Ate/Bev for more than 1 year, and the clinical impact, such as liver function decline, associated with long-term Ate/Bev treatment.

Materials and Methods

1. Study population

This retrospective study included patients with unresectable HCC who received Ate/Bev treatment at three tertiary cancer centers (CHA Bundang Medical Center, Ulsan University Hospital, and Haeundae Paik Hospital) in Korea between May 2020 and April 2022. Patients who received Ate/Bev in later lines, had no baseline clinical data, and had no target lesion according to Response Evaluation Criteria in Solid Tumors (RECIST) ver. 1.1, Child-Pugh class C, or Barcelona Clinical Liver Cancer (BCLC) stage A were excluded. Patients were subsequently divided into two groups based on the treatment duration: the long-term treatment group (≥ 1 year) and the short-term treatment group (< 1 year). This study was conducted in accordance with the ethical guidelines of the Declaration of Helsinki and approved by the participating hospitals’ institutional review boards (CHA Bundang Medical Center, CHA-2020-12-030; Ulsan University Hospital, 2020-12-006; Haeundae Paik Hospital, 2020-12-019-001). The need for informed consent in this study was waived, as Korean regulations do not require consent for retrospective analyses. Data were anonymized and deidentified prior to analysis.

2. Treatment and assessment

Patients were treated with Ate/Bev following the IMbrave150 trial (1,200 mg atezolizumab plus 15 mg/kg bevacizumab intravenously every 3 weeks) [11]. Dose interruptions or reductions were made at the discretion of the attending physicians. Ate/Bev was continued until disease progression or occurrence of unacceptable toxicity. Tumor response was assessed every 6 or 9 weeks according to RECIST ver. 1.1. The objective response rate (ORR) was defined as the combined proportion of patients with a complete response (CR) or partial response (PR). Overall survival (OS) was defined as the time from the date of Ate/Bev treatment initiation to death. Progression-free survival (PFS) was defined as the time from the date of Ate/Bev treatment initiation to disease progression or death from any cause, whichever occurred first. Data from patients free of disease progression, death, or loss to follow-up were excluded at the last follow-up visit.

Blood samples were collected at every visit, and the Child-Pugh score and albumin-bilirubin (ALBI) grade were monitored. Treatment-related adverse events (AEs) were evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) ver. 5.0. The intrahepatic tumor burden, defined as the volume of hepatic tumors occupying the remaining liver, was evaluated using dynamic contrast-enhanced computed tomography images obtained as a baseline study. Two board-certified radiologists, S.J. and C.A., each with over 10 years of experience in liver cancer imaging, conducted the assessment collaboratively and reached a consensus (S1 Fig.).

3. Statistical analysis

Categorical variables were analyzed using frequencies and percentages and compared using Fisher’s exact test or Pearson’s chi-square test, as appropriate. Continuous variables were analyzed using the median and range and compared using a Student’s t-test. Survival analysis was performed using the Kaplan-Meier method, and the log-rank test was used to compare the groups. Statistical analyses were performed using SPSS software ver. 22 (IBM Corp., Armonk, NY). Statistical significance was set at p < 0.05. GraphPad Prism 8.0 software (GraphPad Software Inc., San Diego, CA) was used to create the graphs.

Results

1. Patient characteristics

A total of 246 patients who received first-line Ate/Bev treatment between May 2020 and April 2022 were included in this study (S2 Fig.). The median age at diagnosis was 61 years (interquartile range, 54 to 68 years), and 209 patients (85.0%) were male. Hepatitis B virus infection (n=166, 67.5%)was the most common etiology of HCC, and 207 patients (84.1%) had liver cirrhosis. Baseline α-fetoprotein (AFP) levels at diagnosis were < 400 ng/mL in 150 patients (61.0%), and 241 patients had an Eastern Cooperative Oncology Group (ECOG) performance status of < 2. Portal vein tumor thrombosis (PVTT) was observed in 102 patients (41.5%). More than half of the patients had extrahepatic spread (n=155, 63.0%) and had previously received local therapy (n=165, 67.1%) for HCC. Regarding liver function, 128 patients (52.0%) had Child-Pugh class A5 and 126 patients (51.2%) had ALBI grade 1. Most patients had BCLC stage C (n=206, 83.7%). Additionally, 134 patients (54.5%) had an intrahepatic tumor burden of < 25% (Table 1).

Demographic characteristics of the patients at baseline

2. Characteristics of the long-term treatment group

In the long-term treatment group, more patients had intrahepatic tumor burdens of < 25% compared to the short-term group (73.9% vs. 46.9%, p=0.002). PVTT occurred less frequently in the long-term treatment group (29.0% vs. 46.3%, p=0.013). In addition, more patients had an ECOG performance status of 0 (63.8% vs. 32.8%, p < 0.001) and were classified as Child-Pugh class A5 (66.7% vs. 46.3%, p=0.006) or ALBI grade 1 (68.1% vs. 44.6%, p=0.002) than did those in the short-term group. Other baseline characteristics, including known prognostic factors [19-22] such as C-reactive protein, neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, or protein induced by vitamin K absence or antagonistII demonstrated no significant differences between the longand short-term groups (Table 1).

3. Survival outcomes

At the data cutoff of May 10, 2023, the median follow-up duration was 15.5 months (95% confidence interval [CI], 12.5 to 17). Overall, the median PFS and OS were 6.9 months (95% CI, 4.4 to 8.6) and 15.8 months (95% CI, 12.5 to 19.9), respectively. The ORR was 32.1% (79/246).

Considering the survival outcomes according to the treatment duration, the long-term treatment group demonstrated better PFS (25.0 months vs. 3.5 months, p < 0.001) and OS (not reached vs. 9.8 months, p < 0.001) than the short-term group (S3 Fig.). Moreover, the ORR was significantly higher in the long-term treatment group than in the short-term treatment group (75.4% vs. 15.3%, p < 0.001) (S4 Table).

4. Survival outcomes according to the intrahepatic tumor burden

To further evaluate the impact of the intrahepatic tumor burden on the survival outcomes, we evaluated PFS and OS according to the intrahepatic tumor burden. PFS was significantly different according to the intrahepatic tumor burden. The median PFS of patients with burdens of < 25%, 25%-50%, 50%-75%, and > 75% were 11.3, 4.0, 3.5, and 1.5 months, respectively (p < 0.001) (Fig. 1A). The OS demonstrated a similar pattern. The median OS of patients with a burden of < 25%, 25%-50%, 50%-75%, and > 75% was not reached at 11.1, 4.5, and 3.0 months, respectively (p < 0.001) (Fig. 1B).

Fig. 1.

Survival outcomes according to the intrahepatic tumor burden: PFS (A), OS (B), objec tive response rate (C). CI, confidence interval; CR, complete response; NR, not reached; OS, overall survival; PD, progressive disease; PFS, progression-free survival; PR, partial response; SD, stable disease.

Subsequently, the ORR based on the intrahepatic tumor burden was tested. ORR decreased as the intrahepatic tumor burden increased (p=0.002) (Fig. 1C). The ORRs of patients with tumor burdens of < 25%, 25%-50%, 50%-75%, and > 75% were 41.1%, 26.3%, 18.6%, and 8.3%, respectively. CR was achieved in 7.5% (10/134) and 1.8% (1/57) of patients with tumor burdens of < 25% and 25%-50%, respectively. However, no CR was observed in patients with a tumor burden of 50%-75% and > 75%.

5. Safety of long-term treatment with Ate/Bev

In the long-term treatment group, atezolizumab-related thyroid dysfunction (10.2% vs. 31.9%, p < 0.001), dermatological toxicity (14.7% vs. 29.0%, p=0.010), and arthritis (1.7% vs. 7.2%, p=0.041) occurred more frequently in the long- than in the short-term treatment group. Moreover, the incidence of bevacizumab-related AEs, such as hypertension (22.6% vs. 44.9%, p=0.001) and proteinuria (38.4% vs. 69.6%, p < 0.001), was higher in the long-term treatment group. Although no patient discontinued atezolizumab due to AEs, 34 patients (13.8%) discontinued bevacizumab due to AEs. The rate of bevacizumab discontinuation was higher in the long-term treatment group than in the short-term treatment group (8.5% vs. 27.5%, p < 0.001) (Table 2).

Treatment-related AEs

The median time to onset (mTTO) of atezolizumab-related thyroid dysfunction, dermatologic toxicity, and arthritis was 2.8 months (interquartile range [IQR], 1.8 to 4.3), 3.3 (IQR, 1.4 to 8.4), and 5.5 (IQR, 4.4 to 9.2), respectively. On the other hand, the mTTO of bevacizumab-related hypertension and proteinuria was 4.2 (IQR, 2.1 to 8.3) and 6.8 months (IQR, 3.7 to 10.5), respectively.

6. Changes in liver function in the long-term treatment group

In the long-term treatment group, changes in liver function were tracked using the Child-Pugh score and ALBI grade over a 1-year period. As treatment continued, a fraction of the patients experienced deteriorating liver function. Specifically, 87.0% of the patients were classified as Child-Pugh class A before Ate/Bev treatment; however, 75.4% remained in Child-Pugh class A after 1 year of treatment. While the proportion of patients with Child-Pugh class A5 showed no change from 60.9%, the proportion of patients with Child-Pugh class A6 notably decreased from 26.1% to 14.5% (Fig. 2A). Moreover, the proportion of patients classified as having ALBI grade 1 decreased from 68.1% to 50.7% after 1 year of treatment (p=0.025) (Fig. 2B).

Fig. 2.

Changes in liver function over 12 months of atezolizumab plus bevacizumab treatment in the long-term treatment group (n=69). (A) Child-Pugh (CP) score. (B) Albumin-Bilirubin grade.

7. Proportions of patients maintained on Ate/Bev treatment over 2 years

To track the proportion of patients who maintained Ate/Bev treatment over a 2-year period, we separately evaluated 182 patients who started Ate/Bev treatment by August 2021 (S2 Fig.). Of them, 125 patients (68.7%) maintained treatment for more than 3 months, 87 (47.8%) for more than 6 months, 59 (32.4%) for more than 12 months, 41 (22.5%) for more than 18 months, and 31 (17.0%) for 24 months or more (Fig. 3A). Meanwhile, when the same analysis was conducted with only responders who showed CR or PR, all 61 responders maintained Ate/Bev treatment for more than 3 months, 55 (90.2%) for more than 6 months, 44 (72.1%) for more than 12 months, 36 (59.0%) for more than 18 months, and 28 (45.9%) for 24 months or more (Fig. 3B).

Fig. 3.

Proportions of patients maintained on atezolizumab plus bevacizumab treatment over two years (separate cohort).

The proportion of patients completing 24 months of treatment was 24.8%, 35.6%, 52.5%, and 75.6% for those who received 3, 6, 12, and 18 months of Ate/Bev treatment, respectively; these rates were elevated among responders at 45.9%, 50.9%, 63.6%, and 77.8%, respectively.

Regarding patients with PFS of < 3 months, they had a significantly larger proportion of elevated AFP level (≥ 400 ng/mL) (56.4% vs. 29.7%, p=0.001) and PVTT (50.0% vs. 31.6%, p=0.013) and BCLC stage C (95.6% vs. 76.1%, p < 0.001); and a smaller proportion of ECOG performance status 0 (22.1% vs. 55.3%, p < 0.001), Child-Pugh A5 (38.8% vs. 65.2%, p=0.001) at baseline than those with PFS of 3 or more months (S5 Table).

Discussion

With the advent of immunotherapy, long-term survival has become possible in patients with advanced HCC. However, few studies have reported on how many patients benefit from Ate/Bev in the long term; the characteristics of these patients remain unclear. Moreover, few studies exist on the clinical changes, such as liver function decline, that may occur during long-term exposure to Ate/Bev treatment. In this study, one-third of the patients were treated with Ate/Bev for > 1 year (i.e., the long-term treatment group). This group had a better performance status (ECOG 0), better liver function (Child-Pugh class A5 or ALBI grade 1), lower PVTT prevalence, and a lower intrahepatic tumor burden (< 25%) than the short-term group. More patients in the long-term treatment group experienced atezolizumab- and bevacizumab-related AEs. In addition, some patients in the long-term treatment group experienced liver function deterioration during Ate/Bev treatment.

The long-term treatment group was characterized by a low intrahepatic tumor burden (< 25%). Further analysis of survival outcomes according to the intrahepatic tumor burden of < 25%, 25%-50%, 50%-75%, and > 75% revealed that the PFS, OS, and ORR of Ate/Bev-treated patients were well stratified according to the intrahepatic tumor burden. A recent study [23] reported that Ate/Bev, unlike anti–programmed death-1 monotherapy [24], demonstrated favorable responses upon treatment of intrahepatic lesions, comparable to those of extrahepatic lesions. However, larger intrahepatic tumors (i.e., ≥ 5 cm) were associated with inferior ORR and OS compared to smaller intrahepatic tumors, highlighting the need to verify the therapeutic efficacy of Ate/Bev according to the intrahepatic tumor burden. Our finding supports that increased intrahepatic tumor burden may prognosticate poor survival outcomes with Ate/Bev treatment. For patients with very large intrahepatic tumors, exploring strategies that incorporate pretreatment to control the large intrahepatic mass before Ate/Bev treatment may be necessary to enhance treatment efficacy.

In terms of AEs, the long-term treatment group experienced both atezolizumab- and bevacizumab-related AEs. However, the time-to-onset of AEs caused by the two agents showed different patterns. Atezolizumab-related AEs tended to occur early in the treatment course, whereas bevacizumab-related AEs tended to occur later, which suggest that atezolizumab-related AEs are immunological events that occur relatively early presumably caused by the bystander effects of ICI-induced T-cell activation events following atezolizumab treatment, rather than a consequence of chronic exposure to atezolizumab treatment [17,25]. As atezolizumab-related AEs, such as thyroid and dermatologic toxicities, are generally manageable, early detection and management efforts are needed to achieve long-term benefits without compromising the quality of life. However, later occurrences of bevacizumab-related AEs, such as hypertension and proteinuria, imply the consequences of cumulative stress on the vascular system resulting from prolonged use. Kudo et al. [26] recently reported that the efficacy of Ate/Bev in HCC was comparable between patients whose bevacizumab regimen was interrupted due to AEs and those who did not demonstrate AEs; therefore, an optimal bevacizumab application strategy should be considered in future studies to balance the potential risks and benefits.

Regarding changes in the liver function in the long-term treatment group in our cohort, some patients exhibited deterioration of liver function (i.e., an increase in the Child-Pugh score and ALBI grade). Intriguingly, the patients whose liver function worsened with long-term Ate/Bev treatment mainly including individuals who had already started to deteriorate, with a baseline Child-Pugh score of A6 or higher. This finding suggests that even in Child-Pugh class A patients, it is necessary to monitor A6 patients separately from A5 patients. The STRIDE regimen (tremelimumab plus durvalumab) [27,28] had little effect on liver function deterioration during long-term treatment, suggesting that, unlike dual immunotherapy, long-term immunotherapy combined with an anti-angiogenic agent may affect liver function.

In a separate cohort analysis of patients who started Ate/Bev treatment 2 years ago, 17% remained in treatment for 2 years or more; however, one-third discontinued treatment in the first 3 months, suggesting an urgent need to identify optimal biomarkers that can predict therapeutic response to Ate/Bev to select patients who can achieve a long-term clinical benefit [13,14]. On the other hand, when the same analysis was performed with only responders, 46% of patients continued treatment for 2 years or more, and all patients continued treatment in the first 3 months, suggesting that the combination of immunotherapy with an improved response rate is needed for patients to maintain long-term treatment. In this context, because the recent phase 1b/2 trial demonstrated that the addition of an anti-TIGIT (T-cell immunoglobulin and ITIM domain) antibody, tiragolumab, to Ate/Bev, improved the ORR (11.1% vs. 42.5%) [29], the results of a consecutive phase 3 trial are awaited (ClinicalTrials.gov Identifier: NCT05904886).

Our study has several limitations. First, it was susceptible to unintended biases owing to its retrospective nature, such as selection bias. Second, because all of the patients were of Asian ethnicity and since the majority of patients had chronic hepatitis B virus infection, the general applicability of the results warrants further investigation in diverse populations. Third, owing to the small number of patients who reached 2 years of treatment, liver function assessment according to treatment duration in the long-term treatment group was limited to 1 year. In addition, while adopting the concept of intrahepatic tumor burden in prognostication was a meaningful approach, the underlying mechanism needs to be elucidated in future translational research and warrants further validation in larger, prospective cohorts. Subsequently, constructing a predictive model with expanded cohorts of those who were treated with Ate/Bev with validation would be invaluable. Despite these limitations, our study is the first to demonstrate patterns of liver function deterioration in patients treated with long-term Ate/Bev treatment.

In conclusion, a good performance stage, good liver function, lower PVTT, and lower intrahepatic tumor burden are associated with long-term treatment in patients with unresectable HCC treated with first-line Ate/Bev. A higher proportion of patients experienced atezolizumab-related adverse immunological AEs early in the course of treatment. In addition, a fraction of patients, especially those with Child-Pugh A6, experienced deterioration of liver function during long-term treatment.

Electronic Supplementary Material

Supplementary materials are available at Cancer Research and Treatment website (https://www.e-crt.org).

Notes

Ethical Statement

This study was conducted in accordance with the ethical guidelines of the Declaration of Helsinki and approved by the participating hospitals’ institutional review boards (CHA Bundang Medical Center, CHA-2020-12-030; Ulsan University Hospital, 2020-12-006; Haeundae Paik Hospital, 2020-12-019-001). The need for informed consent in this study was waived, as Korean regulations do not require consent for retrospective analyses. Data were anonymized and de-identified prior to analysis.

Author Contributions

Conceived and designed the analysis: Kim Y, Kim JS, An C, Kim C, Chon HJ.

Collected the data: Kim JS, Kang B, Kim I, Kim H, Lee WS, Sang YB, Jung S, Kim C, Chon HJ.

Contributed data or analysis tools: Kim Y, Kim JS, An C, Kim C, Chon HJ.

Performed the analysis: Kim Y, Kim JS, An C, Kim C, Chon HJ.

Wrote the paper: Kim Y, Kim JS, Kang B, Kim I, Kim H, Lee WS, Sang YB, Jung S, An C, Kim C, Chon HJ.

Conflicts of Interest

Hong Jae Chon has consulting or advisory roles at Eisai, Roche, Bayer, ONO, MSD, BMS, Celgene, Sanofi, Servier, AstraZeneca, SillaJen, Menarini, and GreenCross Cell, and has received research grants from Roche, Dong-A ST, and Boryung Pharmaceuticals. Chan Kim has consulting or advisory roles at Roche, ONO, MSD, BMS, Oncocross, and Virocure, and has received research grants from Boryung Pharmaceuticals, Oncocross, SillaJen, and Virocure.

Acknowledgements

This work was supported by grants from the National Research Foundation of Korea (NRF) funded by the Korean government (MSIT) (NRF-2023R1A2C2004339 to Hong Jae Chon and NRF-2023R1A2C2006375 to Chan Kim).

References

1. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 2008;359:378–90.
2. Kudo M, Finn RS, Qin S, Han KH, Ikeda K, Piscaglia F, et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. Lancet 2018;391:1163–73.
3. Llovet JM, Pinyol R, Kelley RK, El-Khoueiry A, Reeves HL, Wang XW, et al. Molecular pathogenesis and systemic therapies for hepatocellular carcinoma. Nat Cancer 2022;3:386–401.
4. Yang C, Zhang H, Zhang L, Zhu AX, Bernards R, Qin W, et al. Evolving therapeutic landscape of advanced hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2023;20:203–22.
5. Llovet JM, Hernandez-Gea V. Hepatocellular carcinoma: reasons for phase III failure and novel perspectives on trial design. Clin Cancer Res 2014;20:2072–9.
6. Hepatocellular carcinoma. Nat Rev Dis Primers 2021;7:7.
7. Llovet JM, Castet F, Heikenwalder M, Maini MK, Mazzaferro V, Pinato DJ, et al. Immunotherapies for hepatocellular carcinoma. Nat Rev Clin Oncol 2022;19:151–72.
8. Sperandio RC, Pestana RC, Miyamura BV, Kaseb AO. Hepatocellular carcinoma immunotherapy. Annu Rev Med 2022;73:267–78.
9. Lee MS, Ryoo BY, Hsu CH, Numata K, Stein S, Verret W, et al. Atezolizumab with or without bevacizumab in unresectable hepatocellular carcinoma (GO30140): an open-label, multicentre, phase 1b study. Lancet Oncol 2020;21:808–20.
10. Lee WS, Yang H, Chon HJ, Kim C. Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity. Exp Mol Med 2020;52:1475–85.
11. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med 2020;382:1894–905.
12. NCCN clinical practice guidelines in oncology (NCCN Guidelines) for hepatocellular carcinoma, V.2.2023 [Internet]. Plymouth Meeting, PA: National Comprehensive Cancer Network; 2023. [cited 2023 Dec 31]. Available from: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1514.
13. Kim C, Yang H, Kim I, Kang B, Kim H, Kim H, et al. Association of high levels of antidrug antibodies against atezolizumab with clinical outcomes and T-cell responses in patients with hepatocellular carcinoma. JAMA Oncol 2022;8:1825–9.
14. Yang H, Kang B, Ha Y, Lee SH, Kim I, Kim H, et al. High serum IL-6 correlates with reduced clinical benefit of atezolizumab and bevacizumab in unresectable hepatocellular carcinoma. JHEP Rep 2023;5:100672.
15. Zhu AX, Dayyani F, Yen CJ, Ren Z, Bai Y, Meng Z, et al. Alphafetoprotein as a potential surrogate biomarker for atezolizumab+bevacizumab treatment of hepatocellular carcinoma. Clin Cancer Res 2022;28:3537–45.
16. Zhu AX, Abbas AR, de Galarreta MR, Guan Y, Lu S, Koeppen H, et al. Molecular correlates of clinical response and resistance to atezolizumab in combination with bevacizumab in advanced hepatocellular carcinoma. Nat Med 2022;28:1599–611.
17. Song YS, Yang H, Kang B, Cheon J, Kim I, Kim H, et al. Thyroid dysfunction after atezolizumab and bevacizumab is associated with favorable outcomes in hepatocellular carcinoma. Liver Cancer 2024;13:89–98.
18. An HJ, Chon HJ, Kim C. Peripheral blood-based biomarkers for immune checkpoint inhibitors. Int J Mol Sci 2021;22:9414.
19. Zheng Z, Zhou L, Gao S, Yang Z, Yao J, Zheng S. Prognostic role of C-reactive protein in hepatocellular carcinoma: a systematic review and meta-analysis. Int J Med Sci 2013;10:653–64.
20. Cheon J, Yoo C, Hong JY, Kim HS, Lee DW, Lee MA, et al. Efficacy and safety of atezolizumab plus bevacizumab in Korean patients with advanced hepatocellular carcinoma. Liver Int 2022;42:674–81.
21. Li DZ, Guo J, Song QK, Hu XJ, Bao XL, Lu J. Prognostic prediction of the platelet-to-lymphocyte ratio in hepatocellular carcinoma: a systematic review and meta-analysis. Transl Cancer Res 2022;11:4037–50.
22. Zhang D, Liu Z, Yin X, Qi X, Lu B, Liu Y, et al. Prognostic value of PIVKA-II in hepatocellular carcinoma patients receiving curative ablation: a systematic review and meta-analysis. Int J Biol Markers 2018;33:266–74.
23. Cheon J, Jung S, Kang B, Kim H, Kim C, Chon H. Organ-specific responses to atezolizumab plus bevacizumab in patients with advanced hepatocellular carcinoma. J Clin Oncol 2022;40(16 Suppl):4076.
24. Kim HS, Kim CG, Hong JY, Kim IH, Kang B, Jung S, et al. The presence and size of intrahepatic tumors determine the therapeutic efficacy of nivolumab in advanced hepatocellular carcinoma. Ther Adv Med Oncol 2022;14:17588359221113266.
25. Yoest JM. Clinical features, predictive correlates, and pathophysiology of immune-related adverse events in immune checkpoint inhibitor treatments in cancer: a short review. Immunotargets Ther 2017;6:73–82.
26. Kudo M, Tsuchiya K, Shao YY, Finn RS, Galle PR, Ducreux M, et al. Impact of bevacizumab being skipped due to adverse events of special interest for bevacizumab in patients with unresectable hepatocellular carcinoma treated with atezolizumab plus bevacizumab: an exploratory analysis of the phase III IMbrave150 study. Liver Cancer 2023;13:401–12.
27. Abou-Alfa GK, Lau G, Kudo M, Chan SL, Kelley RK, Furuse J, et al. Tremelimumab plus durvalumab in unresectable hepatocellular carcinoma. NEJM Evid 2022;1:EVIDoa2100070.
28. Vogel A, Chan SL, Furuse J, Tak WY, Masi G, Varela M, et al. 79P outcomes by baseline liver function in patients with unresectable hepatocellular carcinoma treated with tremelimumab and durvalumab in the phase III Himalaya study. Ann Oncol 2022;33(Suppl 9):S1463–4.
29. Finn RS, Ryoo BY, Hsu CH, Li D, Burgoyne A, Cotter C, et al. Results from the MORPHEUS-liver study: phase Ib/II randomized evaluation of tiragolumab (tira) in combination with atezolizumab (atezo) and bevacizumab (bev) in patients with unresectable, locally advanced or metastatic hepatocellular carcinoma (uHCC). J Clin Oncol 2023;41(16 Suppl):4010.

Article information Continued

Fig. 1.

Survival outcomes according to the intrahepatic tumor burden: PFS (A), OS (B), objec tive response rate (C). CI, confidence interval; CR, complete response; NR, not reached; OS, overall survival; PD, progressive disease; PFS, progression-free survival; PR, partial response; SD, stable disease.

Fig. 2.

Changes in liver function over 12 months of atezolizumab plus bevacizumab treatment in the long-term treatment group (n=69). (A) Child-Pugh (CP) score. (B) Albumin-Bilirubin grade.

Fig. 3.

Proportions of patients maintained on atezolizumab plus bevacizumab treatment over two years (separate cohort).

Table 1.

Demographic characteristics of the patients at baseline

Variable Total (n=246) Short-term treatment groupa) (n=177) Long-term treatment groupb) (n=69) p-valuec)
Age (yr) 61 (54-68) 62 (53-68) 61 (55-68) 0.810d)
Sex
 Male 209 (85.0) 152 (85.9) 57 (82.6) 0.520
 Female 37 (15.0) 25 (14.1) 12 (17.4)
Etiology of HCC
 Hepatitis B 166 (67.5) 115 (65.0) 51 (73.9) 0.130e)
 Hepatitis C 18 (7.3) 17 (9.6) 1 (1.5)
 Alcohol 31 (12.6) 22 (12.4) 9 (13.0)
 Others 31 (12.6) 23 (13.0) 8 (11.6)
Cirrhosis
 Yes 207 (84.1) 151 (85.3) 56 (81.2) 0.423
 No 39 (15.9) 26 (14.7) 13 (18.8)
AFP (ng/mL)
 < 400 150 (61.0) 103 (58.2) 47 (68.1) 0.152
 ≥ 400 96 (39.0) 74 (41.8) 22 (31.9)
ECOG performance status
 0 102 (41.5) 58 (32.8) 44 (63.8) < 0.001e)
 1 139 (56.5) 115 (65.0) 24 (34.8)
 2 5 (2.0) 4 (2.3) 1 (1.5)
Portal vein tumor thrombosis
 No 144 (58.5) 95 (53.7) 49 (71.0) 0.013
 Yes 102 (41.5) 82 (46.3) 20 (29.0)
Extrahepatic spread
 No 91 (37.0) 62 (35.0) 29 (42.0) 0.467
 Yes 155 (63.0) 115 (65.0) 40 (58.0)
Prior local treatment
 No 81 (32.9) 63 (35.6) 18 (26.1) 0.154
 Yes 165 (67.1) 114 (64.4) 51 (73.9)
Child-Pugh classification
 A5 128 (52.0) 82 (46.3) 46 (66.7) 0.006e)
 A6 62 (25.2) 47 (26.6) 15 (21.7)
 B7 36 (14.6) 33 (18.6) 3 (4.4)
 B8-9 20 (8.1) 15 (8.5) 5 (7.3)
ALBI grade
 1 126 (51.2) 79 (44.6) 47 (68.1) 0.002e)
 2 115 (46.7) 95 (53.7) 20 (29.0)
 3 5 (2.0) 3 (1.7) 2 (2.9)
BCLC stage
 B 41 (16.7) 26 (14.7) 15 (21.7) 0.183
 C 502 (83.3) 151 (85.3) 54 (78.3)
Intrahepatic tumor burden (%)
 < 25 134 (54.5) 83 (46.9) 51(73.9) 0.002e)
 25-50 57 (23.2) 47 (26.6) 10 (14.5)
 50-75 43 (17.5) 36 (20.3) 7 (10.1)
 > 75 12 (4.9) 11 (6.2) 1 (1.5)
PIVKA-II (mAU/mL)
 < 200 120 (48.8) 78 (44.1) 42 (60.9) 0.018
 ≥ 200 126 (51.2) 99 (55.9) 27 (39.1)
NLR (n=244) 2.78 (1.81-4.27) 2.87 (1.98-4.43) 2.12 (1.47-3.93) 0.061d)
PLR (n=244) 0.13 (0.09-0.20) 0.14 (0.10-0.20) 0.10 (0.08-0.17) 0.323d)
CRP (mg/dL) (n=198) 0.52 (0.16-1.66) 0.81 (0.18-2.00) 0.25 (0.09-0.99) 0.172d)

Values are presented as median (interquartile range) or number (%). AFP, α-fetoprotein; ALBI, albumin-bilirubin grade; BCLC, Barcelona Clinical Liver Cancer stage; CRP, C-reactive protein; ECOG, Eastern Cooperative Oncology Group; HCC, hepatocellular carcinoma; NLR, neutrophil-to-lymphocyte ratio; PIVKA-II, protein induced by vitamin K absence or antagonist-II; PLR, platelet-to-lymphocyte ratio.

a)

Patients treated with atezolizumab plus bevacizumab for less than 1 year,

b)

Patients treated with atezolizumab plus bevacizumab for 1 year or more,

c)

Fisher’s exact test or Pearson’s chi-square test, as appropriate,

d)

Student’s t test,

e)

Fisher’s exact test.

Table 2.

Treatment-related AEs

Short-term treatment groupa) (n=177) Long-term treatment groupb) (n=69) p-valuec)
Atezolizumab-related AEs (any grade)
 Diabetes mellitus 6 (3.4) 3 (4.3) 0.713
 Adrenal insufficiency 4 (2.3) 3 (4.3) 0.404d)
 Thyroid toxicity 18 (10.2) 22 (31.9) < 0.001
 Dermatologic toxicity 26 (14.7) 20 (29.0) 0.010
 Colitis 14 (7.9) 10 (14.5) 0.118
 Fatigue 45 (25.4) 13 (18.8) 0.275
 Liver toxicity 60 (33.9) 30 (43.5) 0.161
 Pituitary toxicity 2 (1.1) 4 (5.8) 0.054d)
 Arthritis 3 (1.7) 5 (7.3) 0.041d)
 Pneumonitis 1 (0.6) 1 (1.5) 0.483d)
Bevacizumab-related AEs (any grade)
 Hypertension 40 (22.6) 31 (44.9) 0.001
 Proteinuria 68 (38.4) 48 (69.6) < 0.001
 Palmo-plantar erythrodysesthesia 1 (0.6) 0 > 0.99d)
 Bleeding 14 (7.9) 10 (14.5) 0.118
 Thrombosis 2 (1.1) 0 > 0.99d)
AEs leading to treatment discontinuation
 Atezolizumab 0 0
 Bevacizumab 15 (8.5) 19 (27.5) < 0.001
Grade 5 Aese) 7 (4.0) 1 (1.5) 0.448d)

Values are presented as number (%). AE, adverse event.

a)

Patients treated with atezolizumab plus bevacizumab for less than 1 year,

b)

Patients treated with atezolizumab plus bevacizumab for 1 year or more,

c)

Fisher’s exact test or Pearson’s chi-square test, as appropriate,

d)

Fisher’s exact test,

e)

Grade 5 adverse events in the short-term group included gastrointestinal hemorrhage (in 3 patients), multi-organ dysfunction syndrome, intracranial hemorrhage, duodenal ulcer perforation, and mesenteric vein thrombosis (in 1 patient each); grade 5 events in the long-term treatment group included gastrointestinal hemorrhage (in 1 patient).