Back to Journals » Journal of Hepatocellular Carcinoma » Volume 12

Atezolizumab Plus Bevacizumab in Patients with Unresectable Hepatocellular Carcinoma: Real-World Experience From a US Community Oncology Network

Authors Cosgrove D, Tan R, Osterland AJ , Hernandez S, Ogale S, Mahrus S, Murphy J, Wilson T, Patton G, Loaiza-Bonilla A, Singal AG

Received 8 October 2024

Accepted for publication 13 January 2025

Published 18 April 2025 Volume 2025:12 Pages 791—804

DOI https://doi.org/10.2147/JHC.S492881

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Prof. Dr. Jörg Trojan



David Cosgrove,1 Ruoding Tan,2 Andrew J Osterland,3 Sairy Hernandez,2 Sarika Ogale,2 Sami Mahrus,2 John Murphy,3 Thomas Wilson,3 Gregory Patton,3 Arturo Loaiza-Bonilla,4 Amit G Singal5

1Medical Oncology, Compass Oncology/The US Oncology Network, Vancouver, WA, USA; 2Genentech, South San Francisco, CA, USA; 3Real-World Research, Ontada, Boston, MA, USA; 4Medical Oncology, Capital Health Cancer Center, Pennington, NJ, USA; 5Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA

Correspondence: David Cosgrove, Compass Oncology, 210 SE 136th Ave, Vancouver, WA, 98684, USA, Email [email protected]

Purpose: Atezolizumab plus bevacizumab (atezo-bev) is a preferred first-line (1L) systemic therapy option for unresectable hepatocellular carcinoma (uHCC). However, evidence of its effectiveness in real-world clinical practice, including in patients with impaired liver function, remains limited.
Patients and Methods: This retrospective observational study included adult patients who initiated 1L atezo-bev for uHCC within The US Oncology Network between 1/1/2019 and 8/31/2022 using structured and unstructured electronic health records data. Overall survival (OS) and real-world progression-free survival (rwPFS) were assessed using Kaplan-Meier methods for the overall cohort and in a subgroup of “trial-like” patients with characteristics that were consistent with those of the IMbrave150 Trial (ECOG performance status 0– 1, Child-Pugh class A, albumin-bilirubin grade 1– 2).
Results: Overall, 374 patients met eligibility criteria (mean age 68.8 years, 78.9% male, 31% Child-Pugh class B-C among reported, 18% ECOG performance status ≥ 2 among reported), of whom 132 patients comprised the trial-like subgroup. At a median follow-up of 5.6 months, median (95% CI) OS was 13.2 (9.5, 15.9) months and rwPFS was 6.4 (5.1, 7.7) months. In the trial-like subgroup, median (95% CI) OS was 16.5 (13.2, NR) months and rwPFS was 9.4 (5.7, 12.5) months.
Conclusion: Atezo-bev was used as 1L systemic therapy for HCC in a diverse patient population across US community oncology settings. Real-world effectiveness of atezo-bev among trial-like patients is comparable to that reported in the Phase 3 study. These data can help guide selection of appropriate treatment candidates and maximize the benefits of atezo-bev in routine clinical practice.

Keywords: advanced HCC, first-line, systemic treatment, real-world evidence

Introduction

Hepatocellular carcinoma (HCC) is a primary malignancy of the liver which most commonly occurs in patients with chronic liver disease and cirrhosis.1 Risk factors for HCC include viral hepatitis including hepatitis B virus (HBV) and/or hepatitis C virus (HCV), chronic alcohol consumption, and other conditions such as metabolic dysfunction associated steatotic liver disease (MASLD).1 Patients with HCC generally have a poor prognosis, with overall 5-year survival rates reported to be approximately 20%.2

Upon diagnosis, patients should be evaluated in multidisciplinary settings for potentially curative approaches such as surgical resection, liver transplant or local ablative therapy, although many patients with HCC have advanced disease requiring systemic therapy. Preferred first-line (1L) treatments shifted from tyrosine kinase inhibitors to immunotherapy-based regimens after the introduction of atezolizumab plus bevacizumab (atezo-bev), approved by the US Food and Drug Administration (FDA) in May 2020, followed by tremelimumab plus durvalumab which was approved in October 2022.3 FDA approval of atezo-bev was granted based on results from the Phase 3 IMbrave150 Trial.3,4 Median (95% confidence interval [CI]) progression-free survival (PFS) was 6.8 (5.7–8.3) months in the atezo-bev arm and 4.3 (4.0–5.6) months in the sorafenib arm (hazard ratio [HR]: 0.59; 95% CI: 0.47–0.76; P<0.001).4 Updated IMbrave150 results demonstrated that the median (95% CI) overall survival (OS) was 19.2 (17.0–23.7) months in the atezo-bev arm and 13.4 (11.4–16.9) months in the sorafenib arm (HR: 0.66; 95% CI: 0.52–0.85; P=0.0009) with landmark OS at 18 months of 52% in the atezo-bev arm and 40% in the sorafenib arm.5

Despite the superior efficacy of atezo-bev relative to the previous standard of care demonstrated by the IMbrave150 Trial, there is a continued need to understand the real-world use and effectiveness of atezo-bev in clinical practice. Real-world data is particularly important as clinical practice may vary from clinical trial protocols in terms of the timing and frequency of monitoring, adherence to treatment, and management of complications. Real-world effectiveness may differ from clinical trial efficacy due to differences in patient characteristics. For example, patients with Child Pugh (C-P) class B-C or Eastern Cooperative Oncology Group Performance Status (ECOG PS) >2 are typically excluded from clinical trials. Moreover, trials for HCC have historically lacked racial, ethnic, and socioeconomic diversity, limiting our understanding of treatment responses across different minority groups. While there is some emerging evidence of real-world effectiveness outside the US and in academic institutions, there are limited data in the US community oncology setting on the use and effectiveness of atezo-bev for first-line (1L) systemic treatment of patients with unresectable HCC (uHCC), including those with impaired liver function. Therefore, this study aims to describe demographic and clinical characteristics, treatment patterns, and clinical outcomes of patients diagnosed with uHCC and treated with 1L atezo-bev in real-world practices.

Materials and Methods

Study Design and Data Sources

This was a retrospective observational study of adult patients with HCC treated with 1L atezo-bev within The US Oncology Network, a large community oncology network comprised of over 2400 providers in more than 500 sites of care across the US.6 Data were sourced from structured fields of The US Oncology Network’s iKnowMed (iKM) electronic health record (EHR) and were supplemented with unstructured data abstracted from patient charts for measures such as the physician-documented C-P class, treatment history, and physician-assessed response. The Limited Access Death Master File (LADMF) was also accessed for additional information on vital status and death. Eligibility criteria included patients at least 18 years of age at first diagnosis of HCC, at least 2 visits within the US Oncology Network during the study observation period (June 1, 2020 - November 30, 2022), and initiation of 1L atezo-bev for HCC (index event) during the study identification period (June 1, 2020 - August 31, 2022). Initiation of atezolizumab and bevacizumab were required on the same day or within 42 days of each other. Patients were excluded from the study if they were enrolled in clinical trials, treated for other documented primary cancers during the study observation period, received a liver transplant any time prior to or during the study observation period, or underwent surgery within 6 months prior to the index date.

Patient Characteristics and Outcomes

Demographic and clinical characteristics included age, sex, race and ethnicity (White non-Hispanic, Black non-Hispanic, Hispanic, Asian), ECOG PS, alpha-fetoprotein (AFP), albumin-bilirubin (ALBI) grade (1, 2, 3), C-P class (A, B, C), and liver disease etiology (viral, non-viral). C-P class was reported as documented in the charts. For patients with no physician-documented C-P class, we derived C-P score based on laboratory results and risk factors assessed within 90 days prior to index or the earliest value after the index date (ie, date of atezo-bev initiation).7,8 Treatment regimens during the study observation period were assessed up to third-line (3L). Additional treatment patterns included type and frequency of locoregional therapy (LRT), type and timing of bevacizumab initiation, treatment disposition and reason for discontinuation of 1L atezo-bev. Clinical effectiveness was assessed using progression-free survival (rwPFS) and OS. rwPFS was measured from index to the earliest date of physician-documented progression or death due to any cause. OS was defined as the interval between the index date and date of death due to any cause as documented in the EHR or LADMF.

Statistical Analysis

Descriptive analyses were performed on patient characteristics and treatment patterns. Kaplan-Meier methods were used to assess clinical effectiveness outcomes for the overall cohort and in a “trial-like” cohort of patients with certain clinical characteristics that were consistent with the IMbrave150 Trial (C-P class A, ALBI grade 1–2, and ECOG PS 0–1). Additional exploratory subgroups analyses were conducted by C-P class, ALBI grade, etiology, and race/ethnicity. Patients who did not experience an event were censored on the last contact date during the study observation period. Analyses were conducted using SAS v.9.4 (SAS Institute, Inc; Cary, NC, USA).

Results

Patient Characteristics

Within The US Oncology Network, there were 514 adult patients identified with a documented diagnosis of “hepatocellular carcinoma” or “liver cancer” who initiated 1L atezo-bev for HCC during the study identification period. Among them, 402 patients met additional eligibility criteria using structured EHR data and were selected for chart abstraction. Upon verification of eligibility criteria, 374 patients were included in the study, of whom 132 comprised the “trial-like” subgroup.

Patient characteristics are reported in Table 1. The mean (standard deviation [SD]) age at index was 68.8 (8.9) years, 69.3% were age ≥65 years, 78.9% were male, and 48.4% were non-Hispanic White. ECOG PS was documented at baseline (within 30 days of index) in 291 patients, of whom 239 (82.1%) had an ECOG PS of 0–1 and 52 (17.9%) had ECOG PS ≥2. There were 192 (51.3%) patients with viral disease etiology, and 70 (18.7%) patients with non-viral liver disease. Of the 192 patients with viral liver disease, 172 (89.6%) had HCV and 28 (14.6%) had HBV. Among patients with available data for C-P class (n=332), 132 (39.8%) patients had a documented C-P class in the EHR, and C-P class was derived for an additional 200 (60.2%) patients. As a result, there were 229 (61.2%), 91 (24.3%), and 12 (3.2%) patients from the overall sample with C-P class A, B, and C, respectively. There were 107 (28.6%) patients with ALBI grade 1, 217 (51.0%) with ALBI grade 2, and 30 (8.0%) with ALBI grade 3. Among patients with available data for ALBI who were C-P class A (n=218), 77 (35.3%) were ALBI grade 1 and 136 (62.4%) patients were ALBI grade 2. The median (interquartile range [IQR]) duration of follow-up from initiation of 1L therapy was 5.6 (2.7–12.0) months.

Table 1 Demographics and Clinical Characteristics of Patients Who Initiated 1L Atezolizumab Plus Bevacizumab for uHCC in the US Oncology Network During the Study Identification Period

Treatment Patterns

Prior treatment with locoregional therapies (LRT) was documented in 121 (32.4%) patients. Bevacizumab biosimilars were used in 333 (89.0%) patients, and 346 (92.5%) initiated bevacizumab on the same day as atezolizumab. Initiation of atezolizumab and bevacizumab on separate days but within 2 cycles was observed in 28 (7.5%) patients. Of 296 (79.1%) patients who discontinued 1L atezo-bev during the study period, the primary reason for discontinuation was disease progression or hospice (n=167, 44.7%). Additionally, 11 (2.9%) patients discontinued due to toxicity or an adverse event.

Additional treatment patterns are included in Table 2. Of the 296 patients who discontinued, only 55 (18.6%) patients proceeded to second-line therapy (2L). The most common 2L regimens included cabozantinib (n=21, 38.2%), lenvatinib (n=13, 23.6%), or sorafenib (n=9, 16.4%). Only 14 (3.7%) patients proceeded to 3L during the study period. Treatment sequencing from 1L to 3L is shown in Figure 1.

Table 2 Treatment Patterns of Patients Who Initiated 1L Atezolizumab Plus Bevacizumab for uHCC in the US Oncology Network During the Study Identification Period

Figure 1 Treatment sequencing of patients who initiated 1L atezolizumab plus bevacizumab for uHCC in the US Oncology Network during the study identification period.

Notes: “End of observation” group includes patients who did not initiate a subsequent regimen and had no record of death during the observation period. Of the 178 patients in this group at 2L, n=78 had ongoing 1L therapy. An additional 23 patients did not initiate a subsequent regimen at 3L and had no record of death during the observation period. “Other treatments” in 2L included nivolumab ± ipilimumab, regorafenib, durvalumab, FOLFOX, pembrolizumab + lenvatinib, and ramucirumab.

Clinical Outcomes

During the follow-up period, 224 (59.9%) patients experienced a rwPFS event, and 163 (43.6%) died. Median (95% CI) rwPFS was 6.4 (5.1–7.7) months in the overall cohort and 9.4 (5.7–12.5) months in the “trial-like” subgroup (Figure 2). The estimated rwPFS was 52.2% (95% CI: 46.7–57.5%) at 6 months and 33.4% (95% CI: 27.8–39.1%) at 12 months for all patients (Figure 2). Median (95% CI) OS was 13.2 (9.5–15.9) months in the overall cohort and 16.5 (13.2–NR) months in the “trial-like” subgroup (Figure 3). The estimated OS was 68.6% (95% CI: 63.2–73.3%) at 6 months and 53.5% (95% CI: 47.4–59.2%) at 12 months for all patients (Figure 3).

Figure 2 Kaplan-Meier analysis of median rwPFS for patients who initiated 1L atezolizumab plus bevacizumab for uHCC in the US Oncology Network during the study identification period.

Abbreviations: CI, confidence interval; IQR, interquartile range; rwPFS, real-world progression-free survival.

Figure 3 Kaplan-Meier analysis of median OS for patients who initiated 1L atezolizumab plus bevacizumab for uHCC in the US Oncology Network during the study identification period.

Abbreviations: CI, confidence interval; IQR, interquartile range; NR, not reached; OS, overall survival.

Subgroup Analysis

Unadjusted median (95% CI) rwPFS and OS are reported by C-P class, ALBI grade, ECOG PS, liver disease etiology, and race/ethnicity category in Table 3. As expected, we observed differences in OS by the degree of liver dysfunction and ECOG performance status. Median (95% CI) OS among ALBI grades 1, 2 and 3 were 16.8 (13.8–NR), 9.9 (8.1–15.8), and 2.1 (1.3–4.5) months, respectively, and median (95% CI) OS among ECOG PS 0, 1, and ≥2 were 19.3 (13.8–NR), 9.9 (8.1–15.8), and 3.2 (2.2–6.5) months, respectively (P<0.0001 for both). Statistically significant differences were also observed in rwPFS by ALBI grade (P=0.002) and by ECOG PS (P<0.0001). While OS differed by C-P class (median [95% CI], C-P class A: 16.5 [12.6–NR], C-P class B: 7.5 [4.7–9.9], C-P class C: 4.5 [1.3–NR], P<0.0001), a significant difference in rwPFS by C-P class was not observed (P=0.19). Notably, there were no significant differences in rwPFS or OS by liver disease etiology (P=0.58, P=0.43, respectively) or by race and ethnicity (P=0.74, P=0.85, respectively).

Table 3 Subgroup Analysis of Unadjusted rwPFS and OS for Patients Who Initiated 1L Atezolizumab Plus Bevacizumab for uHCC in the US Oncology Network During the Study Identification Period

Discussion

In this real-world study of patients who initiated 1L atezo-bev for uHCC at community oncology clinics in the United States, we found many patients had clinical features that would have excluded their participation in the IMbrave150 Trial, including over 1 in 4 patients with C-P class B/C cirrhosis and nearly 1 in 5 patients with ECOG PS 2 or more. Additionally, our real-world cohort had a higher proportion of elderly patients and racial ethnic minorities. Furthermore, real-world clinical practice may also vary from clinical trial protocols in terms of the timing and frequency of monitoring, adherence to treatment, management of complications, and measurement of clinical outcomes such as progression. Despite these differences, many of which could lead to less favorable results in the real-world study, patients had a median rwPFS of 6.4 months and OS of 13.2 months, respectively. Subgroup analyses showed that patients with negative prognostic factors such as greater liver dysfunction or poor performance status exhibited worse rwPFS and OS compared to those with preserved liver function and performance status. Differences in patient characteristics of the overall study cohort warranted further evaluation of outcomes among the sub-cohort of patients with trial-like characteristics. Indeed, among trial-like patients, the median rwPFS was 9.4 months and OS was 16.5 months, which is consistent with estimates from the IMbrave150 Trial. However, the trial-like cohort only considers a limited number of patient characteristics (ECOG PS, C-P class, ALBI grade) and other patient characteristics described in Table 1 could be different from the IMbrave150 Trial population. For example, the trial-like cohort consists of older patients (median age: 70 vs 64) and includes a higher proportion of patients with ALBI grade 2 (58% vs 43%) relative to the IMbrave150 trial population, suggesting that the real-world trial-like cohort may have a poorer prognosis.

Findings from this real-world cohort of patients treated at community oncology practices align with results from other US-based studies conducted in other practice settings, including the Veteran Health Administration (VHA) and academic institutions. Using medical records from the National VHA data warehouse from 2017 to 2022, Kaplan et al (2024) reported a median OS of 12.8 (95% CI: 10.6–17.1) months among veterans diagnosed with uHCC who received 1L A+B (N=405).9 Storandt et al (2024) observed a median OS of 21.6 (95% CI: 17.7–34.9) months for patients with C-P class A (n=226) and 6.4 (95% CI: 5.2–9.0) months for those with C-P class B (n=86) treated at multiple academic institutions across the US.10

The present findings are also generally consistent with the results of other observational studies conducted globally including countries outside of the US, though some differences may arise due to regional variations in clinical practice, health systems, and patient populations. For example, D’Alessio et al (2022) conducted a global multi-center retrospective study of A+B patients with uHCC across seven countries, including the US, reporting a median OS of 14.9 (95% CI: 13.6–16.3) months and a median PFS of 6.8 (95% CI: 5.2–8.5) months for all patients (n=216).11 Allaire et al (2024) conducted a registry-based study in France and reported a median PFS of 5 months and a median OS of 23.7 months in a large prospective multicentric cohort (n=545).12 Additionally, Himmelsbach et al (2022) reported a median PFS of 6.5 months in 66 patients with advanced HCC treated with 1L atezo-bev at four cancer centers in Germany and Austria between December 2018 and August 2021.13 De Castro et al (2022) reported median (95% CI) PFS of 5.1 (2.6–7.6) months in 147 patients with advanced HCC who received atezo-bev regardless of prior treatment across six hospitals in Germany and Austria.14 In the subgroup of patients (n=74) with C-P class A, ECOG PS 0–1, and no prior systemic therapy for HCC, median (95% CI) PFS was 8.7 (5.9–11.5) months.14 Lastly, Fulgenzi et al (2022) observed median (95% CI) PFS of 6.9 (6.1–8.3 months) in 296 patients with uHCC, C-P class A, and ECOG PS 0–1 who received 1L atezo-bev at one of 14 tertiary care centers across the USA, Europe and Asia.15 The observed associations between C-P class, ALBI grade, and ECOG PS on clinical outcomes are expected given their prognostic nature and not unique to atezo-bev. Prior studies have demonstrated similar findings across 1L treatment regimens.9–18

Overall, these data underscore the importance of careful patient selection to achieve optimal safety and effectiveness. However, many patients in clinical practice present with greater liver dysfunction and worse performance status, and there are no proven therapies in this patient population. There is currently an ongoing prospective study to generate robust safety and efficacy estimates for atezo-bev in patients with C-P B cirrhosis.19 However, real-world data is helpful to guide decision making and patient discussions about expectation in the interim. The poor observed outcomes in patients with Child Pugh class C cirrhosis or ALBI grade 3, with median survival less than 6 months in both subgroups, reinforce that these patients may be better suited for best supportive care given the high competing risk of mortality from their underlying liver disease.

There has been controversy about the efficacy of immunotherapy in patients with non-viral liver disease, postulated to be driven by T cell exhaustion in patients with MASLD.20 While subgroup analysis of the IMbrave150 Trial suggested differential effect, lack of stratification by etiology precluded robust conclusions. Further, a post-hoc analysis found no significant difference in efficacy after adjusting for baseline differences in the groups.21 While some prior real-world studies have found viral hepatitis treated with 1L atezo-bev to be associated with a more favorable PFS and OS relative to non-viral disease, most have found 1L atezo-bev to be effective regardless of etiology.9–15,22,23 In this study, there was no association between clinical outcomes and etiology observed.

Previous studies have reported racial, ethnic and socioeconomic disparities in the incidence, treatment and outcomes of patients with HCC.24,25 Racial minorities are often under-represented in clinical trials and may face barriers to timely detection and treatment including financial toxicity, transportation, language barriers, medical mistrust and other patient preferences that may impede treatment.26,27 Therefore, real-world data are critical for understanding treatment practices and outcomes in diverse cohorts. While outcomes in our study did not appear to differ significantly across racial and ethnic subgroups as evidenced by overlapping confidence intervals, the sample size may be too small to draw definitive conclusions. It is also likely that patients in this study cohort were engaged in care at community oncology practices which does not exclude the possibility of upstream disparities in access to clinical care.

This study is subject to several limitations inherent to the nature of observational study designs. As this was a retrospective, observational study of EHR data, there is a risk of measurement bias as well as missing data. For instance, Vp classification of macrovascular invasion was not available for the analysis as it was not documented in charts within these US community oncology settings. Documentation of locoregional therapies and esophagogastroduodenoscopy was also limited as services performed outside of the network were not always recorded within the community oncology charts. While C-P class was captured as documented in the EHR in patients with available data, other patients’ C-P class was derived from individual inputs which could include post-treatment data and therefore may not be representative of the patient’s baseline status. Also, our study had a relatively short duration of follow-up, which precluded robust estimates of long-term survival. For the evaluation of rwPFS, this real-world study used provider-documented tumor assessments which may differ from the RECIST criteria used for PFS in clinical trials. Additionally, subgroup analyses of clinical outcomes were not statistically powered to study the differences across subgroups. Lastly, the patient characteristics, disease etiology, treatment patterns and outcomes are reflective of the US community oncology setting which may be different than those observed in academic and/or non-US settings. Despite these limitations, our study adds important effectiveness data from a well characterized set of patients treated in US oncology practices.

Conclusion

In US community oncology settings, atezo-bev is used as 1L treatment for uHCC in a broad and diverse patient population, including those with liver dysfunction, impaired performance status, and racial and ethnic minorities. Our real-world study provides further evidence to support the effectiveness of 1L atezo-bev for uHCC in appropriately selected patients. Among patients with similar clinical characteristics as those included in the IMbrave150 Trial, real-world clinical effectiveness appears similar to that reported from the clinical trial. While further research with larger sample sizes and longer follow-up is warranted, findings from this study can help guide patient selection of appropriate treatment candidates and maximize the benefits of atezo-bev in clinical practice.

Ethics Statement

Institutional Review Board and Compliance/Privacy approval was gained prior to initiation of the retrospective research. Since this project involved the analysis of existing data and records, study information was analyzed in such a manner that research participants could not be directly identified. Patient informed consent was not required due to the nature of the study design. Thus, exemption status and a waiver of informed consent were approved by The US Oncology, Inc. Institutional Review Board. Data were handled in compliance with the Health Insurance Portability and Accountability Act (HIPAA), the Health Information Technology for Economic and Clinical Health (HITECH) Act, and the Declaration of Helsinki.

Acknowledgments

Support for third-party writing assistance for this manuscript was provided by Elyse Viana and Angel Yutig of Nucleus Global. The abstract of this paper was presented as, “Atezolizumab plus bevacizumab (A+B) in patients with unresectable hepatocellular carcinoma (uHCC): Real-world experience from a US community oncology network” at the 2024 ASCO Gastrointestinal Cancers Symposium as a poster presentation with interim findings. The abstract was published in the Journal of Clinical Oncology: https://ascopubs.org/doi/10.1200/JCO.2024.42.3_suppl.447

Funding

This study was sponsored by Genentech Inc, a member of the Roche Group.

Disclosure

DC reports leadership at Compass Oncology and Sarah Cannon Research Institute, honoraria from EMD Serono, and consulting for Daiichi Sankyo, Lilly, Merck KGaA, and Genentech. AGS has served as a consultant or on advisory boards for Genentech, AstraZeneca, Eisai, Bayer, Exelixis, Elevar, Merck, Boston Scientific, Sirtex, HistoSonics, FujiFilm Medical Sciences, Exact Sciences, Roche, Glycotest, Abbott, Freenome, and GRAIL. AGS’s research is supported by NIH R01 MD012565. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. The funding agencies had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; or preparation of the manuscript. RT and SH are employees and stockholders of Genentech Inc, a member of the Roche Group. AO, JM, and GP are employees of McKesson/Ontada. GP also reports locum tenens salary from Texas Oncology and Compass Oncology. TW was an employee of McKesson/Ontada at the time of the study, is a stockholder of McKesson, and reports leadership with Population Health Impact Institute and Trajectory Healthcare, LLC. ALB reports leadership, consulting, and stock in Massive Bio, consulting or advisory roles for Bayer, PSI, BrightInsight, Cardinal Health, Pfizer, Eisai, AstraZeneca, Regeneron, Verily, and Medscape, and honoraria from Guardant Health, Bayer, Amgen, Ipsen, AstraZeneca/Daiichi Sankyo, and Natera. The authors report no other conflicts of interest in this work.

References

1. El-Serag HB. Hepatocellular carcinoma. N Engl J Med. 2011;365(12):1118–1127. doi:10.1056/NEJMra1001683

2. Surveillance, Epidemiology, and End Results (SEER). Cancer stat facts: liver and intrahepatic bile duct cancer. National Cancer Institute. Available from: https://seer.cancer.gov/statfacts/html/livibd.html. Accessed March 5, 2024.

3. Casak S, Donoghue ML, Fashoyin-Aje L, et al. FDA approval summary: atezolizumab plus bevacizumab for the treatment of patients with advanced unresectable or metastatic hepatocellular carcinoma. Clin Cancer Res. 2021;27(7):1836–1841. doi:10.1158/1078-0432.CCR-20-3407

4. 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

5. Finn RS, Qin S, Ikeda M, et al. IMbrave150: updated overall survival (OS) data from a global, randomized, open-label Phase III study of atezolizumab (atezo) + bevacizumab (bev) versus sorafenib (sor) in patients (pts) with unresectable hepatocellular carcinoma (HCC). J Clin Oncol. 2021;39(3_suppl):267. doi:10.1200/JCO.2021.39.3_suppl.267

6. The US Oncology Network. Available from: https://usoncology.com/our-company/. Accessed March 5, 2024.

7. Cabrera R, Singal AG, Colombo M, et al. A real-world observational cohort of patients with hepatocellular carcinoma: design and rationale for TARGET-HCC. Hepatol Commun. 2021;5(3):538–547. doi:10.1002/hep4.1652

8. Aly A, Fulcher N, Seal B, et al. Clinical outcomes by Child-Pugh Class in patients with advanced hepatocellular carcinoma in a community oncology setting. Hepatol Oncol. 2023;10(1):Hep47. doi:10.2217/hep-2023-0002

9. Kaplan DE, Tan R, Xiang C, et al. Overall survival in real-world patients with unresectable hepatocellular carcinoma receiving atezolizumab plus bevacizumab versus sorafenib or lenvatinib as first-line therapy: findings from the National Veterans Health Administration database. Cancers. 2024;16(20):3508.

10. Storandt MH, Zemla TJ, Patell K, et al. Atezolizumab plus bevacizumab as first-line systemic therapy for hepatocellular carcinoma: a multi-institutional cohort study. Oncologist. 2024;29(11):986–996. doi:10.1093/oncolo/oyae142

11. D’Alessio A, Fulgenzi CAM, Nishida N, et al. Preliminary evidence of safety and tolerability of atezolizumab plus bevacizumab in patients with hepatocellular carcinoma and Child-Pugh A and B cirrhosis: a real-world study. Hepatology. 2022;76(4):1000–1012. doi:10.1002/hep.32468

12. Allaire M, Hajer BK, Amaddeo G, et al. THU-466 Treatment with atezolizumab-bevacizumab for hepatocellular carcinoma in the French population outside clinical trials: data from the prospective CHIEF cohort. J Hepatol. 2024;80:S444. doi:10.1016/S0168-8278(24)01393-X

13. Himmelsbach V, Pinter M, Scheiner B, et al. Efficacy and safety of atezolizumab and bevacizumab in the real-world treatment of advanced hepatocellular carcinoma: experience from four tertiary centers. Cancers. 2022;14(7):1722. doi:10.3390/cancers14071722

14. de Castro T, Jochheim LS, Bathon M, et al. Atezolizumab and bevacizumab in patients with advanced hepatocellular carcinoma with impaired liver function and prior systemic therapy: a real-world experience. Ther Adv Med Oncol. 2022;14:17588359221080298. doi:10.1177/17588359221080298

15. Fulgenzi CAM, Cheon J, D’Alessio A, et al. Reproducible safety and efficacy of atezolizumab plus bevacizumab for HCC in clinical practice: results of the AB-real study. Eur J Cancer. 2022;175:204–213. doi:10.1016/j.ejca.2022.08.024

16. Marrero JA, Kudo M, Venook AP, et al. Observational registry of sorafenib use in clinical practice across Child-Pugh subgroups: the GIDEON study. J Hepatol. 2016;65(6):1140–1147. doi:10.1016/j.jhep.2016.07.020

17. Ramaswamy A, Kulkarni A, John G, et al. Survival of trial-like and non-trial-like patients with immunotherapy in advanced hepatocellular carcinoma in real world: a collaborative multicenter Indian study (IMHEP). JCO Glob Oncol. 2023;9:e2300215. doi:10.1200/GO.23.00215

18. Yang X, Chen B, Wang Y, et al. Real-world efficacy and prognostic factors of lenvatinib plus PD-1 inhibitors in 378 unresectable hepatocellular carcinoma patients. Hepatol Int. 2023;17(3):709–719. doi:10.1007/s12072-022-10480-y

19. Genentech, Inc. A study evaluating atezolizumab, with or without bevacizumab, in patients with unresectable hepatocellular carcinoma and Child-Pugh B7 and B8 cirrhosis (Kirros). Available from: https://clinicaltrials.gov/study/NCT06096779. NLM identifier: NCT06096779. Accessed June 27, 2024.

20. Pfister D, Núñez NG, Pinyol R, et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature. 2021;592(7854):450–456. doi:10.1038/s41586-021-03362-0

21. Espinoza M, Muquith M, Lim M, et al. Disease etiology and outcomes after atezolizumab plus bevacizumab in hepatocellular carcinoma: post-hoc analysis of IMbrave150. Gastroenterology. 2023;165(1):286–288.e4. doi:10.1053/j.gastro.2023.02.042

22. Brown TJ, Mamtani R, Gimotty PA, Karasic TB, Yang YX. Outcomes of hepatocellular carcinoma by etiology with first-line atezolizumab and bevacizumab: a real-world analysis. J Cancer Res Clin Oncol. 2023;149(6):2345–2354. doi:10.1007/s00432-023-04590-9

23. Hatanaka T, Kakizaki S, Hiraoka A, et al. Comparative efficacy and safety of atezolizumab and bevacizumab between hepatocellular carcinoma patients with viral and non-viral infection: a Japanese multicenter observational study. Cancer Med. 2023;12(5):5293–5303. doi:10.1002/cam4.5337

24. Wagle N, Park S, Washburn D, et al. Racial and ethnic disparities in hepatocellular carcinoma treatment receipt in the United States: a systematic review and meta-analysis. Cancer Epi Biomarkers Prevention. 2024;33(4):463–470. doi:10.1158/1055-9965.EPI-23-1236

25. Rich NE, Carr C, Yopp AC, et al. Racial and ethnic disparities in survival among patients with hepatocellular carcinoma in the United States: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2022;20(2):e267–e288. doi:10.1016/j.cgh.2020.12.029

26. Jan J, Osho A, Murphy C, et al. Gender, age, racial and ethnic disparities in clinical trial enrollment for primary liver cancer. Gastroenterology. 2022;163(1):14–20.e2. doi:10.1053/j.gastro.2022.03.015

27. Schoenberger H, Rich N, Jones P, et al. Racial and ethnic disparities in barriers to care in patients with hepatocellular carcinoma. Clin Gastroenterol Hepatol. 2023;21(4):1094–1096. doi:10.1016/j.cgh.2021.12.027

Creative Commons License © 2025 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, 4.0) 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.