Impact of neutrophil-to-lymphocyte ratio on survival outcomes among cirrhotic and non-cirrhotic patients with advanced hepatocellular carcinoma under atezolizumab–bevacizumab combination therapy

Spyridon Pantzios, Orestis Sidiropoulos, Antonia Syriha, Ioanna Stathopoulou, Sofia Rellou, Emmanouil Nychas, Georgia Barla, Nikolaos Ptohis, Ioannis Elefsiniotis

“Agioi Anargyroi” General and Oncology Hospital of Kifisia, National and Kapodistrian University of Athens, Greece

Academic Department of Internal Medicine, Hepatogastroenterology Unit, “Agioi Anargyroi” General and Oncology Hospital of Kifisia, National and Kapodistrian University of Athens, Greece (Spyridon Pantzios, Orestis Sidiropoulos, Antonia Syriha, Ioanna Stathopoulou, Sofia Rellou, Emmanouil Nychas, Georgia Barla, Nikolaos Ptohis, Ioannis Elefsiniotis)

Correspondence to: Ioannis Elefsiniotis, Professor of Medicine and Hepatology, National and Kapodistrian University of Athens, Academic Department of Internal Medicine - Hepatogastroenterology Unit, “Agioi Anargyroi” General and Oncology Hospital of Kifisia, Timiou Stavrou and Noufaron 14, Kalyftaki, N. Kifisia, 14564, Athens, Greece, e-mail: ielefs@nurs.uoa.gr
Received 24 February 2025; accepted 19 March 2025; published online 22 April 2025
DOI: https://doi.org/10.20524/aog.2025.0963
© 2025 Hellenic Society of Gastroenterology

Abstract

Background The efficacy of atezolizumab–bevacizumab in patients with hepatocellular carcinoma (HCC) has not been studied separately in cirrhotic and non-cirrhotic patients. Our aim was to evaluate the efficacy of atezolizumab–bevacizumab in these patients, in relation to baseline values of the neutrophil-to-lymphocyte ratio (NLR).

Methods We divided 57 atezolizumab–bevacizumab-treated HCC patients according to baseline NLR (>3: NLR-H, ≤3: NLR-L) and studied overall survival (OS) and progression-free survival (PFS) in 4 groups: group A, non-cirrhotic/NLR-L; group B, non-cirrhotic/NLR-H; group C, cirrhotic/NLR-L; and group D, cirrhotic/NLR-H.

Results The 4 groups were comparable except for etiology, ALBI grade, macrovascular invasion, Barcelona Clinic Liver Cancer stage and prior therapy. Median OS and PFS were 30, 10, 12 and 5 months, and 14, 4, 8 and 2 months, for groups A, B, C, D, respectively (P<0.001). By Cox regression, cirrhotic/NLR-H patients showed significantly worse OS and PFS. Cirrhotic/NLR-L patients had better OS (12 vs. 5 months, P=0.002) and PFS (8 vs. 2 months, P=0.028) compared to cirrhotic/NLR-H. NLR was significantly correlated with OS (P=0.015). Non-cirrhotic/NLR-L patients had better OS (30 vs. 10 months, P=0.006) and PFS (15 vs. 4 months, P=0.01) compared to non-cirrhotic/NLR-H patients. Prior therapy was significantly correlated with better OS (30 vs. 8 months, P<0.001) and PFS (24 vs. 4 months, P<0.001) in non-cirrhotic patients.

Conclusions Cirrhotic/NLR-H HCC patients presented the worst survival. NLR is an independent risk factor for worse survival in cirrhotic patients. Prior therapy is the only factor significantly correlated with OS and PFS in non-cirrhotic patients.

Keywords Hepatocellular carcinoma, immunotherapy, cirrhosis, neutrophil-to-lymphocyte ratio

Ann Gastroenterol 2025; 38 (3): 319-327


Introduction

Liver cancer is a major clinical entity and has become increasingly frequent during recent decades, being the 6th most common type of cancer and the 4th leading cause of cancer-related mortality globally [1]. The most common type of liver cancer is hepatocellular carcinoma (HCC) [2]. The majority of HCC cases are frequently diagnosed in the intermediate or the advanced stage, where potentially curative interventions are not feasible [3].

In 2007, sorafenib was the first agent to receive approval for the systemic treatment of advanced HCC [4]. Since then, more tyrosine kinase inhibitors (TKIs) have gradually been developed and used for this purpose [5-7]. Recently, the introduction of atezolizumab–bevacizumab in unresectable HCC shifted the balance in the HCC treatment landscape towards new immunological pathways. The IMBRAVE150 study showed a survival benefit in patients with unresectable HCC compared to sorafenib in first-line systemic treatment, yielding objective response rates up to 20%, accompanied by a great improvement in patients’ quality of life [8].

Unfortunately, there are only limited data concerning factors that determine the response to immunotherapy in HCC. In recent years, models have been demonstrated that could predict the response to immunotherapy [9], but more complex parameters need to be assessed in order to predict the survival of immunotherapy-treated HCC patients, taking into consideration the complex molecular profile of HCC [10-12]. Another score that has proved valuable in oncology is the neutrophil-to-lymphocyte ratio (NLR) [13,14]. Low NLR values have been associated with objective response and better survival in patients with solid tumors [15-18], and there is a rationale for using it in HCC, as it is a tumor with a complex microenvironment [19-21]. As a result, NLR has been previously correlated in some HCC cohorts with overall survival and response to atezolizumab–bevacizumab therapy [22-24].

Another important variable in HCC therapeutic management is the combined use of surgery or locoregional therapies with immunotherapy. Recently, the IMBRAVE050 study failed to show better survival rates for high-risk HCC patients who received adjuvant atezolizumab–bevacizumab post-resection, compared to active surveillance [25]. However, the administration of locoregional treatment before the initiation of immunotherapy can reshape the tumor microenvironment, making it less immune-resistant and increasing the chances of demonstrating an objective response with immunotherapy, and probably better survival [26,27].

The majority of HCCs used to arise on a substrate of liver cirrhosis [28]. However, the HCC landscape has changed dramatically over the last years. The increasing and more frequent use of antiviral drugs against hepatitis B and C viruses (HBV and HCV), in addition to the growing prevalence of steatotic liver disease, has led to more HCCs appearing in non-cirrhotic and non-viral patients, something that could definitely have a great impact on survival [29,30].

The aim of our study was to evaluate the efficacy of atezolizumab–bevacizumab combination therapy in patients with locally advanced and/or metastatic HCC not amenable to surgery or locoregional treatment, in relation to the baseline NLR values of histologically proved cirrhotic and non-cirrhotic HCC patients.

Patients and methods

We retrospectively evaluated 57 white European patients with locally advanced and/or metastatic HCC who received first-line systemic treatment with atezolizumab–bevacizumab in our center between January 2021 and December 2023. Patients were evaluated for their baseline characteristics on the first day of atezolizumab–bevacizumab treatment. We gathered demographic, clinical and radiological data, while histological information was obtained from all patients through ultrasound-guided liver biopsy. Recorded parameters included sex, age, body mass index (BMI), HCC etiology, presence of diabetes/varices, α-fetoprotein (AFP), baseline NLR, and liver disease severity scores, including Child–Pugh (CP) grade/score, model for end-stage liver disease (MELD)-Na score [31] and albumin–bilirubin (ALBI) grade [32], as well as tumor data concerning the presence of macrovascular invasion (MVI), extrahepatic disease (EHD), Barcelona Clinic Liver Cancer (BCLC) stage and prior HCC therapy at the start of immunotherapy. The diagnosis of HCC was confirmed in all patients with the combined use of AFP, radiological exams (computed tomography [CT] or magnetic resonance imaging [MRI]) and histology. The presence of varices, CP and MELD-Na scores were not evaluated for non-cirrhotic patients.

HCC etiology was divided into viral and non-viral. Chronic HBV and HCV infections were associated with viral HCC. Patients with chronic HCV infection had received prior treatment with direct-acting antivirals, while patients with chronic hepatitis B were appropriately suppressed with antiviral treatment through long term nucleos(t)ide analogs. All patients who declared alcohol use before initiation of treatment were excluded from the study, as were patients who refused to comply with the protocol. All study participants had undergone an ultrasound-guided liver biopsy that was diagnostic for HCC before the initiation of immunotherapy. The presence of liver cirrhosis was evaluated through histological samples, both in patients treated with liver resection and in those who underwent guided biopsy. Data on morphomolecular HCC characteristics were gathered from all liver biopsies, creating 2 distinct HCC subgroups (proliferative and non-proliferative HCCs), according to the recent morphomolecular classification of HCCs [33].

Patients who had received prior systemic treatments were excluded. Prior treatment was allowed if it involved locoregional treatment (transarterial chemoembolization [TACE] or radiofrequency ablation [RFA]) or liver resection, but not both procedures. Patients had to have undergone liver resection within the past 2 years and experienced relapse in an intermediate or advanced stage before the administration of immunotherapy. Furthermore, patients were allowed to have received a maximum of 2 sessions of locoregional treatment with TACE and/or RFA during the previous 2 years and experienced progressive disease before inclusion in the study. Patients received first-line treatment with 1200 mg of intravenous atezolizumab and intravenous bevacizumab at a dose of 15 mg/kg every 21 days, until unacceptable toxicity or loss of clinical benefit [8]. Generally, patients were on a stable dose of both drugs, but treatment was postponed when a grade 3 treatment-related adverse event was detected, and permanently discontinued in patients who exhibited serious adverse events. All 57 patients had to have received at least 3 consecutive cycles of atezolizumab–bevacizumab and had at least 1 tumor assessment in order to be included in this study. Patients underwent tumor assessment every 2 months with CT or MRI. We used the RECIST 1.1 criteria to assess tumor response [34].

We divided a total of 57 HCC patients into 2 groups according to the presence or absence of cirrhosis. Cirrhotic patients accounted for 52.7% of patients (N=30), while the rest of the patients were non-cirrhotic (N=27, 47.3%). We then calculated baseline NLR values for all patients on the first day of atezolizumab–bevacizumab infusion, using the absolute count of neutrophil to lymphocytes in peripheral blood, and divided the patients in those with NLR ≤3 (NLR-L groups) and those with NLR >3 (NLR-H groups). As a result, 4 groups were eventually created: group A (non-cirrhotic, NLR-L, N=13); group B (non-cirrhotic, NLR-H, N=14); group C (cirrhotic, NLR-L, N=11); and group D (cirrhotic, NLR-H, N=19). These groups were compared according to their baseline characteristics, overall survival (OS) and progression-free survival (PFS). We then studied the cirrhotic population separately according to their NLR values and prior treatment status, and performed the same analysis in non-cirrhotic patients.

All patients who participated in the current study gave written informed consent before systemic treatment initiation and a written informed consent form was signed by all living patients at the time of study entry. The protocol of our study was in accordance with the Declaration of Helsinki for human trials and was approved by the Ethics Committee and the Scientific Board of the “Agioi Anargyroi” General and Oncology Hospital of Kifisia, Athens, Greece.

Statistical analysis

We used the IBM SPSS Statistics software (IBM Corp. Released 2012. IBM SPSS statistics for Windows, Version 29.0.2.0, Armonk, NY: IBM Corp.) for the statistical analysis of our data. Variables were separated into numerical variables, shown as mean values ± standard deviation (SD), and categorical variables, which are represented as numbers and percentages. When comparing 2 groups of patients, we used the independent samples’ t-test in order to find significant differences for numerical variables and the chi-square test in order to compare the categorical values of different groups. When 4 groups were compared, we used analysis of variance (1-way ANOVA) to compare groups concerning normally distributed continuous variables, the Kruskal–Wallis H test for non-normally distributed continuous variables, and the chi-square test for categorical variables. We checked normality for continuous variables with the Kolmogorov–Smirnov test. We then did a survival analysis using Kaplan–Meier curves for OS and PFS and compared the median survival of different groups using the log-rank test (Mantel–Cox). If 2 or more groups had statistically significant differences in 1 or more baseline characteristics, we used Cox regression analysis for OS and PFS separately in order to find differences in outcomes between patient groups, adjusting for all covariates in which the groups were not comparable. The Bonferroni correction was used to adjust the significance level for multiple comparisons. Multicollinearity issues were checked while using Cox regression, using values of tolerance and variance inflation factor. Cox regression results are presented using P-values, hazard ratios (HR) and 95% confidence interval (CI) for HR; P-values < 0.05 were considered statistically significant. Lastly, we conducted Cox-regression Kaplan–Meier analysis in order to present survival results at the mean of covariates, in case of comparison between the 2 values of a binary variable.

Results

Of the 57 patients, 45 were males (78.9%). Patients had a mean age of 66.3±11.3 years, a mean BMI of 27.23±4.8 kg/m2, 19 had diabetes (33.3%), 33 viral etiology (57.9%), 29 were ALBI-I (50.8%), 22 had MVI (38.6%), 23 had EHD (40.4%), while 41 were categorized as BCLC-C (71.9%) and 16 as BCLC-B (28.1%) on study entry. In addition, 22 had AFP levels above 400 ng/mL (38.6%), 27 were categorized as proliferative subgroup HCCs (47.4%), 30 as non-proliferative (52.6%) and 31 had received prior HCC therapy (54.4%). In total, 14 patients (24.6%) underwent resection; only 4 were cirrhotic (28.6%) while the remaining 10 resected patients (71.4%) were non-cirrhotic. All resected patients in our study experienced early relapse, defined as HCC recurrence within 2 years post-resection. Locoregional treatment before immunotherapy onset was recorded in a total of 17 patients (29.8%), of whom 8 (47.1%) were cirrhotic, and 9 (52.9%) non-cirrhotic.

We performed our analysis in 3 steps. First of all, we divided the total of 57 HCC patients into 4 different groups, as described above. We then studied these groups according to their baseline characteristics. Taking into consideration that CP and MELD-Na scores were only used in patients with cirrhosis, and that none of the non-cirrhotic patients had signs of portal hypertension, we did not include CP stage and score, MELD-Na score or the presence of varices as baseline characteristics in this first analysis. On the other hand, we used the ALBI grade—which has already been widely used in patients with HCC and not exclusively in cirrhotic patients—as a baseline characteristic to measure hepatic reserve in all teams. The baseline characteristics of the 4 groups are presented in Table 1: the 4 groups were comparable in terms of all the baseline characteristics except for liver disease etiology (P<0.001), ALBI grade (P=0.014), MVI (P<0.001), BCLC stage at treatment initiation (P=0.014) and prior therapy (P=0.009). More specifically, non-cirrhotic patients tended to have non-viral disease, lower ALBI grade, rarely MVI, frequently BCLC-B stage and had often received prior treatment. Median OS (Fig. 1A) was 30, 10, 12 and 5 months for groups A, B, C and D, respectively and the log-rank test showed a statically significant result (P<0.001). We then performed Cox regression analysis for OS, in order to compare the survival of group D to the other 3 groups, after adjusting for all the baseline parameters for which the 4 groups were not comparable (etiology, ALBI, MVI, BCLC, prior therapy). According to the Bonferroni correction, the adjusted alpha level for statistical significance while comparing group D to groups A, B and C was 0.0167 (0.05/3) and we observed a statistically significant difference in OS when comparing group A to group D (HR 0.1, 95%CI 0.020-0.504; P=0.005), but not when comparing group C to group D (P=0.032) or group B to D (P=0.055), while the other covariates failed to achieve statistically significant P-values (Supplementary Fig. 1A). Similarly, median PFS (Fig. 1B) differed significantly (P<0.001) between the 4 groups (14, 4, 8 and 2 months for groups A, B, C and D, respectively). Cox regression for PFS, using the Bonferroni correction, showed a statistically significant difference when comparing group A to D (HR 0.140, 95%CI 0.043-0.459; P=0.006), whereas no statistical significance was found when comparing group B to D (P=0.070) and C to D (P=0.054), and no statistically significant P-values were found for the other covariates (Supplementary Fig. 1B).

Table 1 Baseline characteristics of the 4 groups created according to presence/absence of cirrhosis and baseline NLR status

thumblarge
thumblarge

Figure 1 Overall survival (A) and progression-free survival (B) for each of the 4 groups according to cirrhosis presence/absence and baseline NLR status

OS, overall survival; PFS, progression-free survival; NLR-H, high NLR group; NLR-L, low NLR group

We then conducted a Cox regression analysis for all patients, without categorizing them into groups, in order to assess the impact of cirrhosis and NLR separately for OS and PFS. We found that both presence of cirrhosis (HR 3.173, 95%CI 1.083-9.301; P=0.035) and high NLR (HR 3.076, 95%CI 1.337-7.078; P=0.008) were significantly associated with worse OS, independently of other potential confounders. For PFS, only a high NLR emerged as a statistically significant factor associated with worse PFS (HR 2.54, 95%CI 1.188-5.429; P=0.016), independently of all other confounders (Supplementary Fig. 2A,B).

The second analysis included only the cirrhotic HCC patients of our study (N=30). We divided those patients into 2 groups according to their baseline NLR status, and compared NLR-L and NLR-H patients according to their baseline characteristics (Supplementary Table 1). Those 2 groups were comparable for all baseline parameters, except for CP stage (P=0.021) and number (P=0.007). Median OS was significantly higher (P=0.002) in the cirrhotic NLR-L group (12 months) compared to NRL-H cirrhotic patients (5 months), and similarly PFS was significantly higher (8 months) in cirrhotic NLR-L compared to NLR-H patients (2 months, P=0.028; Fig. 2A,B). Cox regression analysis for OS showed that NLR was significantly associated with OS (HR 3.480, 95%CI 1.274-9.508; P=0.015), independently of CP stage and score, while no statistically significant results were achieved with regard to PFS (Supplementary Fig. 3A,B). We then compared cirrhotic patients according to prior therapy status for HCC: the 2 groups were comparable for all the baseline parameters evaluated (Supplementary Table 2) and did not show differences in median OS (P=0.201) or PFS (P=0.792) (Fig. 2C,D).

thumblarge

Figure 2 Overall survival (A) and progression-free survival (B) in cirrhotic HCC patients according to baseline NLR status. Overall survival (C) and progression-free survival (D) in cirrhotic HCC patients according to prior therapy status

OS, overall survival; PFS, progression-free survival; NLR-H, high NLR group; NLR-L, low NLR group

Finally, we compared the non-cirrhotic HCC patients (N=27) according to their baseline NLR status. The 2 groups were comparable for most of their baseline characteristics (Supplementary Table 3), except for prior therapy (P=0.016). Non-cirrhotic NLR-L patients showed a significantly higher median OS (30 months) compared to NLR-H non-cirrhotic patients (10 months, P=0.006) (Fig. 3A). Cox regression, adjusting for NLR status and prior therapy type, showed that only the latter factor and not NLR was significantly associated with OS (Supplementary Fig. 4A). Regarding PFS, non-cirrhotic NLR-L patients showed significantly higher PFS (15 months) than non-cirrhotic NLR-H patients (4 months, P=0.01, Fig. 3B). Cox regression analysis for PFS, adjusting for NLR and prior therapy type, showed that only prior treatment was significantly associated with PFS (Supplementary Fig. 4B). In our final analysis, we separated our non-cirrhotic patients in 2 groups according to prior HCC therapy status. Supplementary Table 4 shows the baseline characteristics of the 2 groups, which were comparable in terms of most factors, except for BCLC stage on treatment onset (P=0.03) and baseline NLR status (P=0.017). Those 2 groups differed significantly in terms of OS (30 months in patients with prior therapy vs. 8 months in patients without prior therapy, P<0.001) and PFS (24 months vs. 4 months, respectively, P<0.001, Fig. 3C,D). Cox regression analysis showed that only prior therapy, and not NLR or BCLC status, was significantly associated with OS in non-cirrhotic patients, and the same results also applied to PFS (Supplementary Fig. 5A,B). Lastly, we conducted Cox regression Kaplan–Meier analysis in order to present survival results (for both OS and PFS) at the mean of covariates, both for cirrhotic patients (using NLR as grouping variable) and for non-cirrhotic patients (using NLR and prior therapy separately as grouping variables). Results are presented in Supplementary Fig. 6,7.

thumblarge

Figure 3 Overall survival (A) and progression-free survival (B) in non-cirrhotic patients according to baseline NLR status. Overall survival (C) and progression-free survival (D) in non-cirrhotic HCC patients according to prior therapy status

OS, overall survival; PFS, progression-free survival; NLR-H, high NLR group; NLR-L, low NLR group

Discussion

It is noticeable that the majority of randomized trials, as well as real-world data in the HCC management field, do not separate cirrhotic from non-cirrhotic patients in their analysis of advanced HCC, despite the major impact of liver disease severity on the final clinical outcome [8,35,36]. Furthermore, non-cirrhotic patients in most of these studies are wrongly categorized as CP-A, considering that CP only applies to cirrhotic patients. Therefore, clinical studies of HCC should carefully distinguish between cirrhotics and non-cirrhotics when recruiting HCC patients, as survival outcomes may be affected by the presence of cirrhosis. In this study, we used easily obtained biomarkers, such as pre-treatment NLR, and recorded medical data, such as prior HCC treatment, in order to find possible correlations with survival in cirrhotic/non-cirrhotic patients.

In our study, we evaluated the efficacy of atezolizumab–bevacizumab in terms of OS and PFS, in a mixed population of cirrhotic and non-cirrhotic patients with unresectable HCC, according to baseline NLR. As expected, both OS and PFS were worse in cirrhotic compared to non-cirrhotic patients, and cirrhotic patients with high NLR values exhibited the worst outcomes (median OS 5 months, median PFS 2 months), irrespective of the other baseline parameters that could influence the survival outcome. Cirrhosis and high NLR values have been related to worse survival outcomes in untreated as well as treated HCC patients with all treatment modalities [22,37], including atezolizumab–bevacizumab therapy [23,24]. Both reports concerning the prognostic impact of NLR in patients treated with atezolizumab–bevacizumab [23,24] referred to fewer than 50 treated patients, without any histological definition of cirrhosis in most of them, while only 5-12.5% of patients were CP-B stage, and the studies had relatively short observation periods. In our study, high NLR values (>3) were an independent predictor of OS but not PFS, among histologically proven cirrhotic patients with HCC (23.3% in CP-B stage) treated with atezolizumab–bevacizumab. Several reports have highlighted the lack of a strong correlation between tumor-based endpoints, such as objective responses, and/or PFS and OS, especially among cirrhotic patients with advanced HCC under systemic treatment [38-40]. It has been suggested that the surrogacy of PFS with OS is variable, and mainly dependent on systemic treatment class (TKIs or immunotherapy-based) and evaluation treatment point [40].

In our study, we found that cirrhosis was a statistically significant factor for worse OS, as anticipated, along with high NLR values, heralding the importance of the separate evaluation of NLR status in cirrhotic and non-cirrhotic patients. As for PFS, in the total population of patients, only high NLR was independently associated with worse PFS; this suggests that NLR could be a very useful tool for predicting tumor response to immunotherapy, independently of cirrhosis and other confounding factors. Median OS was 12 months in cirrhotic patients with low baseline NLR, compared to only 5 months in those with high baseline NLR, a finding that was statistically significant. It is important to note that previous locoregional therapy (n=8) or resection (n=4) did not affect the survival rates of cirrhotic HCC patients (5-12 months for OS and 2-8 months for PFS).

NLR was not found to be an independent predictor of OS and PFS among non-cirrhotic HCC patients treated with atezolizumab–bevacizumab therapy. It is very interesting to note that 10 of the 27 non-cirrhotic HCC patients in our study population had been previously resected, and early disease recurrence was observed within 2 years from the time of resection, whereas locoregional therapy with incomplete response presented in 9 of 27 patients, prior to the initiation of atezolizumab–bevacizumab therapy. In the multivariate analysis, prior therapy, either resection or locoregional, was an independent predictor of long-term OS and PFS, among treated non-cirrhotic HCC patients. A beneficial reshaping of the tumor microenvironment by prior locoregional or surgical therapy has been suggested in many recently published studies, and trials evaluating combinations of these procedures with immunotherapy are already ongoing. Through this observational study, it could be supposed that prior therapy might possibly affect baseline NLR values prior to the start of immunotherapy, shifting the tumor microenvironment balance towards an immune-friendly environment before the initiation of immunotherapy, given that the majority of non-cirrhotic HCC patients (12/13, 92.3%) with a low baseline NLR had been previously treated with surgery or locoregional treatment, compared to only 50% (7/14, P=0.016) of those with high baseline NLR. This finding needs further investigation and evaluation in larger cohorts.

This study had some limitations. The small number of patients in each group could have led to biases, along with the retrospective analysis of the data. However, the grade of statistical significance of our values, in accordance with results from other studies [23,24], might suggest that NLR should be tested in cohorts of large randomized prospective trials to confirm its prognostic value in relation to survival. Another limitation is the small proportion of viral non-cirrhotic patients. This could be possibly explained by the increasing prevalence of steatotic liver disease in Europe, leading to the appearance of HCC in non-cirrhotic livers. On the other hand, this study was conducted in a single HCC referral center, where laboratory examinations were conducted by the same laboratory, and the same HCC specialists cooperated in multidisciplinary patient management, gathering of patient data and presentation of the results.

Summary Box

What is already known:

  • Hepatocellular carcinoma (HCC) is the 6th most common cancer and the 4th leading cause of cancer-related deaths globally

  • Early-stage diagnosis is rare, with most cases diagnosed at intermediate or advanced stages, limiting treatment options

  • Sorafenib was the first Food & Drug Administration-approved systemic treatment for advanced HCC, followed by other tyrosine kinase inhibitors and, more recently, the combination of atezolizumab and bevacizumab, which have shown better survival outcomes and quality of life in patients with unresectable HCC

  • The neutrophil-to-lymphocyte ratio (NLR) has been associated with better survival and response to immunotherapy in solid tumors, including HCC

What the new findings are:


  • Cirrhotic patients with high baseline NLR values (>3) had significantly worse overall survival (OS) and progression-free survival (PFS), with a median OS of 5 months and PFS of 2 months

  • High NLR was found to be an independent predictor of OS, but not PFS, in cirrhotic patients

  • Non-cirrhotic patients with low baseline NLR had the best survival outcomes (30 months OS and 15 months PFS), and prior locoregional/surgical therapy (though not NLR) was an independent predictor of better prognosis in non-cirrhotic patients

References

1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018:GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68:394-424.

2. Asafo-Agyei KO, Samant H. Hepatocellular Carcinoma. 2023 Jun 12. In:StatPearls [Internet]. Treasure Island (FL):StatPearls Publishing;2025 Jan–.

3. Llovet JM, BrúC, Bruix J. Prognosis of hepatocellular carcinoma:the BCLC staging classification. Semin Liver Dis 1999;19:329-338.

4. Llovet JM, Ricci S, Mazzaferro V, et al;SHARP Investigators Study Group. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 2008;359:378-390.

5. Yamashita T, Kudo M, Ikeda K, et al. REFLECT-a phase 3 trial comparing efficacy and safety of lenvatinib to sorafenib for the treatment of unresectable hepatocellular carcinoma:an analysis of Japanese subset. J Gastroenterol 2020;55:113-122.

6. Bruix J, Qin S, Merle P, et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE):a randomised, double-blind, placebo-controlled, phase 3 trial.Lancet 2017;389:56-66.

7. Abou-Alfa GK, Meyer T, Cheng AL, et al. Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. N Engl J Med 2018;379:54-63.

8. Finn RS, Qin S, Ikeda M, et al;IMbrav150 Investigators. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med 2020;382:1894-1905.

9. Scheiner B, Pomej K, Kirstein MM, et al. Prognosis of patients with hepatocellular carcinoma treated with immunotherapy - development and validation of the CRAFITY score. J Hepatol 2022;76:353-363.

10. Cammarota A, Zanuso V, Manfredi GF, Murphy R, Pinato DJ, Rimassa L. Immunotherapy in hepatocellular carcinoma:how will it reshape treatment sequencing?Ther Adv Med Oncol 2023;15:17588359221148029.

11. Manfredi GF, Celsa C, John C, et al. Mechanisms of resistance to immunotherapy in hepatocellular carcinoma. J Hepatocell Carcinoma 2023;10:1955-1971.

12. Pinto E, Meneghel P, Farinati F, Russo FP, Pelizzaro F, Gambato M. Efficacy of immunotherapy in hepatocellular carcinoma:does liver disease etiology have a role?Dig Liver Dis 2024;56:579-588.

13. Proctor MJ, McMillan DC, Morrison DS, Fletcher CD, Horgan PG, Clarke SJ. A derived neutrophil to lymphocyte ratio predicts survival in patients with cancer. Br J Cancer 2012;107:695-699.

14. Buonacera A, Stancanelli B, Colaci M, Malatino L. Neutrophil to lymphocyte ratio:an emerging marker of the relationships between the immune system and diseases. Int J Mol Sci 2022;23:3636.

15. Wang W, Tong Y, Sun S, et al. Predictive value of NLR and PLR in response to preoperative chemotherapy and prognosis in locally advanced gastric cancer. Front Oncol 2022;12:936206.

16. Bryant AK, Sankar K, Strohbehn GW, et al. Prognostic and predictive value of neutrophil-to-lymphocyte ratio with adjuvant immunotherapy in stage III non-small-cell lung cancer. Lung Cancer 2022;163:35-41.

17. Hamid HKS, Davis GN, Trejo-Avila M, Igwe PO, Garcia-Marín A. Prognostic and predictive value of neutrophil-to-lymphocyte ratio after curative rectal cancer resection:A systematic review and meta-analysis. Surg Oncol 2021;37:101556.

18. Gao S, Tang W, Zuo B, Mulvihill L, Yu J, Yu Y. The predictive value of neutrophil-to-lymphocyte ratio for overall survival and pathological complete response in breast cancer patients receiving neoadjuvant chemotherapy. Front Oncol 2022;12:1065606.

19. Zhou D, Luan J, Huang C, Li J. Tumor-associated macrophages in hepatocellular carcinoma:friend or foe?Gut Liver 2021;15:500-516.

20. Sas Z, Cendrowicz E, Weinhäuser I, Rygiel TP. Tumor microenvironment of hepatocellular carcinoma:challenges and opportunities for new treatment options. Int J Mol Sci 2022;23:3778.

21. Arvanitakis K, Koletsa T, Mitroulis I, Germanidis G. Tumor-associated macrophages in hepatocellular carcinoma pathogenesis, prognosis and therapy. Cancers (Basel) 2022;14:226.

22. Lin S, Hu S, Ran Y, Wu F. Neutrophil-to-lymphocyte ratio predicts prognosis of patients with hepatocellular carcinoma:a systematic review and meta-analysis. Transl Cancer Res 2021;10:1667-1678.

23. Eso Y, Takeda H, Taura K, Takai A, Takahashi K, Seno H. Pretreatment neutrophil-to-lymphocyte ratio as a predictive marker of response to atezolizumab plus bevacizumab for hepatocellular carcinoma. Curr Oncol 2021;28:4157-4166.

24. Wu YL, Fulgenzi CAM, D'Alessio A, et al. Neutrophil-to-lymphocyte and platelet-to-lymphocyte ratios as prognostic biomarkers in unresectable hepatocellular carcinoma treated with atezolizumab plus bevacizumab. Cancers (Basel) 2022;14:5834.

25. Qin S, Chen M, Cheng AL, et al;IMbrav050 investigators. Atezolizumab plus bevacizumab versus active surveillance in patients with resected or ablated high-risk hepatocellular carcinoma (IMbrav050):a randomised, open-label, multicentre, phase 3 trial. Lancet 2023;402:1835-1847.

26. 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.

27. Slovak R, Ludwig JM, Gettinger SN, Herbst RS, Kim HS. Immuno-thermal ablations - boosting the anticancer immune response. J Immunother Cancer 2017;5:78.

28. Simonetti RG, CammàC, Fiorello F, Politi F, D'Amico G, Pagliaro L. Hepatocellular carcinoma. A worldwide problem and the major risk factors. Dig Dis Sci 1991;36:962-972.

29. Samant H, Amiri HS, Zibari GB. Addressing the worldwide hepatocellular carcinoma:epidemiology, prevention and management. J Gastrointest Oncol 2021;12:S361-S373.

30. Akinyemiju T, Abera S, Ahmed M, et al;Global Burden of Disease Liver Cancer Collaboration. The burden of primary liver cancer and underlying etiologies from 1990 2015 at the global, regional, and national level:results from the Global Burden of Disease study 2015. JAMA Oncol 2017;3:1683-1691.

31. Kamath PS, Wiesner RH, Malinchoc M, et al. A model to predict survival in patients with end-stage liver disease. Hepatology 2001;33:464-470.

32. 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;33:550-558.

33. Rebouissou S, Nault JC. Advances in molecular classification and precision oncology in hepatocellular carcinoma. J Hepatol 2020;72:215-229.

34. Lencioni R, Llovet JM. Modified RECIST (mRECIST) assessment for hepatocellular carcinoma. Semin Liver Dis 2010;30:52-60.

35. Kelley RK, Rimassa L, Cheng AL, et al. Cabozantinib plus atezolizumab versus sorafenib for advanced hepatocellular carcinoma (COSMIC-312):a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2022;23:995-1008.

36. Abou-Alfa GK, Lau G, Kudo M, et al. Tremelimumab plus durvalumab in unresectable hepatocellular carcinoma. NEJM Evid 2022;1:EVIDoa2100070.

37. Mouchli M, Reddy S, Gerrard M, Boardman L, Rubio M. Usefulness of neutrophil-to-lymphocyte ratio (NLR) as a prognostic predictor after treatment of hepatocellular carcinoma.“Review article. Ann Hepatol 2021;22:100249.

38. Merino M, Kasamon Y, Theoret M, Pazdur R, Kluetz P, Gormley N. Irreconcilable differences:the divorce between response rates, progression-free survival, and overall survival. J Clin Oncol 2023;41:2706-2712.

39. Llovet JM, Montal R, Villanueva A. Randomized trials and endpoints in advanced HCC:role of PFS as a surrogate of survival. J Hepatol 2019;70:1262-1277.

40. Cabibbo G, Celsa C, Enea M, et al. Progression-free survival early assessment is a robust surrogate endpoint of overall survival in immunotherapy trials of hepatocellular carcinoma. Cancers (Basel) 2020;13:90.

Notes

Conflict of Interest: None