Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Objective

To explore changing trends in circulating immune indicators of hepatocellular carcinoma (HCC) undergoing TACE plus immune checkpoint inhibitors (ICIs) and anti-VEGF antibodies/TKIs and to elucidate the relationship between immune response and tumor prognosis.

Materials

This single-center retrospective study included patients with unresectable HCC undergoing TACE plus ICIs and anti-VEGF antibodies/TKIs from March 11, 2019, to February 15, 2024. Peripheral blood samples were collected at baseline and every cycle, from which blood cell counts and immune indicators were analyzed. The primary outcome was the objective response rate (ORR) at the first evaluation. According to the first evaluation based on mRECIST, patients were classified into PD, SD, and OR groups for analysis. Further subgroup analysis was performed on the OR group based on whether experiencing progression after the first evaluation. Lymphocyte subsets were measured by flow cytometry. Immunoglobulins were measured using the immune turbidimetric method. The neutrophil-to-lymphocyte ratio (NLR) was measured by the complete blood count. Simple linear regression was employed to examine the dynamic trends.

Results

A total of 63 patients were enrolled, with an ORR of 55.6% and a disease control rate (DCR) of 87.3% at the first evaluation. The median overall survival (mOS) was 27.5 months (95% CI: 22.5-32.5 months). In the OR group (n=35), more active immune responses, expressed in a decrease in CD3-CD19+ (p=0.004), CFB (p=0.027), NLR (p<0.001) and an increase in Ig λ (p=0.010), Ig κ (p=0.037), Ig A (p=0.005), Ig G (p=0.006), were related to better prognosis, while similar patterns seen in the OR-nPD subgroup. Concurrently, no significant differences were noted in the PD group (n=8).

Conclusion

The combination therapy may modify the tumor microenvironment of HCC. Changing trends in circulating immune indicators and NLR can serve as potential biomarkers for predicting tumor response and guiding clinical treatment.

Free full text 


Logo of jhepcDove Medical PressThis ArticleSubscribeSubmit a ManuscriptSearchFollowDovepressJournal of Hepatocellular Carcinoma
J Hepatocell Carcinoma. 2024; 11: 2019–2032.
Published online 2024 Oct 22. https://doi.org/10.2147/JHC.S487472
PMCID: PMC11512558
PMID: 39465041

Immune Indicator Changes in Hepatocellular Carcinoma Undergoing TACE Plus ICIs and Anti-VEGF Antibodies/TKIs: A Prognostic Biomarker Analysis

Abstract

Objective

To explore changing trends in circulating immune indicators of hepatocellular carcinoma (HCC) undergoing TACE plus immune checkpoint inhibitors (ICIs) and anti-VEGF antibodies/TKIs and to elucidate the relationship between immune response and tumor prognosis.

Materials

This single-center retrospective study included patients with unresectable HCC undergoing TACE plus ICIs and anti-VEGF antibodies/TKIs from March 11, 2019, to February 15, 2024. Peripheral blood samples were collected at baseline and every cycle, from which blood cell counts and immune indicators were analyzed. The primary outcome was the objective response rate (ORR) at the first evaluation. According to the first evaluation based on mRECIST, patients were classified into PD, SD, and OR groups for analysis. Further subgroup analysis was performed on the OR group based on whether experiencing progression after the first evaluation. Lymphocyte subsets were measured by flow cytometry. Immunoglobulins were measured using the immune turbidimetric method. The neutrophil-to-lymphocyte ratio (NLR) was measured by the complete blood count. Simple linear regression was employed to examine the dynamic trends.

Results

A total of 63 patients were enrolled, with an ORR of 55.6% and a disease control rate (DCR) of 87.3% at the first evaluation. The median overall survival (mOS) was 27.5 months (95% CI: 22.5–32.5 months). In the OR group (n=35), more active immune responses, expressed in a decrease in CD3CD19+ (p=0.004), CFB (p=0.027), NLR (p<0.001) and an increase in Ig λ (p=0.010), Ig κ (p=0.037), Ig A (p=0.005), Ig G (p=0.006), were related to better prognosis, while similar patterns seen in the OR-nPD subgroup. Concurrently, no significant differences were noted in the PD group (n=8).

Conclusion

The combination therapy may modify the tumor microenvironment of HCC. Changing trends in circulating immune indicators and NLR can serve as potential biomarkers for predicting tumor response and guiding clinical treatment.

Keywords: immune indicator, TACE, immune checkpoint inhibitors, tyrosine kinase inhibitors, tumor microenvironment

Introduction

Hepatocellular carcinoma (HCC) is characterized by high incidence and mortality rates, with approximately half of the global new cases and deaths occurring in China each year.1 HCC often presents insidiously, and over 80% of patients are diagnosed at an advanced stage, resulting in a low curative resection rate and poor prognosis.2

According to the Barcelona Clinic Liver Cancer (BCLC) staging system, transarterial chemoembolization (TACE) is the recommended standard treatment for patients with intermediate HCC and is also widely used for advanced HCC.3,4 TACE works by embolizing the tumor blood supply, leading to tumor ischemia and hypoxia, thereby inhibiting tumor progression while chemotherapy drugs can be administered locally and accurately.5 However, the resultant hypoxic environment can stimulate angiogenic pathways, such as vascular endothelial growth factor (VEGF), reducing the number and function of immune cells. Combining immune checkpoint inhibitors (ICIs) and tyrosine kinase inhibitors (TKIs) can improve the hypoxic and immunosuppressive tumor microenvironment (TME) and inhibit tumor angiogenesis.6 Anti-VEGF plays its role as an anti-vascular, anti-angiogenic, or anti-permeability factor in preventing tumor growth and metastasis.7 Therefore, the combination therapy of TACE plus ICIs and anti-VEGF antibodies/TKIs holds the potential for further improving the efficacy of treatment for unresectable HCC (uHCC).8,9 Some studies, such as EMERALD-1, CHANCE001, and CHANCE2201, have further demonstrated that this combination therapy significantly improves the prognosis of patients with uHCC.10–12

The liver, being an immune-privileged organ, often exhibits a lack or even absence of tumor-infiltrating lymphocytes in both the HCC tumor nests and surrounding stroma, making it difficult to generate functional tumor-specific T cells and thus an effective anti-tumor immune response.13–15 TACE can reduce the density of immune-exhausted effector cells and regulatory T cells within the tumor.16 The combination of ICIs and TKIs can further activate T cells.17 Through TACE plus ICIs and anti-VEGF antibodies/TKIs, a non-immunogenic tumor, lacking immune effector cells, can be transformed into an immunogenic tumor with immune effector cell infiltration.8 There are few reports on how various humoral and cellular immune indicators in the peripheral circulation change in highly heterogeneous HCC patients undergoing this combination therapy.

In this study, we aim to explore the changes in immune indicators in the peripheral circulation of HCC patients undergoing this combination therapy and attempt to elucidate the relationship between immune response and tumor prognosis.

Materials and Methods

Patient Selection

This retrospective study was approved by the Institutional Review Board and the Ethics Committee. Since the study was retrospective, informed consent was not required. From March 11, 2019, to February 15, 2024, patients with uHCC who underwent TACE plus ICIs and anti-VEGF antibodies/TKIs were included. The inclusion criteria were as follows: (1) histologically or clinically diagnosed with HCC according to the American Association for the Study of Liver Diseases (AASLD) guidelines;18 (2) BCLC stage B or C; (3) Child-Pugh class A or B, without uncontrollable ascites or hepatic encephalopathy; (4) ECOG performance status of 0 or 1; (5) treatment with TACE plus ICIs and anti-VEGF antibodies/TKIs. The criteria for the timing of combination therapy were defined as simultaneous use of the initial TACE and the first ICI treatment or within a 3-month interval, with TKIs used simultaneously with TACE or ICIs. At least one cycle of ICI treatment should be used after TACE. (6) having complete blood cell count and immune indicators before combination therapy and at the first evaluation, and at least one serological assessment within 4 cycles after combination therapy. The first evaluation is often conducted in the fourth cycle or the third month.

Exclusion criteria were: (1) histologically or clinically confirmed intrahepatic cholangiocarcinoma (ICC), mixed hepatocellular carcinoma, sarcomatoid hepatocellular carcinoma, or fibrolamellar hepatocellular carcinoma; (2) concomitant other malignant tumors; (3) prior systemic treatment with ICIs, anti-VEGF antibodies /TKIs or chemotherapy; (4) incomplete blood or immune indicators that preclude data analysis; (5) follow up time less than 3 months after the combination therapy.

TACE Procedure

The TACE procedures were performed by two interventional radiologists with at least 10 years of experience. Precision TACE was used to reduce the heterogeneity and improve the embolization effect.19 The decision to perform “on-demand” TACE was based on the results of tumor markers and radiological examinations. “On-demand” TACE was discontinued if any of the following conditions occurred: (1) Child-Pugh class C (uncontrollable ascites, severe jaundice, overt hepatic encephalopathy, or hepatorenal syndrome); (2) ECOG score >2; (3) continuous progression of target lesions after three TACE sessions according to the modified Response Evaluation Criteria in Solid Tumors (mRECIST).20

ICIs and Anti-VEGF Antibodies/TKIs Administration

All patients included in the study received ICIs and anti-VEGF antibodies/TKIs combination therapy, all of which were approved by the National Medical Products Administration and available in China. Anti-VEGF antibodies, like bevacizumab, were administered concurrently with ICIs. All oral TKIs were interrupted for two or three days before and after TACE. All drugs were administered based on the guidelines and availability in China following their standard dose and frequency. ICIs and anti-VEGF antibodies/TKIs treatment continued until disease progression or unacceptable toxic effects.

Follow-Up

The primary outcome was the objective response rate (ORR) at the first evaluation. After combination therapy, follow-up evaluations were conducted every 6–9 weeks, including imaging examinations (enhanced abdominal MRI and chest-abdomen plain CT scans) and laboratory examinations (complete blood cell count, comprehensive biochemical analysis, tumor markers, HBV-DNA load analysis, cytokines, immunity index, chest pain panel, and comprehensive thyroid function tests). ICIs were administered every three weeks, with laboratory tests (complete blood cell count, comprehensive biochemical analysis, immunity index, chest pain panel, and comprehensive thyroid function tests) conducted before each cycle. Tumor response was assessed according to mRECIST. The last follow-up date was May 15, 2024.

Explore Systemic Immune Response in Peripheral Blood

Peripheral blood samples were collected before TACE and every three weeks before ICI treatment. Immune indicators include the percentage of CD3+ cells, CD3+CD4+ cells, CD3+CD8+ cells, CD3CD (16+56)+ cells, and CD3CD19+ cells; the CD4+/CD8+ ratio; and the counts of immunoglobulin λ (Ig λ), immunoglobulin κ (Ig κ), complement 4 (C4), complement 3 (C3), immunoglobulin M (Ig M), immunoglobulin A (Ig A), immunoglobulin G (Ig G), and complement B factors (CFB). The neutrophil-to-lymphocyte ratio (NLR) is also included. Immune and peripheral blood indicators were completely collected before combination treatment and each cycle.

The peripheral blood mononuclear cells (PBMCs) were isolated using the Ficoll-Paque method and resuspended at a concentration of 1×10^6 cells/mL. PBMCs were collected and analyzed using flow cytometry (FACScan Caliber, Becton Dickinson, Franklin Lakes, NJ, USA). Immunoglobulins and complements were measured by the immune turbidimetric method. NLR was measured by the complete blood count.

The initial treatment efficacy and tumor response were first assessed at the fourth cycle or the end of the third month according to mRECIST. Compare the baseline indicators of all patients with those at the first evaluation. Classify all patients into PD, SD, and OR (PR+CR) groups based on the first evaluation. Do inter-group and intra-group comparisons for each group. Within the OR group, further classify patients into OR-PD and OR-non-PD (OR-nPD) subgroups based on whether experiencing progression after the first evaluation. Collect immune and peripheral blood indicators at the time of progression. Compare the indicators of both subgroups at the different time points respectively. (Figure 1).

An external file that holds a picture, illustration, etc.
Object name is JHC-11-2019-g0001.jpg

Flowchart of study enrollment.

Statistical Analysis

Continuous variables are expressed as means with 95% confidence intervals. For categorical variables, counts and percentages are presented. Shapiro–Wilk tests were performed to determine the normality of the data distribution. For data that followed a normal distribution, significance testing for differences was conducted using either the chi-square test or the two-tailed paired Student’s t-test. For data that did not follow a normal distribution, the Kruskal–Wallis test or the nonparametric Mann–Whitney U-test was used to test for significant differences. The dynamic changes in the indicators were examined using simple linear regression analysis. Statistics were judged to be significant when p values were less than 0.05. All statistical analyses were performed using SPSS (version 26.0; IBM, Somers, NY) and GraphPad Prism software (version 9.0.2; GraphPad Software, CA, https://www.graphpad.com/).

Results

Patient Characteristics

From March 11, 2019, to February 15, 2024, a total of 163 uHCC patients undergoing TACE plus ICIs and anti-VEGF antibodies/TKIs were screened. Finally, 63 patients with complete follow-up data who met the inclusion criteria were enrolled. Based on the first evaluation, patients were categorized into three groups: PD group (8/63), SD group (20/63), and OR group (35/63), whereas 1 case of CR and 34 cases of PR were included in the OR group. The characteristics of each group were summarized in Table 1.

Table 1

Patient Baseline Characteristics of Each Groups

CharacteristicsOverall (n=63)PD (n=8)SD (n=20)OR (n=35)p value
Age(years)60(57,62)55(43,67)60(55,65)60(57,64)0.38
Gender0.03
 Male52(82.5)8(100.0)13(65.0)31(88.6)
 Female11(17.5)0(0.0)7(35.0)4(11.4)
HBV0.89
 Yes51(81.0)7(87.5)16(80.0)28(80.0)
 No12(19.0)1(12.5)4(20.0)7(20.0)
Cirrhosis0.34
 Yes32(50.8)6(75.0)10(50.0)16(45.7)
 No31(49.2)2(25.0)10(50.0)19(54.3)
AFP0.23
 <400ng/mL43(68.3)5(62.5)11(55.0)27(77.1)
 ≥400ng/mL20(31.7)3(37.5)9(45.0)8(22.9)
PIVKA-II (mAU/mL)8475(4765,12,186)14,277(−1848,30,402)5180(−100,10,460)9033(3783,14,282)0.32
Child-Pugh stage0.33
 A51(81.0)7(87.5)14(70.0)30(77.1)
 B12(19.0)1(12.5)6(30.0)8(22.9)
BCLC stage0.72
 B30(47.6)4(50.0)8(40.0)18(51.4)
 C33(52.4)4(50.0)12(60.0)17(48.6)
ECOG score0.86
 034(54.0)4(50.0)10(50.0)20(57.1)
 129(46.0)4(50.0)10(50.0)15(42.9)
MELD3.63(2.93,4.35)4.07(1.35,6.79)3.29(1.96,4.63)3.74(2.79,4.69)0.77
ALBI−2.29(−2.38,-2.18)−2.27(−2.68,-1.87)−2.25(−2.44,-2.05)−2.31(−2.44,-2.17)0.85
Extrahepatic spread0.64
 Yes16(25.4)1(12.5)6(30.0)9(25.7)
 No47(74.6)7(87.5)14(70.0)26(74.3)
Vascular invasion0.61
 Yes23(36.5)3(37.5)9(45.0)11(31.4)
 No42(63.5)5(62.5)11(55.0)24(68.6)
Up-to-seven criteria0.34
 Within9(14.3)2(25.0)4(20.0)3(8.6)
 Beyond54(85.7)6(75.0)16(80.0)32(91.4)

Abbreviations: HBV, hepatitis B virus; AFP, Alpha-fetoprotein; PIVKA-II, protein induced by vitamin K absence or antagonist-II; BLCL, Barcelona Clinic Liver Cancer; ECOG, Eastern Cooperative Oncology Group; MELD, model for end-stage liver disease score; ALBI, albumin bilirubin score.

Efficacy and Safety

The ORR was 55.6% at the first evaluation, while the disease control rate (DCR) was 87.3%. As of the last follow-up on May 15, 2024, the median follow-up time for the entire cohort was 16.3 months (interquartile range [IQR]: 10.6–25.5 months). The median overall survival (mOS) was 27.5 months (95% confidence interval [CI]: 22.5–32.5 months), and the median progression-free survival (mPFS) was 15.9 months (95% CI: 12.6–19.3 months).

All reported adverse events were mild and well-tolerated, with no treatment-related fatalities during the study. The incidence of adverse events was 61.9% (39/63), including primary fever (59.0%), liver function impairment (20.5%), abdominal pain (48.7%), hand-foot skin reaction (23.1%), hypertension (15.4%), thrombocytopenia (10.3%), proteinuria (17.9%), and immune myocarditis (2.6%).

Systemic Immune Response in Peripheral Blood

Comparisons of Pre-Treatment and Post-Treatment Indicators in the Whole Group

A comparison of baseline indicators with those at the first evaluation revealed significant differences, with a decrease in CD3CD19+ (p=0.009), C4 (p=0.039), CFB (p=0.002), NLR (p=0.006) and an increase in Ig λ (p=0.002), Ig κ (p=0.005), Ig A (p<0.001), Ig G (p=0.001) (Table 2).

Table 2

Comparation Between Baseline and First Evaluation Indicators for All Patients

Baseline-OverallAt First Evaluation-Overallp value
CD3+ (%)72.88(70.46,75.30)73.44(70.44,76.44)0.571
CD3+CD4+ (%)41.74(39.52,43.96)42.01(39.31,44.71)0.750
CD3+CD8+ (%)27.02(24.68,29.36)26.92(24.52,29.33)0.885
CD4+/CD8+1.82(1.55,2.09)1.83(1.58,2.09)0.904
CD3CD(16+56)+ (%)16.67(14.54,18.81)17.72(15.01,20.42)0.419
CD3CD19+ (%)9.80(8.29,11.31)8.20(6.59,9.80)0.009
Ig λ (mg/dl)672.86(611.27,734.44)740.59(684.81,796.37)0.002
Ig κ (mg/dl)1212.59(1120.41,1304.76)1335.95(1246.17,1425.74)0.005
C4 (g/l)0.22(0.20,0.25)0.21(0.19,0.23)0.039
C3 (g/l)0.92(0.86,0.97)0.90(0.85,0.94)0.653
Ig M (g/l)1.23(1.05,1.40)1.26(1.08,1.45)0.584
Ig A (g/l)3.34(2.98,3.69)3.75(3.35,4.16)<0.001
Ig G (g/l)14.15(13.07,15.24)15.61(14.62,16.60)0.001
CFB (mg/dl)45.10(41.90,48.31)40.66(38.17,43.15)0.002
NLR5.72(4.62,6.83)3.78(2.72,4.83)0.006

Inter-Group and Intra-Group Comparisons for Each Group

When comparing among PD, SD, and OR groups, only CD3CD19+ showed a significant difference at baseline (p=0.019). Further intra-group comparisons revealed no significant differences in the PD group, while the SD group showed significant differences in CFB (p=0.008) and NLR (p=0.037). The OR group exhibited significant differences, similar to those in the whole group, with a decrease in CD3CD19+ (p=0.004), CFB (p=0.027), NLR (p<0.001), and an increase in Ig λ (p=0.010), Ig κ (p=0.037), Ig A (p=0.005), Ig G (p=0.006) (Table 3).

Table 3

Inter-Group and Intra-Group Comparisons for Each Group

Baselinep1 valueAt First Evaluationp2 valuep3 value
CD3+ (%)0.9160.647
 PD73.75(66.53,80.98)74.13(64.26,84.01)0.875
 SD73.31(68.31,78.31)71.36(64.91,77.82)0.307
 OR72.44(69.20,75.67)74.47(70.70,78.24)0.064
CD3+CD4+ (%)0.4680.303
 PD39.14(34.27,44.02)39.56(32.94,46.19)0.851
 SD40.76(36.73,44.78)39.70(34.79,44.61)0.439
 OR42.89(39.65,46.14)43.89(40.02,47.77)0.425
CD3+CD8+ (%)0.1480.298
 PD32.32(23.65,41.00)31.82(24.69,38.96)0.744
 SD27.76(23.73,31.78)26.54(21.90,31.18)0.332
 OR25.38(22.24,28.53)26.03(22.78,29.27)0.465
CD4+/CD8+0.1410.255
 PD1.35(0.89,1.80)1.32(0.94,1.70)0.777
 SD1.64(1.28,2.01)1.72(1.30,2.13)0.313
 OR2.04(1.61,2.47)2.02(1.62,2.41)0.572
CD3CD(16+56)+ (%)0.3280.485
 PD18.04(12.64,23.43)19.66(11.07,28.25)0.499
 SD18.62(14.19,23.06)19.22(14.23,24.21)0.765
 OR15.24(12.42,18.07)16.42(12.63,20.20)0.342
CD3CD19+ (%)0.0190.484
 PD7.53(4.59,10.46)5.67(3.53,7.81)0.143
 SD7.44(5.49,9.38)4.28(1.36,7.19)0.412
 OR11.67(9.36,13.99)8.48(6.57,10.40)0.004
Ig λ (mg/dl)0.0640.095
 PD531.13(382.36,679.89)610.38(437.51,783.24)0.139
 SD773.60(644.22,902.98)808.25(690.03,926.47)0.319
 OR647.69(572.05,723.32)731.69(664.52,798.85)0.010
Ig κ (mg/dl)0.5670.586
 PD1141.75(852.18,1431.32)1234.13(999.70,1468.55)0.441
 SD1286.80(1113.61,1459.99)1396.75(1217.72,1575.78)0.080
 OR1186.37(1058.80,1313.94)1324.49(1201.08,1447.89)0.037
C4 (g/l)0.3290.052
 PD0.26(0.17,0.36)0.30(0.16,0.43)0.388
 SD0.22(0.19,0.25)0.21(0.18,0.23)0.144
 OR0.22(0.18,0.26)0.19(0.17,0.22)0.067
C3 (g/l)0.9770.528
 PD0.90(0.72,1.07)0.86(0.68,1.04)0.594
 SD0.92(0.82,1.02)0.87(0.78,0.96)0.146
 OR0.92(0.83,1.00)0.92(0.85,0.99)0.590
Ig M (g/l)0.6860.389
 PD1.49(0.40,2.58)1.43(0.35,2.51)0.310
 SD1.08(0.85,1.30)1.09(0.85,1.33)0.533
 OR1.26(1.05,1.47)1.33(1.10,1.55)0.416
Ig A (g/l)0.4150.944
 PD3.10(2.38,3.82)3.61(2.85,4.37)0.065
 SD3.78(2.96,4.59)4.01(3.04,4.97)0.268
 OR3.14(2.70,3.58)3.64(3.15,4.14)0.005
Ig G (g/l)0.2950.252
 PD12.70(9.01,16.39)13.92(10.36,17.48)0.255
 SD15.40(13.29,17.50)16.59(14.65,18.53)0.058
 OR13.77(12.36,15.19)15.44(14.18,16.71)0.006
CFB (mg/dl)0.4800.426
 PD41.00(30.32,51.68)41.79(34.22,49.35)0.838
 SD44.02(38.90,49.14)38.25(33.84,42.66)0.008
 OR46.66(41.95,51.37)41.77(38.22,45.33)0.027
NLR0.8220.075
 PD4.56(2.33,6.79)6.10(1.99,10.21)0.327
 SD5.77(4.02,7.53)4.28(1.36,7.19)0.037
 OR5.96(4.23,7.69)2.96(2.34,3.58)<0.001

Subgroup Analysis Within the OR Group

The OR group was further divided into OR-PD (16/35) and OR-nPD (19/35) subgroups. Subsequent indicator comparisons in the OR-PD subgroup showed no significant differences among baseline, first evaluation, and progression points (Figure 2, Tables S1 and S2).

An external file that holds a picture, illustration, etc.
Object name is JHC-11-2019-g0002.jpg

The distribution of indicators at different timepoints in the OR-PD subgroup.

Note: There was no significant difference in any indicator at different timepoints in the OR-PD subgroup.

Intriguingly, similar trends were observed in the OR-nPD subgroup with a decrease in CD3CD19+ (p=0.001), C4 (p=0.040), CFB (p=0.029), NLR (p=0.001), and an increase in CD3+ (p=0.013), Ig λ (p=0.004), Ig κ (p=0.021), Ig A (p=0.005), Ig G (p=0.003). In contrast, the OR-PD subgroup showed no significant differences (Figure 3, Table S2).

An external file that holds a picture, illustration, etc.
Object name is JHC-11-2019-g0003.jpg

The distribution of baseline and first evaluation indicators in the OR-nPD subgroup.

Note: *p<0.05; **p<0.01; ***p<0.001; nsp>0.05.

Simple Linear Regression Analysis

For indicators showing significant differences in the OR-nPD subgroup, simple linear regression analysis revealed dynamic changes, including CD3CD19+ (Y = −1.063*X + 10.98, p=0.0498), Ig λ (Y = 37.47*X + 672.7, p=0.0492), Ig A (Y = 0.2174*X + 3.276, p=0.0411) and NLR (Y = −0.6941*X + 5.595, p=0.0038) (Figure 4).

An external file that holds a picture, illustration, etc.
Object name is JHC-11-2019-g0004.jpg

Simple linear regression analysis for statistically significant indicators in the OR-nPD subgroup.

Discussion

This retrospective study demonstrated the safety and efficacy of the combination therapy of TACE plus ICIs and anti-VEGF antibodies/TKIs for uHCC patients, with an ORR of 55.6% and a DCR of 87.3% at the first evaluation. This combination therapy may enhance systemic immune responses, with changes in circulating immune indicators linked to prognosis. It indicated that more active immune responses may correlate with better prognosis, expressed in a decrease in CD3CD19+, C4, CFB, NLR and an increase in CD3+, Ig λ, Ig κ, Ig A, Ig G. These changes can potentially guide personalized treatment strategies by identifying patients who are more likely to benefit from this combined therapy.

In previous studies, low levels of CD3+ cells have been confirmed as an adverse prognostic factor.21–24 In the OR group, especially in the OR-nPD subgroup, a similar trend can be observed: higher levels of CD3+ and CD4+ cells suggest improved immune status and better prognosis. Conversely, during recurrence or metastasis, CD3+ and CD4+ cell levels are significantly reduced, although not statistically significant. Unlike previous studies, changes in CD4/CD8 ratio and CD8+ cells did not show specific patterns related to prognosis.25–27 Rather than solely the frequency of CD3+, CD8+, and CD4+ lymphocytes, biomarkers preferentially expressed on tumor-reactive lymphocytes, such as PD-1hi, CD39, CXCL13, CD103, or the co-expression of PD-1hi and CXCL13, have more predictive value.28 Nonetheless, it is clear that the frequency of CD3+ was positively correlated with prognosis.

In this study, the frequency of B cells was also associated with the progression of HCC patients undergoing combination therapy. Consistent with previous findings, the number of B cells in HCC patients achieving disease remission was significantly reduced compared to those with disease progression (p=0.004).29 Although not statistically significant, circulating B cell counts tended to increase at the time of progression in the OR-PD subgroup. In other words, B cell is possibly another negative regulatory factor.30 The antibodies secreted by B cells can independently activate innate immune responses and induce the cancer immunity cycle.31 B cells migrate from the bone marrow to secondary lymphoid organs, where they undergo activation upon encountering antigens. This activation process includes class switching, during which B cells change the type of immunoglobulin.32 Therefore, in this study, a decrease in circulating B cells was observed to be accompanied by an increase in IgG, IgA, Ig κ, and Ig λ.

Previous studies have used RNA sequencing (RNA-seq) to analyze the whole transcriptome of shCFB (knockdown) cells and shControl (mock) cells, concluding that CFB itself is an early initiator of tumorigenesis and suggesting that increased CFB expression may be an indicator of pancreatic cancer development.33,34 In this study, both the OR group (p=0.027) and OR-nPD subgroup (p=0.029) showed a decreasing trend in CFB expression. This indicates that a decrease in CFB was associated with a better prognosis. Besides CFB, complement factors like C3 and C5 are involved in tumor development.35,36 However, the role of C4 in tumor progression is less studied, likely due to its early involvement in the classical complement pathway. Markiewski et al injected TC-1 tumor cells subcutaneously into mice and found that mice lacking C3 or C4 showed significantly reduced tumor proliferation compared to mice with adequate levels of C3 and C4.37 This suggests that C4 can infect tumor progression.

Elevated NLR, resulting from increased neutrophil count and decreased lymphocyte count, has been associated with poor prognosis in certain malignant tumors.38–41 The TME, involving inflammatory cells, plays a crucial role in tumor formation while NLR serves as an effective indicator of systemic inflammatory response.42 Wu et al reported that NLR ≥ 5 is an independent predictor of poor prognosis for uHCC treated with atezolizumab plus bevacizumab.43 In this study, the PD group exhibited an increase in NLR compared to baseline, while the SD and OR groups showed a decrease. Additionally, the OR-nPD group demonstrated a continuous decrease in NLR (p=0.0038). Zhou et al reported that tumor-associated neutrophils promote HCC cell growth.44 Increased circulating neutrophil levels may elevate levels of proteases, growth factors, VEGF, and intercellular adhesion molecule-1 (ICAM-1), contributing to cancer progression by modulating angiogenesis, cell growth, or inflammation, ultimately leading to lower survival rates in HCC patients.45,46

Additionally, this study included exploratory subgroup analyses based on three different immune checkpoint inhibitors: atezolizumab, camrelizumab, and sintilimab. Consistent with the results mentioned earlier, the atezolizumab and sintilimab subgroups, which had a higher ORR at the first assessment, demonstrated more active immune responses compared to the camrelizumab subgroup, which had a lower ORR (Tables S3 and S4). In this study, atezolizumab and sintilimab were primarily combined with bevacizumab, whereas camrelizumab was more paired with TKIs. In the short term, bevacizumab’s anti-angiogenic effect may be more effective against the ischemic and hypoxic environment caused by TACE. This finding provides a basis for clinical decision-making regarding combined immunotherapy regimens.

This study has some limitations. Firstly, it is a single-center retrospective study, which may introduce selection bias. Secondly, incomplete follow-up data resulted in a small sample size. Future studies should standardize and extend monitoring periods to collect more comprehensive data on dynamic changes in these immune indicators. Lastly, integrating pathological immunohistochemistry results could further investigate reasons for the differences in these immune indicators within the TME and in circulation.

In conclusion, the combination therapy of TACE plus ICIs and anti-VEGF antibodies/TKIs may modify the TME of HCC. Decreases in CD3CD19+, C4, CFB, NLR, along with increases in Ig λ, Ig κ, Ig A, Ig G, were associated with better tumor response, serving as potential biomarkers. Monitoring these trends can help tailor treatments individually and enhance efficacy.

Acknowledgments

Xiao-Yang Xu, Ze Wang and Chen-You Liu contributed equally to this work and share first authorship.We wish to acknowledge the support from all the investigators in this study.

Funding Statement

This work is supported by the Foundation of 2023 Jiangsu Province Natural Science Foundation Project (SBK2023022210), the 2023 Clinical Research Project of the First Affiliated Hospital of Soochow University (BXLC010), and the 2023 Zhou Nursing Research Project of the First Affiliated Hospital of Soochow University (HLYJ-Z-2023-02).

Data Sharing Statement

The datasets used and/or analyzed during the current study are available from the corresponding authors upon reasonable request.

Ethics Statement

This retrospective study was approved by the Institutional Review Board and Human Ethics Committee of the First Affiliated Hospital of Soochow University (Approval Number: 2024352). The need for informed consent was waived by the IRB due to the retrospective nature of this study. However, all participating patients provided written informed consent at each admission. The data collected were used solely for the purposes of this study. The study was performed in accordance with the ethical guidelines of the 1975 Declaration of Helsinki.

Disclosure

The authors of this manuscript declare no relationships with any companies whose products or services may be related to the subject matter of the article. The authors report no conflicts of interest in this work.

References

1. Bray F, Laversanne M, Sung H, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74(3):229–263. 10.3322/caac.21834 [Abstract] [CrossRef] [Google Scholar]
2. Ganesan P, Kulik LM. Hepatocellular Carcinoma: new Developments. Clin Liver Dis. 2023;27(1):85–102. 10.1016/j.cld.2022.08.004 [Abstract] [CrossRef] [Google Scholar]
3. Reig M, Forner A, Rimola J, et al. BCLC strategy for prognosis prediction and treatment recommendation: the 2022 update. J Hepatol. 2022;76(3):681–693. 10.1016/j.jhep.2021.11.018 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
4. Park JW, Chen M, Colombo M, et al. Global patterns of hepatocellular carcinoma management from diagnosis to death: the BRIDGE study. Liver Int. 2015;35(9):2155–2166. 10.1111/liv.12818 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
5. Lencioni R, Llovet JM, Han G, et al. Sorafenib or placebo plus TACE with doxorubicin-eluting beads for intermediate stage HCC: the SPACE trial. J Hepatol. 2016;64(5):1090–1098. 10.1016/j.jhep.2016.01.012 [Abstract] [CrossRef] [Google Scholar]
6. Oura K, Morishita A, Tani J, Masaki T. Tumor immune microenvironment and immunosuppressive therapy in hepatocellular carcinoma: a review. Int J Mol Sci. 2021;22(11):5801. 10.3390/ijms22115801 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
7. Hegde PS, Wallin JJ, Mancao C. Predictive markers of anti-VEGF and emerging role of angiogenesis inhibitors as immunotherapeutics. Semin Cancer Biol. 2018;52(Pt 2):117–124. 10.1016/j.semcancer.2017.12.002 [Abstract] [CrossRef] [Google Scholar]
8. Llovet JM, De Baere T, Kulik L, et al. Locoregional therapies in the era of molecular and immune treatments for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2021;18(5):293–313. 10.1038/s41575-020-00395-0 [Abstract] [CrossRef] [Google Scholar]
9. Chang X, Lu X, Guo J, Teng GJ. Interventional therapy combined with immune checkpoint inhibitors: emerging opportunities for cancer treatment in the era of immunotherapy. Cancer Treat Rev. 2019;74:49–60. 10.1016/j.ctrv.2018.08.006 [Abstract] [CrossRef] [Google Scholar]
10. Lencioni R, Kudo M, Erinjeri J, et al. EMERALD-1: a phase 3, randomized, placebo-controlled study of transarterial chemoembolization combined with durvalumab with or without bevacizumab in participants with unresectable hepatocellular carcinoma eligible for embolization. J Clin Oncol. 2024;42(3_suppl):LBA432–LBA432. 10.1200/JCO.2024.42.3_suppl.LBA432 [CrossRef] [Google Scholar]
11. Zhu HD, Li HL, Huang MS, et al. Transarterial chemoembolization with PD-(L)1 inhibitors plus molecular targeted therapies for hepatocellular carcinoma (CHANCE001). Signal Transduct Target Ther. 2023;8(1):58. 10.1038/s41392-022-01235-0 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
12. Jin ZC, Chen JJ, Zhu XL, et al. Immune checkpoint inhibitors and anti-vascular endothelial growth factor antibody/tyrosine kinase inhibitors with or without transarterial chemoembolization as first-line treatment for advanced hepatocellular carcinoma (CHANCE2201): a target trial emulation study. EClinicalMedicine. 2024;72:102622. 10.1016/j.eclinm.2024.102622 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
13. Donne R, Lujambio A. The liver cancer immune microenvironment: therapeutic implications for hepatocellular carcinoma. Hepatology. 2023;77(5):1773–1796. 10.1002/hep.32740 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
14. Tsilimigras DI, Ntanasis-Stathopoulos I, Moris D, Pawlik TM. Liver tumor microenvironment. Adv Exp Med Biol. 2020;1296:227–241. 10.1007/978-3-030-59038-3_14 [Abstract] [CrossRef] [Google Scholar]
15. Ringelhan M, Pfister D, O’Connor T, Pikarsky E, Heikenwalder M. The immunology of hepatocellular carcinoma. Nat Immunol. 2018;19(3):222–232. 10.1038/s41590-018-0044-z [Abstract] [CrossRef] [Google Scholar]
16. Pinato DJ, Murray SM, Forner A, et al. Trans-arterial chemoembolization as a loco-regional inducer of immunogenic cell death in hepatocellular carcinoma: implications for immunotherapy. J Immunother Cancer. 2021;9(9):e003311. 10.1136/jitc-2021-003311 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
17. Li SJ, Chen JX, Sun ZJ. Improving antitumor immunity using antiangiogenic agents: mechanistic insights, current progress, and clinical challenges. Cancer Commun. 2021;41(9):830–850. 10.1002/cac2.12183 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
18. Singal AG, Llovet JM, Yarchoan M, et al. AASLD practice guidance on prevention, diagnosis, and treatment of hepatocellular carcinoma. Hepatology. 2023;78(6):1922–1965. 10.1097/HEP.0000000000000466 [Abstract] [CrossRef] [Google Scholar]
19. Clinical Guidelines Committee of Chinese College of I. Chinese clinical practice guidelines for transarterial chemoembolization of hepatocellular carcinoma (2023 edition). Zhonghua Yi Xue Za Zhi. 2023;103(34):2674–2694. 10.3760/cma.j.cn112137-20230630-01114 [Abstract] [CrossRef] [Google Scholar]
20. Llovet JM, Lencioni R. mRECIST for HCC: performance and novel refinements. J Hepatol. 2020;72(2):288–306. 10.1016/j.jhep.2019.09.026 [Abstract] [CrossRef] [Google Scholar]
21. Liang Y, Yu M, Zhou C, Zhu X. Variation of PD-L1 expression in locally advanced cervical cancer following neoadjuvant chemotherapy. Diagn Pathol. 2020;15(1):67. 10.1186/s13000-020-00977-1 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
22. Zhu L, Cheng G, Wu M, Chen M, Jin Y. Heterogeneous distribution pattern of CD3+ tumor-infiltrated lymphocytes (TILs) and high combined positive score (CPS) favored the prognosis of resected early stage small-cell lung cancer. Transl Oncol. 2023;34:101697. 10.1016/j.tranon.2023.101697 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
23. Francini E, Ou FS, Lazzi S, et al. The prognostic value of CD3+ tumor-infiltrating lymphocytes for stage II colon cancer according to use of adjuvant chemotherapy: a large single-institution cohort study. Transl Oncol. 2021;14(2):100973. 10.1016/j.tranon.2020.100973 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
24. Han B, Yim J, Lim S, et al. Prognostic impact of the immunoscore based on whole-slide image analysis of CD3+ tumor-infiltrating lymphocytes in diffuse large B-cell lymphoma. Mod Pathol. 2023;36(9):100224. 10.1016/j.modpat.2023.100224 [Abstract] [CrossRef] [Google Scholar]
25. Yang F, Xu GL, Huang JT, et al. Transarterial chemoembolization combined with immune checkpoint inhibitors and tyrosine kinase inhibitors for unresectable hepatocellular carcinoma: efficacy and systemic immune response. Front Immunol. 2022;13:847601. 10.3389/fimmu.2022.847601 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
26. Duffy AG, Ulahannan SV, Makorova-Rusher O, et al. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J Hepatol. 2017;66(3):545–551. 10.1016/j.jhep.2016.10.029 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
27. Zhuang Y, Yuan BY, Chen GW, et al. Association between circulating lymphocyte populations and outcome after stereotactic body radiation therapy in patients with hepatocellular carcinoma. Front Oncol. 2019;9:896. 10.3389/fonc.2019.00896 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
28. Palomero J, Panisello C, Lozano-Rabella M, et al. Biomarkers of tumor-reactive CD4 + and CD8 + TILs associate with improved prognosis in endometrial cancer. J Immunother Cancer. 2022;10(12):e005443. 10.1136/jitc-2022-005443 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
29. Yuan S, Liu Y, Till B, Song Y, Wang Z. Pretreatment peripheral B cells are associated with tumor response to anti-PD-1-based immunotherapy. Front Immunol. 2020;11:563653. 10.3389/fimmu.2020.563653 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
30. Qin Z, Richter G, Schuler T, Ibe S, Cao X, Blankenstein T. B cells inhibit induction of T cell-dependent tumor immunity. Nat Med. 1998;4(5):627–630. 10.1038/nm0598-627 [Abstract] [CrossRef] [Google Scholar]
31. Kim SS, Sumner WA, Miyauchi S, Cohen EEW, Califano JA, Sharabi AB. Role of B cells in responses to checkpoint blockade immunotherapy and overall survival of cancer patients. Clin Cancer Res. 2021;27(22):6075–6082. 10.1158/1078-0432.CCR-21-0697 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
32. Qin M, Wang D, Fang Y, et al. Current perspectives on B lymphocytes in the immunobiology of hepatocellular carcinoma. Front Oncol. 2021;11:647854. 10.3389/fonc.2021.647854 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
33. Lee MJ, Na K, Shin H, et al. Early diagnostic ability of human complement factor B in pancreatic cancer is partly linked to its potential tumor-promoting role. J Proteome Res. 2021;20(12):5315–5328. 10.1021/acs.jproteome.1c00805 [Abstract] [CrossRef] [Google Scholar]
34. Shimazaki R, Takano S, Satoh M, et al. Complement factor B regulates cellular senescence and is associated with poor prognosis in pancreatic cancer. Cell Oncol. 2021;44(4):937–950. 10.1007/s13402-021-00614-z [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
35. Reis ES, Mastellos DC, Ricklin D, Mantovani A, Lambris JD. Complement in cancer: untangling an intricate relationship. Nat Rev Immunol. 2018;18(1):5–18. 10.1038/nri.2017.97 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
36. Rutkowski MJ, Sughrue ME, Kane AJ, Mills SA, Parsa AT. Cancer and the complement cascade. Mol Cancer Res. 2010;8(11):1453–1465. 10.1158/1541-7786.MCR-10-0225 [Abstract] [CrossRef] [Google Scholar]
37. Markiewski MM, DeAngelis RA, Benencia F, et al. Modulation of the antitumor immune response by complement. Nat Immunol. 2008;9(11):1225–1235. 10.1038/ni.1655 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
38. Zheng X, Qian K. Neutrophil-to-lymphocyte ratio predicts therapy outcomes of transarterial chemoembolization combined with tyrosine kinase inhibitors plus programmed cell death ligand 1 antibody for unresectable hepatocellular carcinoma. Anticancer Drugs. 2023;34(6):775–782. 10.1097/CAD.0000000000001458 [Abstract] [CrossRef] [Google Scholar]
39. Mazaki J, Katsumata K, Sujino H, et al. Neutrophil-to-lymphocyte ratio as a prognostic factor for colon cancer in elderly patients: a propensity score analysis. Anticancer Res. 2021;41(9):4471–4478. 10.21873/anticanres.15256 [Abstract] [CrossRef] [Google Scholar]
40. Liu N, Mao J, Tao P, Chi H, Jia W, Dong C. The relationship between NLR/PLR/LMR levels and survival prognosis in patients with non-small cell lung carcinoma treated with immune checkpoint inhibitors. Medicine. 2022;101(3):e28617. 10.1097/MD.0000000000028617 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
41. Capone M, Giannarelli D, Mallardo D, et al. Baseline neutrophil-to-lymphocyte ratio (NLR) and derived NLR could predict overall survival in patients with advanced melanoma treated with nivolumab. J Immunother Cancer. 2018;6(1):74. 10.1186/s40425-018-0383-1 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
42. Powell DR, Huttenlocher A. Neutrophils in the tumor microenvironment. Trends Immunol. 2016;37(1):41–52. 10.1016/j.it.2015.11.008 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
43. 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. 2022;14(23):5834. 10.3390/cancers14235834 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
44. Zhou SL, Yin D, Hu ZQ, et al. A positive feedback loop between cancer stem-like cells and tumor-associated neutrophils controls hepatocellular carcinoma progression. Hepatology. 2019;70(4):1214–1230. 10.1002/hep.30630 [Abstract] [CrossRef] [Google Scholar]
45. Du D, Liu C, Qin M, et al. Metabolic dysregulation and emerging therapeutical targets for hepatocellular carcinoma. Acta Pharm Sin B. 2022;12(2):558–580. 10.1016/j.apsb.2021.09.019 [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
46. Kuang DM, Zhao Q, Wu Y, et al. Peritumoral neutrophils link inflammatory response to disease progression by fostering angiogenesis in hepatocellular carcinoma. J Hepatol. 2011;54(5):948–955. 10.1016/j.jhep.2010.08.041 [Abstract] [CrossRef] [Google Scholar]

Articles from Journal of Hepatocellular Carcinoma are provided here courtesy of Dove Press

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.