This work is licensed under http://creativecommons.org/licenses/by/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
1. Introduction
Comprising 75%–85% of primary liver cancer, hepatocellular carcinoma (HCC) is one of the most fatal cancers which presents a severe challenge to human health and life quality [1]. Despite recent advances in diagnosis and treatment, the resection rate and overall prognosis of HCC patients are still unsatisfactory. Even if the early-stage HCC patients were treated with curative surgery [2], the incidence of tumor relapse ranges from 50% to 70% [3]. Therefore, it is urgent to identify biomarkers to screen out patients with a high risk of relapse. The tumor immune ecosystem is a key determinant and research hotspot of the progression of HCC [4]. In this study, we analyzed the relationship between HLA-DR+ T cells, one of the peripheral blood lymphocyte subsets, and the prognosis of HCC.
The human leukocyte antigen isotype DR (HLA-DR) as an MHC class II molecule is expressed on antigen-presenting cells (APC) and activated T cells [5, 6]. Numerous detailed studies have elucidated the importance of presenting antigens to T cells by professional APC expressing HLA-DR in the immune process [7, 8]. HLA-DR, as a marker of T cell activation, was shown to associate with disease states, such as cancer, HIV, and autoimmune diseases [9–11]. However, immunosuppressive cells expressing HLA-DR, including TAM tumor-associated macrophage (TAM) and regulatory T (Treg) cells, are involved in immune escape [12, 13]. Additionally, previous studies revealed HCC cells are one major source of HLA-DR in tumor tissues [14]. But in HCC, few studies reported the role of HLA-DR+ T cells in prognosis.
T cells, as the most important part of antitumor immunity, have been the focus of the research. Over these years, more and more studies revealed the significance of HLA-DR+ T cells in the development and treatment of HCC. According to a recent study, mucosal-associated invariant T (MAIT) cells, as innate T cells with immunoregulatory activity, upregulated HLA-DR in peripheral blood of HCC patients but not in hepatic sinusoids and were functionally impaired [15]. The activated but functionally impaired MAIT cells might suggest a potential role in HCC pathogenesis. In addition, the CD57+ HLA-DR+ CD8+ T cells, displaying a highly proliferative and cytotoxic phenotype, were correlated with recurrence in HCC patients after liver transplantation [16]. HLA-DR+ T cells in peripheral are increased in HCC patients with anti-CTLA4 treatment [17]. In conclusion, the role of HLA-DR+ T cells in HCC prognosis deserves further investigation.
We aimed to analyze the relationship between the preoperative HLA-DR+ T cell ratio and postoperative recurrence or death in HCC patients. We also analyzed the differences in baseline data between the groups with different HLA-DR+ T cell ratios to investigate the potential value of HLA-DR+ T cells in HCC progression and prognosis.
2. Patients and Methods
2.1. Patient Selection
Patients who received liver resection and were pathologically diagnosed as having HCC at the Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, between January 2013 and December 2021 were retrospectively collected. We eliminated patients with other malignancies, preoperative anti-HCC treatment, macrovascular invasion, TNM stage III, TNM stage VI, and postoperative liver transplantation for non-neoplastic liver disease, as well as patients lacking complete baseline and follow-up data, resulting in 192 patients included in the study. We acquired clinicopathological data from medical records. This study complied with the ethical guidelines of the Helsinki Declaration, and written informed consent was obtained from participants or their immediate families prior to surgery.
2.2. Clinicopathological Variables
The clinicopathological variables of HCC patients incorporated the date of surgery, gender, age, HbsAg, Child-Pugh grade, AFP (alpha-fetoprotein) levels, ALT (alanine transaminase) levels, AST (aspartate transaminase) levels, AST/ALT, GGT (gamma glutamyl transferase) levels, lymphocyte subsets, largest tumor size, tumor number, liver cirrhosis, macro- and microvascular invasion, capsular invasion, tumor satellites, and tumor differentiation. Each subset of lymphocytes was described in terms of its ratio to total lymphocytes. For continuous variables, the upper level of normal (ULN) was used as the cut-off value. AFP positivity was defined as a serum level ≥20 ng/ml. Liver cirrhosis was diagnosed based on pathological findings. The curative surgery referred to complete resection of tumor with negative microscopic margins. The 8th AJCC cancer staging system was used to stage HCC. Partial HCC patients with microvascular invasion received prophylactic TACE (transcatheter arterial chemoembolization) within 1 month after surgery.
2.3. Postoperative Follow-Up
All patients were followed up regularly at the outpatient clinic after discharge. AFP and image studies were performed routinely at 1 month after the operation, then at least 3 months in the first 2 years, and at least 6 months thereafter. Dynamic contrast-enhanced computed tomography (CT) of the upper abdomen or Gd-EOB-DPTA-enhanced magnetic resonance imaging (MRI) was performed if digestive ultrasound revealed HCC recurrence and/or alpha fetoprotein (AFP) was significantly elevated. Further auxiliary inspection including chest CT, lower abdominal CT, pelvic CT, or positron emission tomography (PET)-CT was completed when clinically necessary. The diagnosis of tumor recurrence was confirmed by at least two imaging examinations or liver histopathological examination. Once tumor recurrence is found, resurgical resection, local ablation, TACE, radiotherapy, systemic therapy, and supportive care can be selected according to the characteristics of tumor recurrence. The survival information was obtained from clinical follow-up or telephone follow-up. Follow-up was up to September 1, 2021. The starting point of follow-up was the date of surgery; the time interval from the starting point to the date of the first recurrence, first metastasis, death, or last follow-up was PFS; and the time interval from the starting point to the date of death or last follow-up was OS.
2.4. Statistical Analysis
Continuous variables were categorized by ULN and described together with categorical variables as frequencies (N). The χ2 test or Fisher’s test was performed to compare categorical variables. Using the COX proportional hazard model, we identified independent risk factors associated with postoperative progression or death, with hazard ratios (HR) and 95% confidence intervals (CI). We plotted the Kaplan–Meier curves through the “survival” package of the R language software package. SPSS software version 22.0 (SPSS, Chicago, IL, USA) and R language software version 4.1.2 were used for statistical analysis in this study. Results were considered statistically significant when the
3. Results
3.1. Patient Baseline Information
The baseline clinicopathological characteristics of all HCC patients were outlined in Table 1. A total of 115 (59.9%) patients were less than 60 years old, and 153 (79.7%) patients were male. There were 164 (85.4%) patients who had the history of viral hepatitis B, and 114 (59.4%) patients had liver cirrhosis. There were 187 (97.4%) patients with Child-Pugh grade A, and 5 well selected HCC patients with Child-Pugh grade B who underwent curative surgery were also included in the study according to a comprehensive review [18]. The proportion of patients with AFP positivity (≥20 ng/ml) and a high HLA-DR+ T cell ratio (≥5.8%) were 50.0% and 52.1%, respectively. Except for 2 patients with two tumors, the rest of the patients had one tumor. 190 patients (99.0%) were TNM stage I, and 2 patients were TNM stage II. 116 patients (60.4%) were Edmondson grades I-II. Capsular invasion was noted in 70 (36.5%) patients, and microvascular invasion was noted in 67 (34.9%) patients. After curative surgery, 65 (33.9%) patients received prophylactic TACE. With a median follow-up of 48.3 months in entire study patients, postoperative tumor progression occurred in 81 (42.2%) patients and 32 (16.7%) patients died. Nevertheless, patients with a high HLA-DR+ T cell ratio were less likely to present cancer progression (35%).
Table 1
Comparisons of clinicopathological characteristics among 192 HCC patients with different HLA-DR+ T cell ratio groups.
Variables | Total (N) | High HLADR+ T cell ratio (N) | Low HLADR+ T cell ratio (N) | χ2 | ||
Age | <60 years | 115 | 55 | 60 | 2.082 | 0.149 |
≥60 years | 77 | 45 | 32 | |||
Gender | Male | 153 | 79 | 74 | 0.061 | 0.805 |
Female | 39 | 21 | 18 | |||
HbsAg | Positive | 164 | 84 | 80 | 0.336 | 0.562 |
Negative | 28 | 16 | 12 | |||
Liver cirrhosis | Yes | 114 | 61 | 53 | 0.228 | 0.633 |
No | 78 | 39 | 39 | |||
AFP level | <20 ng/ml | 96 | 52 | 44 | 0.334 | 0.563 |
≥20 ng/ml | 96 | 48 | 48 | |||
AST/ALT | <1 | 123 | 67 | 56 | 0.782 | 0.376 |
≥1 | 69 | 33 | 36 | |||
GGT level | <45 U/L | 129 | 70 | 59 | 0.749 | 0.387 |
≥45 U/L | 63 | 30 | 33 | |||
Child-Pugh grade | A | 187 | 97 | 90 | 1 | |
B | 5 | 3 | 2 | |||
T cell ratio | <77% | 150 | 70 | 80 | 8.062 | 0.005 |
≥77% | 42 | 30 | 12 | |||
CD4+T cell ratio | <41.6% | 124 | 66 | 58| | 0.669 | |
≥41.6% | 68 | 34 | 34 | |||
CD8+ T cell ratio | <29.6% | 126 | 54 | 72 | 12.052 | <0.001 |
≥29.6% | 66 | 46 | 20 | |||
B cell ratio | <18.2% | 154 | 88 | 66 | 7.981 | 0.005 |
≥18.2% | 38 | 12 | 26 | |||
NK cell ratio | <25.6% | 168 | 90 | 78 | 1.193 | 0.275 |
≥25.6% | 24 | 10 | 14 | |||
Largest tumor size | <5 cm | 142 | 71 | 71 | 0.948 | 0.330 |
≥5 cm | 50 | 29 | 21 | |||
Edmondson grade | I-II | 116 | 56 | 60 | 1.702 | 0.192 |
III-IV | 76 | 44 | 32 | | | ||
Microvascular invasion | Yes | 67 | 32 | 35 | 0.770 | 0.380 |
No | 125 | 68 | 57 | |||
Capsular invasion | Yes | 70 | 38 | 32 | 0.214 | 0.644 |
No | 122 | 62 | 60 | |||
Prophylactic TACE after surgery | Yes | 65 | 31 | 34 | 0.759 | 0.384 |
No | 127 | 69 | 58 |
AFP: alpha-fetoprotein; AST: aspartate aminotransferase; ALT: alanine aminotransferase; GGT: gamma glutamyl transferase; TACE: transcatheter arterial chemoembolization.
3.2. The Relationship between Clinicopathological Characteristics and the HLA-DR+ T Cell Ratio
A total of 100 (52.1%) patients had a high HLA-DR+ T cell ratio. The relationship between clinicopathological characteristics and the HLA-DR+ T cell ratio is shown in Table 1. Patients with a high HLA-DR+ T cell ratio had a higher T cell ratio (
[figure(s) omitted; refer to PDF]
3.3. COX Regression Analyses between Clinicopathological Variables Associated with PFS and OS after Curative Surgery for HCC
As indicated by the Kaplan–Meier curves in Figure 2(a), HCC patients with a high HLA-DR+ T cell ratio had better PFS (
[figure(s) omitted; refer to PDF]
Table 2
Univariate and multivariate Cox regression analyses of risk factors associated with progression-free survival (PFS) after curative surgery for hepatocellular carcinoma.
Variables | HR comparison | UV HR (95% CI) | UV | MV HR (95% CI) | MV |
Gender | Male vs. female | 1.264 (0.710–2.249) | 0.425 | ||
Age | ≥60 vs.<60 years | 0.774 (0.491–1.219) | 0.269 | ||
HbsAg positivity | Yes vs. no | 1.499 (0.722–3.113) | 0.277 | ||
Liver cirrhosis | Yes vs. no | 1.535 (0.957–2.463) | 0.076 | ||
AST/ALT | ≥1 vs. <1 | 1.016 (0.634–1.627) | 0.948 | ||
GGT level | ≥45 U/L vs. <45 U/L | 2.104 (1.354–3.271) | 0.001 | 1.692 (1.062–2.696) | 0.027 |
AFP positivity | Yes vs. no | 1.648 (1.057–2.568) | 0.027 | 1.371 (0.857–2.194) | 0.187 |
CD3+ T cell ratio | ≥77% vs. <77% | 0.680 (0.375–1.233) | 0.204 | ||
CD4+ T cell ratio | ≥41.6% vs. <41.6% | 1.129 (0.706–1.803) | 0.613 | ||
CD8+ T cell ratio | ≥29.6% vs. <29.6% | 0.595 (0.363–0.975) | 0.040 | 0.655 (0.387–1.109) | 0.116 |
HLADR+ T cell ratio | ≥5.8% vs.<5.8% | 0.512 (0.328–0.799) | 0.003 | 0.465 (0.288–0.750) | 0.002 |
B cell ratio | ≥18.2% vs. <18.2% | 1.427 (0.861–2.366) | 0.168 | ||
NK cell ratio | ≥25.6% vs. <25.6% | 1.063 (0.575–1.956) | 0.845 | ||
Edmondson grade | III-IV vs. I-II | 1.697 (1.095–2.629) | 0.018 | 1.638 (1.006–2.668) | 0.047 |
Largest tumor size | ≥5 vs. <5 cm | 1.675 (1.044–2.690) | 0.033 | 1.572 (0.930–2.657) | 0.091 |
Microvascular invasion | Yes vs. No | 1.440 (0.924–2.246) | 0.108 | ||
Capsular invasion | Yes vs. No | 1.709 (1.103–2.647) | 0.016 | 1.615 (1.010–2.582) | 0.045 |
Prophylactic TACE after surgery | Yes vs. No | 1.790 (1.154–2.776) | 0.009 | 1.044 (0.633–1.723) | 0.867 |
AFP: alpha-fetoprotein; AST: aspartate aminotransferase; ALT: alanine aminotransferase; GGT: gamma glutamyl transferase; TACE: transcatheter arterial chemoembolization.
Table 3
Univariate and multivariate Cox regression analyses of risk factors associated with overall survival (OS) after curative surgery for hepatocellular carcinoma.
Variables | HR comparison | UV HR (95% CI) | UV | MV HR (95% CI) | MV |
Gender | Male vs. female | 1.895 (0.666–5.394) | 0.231 | ||
Age | ≥60 vs.<60 years | 0.594 (0.282–1.251) | 0.170 | ||
HbsAg positivity | Yes vs. no | 2.223 (0.531–9.302) | 0.274 | ||
Liver cirrhosis | Yes vs. no | 1.836 (0.823–4.096) | 0.138 | ||
AST/ALT | ≥1 vs. <1 | 1.232 (0.588–2.580) | 0.581 | ||
GGT level | ≥45 U/L vs. <45 U/L | 2.459 (1.239–4.880) | 0.010 | 2.156 (1.020–4.558) | 0.044 |
AFP positivity | Yes vs. no | 2.241 (1.085–4.626) | 0.029 | 1.534 (0.716–3.287) | 0.271 |
CD3+ T cell ratio | ≥77% vs. <77% | 0.996 (0.407–2.435) | 0.993 | ||
CD4+ T cell ratio | ≥41.6% vs. <41.6% | 0.412 (0.158–1.072) | 0.069 | ||
CD8+ T cell ratio | ≥29.6% vs. <29.6% | 1.018 (0.500–2.074) | 0.960 | ||
HLADR+ T cell ratio | ≥5.8% vs.<5.8% | 0.861 (0.434–1.707) | 0.567 | ||
B cell ratio | ≥18.2% vs. <18.2% | 1.543 (0.733–3.249) | 0.254 | ||
NK cell ratio | ≥25.6% vs. <25.6% | 1.326 (0.546–3.217) | 0.533 | ||
Edmondson grade | III-IV vs. I-II | 2.541 (1.269–5.089) | 0.008 | 1.582 (0.727–3.441) | 0.247 |
Largest tumor size | ≥5 cm vs. <5 cm | 2.885 (1.440–5.780) | 0.003 | 2.346 (1.100–5.007) | 0.027 |
Microvascular invasion | Yes vs. no | 2.817 (1.409–5.634) | 0.003 | 2.379 (1.085–5.215) | 0.030 |
Capsular invasion | Yes vs. no | 1.757 (0.878–3.515) | 0.111 | ||
Prophylactic TACE after surgery | Yes vs. no | 2.117 (1.068–4.194) | 0.032 | 0.794 (0.345–1.831) | 0.589 |
AFP: alpha-fetoprotein; AST: aspartate aminotransferase; ALT: alanine aminotransferase; GGT: gamma glutamyl transferase; TACE: transcatheter arterial chemoembolization.
3.4. COX Regression Analyses between Clinicopathological Variables Associated with PFS and OS after Curative Surgery for AFP-Positive HCC
The clinicopathological characteristics associated with serum AFP positivity (≥20 ng/ml) versus AFP negativity (<20 ng/ml) among HCC patients were noted in Table 4. AFP-positive HCC patients had more female patients (
Table 4
Comparisons of clinicopathological characteristics among 192 HCC patients with different AFP level groups.
Variables | AFP positivity (N) | AFP negativity (N) | χ2 | ||
Age | <60 years | 62 | 53 | 1.756 | 0.185 |
≥60 years | 34 | 43 | |||
Gender | Male | 71 | 82 | 3.893 | 0.048 |
Female | 25 | 14 | |||
HbsAg | Positive | 87 | 77 | 4.181 | 0.041 |
Negative | 9 | 19 | |||
Liver cirrhosis | Yes | 60 | 54 | 0.777 | 0.378 |
No | 36 | 42 | |||
AST/ALT | <1 | 58 | 65 | 1.109 | 0.292 |
≥1 | 38 | 31 | |||
GGT level | <45 U/L | 61 | 68 | 0.369 | 0.543 |
≥45 U/L | 35 | 28 | |||
Child-Pugh class | A | 93 | 94 | 1 | |
B | 3 | 2 | |||
T cell ratio | <77% | 75 | 75 | 0.000 | 1 |
≥77% | 21 | 21 | |||
CD4+ T cell ratio | <41.6% | 60 | 64 | 0.364 | 0.546 |
≥41.6% | 36 | 32 | |||
CD8+ T cell ratio | <29.6% | 61 | 65 | 0.369 | 0.543 |
≥29.6% | 35 | 31 | |||
HLADR+ T cell ratio | <5.8% | 48 | 44 | 0.334 | 0.563 |
≥5.8% | 48 | 52 | |||
B cell ratio | <18.2% | 70 | 84 | 6.431 | 0.011 |
≥18.2% | 26 | 12 | |||
NK cell ratio | <25.6% | 85 | 83 | 0.190 | 0.663 |
≥25.6% | 11 | 13 | |||
Largest tumor size | <5 cm | 70 | 72 | 0.108 | 0.742 |
≥5 cm | 26 | 24 | |||
Edmondson grade | I-II | 47 | 69 | 10.541 | 0.001 |
III-IV | 49 | 27 | |||
Microvascular invasion | Yes | 40 | 27 | 3.874 | 0.049 |
No | 56 | 69 | |||
Capsular invasion | Yes | 37 | 33 | 0.360 | 0.549 |
No | 59 | 63 | |||
Prophylactic TACE after surgery | Yes | 36 | 29 | 1.140 | 0.286 |
No | 60 | 67 |
AFP: alpha-fetoprotein; AFP positivity: AFP ≥ 20 ng/ml; AFP negativity: AFP < 20 ng/ml; AST: aspartate aminotransferase; ALT: alanine aminotransferase; GGT: gamma glutamyl transferase; TACE: transcatheter arterial chemoembolization.
[figure(s) omitted; refer to PDF]
Table 5
Univariate Cox regression analyses of risk factors associated with progression-free survival and overall survival after curative surgery for AFP-positive (≥20 ng/ml) hepatocellular carcinoma.
Variables | HR comparison | Progression-free survival | Overall survival | ||
UV HR (95% CI) | UV | UV HR (95% CI) | UV | ||
Gender | Male vs. female | 0.927 (0.479–1.793) | 0.822 | 2.967 (0.684–12.878) | 0.146 |
Age | ≥60 vs.<60 years | 0.823 (0.449–1.508) | 0.529 | 0.775 (0.311–1.932) | 0.585 |
HbsAg positivity | Yes vs. no | 0.770 (0.302–1.962) | 0.584 | 0.896 (0.208–3.859) | 0.882 |
Liver cirrhosis | Yes vs. no | 1.386 (0.743–2.587) | 0.305 | 1.180 (0.450–3.096) | 0.737 |
AST/ALT | ≥1 vs. <1 | 0.980 (0.538–1.787) | 0.948 | 1.029 (0.403–2.622) | 0.953 |
GGT level | ≥45 U/L vs. <45 U/L | 2.122 (1.203–3.745) | 0.009 | 1.974 (0.837–4.656) | 0.120 |
CD3+ T cell ratio | ≥77% vs. <77% | 0.616 (0.275–1.380) | 0.239 | 0.686 (0.201–2.345) | 0.548 |
CD4+ T cell ratio | ≥41.6% vs. <41.6% | 1.215 (0.664–2.223) | 0.528 | 0.200 (0.047–0.863) | 0.031 |
CD8+ T cell ratio | ≥29.6% vs. <29.6% | 0.505 (0.261–0.975) | 0.042 | 0.780 (0.310–1.964) | 0.598 |
HLADR+ T cell ratio | ≥5.8% vs. <5.8% | 0.499 (0.278–0.895) | 0.020 | 0.589 (0.244–1.421) | 0.239 |
B cell ratio | ≥18.2% vs. <18.2% | 1.060 (0.569–1.977) | 0.854 | 1.338 (0.551–3.249) | 0.520 |
NK cell ratio | ≥25.6% vs, <25.6% | 1.680 (0.783–3.603) | 0.183 | 2.816 (1.003–7.909) | 0.049 |
Edmondson grade | III-IV vs. I-II | 1.630 (0.908–2.926) | 0.101 | 1.549 (0.648–3.702) | 0.325 |
Largest tumor size | ≥5 vs. <5 cm | 1.467 (0.796–2.703) | 0.219 | 2.438 (1.021–5.825) | 0.045 |
Microvascular invasion | Yes vs. no | 1.538 (0.868–2.729) | 0.140 | 3.514 (1.404–8.797) | 0.007 |
Capsular invasion | Yes vs. no | 1.571 (0.888–2.778) | 0.121 | 1.517 (0.625–3.685) | 0.357 |
AFP: alpha-fetoprotein; AST: aspartate aminotransferase; ALT: alanine aminotransferase; GGT: gamma glutamyl transferase.
Table 6
Univariate Cox regression analyses of risk factors associated with progression-free survival and overall survival after curative surgery for AFP-negative (<20 ng/ml) hepatocellular carcinoma.
Variables | HR comparison | Progression-free survival | Overall survival | ||
UV HR (95% CI) | UV | UV HR (95% CI) | UV | ||
Gender | Male vs. female | 3.477 (0.828–14.578) | 0.088 | 1.856 (0.237–14.538) | 0.556 |
Age | ≥60 vs. <60 years | 0.758 (0.377–1.524) | 0.436 | 0.451 (0.119–1.707) | 0.241 |
Liver cirrhosis | Yes vs. no | 1.627 (0.786–3.367) | 0.190 | 3.203 (0.689–14.893) | 0.138 |
AST/ALT | ≥1 vs. <1 | 0.958 (0.443–2.072) | 0.914 | 1.635 (0.471–5.678) | 0.439 |
GGT level | ≥45 U/L vs. <45 U/L | 1.910 (0.938–3.887) | 0.074 | 3.591 (1.083–11.907) | 0.037 |
CD3+T cell ratio | ≥77% vs. <77% | 0.802 (0.330–1.952) | 0.627 | 1.737 (0.448–6.742) | 0.425 |
CD4+ T cell ratio | ≥41.6% vs. <41.6% | 1.024 (0.483–2.171) | 0.950 | 1.073 (0.280–4.117) | 0.918 |
CD8+ T cell ratio | ≥29.6% vs. <29.6% | 0.672 (0.310–1.458) | 0.314 | 1.087 (0.317–3.736) | 0.894 |
HLADR+ T cell ratio | ≥5.8% vs. <5.8% | 0.548 (0.275–1.093) | 0.088 | 1.351 (0.394–4.629) | 0.632 |
B cell ratio | ≥18.2% vs. <18.2% | 1.910 (0.787–4.633) | 0.152 | 1.353 (0.291–6.278) | 0.700 |
NK cell ratio | ≥25.6% vs. <25.6% | 0.650 (0.227–1.864) | 0.423 | 0.487 (0.062–3.280) | 0.493 |
Edmondson grade | III-IV vs. I-II | 1.525 (0.738–3.149) | 0.254 | 3.500 (1.067–11.481) | 0.039 |
Largest tumor size | ≥5 cm vs. <5 cm | 1.911 (0.901–4.052) | 0.091 | 3.779 (1.128–12.665) | 0.031 |
Microvascular invasion | Yes vs. no | 1.144 (0.544–2.406) | 0.723 | 1.926 (0.587–6.324) | 0.280 |
Capsular invasion | Yes vs. no | 1.724 (0.863–3.445) | 0.123 | 2.136 (0.645–7.706) | 0.214 |
AFP: alpha-fetoprotein; AST: aspartate aminotransferase; ALT: alanine aminotransferase; GGT: gamma glutamyl transferase.
Table 7
Comparisons of clinicopathological characteristics among AFP-positive (≥20 ng/ml) HCC patients with different HLA-DR+ T cell ratio groups.
Variables | Total (N) | High HLADR+ T cell ratio (N) | Low HLADR+ T cell ratio (N) | χ2 | ||
Age | <60 years | 62 | 31 | 31 | 0.000 | 1 |
≥60 years | 34 | 17 | 17 | |||
Gender | Male | 71 | 35 | 36 | 0.054 | 0.816 |
Female | 25 | 13 | 12 | |||
HbsAg | Positive | 87 | 45 | 42 | 0.486 | |
Negative | 9 | 3 | 6 | |||
Liver cirrhosis | Yes | 60 | 29 | 31 | 0.178 | 0.673 |
No | 36 | 19 | 17 | |||
AST/ALT | <1 | 58 | 29 | 29 | 0.000 | 1.000 |
≥1 | 38 | 19 | 19 | |||
GGT level | <45 U/L | 61 | 31 | 30 | 0.045 | 0.832 |
≥45 U/L | 35 | 17 | 18 | |||
Child-Pugh grade | A | 93 | 45 | 48 | 0.242 | |
B | 3 | 3 | 0 | |||
T cell ratio | <77% | 75 | 31 | 44 | 10.301 | 0.001 |
≥77% | 21 | 17 | 4 | |||
CD4+ T cell ratio | <41.6% | 60 | 32 | 28 | 0.771 | 0.399 |
≥41.6% | 36 | 16 | 20 | |||
CD8+ T cell ratio | <29.6% | 61 | 25 | 36 | 5.441 | 0.020 |
≥29.6% | 35 | 23 | 12 | |||
B cell ratio | <18.2% | 70 | 40 | 30 | 5.275 | 0.022 |
≥18.2% | 26 | 8 | 18 | |||
NK cell ratio | <25.6% | 85 | 45 | 40 | 2.567 | 0.109 |
≥25.6% | 11 | 3 | 8 | |||
Largest tumor size | <5 cm | 70 | 32 | 38 | 1.899 | 0.168 |
≥5 cm | 26 | 16 | 10 | |||
Edmondson grade | I-II | 47 | 23 | 24 | 0.042 | 0.838 |
III-IV | 49 | 25 | 24 | |||
Microvascular invasion | Yes | 40 | 17 | 23 | 1.543 | 0.214 |
No | 56 | 31 | 25 | |||
Capsular invasion | Yes | 37 | 19 | 18 | 0.044 | 0.834 |
No | 59 | 29 | 30 | |||
Prophylactic TACE after surgery | Yes | 36 | 18 | 18 | 0.000 | 1.000 |
No | 60 | 30 | 30 |
AFP: alpha-fetoprotein; AST: aspartate aminotransferase; ALT: alanine aminotransferase; GGT: gamma glutamyl transferase; TACE: transcatheter arterial chemoembolization.
Table 8
Comparisons of clinicopathological characteristics among AFP-negative (<20 ng/ml) HCC patients with different HLA-DR+ T cell ratio groups.
Variables | Total (N) | High HLADR+ T cell ratio (N) | Low HLADR+ T cell ratio (N) | χ2 | ||
Age | <60 years | 53 | 24 | 29 | 3.762 | 0.052 |
≥60 years | 33 | 28 | 15 | |||
Gender | Male | 82 | 44 | 38 | 0.058 | 0.809 |
Female | 14 | 8 | 6 | |||
HbsAg | Positive | 77 | 39 | 38 | 1.939 | 0.164 |
Negative | 19 | 13 | 6 | |||
Liver cirrhosis | Yes | 54 | 32 | 22 | 1.289 | 0.256 |
No | 42 | 20 | 22 | |||
AST/ALT | <1 | 65 | 38 | 27 | 1.496 | 0.221 |
≥1 | 31 | 14 | 17 | |||
GGT level | <45 U/L | 68 | 39 | 29 | 0.953 | 0.329 |
≥45 U/L | 28 | 13 | 15 | |||
T cell ratio | <77% | 75 | 39 | 36 | 0.648 | 0.421 |
≥77% | 21 | 13 | 8 | |||
CD4+ T cell ratio | <41.6% | 64 | 34 | 30 | 0.084 | 0.772 |
≥41.6% | 32 | 18 | 14 | |||
CD8+ T cell ratio | <29.6% | 65 | 29 | 36 | 7.397 | 0.007 |
≥29.6% | 31 | 23 | 8 | |||
B cell ratio | <18.2% | 84 | 48 | 36 | 2.398 | 0.122 |
≥18.2% | 12 | 4 | 8 | |||
NK cell ratio | <25.6% | 83 | 45 | 38 | 0.001 | 0.980 |
≥25.6% | 13 | 7 | 6 | |||
Largest tumor size | <5 cm | 62 | 39 | 33 | 0.000 | 1.000 |
≥5 cm | 24 | 13 | 11 | |||
Edmondson grade | I-II | 69 | 33 | 36 | 3.973 | 0.046 |
III-IV | 27 | 19 | 8 | |||
Microvascular invasion | Yes | 27 | 15 | 12 | 0.029 | 0.864 |
No | 69 | 37 | 32 | |||
Capsular invasion | Yes | 33 | 19 | 14 | 0.235 | 0.628 |
No | 63 | 33 | 30 | |||
Prophylactic TACE after surgery | Yes | 29 | 13 | 16 | 1.460 | 0.227 |
No | 67 | 39 | 28 |
AFP: alpha-fetoprotein; AST: aspartate aminotransferase; ALT: alanine aminotransferase; GGT: gamma glutamyl transferase; TACE: transcatheter arterial chemoembolization.
4. Discussion
With the deepening of the research on the immune environment, more and more studies have confirmed that the functional status of T cells plays a pivotal role in the occurrence and development of HCC [4, 19, 20]. As an important molecule for presenting antigens, HLA-DR was mostly used as a target on the surface of APC for HCC-related research [12, 21], whereas HLA-DR+ T cells are shown to be effector T cells in a variety of malignancies and serve as a predictive factor for antitumor treatment [22, 23]. Based on these existing studies, we designed this project to explore the role of HLA-DR+ T cell in the anti-HCC immunity process. To the best of our knowledge, this research first revealed that HLA-DR+ T cell was a predictive marker for recurrence of HCC after hepatectomy.
In this article, we revealed that HCC patients with a high HLA-DR+ T cell ratio were less likely to experience recurrence after curative liver resection. HLA-DR+ cytotoxic T lymphocytes (CTLs) are cytotoxic and express immune signatures of functionally activated cells, such as Granzyme B and IFN-γ [24]. Meanwhile, regulatory T (Treg) cells upregulating HLA-DR expression increase in HCC and exhibit enhanced immunosuppressive activity driven by the hypoxia environment [25, 26]. However, it has been recently reported that Treg cells are differently expressed in HCC according to etiology of underlying liver cirrhosis. Treg cells play an immunosuppressive role in chronic viral liver diseases by hindering the antiviral process, while in autoimmune liver diseases, Treg cells show quantitative and functional defects so that they cannot effectively suppress self-reactive lymphocytes [27–29]. Besides, monitoring the genetic background (HLA) of Treg cells has potential value in assessing the extent of drug side effects during treatment with immune checkpoint inhibitors (ICIs) in patients with HCC [29]. HLA-DR is expressed not only on the surface of CTLs but also on immunosuppressive T cells so that the effect of HLA-DR on tumor immunity may be opposite. It is necessary to separately research the expression and prognostic value of HLA-DR in various T lymphocyte subsets. Besides, through the significance test of difference, we demonstrated that patients in the high HLA-DR+ T cell ratio group had a lower B cell ratio than patients in the low HLA-DR+ T cell ratio group. The role of B cells in T cell activation is bidirectional. Tumor-infiltrating B cells activate T cells through spatial cell-to-cell contacts to enhance antitumor activity in HCC patients [30]. On the contrary, B cell-derived GABA (γ-aminobutyric acid) impairs cytotoxic T cell responses and antitumor immunity [31]. In this article, we revealed that the B cell ratio was negatively correlated with the activated (HLA-DR+) T cell ratio. In conclusion, we believed that HCC patients with a high HLA-DR+ T cell ratio had a low B cell ratio and were less prone to tumor progression postoperatively.
AFP, as a tumor-associated antigen (TAA) of HCC [32], is considered a serum biomarker for diagnosis and a potential target for immunotherapy [33]. We, respectively, analyzed the prognostic significance of the HLA-DR+ T cell ratio in AFP-positive and AFP-negative HCC patients. There was no statistically significant difference in the HLA-DR+ T cell ratio between AFP-positive and AFP-negative HCC patients. But only among AFP-positive HCC patients, we found that patients in the high HLA-DR+ T cell ratio group had a lower rate of postoperative tumor progression than patients in the low HLA-DR+ T cell ratio group. It has been reported that AFP has strong binding properties towards HLA-DR of helper T lymphocyte and T cell response against AFP contributes to the significantly improving survival rate [34]. As part of the normal T cell repertoire, TAA-specific T cell responses are important for controlling HCC in different stages [35, 36]. However, further research will be needed to confirm whether HLA-DR+ T cells are AFP-specific T cells.
This study aimed to find new predictors of postoperative recurrence to optimize postoperative follow-up protocols. For the first time, HLA-DR+ T cell was used as a marker of antitumor immunity activity in the study of HCC. Simultaneously, we conducted stratification studies and demonstrated the specific prognostic value of HLA-DR+ T cells for postoperative recurrence in AFP-positive HCC patients. Moreover, apart from the HLA-DR+ T cell ratio, this study found other predictors for progression of HCC patients after curative resection, such as the GGT level, Edmondson grade, and capsular invasion.
Meanwhile, there were a number of limitations in the current research. First, this retrospective study in a single center may bring about selection bias. Consequently, a large-scale, multicenter, and prospective study is needed to validate the results of this study. Second, the classification of continuous variables is bounded by the ULN of a single medical institution. Whether the results will differ from those of other hospitals is unknown. Third, this study only included HBV, which is a common etiology of HCC in China, so it cannot be confirmed whether HLA-DR+ T cell have the same prognostic value in HCC caused by other pathogenies. Last, some postoperative adjuvant treatments, such as antiviral therapy, targeted therapy, and immunotherapy, were not included in this study. We need to incorporate these factors affecting postoperative recurrence into the study for further stratified analysis.
5. Conclusion
In summary, our study found that the preoperative HLA-DR+ T cell ratio served as a useful prognostic marker for PFS in HCC patients with curative resection, and the same results were obtained in AFP-positive patients. This is beneficial for clinicians to screen patients with poor prognoses based on clinical data in order to improve patient outcomes by developing personalized treatment and follow-up plans.
[1] H. Sung, J. Ferlay, R. L. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal, F. Bray, "Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries," CA: A Cancer Journal for Clinicians, vol. 71 no. 3, pp. 209-249, DOI: 10.3322/caac.21660, 2021.
[2] J. Zhou, S. Huichuan, W. Zheng, C. Wenming, "Guidelines for the diagnosis and treatment of hepatocellular carcinoma," Liver Cancer, vol. 9 no. 6, pp. 682-720, 2020.
[3] P. R. Galle, A. Forner, J. M. Llovet, V. Mazzaferro, F. Piscaglia, J. L. Raoul, P. Schirmacher, V. Vilgrain, "EASL clinical practice guidelines: management of hepatocellular carcinoma," Journal of Hepatology, vol. 69 no. 1, pp. 182-236, DOI: 10.1016/j.jhep.2018.03.019, 2018.
[4] Y. Sun, L. Wu, Y. Zhong, K. Zhou, Y. Hou, Z. Wang, Z. Zhang, J. Xie, C. Wang, D. Chen, Y. Huang, X. Wei, Y. Shi, Z. Zhao, Y. Li, Z. Guo, Q. Yu, L. Xu, G. Volpe, S. Qiu, J. Zhou, C. Ward, H. Sun, Y. Yin, X. Xu, X. Wang, M. A. Esteban, H. Yang, J. Wang, M. Dean, Y. Zhang, S. Liu, X. Yang, J. Fan, "Single-cell landscape of the ecosystem in early-relapse hepatocellular carcinoma," Cell, vol. 184 no. 2, pp. 404-421.e16, DOI: 10.1016/j.cell.2020.11.041, 2021.
[5] M. Houseman, M. Y. Y. Huang, M. Huber, M. Staiger, L. Zhang, A. Hoffmann, C. Lippuner, F. Stuber, "Flow cytometry-based high-throughput RNAi screening for miRNAs regulating MHC class II HLA-DR surface expression," European Journal of Immunology, vol. 52 no. 9, pp. 1452-1463, DOI: 10.1002/eji.202149735, 2022.
[6] B. E. Lippitz, "Cytokine patterns in patients with cancer: a systematic review," The Lancet Oncology, vol. 14 no. 6, pp. 218-228, DOI: 10.1016/s1470-2045(12)70582-x, 2013.
[7] S. A. Erokhina, M. A. Streltsova, L. M. Kanevskiy, M. V. Grechikhina, A. M. Sapozhnikov, E. I. Kovalenko, "HLA-DR-expressing NK cells: effective killers suspected for antigen presentation," Journal of Leukocyte Biology, vol. 109 no. 2, pp. 327-337, DOI: 10.1002/jlb.3ru0420-668rr, 2021.
[8] Y. H. Lee, Y. S. Hyun, H. A. Jo, I. C. Baek, S. M. Kim, H. J. Sohn, T. G Kim, "Comprehensive analysis of mycobacterium tuberculosis antigen-specific CD4 + T cell responses restricted by single HLA class II allotype in an individual," Frontiers in Immunology, vol. 13,DOI: 10.3389/fimmu.2022.897781, 2022.
[9] R. Osuna-Gómez, C. Arqueros, C. Galano, M. Mulet, C. Zamora, A. Barnadas, S. Vidal, "Effector mechanisms of CD8+ HLA-DR+ T cells in breast cancer patients who respond to neoadjuvant chemotherapy," Cancers, vol. 13 no. 24,DOI: 10.3390/cancers13246167, 2021.
[10] L. Váróczy, L. Gergely, Z. Miltenyi, M. Aleksza, A. Illes, "Can CD3+/HLA-DR+ activated T cells predict the prognosis of non-Hodgkin's lymphoma patients?," Immunology Letters, vol. 97 no. 1, pp. 155-157, DOI: 10.1016/j.imlet.2004.10.005, 2005.
[11] P. Y. Chen, C. Y. J. Wu, J. H. Fang, H. C. Chen, L. Y. Feng, C. Y. Huang, K. C. Wei, J. Y. Fang, C. Y Lin, "Functional change of effector tumor-infiltrating CCR5 + CD38 + HLA-DR + CD8 + T cells in glioma microenvironment," Frontiers in Immunology, vol. 10,DOI: 10.3389/fimmu.2019.02395, 2019.
[12] Z. Zong, J. Zou, R. Mao, C. Ma, N. Li, J. Wang, X. Wang, H. Zhou, L. Zhang, Y. Shi, "M1 macrophages induce PD-L1 expression in hepatocellular carcinoma cells through IL-1 β signaling," Frontiers in Immunology, vol. 10,DOI: 10.3389/fimmu.2019.01643, 2019.
[13] A. Machicote, S. Belen, P. Baz, L. A. Billordo, L. Fainboim, "Human CD8 + HLA-DR + regulatory T cells, similarly to classical CD4 + Foxp3 + cells, suppress immune responses via PD-1/PD-L1 Axis," Frontiers in Immunology, vol. 9,DOI: 10.3389/fimmu.2018.02788, 2018.
[14] K. Matoba, N. Iizuka, T. Gondo, T. Ishihara, H. Yamada-Okabe, T. Tamesa, N. Takemoto, K. Hashimoto, K. Sakamoto, T. Miyamoto, S. Uchimura, Y. Hamamoto, M. Oka, "Tumor HLA-DR expression linked to early intrahepatic recurrence of hepatocellular carcinoma," International Journal of Cancer, vol. 115 no. 2, pp. 231-240, DOI: 10.1002/ijc.20860, 2005.
[15] W. Huang, D. Ye, W. He, X. He, X. Shi, Y. Gao, "Activated but impaired IFN- γ production of mucosal-associated invariant T cells in patients with hepatocellular carcinoma," Journal for ImmunoTherapy of Cancer, vol. 9 no. 11,DOI: 10.1136/jitc-2021-003685, 2021.
[16] X. Wei, W. Xie, W. Yin, M. Yang, A. R. Khan, R. Su, W. Shu, B. Pan, G. Fan, K. Wang, F. Yang, D. Lu, C. Li, L. Pan, B. Cen, H. Xie, L. Zhuang, S. Zheng, X. Zeng, W. Chen, X. Xu, "Prediction of tumor recurrence by distinct immunoprofiles in liver transplant patients based on mass cytometry," American journal of cancer research, vol. 12 no. 9, pp. 4160-4176, 2022.
[17] D. Agdashian, M. ElGindi, C. Xie, M. Sandhu, D. Pratt, D. E. Kleiner, W. D. Figg, J. A. Rytlewski, C. Sanders, E. C. Yusko, B. Wood, D. Venzon, G. Brar, A. G. Duffy, T. F. Greten, F. Korangy, "The effect of anti-CTLA4 treatment on peripheral and intra-tumoral T cells in patients with hepatocellular carcinoma," Cancer Immunology, Immunotherapy, vol. 68 no. 4, pp. 599-608, DOI: 10.1007/s00262-019-02299-8, 2019.
[18] A. Granito, L. Bolondi, "Non-transplant therapies for patients with hepatocellular carcinoma and Child-Pugh-Turcotte class B cirrhosis," The Lancet Oncology, vol. 18 no. 2, pp. 101-112, DOI: 10.1016/s1470-2045(16)30569-1, 2017.
[19] C. Zheng, L. Zheng, J. K. Yoo, H. Guo, Y. Zhang, X. Guo, B. Kang, R. Hu, J. Y. Huang, Q. Zhang, Z. Liu, M. Dong, X. Hu, W. Ouyang, J. Peng, Z. Zhang, "Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing," Cell, vol. 169 no. 7, pp. 1342-1356.e16, DOI: 10.1016/j.cell.2017.05.035, 2017.
[20] Q. Zhang, Y. He, N. Luo, S. J. Patel, Y. Han, R. Gao, M. Modak, S. Carotta, C. Haslinger, D. Kind, G. W. Peet, G. Zhong, S. Lu, W. Zhu, Y. Mao, M. Xiao, M. Bergmann, X. Hu, S. P. Kerkar, A. B. Vogt, S. Pflanz, K. Liu, J. Peng, X. Ren, Z. Zhang, "Landscape and dynamics of single immune cells in hepatocellular carcinoma," Cell, vol. 179 no. 4, pp. 829-845.e20, DOI: 10.1016/j.cell.2019.10.003, 2019.
[21] L. Circelli, A. Petrizzo, M. Tagliamonte, R. Heidenreich, M. L. Tornesello, F. M. Buonaguro, L. Buonaguro, "Immunological effects of a novel RNA-based adjuvant in liver cancer patients," Cancer Immunology, Immunotherapy, vol. 66 no. 1, pp. 103-112, DOI: 10.1007/s00262-016-1923-5, 2017.
[22] J. Waidhauser, P. Nerlinger, T. T. Arndt, S. Schiele, F. Sommer, S. Wolf, P. Lohr, S. Eser, G. Muller, R. Claus, B. Markl, A. Rank, "Alterations of circulating lymphocyte subsets in patients with colorectal carcinoma," Cancer Immunology, Immunotherapy, vol. 71 no. 8, pp. 1937-1947, DOI: 10.1007/s00262-021-03127-8, 2022.
[23] K. Trieb, T. Lechleitner, S. Lang, R. Windhager, R. Kotz, S. Dirnhofer, "Evaluation of HLA-DR expression and T-lymphocyte infiltration in osteosarcoma," Pathology, Research & Practice, vol. 194 no. 10, pp. 679-684, DOI: 10.1016/s0344-0338(98)80126-x, 1998.
[24] B. Stankovic, A. K. B. Heidi, S. Renate, "Immune cell composition in human non-small cell lung cancer," Front Immunol, vol. 9, 2018.
[25] S. Suthen, C. J. Lim, P. H. D. Nguyen, C. Dutertre, H. L. H. Lai, M. Wasser, C. Chua, T. K. H. Lim, W. Q. Leow, T. J. Loh, W. K. Wan, Y. H. Pang, G. Soon, P. C. Cheow, J. H. Kam, S. Iyer, A. Kow, W. L. Tam, T. W. H. Shuen, H. C. Toh, Y. Y. Dan, G. K. Bonney, C. Y. Chan, A. Chung, B. K. P. Goh, W. Zhai, F. Ginhoux, P. K. H. Chow, S. Albani, V. Chew, "Hypoxia-driven immunosuppression by Treg and type-2 conventional dendritic cells in HCC," Hepatology, vol. 76 no. 5, pp. 1329-1344, DOI: 10.1002/hep.32419, 2022.
[26] L. A. Ormandy, T. Hillemann, H. Wedemeyer, M. P. Manns, T. F. Greten, F. Korangy, "Increased populations of regulatory T cells in peripheral blood of patients with hepatocellular carcinoma," Cancer Research, vol. 65 no. 6, pp. 2457-2464, DOI: 10.1158/0008-5472.can-04-3232, 2005.
[27] C. M. Bonilla, N. A. McGrath, J. Fu, C. Xie, "Immunotherapy of hepatocellular carcinoma with infection of hepatitis B or C virus," Hepatoma Res, vol. 6,DOI: 10.20517/2394-5079.2020.58, 2020.
[28] P. Lapierre, A. Lamarre, "Regulatory T cells in autoimmune and viral chronic hepatitis," Journal of Immunology Research, vol. 2015,DOI: 10.1155/2015/479703, 2015.
[29] A. Granito, L. Muratori, C. Lalanne, C. Quarneti, S. Ferri, M. Guidi, M. Lenzi, P. Muratori, "Hepatocellular carcinoma in viral and autoimmune liver diseases: role of CD4+ CD25+ Foxp3+ regulatory T cells in the immune microenvironment," World Journal of Gastroenterology, vol. 27 no. 22, pp. 2994-3009, DOI: 10.3748/wjg.v27.i22.2994, 2021.
[30] M. Garnelo, A. Tan, Z. Her, J. Yeong, C. J. Lim, J. Chen, K. H. Lim, A. Weber, P. Chow, A. Chung, L. L. P. Ooi, H. C. Toh, M. Heikenwalder, I. O. L. Ng, A. Nardin, Q. Chen, J. P. Abastado, V. Chew, "Interaction between tumour-infiltrating B cells and T cells controls the progression of hepatocellular carcinoma," Gut, vol. 66 no. 2, pp. 342-351, DOI: 10.1136/gutjnl-2015-310814, 2017.
[31] B. Zhang, A. Vogelzang, M. Miyajima, Y. Sugiura, Y. Wu, K. Chamoto, R. Nakano, R. Hatae, R. J. Menzies, K. Sonomura, N. Hojo, T. Ogawa, W. Kobayashi, Y. Tsutsui, S. Yamamoto, M. Maruya, S. Narushima, K. Suzuki, H. Sugiya, K. Murakami, M. Hashimoto, H. Ueno, T. Kobayashi, K. Ito, T. Hirano, K. Shiroguchi, F. Matsuda, M. Suematsu, T. Honjo, S Fagarasan, "B cell-derived GABA elicits IL-10+ macrophages to limit anti-tumour immunity," Nature, vol. 599 no. 7885, pp. 471-476, DOI: 10.1038/s41586-021-04082-1, 2021.
[32] P. R. Galle, F. Foerster, M. Kudo, S. L. Chan, J. M. Llovet, S. Qin, W. R. Schelman, S. Chintharlapalli, P. B. Abada, M. Sherman, A. X Zhu, "Biology and significance of alpha‐fetoprotein in hepatocellular carcinoma," Liver International, vol. 39 no. 12, pp. 2214-2229, DOI: 10.1111/liv.14223, 2019.
[33] W. Zhu, Y. Peng, L. Wang, Y. Hong, X. Jiang, Q. Li, H. Liu, L. Huang, J. Wu, E. Celis, T. Merchen, E. Kruse, Y. He, "Identification of α -fetoprotein-specific T-cell receptors for hepatocellular carcinoma immunotherapy," Hepatology, vol. 68 no. 2, pp. 574-589, DOI: 10.1002/hep.29844, 2018.
[34] T. Tamai, E. Mizukoshi, M. Kumagai, T. Terashima, N. Iida, M. Kitahara, T. Shimakami, K. Kitamura, K. Arai, T. Yamashita, Y. Sakai, T. Yamashita, M. Honda, K. Fushimi, S. Kaneko, "A novel α -fetoprotein-derived helper T-lymphocyte epitope with strong immunogenicity in patients with hepatocellular carcinoma," Scientific Reports, vol. 10 no. 1,DOI: 10.1038/s41598-020-60843-4, 2020.
[35] C. Zang, Y. Zhao, L. Qin, G. Liu, J. Sun, K. Li, Y. Zhao, S. Sheng, H. Zhang, N. He, P. Zhao, Q. Wang, X. Li, Y. Peng, T. Dong, Y. Zhang, "Distinct tumour antigen-specific T-cell immune response profiles at different hepatocellular carcinoma stages," BMC Cancer, vol. 21 no. 1,DOI: 10.1186/s12885-021-08720-9, 2021.
[36] T. Flecken, N. Schmidt, S. Hild, E. Gostick, O. Drognitz, R. Zeiser, P. Schemmer, H. Bruns, T. Eiermann, D. A. Price, H. E. Blum, C. Neumann‐Haefelin, R. Thimme, "Immunodominance and functional alterations of tumor-associated antigen-specific CD8+ T-cell responses in hepatocellular carcinoma," Hepatology, vol. 59 no. 4, pp. 1415-1426, DOI: 10.1002/hep.26731, 2014.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2023 Rui-Qian Gao et al. This work is licensed under http://creativecommons.org/licenses/by/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Background. HLA-DR+ T cell, accounting for 1.2%–5.8% of peripheral lymphocyte, is a type of activated T lymphocyte. This retrospective study aimed to evaluate the prognostic value of HLA-DR+ T cell for progression-free survival (PFS) and overall survival (OS) in hepatocellular carcinoma (HCC) patients after curative surgery. Patients and Methods. Clinicopathological data of 192 patients who underwent curative resection for hepatocellular carcinoma in the affiliated hospital of Qingdao University between January 2013 and December 2021 were collected and analyzed. Statistical tests used in this study were the chi-square test and Fisher’s exact test. The prognostic value of the HLA-DR+ T cell ratio was analyzed using univariate and multivariate Cox regression analyses. The Kaplan–Meier curves were drawn by the R programming language. Results. HCC patients were divided into high (≥5.8%) and low (<5.8%) HLADR+ T cell ratio groups. Cox regression analysis indicated that a high HLA-DR+ T cell ratio was positively related to the PFS in HCC patients (
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details

1 Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
2 Department of Cosmetic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China