eISSN: 2299-0054
ISSN: 1895-4588
Videosurgery and Other Miniinvasive Techniques
Current issue Archive Manuscripts accepted About the journal Supplements Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
4/2023
vol. 18
 
Share:
Share:
General surgery
Original paper

Transarterial chemoembolization with insertion of radioactive seeds for hepatocellular carcinoma

You-Bin Wang
1
,
Wei Zhang
2
,
Le Bao
1
,
Yun Lu
3
,
Jiao Hong
3

  1. Department of Interventional Radiology, Xuzhou Cancer Hospital, Xuzhou, China
  2. Department of Liver Disease, Daxing Hospital, Xi’an, China
  3. Department of Radiology, Xuzhou Central Hospital, Xuzhou, China
Videosurgery Miniinv 2023; 18 (4): 645–654
Online publish date: 2023/09/22
Article file
- Transarterial.pdf  [0.21 MB]
Get citation
 
PlumX metrics:
 

Introduction

Hepatocellular carcinoma (HCC) is among the most prevalent malignancies and the second leading cause of cancer-associated death at the global level [1, 2]. While hepatectomy is the optimal approach to HCC patient treatment, 30% or fewer patients diagnosed with HCC are eligible for curative treatment owing to advanced disease staging, limited liver reserve capacity, or lack of access to liver transplantation [35].

In patients with inoperable HCC, transarterial chemoembolization (TACE) is commonly employed as an alternative therapeutic modality that is recommended as a first-line management approach for stage B patients under the Barcelona Clinic Liver Cancer (BCLC) guidelines [68]. TACE and subsequent percutaneous ablation (PA) can effectively improve HCC patient outcomes [5], and patients who undergo TACE combined with PA reportedly exhibit significant improvements in progression-free survival (PFS) (45 vs. 4 months) and overall survival (OS) (59 vs. 16 months) relative to those patients treated via TACE alone [5]. According to the BCLC guidelines, PA is primarily used to treat HCC patients with stage A disease [8]. In individuals exhibiting tumors proximal to the liver surface, diaphragm, gallbladder, or major vessels, however, PA is not an appropriate approach [9]. In these patients, some studies have employed radioactive seed insertion (RSI) as an alternative therapeutic strategy [10, 11]. To date there have only been a relatively small number of studies focused on the combination of TACE and RSI as a means of treating HCC.

Aim

This study was designed with the goal of comparing the clinical efficacy and long-term outcomes associated with TACE plus RSI to those associated with TACE alone.

Material and methods

Patient selection

The Institutional Review Board of our institute approved this study and waived the need for written informed consent. From January 2018 to December 2021, a total of 80 patients with HCC underwent treatment in our center via either TACE plus RSI (n = 39) or TACE alone (n = 41). To be eligible for study inclusion, patients had to (a) be diagnosed with HCC, (b) have inoperable disease or have declined to undergo hepatectomy, (c) have 3 or fewer tumors, and (d) not be candidates for PA. Patients were excluded if they (a) had undergone any prior treatments for HCC, (b) exhibited diffuse HCC, or (c) exhibited disease of BCLC stage C or higher. Initially, all patients in this study were scheduled for combined treatment (TACE + RSI) before TACE. However, 41 patients did not receive RSI because of their poor family economic condition and they only received TACE.

Diagnosis

HCC was diagnosed based on contrast-enhanced computed tomography (CT) and/or magnetic resonance imaging (MRI) results exhibiting a pattern of enhancement consistent with arterial hypervascularity and venous/delayed phase washout [12]. In cases where these typical imaging findings were not observed, patients underwent percutaneous biopsy. HCC patients were not candidates for PA in cases where the tumor was < 5 mm from adjacent organs [9].

TACE treatment

TACE was performed under local anesthesia with fluoroscopic guidance using a combination of 5-fluorouracil (150 mg), mitomycin (10 mg), epirubicin (50 mg), and lipiodol (10–20 ml).

Combined TACE and RSI treatment

TACE was performed prior to RSI in all cases. Prior to TACE, abdominal CT images were transferred into the treatment-planning system (TPS; Fei-Tian, Beijing, China) in order to approximate the number of radioactive 125I seeds necessary and to optimize the distribution of those seeds. After manually defining tumor contours on axial CT slices, the TPS system was used to establish the gross tumor volume (GTV). The radiation dose for all patients was in the range 100–140 Gy, with at least 90% of the GTV receiving the selected dose of radioactivity.

All RSI procedures were performed 14 days after TACE with guidance from a 64-row CT instrument (Siemens, Erlangen, Germany) (Photo 1). Multiple 18G puncture needles were inserted into the tumor in accordance with the design of the TPS system, after which 125I seeds were introduced into the tumor at a spacing of 5–10 mm based on the established treatment plan. The needles were withdrawn so that these seeds were deposited in the needle pathways. A further round of CT scanning was used to confirm seed distributions. After the insertion of 125I seeds, we routinely used hemostatic drugs for 1 day to prevent bleeding.

Photo 1

Procedures of combined TACE and RSI treatment for HCC. A – Preoperative contrast-enhanced CT showed the HCC (arrow) at the right lobe. B – TACE treatment for HCC. C, D – Procedures of CT-guided RSI after TACE. E – Post-operative contrast-enhanced CT indicated that the treatment response was CR

/f/fulltexts/WIITM/51497/WIITM-18-51497-g003_min.jpg

Follow-up

Patient follow-up was performed at 1 and 3 months after treatment, and every 3 months thereafter. Follow-up was performed until December 2022. Follow-up analyses included routine blood analyses, liver function tests, MRI or contrast-enhanced CT imaging, and analyses of α-fetoprotein (AFP) levels. If evidence of MRI/CT enhancement in the treated tumor area was observed, TACE was repeated.

Definitions

Treatment responses at 1 month following treatment were evaluated using the modified Response Evaluation Criteria in Solid Tumors (mRECIST) criteria [13]. For details regarding the criteria used to define complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD), see Table I. PFS was defined as the period of time between treatment and disease progression or death, while OS was the period of time from treatment to death or most recent follow-up.

Table I

Modified response evaluation criteria in solid tumors for hepatocellular carcinoma

ParameterDefinitions
Complete responseDisappearance of any intratumoral arterial enhancement in all target lesions
Partial responseAt least a 30% decrease in the sum of diameters of viable (enhancement in the arterial phase) target lesions, taking as reference the baseline sum of the diameters of target lesions
Stable diseaseAny cases that do not qualify for either partial response or progressive disease
Progression diseaseAn increase of at least 20% in the sum of the diameters of viable (enhancing) target lesions, taking as reference the smallest sum of the diameters of viable (enhancing) target lesions recorded since treatment started

Statistical analysis

SPSS 16.0 (SPSS, Inc., IL, USA) was used for all data analyses. Results were reported as means ± standard deviations when normally distributed and otherwise presented as medians (Q1; Q3), and were respectively compared with t-tests and Mann-Whitney U tests. When comparing categorical variables, χ2 tests or Fisher’s exact test were employed. Kaplan-Meier curves and log-rank tests were used for comparisons of OS and PFS rates. Multivariate Cox regression analyses were used to identify factors independently related to survival outcomes. The factors with p < 0.1 on univariate Cox regression analysis were included in the multivariate Cox regression analysis. P < 0.05 was used as the cut-off to define statistical significance.

Results

Patient characteristics

The baseline characteristics of patients in both groups were comparable (Table II). In total, 1790 125I seeds were placed in 39 patients in the combination group (45.9 seeds/patient). No treatment-related complications were reported for any patients.

Table II

Baseline data of the included patients

ParameterCombined treatment (n = 39)TACE alone (n = 41)P-value
Age [years]62.0 ±11.562.4 ±12.60.886
Gender:0.512
 Male2933
 Female108
Hepatitis types:0.200
 B2330
 C56
 None115
AFP value [ng/ml]:
 Before treatment191.2 (Q1: 28.7; Q3: 319.6)164.2 (Q1: 9.6; Q3: 688.9)0.954
 After treatment (at 1 month)84.9 (Q1: 12.1; Q3: 384.0)32.7 (Q1: 4.6; Q3: 366.7)0.405
AFP level:0.071
 ≤ 400 ng/ml3125
 > 400 ng/ml816
NLR:0.695
 ≤ 2.772629
 > 2.771312
Child-Pugh class:0.051
 A3733
 B28
BCLC stage:0.692
 A2528
 B1413
Number of tumors:0.678
 Single2225
 Multiple1716
Tumor diameter [cm]5.2 ±1.55.7 ±2.10.169
Target or immune therapy430.945

[i] AFP – α-fetoprotein, BCLC – Barcelona Clinic Liver Cancer, NLR – neutrophil-to-lymphocyte ratio, TACE – trans-arterial chemo-embolization.

Treatment response rates

Treatment response rates in both groups are summarized in Table III. Patients who underwent combination treatment exhibited significant improvements in both CR (59.0% vs. 22.0%, p = 0.001) and total response (92.3% vs. 58.5%, p = 0.001) rates relative to patients treated via TACE alone.

Table III

Treatment response between 2 groups

ParameterCombined treatment (n = 39)TACE alone (n = 41)P-value
CR23 (59.0%)9 (22.0%)0.001
PR13 (33.3%)15 (36.6%)0.761
SD1 (2.6%)14 (34.1%)0.001
PD2 (5.1%)3 (7.3%)0.686
Total response (CR + PR)36 (92.3%)24 (58.5%)0.001

[i] CR – complete response, PD – progressive disease, PR – partial response, SD – stable disease.

Treatment-related toxicity

There were no significant differences between the two patient groups with respect to the most common forms of treatment-related toxicity, which included fever (38.4% vs. 39.0%, p = 0.959), vomiting (33.3% vs. 43.9%, p = 0.332), and myelosuppression (28.2% vs. 29.3%, p = 0.916) (Table IV).

Table IV

Toxicity effects between 2 groups

VariableCombined treatment (n = 39)TACE alone (n = 41)P-value
Fever15 (38.4%)16 (39.0%)0.959
Vomit13 (33.3%)18 (43.9%)0.332
Myelosuppression11 (28.2%)12 (29.3%)0.916

Progression-free survival

Patients in the combination treatment group exhibited a significant increase in median PFS relative to patients who underwent TACE alone (13 vs. 7 months, p = 0.019, Figure 1). The respective 1- and 3-year PFS rates of patients who underwent combination treatment were 51.2% and 16.0%, whereas for patients who underwent TACE alone, these respective rates were 24.4% and 0.0%.

Figure 1

Comparison of PFS between combined and TACE alone groups

/f/fulltexts/WIITM/51497/WIITM-18-51497-g001_min.jpg

In univariate Cox regression analyses, AFP > 400 ng/ml (p = 0.048) and undergoing TACE alone (p = 0.028) were associated with a risk of shorter PFS. In a multivariate analysis, only undergoing TACE alone remained independently associated with the risk of shorter PFS (p = 0.033, Table V).

Table V

Predictors of progression-free survival

VariablesUnivariate analysisMultivariate analysis
Hazard ratio95% CIP-valueHazard ratio95% CIP-value
Age [years]0.9900.970–1.0100.310
Gender:
 Male1
 Female0.8060.439–1.4770.485
Hepatitis types:
 None1
 B1.0560.588–1.8980.854
 C1.1070.483–2.5360.810
AFP level:
 ≤ 400 ng/ml11
 > 400 ng/ml1.6771.005–2.7960.0481.6420.983–2.7430.058
NLR:
 ≤ 2.771
 > 2.770.7190.423–1.2220.222
Child-Pugh class:
 A1
 B1.3000.642–2.6330.467
BCLC stage:
 A1
 B0.9040.547–1.4930.693
Number of tumors:
 Single1
 Multiple1.0110.627–1.6330.963
Tumor diameter [cm]1.1130.974–1.2730.117
Treatment options:
 Combined treatment11
 TACE alone1.7151.061–2.7710.0281.6881.044–2.7310.033

[i] AFP – α-fetoprotein, BCLC – Barcelona Clinic Liver Cancer, NLR – neutrophil-to-lymphocyte ratio, TACE – trans-arterial chemo-embolization.

Overall survival

Over the course of follow-up, 17 and 37 patients in the combination and TACE only treatment groups died, respectively. Individuals in the combination treatment group exhibited significantly longer median OS relative to the TACE only treatment group (23 vs. 15 months, p = 0.005, Figure 2). The respective 1- and 3-year OS rates of patients who underwent combination treatment were 87.2% and 35.1%, whereas for patients who underwent TACE alone, these respective rates were 73.2% and 13.0%.

Figure 2

Comparison of OS between combined and TACE alone groups

/f/fulltexts/WIITM/51497/WIITM-18-51497-g002_min.jpg

In univariate Cox regression analyses, risk factors for lower OS included being male (p = 0.089), exhibiting an AFP level > 400 ng/ml (p = 0.049), having a larger tumor diameter (p = 0.004), and undergoing TACE alone (p = 0.008). In a multivariate regression analysis, only larger tumor diameter (p = 0.016) and TACE alone (p = 0.029) were associated with significantly reduced OS (Table VI).

Table VI

Predictors of overall survival

VariablesUnivariate analysisMultivariate analysis
Hazard ratio95% CIP-valueHazard ratio95% CIP-value
Age [years]1.0020.978–1.0260.896
Gender:
 Male11
 Female0.4990.224–1.1120.0890.5620.250–1.2620.163
Hepatitis types:
 None1
 B1.3000.645–2.6220.463
 C1.0290.372–2.8490.956
AFP level:
 ≤ 400 ng/ml11
 > 400 ng/ml1.7781.004–3.1500.0491.4570.801–2.6480.218
NLR:
 ≤ 2.771
 > 2.770.8900.488–1.6240.704
Child-Pugh class:
 A1
 B1.5990.751–3.4070.223
BCLC stage:
 A1
 B0.7130.393–1.2930.265
Number of tumors:
 Single1
 Multiple0.6930.392–1.2250.207
Tumor diameter [cm]1.2491.076–1.4510.0041.1951.034–1.3800.016
Treatment options:
 Combined treatment11
 TACE alone2.2241.230–4.0220.0081.9561.071–3.5740.029

[i] AFP – α-fetoprotein, BCLC – Barcelona Clinic Liver Cancer, NLR – neutrophil-to-lymphocyte ratio, TACE – trans-arterial chemo-embolization.

Discussion

While TACE remains a key component of HCC patient care under BCLC guidelines, many other therapeutic approaches can be combined with TACE to enhance its efficacy, including RS, PA, and targeted or immunotherapy [7, 11, 14, 15]. PA is commonly combined with TACE [37], but it is not well suited to use in cases where HCC tumors are proximal to adjacent organs that may otherwise experience thermal injury [9].

Currently, the insertion of 125I seeds is often employed as an approach to treating various forms of cancer [1618]. In contrast to more conventional radiotherapy, these 125I seeds offer advantages including their ability to directly contact the internal regions of the tumor, thereby ensuring the prolonged delivery of low-dose radiation in a sustained manner while remaining easy to manipulate [16]. By continuously emitting X-rays and γ-rays, 125I seeds can readily kill target tumor cells and suppress further tumor development [19]. Following 125I seed implantation, significant increases in peripheral blood CD3+ T cell, CD4+ T cell, natural killer cell, and regulatory T cell percentages have been reported in patients with cancer [19]. Circulating C3, C4, IgM, IgA, and IgG concentrations also reportedly rose, suggesting that 125I seeds can provoke robust cellular and humoral immunity [19].

Here, the relative clinical efficacy and associated long-term outcomes were compared for HCC patients who underwent combination TACE and RSI treatment or TACE alone. The CR and total response rates in the combination group were significantly higher than those of patients treated with TACE alone. In the combination treatment group, the total response rate was up to 92.3%, similar to that reported previously following combination TACE and PA treatment (85.9%) [5]. Hence, 125I seeds may facilitate superior therapeutic efficacy for TACE treatment. RSI can also efficiently treat residual HCC tumor cells following TACE [10, 11].

Combining TACE with RSI also contributed to significant improvements in HCC patient OS and PFS relative to TACE alone. The ability of low-dose irradiation derived from 125I seeds to damage and eliminate cycle-sensitive cells and to alter tumor cell distributions can further render HCC cells more sensitive to chemotherapy, resulting in improved long-term therapeutic efficacy [20]. Patients in the combination treatment group exhibited 1- and 3-year OS rates (87.2% and 35.1%) similar to those in a prior report focused on treating HCC with a combination of TACE and PA (85.9% and 32.6%) [21]. As such, RSI may offer a degree of therapeutic efficacy similar to that of PA when treating individuals diagnosed with HCC.

Cox regression analyses confirmed that combined treatment was predictive of the prolongation of patient OS and PFS. Several factors may account for the superior performance of combined therapeutic regimens. For one, tumor embolization can restrict the tumor blood flow [21]. In addition, TACE can generate high levels of ischemia and inflammatory edema in the peritumoral regions, contributing to greater brachytherapeutic benefits upon 125I seed insertion [10, 11]. The 125I seeds were also able to destroy hypovascular HCC tumor regions refractory to precision TACE treatment [10, 11]. A larger diameter was identified as a risk factor for poorer OS, as reported in another similar study focused on treating HCC via a combination of TACE and PA [21]. Larger tumors translate to greater difficulty when attempting to achieve complete necrotic tumor death.

The present results revealed that RSI was not associated with any additional side effects relative to TACE alone. This may be attributable to the use of TPS prior to RSI to select the optimal number and distribution of 125I seeds so as to minimize any radiation-related damage to adjacent healthy tissue.

There are some limitations to this study. Notably, this retrospective study is susceptible to a high risk of bias, underscoring the need for future prospective validation of these findings in a randomized controlled trial. Second, these results were derived from a single center, and a larger patient cohort derived from multiple centers will be important when conducting further follow-up research. Third, the majority of enrolled patients had previously experienced hepatitis infections, and so additional analyses will be required to ascertain whether combining TACE and RSI can effectively treat patients with HCC resulting from other etiologies.

Conclusions

The present data suggested that combining TACE and RSI can result in significant improvements in treatment response rates, OS, and PFS among HCC patients relative to TACE alone.

Acknowledgments

You-Bin Wang and Wei Zhang contributed equally to this work.

This study was supported by Xuzhou City Important Development Program (KC22150).

Conflict of interest

The authors declare no conflict of interest.

References

1 

Zhang FQ, Sun J, Gu XJ. Repeat resection versus percutaneous ablation for recurrent hepatocellular carcinoma: a meta-analysis. Videosurgery Miniinv 2023; 18: 1-10.

2 

Li Z, Yu Q, Lu X, et al. Efficacy of radiofrequency ablation versus laparoscopic liver resection for hepatocellular carcinoma in China: a comprehensive meta-analysis. Videosurgery Miniinv 2021; 16: 455-71.

3 

Pan T, Mu LW, Wu C, et al. Comparison of combined transcatheter arterial chemoembolization and CT-guided radiofrequency ablation with surgical resection in patients with hepatocellular carcinoma within the up-to-seven criteria: a multicenter case-matched study. J Cancer 2017; 8: 3506-13.

4 

Chen QF, Jia ZY, Yang ZQ, et al. Transarterial chemoembolization monotherapy versus combined transarterial chemoembolization-microwave ablation therapy for hepatocellular carcinoma tumors ≤5 cm: a propensity analysis at a single center. Cardiovasc Intervent Radiol 2017; 40: 1748-55.

5 

Ren Y, Cao Y, Ma H, et al. Improved clinical outcome using transarterial chemoembolization combined with radiofrequency ablation for patients in Barcelona clinic liver cancer stage A or B hepatocellular carcinoma regardless of tumor size: results of a single-center retrospective case control study. BMC Cancer 2019; 19: 983.

6 

Sheta E, El-Kalla F, El-Gharib M, et al. Comparison of single-session transarterial chemoembolization combined with microwave ablation or radiofrequency ablation in the treatment of hepatocellular carcinoma: a randomized-controlled study. Eur J Gastroenterol Hepatol 2016; 28: 1198-203.

7 

Yin X, Zhang L, Wang YH, et al. Transcatheter arterial chemoembolization combined with radiofrequency ablation delays tumor progression and prolongs overall survival in patients with intermediate (BCLC B) hepatocellular carcinoma. BMC Cancer 2014; 14: 849.

8 

Raoul JL, Forner A, Bolondi L, et al. Updated use of TACE for hepatocellular carcinoma treatment: how and when to use it based on clinical evidence. Cancer Treat Rev 2019; 72: 28-36.

9 

Wang H, Wu F, Wen M, et al. Clinical effect of TACE combined with 125I seed implantation and internal irradiation in the treatment of primary liver cancer in special areas. Contemp Med 2020; 26: 46-9.

10 

Li J, Zhang L, Sun Z, et al. Iodine-125 seed implantation for residual hepatocellular carcinoma or cholangiocellular carcinoma in challenging locations after transcatheter arterial chemoembolization: initial experience and findings. J Contemp Brachytherapy 2020; 12: 233-40.

11 

Li J, Zhang L, Xie Q, et al. 125I seeds implantation for treating residual hepatocellular carcinoma located beneath the diaphragm after transcatheter arterial chemoembolization. Brachytherapy 2019; 18: 420-5.

12 

Bruix J, Sherman M; American Association for the Study of Liver Diseases. Management of hepatocellular carcinoma: an update. Hepatology 2011; 53: 1020-2.

13 

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

14 

Fidelman N. A Leap from TACE to TACE potentiated by immune response modulation and angiogenesis inhibition for patients with intermediate-stage hepatocellular carcinoma: LEAP-012 phase 3 randomized control trial protocol. Cardiovasc Intervent Radiol 2022; 45: 413-4.

15 

Tan J, Fan W, Liu T, et al. TREM2+ macrophages suppress CD8+ T-cell infiltration after transarterial chemoembolisation in hepatocellular carcinoma. J Hepatol 2023; 79: 126-40.

16 

Gao FL, Wang Y, Lu J, et al. Insertion of I-125 seed-loaded stent for inoperable hilar cholangiocarcinoma. Videosurgery Miniinv 2021; 16: 678-85.

17 

Chen ZK, Fan J, Li FQ, et al. I-125 seeds with chemotherapy for progressive non-small-cell lung cancer after first-line treatment: a meta-analysis. J Cardiothorac Surg 2022; 17: 75.

18 

Langhans L, Klausen TL, Tvedskov TF, et al. Preparation and administration of I-125 labeled seeds for localization of nonpalpable breast lesions. Curr Radiopharm 2016; 9: 143-9.

19 

Pang Q, Zhou L, Hu XS, et al. Biliary stenting alone versus biliary stenting combined with 125I particles intracavitary irradiation for the treatment of advanced cholangiocarcinoma. Sci Rep 2019; 9: 11348.

20 

Gao FL, Wang Y, Huang XZ, et al. I-125 seeds brachytherapy with transcatheter arterial chemoembolization for subcapsular hepatocellular carcinoma. BMC Gastroenterol 2022; 22: 273.

21 

Zheng L, Li HL, Guo CY, et al. Comparison of the efficacy and prognostic factors of transarterial chemoembolization plus microwave ablation versus transarterial chemoembolization alone in patients with a large solitary or multinodular hepatocellular carcinomas. Korean J Radiol 2018; 19: 237-46.

Copyright: © 2023 Fundacja Videochirurgii This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
  
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.