This was a single-center retrospective study. Local institutional review board approval was obtained. We reviewed the electronic medical records of 95 consecutive patients with hepatitis B-related HCC and Vp4 PVTT, who were administered interventional therapy (125I seed strand [with or without stent] implantation plus transarterial chemoembolization) combined with systemic therapy (lenvatinib plus anti-PD-1 antibody) (group A) or interventional therapy (125I seed strand [with or without stent] implantation plus transarterial chemoembolization) combined with systemic therapy (lenvatinib) (group B) from December 2018 to June 2021. Before treatment initiation, the benefits, and potential adverse events (AEs) related to both combination regimens were explained thoroughly to the patients. The final choices were made by the patients. All patients provided written informed consent.
Patients
Intrahepatic HCC was diagnosed based on the American Association for the Study of Liver Disease guidelines [14]. According to the standard recommended by Shah et al [15], a PVTT was considered to be neoplastic if at least one of the following criteria was met: (a) expansion of the involved vessel (vessel diameter ≥ 1.8 cm for the MPV, ≥ 1.6 cm for the right portal vein (PV), or ≥ 1.8 cm for the left PV; (b) clear evidence of enhancement on dynamic contrast-enhanced CT images during the arterial phase of dynamic imaging, compared with baseline images (≥ 20 HU on CT). Otherwise, the PVTT was bland. The extent of PVTT was classified as follows: Vp0, no PVTT; Vp1, segmental PV invasion; Vp2, right anterior or posterior PV; Vp3, right or left PV; Vp4, main trunk and/or contralateral portal vein branch to the primarily involved lobe [16]. In this study, Vp4 PVTT was classified as follows: Vp4 I PVTT, tumor thrombus extended to main trunk of portal vein, but not extended to contralateral portal vein branch; Vp4 II PVTT, tumor thrombus extended to main trunk and contralateral portal vein branch.
Inclusion criteria were: (1) between 18 and 75 years of age; (2) a single tumor ≥ 5.0 cm or multiple nodular tumors > 3.0 cm; (3) Vp4 PVTT; (4) patent second-order branch of the portal vein prior to PVTT; (5) Child-Pugh class A or B; and (6) an Eastern Cooperative Group performance status (ECOG) score of 0–2. These points represent eligibility criteria for the treatment.
Exclusion criteria were: (1) Vp1-3 PVTT; (2) completely occluded portal vein; (3) hepatic encephalopathy, severe ascites, esophageal, gastric fundal variceal bleeding or other serious medical comorbidities; (4) previous local-regional therapy (transarterial chemoembolization, radiofrequency ablation [RFA], microwave ablation [MWA], cryoablation, yttrium-90 [90Y] radioembolization, stereotactic body radiotherapy [SBRT], hepatic artery infusion chemotherapy [HAIC], or liver transplantation); (5) previous systemic therapy (tyrosine kinase inhibitors [TKIs], systemic chemotherapy, or immunotherapy); or (6) malignant tumor other than HCC.
According to the inclusion and exclusion criteria, 75 patients were included in this study (Group A, n = 41; and Group B, n = 34; Fig. 1). Baseline characteristics are presented in Table 1.
Table 1
Baseline characteristics of the patients in 2 groups, n (%)
Characteristic | Group A (n = 41) | Group B (n = 34) | p-value |
Sex | | | 1.000 |
Male | 36(87.8) | 30(88.2) | |
Female | 5(12.2) | 4(11.8) | |
Age | | | 0.882 |
≥55y | 21(51.2) | 18(52.9) | |
<55y | 20(48.8) | 16(47.1) | |
Tumor size *(mm) | | | 0.321 |
≥10cm | 17(41.5) | 18(52.9) | |
<10cm | 24(58.5) | 16(47.1) | |
PVTT type * | | | 0.163 |
Vp4 I | 28(68.3) | 28(82.4) | |
Vp4 II | 13(31.7) | 6(17.6) | |
Child-Pugh class | | | 1.000 |
A | 39(95.1) | 32(94.1) | |
B | 2(4.9) | 2(5.9) | |
ECOG performance status | | | 0.401 |
0/1 | 39(95.1) | 30(88.2) | |
2 | 2(4.9) | 4(11.8) | |
Serum AFP level | | | 0.150 |
≥400 | 21(51.2) | 23(67.6) | |
<400 | 20(48.8) | 11(32.4) | |
Extrahepatic metastasis | | | 0.722 |
Yes | 5(12.2) | 3(8.8) | |
No | 36(87.8) | 31(91.2) | |
AFP = α-fetoprotein; ECOG = Eastern Cooperative Oncology Group; PVTT = portal vein tumor thrombus. |
*Tumor size, the maximum diameter of the largest target index lesion. |
*Vp4 I = tumor thrombus extended to main trunk of portal vein, but not extended to contralateral portal vein branch; Vp4 II = tumor thrombus extended to main trunk and contralateral portal vein branch |
Interventional Therapy
The protocol for 125I seed strand (with or without stent) implantation and transarterial chemoembolization procedure was the same in both groups.
125 I seed properties
Model 6711 125I seeds (XinKe; Shanghai, China) were used in this study. The radioactivity of each 125I seed was 25.9 MBq with a half-life of 59.4 days. Principal photon emissions were 27.4 and 35.5 keV X-rays and gamma-rays, respectively. The half-value thickness of the tissue for 125I seed was 17 mm, and the incipient dose rate was 7 cGy/h. The 240-day intended dose at 10 mm from the axis of the 125I seed strand was calculated with a radiation calculation software (version 0.1) based on the American Association of Physicists in Medicine TG43U1 brachytherapy formalism [17].
The production process of 125I seed strands was as follows [9]: (a) a 4-F flexible compliant cannula (Boston Scientific, Natick, Massachusetts) was sealed at one end with an alcohol lamp; (b) 125I seeds were loaded into the tube linearly, and the number of 125I seeds loaded (N) was determined as N = L / 4.5 + 4, where L (mm) is the length of the obstructed PV; (c) the other end was cut and sealed.
125 I seed strand (with or without stent) implantation
Vp4 I PVTT cases
In the 2 groups, patients with Vp4 I PVTT received 125I seed strand and intra-main trunk of portal vein (MPV) stent implantations. The method of 125I seed strand and stent implantations in contralateral branch and MPV was adapted the technique proposed by Luo et al [9]. The contralateral secondorder branch was punctured with a 21-gauge Chiba needle (Cook, Bloomington, Indiana) under ultrasound guidance, followed by the insertion of a 0.018-inch wire (Cook) into the MPV. A 6-F NEEF set (Cook) was introduced into the MPV over the wire. Through the outer cannula of the 6-F NEEF set, a 0.035-inch wire (Terumo, Tokyo, Japan) combined with a 4-F Cobra catheter (Cordis, Miami Lakes, Florida) was manipulated across the obstructed MPV into the superior mesenteric vein (SMV). The 4-F Cobra catheter and the 6-F NEEF set were removed, and a 6-F sheath (Cordis) was introduced through the wire. Portography was performed to measure the diameter and length of the obstructed MPV by a 4-F pigtail catheter (Cook) placed in the SMV. Two 0.035-inch stiff wires (Terumo) were inserted into the SMV through the 6F sheath. After the sheath removal, the 6-F NEFF set and a self-expandable stent (Bard, New Jersey, America) of appropriate size were introduced into the MPV over one of the stiff wires, respectively. The stent was deployed from the distal MPV into the contralateral fist-order branch of the portal vein. A 125I seed strand was delivered to the target position via the outer cannula of the 6-F NEFF set and released between the stent and the MPV. Portography was repeated through the 4-F pigtail catheter (Cook). The puncture tract was next occluded by 3 × 140 mm Nester coils (Cook).
Then, another 125I seed strand was implanted into the ipsilateral portal vein branch. This method was adapted the technique proposed by Zhang et al [18]. The ipsilateral second-order portal vein branch was punctured with a 21-gauge Chiba needle (Cook) under ultrasound guidance. With confirmed access, a 0.018-inch wire (Cook) was manipulated to cross the obstructed segment of ipsilateral portal vein branch and positioned into the MPV. A 6-F NEFF set (Cook) was introduced into the ipsilateral portal vein over the 0.018-inch wire. Then, the 0.018-inch wire was replaced by a 0.035-inch wire (Cook). Another 125I seed strand was pushed to the target position of PVTT in ipsilateral portal vein branch by the inner core of the 6-F NEFF set. Then, the outer cannula of the 6-F sheath was retreated slowly until the strand was completely released. The position of the strand should completely cover the macroaxis of PVTT in ipsilateral portal vein branch. The trailing part of the strand was fixed into the hepatic parenchyma through the liver elasticity. Finally, the transhepatic puncture track was occluded by 3 × 140 mm Nester coils (Cook) (Fig. 2a-d).
Vp4 II PVTT cases
Patients with Vp4 II PVTT received 125I seed strand (without stent) implantation. The contralateral second-order portal vein branch was punctured with a 21-gauge Chiba needle (Cook) under ultrasound guidance. A 125I seed strand was implanted from MPV to contralateral branch by the same method which performed in ipsilateral portal vein branch of Vp4 I PVTT cases. The position of this strand should completely cover the macroaxis of PVTT in MPV and contralateral branch. Then, the ipsilateral second-order branch was punctured, another 125I seed strand was implanted into the ipsilateral portal vein branch by the same method and this strand should completely cover the macroaxis of PVTT in ipsilateral portal vein branch (Fig. 3a-d).
Transarterial Chemoembolization
Transarterial chemoembolization was provided after the 125I seed strand (with or without stent) implantation immediately. This method was adapted the technique proposed by Zhang et al [18]. Hepatic angiography was performed to evaluate tumor vascularity. A chemotherapeutic emulsion consisting of 10–50 mg epirubicin (Pharmorubicin; Pfizer, New York) and 4–10 ml lipiodol (Lipiodol; Guerbet, Roissy, France) was slowly injected at a rate of 0.5-1.0 mL/min under fluoroscopic guidance via a 2.4-F microcatheter (Merit Medical, USA) until saturation of the tumor-supplying arteries. The dose of iodized oil was calculated as 1.0-1.5 ml per cm in dimeter of tumor. If the tumor had a rich blood supply, more oil was needed and vice versa. The dose of epirubicin was calculated as 10–50 mg/m2 of body surface area. Then, 350-560-µm gelatin sponge particles (Jingling, Jiangsu, China) were used to embolize the residual feeding artery of tumor.
Systemic Therapy
In group A, all patients received lenvatinib 3 days after the first interventional procedure. Lenvatinib was orally administered at 8 mg/day in patients weighing < 60 kg and at 12 mg/day in those ≥ 60 kg. In patients developing AEs (grade ≥ 2), dose reduction or temporary interruption was maintained until the symptoms resolved to grade 0–1. AEs were assessed by the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE v4.03).
In group A, patients received anti-PD-1 inhibitor injection in 3–7 days after the first interventional procedure. They were monitored regularly, including repeat safety evaluation 2–3 days prior to each anti-PD-1 antibody treatment cycle. Anti-PD-1 antibodies were intravenously administered as follows: nivolumab (Bristol-Myers Squibb, USA) 3 mg/kg or camrelizumab (Hengrui Medicine, China) 200 mg [19] every 2 weeks, or pembrolizumab (MSD, USA) 200 mg, sintilimab (Innovent Biologics, China) 200 mg [20] or toripalimab (Junshi Bioscience, China) 240 mg [21] every 3 weeks. In patients developing AEs (grade 2), temporary interruption was maintained until the symptoms resolved to grade 0–1. In patients developing AEs (grade 3–4), anti-PD-1 inhibitor injection was ceased permanently.
Post-procedure Management
Single photon-emission computer tomography combined with CT (SPECT/CT) was performed on day 1 to evaluate the location and distribution of radiation by the 125I seed strand. Laboratory tests (including hepatic and renal functions, complete blood count, and coagulation parameters) were performed 3–7 days after the initial procedure.
Follow-up and Repeated Transarterial Chemoembolization
The follow-up period was defined as the time from the initial interventional procedure to death or the last follow-up date. Each follow-up session included a detailed medical history, physical examination, laboratory tests, and contrast-enhanced CT or MRI. Follow-up was conducted every 30–45 days after the initial procedure. Patients with residual viable tumors or recurrent tumors in the hepatic parenchyma on CT or MRI images underwent repeated transarterial chemoembolization in case the Child-Pugh status remained at class A or B. No other interventional therapy was provided except for transarterial chemoembolization.
Evaluation
The primary endpoint was overall survival (OS, defined as the time from the initial interventional procedure to death from any cause). Secondary endpoint was progression-free survival (PFS, defined as the time from the initial interventional procedure until tumor progression or death from any cause).
Intrahepatic tumor response was classified as complete response (CR), partial response (PR), stable disease (SD), or progressive disease (PD) according to modified Response Evaluation Criteria in Solid Tumor (mRECIST) criteria.
In Vp4 I PVTT patients who administered 125I seed strand and stent implantations, treatment response in PVTT was evaluated by the rate of stent occlusion and the median stent patency time. Because PVTT changed into an irregular shape and was positioned between the stent and the portal vein wall after stent implantation, it is hard to calculate the diameter of tumor thrombus precisely. Stent occlusion was defined with no contrast medium detected inside the stent on the portal phase of contrast-enhanced CT or contrast-enhanced MRI images, or no blood flow signal detected by color doppler flow imaging (CDFI). Stent patency time was determined from the day of stent placement to stent occlusion or the day of last follow-up.
In Vp4 II PVTT patients who administered 125I seed strand (without stent) implantation, treatment response in PVTT was evaluated by contrast-enhanced CT or MRI, and the product of the largest perpendicular diameters of the PVTT was calculated and compared to the initial value, irrespective of the vascular site [22]. A complete response (CR) was defined as complete PVTT disappearance, a partial response (PR) as a ≥ 30% decrease of PVTT diameter, stable disease (SD) as between a 30% decrease and a 20% increase in thrombus diameter, and progressive disease (PD) as ≥ 20% increase in PVTT diameter.
Overall response rate (ORR) was defined as the percentage of patients who had a best tumor response rating of CR or PR. Disease control rate (DCR) was defined as the percentage of patients achieving CR, PR or SD as the best tumor response.
Statistical Analysis
All statistical analysis was performed with SPSS (version 23.0, Chicago, Illinois). Continuous variables were presented as mean ± standard deviation and were compared by independent or paired samples t test. Categorical variables were presented as frequency and compared by the Chi-square test. Median PFS (mPFS), mOS and median stent patency times were analyzed by the Kaplan-Meier method and the log-rank test. A p-value < 0.05 was considered statistically significant. Factors statistically significant at p-value < 0.10 in univariate analysis were entered a multivariable Cox proportional hazards model.