Next Article in Journal
Crohn’s Disease and Jejunal Artery Aneurysms: A Report of the First Case and a Review of the Literature
Next Article in Special Issue
PD-L1 Gene Polymorphisms rs822336 G>C and rs822337 T>A: Promising Prognostic Markers in Triple Negative Breast Cancer Patients
Previous Article in Journal
External Apical Root Resorption in Orthodontic Patients Who Practice Combat Sports: A Case-Control Observational Pilot Study
Previous Article in Special Issue
Loss of Expression of Antiangiogenic Protein FKBPL in Endometrioid Endometrial Carcinoma: Implications for Clinical Practice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Hepatic Arterial Infusion Chemotherapy with Oxaliplatin Plus Raltitrexed as an Alternative Option in Advanced Hepatocellular Carcinoma Patients with Failure of, or Unsuitability for, Transarterial Chemoembolization

1
Department of Hepatic Oncology, Xiamen Clinical Research Center for Cancer Therapy, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, China
2
Key Laboratory for Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, The Liver Cancer Institute, Zhongshan Hospital, Shanghai Medical School, Fudan University, Shanghai 200032, China
3
Center for Evidence-Based Medicine, Shanghai Medical School, Fudan University, Shanghai 200032, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Medicina 2022, 58(10), 1343; https://doi.org/10.3390/medicina58101343
Submission received: 29 July 2022 / Revised: 16 September 2022 / Accepted: 20 September 2022 / Published: 24 September 2022

Abstract

:
Background and Objectives: To assess the efficacy and safety of hepatic arterial infusion chemotherapy (HAIC) with oxaliplatin plus raltitrexed (HAICROX) as an alternative treatment option for advanced hepatocellular carcinoma (HCC) patients who are ineligible for, or failed, the transarterial chemoembolization (TACE) treatment. Materials and Methods: From July 2020 to November 2021, a total of 35 HCC patients were enrolled and received HAIC with oxaliplatin plus raltitrexed. The overall survival (OS) and time to progression (TTP) were primary and secondary endpoints, respectively. The tumor response was assessed by the modified response evaluation criteria in solid tumors (mRECIST), and the adverse events were investigated using the common terminology criteria for adverse events version 5.0 (CTCAE 5.0). Results: The median OS and TTP were 10 months (95% confidence interval (CI): 5.5–14.6) and 3.5 months (95% CI: 2.3–4.7), respectively. By means of multivariate analysis, anti-programmed cell death protein 1 (anti-PD-1) immunotherapy was found to be an independent prognostic factor for better survival. No patients experienced toxicity-related death. Thrombocytopenia, alanine aminotransferase (ALT), and aspartate aminotransferase (AST) elevation were the most common toxicities. No grade 3 or higher adverse events related to HAICROX were observed. Conclusion: HAICROX showed valuable efficacy and tolerable toxicity in advanced HCC patients who progressed on TACE or were ineligible for TACE. HAICROX is a promising treatment for advanced-stage HCC patients with TACE failure or ineligibility.

1. Introduction

Hepatocellular carcinoma (HCC) is one of the most common malignancies and the fourth leading cause of cancer-related death worldwide [1]. HCC is also the third most common cause of cancer-related deaths in China [2]. As HCC is often asymptomatic, most patients are already in the intermediate and advanced stages when first diagnosed. The prognosis is dismal, with a median untreated survival time of 7–9 months [3]. Although tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors are regarded as preferred treatments [4], TACE is currently recognized as the most commonly used method for the non-surgical treatment of advanced liver cancer [5,6,7]. However, TACE is not suitable for patients with diffuse HCC, an arterioportal/arteriovenous shunt, or main portal vein tumor thrombosis [5], who are considered as TACE-ineligible. Meanwhile, some patients are classified as TACE-refractory, defined as having refractoriness to TACE after more than two TACE procedures within 6 months [8]. However, there are currently no consistent conclusions on later-line treatments with TACE-refractory or -ineligible patients.
Hepatic arterial infusion chemotherapy (HAIC) is a method of local treatment with a high drug concentration in the liver and few systemic adverse reactions. Several studies have shown that the combination therapy of sorafenib (SORA) plus FOLFOX4-HAIC (the infusion of 5-fluorouracil, leucovorin, and oxaliplatin) showed better survival benefits compared with SORA alone [9,10,11]. In addition, Hsu found that HAIC with FOLFOX4 also showed acceptable outcomes in advanced HCC patients who failed, or were unsuitable for, TACE; the overall survival (OS) and progression-free survival (PFS) were 9.0 months (95% CI: 7.6–10.4) and 3.7 months (95% CI: 3.1–4.3), respectively.
However, the transarterial infusion of 5-fluorouracil needs approximately 44–48 h because 5-fluorouracil is a time-dependent chemotherapy drug. Prolonged arterial infusion is inconvenient and increases the risk of intrahepatic catheter thrombosis and displacement [12]. Conversely, raltitrexed is another thymidylate synthase inhibitor with a longer half-life than 5-Fu, reaching 198 h, and can exert a stable anti-tumor effect over a long period of time [13]. It was regarded as a better candidate for HAIC in HCC. A retrospective study showed that hepatic arterial infusion with a low dose of raltitrexed plus oxaliplatin (HAICROX) was effective in patients with advanced HCC with MPVTT; the median survival time was 8.7 months [14]. A phase II prospective study performed HAICROX in patients with intermediate and advanced-stage HCC; the ORR was 18 (51.4%) out of 35 patients, the DCR was 31 (79.5%) out of 35 patients, the median TTP was 6.7 months (95% CI: 4.6–8.8), and no treatment-related grade 3 or 4 toxicities or deaths were found [15]. However, the efficacy and safety of HAICROX treatment for TACE-refractory or -ineligible advanced HCC patients remain unknown.
Here, we carried out this study to investigate the efficacy and safety of HAICROX for advanced HCC patients with TACE failure or ineligibility.

2. Materials and Methods

2.1. Patients

The inclusion criteria were: (1) pathologically or radiologically (contrast-enhanced magnetic resonance imaging (MRI) or computed tomography (CT)) confirmed advanced HCC based on the American Association for the Study of Liver Disease criteria. (2) TACE refractoriness/failure, defined as disease progression after more than two TACE sessions (The Japan Society of Hepatology (JSH) defines refractoriness to TACE as a failure to control target lesions or the appearance of new lesions, even after two or more consecutive TACE sessions). TACE ineligibility was defined as diffuse HCC, with major portal vein cancer metastasis, a severe arterioportal/arteriovenous shunt on angiography, tumor thrombosis in the inferior vena cava or right atrium, having received TACE treatment at least once. (3) Patients had at least one measurable lesion. (4) Patients had a Child–Pugh score of ≤6 and an Eastern Cooperative Oncology Group (ECOG) performance status of ≤ 1.
The exclusion criteria were: uncontrollable infection; hepatic encephalopathy; gastrointestinal bleeding; refractory ascites; serum bilirubin levels > 3.0 mg/dL; ALT and AST more than 3 times the upper normal limit; albumin levels < 2.5 g/dL; platelet count < 50 × 109/L; and serum creatinine levels > 1.5 mg/dL. This study was approved by the ethical review board of our institution. Informed consent was waived for all patients. Between July 2020 and November 2021, a total of 35 patients with advanced-stage HCC, confirmed as being TACE-refractory or -ineligible and treated with HAICROX, were included. The demographic, clinical, and survival information was extracted from electronic medical records.

2.2. HAIC Procedures

For the HAIC with the oxaliplatin plus raltitrexed procedure, the Seldinger technique was performed through the femoral artery or the radial artery, and a catheter and a coaxial microcatheter were inserted into the feeding hepatic artery. The following regimens of ROX were administered: oxaliplatin (67–75 mg/m2 continuous infusion for 4 h) and raltitrexed (2 mg/m2 continuous infusion for 1 h). After priming chemotherapy, we removed the catheter and microcatheter, performed the treatment every 3–4 weeks, and then discontinued the treatment, as previously defined [15].

2.3. Assessment and Follow-Up

Liver function tests; routine blood tests; and determinations of cytokine, alpha-fetoprotein (AFP), and prothrombin (induced by vitamin K absence-II (PIVKA-II)) levels were performed before every treatment period, and contrast-enhanced CT or MRI of the upper abdomen was performed every 2–3 cycles of HAIC therapy to assess the treatment outcome, according to the modified response evaluation criteria in solid tumors (mRECIST). A further examination was performed if patients had suspected extrahepatic spread. As in a previous study [16], the albumin–bilirubin (ALBI) score was calculated using the following formula: ALBI score = (0.66 × log10 total bilirubin (µmol/L)) + (−0.085 × serum albumin (g/L)). The last follow-up date was 13 March 2022.
The ORR was defined as the number of patients who achieved either a partial response (PR) or a complete response (CR). Additionally, the disease control (DCR) was calculated as the sum of the CR, PR, and stable disease (SD). The primary endpoint of our study was OS, and the secondary endpoint was TTP. In this study, the OS was defined as the period from the initiation of HAIC treatment to death or the last known follow-up, and the TTP was defined as the time from the start of HAIC therapy to disease progression (a radiological or clinical evaluation by mRECIST).
The grade of toxicity was recorded and graded according to the National Cancer Institute’s common terminology criteria for adverse events (CTCAE, version 5.0).

2.4. Statistical Analysis

For baseline characteristics, the categorical variables are described as frequencies and percentages, while continuous variables are presented as the mean ± standard deviation (SD). The Kaplan–Meier method was used to estimate the TTP and OS. Univariate analyses were performed using the log-rank test. For the univariate analysis results, variables with a p < 0.05 were entered into the multivariate analysis. The multivariate Cox model was applied to identify independent risk factors. All statistical tests were 2-sided, and p < 0.05 was considered statistically significant. SPSS software (SPSS version 22.0; SPSS, Chicago, IL, USA) was used to perform the statistical analyses.

3. Results

3.1. Patients Characteristics

In total, between July 2020 and November 2021, 35 HCC patients with either TACE unsuitability or refractoriness received HAICROX treatment and were enrolled in this study. The baseline demographics and characteristics of enrolled patients are shown in Table 1.
All patients received TKI treatments; among them, 30 patients were refractory to TACE and 5 patients were ineligible for TACE because of major vascular tumor thrombosis. The average age of patients was 53 ± 11.7 years, and the median follow-up period was 10 months (range: 2–17.2). Most patients with an advanced stage of HCC were male (33/35, 94.3%) and had hepatitis B (30/35, 85.7%), a small number of patients had hepatitis C (2/35, 5.7%), and just one patient (2.9%) had an HBV and HCV co-infection. Most patients (34/35, 97.1%) belonged to Child–Pugh class A, and 21 (60%) and 14 (40%) patients had liver functions of ALBI grade 1 and grade 2, respectively. Twenty (57.1%) patients had ECOG 1. Twenty-five (86.7%) patients had more than three tumors. In total, 21 (60%) patients had portal vein thrombosis (PVTT), while 7 (20%) patients had tumor thrombosis in the inferior vena cava. Nineteen (54.3%) patients had extrahepatic metastasis, including four (11.4%) patients with pulmonary metastasis, four (11.4%) patients with bone metastasis, seven (20%) patients with lymph node metastasis, three (8.6%) patients with abdominal metastasis, and one (2.9%) patient with gallbladder invasion. The median number of previous TACE sessions was 2.7 ± 2.3. Meanwhile, 13 (37.1%) patients had received more than the second line of TKI treatment and 22 (62.9%) patients had received anti-PD-1 immunotherapy.

3.2. Efficacy and Safety

3.2.1. Tumor Response

The mean number of HAIC treatments was 2.3 (range: 1–5). At 2 months after HAIC treatment, 4 (11.4%) patients had a PR, 16 (45.7%) patients had an SD and 15 (42.9%) patients had a PD. Accordingly, the ORR was 11.4% and the DCR was 57.1%. Table 2 lists the treatment outcome details.

3.2.2. Survival Outcome

In total, 21 out of 35 (60%) patients had died at the end of follow-up (March 2022). The mean follow-up time was 10 months (range: 2–17.2). The median OS and TTP were 10 months (95% CI: 5.5–14.6) and 3.5 months (95% CI: 2.3–4.7), respectively (Figure 1). The median OS for patients who achieved a response (PR) to HAIC treatment was not reached, and the median OS for patients with SD and PD (non-respondents) was 8.6 months (95% CI: 5.5–11.8). Patients who achieved clinical benefits (CR + PR + SD) showed a longer OS and TTP compared with people without clinical benefits (median OS, not reached vs. 6.8 months, p = 0.014; median TTP, 6.5 months vs. 2.6 months, p = 0.023) (Figure 2). These results show that achieving a clinical benefit could predict the patients’ survival. In addition, patients with anti-PD-1 combination therapy showed better OS and TTP than without anti-PD-1 immunotherapy (median OS, 15.8 months (95% CI 7.4–24.2) vs. 6.7 months (95% CI 3.7–9.8), p = 0.01; median TTP, 6.5 months (95% CI 1.6–11.4) vs. 2.1 months (95% CI 1.1–3.0), p = 0.043) (Figure 3).

3.2.3. Safety and Toxicity

No treatment-related deaths occurred, and no patients experienced grade 3 or 4 toxicities. The most common toxicities associated with HAIC included AST and ALT elevation (both 34.3%), thrombocytopenia (17.1%), bilirubin elevation (8.6%), general weakness (8.6%), dyspepsia/anorexia (2.9%), nausea/vomiting (2.9%), gastrointestinal (GI) bleeding (2.9%), hyponatremia (2.9%), ascites aggravation (2.9%), and hepatic encephalopathy (2.9%). HAIC-related toxicities are detailed in Table 3.

3.3. Prognostic Factors

In the univariate analysis, tumor size, total bilirubin (TBIL), albumin (ALB), ALBI-grade, AST, GGT, IL-8, PIVKA-II, and treatment with anti-PD-1 immunotherapy were significantly associated with OS. Multivariate Cox analysis demonstrated that treatment with anti-PD-1 immunotherapy was an independent prognostic factor for OS (details are presented in Table 4). In the univariate analysis, AST and receiving later-line TACE or anti-PD-1 treatment were significantly associated with TTP. However, according to multivariate Cox analysis, there were no independent prognostic factors for TTP (Supplementary Table S1).

4. Discussion

TACE still represents a mainstay of treatment and is often the first-line therapy in patients with advanced HCC, but TACE failure or ineligibility remains a challenge for clinicians. This study provides a later-line option for advanced HCC patients with TACE refractoriness/failure or ineligibility. In this study, all patients received TKI treatment and belonged to the TACE-failure or -ineligible advanced HCC group. The median OS and TTP were 10 months (95% CI: 5.5–14.6) and 3.5 months (95% CI: 2.3–4.7), respectively, similar to previous studies [9].
Although TKI- and anti-PD-1-based treatments are regarded as basal therapies for advanced HCC according to many international HCC guidelines, TACE is the most frequently utilized treatment [7,17]. SHARP and Oriental studies showed that sorafenib could delay advanced HCC tumor progress (median TTP, 2.8–5.5 months) and prolong patient survival (median OS, 6.5–10.7 months) [18,19]. Meanwhile, lenvatinib was not inferior to sorafenib in advanced HCC (median OS, 13.6 months vs. 12.3 months) [20]. Additionally, some studies showed that the survival time of TACE treatment was comparable to sorafenib alone [21,22]. Moreover, some reviews have demonstrated that TACE plus sorafenib/lenvatinib was more effective than sorafenib/lenvatinib or TACE alone in advanced HCC patients [23,24,25,26]. In addition, patients with diffuse HCC, portal vein tumor thrombosis, or an arterioportal/arteriovenous shunt are not suitable for TACE. Furthermore, some HCC patients became TACE-refractory after repeatedly undergoing TACE treatment. More importantly, multiple TACE treatments may cause liver function abnormalities. For these patients, switching to other TKIs or PD-1 inhibitor treatments may be a good choice, such as using sorafenib [27], lenvatinib [28], and apatinib [29,30]. However, some patients may not withstand TKI treatment, and the treatment efficacy was poor (only prolonging OS by 3–6 months). Other TKIs, for example, regorafenib [31], apatinib [32], and cabozantinib [33], may also be selected for advanced HCC. However, the treatment effects were also limited, and the ORR was 2–18.8% [34]. Although several PD-1/PD-L1 inhibitors served as second-line treatments for unresectable HCC patients with a median OS of 13.9–15.6 months [35,36,37], they are not suitable for patients with a high tumor burden and a short life expectancy of less than 3 months.
HAIC has been used classically for unresectable colorectal liver metastases [38], and is also widely performed in HCC patients. Some studies showed that HAIC with FOLFOX4 is superior to sorafenib for advanced HCC patients [39,40]. JSH consensus statements proposed that HAIC is another choice for patients with a liver function of Child–Pugh class B or worse at the time of TACE failure/refractoriness [41]. In the present study, instead of 5-FU, raltitrexed was used in the treatment of HAIC, which shortened the infusion time, improved patient comfort, and reduced hospitalization hours. A phase II prospective study showed that raltitrexed-plus-oxaliplatin-based HAIC led to a higher ORR (51.4%, 18 of 35 patients) and was considered safe and tolerable in patients with unresectable HCC [15]. Another retrospective study highlighted the efficacy of low-dose continuous HAICROX for advanced HCC patients with MPVTT, where the median survival time was 8.7 months [14]. However, the survival benefits of HAICROX treatments as a later-line therapy in TACE-TKI-failed or -refractory HCC remain unknown. In this study, we treated TACE-failed or -ineligible advanced HCC patients with HAIC with raltitrexed plus oxaliplatin. Taking the patient’s liver function and physical strength into consideration, the dosage of raltitrexed and oxaliplatin was reduced by one-third compared to the dosage in previous HAICROX studies [15]. The ORR and DCR were significantly lower in our study compared to previous results (ORR, 51.4%, 18 of 35 patients; DCR, 88.6%, 31 of 35 patients) [15], but our patients belonged to a less favorable subset of clinical baseline characteristics, such as a greater tumor burden (71.4% of patients with more than three tumors) and they were at a later BCLC stage (all of them were BCLC stage cases, 60% of patients had vascular invasion, and 54.3% of patients with extrahepatic metastasis). In fact, all of our patients were classified as TACE-failed (85.7% of patients) or TACE-unsuitable (14.3% of patients) and had received first or second lines of TKI therapy. However, our survival outcomes, including OS and TTP, were 10.0 months (95% CI: 5.5–14.6) and 3.5 months, respectively (95% CI: 2.3–4.7), comparable to those receiving HAIC with modified FOLFOX, as reported by Hsu SJ [9]. Of note, the patients in our study who received clinical benefits from HAICROX had improved OS and TTP compared to non-responders (median OS, not reached vs. 6.8 months, p = 0.014; median TTP, 6.5 months vs. 2.6 months, p = 0.023).
The results from the Check Mate 459 [42] and KEYNOTE-240 [36] trials show that anti-PD-1 immunotherapy with nivolumab or pembrolizumab as a single agent did not meet the setting primary endpoints (OS). However, in this study, anti-PD-1 treatment combination therapy was identified as an independent prognostic factor, and the patients who accepted anti-PD-1 immunotherapy and HAICROX plus TKI treatments achieved better survival outcomes; the median OS was 15.8 vs. 6.7 months (p = 0.01; HR = 0.329; 95% CI: 0.135–0.802) and the median TTP was 6.5 vs. 2.1 months (p = 0.043; HR 0.324; 95% CI: 0.113–0.926), which implied that the combination therapy may be a valuable choice and should be verified in our future studies. Consistent with another study, HAIC combined with anti-PD-1 immunotherapy (HAICAP) was superior to HAIC treatment alone for advanced hepatocellular carcinoma [43]. The potential mechanism may be that HAIC can improve anti-tumor immunity by releasing the neoplasm antigens from killed liver tumor cells [44] and increase PD-1/PD-L1 expression in the tumor microenvironment (TME). The combination of anti-PD1 therapy and locoregional therapy may also reduce the proportion of Tregs and the aggregation of myeloid-derived suppressor cells (MDSCs) [45].
Compared to previous research [14,15], HAICROX in TACE-refractory or -failed advanced HCC showed good tolerance and safety in our study. The most frequent adverse events were thrombocytopenia (17.1%), alanine aminotransferase (ALT), and aspartate aminotransferase (AST) elevation (34.29%, both), which can be managed and alleviated through dose reduction. Overall, these results show that HAICROX is a promising treatment for advanced HCC patients who failed or are unsuited to TACE combined with TKI treatments, and the subsequent HAICROX therapy prolonged the OS and TTP of advanced-stage HCC patients to some extent.
However, this study has several limitations. Firstly, this is a retrospective study, and all clinical data were obtained from a single medical center. Secondly, the categories of PD-1 inhibitors varied, which might influence the uniformity of the treatment procedure. Thirdly, our study was a single-arm study, and the sample size was small. Thus, more randomized controlled trials are required to verify the efficacy of HAICROX in this clinical setting.
In conclusion, this result highlighted that HAICROX largely prolonged the OS and TTP of advanced HCC patients who progressed on TACE-based treatment or were unsuitable/ineligible for TACE. The results provide a promising option for the later-line treatment for the majority of advanced HCC patients with TACE failure or ineligibility.

5. Conclusions

HAICROX showed valuable efficacy and tolerable toxicity, and it is an alternative option for advanced-stage HCC patients with TACE failure or ineligibility.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/medicina58101343/s1, Table S1: Univariate and Multivariate Analyses of Prognostic Factors for TTP of 35 patients.

Author Contributions

Conceptualization, Y.W. and B.Z.; methodology, Z.Z., S.Z. and B.Z.; software, Z.Z., Y.W. and M.W.; validation, T.Z. and H.C.; formal analysis, Y.W. and S.Z.; investigation, Y.W., Z.Z. and S.Z.; resources, Y.W., S.Z., Z.Z., X.C., G.C., T.Z., H.C. and X.G.; data curation, S.Z.; writing—original draft preparation, Y.W.; writing—review and editing, S.Z. and B.Z.; visualization, Y.W.; supervision, B.Z.; project administration, Y.W., X.X. and B.Z.; funding acquisition, Y.W., Z.Z. and B.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded in part by the Xiamen Medical and Health Key Project (3502Z20191105 to Boheng Zhang), the Foundation of the Bethune Charitable Foundation (BCF-MSD-HCC-20201127-001 to Zhenzhen Zhang), and the Foundation of the Xiamen Branch, Zhongshan Hospital, Fudan University of China (2020ZSXMYS02 to Yanfang Wu).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Ethics Committee of Zhongshan Hospital, Fudan University (Xiamen Branch) (protocol # B2019-026, 23 March 2020).

Informed Consent Statement

Written informed consent was obtained from the patients to publish this paper.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
  2. Chen, W.; Zheng, R.; Baade, P.D.; Zhang, S.; Zeng, H.; Bray, F.; Jemal, A.; Yu, X.Q.; He, J. Cancer statistics in China, 2015. CA Cancer J Clin. 2016, 66, 115–132. [Google Scholar] [CrossRef] [PubMed]
  3. Giannini, E.G.; Farinati, F.; Ciccarese, F.; Pecorelli, A.; Rapaccini, G.L.; Di Marco, M.; Benvegnù, L.; Caturelli, E.; Zoli, M.; Borzio, F.; et al. Prognosis of untreated hepatocellular carcinoma. Hepatology 2015, 61, 184–190. [Google Scholar] [CrossRef] [PubMed]
  4. European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. J. Hepatol. 2018, 69, 182–236. [Google Scholar] [CrossRef] [PubMed]
  5. Park, J.W.; Chen, M.; Colombo, M.; Roberts, L.R.; Schwartz, M.; Chen, P.J.; Kudo, M.; Johnson, P.; Wagner, S.; Orsini, L.S.; et al. Global patterns of hepatocellular carcinoma management from diagnosis to death: The BRIDGE Study. Liver Int. 2015, 35, 2155–2166. [Google Scholar] [CrossRef]
  6. Llovet, J.M.; Real, M.I.; Montaña, X.; Planas, R.; Coll, S.; Aponte, J.; Ayuso, C.; Sala, M.; Muchart, J.; Solà, R.; et al. Barcelona Liver Cancer Group. Arterial embolisation or chemoembolisation versus symptomatic treatment in patients with unresectable hepatocellular carcinoma: A randomised controlled trial. Lancet 2002, 359, 1734–1739. [Google Scholar] [CrossRef]
  7. Lencioni, R.; de Baere, T.; Soulen, M.C.; Rilling, W.S.; Geschwind, J.F. Lipiodol transarterial chemoembolization for hepatocellular carcinoma: A systematic review of efficacy and safety data. Hepatology 2016, 64, 106–116. [Google Scholar] [CrossRef] [PubMed]
  8. Jeon, M.Y.; Kim, H.S.; Lim, T.S.; Han, D.H.; Kim, B.K.; Park, J.Y.; Kim, D.Y.; Ahn, S.H.; Choi, G.H.; Choi, J.S.; et al. Refractoriness to transarterial chemoembolization in patients with recurrent hepatocellular carcinoma after curative resection. PLoS ONE 2019, 14, e0214613. [Google Scholar] [CrossRef] [PubMed]
  9. Hsu, S.J.; Xu, X.; Chen, M.P.; Zhao, Z.Y.; Wang, Y.; Yin, X.; Zhang, L.; Ge, N.L.; Chen, Y.; Wang, Y.H.; et al. Hepatic Arterial Infusion Chemotherapy with Modified FOLFOX as an Alternative Treatment Option in Advanced Hepatocellular Carcinoma Patients with Failed or Unsuitability for Transarterial Chemoembolization. Acad. Radiol. 2021, 28 (Suppl. S1), S157–S166. [Google Scholar] [CrossRef] [PubMed]
  10. Lyu, N.; Kong, Y.; Mu, L.; Lin, Y.; Li, J.; Liu, Y.; Zhang, Z.; Zheng, L.; Deng, H.; Li, S.; et al. Hepatic arterial infusion of oxaliplatin plus fluorouracil/leucovorin vs. sorafenib for advanced hepatocellular carcinoma. J. Hepatol. 2018, 69, 60–69. [Google Scholar] [CrossRef] [PubMed]
  11. Lyu, N.; Lin, Y.; Kong, Y.; Zhang, Z.; Liu, L.; Zheng, L.; Mu, L.; Wang, J.; Li, X.; Pan, T.; et al. FOXAI: A phase II trial evaluating the efficacy and safety of hepatic arterial infusion of oxaliplatin plus fluorouracil/leucovorin for advanced hepatocellular carcinoma. Gut 2018, 67, 395–396. [Google Scholar] [CrossRef] [PubMed]
  12. Kudo, M.; Ueshima, K.; Yokosuka, O.; Ogasawara, S.; Obi, S.; Izumi, N.; Aikata, H.; Nagano, H.; Hatano, E.; Sasaki, Y.; et al. Sorafenib plus low-dose cisplatin and fluorouracil hepatic arterial infusion chemotherapy versus sorafenib alone in patients with advanced hepatocellular carcinoma (SILIUS): A randomised, open label, phase 3 trial. Lancet Gastroenterol. Hepatol. 2018, 3, 424–432. [Google Scholar] [CrossRef]
  13. Hagner, N.; Joerger, M. Cancer chemotherapy: Targeting folic acid synthesis. Cancer Manag. Res. 2010, 2, 293–301. [Google Scholar] [CrossRef]
  14. Zhu, L.Z.; Xu, S.; Qian, H.L. Transarterial embolization and low-dose continuous hepatic arterial infusion chemotherapy with oxaliplatin and raltitrexed for hepatocellular carcinoma with major portal vein tumor thrombus. World J. Gastroenterol. 2018, 24, 2501–2507. [Google Scholar] [CrossRef] [PubMed]
  15. Chen, S.; Zhang, K.; Liu, W.; Yu, W. Hepatic arterial infusion of oxaliplatin plus raltitrexed in patients with intermediate and advanced stage hepatocellular carcinoma: A phase II, single-arm, prospective study. Eur. J. Cancer 2020, 134, 90–98. [Google Scholar] [CrossRef] [PubMed]
  16. Johnson, P.J.; Berhane, S.; Kagebayashi, C.; Satomura, S.; Teng, M.; Reeves, H.L.; O’Beirne, J.; Fox, R.; Skowronska, A.; Palmer, D.; et al. Assessment of liver function in patients with hepatocellular carcinoma: A new evidence-based approach-the ALBI grade. J. Clin. Oncol. 2015, 33, 550–558. [Google Scholar] [CrossRef] [PubMed]
  17. Llovet, J.M.; Bruix, J. Systematic review of randomized trials for unresectable hepatocellular carcinoma: Chemoembolization improves survival. Hepatology 2003, 37, 429–442. [Google Scholar] [CrossRef]
  18. Llovet, J.M.; Ricci, S.; Mazzaferro, V.; Hilgard, P.; Gane, E.; Blanc, J.F.; de Oliveira, A.C.; Santoro, A.; Raoul, J.L.; Forner, A.; et al. Sorafenib in advanced hepatocellular carcinoma. N. Engl. J. Med. 2008, 359, 378–390. [Google Scholar] [CrossRef]
  19. Cheng, A.L.; Kang, Y.K.; Chen, Z.; Tsao, C.J.; Qin, S.; Kim, J.S.; Luo, R.; Feng, J.; Ye, S.; Yang, T.S.; et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: A phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009, 10, 25–34. [Google Scholar] [CrossRef]
  20. Kudo, M.; Finn, R.S.; Qin, S.; Han, K.H.; Ikeda, K.; Piscaglia, F.; Baron, A.; Park, J.W.; Han, G.; Jassem, J.; et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: A randomised phase 3 non-inferiority trial. Lancet 2018, 391, 1163–1173. [Google Scholar] [CrossRef] [Green Version]
  21. Nishikawa, H.; Osaki, Y.; Iguchi, E.; Takeda, H.; Nakajima, J.; Matsuda, F.; Sakamoto, A.; Henmi, S.; Hatamaru, K.; Saito, S.; et al. Comparison of the efficacy of transcatheter arterial chemoembolization and sorafenib for advanced hepatocellular carcinoma. Exp. Ther. Med. 2012, 4, 381–386. [Google Scholar] [CrossRef] [PubMed]
  22. Pinter, M.; Hucke, F.; Graziadei, I.; Vogel, W.; Maieron, A.; Königsberg, R.; Stauber, R.; Grünberger, B.; Müller, C.; Kölblinger, C.; et al. Advanced-stage hepatocellular carcinoma: Transarterial chemoembolization versus sorafenib. Radiology 2012, 263, 590–599. [Google Scholar] [CrossRef] [PubMed]
  23. Wu, F.X.; Chen, J.; Bai, T.; Zhu, S.L.; Yang, T.B.; Qi, L.N.; Zou, L.; Li, Z.H.; Ye, J.Z.; Li, L.Q. The safety and efficacy of transarterial chemoembolization combined with sorafenib and sorafenib mono-therapy in patients with BCLC stage B/C hepatocellular carcinoma. BMC Cancer 2017, 17, 645. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, X.; Wang, K.; Wang, M.; Yang, G.; Ye, X.; Wu, M.; Cheng, S. Transarterial chemoembolization (TACE) combined with sorafenib versus TACE for hepatocellular carcinoma with portal vein tumor thrombus: A systematic review and meta-analysis. Oncotarget 2017, 8, 29416–29427. [Google Scholar] [CrossRef]
  25. Zhang, X.P.; Wang, K.; Li, N.; Zhong, C.Q.; Wei, X.B.; Cheng, Y.Q.; Gao, Y.Z.; Wang, H.; Cheng, S.Q. Survival benefit of hepatic resection versus transarterial chemoembolization for hepatocellular carcinoma with portal vein tumor thrombus: A systematic review and meta-analysis. BMC Cancer 2017, 17, 902. [Google Scholar] [CrossRef]
  26. Wu, J.Y.; Yin, Z.Y.; Bai, Y.N.; Chen, Y.F.; Zhou, S.Q.; Wang, S.J.; Zhou, J.Y.; Li, Y.N.; Qiu, F.N.; Li, B.; et al. Lenvatinib Combined with Anti-PD-1 Antibodies Plus Transcatheter Arterial Chemoembolization for Unresectable Hepatocellular Carcinoma: A Multicenter Retrospective Study. J Hepatocell Carcinoma 2021, 8, 1233–1240. [Google Scholar] [CrossRef]
  27. Kodama, K.; Kawaoka, T.; Aikata, H.; Uchikawa, S.; Inagaki, Y.; Hatooka, M.; Morio, K.; Nakahara, T.; Murakami, E.; Tsuge, M.; et al. Comparison of clinical outcome of hepatic arterial infusion chemotherapy and sorafenib for advanced hepatocellular carcinoma according to macrovascular invasion and transcatheter arterial chemoembolization refractory status. J. Gastroenterol. Hepatol. 2018, 33, 1780–1786. [Google Scholar] [CrossRef]
  28. Shimose, S.; Kawaguchi, T.; Tanaka, M.; Iwamoto, H.; Miyazaki, K.; Moriyama, E.; Suzuki, H.; Niizeki, T.; Shirono, T.; Nakano, M.; et al. Lenvatinib prolongs the progression-free survival time of patients with intermediate-stage hepatocellular carcinoma refractory to transarterial chemoembolization: A multicenter cohort study using data mining analysis. Oncol Lett. 2020, 20, 2257–2265. [Google Scholar] [CrossRef]
  29. Cao, Y.; Ouyang, T.; Xiong, F.; Kan, X.; Chen, L.; Liang, B.; Zheng, C. Efficacy of apatinib in patients with sorafenib-transarterial chemoembolization refractory hepatocellular carcinoma: A retrospective study. Hepatol. Int. 2021, 15, 1268–1277. [Google Scholar] [CrossRef]
  30. Qiu, Z.; Shen, L.; Chen, S.; Qi, H.; Cao, F.; Xie, L.; Fan, W. Efficacy Of Apatinib In Transcatheter Arterial Chemoembolization (TACE) Refractory Intermediate And Advanced-Stage Hepatocellular carcinoma: A Propensity Score Matching Analysis. Cancer Manag. Res. 2019, 11, 9321–9330. [Google Scholar] [CrossRef] [Green Version]
  31. Bruix, J.; Qin, S.; Merle, P.; Granito, A.; Huang, Y.H.; Bodoky, G.; Pracht, M.; Yokosuka, O.; Rosmorduc, O.; Breder, V.; et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 389, 56–66. [Google Scholar] [CrossRef]
  32. Qin, S.; Li, Q.; Gu, S.; Chen, X.; Lin, L.; Wang, Z.; Xu, A.; Chen, X.; Zhou, C.; Ren, Z.; et al. Apatinib as second-line or later therapy in patients with advanced hepatocellular carcinoma (AHELP): A multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Gastroenterol. Hepatol. 2021, 6, 559–568. [Google Scholar] [CrossRef]
  33. Abou-Alfa, G.K.; Meyer, T.; Cheng, A.L.; El-Khoueiry, A.B.; Rimassa, L.; Ryoo, B.Y.; Cicin, I.; Merle, P.; Chen, Y.; Park, J.W.; et al. Cabozantinib in Patients with Advanced and Progressing Hepatocellular Carcinoma. N. Engl. J. Med. 2018, 379, 54–63. [Google Scholar] [CrossRef] [PubMed]
  34. Da Fonseca, L.G.; Reig, M.; Bruix, J. Tyrosine Kinase Inhibitors and Hepatocellular Carcinoma. Clin. Liver Dis. 2020, 24, 719–737. [Google Scholar] [CrossRef] [PubMed]
  35. Yau, T.; Hsu, C.; Kim, T.Y.; Choo, S.P.; Kang, Y.K.; Hou, M.M.; Numata, K.; Yeo, W.; Chopra, A.; Ikeda, M.; et al. Nivolumab in advanced hepatocellular carcinoma: Sorafenib-experienced Asian cohort analysis. J. Hepatol. 2019, 71, 543–552. [Google Scholar] [CrossRef] [PubMed]
  36. Finn, R.S.; Ryoo, B.Y.; Merle, P.; Kudo, M.; Bouattour, M.; Lim, H.Y.; Breder, V.; Edeline, J.; Chao, Y.; Ogasawara, S.; et al. Pembrolizumab As Second-Line Therapy in Patients With Advanced Hepatocellular Carcinoma in KEYNOTE-240: A Randomized, Double-Blind, Phase III Trial. J. Clin. Oncol. 2020, 38, 193–202. [Google Scholar] [CrossRef]
  37. Qin, S.; Ren, Z.; Meng, Z.; Chen, Z.; Chai, X.; Xiong, J.; Bai, Y.; Yang, L.; Zhu, H.; Fang, W.; et al. Camrelizumab in patients with previously treated advanced hepatocellular carcinoma: A multicentre, open-label, parallel-group, randomised, phase 2 trial. Lancet Oncol. 2020, 21, 571–580. [Google Scholar] [CrossRef]
  38. Kemeny, N.E.; Niedzwiecki, D.; Hollis, D.R.; Lenz, H.J.; Warren, R.S.; Naughton, M.J.; Weeks, J.C.; Sigurdson, E.R.; Herndon, J. E 2nd.; Zhang, C.; et al. Hepatic arterial infusion versus systemic therapy for hepatic metastases from colorectal cancer: A randomized trial of efficacy, quality of life, and molecular markers (CALGB 9481). J. Clin. Oncol. 2006, 24, 1395–1403. [Google Scholar] [CrossRef]
  39. Long, G.B.; Xiao, C.W.; Zhao, X.Y.; Zhang, J.; Li, X. Effects of hepatic arterial infusion chemotherapy in the treatment of hepatocellular carcinoma: A meta-analysis. Medicine 2020, 99, e20745. [Google Scholar] [CrossRef]
  40. Liu, M.; Shi, J.; Mou, T.; Wang, Y.; Wu, Z.; Shen, A. Systematic review of hepatic arterial infusion chemotherapy versus sorafenib in patients with hepatocellular carcinoma with portal vein tumor thrombosis. J. Gastroenterol. Hepatol. 2020, 35, 1277–1287. [Google Scholar] [CrossRef]
  41. Kudo, M.; Kawamura, Y.; Hasegawa, K.; Tateishi, R.; Kariyama, K.; Shiina, S.; Toyoda, H.; Imai, Y.; Hiraoka, A.; Ikeda, M.; et al. Management of Hepatocellular Carcinoma in Japan: JSH Consensus Statements and Recommendations 2021 Update. Liver Cancer 2021, 10, 181–223. [Google Scholar] [CrossRef] [PubMed]
  42. Yau, T.; Park, J.W.; Finn, R.S.; Cheng, A.L.; Mathurin, P.; Edeline, J.; Kudo, M.; Harding, J.J.; Merle, P.; Rosmorduc, O.; et al. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): A randomised, multicentre, open-label, phase 3 trial. Lancet Oncol. 2022, 23, 77–90. [Google Scholar] [CrossRef]
  43. Mei, J.; Li, S.H.; Li, Q.J.; Sun, X.Q.; Lu, L.H.; Lin, W.P.; Zheng, L.; Chen, M.S.; Shi, M.; Wei, W.; et al. Anti-PD-1 Immunotherapy Improves the Efficacy of Hepatic Artery Infusion Chemotherapy in Advanced Hepatocellular Carcinoma. J. Hepatocell. Carcinoma 2021, 8, 167–176. [Google Scholar] [CrossRef] [PubMed]
  44. Galluzzi, L.; Buqué, A.; Kepp, O.; Zitvogel, L.; Kroemer, G. Immunogenic cell death in cancer and infectious disease. Nat. Rev. Immunol. 2017, 17, 97–111. [Google Scholar] [CrossRef]
  45. Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014, 211, 781–790. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Kaplan–Meier curves for overall survival (OS) (a) and time to progression (TTP) (b) in 35 patients undergoing HAIC with oxaliplatin plus raltitrexed (HAICROX).
Figure 1. Kaplan–Meier curves for overall survival (OS) (a) and time to progression (TTP) (b) in 35 patients undergoing HAIC with oxaliplatin plus raltitrexed (HAICROX).
Medicina 58 01343 g001
Figure 2. Kaplan–Meier curves for overall survival (OS) (a) and TTP (b) for patients with clinical benefits (CR + PR + SD) and without clinical benefits (PD) in HCC patients treated with HAICROX, who experienced TACE treatment failure or unsuitability.
Figure 2. Kaplan–Meier curves for overall survival (OS) (a) and TTP (b) for patients with clinical benefits (CR + PR + SD) and without clinical benefits (PD) in HCC patients treated with HAICROX, who experienced TACE treatment failure or unsuitability.
Medicina 58 01343 g002
Figure 3. Kaplan–Meier curves for OS (a) and TTP (b) in patients, stratified by treatment with or without anti-PD-1 immunotherapy.
Figure 3. Kaplan–Meier curves for OS (a) and TTP (b) in patients, stratified by treatment with or without anti-PD-1 immunotherapy.
Medicina 58 01343 g003
Table 1. Clinical characteristics of 35 advanced HCC patients with TACE refractoriness or ineligibility.
Table 1. Clinical characteristics of 35 advanced HCC patients with TACE refractoriness or ineligibility.
Characteristicn= 35
Age 153.0 ± 11.7
Gender
Male33 (94.3%)
Female2 (5.7%)
HBV
Negative5 (14.3%)
Positive30 (85.7%)
HCV
Negative33 (94.3%)
Positive2 (5.7 %)
HBV + HCV coinfection1 (2.9%)
Child–Pugh
A34 (97.1%)
B1 (2.9%)
ECOG
015 (42.9%)
120 (57.1%)
Tumor size (mm) 183.8 ± 44.8
Tumor number
≤310 (28.6%)
>325 (71.4%)
Tumor thrombosis
None14 (40%)
Inferior vena cava (IVC)7 (20%)
Main portal vein3 (8.6%)
Branch of the portal vein9 (25.7%)
Distant branch of portal vein2 (5.7%)
Extrahepatic metastasis
Yes19 (54.3%)
No16 (45.7%)
Number of HAIC treatments 2.3 ± 1.1
Number of previous TACE treatments 2.7 ± 2.3
Previous TKI lines
1st22 (62.9%)
≥2nd13 (37.1%)
Later-line treatment
Anti-PD-122 (62.9%)
TACE10 (28.6%)
TBIL (µmol/L) 212.2 (8.5–19.2)
ALT (U/L) 236 (22–48)
AST (U/L) 244 (35–68)
Albumin (g/L) 140.3 ± 5.2
ALP (U/L) 2127 (92–198.5)
GGT (U/L) 2160 (78–207)
WBC (×109/L) 25.76 (4.01–6.74)
Neu (×109/L) 23.5 (2.1–4.6)
PLT (×109/L) 2161 (101–192)
L (×109/L) 21.1 (0.8–1.6)
NLR 22.8 (1.8–5.1)
PLR 2128.1 (91.8–151.5)
IL-6 (pg/mL) 210.4 (5.9–18.6)
IL-8 (pg/mL) 236.95 (17.75–63.73)
TNF (pg/mL) 28 (6.7–14.7)
IL-2R(U/mL) 2557.5 (340–927)
ALBI (grade1/grade2/grade3)21 (60%)/14 (40%)/0
AFP (ng/mL) 2457.2 (75.3–4917)
PIVKA-II (mAU/mL) 24139 (703–28,798)
TBIL, total bilirubin; ALT, alanine aminotransferase; AST, aspartate aminotransferase; ALP, alkaline phosphatase; GGT, γ-glutamyl transpeptidase; WBC, white blood cell count; Neu, neutrophil cell count; PLT, platelet count; L, lymphocyte cell count; NLR, neutrophil–lymphocyte ratio; PLR, platelet–lymphocyte ratio; IL-6, interleukin-6; IL-8, interleukin-8; TNF, tumor necrosis factor; IL-2R, interleukin-2R; ALBI, albumin–bilirubin grade; AFP, alpha-fetoprotein; PIVKA-II, prothrombin induced by vitamin K absence II. 1 Data are presented as the mean ± SD. 2 Data are presented as the median (interquartile range).
Table 2. Tumor response assessed according to mRECIST after two months of HAIC treatment.
Table 2. Tumor response assessed according to mRECIST after two months of HAIC treatment.
Tumor Responsesn = 35 (%)
CR0
PR4 (11.4%)
SD16 (45.7%)
PD15 (42.9%)
ORR4 (11.4%)
DCR20 (57.1%)
CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease; ORR, objective response rate; DCR, disease control rate.
Table 3. HAICROX associated with adverse events in 35 HCC patients.
Table 3. HAICROX associated with adverse events in 35 HCC patients.
Adverse EventGrades I
n (%)
Grades II
n (%)
Grades III
n (%)
Grades IV
n (%)
Thrombocytopenia4 (11.4%)2 (5.7%)00
Dyspepsia/anorexia1 (2.9%)000
Nausea/vomiting1 (2.9%)000
GI bleeding01 (2.9%)00
Fatigue3 (2.9%)000
General weakness3 (8.6%)000
AST elevation11 (31.4%)1 (2.9%)00
ALT elevation11 (31.4%)1 (2.9%)00
Bilirubin elevation3 (8.6%)000
Hyponatremia1 (2.9%)000
Ascites aggravation1 (2.9%)000
Hepatic encephalopathy1 (2.9%)000
GI bleeding, gastrointestinal bleeding; AST, aspartate aminotransferase; ALT, alanine aminotransferase.
Table 4. Prognostic factors associated with overall survival in 35 patients (* p < 0.05 is considered statistically significant).
Table 4. Prognostic factors associated with overall survival in 35 patients (* p < 0.05 is considered statistically significant).
Univariate Analysis Multivariate Analysis
HR95% CIp ValueHR95% CIp Value
Gender (M/F)0.5450.21–1.4140.212
Age (<50 vs. ≥50)0.610.257–1.4520.081
Etiology (HBC vs. HCV vs. unknown)0.1280.013–1.2860.173
TB (µmol/L)1.0661.014–1.1210.007 *1.0760.978–1.1850.134
Albumin (g/L)0.8790.800–0.9660.013 *1.0130.822–1.2480.905
ALBI-grade2.711.105–6.6470.029 *2.9820.259–33.0460.385
ALT (U/L)1.0240.999–1.0490.059
AST (U/L)1.0181.009–1.026<0.001 *1.0190.998–1.0400.082
ALP (U/L)1.0081.003–1.0140.093
GGT (U/L)1.0041.001–1.0080.027 *0.9970.988–1.0070.554
WBC (×109/L)0.9960.795–1.2480.971
PLT (×109/L)0.9970.989–1.0050.437
Neu (×109/L)1.0940.791–1.5120.558
L (×109/L)0.5410.391–1.6370.113
NLR1.0940.853–1.4030.48
PLR0.9990.993–1.0060.867
IL-61.010.985–1.0350.444
IL-81.0081.002–1.0140.007 *0.9980.986–1.0090.689
IL-2R1.0011–1.0020.142
TNF1.0040.996–1.0130.284
AFP (<400 mg/L, ≥400 mg/L)2.2420.924–5.4390.741
PIVKA-II (<400 mAU/mL, ≥400 mAU/mL)2.2860.668–7.8280.001 *0.5540.095–3.2360.512
Tumor size (mm)1.011–1.020.04 *1.010.992–1.0280.28
Tumor number (1–3/>3)1.7390.634–4.7660.282
Tumor thrombosis (yes, no)1.150.469–2.8190.759
Extrahepatic metastasis1.20.508–2.8370.677
TACE times (1–2/>2)1.1150.461–2.6960.809
TKIs lines (1st/>2nd)1.3950.590–3.30.449
ECOG (0/1)0.6320.268–1.4910.295
Child–Pugh class (A/B)3.3180.404–27.2790.264
Times of HAIC (1, 2/>2)0.7220.449–1.1610.179
Later-line treatment
TACE (0, ≥1)1.3950.590–3.3000.449
Anti-PD-1 (yes, no)0.3290.135–0.8020.014 *0.2670.075–0.9530.042 *
TBIL, total bilirubin; ALT, alanine aminotransferase; AST, aspartate aminotransferase; ALP, alkaline phosphatase; GGT, γ-glutamyl transpeptidase; WBC, white blood cell count; Neu, neutrophil cell count; PLT, platelet count; L, lymphocyte cell count; NLR, neutrophil–lymphocyte ratio; PLR, platelet–lymphocyte ratio; IL-6, interleukin-6; IL-8, interleukin-8; TNF, tumor necrosis factor; IL-2R, interleukin-2R; ALBI, albumin–bilirubin grade; AFP, alpha-fetoprotein; PIVKA-II, prothrombin induced by vitamin K absence-II; TACE, transarterial chemoembolization; TKIs, tyrosine kinase inhibitors; ECOG, Eastern Cooperative Oncology Group; HAIC, hepatic arterial infusion chemotherapy; anti-PD-1, anti-programmed cell death protein 1 immunotherapy.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wu, Y.; Zheng, S.; Zhang, Z.; Chen, G.; Chen, X.; Zheng, T.; Guo, X.; Chen, H.; Wang, M.; Xie, X.; et al. Hepatic Arterial Infusion Chemotherapy with Oxaliplatin Plus Raltitrexed as an Alternative Option in Advanced Hepatocellular Carcinoma Patients with Failure of, or Unsuitability for, Transarterial Chemoembolization. Medicina 2022, 58, 1343. https://doi.org/10.3390/medicina58101343

AMA Style

Wu Y, Zheng S, Zhang Z, Chen G, Chen X, Zheng T, Guo X, Chen H, Wang M, Xie X, et al. Hepatic Arterial Infusion Chemotherapy with Oxaliplatin Plus Raltitrexed as an Alternative Option in Advanced Hepatocellular Carcinoma Patients with Failure of, or Unsuitability for, Transarterial Chemoembolization. Medicina. 2022; 58(10):1343. https://doi.org/10.3390/medicina58101343

Chicago/Turabian Style

Wu, Yanfang, Susu Zheng, Zhenzhen Zhang, Guobin Chen, Xiaochun Chen, Tanghui Zheng, Xinkun Guo, Hong Chen, Meixia Wang, Xiaoying Xie, and et al. 2022. "Hepatic Arterial Infusion Chemotherapy with Oxaliplatin Plus Raltitrexed as an Alternative Option in Advanced Hepatocellular Carcinoma Patients with Failure of, or Unsuitability for, Transarterial Chemoembolization" Medicina 58, no. 10: 1343. https://doi.org/10.3390/medicina58101343

Article Metrics

Back to TopTop