The purpose of this study was to compare the efficacy and safety of drug-eluting beads transarterial chemoembolization plus camrelizumab (D-TACE-C) with conventional transarterial chemoembolization plus camrelizumab (C-TACE-C) in the treatment of patients with unresectable hepatocellular carcinoma (HCC).This was a retrospective study that evaluated the consecutive medical records of patients with unresectable HCC who had undergone D-TACE-C or C-TACE-C from April 2020 to August 2021. Efficacy of treatment was evaluated using tumor response, progression-free survival (PFS) and survival rates. The adverse events were recorded.A total of 54 patients were included in this study, including 27 patients who had received D-TACE-C treatment, and 27 patients who had received C-TACE-C treatment. The median PFS and DCR in the D-TACE-C group were significantly longer than those for the C-TACE-C group (PFS: 10 vs. 3 months, P=.017; DCR: 70.4% vs. 40.7%, P = .028). Cox regression analysis showed that D-TACE-C was the only protective factor for PFS. The 6-month and 12-month survival rates in D-TACE-C group and C-TACE-C group were 85.2% versus 79.4% (P = .646) and 65.2% versus 65.1% (P = .903), respectively. Reactive cutaneous capillary endothelial proliferation was the most common adverse event associated with the treatment. There was no significant difference in the adverse events related to TACE and camrelizumab between the two groups. No treatment-related deaths occurred in this study.D-TACE-C is a safe and well-tolerated treatment, and may produce better PFS and tumor response in patients with unresectable HCC than C-TACE-C.
Camrelizumab (a PD-1 inhibitor) has been used as a potential therapy in unresectable advanced esophageal squamous cell carcinoma (ESCC) along with adjuvant treatment in locally advanced ESCC, exhibiting an acceptable efficacy and safety profile. This pilot study was designed to further investigate the clinical value and tolerance of neoadjuvant camrelizumab plus chemotherapy in locally advanced ESCC.A total of 16 patients with locally advanced ESCC were recruited. Patients received 2 cycles of neoadjuvant therapy including 2 doses of camrelizumab concurrent with 2 cycles of paclitaxel plus carboplatin followed by surgery 4 weeks afterward. Then, the treatment response after neoadjuvant therapy, R0 resection rate, tumor regression grade (TRG), and pathological complete remission (pCR) rate were measured. Besides, adverse events were documented. At last, progression-free survival (PFS) and overall survival (OS) were assessed.Generally, objective remission rate (ORR) was 81.3% whereas disease control rate (DCR) was 100% after neoadjuvant therapy. Concerning TRG grade, 31.3, 37.5, 18.8, and 12.5% patients reached TRG0, TRG1, TRG2, and TRG3, respectively. Then, pCR rate and R0 resection rate were 31.3 and 93.8%, respectively. Besides, mean PFS and OS were 18.3 months (95%CI: (16.2-20.5) months) and 19.2 months (95%CI: (17.7-20.7) months), respectively, with a 1-year PFS of 83% and OS of 90.9%. Adverse events included white blood cell decrease (37.5%), neutrophil decrease (31.3%), reactive cutaneous capillary endothelial proliferation (37.5%), and nausea or vomiting (25.0%), which were relatively mild and manageable.Neoadjuvant camrelizumab plus chemotherapy exhibits good efficacy and acceptable tolerance in patients with locally advanced ESCC.
BACKGROUND Radiotherapy has been widely used in the treatment of hepatocellular carcinoma (HCC). However, whether the patients should receive radiotherapy before or after surgical treatment has not been studied. The objective of the study was to compare the efficacy of the treatment in HCC patients who received pre-surgery and post-surgery radiotherapy. METHODS Data from the Surveillance, Epidemiology, and End Results (SEER) database were analyzed. Patients with surgery combined with radiotherapy were included into the study. The outcome measures were overall survival (OS) and cancer-specific survival (CSS). Propensity score matching (PSM) was used to reduce selection bias. RESULTS Before PSM, the median OS (mOS: 82 months) and median CSS (mCSS: NA) in the pre-surgery group were longer than in the post-surgery group (mOS: 21 months; mCSS: 20 months; P<0.001 for both). After PSM, the mOS and mCSS in the pre-surgery group were longer than in the post-surgery group (mOS: 45 vs. 26 months, P=0.011; mCSS: 60 vs. 26 months, P=0.003). The subgroup analysis documented that patients with single tumor, liver resection, and American Joint Committee on Cancer (AJCC) stage I and II had longer mOS and mCSS if they received pre-surgery rather than post-surgery radiotherapy (all P<0.05). Multivariate regression analysis showed patients with post-surgery radiotherapy had a higher risk of mortality than patients with post-surgery radiotherapy. CONCLUSION HCC patients with single tumor, AJCC stage I and II, or with liver resection who received pre-surgery radiotherapy have better survival benefits than patients receiving post-surgery radiotherapy, particularly if internal radiotherapy was used.
Objective To determine if iodine-125 seed implantation improved the efficacy of transarterial chemoembolization (TACE) in patients with hepatocellular carcinoma (HCC) (≤5 cm). Methods We retrospectively reviewed the medical records of 83 consecutive patients with HCC (≤5 cm) who underwent TACE or TACE–iodine-125 from January 2014 to July 2017. The primary endpoint was progression-free survival (PFS). The secondary endpoints were overall survival (OS) and objective response rate (ORR) at 3 months after the first TACE treatment. PFS and OS were calculated using the Kaplan–Meier method and compared using log-rank tests. Independent risk factors for PFS and OS were analyzed using a Cox proportional hazards model. Results Thirty-five patients received TACE–iodine-125 and 48 received TACE alone. The median OS and PFS were both significantly longer in the TACE–iodine-125 compared with the TACE-alone group (42 vs 23 months and 16 vs 8 months, respectively). The ORR was significantly higher in the TACE–iodine-125 compared with the TACE-alone group. There was no significant difference in adverse events, apart from decreased white cell count, between the two groups. Conclusion TACE–iodine-125 might be an effective and safe alternative treatment for patients with HCC (≤5 cm).
Object: This study aimed to compare the efficacy and safety of transarterial chemoembolization (TACE) combining with apatinib (TACE-apatinib) and TACE-alone for patients with advanced hepatocellular carcinoma (HCC) with hepatic arterioportal shunts (APS). Materials and Methods: This retrospective study evaluated the medical records of patients with advanced HCC with APS who underwent TACE-apatinib or TACE-alone from June 2015 to January 2019. The occlusion of the shunt was performed during the TACE procedure. The time to tumor progression (TTP) and overall survival (OS) of study patients were evaluated. The modified Response Evaluation Criteria in solid tumors (mRECIST) was used to evaluate the treatment response. The apatinib-related adverse events were recorded. Results: Fifty-eight patients were included in this study. Twenty-seven patients underwent the treatment of TACE-apatinib, and 31 received TACE-alone treatment. The median overall survival (OS) and median time of tumor progression (TTP) in the TACE-apatinib group were significantly longer than those of the TACE-alone group (OS: 12.0 vs. 9.0 months, P = 0.000; TTP: 9.0 vs. 5.0 months, P = 0.041). Multivariate analysis revealed that TACE-apatinib was a protective factor for OS, and there was no independent risk factor for TTP. In the TACE-apatinib group, the grade 3 apatinib-related adverse events occurred in four patients. Conclusion: TACE-apatinib was an efficacious and safe treatment for patients with advanced HCC with APS, and apatinib improved the efficacy of TACE in the treatment of these patients.
Background: Hepatocellular carcinoma (HCC) is a tumor with high morbidity and high mortality worldwide. DNA methylation, one of the most common epigenetic changes, might serve a vital regulatory role in cancer. Methods: To identify categories based on DNA methylation data, consensus clustering was employed. The risk signature was yielded by systematic bioinformatics analyses based on the remarkably methylated CpG sites of cluster 1. Kaplan–Meier analysis, variable regression analysis, and ROC curve analysis were further conducted to validate the prognosis predictive ability of risk signature. Gene set enrichment analysis (GSEA) was performed for functional annotation. To uncover the context of tumor immune microenvironment (TIME) of HCC, we employed the ssGSEA algorithm and CIBERSORT method and performed TIMER database exploration and single-cell RNA sequencing analysis. Additionally, quantitative real-time polymerase chain reaction was employed to determine the LRRC41 expression and preliminarily explore the latent role of LRRC41 in prognostic prediction. Finally, mutation data were analyzed by employing the “maftools” package to delineate the tumor mutation burden (TMB). Results: HCC samples were assigned into seven subtypes with different overall survival and methylation levels based on 5′-cytosine-phosphate-guanine-3′ (CpG) sites. The risk prognostic signature including two candidate genes (LRRC41 and KIAA1429) exhibited robust prognostic predictive accuracy, which was validated in the external testing cohort. Then, the risk score was significantly correlated with the TIME and immune checkpoint blockade (ICB)–related genes. Besides, a prognostic nomogram based on the risk score and clinical stage presented powerful prognostic ability. Additionally, LRRC41 with prognostic value was corroborated to be closely associated with TIME characterization in both expression and methylation levels. Subsequently, the correlation regulatory network uncovered the potential targets of LRRC41 and KIAA1429. Finally, the methylation level of KIAA1429 was correlated with gene mutation status. Conclusion: In summary, this is the first to identify HCC samples into distinct clusters according to DNA methylation and yield the CpG-based prognostic signature and quantitative nomogram to precisely predict prognosis. And the pivotal player of DNA methylation of genes in the TIME and TMB status was explored, contributing to clinical decision-making and personalized prognosis monitoring of HCC.
To investigate the change of tumor feeding artery diameter and the efficacy of metastasis inhibition after transarterial chemoembolization (TACE) combined with apatinib or TACE monotherapy for patients with advanced hepatocellular carcinoma who without metastasis.A total of 616 consecutive patients who received the treatment of TACE-apatinib or TACE in our center was enrolled. Propensity score matching (PSM) analysis was used to reduce bias. The overall survival (OS), OS-after-metastasis (OSM), time to progression (TTP), time to metastasis (TTM), time to vessel or organ metastasis (TVOM), time to lymph node metastasis, and tumor feeding artery diameter between the two treatment groups were compared.A total of 113 pairs of patients were eligible after the PSM. Time to lymph node metastasis between the two groups was not significantly different (P > 0.05). The tumor feeding artery diameter was significantly smaller after TACE-apatinib management (P < 0.001). Median OS (P < 0.001) and OSM (P < 0.001) were significantly longer in the TACE-apatinib group compared with the TACE group. Median TTP (P < 0.001), TTM (P < 0.001), and TVOM (P < 0.001) were significantly prolonged in TACE-apatinib group.TACE-apatinib treatment could improve the prognosis compared with TACE alone, and inhibit metastasis after TACE procedure with contracted tumor feeding artery for advanced HCCs without metastasis.
To compare the efficacy and safety of transcatheter arterial chemoembolization (TACE) combined with sorafenib and camrelizumab or with sorafenib alone in patients with intermediate or advanced hepatocellular carcinoma (HCC).
Abstract Background There are no exact treatments been recommended for gastrointestinal tumor liver metastasis patients. This study intends to assess the safety and efficacy of transarterial chemoembolization (TACE) plus apatinib as a combination treatment for gastrointestinal tumor liver metastasis patients. Methods From January 2015 to December 2019, 53 patients with gastrointestinal tumor liver metastases were included in the analysis. Results The median progression-free survival (mPFS) and median overall survival (mOS) numbers were 7 months and 17 months, respectively. The mPFS of metastatic sites was 12 months. The disease control rate (DCR) and objective response rate (ORR) values were 86.8% (46/53) and 49.1% (26/53), respectively. The multivariate regression analysis showed that Child-Pugh and Eastern Cooperative Oncology Group (ECOG) were independent predictors for OS. The independent predictors for overall PFS include carbohydrate antigen 724 (CA 724), CA 199, TACE session, Child-Pugh, and ECOG; while for liver tumor PFS, the independent predictors are TACE session and ECOG scores. There were no changes of leukocyte, lymphocyte, carcinoemnryonicantigen (CEA), CA 724, CA 199, CA 125 of patients before receiving the treatments and one month after receiving the treatment (all P>0.05). Common adverse events at any grade were poor appetite (64.2%), hypertension (56.6%), and hand-foot syndrome (50.9%). Some patients had III or IV-grade adverse events after receiving the TACE-apatinib combination treatment. However, these adverse events alleviated after reducing apatinib administration or receiving symptomatic treatments. Conclusions The TACE-apatinib combination might be an effective treatment option against gastrointestinal tumors liver metastases with promising safety.