Next Article in Journal
An Integrated Computational and Experimental Approach to Formulate Tamanu Oil Bigels as Anti-Scarring Agent
Next Article in Special Issue
Towards Effective Targeted Alpha Therapy for Neuroendocrine Tumours: A Review
Previous Article in Journal
From PROTAC to TPD: Advances and Opportunities in Targeted Protein Degradation
Previous Article in Special Issue
Alpha-Emitting Radionuclides: Current Status and Future Perspectives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Safety and Efficacy of Selective Internal Radionuclide Therapy with 90Y Glass Microspheres in Patients with Progressive Hepatocellular Carcinoma after the Failure of Repeated Transarterial Chemoembolization

1
Department of Nuclear Medicine, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
2
MVZ Radiologie, Nuklearmedizin und Strahlentherapie Essen GmbH, Ruüttenscheider Str. 191, 45131 Essen, Germany
3
Department of Nuclear Medicine, Clinic for Radiology and Nuclear Medicine, University Hospital, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
4
Department of Nuclear Medicine, Saarland University Medical Center, Kirrberger Straße, 66421 Homburg, Germany
5
Department of Internal Medicine, University Hospital Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
6
Department of Internal Medicine, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
7
Department of Diagnostic and Interventional Radiology and Neuroradiology, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
8
MVZ CDT Strahleninstitut GmbH, Turiner Straße 2, 50668 Cologne, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2024, 17(1), 101; https://doi.org/10.3390/ph17010101
Submission received: 29 October 2023 / Revised: 4 January 2024 / Accepted: 8 January 2024 / Published: 11 January 2024
(This article belongs to the Special Issue Therapeutic Radionuclides in Nuclear Medicine)

Abstract

:
Transarterial chemoembolization (TACE) is currently the standard of care in patients with unresectable hepatocellular carcinoma (HCC), and selective internal radionuclide therapy (SIRT) with 90Y microspheres is mainly used as an alternative modality in patients considered poor candidates for TACE. Treatment with sorafenib is the recommended option for patients with progressive disease after TACE. This study aims to evaluate the safety and efficacy of SIRT with glass microspheres in patients with progressive HCC after repeated TACE who are not eligible for treatment with sorafenib. Forty-seven patients with progressive HCC after a median of three TACE sessions (range 2–14) underwent SIRT (3.5 ± 1.5 GBq; liver target dose 110–120 Gy). Toxicity was recorded 4 and 12 weeks after treatment and reported according to the Common Terminology Criteria for Adverse Events Version 5.0. Treatment response was assessed three months after SIRT using multiphase computed tomography and modified criteria in solid tumors (mRECIST). Survival analyses were performed using Kaplan–Meier curves and a Cox proportional hazards model for uni- and multivariate analyses. Significant but reversible hepatotoxicity (≥grade 3) occurred in five patients (11%). No radioembolization-induced liver disease (REILD) was observed. The number of previous TACE sessions and cumulative administered activity did not predict the incidence of post-SIRT significant hepatotoxicity. Treatment responses consisted of partial responses in 26 (55%), stable disease in 12 (26%), and progressive disease in 9 (19%) patients. The median overall survival (OS) was 11 months (95% confidence interval (CI), 9–13), and objective responses to SIRT were associated with a longer OS (p = 0.008). Significant hepatotoxicity (≥grade 3) after SIRT was a contributor to impaired survival (median OS 6 months (95% CI, 4–8) vs. 12 months (95% CI, 10–14), p < 0.001). SIRT with glass microspheres is a safe and effective salvage treatment for patients with progressive HCC refractory to TACE who are considered poor candidates for sorafenib treatment.

1. Introduction

The current Practice Guidance by the American Association for the Study of Liver Diseases recommends further diagnostic work-up for hepatocellular carcinoma (HCC) in the presence of a hepatic lesion >1 cm and an increase in alpha-fetoprotein >20 ng/mL in serum. Multiphase CT or MRI are used as imaging modalities, and histological analysis represents the gold standard [1]. In unresectable HCC, palliative liver-directed treatment options like transarterial chemoembolization (TACE) or selective internal radionuclide therapy (SIRT) can significantly reduce hepatic tumor burden and may increase survival in patients with liver-dominant disease [2,3,4].
TACE is currently the standard treatment for patients with locally advanced HCC without vascular invasion or extrahepatic spread (intermediate stage) [3,5,6,7]. However, a sufficient response after a single TACE session is rare, and often repeated TACE is required to achieve a good response [8]. Accordingly, at least two TACE sessions should be performed before abandoning the procedure [9]. Despite increasing evidence supporting the favorable efficacy of SIRT with 90Yttrium (90Y) microspheres in patients with intermediate to advanced HCC [10,11,12,13,14,15], the lack of prospective randomized clinical trials has currently limited its role as an alternative method for patients considered poor candidates for TACE. For patients refractory to repeated TACE, systemic treatment with the multikinase inhibitor sorafenib is recommended, but sometimes with suboptimal tolerability outweighing the survival benefits [16,17,18]. This leaves SIRT as the only treatment option after failure of TACE in this setting [19,20].
Repeated TACE can be associated with vascular injury and sometimes with liver function deterioration. Furthermore, profound TACE-induced tumor dearterialization may reduce the selective deployment of 90Y microspheres inside the tumor vasculature [21,22,23]. Therefore, SIRT might be associated with a higher risk of treatment failure and severe hepatic toxicity in patients previously treated with repeated TACE. Conversely, a progressive tumor probably develops new tumor vessels, which might compensate for the TACE-induced devascularization. Thus, this study aims to assess the safety and efficacy of SIRT with glass 90Y microspheres (TheraSphere™, Boston Scientific Corporation, Ottawa, ON, Canada) in patients with progressive HCC refractory to repeated TACE but not eligible for sorafenib treatment.

2. Results

2.1. Toxicity

The mean treatment activity per patient was 3.3 ± 1.5 GBq, and the mean follow-up time was 17 ± 2 months. Three of the 47 patients were still alive at the time of analysis. Recorded acute adverse events were as follows: fatigue in 18 (38%), nausea without vomiting in 10 (21%), fever in 9 (19%), and transient abdominal pain in 5 patients (11%). No patient needed hospitalization due to the reported adverse events, and all symptoms resolved within the first six weeks after SIRT. Prior to the treatment, 24 patients had impaired liver function (grade I: 24, grade II: 7, ≥grade III: 0). Post-SIRT hepatotoxicity was defined as newly impaired liver function (albumin, bilirubin, AST/ALT, INR, ascites) or as deterioration in CTCAE-grading after SIRT. Post-SIRT hepatotoxicity was observed in 36 patients. In 10 patients, liver function parameters deteriorated (grade II), and in 5 patients, a significant new hepatic toxicity of grade III–IV occurred. Detailed information about toxicity after treatment is given in Table 1: 12 patients showed elevated liver transaminase (10 grade I, 2 grade II) within six weeks after treatment, and 20 patients had biliary toxicity (4 grade I; 12 grade II and 4 grade III–IV). Fourteen patients showed relevant hepatic toxicity (grade II) based on both liver transaminase and bilirubin concentrations. Portal vein thrombosis and high hepatic tumor load (≥25%) were the independent contributing factors to treatment-induced significant hepatotoxicity, as depicted in Table 2.
All five patients with newly induced hepatic toxicity had ≤3 TACE sessions prior to SIRT, and no increase in the incidence of significant toxicity was observed in patients with >3 prior TACE sessions (Figure 1). High cumulative activity (≥3.5 GBq) during SIRT and a higher number of previous TACE sessions was also not associated with increased hepatic toxicity (p = 0.706). Significant hepatic toxicity was resolved within 12 weeks in all but one patient who died because of acute renal failure. No severe radioembolization-induced liver disease (REILD) was documented, which was defined as new relevant serum total bilirubin elevation (≥3 mg/dL) combined with new ascites 1–2 months after treatment without tumor progression or bile duct obstruction. No radiation-induced pneumonitis, gastroduodenal ulceration, or other organ toxicity was observed.

2.2. Response and Survival

Restaging according to mRECIST yielded a partial remission (PR) in 26 (55%), stable disease (SD) in 12 (26%), and progressive disease (PD) in 9 (19%) patients. Complete remission was not observed in our cohort. An example of a patient with a partial response according to mRECIST is displayed in Figure 2.
The median time to progression after SIRT was 7 months (95% CI, 6–8) and the median OS was 11 months (95% CI, 9–13). Patients showing objective responses to SIRT (i.e., PR) had a median OS of 14 months (95% CI, 11–17) as opposed to 7 months (95% CI, 5–9) in the remaining patients (p = 0.008), as illustrated in Figure 3A. In contrast, progression after SIRT (i.e., PD) was associated with a shorter survival (median OS, 8 months (95% CI, 2–14) versus 12 months (95% CI, 9–15), p = 0.013, Figure 3B).
Significant hepatotoxicity (≥grade III) after SIRT was also a contributor to impaired survival (median OS, 6 months (95% CI, 4–8) versus 12 months (95% CI, 10–14), p < 0.001, Figure 4A). Patients developing grade II hepatotoxicity showed a trend towards a shorter survival (median OS, 7 months (95% CI, 6–8) versus 14 (95% CI, 11–17), p = 0.007, Figure 4B).
Among the baseline characteristics, a relevant decrease in hepatic functional reserve (≥grade II) was the only independent predictor of survival, as depicted by the multivariate analysis (HR; 95% CI, 5.5 (1.5–19.9), p = 0.009). The analysis of various baseline factors for potential contribution to OS is shown in Table 3.

3. Discussion

This retrospective study provides the first results regarding the risk factors for the hepatotoxicity of SIRT with glass-based 90Y microspheres (TheraSphere) in a well-characterized patient cohort (n = 47) with unresectable HCC failing repeated TACE. In clinical practice, previous TACE is considered a major risk factor for serious SIRT-induced toxicity. Fitting to this, the detected rate of significant hepatotoxicity in our cohort (11%) was higher than the previously reported rate of 4–9% in treatment-naive patients or heterogeneous cohorts regarding previous treatment modalities [2,24,25,26,27,28,29,30,31,32,33]. However, treatment-induced hepatotoxicity was almost always reversible, and liver function parameters returned to pre-treatment levels in all but one patient.
Portal vein thrombosis (PVT) is an absolute contraindication for TACE. On the contrary, SIRT with glass microspheres has a very low embolic tendency and can be safely applied to patients with PVT [34,35,36]. Although PVT was associated with a higher incidence of moderate hepatotoxicity in our cohort, the survival outcome of patients who had developed PVT under TACE did not differ from patients without PVT. Among all other analyzed baseline characteristics, only a hepatic tumor load of >25% was a risk factor for significant hepatotoxicity. This observation is in line with the reported data after treatment with 90Y resin microspheres in a large prospective observational study (CIRT study). In a recent study on 1027 patients in a heterogenous patient cohort with various liver tumors, including HCC in 422 patients, a tumor load of >20% was a significant predictor of increased hepatotoxicity (p = 0.0283) [37]. Notably, the number of previous TACE sessions was not a predictor of hepatotoxicity in our cohort, encouraging the consideration of SIRT in patients heavily pre-treated with TACE, similar to previous findings in a smaller patient group (n = 29), indicating the suitability of SIRT after TACE not to be limited by increased risk of toxicity [38].
Achievement of disease control in 81% is promising and compares favorably with the other treatment modalities. Sorafenib, as the recommended agent for TACE-refractory HCC lesions [16,17,39,40], is commonly associated with adverse effects leading to treatment interruption or even permanent drug discontinuation [16,17,39]. Moreover, an objective radiological response is rarely observed after treatment with sorafenib. The reported overall survival in the main clinical phase III trial of sorafenib (SHARP trial) was 10.7 months [17]. Although comparing our retrospective data with results from prospective trials is of limited validity, the objective response rate (ORR) of 55% and median OS of 11 months (CI 95%, 9–13) in our patients ineligible for treatment with sorafenib is very encouraging. Furthermore, responders (i.e., PR) had a significantly longer survival in our cohort (median OS 14 versus 7 months, p = 0.008), underlining the impact of ORR on the survival outcomes of HCC patients after SIRT [15]. The rate of hepatotoxicity in our study was lower than the reported rate after sorafenib treatment. Johnson et al. analyzed the efficacy and safety of SIRT as a salvage therapy after ≥1 TACE, resulting in a slightly inferior OS of 8.4 months; however, BCLC stage C was more prevalent in their cohort (72.5% vs. 47%) [38]. Fitting to this, in a study by Reeves et al., a subgroup of BCLC stage B patients with 1–7 TACE before SIRT (n = 7) reached an OS of 14.8 months [41].
In addition to tumor progression, therapy-induced hepatotoxicity may affect the survival of patients with HCC [15,17]. In a retrospective study, grade II toxicity has been suggested as a risk factor for poor survival outcomes. Correspondingly, relevant hepatotoxicity impaired the survival outcome in our cohort (p < 0.007). Significant hyperbilirubinemia (grade III/IV), as a hallmark of REILD, has been reported in 14% of patients undergoing 90Y glass microsphere SIRT [24]. Although 9% of our cohort developed grade III transient biliary toxicity, no REILD was observed in our cohort.
Our findings support the application of SIRT with glass microspheres after undergoing repeated TACE who are ineligible for treatment with sorafenib. SIRT could induce disease stabilization in most patients, leading to an improved survival outcome. Hepatotoxicity was reversible and the number of previous TACE was not a risk factor. However, the retrospective design and small patient group limit the statistical power and ability to generalize from our results regarding the subgroup analysis and baseline factors with a potential impact on survival. Furthermore, pathological data were, unfortunately, not available to be included in this clinical observational study. It would be interesting to analyze pathological parameters and the treatment efficacy, which might be the subject of further studies.

4. Materials and Methods

4.1. Patient Characteristics

Forty-seven patients with TACE-refractory HCC (38 men, 9 women; age range: 40–85 years; mean age: 69 years) treated with SIRT in the Department of Nuclear Medicine, University Duisburg-Essen, were included in this retrospective analysis [42]. The decision to perform SIRT was based on interdisciplinary consent after discussion in a multidisciplinary tumor board. All patients had progressive liver tumors despite repeated TACE procedures (median: 3, range 2–14) and were not suitable for sorafenib treatment. Apart from repeated TACE, previous treatments were comprised of radiofrequency ablation (n = 9), surgical resection (n = 7), liver transplantation (n = 4), and transarterial embolization or ethanol injection (n = 2). All patients fulfilled the general inclusion criteria for radioembolization [43,44]. Twenty-three patients presented with a unilobar, and 24 patients with a bilobar hepatic tumor spread. At the time of SIRT, 25 patients were classified as stage B and 22 as stage C according to the Barcelona Clinic Liver Cancer (BCLC) staging classification. In 39 patients, HCC was confined to the liver, while 8 patients showed a liver-dominant disease with extrahepatic metastases. In these eight patients, the tumor board identified the extrahepatic metastases as not predominately prognostically relevant regarding survival, size, quantity, and localization of the metastases. The baseline patient characteristics are presented in Table 4. The local committee on ethics approved this retrospective study, and all subjects signed a written informed consent to treatment prior to evaluation and radioembolization session.

4.2. Radioembolization Procedure

Intra-abdominal and excessive pulmonary (lung-shunt fraction) deposition were excluded prior to radioembolization by a pre-treatment diagnostic angiogram with planar and SPECT/CT 99mTechnetium-HSA (human serum albumin microspheres) imaging after an intra-arterial injection of 150 MBq of 99mTc-HSA [45]. Radioembolization was performed within an interval of 1–4 weeks following diagnostic angiography using glass-based 90Y microspheres (TheraSphere™). The prescription of activity was derived from the MIRD-based dose calculation method provided by the manufacturer (Boston Scientific Corporation, Marlborough, MA, USA, former BTG plc, London, UK, former Nordion Inc., Ottawa, ON, Canada) to achieve a standard target dose of 100–120 Gy. The liver was treated in a single session (unilobar, n = 23, whole liver, n = 1) or in a sequential lobar fashion (n = 23 patients). Post-treatment 90Y bremsstrahlung imaging was performed to document target accumulation. Parameters for liver function (albumin, bilirubin, AST/ALT, INR, ascites) were determined before as well as 4 and 12 weeks after each SIRT. Hepatic toxicity was classified according to the Common Terminology Criteria for Adverse Events Version 5.0 (CTCAE v.5.0). Morphological response to SIRT was assessed using contrast-agent-enhanced computed tomography (CT, early arterial and venous phase) 3 months after SIRT using modified response criteria in solid tumors (mRECIST) [46,47].

4.3. Statistical Analysis

Statistical analyses were performed using the SPSS software package version 29.0 (IBM, Armonk, NY, USA). Graph-Pad Prism version 10.1 (GraphPad Software, San Diego, CA, USA) was used to plot graphs. The results were presented as mean ± standard deviation for continuous variables; categorical variables are presented as frequencies with respective percentages. The association of treatment-induced hepatic toxicity (grade I–IV) with the baseline characteristics of the study population, number of previous TACE sessions, and administered activity were examined, applying non-parametric tests for independent samples as well as multiple regression analysis. Survival assessment from the start of radioembolization was performed using the Kaplan–Meier method. Overall survival (OS) was assessed from the first radioembolization session, and the death of patients was considered as an event for OS irrespective of the cause. Survival outcomes were stratified by various variables and compared using the log-rank test. Multivariate analysis (Cox proportional hazards model) was performed with those variables showing at least a trend (p < 0.1) of influence on the univariate analysis (log-rank test). A p value < 0.05 was considered significant.

5. Conclusions

SIRT with glass microspheres is an effective salvage treatment in patients with progressive HCC refractory to TACE who are ineligible for treatment with sorafenib. SIRT provides disease stabilization and improves survival. The rate of significant hepatotoxicity was acceptable, considering the lack of alternative treatment options. Furthermore, the number of previous TACE sessions should not preclude the consideration of SIRT in heavily pre-treated patients fulfilling the established prerequisites.

Author Contributions

Conceptualization, A.S.; methodology, A.S., A.B. (Alexander Bellendorf) and N.M.; validation, S.P.M., S.E. and A.B. (Andreas Bockisch); formal analysis, A.B. (Alexander Bellendorf), N.M. and A.S.; investigation, A.B. (Alexander Bellendorf), T.D.P., H.G. and A.S.; resources, S.P.M., J.G., J.B. and A.B. (Andreas Bockisch); data curation, A.B. (Alexander Bellendorf) and H.G.; writing—original draft preparation, A.B. (Alexander Bellendorf) and N.M.; writing—review and editing, S.E., S.P.M., A.B. (Andreas Bockisch) and A.S.; visualization, A.B. (Alexander Bellendorf) and N.M.; supervision, A.S. and S.P.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This retrospective study was approved by the institutional review board.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The datasets analyzed and/or analyzed during the current study are available from the corresponding author on reasonable request.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Marrero, J.A.; Kulik, L.M.; Sirlin, C.B.; Zhu, A.X.; Finn, R.S.; Abecassis, M.M.; Roberts, L.R.; Heimbach, J.K. Diagnosis, Staging, and Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology 2018, 68, 723–750. [Google Scholar] [CrossRef] [PubMed]
  2. Salem, R.; Lewandowski, R.J.; Mulcahy, M.F.; Riaz, A.; Ryu, R.K.; Ibrahim, S.; Atassi, B.; Baker, T.; Gates, V.; Miller, F.H.; et al. Radioembolization for hepatocellular carcinoma using Yttrium-90 microspheres: A comprehensive report of long-term outcomes. Gastroenterology 2010, 138, 52–64. [Google Scholar] [CrossRef]
  3. Cammà, C.; Schepis, F.; Orlando, A.; Albanese, M.; Shahied, L.; Trevisani, F.; Andreone, P.; Craxì, A.; Cottone, M. Transarterial chemoembolization for unresectable hepatocellular carcinoma: Meta-analysis of randomized controlled trials. Radiology 2002, 224, 47–54. [Google Scholar] [CrossRef]
  4. Brown, Z.J.; Tsilimigras, D.I.; Ruff, S.M.; Mohseni, A.; Kamel, I.R.; Cloyd, J.M.; Pawlik, T.M. Management of Hepatocellular Carcinoma: A Review. JAMA Surg. 2023, 158, 410–420. [Google Scholar] [CrossRef] [PubMed]
  5. 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]
  6. Dalzell, C.G.; Taylor, A.C.; White, S.B. New Insights on Liver-Directed Therapies in Hepatocellular Carcinoma. Cancers 2023, 15, 5749. [Google Scholar] [CrossRef]
  7. Vogl, T.J.; Naguib, N.N.; Nour-Eldin, N.E.; Rao, P.; Emami, A.H.; Zangos, S.; Nabil, M.; Abdelkader, A. Review on transarterial chemoembolization in hepatocellular carcinoma: Palliative, combined, neoadjuvant, bridging, and symptomatic indications. Eur. J. Radiol. 2009, 72, 505–516. [Google Scholar] [CrossRef] [PubMed]
  8. Terzi, E.; Golfieri, R.; Piscaglia, F.; Galassi, M.; Dazzi, A.; Leoni, S.; Giampalma, E.; Renzulli, M.; Bolondi, L. Response rate and clinical outcome of HCC after first and repeated cTACE performed “on demand”. J. Hepatol. 2012, 57, 1258–1267. [Google Scholar] [CrossRef]
  9. Georgiades, C.; Geschwind, J.F.; Harrison, N.; Hines-Peralta, A.; Liapi, E.; Hong, K.; Wu, Z.; Kamel, I.; Frangakis, C. Lack of response after initial chemoembolization for hepatocellular carcinoma: Does it predict failure of subsequent treatment? Radiology 2012, 265, 115–123. [Google Scholar] [CrossRef]
  10. Lance, C.; McLennan, G.; Obuchowski, N.; Cheah, G.; Levitin, A.; Sands, M.; Spain, J.; Srinivas, S.; Shrikanthan, S.; Aucejo, F.N.; et al. Comparative analysis of the safety and efficacy of transcatheter arterial chemoembolization and yttrium-90 radioembolization in patients with unresectable hepatocellular carcinoma. J. Vasc. Interv. Radiol. 2011, 22, 1697–1705. [Google Scholar] [CrossRef]
  11. Salem, R.; Lewandowski, R.J.; Kulik, L.; Wang, E.; Riaz, A.; Ryu, R.K.; Sato, K.T.; Gupta, R.; Nikolaidis, P.; Miller, F.H.; et al. Radioembolization results in longer time-to-progression and reduced toxicity compared with chemoembolization in patients with hepatocellular carcinoma. Gastroenterology 2011, 140, 497–507.e492. [Google Scholar] [CrossRef]
  12. Sangro, B.; Salem, R. Transarterial chemoembolization and radioembolization. Semin. Liver Dis. 2014, 34, 435–443. [Google Scholar] [CrossRef] [PubMed]
  13. Lewandowski, R.J.; Kulik, L.M.; Riaz, A.; Senthilnathan, S.; Mulcahy, M.F.; Ryu, R.K.; Ibrahim, S.M.; Sato, K.T.; Baker, T.; Miller, F.H.; et al. A comparative analysis of transarterial downstaging for hepatocellular carcinoma: Chemoembolization versus radioembolization. Am. J. Transplant. 2009, 9, 1920–1928. [Google Scholar] [CrossRef] [PubMed]
  14. Memon, K.; Lewandowski, R.J.; Riaz, A.; Salem, R. Yttrium 90 microspheres for the treatment of hepatocellular carcinoma. Recent. Results Cancer Res. 2013, 190, 207–224. [Google Scholar] [CrossRef]
  15. Mazzaferro, V.; Sposito, C.; Bhoori, S.; Romito, R.; Chiesa, C.; Morosi, C.; Maccauro, M.; Marchianò, A.; Bongini, M.; Lanocita, R.; et al. Yttrium-90 radioembolization for intermediate-advanced hepatocellular carcinoma: A phase 2 study. Hepatology 2013, 57, 1826–1837. [Google Scholar] [CrossRef] [PubMed]
  16. 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]
  17. 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] [PubMed]
  18. Keating, G.M. Sorafenib: A Review in Hepatocellular Carcinoma. Target. Oncol. 2017, 12, 243–253. [Google Scholar] [CrossRef]
  19. Vilgrain, V.; Pereira, H.; Assenat, E.; Guiu, B.; Ilonca, A.D.; Pageaux, G.P.; Sibert, A.; Bouattour, M.; Lebtahi, R.; Allaham, W.; et al. Efficacy and safety of selective internal radiotherapy with yttrium-90 resin microspheres compared with sorafenib in locally advanced and inoperable hepatocellular carcinoma (SARAH): An open-label randomised controlled phase 3 trial. Lancet Oncol. 2017, 18, 1624–1636. [Google Scholar] [CrossRef]
  20. Chow, P.K.H.; Gandhi, M.; Tan, S.B.; Khin, M.W.; Khasbazar, A.; Ong, J.; Choo, S.P.; Cheow, P.C.; Chotipanich, C.; Lim, K.; et al. SIRveNIB: Selective Internal Radiation Therapy Versus Sorafenib in Asia-Pacific Patients with Hepatocellular Carcinoma. J. Clin. Oncol. 2018, 36, 1913–1921. [Google Scholar] [CrossRef]
  21. Clark, T.W. Complications of hepatic chemoembolization. Semin. Intervent Radiol. 2006, 23, 119–125. [Google Scholar] [CrossRef] [PubMed]
  22. Sun, Z.; Li, G.; Ai, X.; Luo, B.; Wen, Y.; Zhao, Z.; Dong, S.; Guan, J. Hepatic and biliary damage after transarterial chemoembolization for malignant hepatic tumors: Incidence, diagnosis, treatment, outcome and mechanism. Crit. Rev. Oncol. Hematol. 2011, 79, 164–174. [Google Scholar] [CrossRef]
  23. Binzaqr, S.; Debordeaux, F.; Blanc, J.F.; Papadopoulos, P.; Hindie, E.; Lapouyade, B.; Pinaquy, J.B. Efficacy of Selective Internal Radiation Therapy for Hepatocellular Carcinoma Post-Incomplete Response to Chemoembolization. Pharmaceuticals 2023, 16, 1676. [Google Scholar] [CrossRef] [PubMed]
  24. Sangro, B.; Gil-Alzugaray, B.; Rodriguez, J.; Sola, I.; Martinez-Cuesta, A.; Viudez, A.; Chopitea, A.; Inarrairaegui, M.; Arbizu, J.; Bilbao, J.I. Liver disease induced by radioembolization of liver tumors: Description and possible risk factors. Cancer 2008, 112, 1538–1546. [Google Scholar] [CrossRef] [PubMed]
  25. Kennedy, A.S.; McNeillie, P.; Dezarn, W.A.; Nutting, C.; Sangro, B.; Wertman, D.; Garafalo, M.; Liu, D.; Coldwell, D.; Savin, M.; et al. Treatment parameters and outcome in 680 treatments of internal radiation with resin 90Y-microspheres for unresectable hepatic tumors. Int. J. Radiat. Oncol. Biol. Phys. 2009, 74, 1494–1500. [Google Scholar] [CrossRef] [PubMed]
  26. Atassi, B.; Bangash, A.K.; Lewandowski, R.J.; Ibrahim, S.; Kulik, L.; Mulcahy, M.F.; Murthy, R.; Ryu, R.K.; Sato, K.T.; Miller, F.H.; et al. Biliary sequelae following radioembolization with Yttrium-90 microspheres. J. Vasc. Interv. Radiol. 2008, 19, 691–697. [Google Scholar] [CrossRef] [PubMed]
  27. Piana, P.M.; Gonsalves, C.F.; Sato, T.; Anne, P.R.; McCann, J.W.; Bar Ad, V.; Eschelman, D.J.; Parker, L.; Doyle, L.A.; Brown, D.B. Toxicities after radioembolization with yttrium-90 SIR-spheres: Incidence and contributing risk factors at a single center. J. Vasc. Interv. Radiol. 2011, 22, 1373–1379. [Google Scholar] [CrossRef] [PubMed]
  28. Lewandowski, R.J.; Minocha, J.; Memon, K.; Riaz, A.; Gates, V.L.; Ryu, R.K.; Sato, K.T.; Omary, R.; Salem, R. Sustained safety and efficacy of extended-shelf-life (90)Y glass microspheres: Long-term follow-up in a 134-patient cohort. Eur. J. Nucl. Med. Mol. Imaging 2014, 41, 486–493. [Google Scholar] [CrossRef]
  29. Salem, R.; Lewandowski, R.J.; Atassi, B.; Gordon, S.C.; Gates, V.L.; Barakat, O.; Sergie, Z.; Wong, C.Y.; Thurston, K.G. Treatment of unresectable hepatocellular carcinoma with use of 90Y microspheres (TheraSphere): Safety, tumor response, and survival. J. Vasc. Interv. Radiol. 2005, 16, 1627–1639. [Google Scholar] [CrossRef]
  30. Carr, B.I. Hepatic arterial 90Yttrium glass microspheres (Therasphere) for unresectable hepatocellular carcinoma: Interim safety and survival data on 65 patients. Liver Transpl. 2004, 10, S107–S110. [Google Scholar] [CrossRef]
  31. Goin, J.E.; Salem, R.; Carr, B.I.; Dancey, J.E.; Soulen, M.C.; Geschwind, J.F.; Goin, K.; Van Buskirk, M.; Thurston, K. Treatment of unresectable hepatocellular carcinoma with intrahepatic yttrium 90 microspheres: Factors associated with liver toxicities. J. Vasc. Interv. Radiol. 2005, 16, 205–213. [Google Scholar] [CrossRef]
  32. Hilgard, P.; Hamami, M.; Fouly, A.E.; Scherag, A.; Muller, S.; Ertle, J.; Heusner, T.; Cicinnati, V.R.; Paul, A.; Bockisch, A.; et al. Radioembolization with yttrium-90 glass microspheres in hepatocellular carcinoma: European experience on safety and long-term survival. Hepatology 2010, 52, 1741–1749. [Google Scholar] [CrossRef]
  33. Szyszko, T.; Al-Nahhas, A.; Tait, P.; Rubello, D.; Canelo, R.; Habib, N.; Jiao, L.; Wasan, H.; Bansi, D.; Thillainayagam, A.; et al. Management and prevention of adverse effects related to treatment of liver tumours with 90Y microspheres. Nucl. Med. Commun. 2007, 28, 21–24. [Google Scholar] [CrossRef] [PubMed]
  34. Kulik, L.M.; Carr, B.I.; Mulcahy, M.F.; Lewandowski, R.J.; Atassi, B.; Ryu, R.K.; Sato, K.T.; Benson, A., 3rd; Nemcek, A.A., Jr.; Gates, V.L.; et al. Safety and efficacy of 90Y radiotherapy for hepatocellular carcinoma with and without portal vein thrombosis. Hepatology 2008, 47, 71–81. [Google Scholar] [CrossRef] [PubMed]
  35. Memon, K.; Kulik, L.; Lewandowski, R.J.; Mulcahy, M.F.; Benson, A.B.; Ganger, D.; Riaz, A.; Gupta, R.; Vouche, M.; Gates, V.L.; et al. Radioembolization for hepatocellular carcinoma with portal vein thrombosis: Impact of liver function on systemic treatment options at disease progression. J. Hepatol. 2013, 58, 73–80. [Google Scholar] [CrossRef] [PubMed]
  36. Salem, R.; Lewandowski, R.; Roberts, C.; Goin, J.; Thurston, K.; Abouljoud, M.; Courtney, A. Use of Yttrium-90 glass microspheres (TheraSphere) for the treatment of unresectable hepatocellular carcinoma in patients with portal vein thrombosis. J. Vasc. Interv. Radiol. 2004, 15, 335–345. [Google Scholar] [CrossRef]
  37. Maleux, G.; Albrecht, T.; Arnold, D.; Bargellini, I.; Cianni, R.; Helmberger, T.; Kolligs, F.; Munneke, G.; Peynircioglu, B.; Sangro, B.; et al. Predictive Factors for Adverse Event Outcomes After Transarterial Radioembolization with Yttrium-90 Resin Microspheres in Europe: Results from the Prospective Observational CIRT Study. Cardiovasc. Intervent Radiol. 2023, 46, 852–867. [Google Scholar] [CrossRef]
  38. Johnson, G.E.; Monsky, W.L.; Valji, K.; Hippe, D.S.; Padia, S.A. Yttrium-90 Radioembolization as a Salvage Treatment following Chemoembolization for Hepatocellular Carcinoma. J. Vasc. Interv. Radiol. 2016, 27, 1123–1129. [Google Scholar] [CrossRef]
  39. Iavarone, M.; Cabibbo, G.; Piscaglia, F.; Zavaglia, C.; Grieco, A.; Villa, E.; Camma, C.; Colombo, M.; on behalf of the SOFIA (SOraFenib Italian Assessment) Study Group. Field-practice study of sorafenib therapy for hepatocellular carcinoma: A prospective multicenter study in Italy. Hepatology 2011, 54, 2055–2063. [Google Scholar] [CrossRef]
  40. Bruix, J.; Sherman, M.; American Association for the Study of Liver, D. Management of hepatocellular carcinoma: An update. Hepatology 2011, 53, 1020–1022. [Google Scholar] [CrossRef]
  41. Reeves, H.L.; Reicher, J.; Priona, G.; Manas, D.M.; Littler, P. Selective internal radiation therapy (SIRT) for hepatocellular carcinoma (HCC): Informing clinical practice for multidisciplinary teams in England. Frontline Gastroenterol. 2023, 14, 45–51. [Google Scholar] [CrossRef]
  42. Bruix, J.; Sherman, M.; Llovet, J.M.; Beaugrand, M.; Lencioni, R.; Burroughs, A.K.; Christensen, E.; Pagliaro, L.; Colombo, M.; Rodés, J. Clinical management of hepatocellular carcinoma. Conclusions of the Barcelona-2000 EASL conference. European Association for the Study of the Liver. J. Hepatol. 2001, 35, 421–430. [Google Scholar] [CrossRef] [PubMed]
  43. Salem, R.; Thurston, K.G. Radioembolization with 90Yttrium microspheres: A state-of-the-art brachytherapy treatment for primary and secondary liver malignancies. Part 1: Technical and methodologic considerations. J. Vasc. Interv. Radiol. 2006, 17, 1251–1278. [Google Scholar] [CrossRef] [PubMed]
  44. Kennedy, A.; Nag, S.; Salem, R.; Murthy, R.; McEwan, A.J.; Nutting, C.; Benson, A., 3rd; Espat, J.; Bilbao, J.I.; Sharma, R.A.; et al. Recommendations for radioembolization of hepatic malignancies using yttrium-90 microsphere brachytherapy: A consensus panel report from the radioembolization brachytherapy oncology consortium. Int. J. Radiat. Oncol. Biol. Phys. 2007, 68, 13–23. [Google Scholar] [CrossRef] [PubMed]
  45. Sabet, A.; Ahmadzadehfar, H.; Muckle, M.; Haslerud, T.; Wilhelm, K.; Biersack, H.J.; Ezziddin, S. Significance of oral administration of sodium perchlorate in planning liver-directed radioembolization. J. Nucl. Med. 2011, 52, 1063–1067. [Google Scholar] [CrossRef]
  46. Lencioni, R.; Llovet, J.M. Modified RECIST (mRECIST) assessment for hepatocellular carcinoma. Semin. Liver Dis. 2010, 30, 52–60. [Google Scholar] [CrossRef]
  47. Bruix, J.; Sherman, M. Management of hepatocellular carcinoma. Hepatology 2005, 42, 1208–1236. [Google Scholar] [CrossRef]
Figure 1. The relation of significant (grade ≥3) hepatotoxicity and (1) administered activity and (2) number of previous transarterial chemoembolization (TACE) sessions.
Figure 1. The relation of significant (grade ≥3) hepatotoxicity and (1) administered activity and (2) number of previous transarterial chemoembolization (TACE) sessions.
Pharmaceuticals 17 00101 g001
Figure 2. Computer tomography images of a patient showing a partial response after selective internal radiotherapy (red arrow indicates the tumor lesion).
Figure 2. Computer tomography images of a patient showing a partial response after selective internal radiotherapy (red arrow indicates the tumor lesion).
Pharmaceuticals 17 00101 g002
Figure 3. (A) Objective remission (partial response (PR)) after selective internal radiotherapy prolongs overall survival (OS), and (B) early progressive disease (PD) impairs overall survival.
Figure 3. (A) Objective remission (partial response (PR)) after selective internal radiotherapy prolongs overall survival (OS), and (B) early progressive disease (PD) impairs overall survival.
Pharmaceuticals 17 00101 g003
Figure 4. (A) Significant (grade ≥ 3) toxicity after selective internal radiotherapy reduced survival, and (B) moderate changes in liver function (CTC II) impaired overall survival (OS).
Figure 4. (A) Significant (grade ≥ 3) toxicity after selective internal radiotherapy reduced survival, and (B) moderate changes in liver function (CTC II) impaired overall survival (OS).
Pharmaceuticals 17 00101 g004
Table 1. Toxicity after SIRT according to the CTCAE v.5.0.
Table 1. Toxicity after SIRT according to the CTCAE v.5.0.
Toxicity
(Grade)
Post-SIRT Altered LFT, n (%)SIRT-Induced Toxicity, n (%)
IIIIII–IVIIIIII–IV
Bilirubin9 (19)14 (30)4 (9)4 (9)12 (26)4 (9)
Albumin7 (15)6 (13)0 (0)4 (9)5 (11)0 (0)
AST/ALT33 (70)4 (9)0 (0)12 (26)2 (4)0 (0)
AST30 (70)4 (9)0 (0)10 (21)2 (4)0 (0)
ALT24 (51)0 (0)0 (0)12 (26)0 (0)0 (0)
INR13 (28)3 (6)0 (0)9 (19)3 (6)0 (0)
Creatinine0 (0)0 (0)3 (6)0 (0)0 (0)2 (4)
Ascites8 (17)3 (6)0 (0)3 (6)3 (6)0 (0)
Toxicity of any kind22 (47)14 (30)7 (15)21 (45)10 (21)5 (11)
SIRT: selective internal radiation therapy; LFT: liver function test; AST: aspartate aminotransferase; ALT: alanine transaminase; INR: International Normalized Ratio.
Table 2. Contributing factors to toxicity after SIRT.
Table 2. Contributing factors to toxicity after SIRT.
Statistical Analysis
Univariate
p Value
Multivariate
p Value (95% CI)
Age≤65 years0.402
>65 years
Tumor load≤25%0.0410.0290.023–0.398
>25%
Cumulative activity<3.5 GBq0.706
≥3.5 GBq
Tumor spreadUnilobar0.227
Bilobar
HepatitisNo0.256
Yes
BCLC stagingB0.307
C
Child classificationA0.559
B
Lymph node involvementNo0.224
Yes
HepatitisNo0.256
Yes
PVTNo0.0740.0290.024–0.420
Yes
Altered LFTGrade 0–I0.283
Grade II–IV
Pre-treatmentRFA0.364
Resection/LT
Embolization/PEI
BCLC: Barcelona Clinic Liver Cancer; Child classification: Child–Pugh system; PVT: portal vein thrombosis; LFT: liver function test; RFA: radiofrequency ablation; LT: liver transplantation; PEI: percutaneous ethanol ablation.
Table 3. Contributing factors to survival after SIRT.
Table 3. Contributing factors to survival after SIRT.
Survival AnalysisStatistical Analysis
Median OSHR
(95% CI)
Univariate
p Value
Multivariate
HR (95% CI),
p Value
Age≤65 years1411–170.685
>65 years145–23
Tumor load≤25%1410–180.518
>25%1411–17
Cumulative activity<3.5 GBq148–200.323
≥3.5 GBq1413–15
Tumor spreadUnilobar146–220.620
Bilobar1412–16
HepatitisNo1510–200.247
Yes113–19
BCLC stagingB1311–150.389
C83–14
Child classificationA1210–150.736
B106–14
Lymph node involvementNo119–130.686
Yes110–39
HepatitisNo129–150.288
Yes104–16
PVTNo1311–150.352
Yes73–11
Altered LFTGrade 0–I1210–140.0025.50.009
Grade II–IV65–8(1.5–19.9)
Toxicity after SIRTGrade 0–II1412–17<0.001
Grade III–IV65–7
OS: overall survival; HR: hazard ratio; CI: confidence interval; BCLC: Barcelona Clinic Liver Cancer; Child classification: Child–Pugh system; PVT: portal vein thrombosis; LFT: liver function test; SIRT: selective internal radiation therapy.
Table 4. Baseline patient characteristics.
Table 4. Baseline patient characteristics.
All Patients (n = 47)
Age
 >65 years32 (68)
 ≤65 years15 (32)
Hepatic tumor load
 >25%23 (49)
 ≤25%24 (51)
Cumulative applied activity during SIRT session(s)
 ≥3.5 GBq21 (45)
 <3.5 GBq26 (55)
Hepatic tumor spread
 Bilobar24 (51)
 Unilobar23 (49)
BCLC staging
 Stage C22 (47)
 Stage B25 (53)
Child classification
 Child B21 (45)
 Child A26 (55)
Extrahepatic lymph node metastasis
 No39 (83)
 Yes8 (17)
Hepatitis
 Yes21 (45)
 No26 (55)
Etiology of hepatitis
 Alcohol-related6 (13)
 NASH12 (26)
 Viral18 (38)
 Cryptogenic 11 (23)
PVT
 Yes16 (33)
 No31 (66)
Pre-treatment
 RFA9 (19)
 Embolization/PEI2 (4)
 Resection/LT11 (24)
LFT in all patients
 Total bilirubin (mg/dL, normal range: 0.3–1.0)1.0 ± 0.5
 Albumin (g/dL, normal range: 3.4–5.4)3.9 ± 0.5
 AST (U/L, normal range: 5–40)77.6 ± 62.4
 ALT (U/L, normal range: 7–56)59.5 ± 41.1
 INR (normal range: 0.8–1.1)1.1 ± 0.1
Altered LFT (CTC I)
 Total bilirubin (>ULN–1.5 × ULN, mg/dL)16 (34)
 Albumin (<LLN–3 g/dL)6 (13)
 AST/ALT (>ULN–3 × ULN, U/L)26 (55)
 INR (>1.2–1.5 × baseline)4 (9)
 Ascites5 (11)
Altered LFT (CTC II)
 Total bilirubin (>1.5–3.0 × ULN, mg/dL)2 (4)
 Albumin (3–2 g/dL)2 (4)
 AST/ALT (>3–5 × ULN, U/L)2 (4)
 INR (>1.5–2.5)0 (0)
 Ascites0 (0)
Data presented as n (%). SIRT: selective internal radiotherapy; GBq: gigabecquerel; BCLC: Barcelona Clinic Liver Cancer; Child classification: Child–Pugh system; NASH: non-alcoholic steatohepatitis; PVT: portal vein thrombosis; RFA: radiofrequency ablation; PEI: percutaneous ethanol ablation; LT: liver transplantation; LFT: liver function test; CTC: Common Terminology for Common Adverse Events (v.5.0); ULN: upper limit of normal; LLN: lower limit of normal; AST: aspartate aminotransferase; ALT: alanine transaminase; INR: International Normalized Ratio.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bellendorf, A.; Mader, N.; Mueller, S.P.; Ezziddin, S.; Bockisch, A.; Grafe, H.; Best, J.; Goebel, J.; Pöppel, T.D.; Sabet, A. Safety and Efficacy of Selective Internal Radionuclide Therapy with 90Y Glass Microspheres in Patients with Progressive Hepatocellular Carcinoma after the Failure of Repeated Transarterial Chemoembolization. Pharmaceuticals 2024, 17, 101. https://doi.org/10.3390/ph17010101

AMA Style

Bellendorf A, Mader N, Mueller SP, Ezziddin S, Bockisch A, Grafe H, Best J, Goebel J, Pöppel TD, Sabet A. Safety and Efficacy of Selective Internal Radionuclide Therapy with 90Y Glass Microspheres in Patients with Progressive Hepatocellular Carcinoma after the Failure of Repeated Transarterial Chemoembolization. Pharmaceuticals. 2024; 17(1):101. https://doi.org/10.3390/ph17010101

Chicago/Turabian Style

Bellendorf, Alexander, Nicolai Mader, Stefan P. Mueller, Samer Ezziddin, Andreas Bockisch, Hong Grafe, Jan Best, Juliane Goebel, Thorsten D. Pöppel, and Amir Sabet. 2024. "Safety and Efficacy of Selective Internal Radionuclide Therapy with 90Y Glass Microspheres in Patients with Progressive Hepatocellular Carcinoma after the Failure of Repeated Transarterial Chemoembolization" Pharmaceuticals 17, no. 1: 101. https://doi.org/10.3390/ph17010101

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop