Next Article in Journal
The Impact of COVID-19 on Academic Cancer Clinical Trials in Canada and the Initial Response from Cancer Centers
Next Article in Special Issue
Intensive Follow-Up Program and Oncological Outcomes of Biliary Tract Cancer Patients after Curative-Intent Surgery: A Twenty-Year Experience in a Single Tertiary Medical Center
Previous Article in Journal
Operationalizing Leadership and Clinician Buy-In to Implement Evidence-Based Tobacco Treatment Programs in Routine Oncology Care: A Mixed-Method Study of the U.S. Cancer Center Cessation Initiative
Previous Article in Special Issue
Endobiliary Radiofrequency Ablation Combined with Gemcitabine and Cisplatin in Patients with Unresectable Extrahepatic Cholangiocarcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Optimization of the Clinical Effectiveness of Radioembolization in Hepatocellular Carcinoma with Dosimetry and Patient-Selection Criteria

1
Department of Nuclear Medicine, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, 1200 Brussels, Belgium
2
Department of Gastroenterology, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, 1200 Brussels, Belgium
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2022, 29(4), 2422-2434; https://doi.org/10.3390/curroncol29040196
Submission received: 10 March 2022 / Revised: 26 March 2022 / Accepted: 28 March 2022 / Published: 29 March 2022
(This article belongs to the Special Issue Hepatobiliary Malignancies: Recent Advancements and Future Directions)

Abstract

:
Selective internal radiation therapy (SIRT) is part of the treatment strategy for hepatocellular carcinoma (HCC). Strong clinical data demonstrated the effectiveness of this therapy in HCC with a significant improvement in patient outcomes. Recent studies demonstrated a strong correlation between the tumor response and the patient outcome when the tumor-absorbed dose was assessed by nuclear medicine imaging. Dosimetry plays a key role in predicting the clinical response and can be optimized using a personalized method of activity planning (multi-compartmental dosimetry). This paper reviews the main clinical results of SIRT in HCC and emphasizes the central role of dosimetry for improving it effectiveness. Moreover, some patient and tumor characteristics predict a worse outcome, and toxicity related to SIRT treatment of advanced HCC patient selection based on the performance status, liver function, tumor characteristics, and tumor targeting using technetium-99m macro-aggregated albumin scintigraphy can significantly improve the clinical performance of SIRT.

1. Introduction

Liver radioembolization (RE) or selective internal radiation therapy (SIRT) is part of the treatment strategy for hepatocellular carcinoma (HCC) [1]. This treatment involves the injection of radioactive microspheres via the liver arterial blood supply of the tumor(s). These microspheres are trapped in the arterioles of the tumor(s) and the targeted liver parenchyma. The liver parenchyma is primarily supplied by the portal vein, while HCC perfusion is primarily supplied by the hepatic arteries. This preferential vascularization allows a high irradiation of tumors while limiting radiation of the healthy liver [2]. The tumor-absorbed dose can range from 100 to 1000 Gy [3]. In comparison, the dose that can be delivered to tumors is limited to a maximum of 70 Gy, with external beam radiotherapy to avoid irreversible liver damage [4]. Yttrium-90 (90Y)-resin microspheres (Sir-Spheres®; Sirtex Medical Ltd., Sydney, Australia), 90Y-glass microspheres (Therasphere®; Boston Scientific, Boston, MA, USA), and holmium-166-poly-L-lactic acid microspheres (QuiremSpheres®; Quirem Medical B.V., Deventer, The Netherlands) are the three commercially available radioactive microspheres, differing by their physical and irradiation properties [5].
SIRT is planned in two phases. First, a simulation is always performed to evaluate the feasibility of the treatment. An interventional radiologist catheterizes the liver artery(ies) and evaluates the arterial feeding of the tumor(s). A non-therapeutic nuclear medicine agent, technetium-99m macro-aggregated albumin (MAA), is injected into the liver artery(ies) supplying the tumor(s) for simulating the distribution of the radioactive microspheres. Thereafter, the MAA distribution is assessed by nuclear imaging using single-photon emission computed tomography combined with computed tomography (MAA SPECT/CT). This imaging confirms the accurate targeting of the tumor(s) and the absence of risk of toxicity (digestive or lung irradiation). Then, the phase of treatment is scheduled with injection of the radioactive microspheres in the same technical conditions. The recommended methods for calculating the amount of radioactive microspheres needed for the treatment (activity) differ between the different available microspheres [6,7,8]. These methods are semi-empirical, based on the body surface area for resin microspheres, and using a mono-compartmental model (based on the liver volume) for glass and holmium-166-poly-L-lactic acid microspheres [9]. During the workup, the MAA distribution in the tumor, the healthy liver, and the lung compartments can also be evaluated to perform a more personalized method of activity planning (multi-compartmental or partition model) [1].
After therapy, the tumor and the healthy liver absorbed doses are determined with nuclear medicine imaging. With 90Y microspheres, the absorbed doses are ideally evaluated using positron emission tomography combined with computed tomography (90Y PET/CT). 90Y PET/CT accurately predicts the absorbed doses [10].

2. Clinical Results of SIRT in HCC

The treatment options for HCC depend on the Barcelona Clinic Liver Cancer (BCLC) staging system [11]. This classification takes into account the tumor characteristics (i.e., size, number of tumors, portal vein invasion, or extra hepatic spread), underlying liver function (via Child–Pugh score) and patient performance status (via Eastern Cooperative Oncology Group (ECOG) scale) [12]. The BCLC stage is a well-established accurate predictor of patient survival and in routine clinical use worldwide to help determine the best treatment options.
Recent recommendations of the European Society for Medical Oncology consider SIRT as an alternative treatment for patients with BCLC stages A, B, and C [13,14]. For BCLC stage A patients, a recent large, retrospective study demonstrated that SIRT was very efficient to address unresectable solitary HCC alone or for use as a neoadjuvant bridge in a curative surgical approach [15]. For intermediate HCC (i.e., BCLC B), transarterial chemoembolization is recommended for first-line therapy. However, a meta-analysis of previous prospective randomized studies comparing SIRT to transarterial chemoembolization demonstrated similar survival outcomes [16]. Moreover, a randomized study comparing SIRT to transarterial chemoembolization in a population of BCLC A and B patients demonstrated similar survival times but showed that the former was associated with a longer time to progression [17].
Considering advanced stage patients (i.e., BCLC C), systemic therapies are often preferred; these include immunotherapy (e.g., atezolizumab plus bevacizumab) or targeted therapy (e.g., sorafenib, regorafenib). Patients treated with atezolizumab plus bevacizumab demonstrated superior survival and progression-free survival compared to patients treated with sorafenib [18]. However, randomized controlled trials comparing SIRT to sorafenib have failed to demonstrate a superior outcome with SIRT [19,20,21]. Consequently, the place of SIRT in advanced HCC is an alternative and possibilities for therapy optimization should be investigated.
The main results of prospective and randomized studies published to date that have compared SIRT to alternative therapies in HCC patients are summarized in Table 1.
Controlled trials currently investigate the combination of SIRT plus immunotherapy in patients with intermediate and advanced stages of HCC. Preliminary results of the combination of nivolumab three weeks after SIRT demonstrated a favorable tolerability and encouraging response rates [24,25]. A randomized trial (NCT04541173) is also investigating the safety and effectiveness of SIRT followed by the combination of atezolizumab plus bevacizumab. In theory, the combination of immunotherapy after SIRT may give a synergistic clinical effect and improve tumor control and patient survival. Ionizing radiation may induce the release of tumor-associated antigens targeted by antigen presenting cells and result in a stimulation of the immune response, boosting the effects of immunotherapy [26]. SIRT must be performed before the initiation of immunotherapy, when the biological effects of ionizing radiations are effective.

3. Clinical Dosimetry in SIRT

Tumor dosimetry is a predictive factor of SIRT efficiency. Previous data have demonstrated a correlation between tumor-absorbed dose and radiological response [27,28,29]; indeed, a high tumor-absorbed dose is associated with a high probability of tumor control. In addition, a multitude of clinical data demonstrating strong correlation between tumor-absorbed dose, radiological response, and survival of HCC patients are currently available (overview in Table 2). Table 3 and Table 4 summarize the main studies reporting a tumor-absorbed dose threshold associated with SIRT efficiency in HCC. Studies comparing glass to resin microspheres have indicated that the tumor-absorbed dose cut-off is usually two-fold, which is explained by their different physical and radioactive properties [5,30].

4. Personalized Dosimetry in SIRT

To improve the clinical results of RE, the activity prescription can be more personalized and optimized to reach higher tumor-absorbed doses. As previously described, the recommended activity prescription is calculated using semi-empirical methods. While these methods are safe, they can induce suboptimal absorbed doses to tumors [41]. A recent prospective study confirmed the clinical benefits of performing multi-compartmental dosimetry (known as “the partition model”) [40]. In the partition model, activity planning is based upon the MAA distribution in the different compartments (Figure 1), simulating an absorbed dose under the threshold of toxicity for the healthy liver and above the efficacy threshold for the tumor(s).
The dose to the healthy liver can be accurately predicted with MAA SPECT/CT, controlling the risk of liver toxicity [42]. Indeed, an excess of liver radiation can induce liver damage (i.e., RE-induced liver disease). The toxicity threshold doses have been well-demonstrated through non-tumoral, whole-liver dose (reaching 90 Gy for glass microspheres and 40–50 Gy for resin microspheres) [43,44]. As such, with MAA SPECT/CT dosimetry simulating an absorbed dose to the healthy liver under these thresholds, the activity can be planned safely. Moreover, the external beam radiotherapy models have shown that no liver damage can occur if the treated liver volume does not exceed 40% [45]. When a small part of the liver volume is targeted by the treatment, the planned activity can be increased for performing a safe radiation segmentectomy. For treatments applied to a majority of the liver (>60%), the planned activity can be adjusted to reach the maximal tolerable liver absorbed dose. With this method, the planned activity would be the highest possible and would therefore increase the activity in the tumor compartment to maximize the tumor-control probability.
Moreover, a large HCC tumor size (≥5 cm) was a factor of poor prognosis in some studies [46,47,48]. These studies included patients treated by glass microspheres, using the recommended method of activity planning (80–150 Gy to the targeted liver). Given this, Garin et al. [33] demonstrated a significant lower response rate in large HCC tumors (size ≥ 5 cm) using this same method of activity planning, probably because of tumor underdosing. More interestingly, using an optimized method of activity planning increasing the tumor-absorbed dose, Garin et al. [49] demonstrated a high response rate in large HCC tumors and no correlation between the tumor size (≥5 cm) and the patient survival.
A recent prospective trial performed with patients with HCC, mostly with advanced stage disease, demonstrated better outcome achieved with personalized dosimetry and MAA imaging (using glass microspheres) [40]. When an approach reaching a maximum dose of 120 Gy to the targeted healthy liver, and at least 205 Gy to tumors (>250 Gy if possible) was used, the clinical outcome was highly improved as compared to patients treated with the standard (120 Gy to the targeted liver) dosimetric approach. The main results of this trial are summarized in Table 5. The median activity was increased by 38%, as shown upon comparison of the standard method to this personalized method of activity planning. Similarly, in a retrospective study using personalized dosimetry with a whole, normal liver dose reaching 40 to 70 Gy (glass microspheres), the median survival was 14.1 mo in HCC patients with portal vein invasion (95% confidence interval (CI): 10.7–17.5 mo) [50]. These results were higher than expected, considering other published data from a similar population treated with a standard dosimetric approach (median: 10.4 mo, 95%CI: 7.2–16.6) [48].
However, using this optimized method of activity planning, patients with risk factors for RE-induced liver disease must be carefully evaluated before treatment to limit the liver toxicity probability. For this purpose, 99mTc–mebrofenin scintigraphy with SPECT/CT can evaluate and quantify the global and regional liver functions and predict the risk of post-radiation liver damage. In patients who undergo major liver resection, the remnant liver uptake of mebrofenin correlated well with the risk of postoperative liver failure (cut-off value: 2.69%/min/m2) [51]. This technique could also be applied to SIRT for evaluating liver function in patients with risk factors (e.g., advanced cirrhosis, large tumor involvement, etc.). Indeed, the mebrofenin liver uptake of the non-treated liver was also predictive of RE-induced liver disease in some case series [52,53].

5. Optimization of Tumor Targeting

Better tumor targeting is highly valuable because it will improve the tumor-absorbed dose and effectiveness of the treatment. New microcatheters used in interventional radiology allow for more selective angiography, delivering higher activities in the vicinity of tumors and sparing the healthy liver. Interventional radiologists are able to perform this kind of selective approach more and more, splitting the activity among multiple injections for the different arterial branches of the tumor [54]. For this purpose, a cone-beam CT can be performed during the liver arteriography for precisely identifying the feeding arteries of a tumor [55].
Moreover, the innovative new anti-reflux catheters could also improve tumor targeting. In a retrospective analysis of neuroendocrine and HCC tumors, the anti-reflux catheters were found to provide significantly better tumor targeting than the classic end-hole catheters [56]. Some drugs infused during the treatment can also increase the tumor-to-normal-liver ratio (i.e., the tumor targeting). The co-infusion of angiotensin II during SIRT was also shown to significantly increase tumor targeting (tumor-to-normal-liver ratio × 3) by decreasing the healthy liver arterial flow, while the tumor arterial flow increased [57]. However, this effect was short-lived (a few minutes) and rapidly reversed despite the continuous infusion of angiotensin II due to liver arterial vasodilation triggered by the low arterial flow (i.e., a vascular escape mechanism) [58].
For clinical application of SIRT, the arterial vasoconstriction needs to be longer to facilitate injection of all radioactive microspheres before activation of this opposing effect. Alternative drugs, such as sodium acetate and dopexamine, could induce a longer vasoconstrictive effect in the liver artery [59]. These mesenteric vasodilators induce an increase in portal blood flow, resulting in a reflex vasoconstriction of the liver artery (i.e., the hepatic arterial buffer response) [60], an effect to which tumors are not susceptible due to their anarchic vascularization. Hence, the arterial flow would be redirected in tumors preferentially, and the tumor-to-normal-liver ratio would be increased. For this purpose, dopexamine seems to be a good candidate. This analogue of dopamine is responsible for vasodilation of the mesenteric arteries, inducing a reduction in the liver arterial flow to a factor of four in an animal model [61]. Moreover, this drug has a short half-life and is well-tolerated at low infusion rates [62]. Future investigations are needed to evaluate this effect more thoroughly.

6. Good HCC Candidates for SIRT

The collective research efforts have provided a good understanding of the factors responsible for treatment ineffectiveness in HCC, helping clinicians to select the best candidates for SIRT. Currently, using tumor dosimetry, MAA imaging can generally select patients who will respond well to SIRT (high tumor uptake and high absorbed dose) or those who will not respond (low tumor uptake, low absorbed dose) [63]. The interest of this dosimetry applied to MAA SPECT/CT was confirmed in the recent DOSISPHERE randomized controlled trial [40] and was also well-illustrated in a retrospective study of 41 patients treated for advanced HCC with portal vein thrombosis. The overall survival was only 4.3 mo when the tumor-absorbed dose was less than 205 Gy and 18.2 mo when at least 205 Gy (glass microspheres) [49]. Moreover, patients with portal vein thrombosis and poor targeting via MAA imaging had a very poor prognosis.
HCC is a heterogeneous group of tumors with different behaviors; some can be very aggressive, with a tumor doubling time ranging from 3 mo to 1 year [64]. [18F]-Fluorodeoxyglucose (FDG) PET/CT has low sensibility, with a significant uptake in less than 50–65% of the cases [65]. However, data have indicated that HCC tumors with high [18F]FDG uptake are more aggressive, with patients at higher risk of recurrence and poorer survival [66]. SIRT is less effective in this population, with a significant reduction of the local control, progression-free survival (PFS) and overall survival (OS) [67,68]. In advanced HCC, randomized trials have failed to demonstrate a superior PFS and OS in patients treated by RE compared to sorafenib despite a significant increase of the tumor response rate in the RE arm (Table 1). Loco-regional therapies such as SIRT may be less effective for patients with aggressive HCC tumors, and [18F]FDG PET/CT could be useful to identify these patients. Decompensated liver function is also a strong predictor of poor survival. The baseline bilirubin level, the Child–Pugh score, and the albumin–bilirubin grade were independent predictors of poor survival in patients treated with SIRT [50,69,70]. The median overall survival rates reported for advanced HCC patients treated with sorafenib range from 6.5 mo to 14.7 mo [71,72,73,74]. To compare, some markers of poor prognosis have been identified in large retrospective studies of advanced HCC patients treated with SIRT (Table 6). Patients with poor performance status (ECOG 2 or more), extrahepatic metastases, portal vein thrombosis extending to the main left/right branch, tumor burden > 50% of the liver volume, and a baseline alteration of the liver function (albumin–bilirubin score of 3 or bilirubin level of 2–3 mg/dL) have reported median survival rates that fall between 4.3 and 8.2 mo (Table 6). Lescure et al. demonstrated also a strong correlation between the ALBI score (grade 2 or 3) and the risk of REILD [75].
In these groups of patients, RE would be ineffective and potentially toxic; alternative systemic therapies should be suggested.

7. Conclusions

SIRT is an effective therapy in HCC and can significantly improve the outcome of patients. Dosimetry plays a key role in predicting its effectiveness and can be optimized using a personalized method of activity planning (i.e., multi-compartmental dosimetry). Selection of patients based on performance status, liver function, tumor characteristics, and tumor targeting as assessed by MAA imaging can also improve the clinical performance of SIRT.

Author Contributions

Conceptualization, P.d. and F.J.; methodology, P.d., S.W. and F.J.; software, P.d.; validation, all authors; formal analysis, P.d., S.W. and F.J.; investigation, P.d., S.W., I.B. and F.J.; resources, R.L. and F.J.; data curation, P.d., F.J., S.W., M.H. and I.B.; writing—original draft preparation, P.d., S.W. and F.J.; writing—review and editing, all authors; visualization, all authors; supervision, P.d. and F.J., project administration, P.d. and F.J., funding acquisition, F.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Padia, S.A.; Lewandowski, R.J.; Johnson, G.E.; Sze, D.Y.; Ward, T.J.; Gaba, R.C.; Baerlocher, M.O.; Gates, V.L.; Riaz, A.; Brown, D.B.; et al. Radioembolization of Hepatic Malignancies: Background, Quality Improvement Guidelines, and Future Directions. J. Vasc. Interv. Radiol. 2017, 28, 1–15. [Google Scholar] [CrossRef] [PubMed]
  2. Lewandowski, R.J.; Salem, R. Yttrium-90 radioembolization of hepatocellular carcinoma and metastatic disease to the liver. Semin. Interv. Radiol. 2006, 23, 64–72. [Google Scholar] [CrossRef] [Green Version]
  3. Kennedy, A.S.; Nutting, C.; Coldwell, D.; Gaiser, J.; Drachenberg, C. Pathologic response and microdosimetry of (90)Y microspheres in man: Review of four explanted whole livers. Int. J. Radiat. Oncol. Biol. Phys. 2004, 60, 1552–1563. [Google Scholar] [CrossRef]
  4. Kennedy, A. Radioembolization of hepatic tumors. J. Gastrointest. Oncol. 2014, 5, 178–189. [Google Scholar] [CrossRef] [PubMed]
  5. D’Abadie, P.; Hesse, M.; Louppe, A.; Lhommel, R.; Walrand, S.; Jamar, F. Microspheres Used in Liver Radioembolization: From Conception to Clinical Effects. Molecules 2021, 26, 3966. [Google Scholar] [CrossRef] [PubMed]
  6. SIR-Spheres. Instructions for Use; SIRTeX Medical Limited: North Sydney, Australia, 2019; Available online: https://www.sirtex.com/us/clinicians/instructions-for-use/ (accessed on 9 March 2022).
  7. Therasphere. Instructionsforuse. 2022. Available online: https://www.bostonscientific.com/en-EU/products/selective-internal-radiation-therapy/therasphere-y90-glass-microspheres.html (accessed on 9 March 2022).
  8. QuiremSpheres. Instruction for Use. 2022. Available online: https://www.quirem.com/wp-content/uploads/2022/01/LC-8004307-IFU-QuiremSpheres-Multi-Language.pdf (accessed on 9 March 2022).
  9. Bastiaannet, R.; Kappadath, S.C.; Kunnen, B.; Braat, A.; Lam, M.; de Jong, H. The physics of radioembolization. EJNMMI Phys. 2018, 5, 22. [Google Scholar] [CrossRef]
  10. Hesse, M.; d’Abadie, P.; Lhommel, R.; Jamar, F.; Walrand, S. Yttrium-90 TOF-PET-Based EUD Predicts Response Post Liver Radioembolizations Using Recommended Manufacturer FDG Reconstruction Parameters. Front. Oncol. 2021, 11, 592529. [Google Scholar] [CrossRef] [PubMed]
  11. Reig, M.; Forner, A.; Rimola, J.; Ferrer-Fabrega, J.; Burrel, M.; Garcia-Criado, A.; Kelley, R.K.; Galle, P.R.; Mazzaferro, V.; Salem, R.; et al. BCLC strategy for prognosis prediction and treatment recommendation Barcelona Clinic Liver Cancer (BCLC) staging system. The 2022 update. J. Hepatol. 2021, 76, 681–693. [Google Scholar] [CrossRef]
  12. Couri, T.; Pillai, A. Goals and targets for personalized therapy for HCC. Hepatol. Int. 2019, 13, 125–137. [Google Scholar] [CrossRef]
  13. Vogel, A.; Martinelli, E.; on behalf of the ESMO Guidelines Committee. Updated treatment recommendations for hepatocellular carcinoma (HCC) from the ESMO Clinical Practice Guidelines. Ann. Oncol. 2021, 32, 801–805. [Google Scholar] [CrossRef]
  14. Lee, J.J.X.; Tai, D.W.; Choo, S.P. Locoregional therapy in hepatocellular carcinoma: When to start and when to stop and when to revisit. ESMO Open 2021, 6, 100129. [Google Scholar] [CrossRef] [PubMed]
  15. Salem, R.; Johnson, G.E.; Kim, E.; Riaz, A.; Bishay, V.; Boucher, E.; Fowers, K.; Lewandowski, R.; Padia, S.A. Yttrium-90 Radioembolization for the Treatment of Solitary, Unresectable HCC: The LEGACY Study. Hepatology 2021, 74, 2342–2352. [Google Scholar] [CrossRef] [PubMed]
  16. Casadei Gardini, A.; Tamburini, E.; Inarrairaegui, M.; Frassineti, G.L.; Sangro, B. Radioembolization versus chemoembolization for unresectable hepatocellular carcinoma: A meta-analysis of randomized trials. Onco Targets Ther. 2018, 11, 7315–7321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Salem, R.; Gordon, A.C.; Mouli, S.; Hickey, R.; Kallini, J.; Gabr, A.; Mulcahy, M.F.; Baker, T.; Abecassis, M.; Miller, F.H.; et al. Y90 Radioembolization Significantly Prolongs Time to Progression Compared With Chemoembolization in Patients With Hepatocellular Carcinoma. Gastroenterology 2016, 151, 1155–1163 e1152. [Google Scholar] [CrossRef] [Green Version]
  18. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O.; et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [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. Venerito, M.; Pech, M.; Canbay, A.; Donghia, R.; Guerra, V.; Chatellier, G.; Pereira, H.; Gandhi, M.; Malfertheiner, P.; Chow, P.K.H.; et al. NEMESIS: Noninferiority, Individual-Patient Metaanalysis of Selective Internal Radiation Therapy with (90)Y Resin Microspheres Versus Sorafenib in Advanced Hepatocellular Carcinoma. J. Nucl. Med. 2020, 61, 1736–1742. [Google Scholar] [CrossRef] [PubMed]
  22. Pitton, M.B.; Kloeckner, R.; Ruckes, C.; Wirth, G.M.; Eichhorn, W.; Worns, M.A.; Weinmann, A.; Schreckenberger, M.; Galle, P.R.; Otto, G.; et al. Randomized comparison of selective internal radiotherapy (SIRT) versus drug-eluting bead transarterial chemoembolization (DEB-TACE) for the treatment of hepatocellular carcinoma. Cardiovasc. Interv. Radiol. 2015, 38, 352–360. [Google Scholar] [CrossRef] [Green Version]
  23. Ricke, J.; Klumpen, H.J.; Amthauer, H.; Bargellini, I.; Bartenstein, P.; de Toni, E.N.; Gasbarrini, A.; Pech, M.; Peck-Radosavljevic, M.; Popovic, P.; et al. Impact of combined selective internal radiation therapy and sorafenib on survival in advanced hepatocellular carcinoma. J. Hepatol. 2019, 71, 1164–1174. [Google Scholar] [CrossRef]
  24. De La Torre-Alaez, M.; Matilla, A.; Varela, M.; Inarrairaegui, M.; Reig, M.; Lledo, J.; Arenas, J.; Lorente, S.; Testillano, M.; Gomez-Martin, C.; et al. Nivolumab after selective internal radiation therapy using sir spheres resin microspheres in patients with hepatcocellular carcinoma: The NASIR HCC trial. In Proceedings of the International Liver Cancer Association 2020 Virtual Conference, Oral Communication, Virtual Conference, 11–13 September 2020. [Google Scholar]
  25. Tai, D.; Loke, K.; Gogna, A.; Kaya, N.; Tan, S.; Hennedige, T.; Ng, D.; Irani, F.; Lee, J.; Lim, J.; et al. Radioembolization with Y90-resin micropsheres followed by nivolumab for advanced hepatocellular carcinoma (CA 209-678): A single arm, single centre, phase 2 trial. Lancet Gastroenterol. Hepatol. 2021, 6, 1025–1035. [Google Scholar] [CrossRef]
  26. Lee, Y.H.; Tai, D.; Yip, C.; Choo, S.P.; Chew, V. Combinational Immunotherapy for Hepatocellular Carcinoma: Radiotherapy, Immune Checkpoint Blockade and Beyond. Front. Immunol. 2020, 11, 568759. [Google Scholar] [CrossRef] [PubMed]
  27. Strigari, L.; Sciuto, R.; Rea, S.; Carpanese, L.; Pizzi, G.; Soriani, A.; Iaccarino, G.; Benassi, M.; Ettorre, G.M.; Maini, C.L. Efficacy and toxicity related to treatment of hepatocellular carcinoma with 90Y-SIR spheres: Radiobiologic considerations. J. Nucl. Med. 2010, 51, 1377–1385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Hermann, A.L.; Dieudonne, A.; Ronot, M.; Sanchez, M.; Pereira, H.; Chatellier, G.; Garin, E.; Castera, L.; Lebtahi, R.; Vilgrain, V.; et al. Relationship of Tumor Radiation-absorbed Dose to Survival and Response in Hepatocellular Carcinoma Treated with Transarterial Radioembolization with (90)Y in the SARAH Study. Radiology 2020, 296, 673–684. [Google Scholar] [CrossRef] [PubMed]
  29. Dewaraja, Y.K.; Devasia, T.; Kaza, R.K.; Mikell, J.K.; Owen, D.; Roberson, P.L.; Schipper, M.J. Prediction of Tumor Control in (90)Y Radioembolization by Logit Models with PET/CT-Based Dose Metrics. J. Nucl. Med. 2020, 61, 104–111. [Google Scholar] [CrossRef]
  30. D’Abadie, P.; Hesse, M.; Jamar, F.; Lhommel, R.; Walrand, S. (90)Y TOF-PET based EUD reunifies patient survival prediction in resin and glass microspheres radioembolization of HCC tumours. Phys. Med. Biol. 2018, 63, 245010. [Google Scholar] [CrossRef]
  31. Chiesa, C.; Maccauro, M.; Romito, R.; Spreafico, C.; Pellizzari, S.; Negri, A.; Sposito, C.; Morosi, C.; Civelli, E.; Lanocita, R.; et al. Need, feasibility and convenience of dosimetric treatment planning in liver selective internal radiation therapy with (90)Y microspheres: The experience of the National Tumor Institute of Milan. Q. J. Nucl. Med. Mol. Imaging 2011, 55, 168–197. [Google Scholar]
  32. Garin, E.; Lenoir, L.; Rolland, Y.; Edeline, J.; Mesbah, H.; Laffont, S.; Poree, P.; Clement, B.; Raoul, J.L.; Boucher, E. Dosimetry based on 99mTc-macroaggregated albumin SPECT/CT accurately predicts tumor response and survival in hepatocellular carcinoma patients treated with 90Y-loaded glass microspheres: Preliminary results. J. Nucl. Med. 2012, 53, 255–263. [Google Scholar] [CrossRef] [Green Version]
  33. Garin, E.; Rolland, Y.; Pracht, M.; Le Sourd, S.; Laffont, S.; Mesbah, H.; Haumont, L.A.; Lenoir, L.; Rohou, T.; Brun, V.; et al. High impact of macroaggregated albumin-based tumour dose on response and overall survival in hepatocellular carcinoma patients treated with (90) Y-loaded glass microsphere radioembolization. Liver. Int. 2017, 37, 101–110. [Google Scholar] [CrossRef] [Green Version]
  34. Kappadath, S.C.; Mikell, J.; Balagopal, A.; Baladandayuthapani, V.; Kaseb, A.; Mahvash, A. Hepatocellular Carcinoma Tumor Dose Response After (90)Y-radioembolization With Glass Microspheres Using (90)Y-SPECT/CT-Based Voxel Dosimetry. Int. J. Radiat. Oncol. Biol. Phys. 2018, 102, 451–461. [Google Scholar] [CrossRef]
  35. Allimant, C.; Kafrouni, M.; Delicque, J.; Ilonca, D.; Cassinotto, C.; Assenat, E.; Ursic-Bedoya, J.; Pageaux, G.P.; Mariano-Goulart, D.; Aho, S.; et al. Tumor Targeting and Three-Dimensional Voxel-Based Dosimetry to Predict Tumor Response, Toxicity, and Survival after Yttrium-90 Resin Microsphere Radioembolization in Hepatocellular Carcinoma. J. Vasc. Interv. Radiol. 2018, 29, 1662–1670 e1664. [Google Scholar] [CrossRef] [PubMed]
  36. Chan, K.T.; Alessio, A.M.; Johnson, G.E.; Vaidya, S.; Kwan, S.W.; Monsky, W.; Wilson, A.E.; Lewis, D.H.; Padia, S.A. Prospective Trial Using Internal Pair-Production Positron Emission Tomography to Establish the Yttrium-90 Radioembolization Dose Required for Response of Hepatocellular Carcinoma. Int. J. Radiat. Oncol. Biol Phys. 2018, 101, 358–365. [Google Scholar] [CrossRef] [PubMed]
  37. D’Abadie, P.; Walrand, S.; Hesse, M.; Annet, L.; Borbath, I.; Van den Eynde, M.; Lhommel, R.; Jamar, F. Prediction of tumor response and patient outcome after radioembolization of hepatocellular carcinoma using 90Y-PET-computed tomography dosimetry. Nucl. Med. Commun. 2021, 42, 747–754. [Google Scholar] [CrossRef]
  38. Son, M.H.; Ha, L.N.; Bang, M.H.; Bae, S.; Giang, D.T.; Thinh, N.T.; Paeng, J.C. Diagnostic and prognostic value of (99m)Tc-MAA SPECT/CT for treatment planning of (90)Y-resin microsphere radioembolization for hepatocellular carcinoma: Comparison with planar image. Sci. Rep. 2021, 11, 3207. [Google Scholar] [CrossRef] [PubMed]
  39. Nodari, G.; Popoff, R.; Riedinger, J.M.; Lopez, O.; Pellegrinelli, J.; Dygai-Cochet, I.; Tabouret-Viaud, C.; Presles, B.; Chevallier, O.; Gehin, S.; et al. Impact of contouring methods on pre-treatment and post-treatment dosimetry for the prediction of tumor control and survival in HCC patients treated with selective internal radiation therapy. EJNMMI Res. 2021, 11, 24. [Google Scholar] [CrossRef] [PubMed]
  40. Garin, E.; Tselikas, L.; Guiu, B.; Chalaye, J.; Edeline, J.; de Baere, T.; Assenat, E.; Tacher, V.; Robert, C.; Terroir-Cassou-Mounat, M.; et al. Personalised versus standard dosimetry approach of selective internal radiation therapy in patients with locally advanced hepatocellular carcinoma (DOSISPHERE-01): A randomised, multicentre, open-label phase 2 trial. Lancet Gastroenterol. Hepatol. 2021, 6, 17–29. [Google Scholar] [CrossRef]
  41. Kafrouni, M.; Allimant, C.; Fourcade, M.; Vauclin, S.; Delicque, J.; Ilonca, A.D.; Guiu, B.; Manna, F.; Molinari, N.; Mariano-Goulart, D.; et al. Retrospective Voxel-Based Dosimetry for Assessing the Ability of the Body-Surface-Area Model to Predict Delivered Dose and Radioembolization Outcome. J. Nucl. Med. 2018, 59, 1289–1295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. D’Abadie, P.; Walrand, S.; Hesse, M.; Amini, N.; Lhommel, R.; Sawadogo, K.; Jamar, F. Accurate non-tumoral 99mTc-MAA absorbed dose prediction to plan optimized activities in liver radioembolization using resin microspheres. Phys. Med. 2021, 89, 250–257. [Google Scholar] [CrossRef]
  43. Garin, E.; Palard, X.; Rolland, Y. Personalised Dosimetry in Radioembolisation for HCC: Impact on Clinical Outcome and on Trial Design. Cancers 2020, 12, 1557. [Google Scholar] [CrossRef]
  44. Chiesa, C.; Mira, M.; Bhoori, S.; Bormolini, G.; Maccauro, M.; Spreafico, C.; Cascella, T.; Cavallo, A.; De Nile, M.C.; Mazzaglia, S.; et al. Radioembolization of hepatocarcinoma with (90)Y glass microspheres: Treatment optimization using the dose-toxicity relationship. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 3018–3032. [Google Scholar] [CrossRef]
  45. Chiesa, C.; Sjogreen-Gleisner, K.; Walrand, S.; Strigari, L.; Flux, G.; Gear, J.; Stokke, C.; Gabina, P.M.; Bernhardt, P.; Konijnenberg, M. EANM dosimetry committee series on standard operational procedures: A unified methodology for (99m)Tc-MAA pre- and (90)Y peri-therapy dosimetry in liver radioembolization with (90)Y microspheres. EJNMMI Phys. 2021, 8, 77. [Google Scholar] [CrossRef] [PubMed]
  46. Abouchaleh, N.; Gabr, A.; Ali, R.; Al Asadi, A.; Mora, R.A.; Kallini, J.R.; Mouli, S.; Riaz, A.; Lewandowski, R.J.; Salem, R. (90)Y Radioembolization for Locally Advanced Hepatocellular Carcinoma with Portal Vein Thrombosis: Long-Term Outcomes in a 185-Patient Cohort. J. Nucl. Med. 2018, 59, 1042–1048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Zu, Q.; Schenning, R.C.; Jahangiri, Y.; Tomozawa, Y.; Kolbeck, K.J.; Kaufman, J.A.; Al-Hakim, R.; Naugler, W.E.; Nabavizadeh, N.; Kardosh, A.; et al. Yttrium-90 Radioembolization for BCLC Stage C Hepatocellular Carcinoma Comparing Child-Pugh A Versus B7 Patients: Are the Outcomes Equivalent? Cardiovasc. Intervent. Radiol. 2020, 43, 721–731. [Google Scholar] [CrossRef] [PubMed]
  48. 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]
  49. Garin, E.; Rolland, Y.; Edeline, J.; Icard, N.; Lenoir, L.; Laffont, S.; Mesbah, H.; Breton, M.; Sulpice, L.; Boudjema, K.; et al. Personalized dosimetry with intensification using 90Y-loaded glass microsphere radioembolization induces prolonged overall survival in hepatocellular carcinoma patients with portal vein thrombosis. J. Nucl. Med. 2015, 56, 339–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Spreafico, C.; Sposito, C.; Vaiani, M.; Cascella, T.; Bhoori, S.; Morosi, C.; Lanocita, R.; Romito, R.; Chiesa, C.; Maccauro, M.; et al. Development of a prognostic score to predict response to Yttrium-90 radioembolization for hepatocellular carcinoma with portal vein invasion. J. Hepatol. 2018, 68, 724–732. [Google Scholar] [CrossRef]
  51. De Graaf, W.; van Lienden, K.P.; Dinant, S.; Roelofs, J.J.; Busch, O.R.; Gouma, D.J.; Bennink, R.J.; van Gulik, T.M. Assessment of future remnant liver function using hepatobiliary scintigraphy in patients undergoing major liver resection. J. Gastrointest. Surg. 2010, 14, 369–378. [Google Scholar] [CrossRef] [Green Version]
  52. Braat, M.; de Jong, H.W.; Seinstra, B.A.; Scholten, M.V.; van den Bosch, M.; Lam, M. Hepatobiliary scintigraphy may improve radioembolization treatment planning in HCC patients. EJNMMI Res. 2017, 7, 2. [Google Scholar] [CrossRef] [Green Version]
  53. Bennink, R.J.; Cieslak, K.P.; van Delden, O.M.; van Lienden, K.P.; Klumpen, H.J.; Jansen, P.L.; van Gulik, T.M. Monitoring of Total and Regional Liver Function after SIRT. Front. Oncol. 2014, 4, 152. [Google Scholar] [CrossRef] [Green Version]
  54. Tong, A.K.; Kao, Y.H.; Too, C.W.; Chin, K.F.; Ng, D.C.; Chow, P.K. Yttrium-90 hepatic radioembolization: Clinical review and current techniques in interventional radiology and personalized dosimetry. Br. J. Radiol. 2016, 89, 20150943. [Google Scholar] [CrossRef]
  55. Louie, J.D.; Kothary, N.; Kuo, W.T.; Hwang, G.L.; Hofmann, L.V.; Goris, M.L.; Iagaru, A.H.; Sze, D.Y. Incorporating cone-beam CT into the treatment planning for yttrium-90 radioembolization. J. Vasc. Interv. Radiol. 2009, 20, 606–613. [Google Scholar] [CrossRef] [PubMed]
  56. D’Abadie, P.; Walrand, S.; Goffette, P.; Amini, N.; Maanen, A.V.; Lhommel, R.; Jamar, F. Antireflux catheter improves tumor targeting in liver radioembolization with resin microspheres. Diagn. Interv. Radiol. 2021, 27, 768–773. [Google Scholar] [CrossRef] [PubMed]
  57. Sasaki, Y.; Imaoka, S.; Hasegawa, Y.; Nakano, S.; Ishikawa, O.; Ohigashi, H.; Taniguchi, K.; Koyama, H.; Iwanaga, T.; Terasawa, T. Changes in distribution of hepatic blood flow induced by intra-arterial infusion of angiotensin II in human hepatic cancer. Cancer 1985, 55, 311–316. [Google Scholar] [CrossRef]
  58. Macedo, M.P.; Lautt, W.W. Shear-induced modulation of vasoconstriction in the hepatic artery and portal vein by nitric oxide. Am. J. Physiol. 1998, 274, G253–G260. [Google Scholar] [CrossRef]
  59. Walrand, S.; Hesse, M.; d’Abadie, P.; Jamar, F. Hepatic Arterial Buffer Response in Liver Radioembolization and Potential Use for Improved Cancer Therapy. Cancers 2021, 13, 1537. [Google Scholar] [CrossRef]
  60. Lautt, W.W. Hepatic Circulation: Physiology and Pathophysiology. In Colloquium Series on Integrated Systems Physiology: From Molecule to Function to Disease; Morgan & Claypool: San Rafael, CA, USA, 2009. [Google Scholar]
  61. Biro, G.P.; Douglas, J.R.; Keon, W.J.; Taichman, G.C. Changes in regional blood flow distribution induced by infusions of dopexamine hydrochloride or dobutamine in anesthetized dogs. Am. J. Cardiol. 1988, 62, 30C–36C. [Google Scholar] [CrossRef]
  62. Fitton, A.; Benfield, P. Dopexamine hydrochloride. A review of its pharmacodynamic and pharmacokinetic properties and therapeutic potential in acute cardiac insufficiency. Drugs 1990, 39, 308–330. [Google Scholar] [CrossRef]
  63. Levillain, H.; Bagni, O.; Deroose, C.M.; Dieudonne, A.; Gnesin, S.; Grosser, O.S.; Kappadath, S.C.; Kennedy, A.; Kokabi, N.; Liu, D.M.; et al. International recommendations for personalised selective internal radiation therapy of primary and metastatic liver diseases with yttrium-90 resin microspheres. Eur.J. Nucl. Med. Mol. Imaging 2021, 48, 1570–1584. [Google Scholar] [CrossRef] [PubMed]
  64. Rich, N.E.; John, B.V.; Parikh, N.D.; Rowe, I.; Mehta, N.; Khatri, G.; Thomas, S.M.; Anis, M.; Mendiratta-Lala, M.; Hernandez, C.; et al. Hepatocellular Carcinoma Demonstrates Heterogeneous Growth Patterns in a Multicenter Cohort of Patients with Cirrhosis. Hepatology 2020, 72, 1654–1665. [Google Scholar] [CrossRef]
  65. Sacks, A.; Peller, P.J.; Surasi, D.S.; Chatburn, L.; Mercier, G.; Subramaniam, R.M. Value of PET/CT in the management of primary hepatobiliary tumors, part 2. AJR Am. J. Roentgenol. 2011, 197, W260–W265. [Google Scholar] [CrossRef]
  66. Sun, D.W.; An, L.; Wei, F.; Mu, L.; Shi, X.J.; Wang, C.L.; Zhao, Z.W.; Li, T.F.; Lv, G.Y. Prognostic significance of parameters from pretreatment (18)F-FDG PET in hepatocellular carcinoma: A meta-analysis. Abdom. Radiol. 2016, 41, 33–41. [Google Scholar] [CrossRef] [PubMed]
  67. Abuodeh, Y.; Naghavi, A.O.; Ahmed, K.A.; Venkat, P.S.; Kim, Y.; Kis, B.; Choi, J.; Biebel, B.; Sweeney, J.; Anaya, D.A.; et al. Prognostic value of pre-treatment F-18-FDG PET-CT in patients with hepatocellular carcinoma undergoing radioembolization. World J. Gastroenterol. 2016, 22, 10406–10414. [Google Scholar] [CrossRef] [PubMed]
  68. Jreige, M.; Mitsakis, P.; Van Der Gucht, A.; Pomoni, A.; Silva-Monteiro, M.; Gnesin, S.; Boubaker, A.; Nicod-Lalonde, M.; Duran, R.; Prior, J.O.; et al. (18)F-FDG PET/CT predicts survival after (90)Y transarterial radioembolization in unresectable hepatocellular carcinoma. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 1215–1222. [Google Scholar] [CrossRef] [PubMed]
  69. Antkowiak, M.; Gabr, A.; Das, A.; Ali, R.; Kulik, L.; Ganger, D.; Moore, C.; Abecassis, M.; Katariya, N.; Mouli, S.; et al. Prognostic Role of Albumin, Bilirubin, and ALBI Scores: Analysis of 1000 Patients with Hepatocellular Carcinoma Undergoing Radioembolization. Cancers 2019, 11, 879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Mohammadi, H.; Abuodeh, Y.; Jin, W.; Frakes, J.; Friedman, M.; Biebel, B.; Choi, J.; El-Haddad, G.; Kis, B.; Sweeney, J.; et al. Using the Albumin-Bilirubin (ALBI) grade as a prognostic marker for radioembolization of hepatocellular carcinoma. J. Gastrointest. Oncol. 2018, 9, 840–846. [Google Scholar] [CrossRef]
  71. 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] [Green Version]
  72. 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]
  73. 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]
  74. Cheng, A.L.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.Y.; Lim, H.Y.; Kudo, M.; Breder, V.; Merle, P.; et al. Updated efficacy and safety data from IMbrave150: Atezolizumab plus bevacizumab vs. sorafenib for unresectable hepatocellular carcinoma. J. Hepatol. 2021, 76, 862–873. [Google Scholar] [CrossRef]
  75. Lescure, C.; Estrade, F.; Pedrono, M.; Campillo-Gimenez, B.; Le Sourd, S.; Pracht, M.; Palard, X.; Bourien, H.; Muzellec, L.; Uguen, T.; et al. ALBI Score Is a Strong Predictor of Toxicity Following SIRT for Hepatocellular Carcinoma. Cancers 2021, 13, 3794. [Google Scholar] [CrossRef]
  76. Ali, R.; Gabr, A.; Abouchaleh, N.; Al Asadi, A.; Mora, R.A.; Kulik, L.; Abecassis, M.; Riaz, A.; Salem, R.; Lewandowski, R.J. Survival Analysis of Advanced HCC Treated with Radioembolization: Comparing Impact of Clinical Performance Status Versus Vascular Invasion/Metastases. Cardiovasc. Intervent. Radiol. 2018, 41, 260–269. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Multi-compartment dosimetry (partition model) using technetium-99m macro-aggregated albumin single-photon emission computed tomography combined with computed tomography for activity planning. The absorbed doses in these different compartments can be simulated before treatment and enable optimization of the activity planned.
Figure 1. Multi-compartment dosimetry (partition model) using technetium-99m macro-aggregated albumin single-photon emission computed tomography combined with computed tomography for activity planning. The absorbed doses in these different compartments can be simulated before treatment and enable optimization of the activity planned.
Curroncol 29 00196 g001
Table 1. Prospective and randomized studies in hepatocellular carcinoma.
Table 1. Prospective and randomized studies in hepatocellular carcinoma.
StudiesGroupsNb of PatientsBCLC ScoreAdverse Events (≥Grade 3)RRTTP (mo)PFS (mo)OS
(mo)
Pitton et al., 2015 [22] SIRT (resin)12B: 100%NANA12.4619.7
TACE12A: 8%
B: 92%
NANA11.27.226.3
Salem et al., 2016 [17]SIRT (glass)24A: 75%
B: 25%
NA87%>26 *NA18.6
TACE21A: 81%
B: 19%
NA74%4.8NA17.7
SARAH [19] SIRT (resin)237C: 100%41%19% *NA4.19.9
Sorafenib222C: 100%63% *12%NA3.79.9
SIRveNIB [20] SIRT (resin)130B: 61%
C: 39%
28%23% *6.16.38.8
sorafenib162B: 54%
C: 45%
51% *2%5.45.210
SORAMIC [23] SIRT (resin) + sorafenib114A: 4%
B: 28%
C: 68%
65% *NANANA14
sorafenib174A: 2%
B: 28%
C: 70%
54%NANANA11.1
* Statistically significant differences using a Kaplan–Meier method and the log-rank test (p < 0.05). Nb, Number; mo, months; BCLC, Barcelona Clinic Liver Cancer; NA, not available; OS, overall survival; PFS, progression-free survival; RR, response rate; SIRT, selective internal radiation therapy; TACE, transarterial chemoembolization; RR, response rate; TTP, time to progression.
Table 2. Main studies reporting a correlation between tumor dosimetry in SIRT and clinical response.
Table 2. Main studies reporting a correlation between tumor dosimetry in SIRT and clinical response.
StudyStudy DesignType of MicrospheresNb of PatientsCorrelation with Radiological ResponseCorrelation with PFSCorrelation with OS
Strigari et al., 2010 [27]RetrospectiveResin73NANA
Chiesa et al., 2011 [31]RetrospectiveGlass46NANA
Garin et al., 2012 [32]RetrospectiveGlass36
Garin et al., 2017 [33]RetrospectiveGlass85NA
Kappadath et al., 2018 [34]RetrospectiveGlass34NANA
Allimant et al., 2018 [35]RetrospectiveResin38NA
Chan et al., 2018 [36]ProspectiveGlass27NANA
Hermann et al., 2020 [28]Prospective +Resin121NA
Dewaraja et al., 2020 [29]RetrospectiveGlass28NANA
d’Abadie et al., 2021 [37]RetrospectiveResin and glass45
Son et al., 2021 [38]Prospective +Resin34NANA
Nodari et al., 2021 [39]RetrospectiveResin and glass48NA
Garin et al., 2021 [40]Prospective, randomized, multicenterGlass56
Nb, Number; OS:, overall survival; PFS, progression-free survival; ✓, significant correlation with tumor dosimetry; NA, not available. + Secondary analysis of prospectively acquired data.
Table 3. Main studies reporting threshold absorbed doses correlated with clinical outcome in hepatocellular carcinoma using glass microspheres.
Table 3. Main studies reporting threshold absorbed doses correlated with clinical outcome in hepatocellular carcinoma using glass microspheres.
StudyNb of PatientsNb of TumorsDosimetry Performed withCriteria for Radiological Response AssessmentTD Threshold
for Radiological Response
Median PFS above and under the TD ThresholdMedian OS above and under the
TD Threshold
Chiesa et al., 2011 [31]4691MAA SPECT/CTEASL257 Gy
(Se: 85%, Sp: 70%)
NANA
Garin et al., 2012 [32]3658MAA SPECT/CTEASL205 Gy
(Se: 100%, Sp: 75%)
14 mo vs. 5.2 mo *18 mo vs. 9 mo *
Garin et al., 2017 [33]85132MAA SPECT/CTEASL205 Gy
(Se: 98%, Sp NA)
NA21 mo vs. 6.5 mo *
Kappadath et al., 2018 [34] 345390Y SPECT/CTmodified RECIST 1.1160 Gy
(50% response)
NANA
Chan et al., 2018 [36]273890Y PET/CTmodified RECIST 1.1200 Gy
(Se: 66%, Sp: 100%)
NANA
d’Abadie et al., 2021 [37] 267390Y PET/CTmodified RECIST 1.1118 Gy
(Se: 93%, Sp: 75%)
5.5 mo vs. 1.8 mo *14.6 mo vs. 5.5 mo *
Nodari et al., 2021 [39] 23NA90Y PET/CTNA156 Gy
(Se and Sp NA)
NA23 mo vs. 14 mo *
* Statistically significant differences using a Kaplan–Meier method and the log-rank test (p-value < 0.05). NB, Number; mo, months EASL, European Association for the Study of the Liver; MAA SPECT/CT, technetium-99m macro-aggregated albumin single-photon emission computed tomography combined with computed tomography; NA, not available; OS:, overall survival; PFS, progression-free survival; RECIST, Response Evaluation Criteria in Solid Tumors; Se, sensitivity; SIRT, selective internal radiation therapy; Sp, specificity; TD, tumor-absorbed dose threshold; 90Y PET/CT, yttrium-90 positron emission tomography combined with computed tomography; 90Y SPECT/CT, yttrium-90 single-photon emission computed tomography combined with computed tomography.
Table 4. Main studies reporting threshold absorbed doses correlated with clinical outcome in hepatocellular carcinoma using resin microspheres.
Table 4. Main studies reporting threshold absorbed doses correlated with clinical outcome in hepatocellular carcinoma using resin microspheres.
StudyNb of PatientsNb of TumorsDosimetry Performed withCriteria for Radiological Response AssessmentTD Thresholdfor Radiological ResponseMedian PFS above and under the TD ThresholdMedian OS above and under the TD Threshold
Allimant et al., 2018 [35]384290Y PET/CTmodified RECIST 1.161 Gy
(Se: 76%, Sp: 75%)
12.1 mo vs. 6.3 mo *+NA
Hermann et al., 2020 [28] 121NAMAA SPECT/CTRECIST 1.1100 Gy
(72% response)
NA14.1 mo vs. 6.1 mo *
d’Abadie et al., 2021 [37]196090Y PET/CTmodified RECIST 1.161 Gy
(Se: 87%, Sp: 64%)
4.6 mo vs. 1.6 mo *16 mo vs. 5.3 mo *
Son et al.,2021 [38] 3445MAA SPECT/CTmodified RECIST 1.1125 Gy
(Se: 86%, Sp: 75%)
NANA
Nodari et al., 2021 [39] 25NA90Y PET/CTNA98 Gy
(Se and Sp NA)
NA23 mo vs. 14 mo *
* Statistically significant differences using a Kaplan–Meier method and the log-rank test (p-value < 0.05). + Reported for complete tumor targeting (25 patients). Nb, Number; mo, months; EASL, European Association for the Study of the Liver; MAA SPECT/CT, technetium-99m macro-aggregated albumin single-photon emission computed tomography combined with computed tomography; NA, not available; OS, overall survival; PFS, progression-free survival; RECIST, Response Evaluation Criteria in Solid Tumors; Se, sensitivity; SIRT, selective internal radiation therapy; Sp, specificity; TD, tumor-absorbed doses; 90Y PET/CT, yttrium-90 positron emission tomography combined with computed tomography; 90Y SPECT/CT, yttrium-90 single-photon emission computed tomography combined with computed tomography.
Table 5. Main results of the DOSISPHERE-01 randomized controlled trial [40].
Table 5. Main results of the DOSISPHERE-01 randomized controlled trial [40].
Personalized DosimetryStandard Dosimetry
Number of patients2828
Activity planned in GBq, median3.6 *2.6
Response rate at 3 mo, EASL criteria71% *36%
Curative surgery intent after SIRT36% *4%
REILD9%10%
Overall survival in mo, median26.6 +10.7
* Statistically significant differences using a chi-square or Fisher’s exact tests (p < 0.05). + Statistically significant differences using a using a Kaplan–Meier method and the log-rank test (p < 0.05). EASL, European Association for the Study of the Liver; SIRT, selective internal radiation therapy; REILD, radioembolization-induced liver disease.
Table 6. Studies reporting factors of poor prognosis in advanced HCC treated by SIRT.
Table 6. Studies reporting factors of poor prognosis in advanced HCC treated by SIRT.
StudyNb of PatientsParameter Related to Worse PrognosisMedian Survival
(95% CI Interval)
Ali et al.,
2018 [76]
547ECOG 24.3 mo (2.5–7.8)
Extrahepatic metastases7.4 mo (6.0–9.0)
PVT7.3 mo (6.3–8.0)
Spreafico et al., 2018 [50] 120Bilirubin > 1.2 mg/dL9.5 mo (8.8–10.2)
PVT extended to right/left main branch8.2 mo (5.7–10.8)
Tumor burden > 50% liver volume6.4 mo (5.2–7.6)
Abouchaleh et al., 2018 [46]185ECOG 22.5 mo (2–4.6)
Bilirubin 2–3 mg/dL5 mo (2.2–9.7)
PVT extended to right/left main branch7.7 mo (5.3–10.4)
Antkowiak et al., 2019 [69]541Bilirubin 2–3 mg/dL8 mo (6.7–21)
ALBI grade 36.7 mo (5.7–8.8)
Zu et al.,
2020 [47]
91CHILD B76 mo (4.4–7.6)
Lescure et al.,
2021 [75]
222ALBI grade 38.1 mo (4.1–12.1)
Nb, number; mo, months; ALBI, albumin–bilirubin; ECOG, Eastern Cooperative Oncology Group; PVT, portal vein thrombosis.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

d’Abadie, P.; Walrand, S.; Lhommel, R.; Hesse, M.; Borbath, I.; Jamar, F. Optimization of the Clinical Effectiveness of Radioembolization in Hepatocellular Carcinoma with Dosimetry and Patient-Selection Criteria. Curr. Oncol. 2022, 29, 2422-2434. https://doi.org/10.3390/curroncol29040196

AMA Style

d’Abadie P, Walrand S, Lhommel R, Hesse M, Borbath I, Jamar F. Optimization of the Clinical Effectiveness of Radioembolization in Hepatocellular Carcinoma with Dosimetry and Patient-Selection Criteria. Current Oncology. 2022; 29(4):2422-2434. https://doi.org/10.3390/curroncol29040196

Chicago/Turabian Style

d’Abadie, Philippe, Stephan Walrand, Renaud Lhommel, Michel Hesse, Ivan Borbath, and François Jamar. 2022. "Optimization of the Clinical Effectiveness of Radioembolization in Hepatocellular Carcinoma with Dosimetry and Patient-Selection Criteria" Current Oncology 29, no. 4: 2422-2434. https://doi.org/10.3390/curroncol29040196

Article Metrics

Back to TopTop