Next Article in Journal
Biomarkers of Trastuzumab-Induced Cardiac Toxicity in HER2- Positive Breast Cancer Patient Population
Previous Article in Journal
Circulating Exosome Cargoes Contain Functionally Diverse Cancer Biomarkers: From Biogenesis and Function to Purification and Potential Translational Utility
Previous Article in Special Issue
Pulsed Microwave Liver Ablation: An Additional Tool to Treat Hepatocellular Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Intraarterial Therapies for the Management of Hepatocellular Carcinoma

1
Division of Vascular and Interventional Radiology, Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
2
Division of Gastroenterology and Hepatology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
3
Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
4
Vascular and Interventional Radiology Division, Department of Radiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
5
Section of Vascular and Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT 06510, USA
6
Division of Vascular and Interventional Radiology, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
7
Department of Clinical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
8
Vascular and Interventional Radiology, Radiology Associates of Florida, Sarasota, FL 34239, USA
9
Experimental Therapeutics Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(14), 3351; https://doi.org/10.3390/cancers14143351
Submission received: 28 May 2022 / Revised: 5 July 2022 / Accepted: 7 July 2022 / Published: 10 July 2022
(This article belongs to the Special Issue Multimodal and Multi-Sequential Treatment of Hepatocellular Carcinoma)

Abstract

:

Simple Summary

Hepatocellular carcinoma is the most common liver cancer, leading to approximately 700,000 deaths worldwide and 30,000 deaths in the United States every year. Transarterial therapies play a crucial role in the management of these patients, with significant development in techniques over the last couple of decades. The aim of this review is to discuss the different types of transarterial therapies with regards to the pre-procedure, procedural, and post-procedural patient management, along with giving a review of evidence from the literature.

Abstract

Image-guided locoregional therapies play a crucial role in the management of patients with hepatocellular carcinoma (HCC). Transarterial therapies consist of a group of catheter-based treatments where embolic agents are delivered directly into the tumor via their supplying arteries. Some of the transarterial therapies available include bland embolization (TAE), transarterial chemoembolization (TACE), drug-eluting beads–transarterial chemoembolization (DEB–TACE), selective internal radioembolization therapy (SIRT), and hepatic artery infusion (HAI). This article provides a review of pre-procedural, intra-procedural, and post-procedural aspects of each therapy, along with a review of the literature. Newer embolotherapy options and future directions are also briefly discussed.

1. Introduction

Image-guided locoregional therapies play a crucial role in the management of patients with hepatic tumors. Transarterial therapies consist of a group of catheter-based treatments where embolic agents or chemotherapeutic agents are directed into the target tumor via their supplying arteries [1,2]. In the current treatment algorithm of HCC, there is a role supporting the use of bland embolization (TAE), transarterial chemoembolization (TACE), drug-eluting beads–transarterial chemoembolization (DEB–TACE), selective internal radioembolization therapy (SIRT), and hepatic artery infusion (HAI) [3]. The physiological principle behind image-guided intraarterial therapies is based on the vascular supply to the tumors. Although the majority of hepatic blood supply is provided by the portal venous system, the hepatic arteries are the predominant blood supply to HCCs. Therefore, selective and local delivery of tumorocidal agents directly into the tumor spares the adjacent hepatic parenchyma and minimizes systemic complications and toxicities [1,2]. Extensive high-quality evidence is available to support the use of image-guided locoregional therapies for the management of hepatic tumors, and this has led to the addition of locoregional procedures to National Comprehensive Cancer Network (NCCN) and Barcelona Clinic Liver Cancer (BCLC) treatment guidelines [4,5]. This article provides a review of pre-procedural, intra-procedural, and post-procedural aspects of each therapy, along with a review of the literature.

2. Methodology for Review

We searched the MEDLINE/PubMed database, using the following terms to identify all original research articles published related to TAE, TACE, DEB–TACE, SIRT, and HAI:
((Therapeutic Chemoembolization OR Chemoembolizations, Therapeutic OR Therapeutic Chemoembolizations) OR (radioembolization OR selective internal radiation therapy OR radiation segmentectomy) OR (hepatic arter * infusion OR chemoinfusion) OR (Embolotherapy OR Embolotherapies OR Therapeutic Embolization OR Embolizations, Therapeutic OR Therapeutic Embolizations)) AND (Carcinomas, Hepatocellular OR Hepatocellular Carcinomas OR Liver Cell Carcinoma, Adult OR Liver Cancer, Adult OR Adult Liver Cancer OR Adult Liver Cancers OR Cancer, Adult Liver OR Cancers, Adult Liver OR Liver Cancers, Adult OR Liver Cell Carcinoma OR Carcinoma, Liver Cell OR Carcinomas, Liver Cell OR Cell Carcinoma, Liver OR Cell Carcinomas, Liver OR Liver Cell Carcinomas OR Hepatocellular Carcinoma OR Hepatoma OR Hepatomas) Filters: Clinical Study, Clinical Trial, Clinical Trial Protocol, Clinical Trial, Phase I, Clinical Trial, Phase II, Clinical Trial, Phase III, Clinical Trial, Phase IV, Comparative Study, Controlled Clinical Trial, Meta-Analysis, Multicenter Study, Observational Study, Randomized Controlled Trial, Systematic Review, Technical Report.
The results from this search were reviewed by two authors (T.G. and A.S.) to select the key references to write the review article.

3. Treatment Options for Hepatocellular Carcinoma

Given the numerous surgical, medical, and minimally invasive options available for the treatment of HCC, many patients with hepatocellular carcinoma (HCC) benefit when a multidisciplinary approach to treatment is undertaken. This ensures a standardized workup and access to the most up-to-date treatment options based on individual patient parameters, including social factors; underlying liver function; tumoral factors, such as tumor size; invasion of blood vessels and extrahepatic structures; transplant eligibility; institutional experience; and access to various therapies.

3.1. Surgical and Systemic Therapies

Surgical resection and liver transplantation are classically considered to be the only potentially curative treatment options for HCC. Partial hepatectomy is only offered to patients with solitary HCC without any evidence of macrovascular invasion or cirrhosis, and without significant stigmata of portal hypertension, while transplantation is reserved for patients with early HCC within the United Network for Organ Sharing (UNOS) criteria, an adapted version of the Milan criteria [6]. In patients who meet the stringent criteria for future transplantation, “bridging” therapy can prevent disease progression and reduce the dropout rate from the transplant waiting list [7]. Locoregional therapies such as radiofrequency ablation, microwave ablation, TAE, TACE, DEB–TACE, and SIRT have all been shown to be useful as bridge therapies [8,9,10,11,12,13,14,15,16,17,18,19]. “Downstaging” therapies are used in cases of advanced HCC in patients without distant metastasis to reduce the intrahepatic tumor burden [7,20,21]. Locoregional therapies are commonly used for this purpose, allowing many previously ineligible patients to become eligible for liver transplantation under the Milan or UNOS criteria [22]. These therapies have been shown to improve the 1- and 5-year survival after transplant compared to liver transplantation alone [23].
For patients with advanced HCC, systemic therapy can be considered as the first-line treatment [24,25]. Currently, the United States Food and Drug Administration (FDA) has approved six agents for use as systemic therapies. These include atezolizumab + bevacizumab, sorafenib, lenvatinib, regorafenib, cabozantinib, ramucirumab, and nivolumab [26,27,28,29,30].

3.2. Locoregional Therapies

According to the BCLC algorithm, the choice of locoregional therapy in patients with early stage HCC is based on the size of the tumor, underlying liver disease, and liver function. Locoregional therapies can generally be subdivided into (1) percutaneous and (2) intraarterial modalities. Generally, ablative techniques are used in tumors less than ≤3 cm located remotely from the hilar bile ducts [31,32,33,34,35,36]. Ablation involves the direct application of thermal or nonthermal/chemical energy into tissues in order to induce cellular injury and apoptosis. The most commonly utilized ablation technologies within the liver include radiofrequency ablation, microwave ablation, cryoablation, and percutaneous ethanol injection.
Transarterial therapies rely on the arterial blood supply of targeted tumors and include TAE, TACE, DEB–TACE, SIRT, and HAI (Figure 1). These techniques are generally considered when treating larger tumors, multifocal disease, or for tumors centrally located or adjacent to major vasculature/bile ducts. The efficacy of TAE stems from a reduction of blood flow to the supplying artery, thus leading to ischemia in the region of the tumor that can then lead to tissue necrosis. With TACE and DEB–TACE, a high concentration of chemotherapy is additionally delivered to tumor cells [37], whereas the mechanism of action for SIRT is the emission of high-dose beta-radiations into the capillary bed of the tumor [3,38]. In HAI, chemotherapy is administered non-selectively to the liver and embedded tumors via the proper hepatic artery.
Radiation therapies are also considered locoregional therapies in patients with inoperable or unresectable HCC. The two commonly used types of radiation therapy are external beam radiation therapy (EBRT) and stereotactic body radiation therapy (SBRT). In EBRT, a high dose of radiation is delivered to the target tissue (liver tumor), while a lower dose is delivered to the normal liver parenchyma surrounding the tumor. SBRT is a more advanced version of EBRT which helps to deliver a larger amount of ablative dose to the target tissue (liver tumor) without further increasing the dose delivered to the normal liver parenchyma [39,40]. Proton beam therapy (PBT) is another radiation therapy option in which a finite range of a radiation dose is delivered to the target tissue (liver tumor); however, it causes lower delivery of radiation to the normal liver tissue at the margins, providing a dosimetric advantage compared to SBRT [41]. Evidence from multiple studies in the literature has shown that SBRT is useful in the treatment of patients with unresectable, locally advanced, or recurrent HCC [42,43,44,45]. EBRT and SBRT are currently not a part of BCLC algorithm for treatment of the HCC. An in-depth discussion of radiation therapies is beyond the focus of this review article.

4. Indications and Patient Selection for Treatment with Transarterial Therapies

Transarterial therapies can be used to reduce the burden of tumor within the transplant criteria (downstaging), to control the growth of tumor in patients who are currently on the transplant waiting list (bridging), and to increase the survival of patients who are not eligible to undergo a transplant (palliative). A multidisciplinary tumor board consisting of oncologists, surgeons, diagnostic and interventional radiologists, and hepatologists optimally can decide in aggregate if transarterial therapy is indicated for each individual patient [46]. Figure 2 shows the BCLC-approach indications for transarterial therapies in HCC [5].

5. BCLC Staging and Predictors of Response

The BCLC scoring system classifies patients into five stages (BCLC 0, BCLC A, BCLC B, BCLC C, and BCLC D; Table 1) on the basis of size and extent of the primary HCC lesion, performance status of the patient, and presence of vascular invasion and extrahepatic disease [5]. In some studies, the BCLC scoring system has been shown to outperform other prognostic scoring systems in patients undergoing surgical treatment [47,48]. Patients with very early and early BCLC grades have a low tumor burden (≤3 tumors, <3 cm), preserved liver function (Child–Pugh A), and are fully active (ECOG 0). These patients have been shown to have a life expectancy of more than 5 years. Patients are classified into intermediate stage (BCLC B) if they have a multinodular tumor with preserved liver function (Child–Pugh A to B7) and normal functional status (ECOG 0). Advanced stage (BCLC C) includes patients with macrovascular invasion (i.e., tumor thrombus) or extrahepatic disease, along with preserved liver function and worsening performance status (ECOG 1 and 2). The life expectancy of patients in this group has been shown to be approximately 10 months. Terminal-stage disease (BCLC D) includes patients with any tumor burden but with the presence of poor liver function (Child–Pugh C) or a poor performance status (ECOG >2). BCLC-D patients have a life expectancy of only 3 months [5]. The presence of liver decompensation (ascites, jaundice, and encephalopathy) is considered as non-preserved liver function regardless of the Child–Pugh and model for end-stage liver disease (MELD) score. The BCLC staging system has been adopted widely due to its simplicity and prognostic reproducibility. However, the BCLC system has been shown to be conservative in terms of intermediate- and advanced-stage management.

5.1. Eastern Cooperative Oncology Group (ECOG)

The ECOG Performance Status Scale is used to assess a patient’s level of function by looking at his/her ability to perform physical activities, complete daily activities, and take care of him/herself. The ECOG Performance Status Scale is an essential component of BCLC classification. Table 2 shows the ECOG Performance Status Scale [49].

5.2. Child–Pugh Classification

Based on clinical and laboratory information, Child–Pugh scoring predicts the prognosis of patients with cirrhosis by estimating their liver function. Table 3 demonstrates the Child–Pugh classification of severity of cirrhosis. Patients with a score of 5 to 6 are classified in class A and are said to have well-compensated cirrhosis, those with a score of 7–9 are classified in class B and are said to have significant functional compromise of the liver, and those with a score of > 9 are classified in class C and are said to have decompensated cirrhosis [50]. According to several studies, TACE is recommended for Child–Pugh class A and highly selective class B cirrhosis [51].

5.3. Albumin–Bilirubin (ALBI) Score

A significant limitation of the Child–Pugh classification is its subjective assessment due to the inclusion of ascites and encephalopathy in the score that can easily be modified by medication(s). Therefore, a new score called ALBI was developed that includes only objective parameters, serum albumin and bilirubin, to assess the liver function. The formula for calculating the ALBI score is as follows:
ALBI score = (log10 bilirubin 0.66) (albumin 0.085)
where bilirubin is in mmol L−1, and albumin is in g L−1.
Table 4 shows ALBI grading according to the ALBI score. Child–Pugh class A can be subdivided into ALBI Grades 1 and 2 [52]. According to several retrospective studies, ALBI is superior to the Child–Pugh classification in regard to prognosis identification, accuracy, and stratification [53]. The ALBI score has shown an excellent prognostic and predictive ability for patients undergoing palliative transarterial therapies [54]. It has also been used to categorize patients belonging to BCLC-B class undergoing TACE [55].

5.4. Platelet–ALBI (pALBI) Score

In order to include the presence of paraneoplastic syndrome in patients with HCC, the pALBI score was developed. The pALBI score includes platelet counts as an indicator of a paraneoplastic syndrome of HCC. The formula for the pALBI score is given below:
PALBI score = 2.02 × log10 bilirubin − 0.37 × (log10 bilirubin)2 − 0.04 × albumin − 3.48 × log10 platelets + 1.01 × (log10 platelets)2
Table 5 shows the pALBI grading according to the pALBI score [56]. According to a study by Liu et al., a higher C-index was seen for the PALBI grade than the ALBI grade, thus indicating its usefulness as a robust statistical model that provides improved discriminatory power [57].

5.5. The Neutrophil-to-Lymphocyte Ratio (NLR)

In patients with inflammation, neutrophils have been shown to reduce the activity of different types of lymphocytes. The ratio of neutrophils to lymphocytes can be used as a marker of inflammation and immune response [58]. A couple of studies have highlighted that a higher lymphocyte count correlates with treatment response and prognosis [59,60]. As per the study conducted by Wang et al., NLR values increased three days post-TACE and then fell back to baseline after one month; a higher NLR (>2.4) was associated with overall survival of 15.6 months vs. a lower NLR (≤2.4) having an overall survival of 27.1 months [61].

5.6. MELD Score

The MELD score was first developed to predict the survival of patients with liver cirrhosis who underwent placement of a transjugular intrahepatic portosystemic shunt for their disease management. Currently, the MELD system is used to rank liver-transplantation candidates’ priority. The MELD score is calculated by measuring three objective variables: total bilirubin, creatinine, and INR [62]. In a study by Sawhney et al., a MELD score of higher than ten is associated with higher mortality after TACE (Hazard ratio: 4.9, p = 0.001) [63].

5.7. Assessment for Retreatment with Transarterial Chemoembolization (ART) Score

The ART score is used to predict the survival of patients with HCC after their first TACE treatment by looking at the increase in Child–Pugh score (1.5 point for 1 point increase, 3 points for ≥2 points increase, and 0 points for no increase), presence or absence of radiological response (1 point for no response, and 0 points for presence of response), and elevation of aspartate transaminase (4 points for >25% increase, and 0 points for ≤25% increase) [64]. A high ART score after the first TACE was found to be associated with the risk of major adverse events upon the second TACE (p = 0.011) [64]. When the patients where stratified on the basis of their ART score in the initial validation study, patients with a score of 0–1.5 points were found to have a median OS of 23.7 months, and those with ≥2.5 points were found to have a median OS of 6.6 months [64]. These findings were confirmed in an unrelated study conducted by Abbasi et al. [65].

5.8. ASARA Scoring System

More recently, the ASARA scoring system, a predictive model for stratifying candidates based on liver function for suitability for initial/repeated TACE procedure, has come into the picture. AFP ≥ 400 ng mL−1, tumor size (maximum diameter >7 cm), increase in ALBI score, no tumor response, and increase in AST ≥25% are each assigned a score of 1 (total score range of 0–5) [66]. The median OS for patients with a score of ≤2 was significantly higher than those with a score of >2 (p = 0.006). This model suggests that a score of >2 predicts TACE failure; thus, switching to systemic therapy should be considered in such scenarios [66]. For patients with Child–Pugh class B and impaired hepatic function, the modified AS (ARA) model could be applied to screen individuals for initial TACE treatment (cutoff value >1 predicts worse prognosis, p = 0.004) [66].

5.9. Hepatoma Arterial-Embolization Prognostic (HAP) Score

The HAP scoring system stratifies patients into low (HAP class A), intermediate (HAP class B), high (HAP class C), and very high risk (HAP class D) groups for predicting survival after TACE [67]. An albumin level of <36 g dL−1, AFP levels of >400 ng mL−1, bilirubin levels of >17 μmol L−1, and a maximum tumor diameter of >7 cm are each given 1 point in this scoring system. The OS for class A (score 0), class B (score 1), class C (score 2), and class D (≥2) were 25.5, 18.1, 8.9, and 5.9 months, respectively, in the validation model of this scoring system [67]. According to a few studies, HAP classification might be a better predictor of survival than Child–Pugh, CLIP, BCLC, and MELD scoring systems [67,68].

5.10. Selection for Transarterial Chemoembolization Treatment (STATE) Score

The STATE score has been developed to identify unbefitting patients for the initial TACE session. The STATE score = serum albumin (9g L−1) − 12 (if up-to-7-out) − 12 (if CRP >1 m dL−1). Patients with a total score of <18 points had a poorer prognosis as opposed to patients with a score of ≥18 (median OS: 5.3 months vs. 19.5 months, p < 0.001) [69]. Additionally, patients with a score of <18 points had a significantly greater rate of serious adverse events following their initial TACE session and higher short-term mortality (number needed to harm = 4) [69]. These findings have been confirmed in a few other studies [68,70].

5.11. Aspartate-Aminotransferase-to-Platelet Index (APRI) Score

The APRI scoring system was developed by Wai et al. for predicting significant fibrosis and cirrhosis in patients with chronic hepatitis C [71]. Lately, the APRI score has been used to predict survival in patients with HCC. A meta-analysis suggested a significant association between high APRI levels and poor prognosis in patients with HCC (HR 1.59, p < 0.001) [72]. Furthermore, another study indicated that the APRI score has indistinguishable accuracy but higher negative predictive value and sensitivity for predicting post-TACE acute liver function deterioration when compared to Child–Pugh scoring [73].

5.12. Tumor Size, Tumor Number, Baseline AFP, Child–Pugh Class, and Objective Radiological Response (SNACOR) Score

The SNACOR classification is another predictive model for the prognostication of patients with HCC who underwent TACE. Tumor size (<5 cm = 1 point, ≥5 cm = 2 points), tumor number (<4 = 1 point, ≥4 = 2 points), baseline AFP levels (<400 ng mL−1 = 1 point, ≥400 ng mL−1 = 2 points), Child–Pugh class (A = 1 point, B = 2 points), and response evaluation (complete response + partial response = 1 point, stable disease + progressive disease = 2 points) are the components of this scoring system. The patients are classified into three groups based on their total score: low-risk (0–2), intermediate (3–6), and high-risk (7–10) groups [74]. This model suggested that the patients in the intermediate (HR 2.13) or high-risk (HR 6.17) groups had a significant risk of death compared to the low-risk group (p < 0.001) and might not be candidates for retreatment with TACE [74]. A validation study described this model as adequate to distinguish favorable vs. impaired prognosis groups with moderate performance [75].

5.13. Okuda Score

The Okuda prediction model, one of the oldest staging systems, is a scoring scheme based on tumor size, ascites, serum bilirubin, and serum albumin levels; each scored 0/1. Based on the total score, patients are classified into three stages: Stage I (0 points), Stage II (1–2 points), and Stage III (3–4 points) [76]. In the original cohort of 850 patients diagnosed with HCC in 1975–1983, the median OS between the three stages was 11.5 months (Stage I) vs. 3 months (Stage II) vs. 0.9 months (Stage III) [76]. However, Levy et al. found no significant difference in the median OS between Stage II and Stage III patients (3.2 months vs. 3.1 months, p = −0.43) in their cohort of patients [77].

5.14. Cancer of the Liver Italian Program (CLIP) Score

The CLIP scoring system considers the residual liver function and tumor characteristics for the prognostic assessment of HCC patients [78]. In a study by Li et al., patients with a CLIP score of 0–2 were found to have a longer OS than those with CLIP scores of 3–5 (13 months vs. 4 months, p = 0.001), making them better candidates for TACE [79]. A recent study suggests that the CLIP system is superior for predicting survival in patients with unresectable HCC treated with TACE compared to the Okuda classification system and is easier to implement [68].

6. Patient Preparation

6.1. Pretreatment Imaging

A contrast enhanced multiphasic MDCT or MRI can be used for the diagnosis of HCC in lesions >1 cm and should be performed preferably within three months of the procedure [80]. Imaging should be obtained to assess factors such as vascular anatomy, lesion size, number of lesions, macrovascular invasion, and extrahepatic spread, which help determine eligibility for locoregional and/or systemic therapies [81]. MDCT is considered superior for delineating the vascular anatomy of the hepatic vasculature, as well as identifying parasitic tumor feeders; however, MRI demonstrates superior evaluation of lesion enhancement, morphology, and classification [82].

6.2. Pre-Procedural Labs and Tumor Markers

The following tests are recommended for diagnosing the extent of liver function and obtaining baseline liver function for potential postprocedural hepatotoxicity [46]:
  • Aminotransferase,
  • Cholinesterase,
  • Alkaline phosphatase,
  • Gamma-glutamyl transferase,
  • Bilirubin,
  • Albumin,
  • Prothrombin time,
  • Creatinine,
  • Electrolytes.
In addition to the above tests, testing for tumor markers is also essential. Alpha-fetoprotein is the most commonly used tumor marker for HCC, as it is able to predict disease prognosis and aids in monitoring tumor recurrence [83,84]. More recently, vitamin K absence, antagonist-II (PIVKA-II), has shown to increase HCC detection rates and can be obtained in adjunct to AFP and imaging [85].

6.3. Pre-Procedural Preparation

Fasting for six to eight hours is required for preparation for conscious sedation or general anesthesia.
Proton pump inhibitors may be given to reduce the chances of post-procedure gastritis/duodenitis and mitigate the significance of non-target embolization. Dexamethasone (20 mg intravenously) may also be given to reduce the risk of post-embolization syndrome. Intravenous fluids should be considered in those without cardiac contraindications. Anxiolytics have also been found to be beneficial pre-procedure [46]. If predisposing factors for liver abscess listed below are found, then an antimicrobial regimen such as 400 mg of moxifloxacin by mouth daily, beginning three days pre-procedure to seventeen days post-embolization, should be prescribed; however, longer courses are also utilized [86].
  • Biliary tree canulation/dilation/sphincterotomy;
  • Presence of biliary prosthesis (e.g., plastic and metallic stents);
  • Presence of a bilioenteric anastomosis;
  • Presence of TIPS [87];
  • Biliary or gallbladder stone.
Patients without the above risk factors are typically given a single dose of cefazolin immediately prior to the procedure. The pre-procedural target international normalized ratio (INR) and platelet values are desired to be INR ≤2–3 and a platelet count of >20,000 μL−1, as, according to the Society of Interventional Radiology Consensus Guidelines, these procedures are classified to be low risk for bleeding [88].

7. Techniques: Angiography and Embolization Technique

7.1. Initial Angiography

Either the common femoral artery (CFA) or radial artery (RA) access can be used for performing transarterial embolization [89]. A 4–6 Fr vascular sheath is used with either approach. If the RA access is selected, a cocktail of heparin and one or more vasodilators (nitroglycerin, verapamil, or nicardipine) is generally administered intraarterially via the sheath once access has been achieved [90].
Mesenteric angiography is performed to visualize and define the relevant vascular anatomy and identify tumoral arterial supply, as well as collaterals and arterial supply at risk of non-target embolization [91]. A superior mesenteric angiogram is obtained first to exclude aberrant vascular anatomy and confirm portal vein patency on delayed imaging. Then a celiac arteriogram is performed to identify target (hepatic artery branches) and non-target arteries and hypervascular tumors [92]. In situations where, extrahepatic arterial recruitment is suspected, extrahepatic vessels (phrenic, gastric, and internal mammary) are also evaluated [93]. Frequently, intraprocedural cone-beam CT technology is utilized to identify and/or confirm arterial supply of targeted tumors [94].
Once the procedure is completed, all the catheters and sheaths are removed, and hemostasis is achieved by either manual compression or with the help of a closure device [95].

7.2. Embolization Techniques

7.2.1. Bland Embolization (TAE)

In TAE, after following the steps described in the previous section, lobar embolization or subselective embolization can be performed depending on the arterial distribution, location of the tumor, and tumor burden. TAE is usually performed by using embolic agents ranging from 40 to 120 µm in size that may be spherical or non-spherical, resorbable or non-resorbable, calibrated or non-calibrated [96,97]. Particles are mixed with iodinated contrast, normal saline solution, and an antibiotic in some cases. Embolization is performed to the endpoint of stasis of flow in the target vessel(s).

7.2.2. Transarterial Chemoembolization (TACE)

In TACE, following the above steps, a microcatheter is used to subselectively catheterize target branches. The goal of subselective catheterization is to maximize the delivery of chemoembolic material to tumor, while minimizing, as much as possible, delivery to the normal liver. Once the catheter position is optimized, conventional or DEB–TACE can be performed under real-time fluoroscopic observation. For conventional TACE, the prescribed chemotherapy is combined with ethiodized oil and contrast and then continuously administered until the peritumoral venous plexus is visualized. However, this approach might be difficult to achieve in big tumors. For DEB–TACE, the endpoint of administration is sub-stasis of flow within the supplying vessel. Caution must be used to prevent over-embolization, resulting in reflux into non-target liver and non-target organs (i.e., bowel, pancreas, and spleen). In cTACE, particles or gelfoam are often delivered after chemoembolic material to prevent “washout” of chemotherapy. If multiple tumors are present, the best approach is to target each individual tumor as selectively as possible. Classically, only one lobe (right or left) is treated in a single setting; however, this approach is variable with the ability to perform subselective targeted delivery of chemoembolic material. If lipiodol or radiodense beads are utilized, non-contrast cone-beam CT or non-contrast CT can be obtained post-delivery to visualize the extent of lipiodol deposition. Figure 3A–D shows CT and MRI of a patient who underwent cTACE. Figure 4A–F shows the CT and angiograms of a patient who underwent DEB–TACE.

7.2.3. Selective Internal Radiation Therapy (SIRT)

With SIRT/Y-90-radioembolization, the added step of technetium-99 m macroaggregated albumin (Tc-99 m MAA) shunt study, or “mapping”, prior to the SIRT is necessary. Following diagnostic angiography and subselective catheterization of supplying tumoral feeders, approximately 4 mCi of Tc-99 m MAA is delivered into the lobe or segment containing the target tumor. The target artery may be identified with the aid of cone-beam CT. After the injection of Tc-99 m MAA, the patient is taken for planar nuclear imaging or SPECT/CT in order to assess for any shunting (specifically to the lung) and assess technical success by visualizing Tc-99 m MAA deposition within targeted tumors. The patient then returns for Y-90 delivery 1–4 weeks following the shunt study (although “same-day” mapping and delivery are also performed at select centers). The prescribed dose of Y-90 depends on multiple factors such as tumor size and number, liver function, and the microparticle of choice. Y-90-radioembolization is performed from the same catheter position utilized for the Tc-99m MAA shunt study. SPECT/CT is then again sometimes obtained post-procedure to assess Y-90 distribution and dose delivery to the targeted tumor. Figure 5A–D shows the CT, SPECT, and angiogram of a patient who underwent segment-two segmentectomy.

7.2.4. Hepatic Artery Infusion (HAI)

In hepatic artery infusion (HAI), chemotherapy is delivered to the tumor by infusing it into the hepatic artery by placing either a surgically implanted pump/port or by placing a percutaneous catheter, which can be connected to an external pump [98,99,100]. For percutaneously placed catheters, the Seldinger technique is used to access the artery and place infusion catheters. Left subclavian and right femoral artery accesses are frequently used. The most commonly used site for access is the common femoral artery, as it is easier to access due to its superficial and less torturous course. In the “fixed-catheter-tip” technique, the distal tip of the catheter is fixed to the gastroduodenal artery, and the chemotherapy is infused to the tumor with the help of a side hole which is positioned into the proper hepatic artery [101]. Another technique which can be used is the “long tapered catheter placement” technique [102]. In this technique, the catheter is positioned distally into the segmental hepatic artery, and the side hole of the catheter is placed at the origin of the proper hepatic artery [103].

8. Mechanism of Action of Transarterial Therapies

8.1. Bland Embolization (TAE)

While the portal vein and hepatic artery offer duality in terms of blood supply, HCC generally relies on hepatic arteries to meet oxygen requirements. TAE involves delivering embolic particles into the arterial supply of targeted tumors, thereby creating hypoxia and subsequent infarction seen on follow-up contrast enhanced imaging (CT or MRI) [104]. TAE is similar to other catheter-directed embolic interventions, such as DEB–TACE or cTACE, but may be used in patients in whom chemotherapy is contraindicated or if chemotherapeutic agents are unavailable.

8.2. Transarterial Chemoembolization (TACE)

Delivering embolic particles within a tumor’s vascular bed results in localized tissue hypoxia; however, the concurrent addition of chemotherapy may have additive antitumor effects [104]. An animal study combining cTACE with radiotherapy has shown promising results; this two-pronged approach could potentially help target cancer cells not reached by Y-90 beads due to arterial inflow limitation [105]. TACE is commonly used in cases of unresectable HCC and is first-line therapy for patients with intermediate-stage HCC [106]. Conventional TACE is referred to as Lipiodol®-based TACE. The two-part process begins with transcatheter delivery of chemotherapy, followed by particle embolization. The most common chemotherapeutic agents include cisplatin (100 mg), doxorubicin (50 mg), and mitomycin C (10 mg). These agents are mixed with lipiodol (Ethiodol) in a 1:1 to >5:1 (lipiodol:chemotherapy) volume ratio [107]. A higher mixing ratio (larger volume of iodized oil) ensures the creation of a water-in-oil-type emulsion. A water-in-oil-type emulsion is advantageous in the selective delivery of an anticancer drug to the tumor. Moreover, cTACE may be repeated serially when there is clear evidence of a viable tumor, with some studies recommending a minimum of three sessions before abandoning cTACE in patients with nonresponding intermediate-stage HCC [108].

8.3. Drug-Eluting Beads–Transarterial Chemoembolization (DEB–TACE)

With the advent of chemotherapy-loaded embolic spheres, an additional modality of chemotherapy delivery became available. Drug-eluting beads dually embolize selective tumor-feeding vessels and deliver chemotherapy in a concentrated fashion [106]. In this sense, DEB–TACE is different than conventional TACE, as the previously described two-part process (cTACE) is exchanged for a single-step modality (DEB–TACE). In randomized trials comparing cTACE and DEB–TACE, the major difference were less post-procedural pain and shorter hospital admission in DEB–TACE, this finding can be explained by the deeper penetration of chemoembolic material into the liver capsule and peritumoral portal venous system with cTACE [109].

8.4. Selective Internal Radiation Therapy (SIRT)

Radiation therapy has been a staple of oncologic therapy, and its usage has typically been reserved for external beam radiation. However, the collateral damage to surrounding parenchyma limits its use in patients with already tenuous liver function. The localized delivery of radiation-emitting microspheres is a means by which to decrease this collateral damage. The most commonly used radioisotopes include yttrium-90 (Y-90) and Holmium-166 (166-HO). SIRT is effective in cases of unresectable tumors or chemotherapy-resistant disease. SIRT, specifically Y-90, has proven to be efficacious in downstaging HCC for purposes of transplant eligibility [110]. Furthermore, 166-HO is a more recently isolated radioisotope and has additional benefits beyond Y-90. Most interesting, perhaps, is that 166-HO microspheres emit high-energy beta particles, in addition to gamma radiation, therefore offering single-photon emission computed tomography compatibility. Moreover, 166-HO microspheres are also paramagnetic and therefore hyper-resonant on MRI (magnetic resonance imaging). SIRT is delivered via two approaches: lobar/sub-lobar and segmentectomy. The lobar/sub-lobar approach is reserved for instances of multifocal disease, whereas a segmentectomy is chosen in cases of solitary tumors measuring 5 cm or less and that are accessible angiographically.
Currently, two manufactured agents are utilized for SIRT: glass beads and resin beads. Glass beads (TheraSphere, Boston Scientific, Marlborough, MA) are 20–30 microns in size and deliver a large activity to the tumor bed (~2500 Bq). However, resin beads (SIR-Spheres, Sirtex, Woburn, MA, USA) are larger in size (20–60 microns) and deliver a lower level of activity to the tumor bed (50 Bq) but produce a greater degree of embolization [111]. The recommended delivery dose for glass beads is a tumoral dose of 400 Gy for successful radiation segmentectomy and 150 Gy for resin bead; however, dosimetry data are continuing to evolve rapidly and are decided based on personalized dosimetry in most cases [112,113].

8.5. Hepatic Artery Infusion (HAI)

HAI is a widely employed alternative to systemic chemotherapy, as it directly delivers chemotherapeutic agents to the tumor feeding vessels and reduces the systemic toxic side effects of these drugs through the first-pass effect of the liver [114]. In a recently conducted phase-three study for large unresectable HCC without macrovascular invasion, comparing HAI with FOLFOX (fluorouracil, leucovorin, and oxaliplatin) vs. TACE, HAI with FOLFOX (median overall survival 13.3 months) showed a significantly improved overall survival rate as compared to TACE (median overall survival of 10.8 months). The infusion was performed once every three weeks for up to six courses [100]. A study comparing treatment with sorafenib alone vs. sorafenib combined with FOLFOX HAI (soraHAIC) reported a tolerable safety profile and improved efficacy with soraHAIC (median overall survival 13.37 months) vs. sorafenib alone (median overall survival of 7.13 months) [115]. The SILIUS trial consisting of 206 patients compared sorafenib alone vs. sorafenib plus low-dose cisplatin and FU HAI. The study found no significant survival benefit from combining sorafenib with low-dose cisplatin and FU (median overall survival with sorafenib alone was 11.8 months vs. overall combination survival of 11.5 months) [116]. The use of sorafenib with HAI compared to sorafenib along with HCCs with major portal vein tumor thrombus was recently studied in a randomized controlled trial which showed a better median overall survival, higher objective response rate, and longer median progression-free survival in the sorafenib with HAI group [117]. However, the survival benefit of combining sorafenib with HAI has largely been shown to be variable, thus necessitating more studies.

9. Contraindications to Transarterial Embolization

9.1. Bland Embolization (TAE) and Transarterial Chemoembolization (TACE)

There are no absolute contraindications to TAE or chemoembolization procedures; however, the presence of various factors can make patient candidacy unfavorable to undergo these procedures. Relative contraindications include contraindications to arteriography; absence of portal venous blood flow; extensive tumor with replacement of both the lobes of liver; decompensated cirrhosis, which is indicated by Child–Pugh class C, or Child–Pugh class B score >8, along with jaundice; clinically overt hepatic encephalopathy, refractory ascites, and/or hepatorenal syndrome; tumor size more than 10 cm; active cardiovascular or lung disease; untreated gastroesophageal varices at the risk of bleeding; bile duct occlusion or incompetent papilla; poor performance status; active systemic infection; contraindications to chemotherapy; or poor tolerance to prior procedures [81,92,118,119].

9.2. Selective Internal Radiation Therapy (SIRT)

Contraindications to SIRT include the history of prior liver irradiation, significant liver dysfunction/decompensation, metastasis to organs other than the liver, pregnancy, irregularities in hepatic venous anatomy that preclude radioembolization, capecitabine treatment three months before the procedure, pathological shunt fraction causing a lung dose of ≥30 Gy in a single application or presence of flow to the gastrointestinal tract from the arterial supply of the tumor that cannot be corrected by transcatheter techniques, and abnormal laboratory values. Although not absolute guidelines, a white blood cell count less than 2500 cc3, neutrophil count less than 1500 cc3, platelet count less than 60,000 cc3, alanine transaminase or aspartate transaminase more than five times the normal value, bilirubin more than 2 mg dL−1, albumin less than 3 mg dL−1, and creatinine more than 2.5 mg dL−1 have been proposed as limits to therapy [120].

10. Post-Procedural Follow-Up

The post-procedural follow-up after the embolization procedure is similar for many patients regardless of the type of transarterial therapy they are receiving. The follow-up includes a clinic visit to reassess the performance status of the patient and ensure symptomatic management; the laboratory tests include a comprehensive metabolic panel, complete blood count, international normalized ratio, normal tumor markers, and multiphasic contrast enhance MRI or CT of the abdomen with intravenous iodine-based contrast for follow-up. Follow-up after liver-directed therapy is recommended at 1, 3, 6, and 12 months in the first year after the procedure and every 6 months thereafter. Of note, post-treatment imaging of HCC following radioembolization is recommended at 6–8 weeks following delivery, as the localized post-radioembolization parenchymal enhancement imitates the treated disease, making response difficult or impossible to assess before these changes evolve. Additionally, the follow-up schedule should be tailored for each patient according to their treatment goals, transplant status, local imaging availability, etc. [121].

11. Complications

11.1. Bland Embolization (TAE) and Transarterial Chemoembolization (TACE)

The most common complication associated with embolization is postembolization syndrome, which can be seen in up to 90 percent of patients and can present with fatigue, right upper quadrant pain, low-grade fever, nausea, vomiting, and ileus [122,123,124,125]. Symptoms due to the post-embolization syndrome are generally self-limiting, and recovery is often seen within 7 to 10 days [126]. Other less common but serious complications associated with embolization are most often related to treatment-induced ischemic damage, leading to liver failure [127,128,129,130], hepatic abscess [131], acute cholecystitis, biliary duct injury [130,132], gastroduodenal ulceration [126,130], renal dysfunction [130], pulmonary lipoid embolization [133,134], cerebral lipoid embolization [135], interstitial pneumonia [136], tumor lysis syndrome [130,137], and risk of reactivation of hepatitis B infection [138,139].

11.2. Selective Internal Radiation Therapy (SIRT)

Similar to cTACE, post-embolization syndrome is the most common complication associated with SIRT; however, it is less severe in patients undergoing SIRT, and the incidence is lower [140,141,142,143]. Other complications associated with SIRT include hepatic dysfunction, which can manifest as liver failure; radiation-induced liver disease (RILD), which can be seen in 5 to 23 percent of patients [141,144]; hepatic fibrosis [145]; portal hypertension [145,146]; radiation pneumonitis in less than 1 percent of patients [141]; gastric or duodenal injury in less than 5 percent of patients; and lymphopenia [147]. Of note, delayed hepatotoxicity can be seen in 13% of patients following SIRT, especially in patients with a tumor burden of >50% and cirrhosis [148].

12. Outcomes

12.1. Imaging Response Criteria

The main goal of treatment is to improve the survival of patients with cancers, and, therefore, it is important to detect the change in tumor burden in patients undergoing treatment, as it has been found to be associated with increased survival. The tumor burden is evaluated with the help of standard response criteria, which require critical interrogation and cross-verification. A number of criteria have been developed to assess the treatment response in patients undergoing treatment [149,150,151]. These criteria involve one-, two-, and three-dimensional methods that can be used to measure the response after treatment [152]. Each of these methods is further subdivided into two or more categories based on the ratio of total size to the size of the enhancing component. During the Response Evaluation Criteria in Solid Tumors (RECIST) measurements, the sum of the longest one-dimensional diameter is evaluated tumor response; however, now the commonly used criteria are the modified RECIST (mRECIST) criteria, which measure the changes in tumor enhancement that can serve as a biomarker for the tumor viability [153,154]. The World Health Organization (WHO) has introduced an evaluation system for solid liver tumors in which the diameters of the tumor lesions are measured and then multiplied to evaluate the change in lesion size. In contrast to the WHO evaluation system, the European Association for the Study of the Liver (EASL) recommends measuring changes in contrast uptake of the tumor tissue to measure tumor response [150,155]. However, the criteria based on one- or two-dimensional methods are flawed due to the lack of reproducibility and inaccuracy when assessing necrotic or heterogeneous tumor lesions [156]. Due to these limitations, three-dimensional quantitative image analysis techniques have been developed, which are said to provide a reproducible assessment that is biologically accurate and provides a more clinical predictable tumor evaluation [157]. Volumetric analysis (vRECIST) includes the measurement of whole tumor volume, and therefore the change here reflects the true extent and distribution of the tumor tissue [94,158]. The quantitative EASL (qEASL) assesses changes in enhancing tumor volume and therefore goes beyond the boundaries calculated in vRECIST [94,157,159,160]. The PET Response Criteria in Solid Tumors (PERCIST) is another set of response assessment criterion developed to assess tumor response by using PET scans [161]. A liver-tissue classification using 3D multi-parameter MRI-based deep neural networks has been developed to better delineate the liver tissue for better TACE targeting has shown promising results [162]. An artificial intelligence concept using the qEASL criteria was developed to predict TACE response (overall accuracy of 78%) [163]. A study conducted by Miszczuk et al. reports that lipiodol can be a potential imaging biomarker after cTACE for tumor response. Patients with a higher overall lipiodol coverage showed higher response rates (p < 0.05) upon 30-day follow-up imaging studies [164,165,166,167]. Although the mainstay of tumoral response evaluation is currently the assessment of imaging response (such as with mRECIST) and chemical biomarker response (AFP), precision-medicine-based studies have recently gained momentum, thus allowing clinicians to individualize treatment plans based on genomics [168]. For example, a recent genome-based study looking at patients who underwent TACE for HCC showed that an increased PMK2 expression post-TACE was associated with attenuated survival [169].

12.2. Overall Survival, Progression-Free Survival, and Hepatic Progression-Free Survival

Overall survival (OS) is the most commonly used primary endpoint in oncology and clinical trials reporting results utilizing first- and second-line systemic therapies to treat advanced HCC [170]. However, other surrogate endpoints have been identified, such as objective response rate (ORR) and progression-free survival (PFS), which have helped better defined clinical outcomes and have led to accelerated regulatory approval of drugs and therapies [171,172]. OS is defined as the time after the treatment of a disease for which the patient lives. PFS is the length of time during and after the treatment of a disease when the patient is alive and the disease has not worsened [170]. In a clinical trial, measuring PFS is one way of determining how well a new treatment works and how durable its treatment effect is [170]. Likewise, hepatic progression-free survival (HPFS) is calculated from the first regional treatment until the first date of documented progression in the liver and serves as a surrogate of treatment [173].
TAE has been investigated in multiple studies, and 1-year reported overall survival of patients with nonresectable HCC ranges from 42 to 86%, whereas 2-year OS ranges from 25 to 51% [174,175,176,177,178,179]. In a meta-analysis conducted by Marelli et al., TAE was compared to TACE, and no survival benefit was seen in patients who underwent TACE compared to those who underwent TAE [130]. The use of smaller calibrated microspheres has been shown to lead to better outcomes by causing improved tumor-control rates. The use of 300–500 μm beads had an OS of 15.1 months compared to 11.1 months in patients who underwent TAE with 500–700 μm beads [180]. In terms of recurrent HCC, the use of TAE has shown to have a median OS of 46 months, with 1-, 2-, and 5-year survival rates of 86%, 74%, and 47%, respectively [181].
TACE has been shown to improve the OS in patients with unresectable HCC compared to best supportive care in patients with well-preserved liver function [178,182]. The most extensive series of TACE has been reported from Japan, which studied the outcomes of 8510 patients undergoing TACE [183]. In this series, the median OS was 34 months [183]. In the first head-to-head trial comparing TACE with DEB–TACE, no difference in median OS was found between the two groups [184]. Subsequently, multiple randomized controlled trials have been performed to compare TACE and DEB–TACE; however, no significant difference in OS has been found [184,185,186]. Studies have also reported no significant difference in OS or PFS based on the chemotherapeutic regimen used for TACE [187,188,189]. Per the TACTICS trial, sorafenib combined with TACE significantly increases the PFS as compared to TACE alone in unresectable HCC (25.2 vs. 13.5 months, p = 0.006) [190]. The presence of portal vein thrombosis is not a contraindication to TACE. A study evaluating the safety of TACE in patients with portal vein thrombosis showed no evidence of TACE-related hepatic infarction or acute liver failure and a 0% 30-day mortality rate [191].
In the largest retrospective series of 345 HAI procedures, patients undergoing the procedure had significantly worse outcomes if they had more advanced disease [192]. The median OS was found to be 28.6 months in another study where 253 HAI procedures were investigated [193]. However, HAI has shown to have a lower OS rates when compared to TACE [194].
Patients undergoing SIRT with unresectable HCC have a median OS of up to 20.5 months [195,196,197,198,199]. In patients with unresectable HCC without portal vein thrombosis, SIRT and TACE have been shown to have a similar median OS rate (20.5 months vs. 17.5 months, respectively) [199]. In patients with portal vein thrombosis, the use of SIRT has shown to have a median OS ranging from 5.6 to 17 months depending on the extent of portal vein involvement, functional status of the patient, and underlying liver dysfunction [195,196,198,200]. The use of SIRT compared to sorafenib in patients with advanced HCC has not been shown to have improved outcomes [201,202]. Studies reporting outcomes after SIRT segmentectomy have reported significant improvements in time-to-progression, complete response rate, local tumor control, and PFS compared to TACE [112,203,204,205]. More recently, a multiregional study comparing SIRT with TACE found no significant difference between the two in downgrading the disease prior to liver transplantation [206]. The outcomes of radiation segmentectomy have been shown to be similar to ablation and surgical resection in patients with tumors of 3 cm or smaller [203].
In a meta-analysis including 55 randomized controlled trials comparing the effectiveness of different transarterial therapies in patients with HCC, all the embolization techniques provided a significant increase in survival when compared to the best supportive care. However, there was no significant increased benefit of TACE, DEB–TACE, SIRT, or TACE combined with systemic therapy when compared to BE alone. The combination of TACE with radiotherapy or local ablation showed the best survival. SIRT was found to cause the fewest side effects, whereas patients receiving TACE combined with systemic therapy had the highest number of reported complications [207].

13. New Developments and Future Directions

13.1. Newer Drug-Eluting Beads

Drug-eluting beads (DEBs) are beads loaded with different chemotherapeutic agents. A number of different DEBs have been successfully developed and have been used in the management of hypervascular HCCs [208,209]. These beads are composed of biocompatible polymers, which can bind chemotherapeutic agents after the formation of covalent bonds [210]. As pharmacokinetics of these DEBs are better studied and further refined, the ability to deliver chemotherapeutics more efficiently and more effectively will also undoubtedly improve, potentially improving outcomes while also decreasing side effects and complications. The therapeutic potential of DEBs in precision oncology is perhaps one of the most exciting areas of current and future development in interventional oncology.

13.2. Radiopaque Beads

In recent years, imageable radiopaque beads have been developed, which are made radiopaque by incorporating a radio-absorber, such as iodine, zinc, tantalum, bismuth, or barium, into the beads’ manufacturing process [211,212,213,214,215]. Radiopaque beads provide real-time feedback during the embolization procedure and provide invaluable information during follow-up imaging [216,217]. A new investigational radiopaque SIRT embolic has been developed with a radiopaque bland glass microsphere which has shown strong fluoroscopic and CBCT radiopacity in preclinical testing. The use of these microspheres will allow for intra-procedural real-time confirmation of tumor targeting [218].

13.3. Immunoembolization

Immunoembolization is an exciting new area which involves the delivery of systemic immunotherapy agents such as granulocyte-macrophage colony-stimulating factor (GM-CSF) into the arteries supplying the HCC [219]. Immunoembolization is hypothesized to work by producing necrosis of the HCC tumor cells, thus leading to the stimulation of the antigen-presenting cells (APCs) on the tumor surface. These APCs can then increase the update of tumor antigens, which are released from the necrotic tumor cells, and lead to activation of the T cells present in the tumor microenvironment [220,221,222,223]. The development of this inflammatory response can lead to destruction of the tumor cells which did not undergo necrosis during the initial embolization. Additionally, local immune system stimulation can generate a systemic immune response against the tumor cells, which can lead to suppression of extrahepatic metastasis and prevent further tumor growth and dissemination [224,225].

13.4. Nanoparticles

A lot of focus in the recent years has been put into the development of nanoparticles that can be used for the diagnosis and treatment of patients with HCC [226]. The introduction of novel theranostic nanoparticles into the interventional radiology field can help diagnose, localize, and stage disease. Theranostic nanoparticles may also provide invaluable information regarding treatment response [227,228]. Such nanoparticles are designed to carry therapeutic agents to the tumor via molecular and/or external stimuli [229,230,231].

13.5. Combination with Systemic Therapies

The use of systemic therapies in combination with transarterial embolization techniques is an area of significant clinical and research focus [232]. Some of the many targeted systemic therapies that have been studied include, but are not limited to, MAPK Inhibitors, including sunitinib, sorafenib, imatinib, cabozantinib, lenvatinib, and selumetinib [233,234]. Others include immunotherapeutic agents such as ipilimumab, nivolumab, and pembrolizumab [235,236,237]. A majority of the studies have shown positive results, with improved outcomes and acceptable safety profiles [238].

13.6. Combination with Percutaneous Therapies

Combining embolization techniques with ablation for hepatocellular may enhance the therapeutic benefit of each and result in improved patient survival [239]. A majority of studies have demonstrated the safety and efficacy of a two-step or single-session transarterial and percutaneous ablation treatment for unresectable hepatic metastases, specifically for lesions >3 cm in diameter [240].

13.7. Pre-Operative TACE

TACE can be used as neoadjuvant therapy for the treatment of large HCCs prior to surgical resection [241]. Pre-operative TACE can also facilitate curative resection in patients with large HCCs who are otherwise not suitable candidates for liver resection [242,243]. However, the outcomes for the use of pre-operative TACE are mixed, with some studies showing improved survival outcomes and others not [244,245,246].

14. Conclusions

Transarterial therapies remain an important and increasingly utilized therapeutic option in the management of patients with HCC. They are unique, as they provide highly effective tumor control while preserving normal liver parenchyma and reducing toxicity. Over the last couple of decades, the evidence to support the use of transarterial therapies for managing HCC has grown to where they are now within commonly utilized treatment guidelines. A multidisciplinary approach for selecting appropriate patients and tumors is critical to optimizing clinical outcomes and technical success of the various transarterial therapies that are currently available. Advances in therapeutics, pharmacokinetics, drug delivery, and imaging will all serve to further advance transarterial therapies for HCC in the future.

Author Contributions

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

Funding

This research received no external funding.

Conflicts of Interest

T.G. serves on the editorial board of RadioGraphics and Pediatrics Oncall. T.G. receives conference travel support from Siemens Healtineers. J.C. is supported by grants from the Society of Interventional Oncology, Guerbet Pharmaceuticals, Philips Healthcare, Boston Scientific, Yale Center for Clinical Investigation, and the National Institutes of Health (R01CA206180); reports personal fees from Guerbet Pharmaceuticals, Bayer, AstraZeneca and Philips Healthcare; and reports nonfinancial support from Guerbet Pharmaceuticals outside the submitted work. N.N. is consultant to Embolx, RenovoRx, and CAPS Medical.

References

  1. Gyves, J.W.; Ziessman, H.A.; Ensminger, W.D.; Thrall, J.H.; Niederhuber, J.E.; Keyes, J.W.; Walker, S. Definition of Hepatic Tumor Microcirculation by Single Photon Emission Computerized Tomography (SPECT). J. Nucl. Med. 1984, 25, 972–977. [Google Scholar] [PubMed]
  2. Bierman, H.R.; Byron, R.L.; Kelley, K.H.; Grady, A. Studies on the Blood Supply of Tumors in Man. III. Vascular Patterns of the Liver by Hepatic Arteriography in Vivo. J. Natl. Cancer Inst. 1951, 12, 107–131. [Google Scholar] [PubMed]
  3. Liapi, E.; Geschwind, J.-F.H. Intra-Arterial Therapies for Hepatocellular Carcinoma: Where Do We Stand? Ann. Surg. Oncol. 2010, 17, 1234–1246. [Google Scholar] [CrossRef] [PubMed]
  4. Benson, A.B.; D’Angelica, M.I.; Abbott, D.E.; Anaya, D.A.; Anders, R.; Are, C.; Bachini, M.; Borad, M.; Brown, D.; Burgoyne, A.; et al. Hepatobiliary Cancers, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2021, 19, 541–565. [Google Scholar] [CrossRef]
  5. Reig, M.; Forner, A.; Rimola, J.; Ferrer-Fàbrega, J.; Burrel, M.; Garcia-Criado, Á.; Kelley, R.K.; Galle, P.R.; Mazzaferro, V.; Salem, R.; et al. BCLC Strategy for Prognosis Prediction and Treatment Recommendation: The 2022 Update. J. Hepatol. 2021, 76, 681–693. [Google Scholar] [CrossRef]
  6. OPTN/UNOS Policy Notice Modification to Hepatocellular Carcinoma (HCC) Extension Criteria. Available online: https://optn.transplant.hrsa.gov/media/2411/modification-to-hcc-auto-approval-criteria_policy-notice.pdf. (accessed on 28 April 2022).
  7. Fujiki, M.; Aucejo, F.; Kim, R. General Overview of Neo-Adjuvant Therapy for Hepatocellular Carcinoma before Liver Transplantation: Necessity or Option?: Neo-Adjuvant Therapy for Hepatocellular Carcinoma. Liver Int. 2011, 31, 1081–1089. [Google Scholar] [CrossRef]
  8. Pompili, M.; Mirante, V.G.; Rondinara, G.; Fassati, L.R.; Piscaglia, F.; Agnes, S.; Covino, M.; Ravaioli, M.; Fagiuoli, S.; Gasbarrini, G.; et al. Percutaneous Ablation Procedures in Cirrhotic Patients with Hepatocellular Carcinoma Submitted to Liver Transplantation: Assessment of Efficacy at Explant Analysis and of Safety for Tumor Recurrence. Liver Transplant. 2005, 11, 1117–1126. [Google Scholar] [CrossRef]
  9. Mazzaferro, V.; Battiston, C.; Perrone, S.; Pulvirenti, A.; Regalia, E.; Romito, R.; Sarli, D.; Schiavo, M.; Garbagnati, F.; Marchianò, A.; et al. Radiofrequency Ablation of Small Hepatocellular Carcinoma in Cirrhotic Patients Awaiting Liver Transplantation: A Prospective Study. Ann. Surg. 2004, 240, 900–909. [Google Scholar] [CrossRef]
  10. Yao, F.Y.; Bass, N.M.; Nikolai, B.; Merriman, R.; Davern, T.J.; Kerlan, R.; Ascher, N.L.; Roberts, J.P. A Follow-up Analysis of the Pattern and Predictors of Dropout from the Waiting List for Liver Transplantation in Patients with Hepatocellular Carcinoma: Implications for the Current Organ Allocation Policy. Liver Transplant. 2003, 9, 684–692. [Google Scholar] [CrossRef]
  11. Tsochatzis, E.; Garcovich, M.; Marelli, L.; Papastergiou, V.; Fatourou, E.; Rodriguez-Peralvarez, M.L.; Germani, G.; Davies, N.; Yu, D.; Luong, T.V.; et al. Transarterial Embolization as Neo-Adjuvant Therapy Pretransplantation in Patients with Hepatocellular Carcinoma. Liver Int. 2013, 33, 944–949. [Google Scholar] [CrossRef]
  12. 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] [PubMed]
  13. Richard, H.M.; Silberzweig, J.E.; Mitty, H.A.; Lou, W.Y.; Ahn, J.; Cooper, J.M. Hepatic Arterial Complications in Liver Transplant Recipients Treated with Pretransplantation Chemoembolization for Hepatocellular Carcinoma. Radiology 2000, 214, 775–779. [Google Scholar] [CrossRef] [PubMed]
  14. Graziadei, I.W.; Sandmueller, H.; Waldenberger, P.; Koenigsrainer, A.; Nachbaur, K.; Jaschke, W.; Margreiter, R.; Vogel, W. Chemoembolization Followed by Liver Transplantation for Hepatocellular Carcinoma Impedes Tumor Progression While on the Waiting List and Leads to Excellent Outcome. Liver Transplant. 2003, 9, 557–563. [Google Scholar] [CrossRef] [PubMed]
  15. Hayashi, P.H.; Ludkowski, M.; Forman, L.M.; Osgood, M.; Johnson, S.; Kugelmas, M.; Trotter, J.F.; Bak, T.; Wachs, M.; Kam, I.; et al. Hepatic Artery Chemoembolization for Hepatocellular Carcinoma in Patients Listed for Liver Transplantation. Am. J. Transplant. 2004, 4, 782–787. [Google Scholar] [CrossRef] [PubMed]
  16. Nicolini, D.; Svegliati-Baroni, G.; Candelari, R.; Mincarelli, C.; Mandolesi, A.; Bearzi, I.; Mocchegiani, F.; Vecchi, A.; Montalti, R.; Benedetti, A.; et al. Doxorubicin-Eluting Bead vs Conventional Transcatheter Arterial Chemoembolization for Hepatocellular Carcinoma before Liver Transplantation. World J. Gastroenterol 2013, 19, 5622–5632. [Google Scholar] [CrossRef]
  17. Kulik, L.M.; Atassi, B.; van Holsbeeck, L.; Souman, T.; Lewandowski, R.J.; Mulcahy, M.F.; Hunter, R.D.; Nemcek, A.A.; Abecassis, M.M.; Haines, K.G.; et al. Yttrium-90 Microspheres (TheraSphere) Treatment of Unresectable Hepatocellular Carcinoma: Downstaging to Resection, RFA and Bridge to Transplantation. J. Surg. Oncol. 2006, 94, 572–586. [Google Scholar] [CrossRef]
  18. Sandroussi, C.; Dawson, L.A.; Lee, M.; Guindi, M.; Fischer, S.; Ghanekar, A.; Cattral, M.S.; McGilvray, I.D.; Levy, G.A.; Renner, E.; et al. Radiotherapy as a Bridge to Liver Transplantation for Hepatocellular Carcinoma. Transplant. Int. 2010, 23, 299–306. [Google Scholar] [CrossRef]
  19. Lu, L.; Zeng, J.; Wen, Z.; Tang, C.; Xu, N. Transcatheter Arterial Chemoembolisation Followed by Three-Dimensional Conformal Radiotherapy versus Transcatheter Arterial Chemoembolisation Alone for Primary Hepatocellular Carcinoma in Adults. Cochrane Database Syst. Rev. 2019, 2, CD012244. [Google Scholar] [CrossRef]
  20. Toso, C.; Mentha, G.; Kneteman, N.M.; Majno, P. The Place of Downstaging for Hepatocellular Carcinoma. J. Hepatol 2010, 52, 930–936. [Google Scholar] [CrossRef]
  21. Yao, F.Y.; Fidelman, N. Reassessing the Boundaries of Liver Transplantation for Hepatocellular Carcinoma: Where Do We Stand with Tumor down-Staging? Hepatology 2016, 63, 1014–1025. [Google Scholar] [CrossRef]
  22. Parikh, N.D.; Waljee, A.K.; Singal, A.G. Downstaging Hepatocellular Carcinoma: A Systematic Review and Pooled Analysis. Liver Transplant. 2015, 21, 1142–1152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Kulik, L.; Heimbach, J.K.; Zaiem, F.; Almasri, J.; Prokop, L.J.; Wang, Z.; Murad, M.H.; Mohammed, K. Therapies for Patients with Hepatocellular Carcinoma Awaiting Liver Transplantation: A Systematic Review and Meta-Analysis. Hepatology 2018, 67, 381–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. 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] [Green Version]
  25. European Association for the Study of the Liver. Electronic address: [email protected]; European Association for the Study of the Liver EASL Clinical Practice Guidelines: Management of Hepatocellular Carcinoma. J. Hepatol. 2018, 69, 182–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Bruix, J.; Qin, S.; Merle, P.; Granito, A.; Huang, Y.-H.; Bodoky, G.; Pracht, M.; Yokosuka, O.; Rosmorduc, O.; Breder, V.; et al. Regorafenib for Patients with Hepatocellular Carcinoma Who Progressed on Sorafenib Treatment (RESORCE): A Randomised, Double-Blind, Placebo-Controlled, Phase 3 Trial. Lancet 2017, 389, 56–66. [Google Scholar] [CrossRef] [Green Version]
  27. Zhu, A.X.; Kang, Y.-K.; Yen, C.-J.; Finn, R.S.; Galle, P.R.; Llovet, J.M.; Assenat, E.; Brandi, G.; Pracht, M.; Lim, H.Y.; et al. Ramucirumab after Sorafenib in Patients with Advanced Hepatocellular Carcinoma and Increased α-Fetoprotein Concentrations (REACH-2): A Randomised, Double-Blind, Placebo-Controlled, Phase 3 Trial. Lancet Oncol 2019, 20, 282–296. [Google Scholar] [CrossRef]
  28. Abou-Alfa, G.K.; Meyer, T.; Cheng, A.-L.; El-Khoueiry, A.B.; Rimassa, L.; Ryoo, B.-Y.; Cicin, I.; Merle, P.; Chen, Y.; Park, J.-W.; et al. Cabozantinib in Patients with Advanced and Progressing Hepatocellular Carcinoma. N. Engl. J. Med. 2018, 379, 54–63. [Google Scholar] [CrossRef]
  29. Finn, R.S.; Ryoo, B.-Y.; Merle, P.; Kudo, M.; Bouattour, M.; Lim, H.Y.; Breder, V.; Edeline, J.; Chao, Y.; Ogasawara, S.; et al. Pembrolizumab As Second-Line Therapy in Patients With Advanced Hepatocellular Carcinoma in KEYNOTE-240: A Randomized, Double-Blind, Phase III Trial. J. Clin. Oncol. 2020, 38, 193–202. [Google Scholar] [CrossRef]
  30. Yau, T.; Park, J.W.; Finn, R.S.; Cheng, A.-L.; Mathurin, P.; Edeline, J.; Kudo, M.; Han, K.-H.; Harding, J.J.; Merle, P.; et al. CheckMate 459: A Randomized, Multi-Center Phase III Study of Nivolumab (NIVO) vs Sorafenib (SOR) as First-Line (1L) Treatment in Patients (Pts) with Advanced Hepatocellular Carcinoma (AHCC). Ann. Oncol. 2019, 30, v874–v875. [Google Scholar] [CrossRef]
  31. Lencioni, R.A.; Allgaier, H.-P.; Cioni, D.; Olschewski, M.; Deibert, P.; Crocetti, L.; Frings, H.; Laubenberger, J.; Zuber, I.; Blum, H.E.; et al. Small Hepatocellular Carcinoma in Cirrhosis: Randomized Comparison of Radio-Frequency Thermal Ablation versus Percutaneous Ethanol Injection. Radiology 2003, 228, 235–240. [Google Scholar] [CrossRef]
  32. Huang, J.; Yan, L.; Cheng, Z.; Wu, H.; Du, L.; Wang, J.; Xu, Y.; Zeng, Y. A Randomized Trial Comparing Radiofrequency Ablation and Surgical Resection for HCC Conforming to the Milan Criteria. Ann. Surg. 2010, 252, 903–912. [Google Scholar] [CrossRef] [PubMed]
  33. Huang, G.-T.; Lee, P.-H.; Tsang, Y.-M.; Lai, M.-Y.; Yang, P.-M.; Hu, R.-H.; Chen, P.-J.; Kao, J.-H.; Sheu, J.-C.; Lee, C.-Z.; et al. Percutaneous Ethanol Injection versus Surgical Resection for the Treatment of Small Hepatocellular Carcinoma: A Prospective Study. Ann. Surg. 2005, 242, 36–42. [Google Scholar] [CrossRef] [PubMed]
  34. Ng, K.K.C.; Chok, K.S.H.; Chan, A.C.Y.; Cheung, T.T.; Wong, T.C.L.; Fung, J.Y.Y.; Yuen, J.; Poon, R.T.P.; Fan, S.T.; Lo, C.M. Randomized Clinical Trial of Hepatic Resection versus Radiofrequency Ablation for Early-Stage Hepatocellular Carcinoma. Br. J. Surg. 2017, 104, 1775–1784. [Google Scholar] [CrossRef]
  35. Shibata, T.; Iimuro, Y.; Yamamoto, Y.; Maetani, Y.; Ametani, F.; Itoh, K.; Konishi, J. Small Hepatocellular Carcinoma: Comparison of Radio-Frequency Ablation and Percutaneous Microwave Coagulation Therapy. Radiology 2002, 223, 331–337. [Google Scholar] [CrossRef] [PubMed]
  36. Zhang, L.; Wang, N.; Shen, Q.; Cheng, W.; Qian, G.-J. Therapeutic Efficacy of Percutaneous Radiofrequency Ablation versus Microwave Ablation for Hepatocellular Carcinoma. PLoS ONE 2013, 8, e76119. [Google Scholar] [CrossRef] [PubMed]
  37. Ramsey, D.E.; Kernagis, L.Y.; Soulen, M.C.; Geschwind, J.-F.H. Chemoembolization of Hepatocellular Carcinoma. J. Vasc. Interv. Radiol. 2002, 13, S211–S221. [Google Scholar] [CrossRef]
  38. Ibrahim, S.-M.; Lewandowski, R.-J.; Sato, K.-T.; Gates, V.-L.; Kulik, L.; Mulcahy, M.-F.; Ryu, R.-K.; Omary, R.-A.; Salem, R. Radioembolization for the Treatment of Unresectable Hepatocellular Carcinoma: A Clinical Review. World J. Gastroenterol 2008, 14, 1664–1669. [Google Scholar] [CrossRef]
  39. Hawkins, M.A.; Dawson, L.A. Radiation Therapy for Hepatocellular Carcinoma: From Palliation to Cure. Cancer 2006, 106, 1653–1663. [Google Scholar] [CrossRef]
  40. Hoffe, S.E.; Finkelstein, S.E.; Russell, M.S.; Shridhar, R. Nonsurgical Options for Hepatocellular Carcinoma: Evolving Role of External Beam Radiotherapy. Cancer Control 2010, 17, 100–110. [Google Scholar] [CrossRef] [Green Version]
  41. Hasan, S.; Abel, S.; Verma, V.; Webster, P.; Arscott, W.T.; Wegner, R.E.; Kirichenko, A.; Simone II, C.B. Proton Beam Therapy versus Stereotactic Body Radiotherapy for Hepatocellular Carcinoma: Practice Patterns, Outcomes, and the Effect of Biologically Effective Dose Escalation. J. Gastrointest. Oncol. 2019, 10, 999–1009. [Google Scholar] [CrossRef]
  42. Andolino, D.L.; Johnson, C.S.; Maluccio, M.; Kwo, P.; Tector, A.J.; Zook, J.; Johnstone, P.A.S.; Cardenes, H.R. Stereotactic Body Radiotherapy for Primary Hepatocellular Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2011, 81, e447–e453. [Google Scholar] [CrossRef] [PubMed]
  43. Huang, W.-Y.; Jen, Y.-M.; Lee, M.-S.; Chang, L.-P.; Chen, C.-M.; Ko, K.-H.; Lin, K.-T.; Lin, J.-C.; Chao, H.-L.; Lin, C.-S.; et al. Stereotactic Body Radiation Therapy in Recurrent Hepatocellular Carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2012, 84, 355–361. [Google Scholar] [CrossRef] [PubMed]
  44. Kang, J.-K.; Kim, M.-S.; Cho, C.K.; Yang, K.M.; Yoo, H.J.; Kim, J.H.; Bae, S.H.; Jung, D.H.; Kim, K.B.; Lee, D.H.; et al. Stereotactic Body Radiation Therapy for Inoperable Hepatocellular Carcinoma as a Local Salvage Treatment after Incomplete Transarterial Chemoembolization. Cancer 2012, 118, 5424–5431. [Google Scholar] [CrossRef] [PubMed]
  45. Bujold, A.; Massey, C.A.; Kim, J.J.; Brierley, J.; Cho, C.; Wong, R.K.S.; Dinniwell, R.E.; Kassam, Z.; Ringash, J.; Cummings, B.; et al. Sequential Phase I and II Trials of Stereotactic Body Radiotherapy for Locally Advanced Hepatocellular Carcinoma. J. Clin. Oncol. 2013, 31, 1631–1639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Lucatelli, P.; Burrel, M.; Guiu, B.; de Rubeis, G.; van Delden, O.; Helmberger, T. CIRSE Standards of Practice on Hepatic Transarterial Chemoembolisation. Cardiovasc. Interv. Radiol. 2021, 44, 1851–1867. [Google Scholar] [CrossRef] [PubMed]
  47. Yu, S.J. A Concise Review of Updated Guidelines Regarding the Management of Hepatocellular Carcinoma around the World: 2010–2016. Clin. Mol. Hepatol. 2016, 22, 7–17. [Google Scholar] [CrossRef]
  48. Torzilli, G.; Belghiti, J.; Kokudo, N.; Takayama, T.; Capussotti, L.; Nuzzo, G.; Vauthey, J.-N.; Choti, M.A.; De Santibanes, E.; Donadon, M.; et al. A Snapshot of the Effective Indications and Results of Surgery for Hepatocellular Carcinoma in Tertiary Referral Centers: Is It Adherent to the EASL/AASLD Recommendations?: An Observational Study of the HCC East-West Study Group. Ann. Surg. 2013, 257, 929–937. [Google Scholar] [CrossRef]
  49. Oken, M.M.; Creech, R.H.; Tormey, D.C.; Horton, J.; Davis, T.E.; McFadden, E.T.; Carbone, P.P. Toxicity and Response Criteria of the Eastern Cooperative Oncology Group. Am. J. Clin. Oncol. 1982, 5, 649–655. [Google Scholar] [CrossRef]
  50. Tonko, S.; Dufour, J.-F. Hepatology scores. Ther. Umsch. 2013, 70, 577–579. [Google Scholar] [CrossRef]
  51. Heimbach, J.K.; Kulik, L.M.; Finn, R.S.; Sirlin, C.B.; Abecassis, M.M.; Roberts, L.R.; Zhu, A.X.; Murad, M.H.; Marrero, J.A. AASLD Guidelines for the Treatment of Hepatocellular Carcinoma. Hepatology 2018, 67, 358–380. [Google Scholar] [CrossRef] [Green Version]
  52. Johnson, P.J.; Berhane, S.; Kagebayashi, C.; Satomura, S.; Teng, M.; Reeves, H.L.; O’Beirne, J.; Fox, R.; Skowronska, A.; Palmer, D.; et al. Assessment of Liver Function in Patients with Hepatocellular Carcinoma: A New Evidence-Based Approach-the ALBI Grade. J. Clin. Oncol. 2015, 33, 550–558. [Google Scholar] [CrossRef] [PubMed]
  53. Na, S.K.; Yim, S.Y.; Suh, S.J.; Jung, Y.K.; Kim, J.H.; Seo, Y.S.; Yim, H.J.; Yeon, J.E.; Byun, K.S.; Um, S.H. ALBI versus Child-Pugh Grading Systems for Liver Function in Patients with Hepatocellular Carcinoma. J. Surg. Oncol. 2018, 117, 912–921. [Google Scholar] [CrossRef] [PubMed]
  54. Hickey, R.; Mouli, S.; Kulik, L.; Desai, K.; Thornburg, B.; Ganger, D.; Baker, T.; Abecassis, M.; Ralph Kallini, J.; Gabr, A.; et al. Independent Analysis of Albumin-Bilirubin Grade in a 765-Patient Cohort Treated with Transarterial Locoregional Therapy for Hepatocellular Carcinoma. J. Vasc. Interv. Radiol. 2016, 27, 795–802. [Google Scholar] [CrossRef] [PubMed]
  55. Waked, I.; Berhane, S.; Toyoda, H.; Chan, S.L.; Stern, N.; Palmer, D.; Tada, T.; Yeo, W.; Mo, F.; Bettinger, D.; et al. Transarterial Chemo-Embolisation of Hepatocellular Carcinoma: Impact of Liver Function and Vascular Invasion. Br. J. Cancer 2017, 116, 448–454. [Google Scholar] [CrossRef]
  56. Huo, T.-I.; Liu, P.-H.; Hsu, C.-Y. ALBI Score as a Novel Tool in Staging and Treatment Planning for Hepatocellular Carcinoma: Is It Sufficient? Liver Cancer 2017, 6, 375–376. [Google Scholar] [CrossRef]
  57. Liu, P.-H.; Hsu, C.-Y.; Hsia, C.-Y.; Lee, Y.-H.; Chiou, Y.-Y.; Huang, Y.-H.; Lee, F.-Y.; Lin, H.-C.; Hou, M.-C.; Huo, T.-I. ALBI and PALBI Grade Predict Survival for HCC across Treatment Modalities and BCLC Stages in the MELD Era. J. Gastroenterol. Hepatol. 2017, 32, 879–886. [Google Scholar] [CrossRef]
  58. Petrie, H.T.; Klassen, L.W.; Kay, H.D. Inhibition of Human Cytotoxic T Lymphocyte Activity in Vitro by Autologous Peripheral Blood Granulocytes. J. Immunol. 1985, 134, 230–234. [Google Scholar]
  59. el-Hag, A.; Clark, R.A. Immunosuppression by Activated Human Neutrophils. Dependence on the Myeloperoxidase System. J. Immunol. 1987, 139, 2406–2413. [Google Scholar]
  60. Niemi, K.M.; Kero, M.; Kanerva, L.; Mattila, R. Epidermolysis Bullosa Simplex. A New Histologic Subgroup. Arch. Dermatol. 1983, 119, 138–141. [Google Scholar] [CrossRef]
  61. Wang, C.; Wang, M.; Zhang, X.; Zhao, S.; Hu, J.; Han, G.; Liu, L. The Neutrophil-to-Lymphocyte Ratio Is a Predictive Factor for the Survival of Patients with Hepatocellular Carcinoma Undergoing Transarterial Chemoembolization. Ann. Transl. Med. 2020, 8, 541. [Google Scholar] [CrossRef]
  62. Malinchoc, M.; Kamath, P.S.; Gordon, F.D.; Peine, C.J.; Rank, J.; ter Borg, P.C. A Model to Predict Poor Survival in Patients Undergoing Transjugular Intrahepatic Portosystemic Shunts. Hepatology 2000, 31, 864–871. [Google Scholar] [CrossRef] [PubMed]
  63. Sawhney, S.; Montano-Loza, A.J.; Salat, P.; McCarthy, M.; Kneteman, N.; Meza-Junco, J.; Owen, R. Transarterial Chemoembolization in Patients with Hepatocellular Carcinoma: Predictors of Survival. Can. J. Gastroenterol. 2011, 25, 426–432. [Google Scholar] [CrossRef] [PubMed]
  64. Sieghart, W.; Hucke, F.; Pinter, M.; Graziadei, I.; Vogel, W.; Müller, C.; Heinzl, H.; Trauner, M.; Peck-Radosavljevic, M. The ART of Decision Making: Retreatment with Transarterial Chemoembolization in Patients with Hepatocellular Carcinoma. Hepatology 2013, 57, 2261–2273. [Google Scholar] [CrossRef] [PubMed]
  65. Abbasi, A.H.; Abid, S.; Haq, T.U.; Awan, S. Role of Assessment for Retreatment with Transarterial Chemoembolization Score in Decision of Retreatment with Trans-Arterial Chemo-Embolization Sessions in Patients with Hepatocellular Carcinoma. J. Ayub Med. Coll. Abbottabad 2017, 29, 378–383. [Google Scholar] [PubMed]
  66. Jia, K.-F.; Wang, H.; Yu, C.-L.; Yin, W.-L.; Zhang, X.-D.; Wang, F.; Sun, C.; Shen, W. ASARA, a Prediction Model Based on Child-Pugh Class in Hepatocellular Carcinoma Patients Undergoing Transarterial Chemoembolization. Hepatobiliary Pancreat Dis. Int. 2022. [Google Scholar] [CrossRef]
  67. Kadalayil, L.; Benini, R.; Pallan, L.; O’Beirne, J.; Marelli, L.; Yu, D.; Hackshaw, A.; Fox, R.; Johnson, P.; Burroughs, A.K.; et al. A Simple Prognostic Scoring System for Patients Receiving Transarterial Embolisation for Hepatocellular Cancer. Ann. Oncol. 2013, 24, 2565–2570. [Google Scholar] [CrossRef]
  68. Georgiades, C.S.; Liapi, E.; Frangakis, C.; Park, J.; Kim, H.W.; Hong, K.; Geschwind, J.-F.H. Prognostic Accuracy of 12 Liver Staging Systems in Patients with Unresectable Hepatocellular Carcinoma Treated with Transarterial Chemoembolization. J. Vasc. Interv. Radiol. 2006, 17, 1619–1624. [Google Scholar] [CrossRef]
  69. Hucke, F.; Pinter, M.; Graziadei, I.; Bota, S.; Vogel, W.; Müller, C.; Heinzl, H.; Waneck, F.; Trauner, M.; Peck-Radosavljevic, M.; et al. How to STATE Suitability and START Transarterial Chemoembolization in Patients with Intermediate Stage Hepatocellular Carcinoma. J. Hepatol. 2014, 61, 1287–1296. [Google Scholar] [CrossRef]
  70. El Khaddari, S.; Gaudin, J.-L.; Abidi, H.; Picaud, G.; Rode, A.; Souquet, J.-C. Chemoembolization in hepatocellular carcinoma: Multivariate analysis of survival prog.gnostic factors after the first session. Gastroenterol. Clin. Biol. 2002, 26, 728–734. [Google Scholar]
  71. Wai, C.-T.; Greenson, J.K.; Fontana, R.J.; Kalbfleisch, J.D.; Marrero, J.A.; Conjeevaram, H.S.; Lok, A.S.-F. A Simple Noninvasive Index Can Predict Both Significant Fibrosis and Cirrhosis in Patients with Chronic Hepatitis C. Hepatology 2003, 38, 518–526. [Google Scholar] [CrossRef] [Green Version]
  72. Zhang, X.; Svn, Z.; Liv, M.; Liu, M.; Zhang, Y.; Sun, Q. Assessment of Prognostic Value of Aspartate Aminotransferase-to-Platelet Ratio Index in Patients With Hepatocellular Carcinoma: Meta-Analysis of 28 Cohort Studies. Front. Med. Lausanne 2021, 8, 756210. [Google Scholar] [CrossRef] [PubMed]
  73. Song, Y.P.; Zhao, Q.Y.; Li, S.; Wang, H.; Wu, P.H. Non-invasive fibrosis indexes in predicting acute liver function deterioration after transcatheter arterial chemoembolization. Zhonghua Yi Xue Za Zhi 2016, 96, 716–719. [Google Scholar] [CrossRef] [PubMed]
  74. Kim, B.K.; Shim, J.H.; Kim, S.U.; Park, J.Y.; Kim, D.Y.; Ahn, S.H.; Kim, K.M.; Lim, Y.-S.; Han, K.-H.; Lee, H.C. Risk Prediction for Patients with Hepatocellular Carcinoma Undergoing Chemoembolization: Development of a Prediction Model. Liver Int. 2016, 36, 92–99. [Google Scholar] [CrossRef] [PubMed]
  75. Mähringer-Kunz, A.; Weinmann, A.; Schmidtmann, I.; Koch, S.; Schotten, S.; Pinto dos Santos, D.; Pitton, M.B.; Dueber, C.; Galle, P.R.; Kloeckner, R. Validation of the SNACOR Clinical Scoring System after Transarterial Chemoembolisation in Patients with Hepatocellular Carcinoma. BMC Cancer 2018, 18, 489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Okuda, K.; Ohtsuki, T.; Obata, H.; Tomimatsu, M.; Okazaki, N.; Hasegawa, H.; Nakajima, Y.; Ohnishi, K. Natural History of Hepatocellular Carcinoma and Prognosis in Relation to Treatment. Study of 850 Patients. Cancer 1985, 56, 918–928. [Google Scholar] [CrossRef]
  77. Levy, I.; Sherman, M. Liver Cancer Study Group of the University of Toronto Staging of Hepatocellular Carcinoma: Assessment of the CLIP, Okuda, and Child-Pugh Staging Systems in a Cohort of 257 Patients in Toronto. Gut 2002, 50, 881–885. [Google Scholar] [CrossRef] [Green Version]
  78. A New Prognostic System for Hepatocellular Carcinoma: A Retrospective Study of 435 Patients: The Cancer of the Liver Italian Program (CLIP) Investigators. Hepatology 1998, 28, 751–755. [CrossRef]
  79. Li, L.; Gou, C.-Y.; Li, J.-Y.; Achakzai, R.; Li, X.-H. Cancer of the Liver Italian Program Score Helps Identify Potential Candidates for Transarterial Chemoembolization in Patients with Barcelona Clinic Liver Cancer Stage C. Hepatobiliary Pancreat. Dis. Int. 2016, 15, 152–157. [Google Scholar] [CrossRef]
  80. Mitchell, D.G.; Bruix, J.; Sherman, M.; Sirlin, C.B. LI-RADS (Liver Imaging Reporting and Data System): Summary, Discussion, and Consensus of the LI-RADS Management Working Group and Future Directions. Hepatology 2015, 61, 1056–1065. [Google Scholar] [CrossRef]
  81. Lencioni, R.; Petruzzi, P.; Crocetti, L. Chemoembolization of Hepatocellular Carcinoma. Semin. Interv. Radiol. 2013, 30, 3–11. [Google Scholar] [CrossRef] [Green Version]
  82. Schraml, C.; Kaufmann, S.; Rempp, H.; Syha, R.; Ketelsen, D.; Notohamiprodjo, M.; Nikolaou, K. Imaging of HCC-Current State of the Art. Diagnostics 2015, 5, 513–545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Tangkijvanich, P.; Anukulkarnkusol, N.; Suwangool, P.; Lertmaharit, S.; Hanvivatvong, O.; Kullavanijaya, P.; Poovorawan, Y. Clinical Characteristics and Prognosis of Hepatocellular Carcinoma: Analysis Based on Serum Alpha-Fetoprotein Levels. J. Clin. Gastroenterol. 2000, 31, 302–308. [Google Scholar] [CrossRef] [PubMed]
  84. Arai, T.; Kobayashi, A.; Ohya, A.; Takahashi, M.; Yokoyama, T.; Shimizu, A.; Motoyama, H.; Furusawa, N.; Notake, T.; Kitagawa, N.; et al. Assessment of Treatment Outcomes Based on Tumor Marker Trends in Patients with Recurrent Hepatocellular Carcinoma Undergoing Trans-Catheter Arterial Chemo-Embolization. Int. J. Clin. Oncol. 2014, 19, 871–879. [Google Scholar] [CrossRef] [PubMed]
  85. Yu, R.; Tan, Z.; Xiang, X.; Dan, Y.; Deng, G. Effectiveness of PIVKA-II in the Detection of Hepatocellular Carcinoma Based on Real-World Clinical Data. BMC Cancer 2017, 17, 608. [Google Scholar] [CrossRef] [PubMed]
  86. Chehab, M.A.; Thakor, A.S.; Tulin-Silver, S.; Connolly, B.L.; Cahill, A.M.; Ward, T.J.; Padia, S.A.; Kohi, M.P.; Midia, M.; Chaudry, G.; et al. Adult and Pediatric Antibiotic Prophylaxis during Vascular and IR Procedures: A Society of Interventional Radiology Practice Parameter Update Endorsed by the Cardiovascular and Interventional Radiological Society of Europe and the Canadian Association for Interventional Radiology. J. Vasc. Interv. Radiol. 2018, 29, 1483–1501.e2. [Google Scholar] [CrossRef] [Green Version]
  87. Miura, J.T.; Rilling, W.S.; White, S.B.; Hieb, R.A.; Tutton, S.M.; Patel, P.J.; Gamblin, T.C.; Hohenwalter, E.J. Safety and Efficacy of Transarterial Chemoembolization in Patients with Transjugular Intrahepatic Portosystemic Shunts. HPB Oxf. 2015, 17, 707–712. [Google Scholar] [CrossRef] [Green Version]
  88. Patel, I.J.; Rahim, S.; Davidson, J.C.; Hanks, S.E.; Tam, A.L.; Walker, T.G.; Wilkins, L.R.; Sarode, R.; Weinberg, I. Society of Interventional Radiology Consensus.s.s Guidelines for the Periprocedural Management of Thrombotic and Bleeding Risk in Patients Undergoing Percutaneous Image-Guided Interventions-Part II: Recommendations: Endorsed by the Canadian Association for Interventional Radiology and the Cardiovascular and Interventional Radiological Society of Europe. J. Vasc. Interv. Radiol. 2019, 30, 1168–1184.e1. [Google Scholar] [CrossRef] [Green Version]
  89. Kis, B.; Mills, M.; Hoffe, S.E. Hepatic Radioembolization from Transradial Access: Initial Experience and Comparison to Transfemoral Access. Diagn. Interv. Radiol. 2016, 22, 444–449. [Google Scholar] [CrossRef]
  90. Vuurmans, T.; Hilton, D. Brewing the Right Cocktail for Radial Intervention. Indian Heart J. 2010, 62, 221–225. [Google Scholar]
  91. Zimmermann, M.; Schulze-Hagen, M.; Pedersoli, F.; Isfort, P.; Heinzel, A.; Kuhl, C.; Bruners, P. Y90-Radioembolization via Variant Hepatic Arteries: Is There a Relevant Risk for Non-Target Embolization? WJR 2019, 11, 102–109. [Google Scholar] [CrossRef]
  92. Gaba, R.C. Chemoembolization Practice Patterns and Technical Methods Among Interventional Radiologists: Results of an Online Survey. Am. J. Roentgenol. 2012, 198, 692–699. [Google Scholar] [CrossRef] [PubMed]
  93. Abdelmaksoud, M.H.K.; Louie, J.D.; Kothary, N.; Hwang, G.L.; Kuo, W.T.; Hofmann, L.V.; Hovsepian, D.M.; Sze, D.Y. Embolization of Parasitized Extrahepatic Arteries to Reestablish Intrahepatic Arterial Supply to Tumors before Yttrium-90 Radioembolization. J. Vasc. Interv. Radiol. 2011, 22, 1355–1362. [Google Scholar] [CrossRef] [PubMed]
  94. Tacher, V.; Lin, M.; Duran, R.; Yarmohammadi, H.; Lee, H.; Chapiro, J.; Chao, M.; Wang, Z.; Frangakis, C.; Sohn, J.H.; et al. Comparison of Existing Response Criteria in Patients with Hepatocellular Carcinoma Treated with Transarterial Chemoembolization Using a 3D Quantitative Approach. Radiology 2016, 278, 275–284. [Google Scholar] [CrossRef]
  95. Noori, V.J.; Eldrup-Jørgensen, J. A Systematic Review of Vascular Closure Devices for Femoral Artery Puncture Sites. J. Vasc. Surg. 2018, 68, 887–899. [Google Scholar] [CrossRef]
  96. Rand, T.; Loewe, C.; Schoder, M.; Schmook, M.T.; Peck-Radosavljevic, M.; Kettenbach, J.; Wolf, F.; Schneider, B.; Lammer, J. Arterial Embolization of Unresectable Hepatocellular Carcinoma with Use of Microspheres, Lipiodol, and Cyanoacrylate. Cardiovasc. Interv. Radiol 2005, 28, 313–318. [Google Scholar] [CrossRef]
  97. Lencioni, R.; Crocetti, L. Local-Regional Treatment of Hepatocellular Carcinoma. Radiology 2012, 262, 43–58. [Google Scholar] [CrossRef]
  98. Datta, J.; Narayan, R.R.; Kemeny, N.E.; D’Angelica, M.I. Role of Hepatic Artery Infusion Chemotherapy in Treatment of Initially Unresectable Colorectal Liver Metastases: A Review. JAMA Surg. 2019, 154, 768. [Google Scholar] [CrossRef]
  99. Liu, Z.-C.; Lu, X.-F.; Yang, H.; Liu, H.-D.; Song, X.; Ning, S.-L.; Xu, Y.-F.; Chen, Y.-X. Clinical Outcomes of Patients with Severe Hepatic Hereditary Hemorrhagic Telangiectasia After Banding of the Hepatic Artery and Banding/Ligation of Branches of the Hepatic Artery. Eur. J. Vasc. Endovasc. Surg. 2016, 51, 594–601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Li, Q.-J.; He, M.-K.; Chen, H.-W.; Fang, W.-Q.; Zhou, Y.-M.; Xu, L.; Wei, W.; Zhang, Y.-J.; Guo, Y.; Guo, R.-P.; et al. Hepatic Arterial Infusion of Oxaliplatin, Fluorouracil, and Leucovorin Versus Transarterial Chemoembolization for Large Hepatocellular Carcinoma: A Randomized Phase III Trial. JCO 2022, 40, 150–160. [Google Scholar] [CrossRef]
  101. Choi, S.Y.; Kim, A.H.; Kim, K.A.; Won, J.Y.; Lee, D.Y.; Lee, K.-H. A Simplified Technique of Percutaneous Hepatic Artery Port-Catheter Insertion for the Treatment of Advanced Hepatocellular Carcinoma with Portal Vein Invasion. Korean J. Radiol. 2010, 11, 648. [Google Scholar] [CrossRef] [Green Version]
  102. Seki, H.; Shiina, M. Placement of a Long Tapered Side-Hole Catheter in the Hepatic Artery: Technical Advantages, Catheter Stability, and Arterial Patency. Am. J. Roentgenol. 2006, 187, 1312–1320. [Google Scholar] [CrossRef] [PubMed]
  103. Chevallier, O.; Mvouama, S.; Pellegrinelli, J.; Guillen, K.; Manfredi, S.; Ghiringhelli, F.; Falvo, N.; Midulla, M.; Loffroy, R. Percutaneous Implantation of a Microcatheter-Port System for Hepatic Arterial Infusion Chemotherapy of Unresectable Liver Tumors: Technical Feasibility, Functionality, and Complications. Diagnostics 2021, 11, 399. [Google Scholar] [CrossRef] [PubMed]
  104. Tsochatzis, E.A.; Fatourou, E.; O’Beirne, J.; Meyer, T.; Burroughs, A.K. Transarterial Chemoembolization and Bland Embolization for Hepatocellular Carcinoma. World J. Gastroenterol. 2014, 20, 3069–3077. [Google Scholar] [CrossRef] [PubMed]
  105. Geschwind, J.-F.H.; Nezami, N. Combining Chemotherapy and Radiation Therapy for Liver Cancer: Is the Solution an Intraarterial Approach? Cardiovasc. Interv. Radiol. 2020, 43, 1538–1539. [Google Scholar] [CrossRef]
  106. European Association for the Study of the Liver; European Organisation for Research and Treatment of Cancer. EASL–EORTC Clinical Practice Guidelines: Management of Hepatocellular Carcinoma. J. Hepatol. 2012, 56, 908–943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Shin, S.W. The Current Practice of Transarterial Chemoembolization for the Treatment of Hepatocellular Carcinoma. Korean J. Radiol. 2009, 10, 425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Chen, S.; Peng, Z.; Zhang, Y.; Chen, M.; Li, J.; Guo, R.; Li, J.; Li, B.; Mei, J.; Feng, S.; et al. Lack of Response to Transarterial Chemoembolization for Intermediate-Stage Hepatocellular Carcinoma: Abandon or Repeat? Radiology 2021, 298, 680–692. [Google Scholar] [CrossRef] [PubMed]
  109. Zhang, L.; Sun, J.-H.; Ji, J.-S.; Zhong, B.-Y.; Zhou, G.-H.; Song, J.-J.; Hou, Z.-H.; Huang, P.; Zhang, S.; Li, Z.; et al. Imaging Changes and Clinical Complications After Drug-Eluting Bead Versus Conventional Transarterial Chemoembolization for Unresectable Hepatocellular Carcinoma: Multicenter Study. AJR Am. J. Roentgenol. 2021, 217, 933–943. [Google Scholar] [CrossRef]
  110. Saini, A.; Wallace, A.; Alzubaidi, S.; Knuttinen, M.G.; Naidu, S.; Sheth, R.; Albadawi, H.; Oklu, R. History and Evolution of Yttrium-90 Radioembolization for Hepatocellular Carcinoma. JCM 2019, 8, 55. [Google Scholar] [CrossRef] [Green Version]
  111. Vente, M.A.D.; Wondergem, M.; van der Tweel, I.; van den Bosch, M.A.A.J.; Zonnenberg, B.A.; Lam, M.G.E.H.; van Het Schip, A.D.; Nijsen, J.F.W. Yttrium-90 Microsphere Radioembolization for the Treatment of Liver Malignancies: A Structured Meta-Analysis. Eur. Radiol. 2009, 19, 951–959. [Google Scholar] [CrossRef] [Green Version]
  112. Gabr, A.; Riaz, A.; Johnson, G.E.; Kim, E.; Padia, S.; Lewandowski, R.J.; Salem, R. Correlation of Y90-Absorbed Radiation Dose to Pathological Necrosis in Hepatocellular Carcinoma: Confirmatory Multicenter Analysis in 45 Explants. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 580–583. [Google Scholar] [CrossRef] [PubMed]
  113. Cheng, B.; Sethi, I.; Villalobos, A.; Wagstaff, W.; Schuster, D.M.; Bercu, Z.; Brandon, D.; Kokabi, N. Determination of Tumour Dose Response Threshold and Implication on Survival in Patients with HCC Treated with Y90 Radiation Segmentectomy: A Simple Semi-Quantitative Analysis. Nucl. Med. Commun. 2021; Publish Ahead of Print. [Google Scholar] [CrossRef] [PubMed]
  114. 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] [Green Version]
  115. He, M.; Li, Q.; Zou, R.; Shen, J.; Fang, W.; Tan, G.; Zhou, Y.; Wu, X.; Xu, L.; Wei, W.; et al. Sorafenib Plus Hepatic Arterial Infusion of Oxaliplatin, Fluorouracil, and Leucovorin vs Sorafenib Alone for Hepatocellular Carcinoma With Portal Vein Invasion: A Randomized Clinical Trial. JAMA Oncol. 2019, 5, 953. [Google Scholar] [CrossRef]
  116. Kudo, M.; Ueshima, K.; Yokosuka, O.; Ogasawara, S.; Obi, S.; Izumi, N.; Aikata, H.; Nagano, H.; Hatano, E.; Sasaki, Y.; et al. Sorafenib plus Low-Dose Cisplatin and Fluorouracil Hepatic Arterial Infusion Chemotherapy versus Sorafenib Alone in Patients with Advanced Hepatocellular Carcinoma (SILIUS): A Randomised, Open Label, Phase 3 Trial. Lancet Gastroenterol. Hepatol. 2018, 3, 424–432. [Google Scholar] [CrossRef]
  117. Zheng, K.; Zhu, X.; Fu, S.; Cao, G.; Li, W.-Q.; Xu, L.; Chen, H.; Wu, D.; Yang, R.; Wang, K.; et al. Sorafenib Plus Hepatic Arterial Infusion Chemotherapy versus Sorafenib for Hepatocellular Carcinoma with Major Portal Vein Tumor Thrombosis: A Randomized Trial. Radiology 2022, 303, 455–464. [Google Scholar] [CrossRef]
  118. Raoul, J.-L.; Sangro, B.; Forner, A.; Mazzaferro, V.; Piscaglia, F.; Bolondi, L.; Lencioni, R. Evolving Strategies for the Management of Intermediate-Stage Hepatocellular Carcinoma: Available Evidence and Expert Opinion on the Use of Transarterial Chemoembolization. Cancer Treat. Rev. 2011, 37, 212–220. [Google Scholar] [CrossRef]
  119. Sieghart, W.; Hucke, F.; Peck-Radosavljevic, M. Transarterial Chemoembolization: Modalities, Indication, and Patient Selection. J. Hepatol 2015, 62, 1187–1195. [Google Scholar] [CrossRef] [Green Version]
  120. Kennedy, A.; Nag, S.; Salem, R.; Murthy, R.; McEwan, A.J.; Nutting, C.; Benson, A.; 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. Radiat Oncol. Biol. Phys. 2007, 68, 13–23. [Google Scholar] [CrossRef]
  121. Maas, M.; Beets-Tan, R.; Gaubert, J.-Y.; Gomez Munoz, F.; Habert, P.; Klompenhouwer, L.G.; Vilares Morgado, P.; Schaefer, N.; Cornelis, F.H.; Solomon, S.B.; et al. Follow-up after Radiological Intervention in Oncology: ECIO-ESOI Evidence and Consensus-Based Recommendations for Clinical Practice. Insights Imaging 2020, 11, 83. [Google Scholar] [CrossRef]
  122. Groupe d’Etude et de Traitement du Carcinome Hépatocellulaire A Comparison of Lipiodol Chemoembolization and Conservative Treatment for Unresectable Hepatocellular Carcinoma. N. Engl. J. Med. 1995, 332, 1256–1261. [CrossRef] [PubMed]
  123. Paye, F.; Farges, O.; Dahmane, M.; Vilgrain, V.; Flejou, J.F.; Belghiti, J. Cytolysis Following Chemoembolization for Hepatocellular Carcinoma. Br. J. Surg. 1999, 86, 176–180. [Google Scholar] [CrossRef] [PubMed]
  124. Wigmore, S.J.; Redhead, D.N.; Thomson, B.N.J.; Currie, E.J.; Parks, R.W.; Madhavan, K.K.; Garden, O.J. Postchemoembolisation Syndrome--Tumour Necrosis or Hepatocyte Injury? Br. J. Cancer 2003, 89, 1423–1427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Castells, A.; Bruix, J.; Ayuso, C.; Brú, C.; Montanyà, X.; Boix, L.; Rodès, J. Transarterial Embolization for Hepatocellular Carcinoma. Antibiotic Prophylaxis and Clinical Meaning of Postembolization Fever. J. Hepatol. 1995, 22, 410–415. [Google Scholar] [CrossRef]
  126. Leung, D.A.; Goin, J.E.; Sickles, C.; Raskay, B.J.; Soulen, M.C. Determinants of Postembolization Syndrome after Hepatic Chemoembolization. J. Vasc. Interv. Radiol. 2001, 12, 321–326. [Google Scholar] [CrossRef]
  127. Chan, A.O.; Yuen, M.-F.; Hui, C.-K.; Tso, W.-K.; Lai, C.-L. A Prospective Study Regarding the Complications of Transcatheter Intraarterial Lipiodol Chemoembolization in Patients with Hepatocellular Carcinoma. Cancer 2002, 94, 1747–1752. [Google Scholar] [CrossRef]
  128. Chung, J.W.; Park, J.H.; Han, J.K.; Choi, B.I.; Han, M.C.; Lee, H.S.; Kim, C.Y. Hepatic Tumors: Predisposing Factors for Complications of Transcatheter Oily Chemoembolization. Radiology 1996, 198, 33–40. [Google Scholar] [CrossRef]
  129. Garwood, E.R.; Fidelman, N.; Hoch, S.E.; Kerlan, R.K.; Yao, F.Y. Morbidity and Mortality Following Transarterial Liver Chemoembolization in Patients with Hepatocellular Carcinoma and Synthetic Hepatic Dysfunction. Liver Transplant. 2013, 19, 164–173. [Google Scholar] [CrossRef]
  130. Marelli, L.; Stigliano, R.; Triantos, C.; Senzolo, M.; Cholongitas, E.; Davies, N.; Tibballs, J.; Meyer, T.; Patch, D.W.; Burroughs, A.K. Transarterial Therapy for Hepatocellular Carcinoma: Which Technique Is More Effective? A Systematic Review of Cohort and Randomized Studies. Cardiovasc. Interv. Radiol. 2007, 30, 6–25. [Google Scholar] [CrossRef]
  131. Song, S.Y.; Chung, J.W.; Han, J.K.; Lim, H.G.; Koh, Y.H.; Park, J.H.; Lee, H.S.; Kim, C.Y. Liver Abscess after Transcatheter Oily Chemoembolization for Hepatic Tumors: Incidence, Predisposing Factors, and Clinical Outcome. Vasc. Interv. Radiol. 2001, 12, 313–320. [Google Scholar] [CrossRef]
  132. Kim, H.K.; Chung, Y.H.; Song, B.C.; Yang, S.H.; Yoon, H.K.; Yu, E.; Sung, K.B.; Lee, Y.S.; Lee, S.G.; Suh, D.J. Ischemic Bile Duct Injury as a Serious Complication after Transarterial Chemoembolization in Patients with Hepatocellular Carcinoma. J. Clin. Gastroenterol. 2001, 32, 423–427. [Google Scholar] [CrossRef] [PubMed]
  133. Wu, G.-C.; Perng, W.-C.; Chen, C.-W.; Chian, C.-F.; Peng, C.-K.; Su, W.-L. Acute Respiratory Distress Syndrome after Transcatheter Arterial Chemoembolization of Hepatocellular Carcinomas. Am. J. Med. Sci. 2009, 338, 357–360. [Google Scholar] [CrossRef] [PubMed]
  134. Kwok, P.C.H.; Lam, T.W.; Lam, C.L.; Lai, A.K.H.; Lo, H.Y.; Chan, S.C.H. Rare Pulmonary Complications after Transarterial Chemoembolisation for Hepatocellular Carcinoma: Two Case Reports. Hong Kong J. 2003, 9, 457–460. [Google Scholar]
  135. Kim, J.-T.; Heo, S.-H.; Choi, S.-M.; Lee, S.-H.; Park, M.-S.; Kim, B.-C.; Kim, Y.; Kim, M.-K.; Cho, K.-H. Cerebral Embolism of Iodized Oil (Lipiodol) after Transcatheter Arterial Chemoembolization for Hepatocellular Carcinoma. J. Neuroimaging 2009, 19, 394–397. [Google Scholar] [CrossRef]
  136. Jouneau, S.; Vauléon, E.; Caulet-Maugendre, S.; Polard, E.; Volatron, A.-C.; Meunier, C.; Tattevin, P.; Montani, D.; Garin, E.; Raoul, J.-L.; et al. 131I-Labeled Lipiodol-Induced Interstitial Pneumonia: A Series of 15 Cases. Chest 2011, 139, 1463–1469. [Google Scholar] [CrossRef]
  137. Brown, D.B.; Chapman, W.C.; Cook, R.D.; Kerr, J.R.; Gould, J.E.; Pilgram, T.K.; Darcy, M.D. Chemoembolization of Hepatocellular Carcinoma: Patient Status at Presentation and Outcome over 15 Years at a Single Center. AJR Am. J. Roentgenol 2008, 190, 608–615. [Google Scholar] [CrossRef]
  138. Toyoda, H.; Kumada, T.; Tada, T.; Sone, Y.; Fujimori, M. Transarterial Chemoembolization for Hepatitis B Virus-Associated Hepatocellular Carcinoma: Improved Survival after Concomitant Treatment with Nucleoside Analogues. J. Vasc. Interv. Radiol. 2012, 23, 317–322.e1. [Google Scholar] [CrossRef]
  139. Perrillo, R.P. Reactivated Hepatitis B Due to Medical Interventions: The Clinical Spectrum Expands. Antivir. Ther. 2011, 16, 947–949. [Google Scholar] [CrossRef] [Green Version]
  140. 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]
  141. Riaz, A.; Lewandowski, R.J.; Kulik, L.M.; Mulcahy, M.F.; Sato, K.T.; Ryu, R.K.; Omary, R.A.; Salem, R. Complications Following Radioembolization with Yttrium-90 Microspheres: A Comprehensive Literature Review. J. Vasc. Interv. Radiol. 2009, 20, 1121–1130; quiz 1131. [Google Scholar] [CrossRef]
  142. Riaz, A.; Awais, R.; Salem, R. Side Effects of Yttrium-90 Radioembolization. Front. Oncol. 2014, 4, 198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Salem, R.; Lewandowski, R.J.; Atassi, B.; Gordon, S.C.; Gates, V.L.; Barakat, O.; Sergie, Z.; Wong, C.-Y.O.; 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] [PubMed]
  144. Lam, M.G.E.H.; Abdelmaksoud, M.H.K.; Chang, D.T.; Eclov, N.C.; Chung, M.P.; Koong, A.C.; Louie, J.D.; Sze, D.Y. Safety of 90Y Radioembolization in Patients Who Have Undergone Previous External Beam Radiation Therapy. Int. J. Radiat Oncol. Biol. Phys. 2013, 87, 323–329. [Google Scholar] [CrossRef] [PubMed]
  145. Jakobs, T.F.; Saleem, S.; Atassi, B.; Reda, E.; Lewandowski, R.J.; Yaghmai, V.; Miller, F.; Ryu, R.K.; Ibrahim, S.; Sato, K.T.; et al. Fibrosis, Portal Hypertension, and Hepatic Volume Changes Induced by Intra-Arterial Radiotherapy with 90yttrium Microspheres. Dig. Dis. Sci. 2008, 53, 2556–2563. [Google Scholar] [CrossRef]
  146. Ayav, A.; Habib, N.; Jiao, L.R. Portal Hypertension Secondary to 90Yttrium Microspheres: An Unknown Complication. J. Clin. Oncol. 2005, 23, 8275–8276. [Google Scholar] [CrossRef]
  147. Carr, B.I.; Metes, D.M. Peripheral Blood Lymphocyte Depletion after Hepatic Arterial 90Yttrium Microsphere Therapy for Hepatocellular Carcinoma. Int. J. Radiat Oncol. Biol. Phys. 2012, 82, 1179–1184. [Google Scholar] [CrossRef]
  148. Currie, B.M.; Hoteit, M.A.; Ben-Josef, E.; Nadolski, G.J.; Soulen, M.C. Radioembolization-Induced Chronic Hepatotoxicity: A Single-Center Cohort Analysis. J. Vasc. Interv. Radiol. 2019, 30, 1915–1923. [Google Scholar] [CrossRef]
  149. Lencioni, R.; Llovet, J. Modified RECIST (MRECIST) Assessment for Hepatocellular Carcinoma. Semin. Liver Dis. 2010, 30, 052–060. [Google Scholar] [CrossRef] [Green Version]
  150. 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. J. Hepatol. 2001, 35, 421–430. [Google Scholar] [CrossRef]
  151. Eisenhauer, E.A.; Therasse, P.; Bogaerts, J.; Schwartz, L.H.; Sargent, D.; Ford, R.; Dancey, J.; Arbuck, S.; Gwyther, S.; Mooney, M.; et al. New Response Evaluation Criteria in Solid Tumours: Revised RECIST Guideline (Version 1.1). Eur. J. Cancer 2009, 45, 228–247. [Google Scholar] [CrossRef]
  152. Tirkes, T.; Hollar, M.A.; Tann, M.; Kohli, M.D.; Akisik, F.; Sandrasegaran, K. Response Criteria in Oncologic Imaging: Review of Traditional and New Criteria. RadioGraphics 2013, 33, 1323–1341. [Google Scholar] [CrossRef] [PubMed]
  153. James, K.; Eisenhauer, E.; Christian, M.; Terenziani, M.; Vena, D.; Muldal, A.; Therasse, P. Measuring Response in Solid Tumors: Unidimensional Versus Bidimensional Measurement. JNCI J. Natl. Cancer Inst. 1999, 91, 523–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Bruix, J.; Sherman, M. Management of Hepatocellular Carcinoma. Hepatology 2005, 42, 1208–1236. [Google Scholar] [CrossRef] [PubMed]
  155. Miller, A.B.; Hoogstraten, B.; Staquet, M.; Winkler, A. Reporting Results of Cancer Treatment. Cancer 1981, 47, 207–214. [Google Scholar] [CrossRef]
  156. Lim, H.K.; Han, J.K. Hepatocellular Carcinoma: Evaluation of Therapeutic Response to Interventional Procedures. Abdom. Imaging 2002, 27, 168–179. [Google Scholar] [CrossRef]
  157. Chapiro, J.; Lin, M.; Duran, R.; Schernthaner, R.E.; Geschwind, J.-F. Assessing Tumor Response after Loco-Regional Liver Cancer Therapies: The Role of 3D MRI. Expert Rev. Anticancer Ther. 2015, 15, 199–205. [Google Scholar] [CrossRef] [Green Version]
  158. Zhao, Y.; Duran, R.; Bai, W.; Sahu, S.; Wang, W.; Kabus, S.; Lin, M.; Han, G.; Geschwind, J.-F. Which Criteria Applied in Multi-Phasic CT Can Predict Early Tumor Response in Patients with Hepatocellular Carcinoma Treated Using Conventional TACE: RECIST, MRECIST, EASL or QEASL? Cardiovasc. Interv. Radiol. 2018, 41, 433–442. [Google Scholar] [CrossRef] [Green Version]
  159. Chapiro, J.; Duran, R.; Lin, M.; Schernthaner, R.E.; Wang, Z.; Gorodetski, B.; Geschwind, J.-F. Identifying Staging Markers for Hepatocellular Carcinoma before Transarterial Chemoembolization: Comparison of Three-Dimensional Quantitative versus Non-Three-Dimensional Imaging Markers. Radiology 2015, 275, 438–447. [Google Scholar] [CrossRef]
  160. Chapiro, J.; Wood, L.D.; Lin, M.; Duran, R.; Cornish, T.; Lesage, D.; Charu, V.; Schernthaner, R.; Wang, Z.; Tacher, V.; et al. Radiologic-Pathologic Analysis of Contrast-Enhanced and Diffusion-Weighted MR Imaging in Patients with HCC after TACE: Diagnostic Accuracy of 3D Quantitative Image Analysis. Radiology 2014, 273, 746–758. [Google Scholar] [CrossRef] [Green Version]
  161. O, J.H.; Lodge, M.A.; Wahl, R.L. Practical PERCIST: A Simplified Guide to PET Response Criteria in Solid Tumors 1.0. Radiology 2016, 280, 576–584. [Google Scholar] [CrossRef] [Green Version]
  162. Zhang, F.; Yang, J.; Nezami, N.; Laage-Gaupp, F.; Chapiro, J.; De Lin, M.; Duncan, J. Liver Tissue Classification Using an Auto-Context-Based Deep Neural Network with a Multi-Phase Training Framework. Patch Based Tech. Med. Imaging 2018, 11075, 59–66. [Google Scholar] [CrossRef]
  163. Abajian, A.; Murali, N.; Savic, L.J.; Laage-Gaupp, F.M.; Nezami, N.; Duncan, J.S.; Schlachter, T.; Lin, M.; Geschwind, J.-F.; Chapiro, J. Predicting Treatment Response to Intra-Arterial Therapies for Hepatocellular Carcinoma with the Use of Supervised Machine Learning—An Artificial Intelligence Concept. J. Vasc. Interv. Radiol. 2018, 29, 850–857.e1. [Google Scholar] [CrossRef]
  164. Miszczuk, M.A.; Chapiro, J.; Geschwind, J.-F.H.; Thakur, V.; Nezami, N.; Laage-Gaupp, F.; Kulon, M.; van Breugel, J.M.M.; Fereydooni, A.; Lin, M.; et al. Lipiodol as an Imaging Biomarker of Tumor Response After Conventional Transarterial Chemoembolization: Prospective Clinical Validation in Patients with Primary and Secondary Liver Cancer. Transl. Oncol. 2020, 13, 100742. [Google Scholar] [CrossRef] [PubMed]
  165. Nezami, N.; VAN Breugel, J.M.M.; Konstantinidis, M.; Chapiro, J.; Savic, L.J.; Miszczuk, M.A.; Rexha, I.; Lin, M.; Hong, K.; Georgiades, C. Lipiodol Deposition and Washout in Primary and Metastatic Liver Tumors After Chemoembolization. In Vivo 2021, 35, 3261–3270. [Google Scholar] [CrossRef] [PubMed]
  166. Borde, T.; Nezami, N.; Gaupp, F.L.; Savic, L.J.; Taddei, T.; Jaffe, A.; Strazzabosco, M.; Lin, M.; Duran, R.; Georgiades, C.; et al. Optimization of the BCLC Staging System for Locoregional Therapy for Hepatocellular Carcinoma by Using Quantitative Tumor Burden Imaging Biomarkers at MRI. Radiology 2022, 304, 228–237. [Google Scholar] [CrossRef]
  167. Letzen, B.S.; Malpani, R.; Miszczuk, M.; de Ruiter, Q.M.B.; Petty, C.W.; Rexha, I.; Nezami, N.; Laage-Gaupp, F.; Lin, M.; Schlachter, T.R.; et al. Lipiodol as an Intra-Procedural Imaging Biomarker for Liver Tumor Response to Transarterial Chemoembolization: Post-Hoc Analysis of a Prospective Clinical Trial. Clin. Imaging 2021, 78, 194–200. [Google Scholar] [CrossRef]
  168. Weiss, C.R.; Nezami, N. One Step Closer to Precision Medicine for Transarterial Therapy of HCC. Radiology 2020, 297, 235–236. [Google Scholar] [CrossRef]
  169. Martin, S.P.; Fako, V.; Dang, H.; Dominguez, D.A.; Khatib, S.; Ma, L.; Wang, H.; Zheng, W.; Wang, X.W. PKM2 Inhibition May Reverse Therapeutic Resistance to Transarterial Chemoembolization in Hepatocellular Carcinoma. J. Exp. Clin. Cancer Res. 2020, 39, 99. [Google Scholar] [CrossRef]
  170. Llovet, J.M.; Montal, R.; Villanueva, A. Randomized Trials and Endpoints in Advanced HCC: Role of PFS as a Surrogate of Survival. J. Hepatol. 2019, 70, 1262–1277. [Google Scholar] [CrossRef] [Green Version]
  171. Beaver, J.A.; Howie, L.J.; Pelosof, L.; Kim, T.; Liu, J.; Goldberg, K.B.; Sridhara, R.; Blumenthal, G.M.; Farrell, A.T.; Keegan, P.; et al. A 25-Year Experience of US Food and Drug Administration Accelerated Approval of Malignant Hematology and Oncology Drugs and Biologics: A Review. JAMA Oncol. 2018, 4, 849. [Google Scholar] [CrossRef]
  172. Kemp, R.; Prasad, V. Surrogate Endpoints in Oncology: When Are They Acceptable for Regulatory and Clinical Decisions, and Are They Currently Overused? BMC Med. 2017, 15, 134. [Google Scholar] [CrossRef] [PubMed]
  173. Abbott, A.M.; Doepker, M.P.; Kim, Y.; Perez, M.C.; Gandle, C.; Thomas, K.L.; Choi, J.; Shridhar, R.; Zager, J.S. Hepatic Progression-Free and Overall Survival After Regional Therapy to the Liver for Metastatic Melanoma. Am. J. Clin. Oncol. 2018, 41, 747–753. [Google Scholar] [CrossRef]
  174. Lin, D.Y.; Liaw, Y.F.; Lee, T.Y.; Lai, C.M. Hepatic Arterial Embolization in Patients with Unresectable Hepatocellular Carcinoma--a Randomized Controlled Trial. Gastroenterology 1988, 94, 453–456. [Google Scholar] [CrossRef]
  175. Meyer, T.; Kirkwood, A.; Roughton, M.; Beare, S.; Tsochatzis, E.; Yu, D.; Davies, N.; Williams, E.; Pereira, S.P.; Hochhauser, D.; et al. A Randomised Phase II/III Trial of 3-Weekly Cisplatin-Based Sequential Transarterial Chemoembolisation vs Embolisation Alone for Hepatocellular Carcinoma. Br. J. Cancer 2013, 108, 1252–1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Bruix, J.; Llovet, J.M.; Castells, A.; Montañá, X.; Brú, C.; Ayuso, M.C.; Vilana, R.; Rodés, J. Transarterial Embolization versus Symptomatic Treatment in Patients with Advanced Hepatocellular Carcinoma: Results of a Randomized, Controlled Trial in a Single Institution. Hepatology 1998, 27, 1578–1583. [Google Scholar] [CrossRef] [PubMed]
  177. Chang, J.M.; Tzeng, W.S.; Pan, H.B.; Yang, C.F.; Lai, K.H. Transcatheter Arterial Embolization with or without Cisplatin Treatment of Hepatocellular Carcinoma. A Randomized Controlled Study. Cancer 1994, 74, 2449–2453. [Google Scholar] [CrossRef]
  178. Llovet, J.M.; Real, M.I.; Montaña, X.; Planas, R.; Coll, S.; Aponte, J.; Ayuso, C.; Sala, M.; Muchart, J.; Solà, R.; et al. Arterial Embolisation or Chemoembolisation versus Symptomatic Treatment in Patients with Unresectable Hepatocellular Carcinoma: A Randomised Controlled Trial. Lancet 2002, 359, 1734–1739. [Google Scholar] [CrossRef]
  179. Malagari, K.; Pomoni, M.; Kelekis, A.; Pomoni, A.; Dourakis, S.; Spyridopoulos, T.; Moschouris, H.; Emmanouil, E.; Rizos, S.; Kelekis, D. Prospective Randomized Comparison of Chemoembolization with Doxorubicin-Eluting Beads and Bland Embolization with BeadBlock for Hepatocellular Carcinoma. Cardiovasc. Intervent. Radiol. 2010, 33, 541–551. [Google Scholar] [CrossRef]
  180. Prajapati, H.J.; Xing, M.; Spivey, J.R.; Hanish, S.I.; El-Rayes, B.F.; Kauh, J.S.; Chen, Z.; Kim, H.S. Survival, Efficacy, and Safety of Small versus Large Doxorubicin Drug-Eluting Beads TACE Chemoembolization in Patients with Unresectable HCC. AJR Am. J. Roentgenol. 2014, 203, W706–W714. [Google Scholar] [CrossRef]
  181. Covey, A.M.; Maluccio, M.A.; Schubert, J.; BenPorat, L.; Brody, L.A.; Sofocleous, C.T.; Getrajdman, G.I.; Fong, Y.; Brown, K.T. Particle Embolization of Recurrent Hepatocellular Carcinoma after Hepatectomy. Cancer 2006, 106, 2181–2189. [Google Scholar] [CrossRef]
  182. Lo, C.-M.; Ngan, H.; Tso, W.-K.; Liu, C.-L.; Lam, C.-M.; Poon, R.T.-P.; Fan, S.-T.; Wong, J. Randomized Controlled Trial of Transarterial Lipiodol Chemoembolization for Unresectable Hepatocellular Carcinoma. Hepatology 2002, 35, 1164–1171. [Google Scholar] [CrossRef] [PubMed]
  183. Takayasu, K.; Arii, S.; Ikai, I.; Omata, M.; Okita, K.; Ichida, T.; Matsuyama, Y.; Nakanuma, Y.; Kojiro, M.; Makuuchi, M.; et al. Prospective Cohort Study of Transarterial Chemoembolization for Unresectable Hepatocellular Carcinoma in 8510 Patients. Gastroenterology 2006, 131, 461–469. [Google Scholar] [CrossRef] [PubMed]
  184. Lammer, J.; Malagari, K.; Vogl, T.; Pilleul, F.; Denys, A.; Watkinson, A.; Pitton, M.; Sergent, G.; Pfammatter, T.; Terraz, S.; et al. Prospective Randomized Study of Doxorubicin-Eluting-Bead Embolization in the Treatment of Hepatocellular Carcinoma: Results of the PRECISION V Study. Cardiovasc. Interv. Radiol. 2010, 33, 41–52. [Google Scholar] [CrossRef] [Green Version]
  185. Facciorusso, A.; Di Maso, M.; Muscatiello, N. Drug-Eluting Beads versus Conventional Chemoembolization for the Treatment of Unresectable Hepatocellular Carcinoma: A Meta-Analysis. Dig. Liver Dis. 2016, 48, 571–577. [Google Scholar] [CrossRef]
  186. On Behalf of the PRECISION ITALIA STUDY GROUP; Golfieri, R.; Giampalma, E.; Renzulli, M.; Cioni, R.; Bargellini, I.; Bartolozzi, C.; Breatta, A.D.; Gandini, G.; Nani, R.; et al. Randomised Controlled Trial of Doxorubicin-Eluting Beads vs Conventional Chemoembolisation for Hepatocellular Carcinoma. Br. J. Cancer 2014, 111, 255–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Erhardt, A.; Kolligs, F.; Dollinger, M.; Schott, E.; Wege, H.; Bitzer, M.; Gog, C.; Lammert, F.; Schuchmann, M.; Walter, C.; et al. TACE plus Sorafenib for the Treatment of Hepatocellular Carcinoma: Results of the Multicenter, Phase II SOCRATES Trial. Cancer Chemother. Pharmacol. 2014, 74, 947–954. [Google Scholar] [CrossRef]
  188. Jin, B.; Wang, D.; Lewandowski, R.J.; Riaz, A.; Ryu, R.K.; Sato, K.T.; Larson, A.C.; Salem, R.; Omary, R.A. Chemoembolization Endpoints: Effect on Survival Among Patients With Hepatocellular Carcinoma. Am. J. Roentgenol. 2011, 196, 919–928. [Google Scholar] [CrossRef]
  189. Habbel, V.S.A.; Zeile, M.; Stavrou, G.A.; Wacker, F.; Brüning, R.; Oldhafer, K.-J.; Rodt, T. Correlation between SACE (Subjective Angiographic Chemoembolization Endpoint) Score and Tumor Response and Its Impact on Survival after DEB-TACE in Patients with Hepatocellular Carcinoma. Abdom. Radiol. 2019, 44, 3463–3479. [Google Scholar] [CrossRef]
  190. Kudo, M.; Ueshima, K.; Ikeda, M.; Torimura, T.; Tanabe, N.; Aikata, H.; Izumi, N.; Yamasaki, T.; Nojiri, S.; Hino, K.; et al. Randomised, Multicentre Prospective Trial of Transarterial Chemoembolisation (TACE) plus Sorafenib as Compared with TACE Alone in Patients with Hepatocellular Carcinoma: TACTICS Trial. Gut 2020, 69, 1492–1501. [Google Scholar] [CrossRef]
  191. Georgiades, C.S.; Hong, K.; D’Angelo, M.; Geschwind, J.-F.H. Safety and Efficacy of Transarterial Chemoembolization in Patients with Unresectable Hepatocellular Carcinoma and Portal Vein Thrombosis. J. Vasc. Interv. Radiol. 2005, 16, 1653–1659. [Google Scholar] [CrossRef]
  192. Ha, B.Y.; Ahmed, A.; Sze, D.Y.; Razavi, M.K.; Simpson, N.; Keeffe, E.B.; Nguyen, M.H. Long-Term Survival of Patients with Unresectable Hepatocellular Carcinoma Treated with Transcatheter Arterial Chemoinfusion. Aliment. Pharmacol. Ther. 2007, 26, 839–846. [Google Scholar] [CrossRef] [PubMed]
  193. Kim, P.; Prapong, W.; Sze, D.Y.; So, S.K.; Razavi, M.K. Treatment of Hepatocellular Carcinoma with Sub-Selective Transcatheter Arterial Oily Chemoinfusion. Tech. Vasc. Interv. Radiol. 2002, 5, 127–131. [Google Scholar] [CrossRef] [PubMed]
  194. Kim, J.H.; Yoon, H.-K.; Kim, S.Y.; Kim, K.M.; Ko, G.-Y.; Gwon, D.I.; Sung, K.-B. Transcatheter Arterial Chemoembolization vs. Chemoinfusion for Unresectable Hepatocellular Carcinoma in Patients with Major Portal Vein Thrombosis. Aliment. Pharmacol. Ther. 2009, 29, 1291–1298. [Google Scholar] [CrossRef] [PubMed]
  195. Hilgard, P.; Hamami, M.; Fouly, A.E.; Scherag, A.; Müller, 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]
  196. Sangro, B.; Carpanese, L.; Cianni, R.; Golfieri, R.; Gasparini, D.; Ezziddin, S.; Paprottka, P.M.; Fiore, F.; Van Buskirk, M.; Bilbao, J.I.; et al. Survival after Yttrium-90 Resin Microsphere Radioembolization of Hepatocellular Carcinoma across Barcelona Clinic Liver Cancer Stages: A European Evaluation. Hepatology 2011, 54, 868–878. [Google Scholar] [CrossRef]
  197. Kim, D.Y.; Park, B.J.; Kim, Y.H.; Han, K.-H.; Cho, S.B.; Cho, K.R.; Uhm, S.-H.; Choe, J.-G.; Choi, J.Y.; Chun, H.J.; et al. Radioembolization With Yttrium-90 Resin Microspheres in Hepatocellular Carcinoma: A Multicenter Prospective Study. Am. J. Clin. Oncol. 2015, 38, 495–501. [Google Scholar] [CrossRef]
  198. 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]
  199. 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.e2. [Google Scholar] [CrossRef] [Green Version]
  200. 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]
  201. Garin, E.; Lenoir, L.; Edeline, J.; Laffont, S.; Mesbah, H.; Porée, P.; Sulpice, L.; Boudjema, K.; Mesbah, M.; Guillygomarc’h, A.; et al. Boosted Selective Internal Radiation Therapy with 90Y-Loaded Glass Microspheres (B-SIRT) for Hepatocellular Carcinoma Patients: A New Personalized Promising Concept. Eur. J. Nucl. Med. Mol. Imaging 2013, 40, 1057–1068. [Google Scholar] [CrossRef] [Green Version]
  202. 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]
  203. Lewandowski, R.J.; Gabr, A.; Abouchaleh, N.; Ali, R.; Al Asadi, A.; Mora, R.A.; Kulik, L.; Ganger, D.; Desai, K.; Thornburg, B.; et al. Radiation Segmentectomy: Potential Curative Therapy for Early Hepatocellular Carcinoma. Radiology 2018, 287, 1050–1058. [Google Scholar] [CrossRef] [PubMed]
  204. Padia, S.A.; Geisinger, M.A.; Newman, J.S.; Pierce, G.; Obuchowski, N.A.; Sands, M.J. Effectiveness of Coil Embolization in Angiographically Detectable versus Non-Detectable Sources of Upper Gastrointestinal Hemorrhage. J. Vasc. Interv. Radiol. 2009, 20, 461–466. [Google Scholar] [CrossRef] [PubMed]
  205. Biederman, D.M.; Titano, J.J.; Korff, R.A.; Fischman, A.M.; Patel, R.S.; Nowakowski, F.S.; Lookstein, R.A.; Kim, E. Radiation Segmentectomy versus Selective Chemoembolization in the Treatment of Early-Stage Hepatocellular Carcinoma. J. Vasc. Interv. Radiol. 2018, 29, 30–37.e2. [Google Scholar] [CrossRef]
  206. Mehta, N.; Frenette, C.; Tabrizian, P.; Hoteit, M.; Guy, J.; Parikh, N.; Ghaziani, T.T.; Dhanasekaran, R.; Dodge, J.L.; Natarajan, B.; et al. Downstaging Outcomes for Hepatocellular Carcinoma: Results From the Multicenter Evaluation of Reduction in Tumor Size before Liver Transplantation (MERITS-LT) Consortium. Gastroenterology 2021, 161, 1502–1512. [Google Scholar] [CrossRef]
  207. Katsanos, K.; Kitrou, P.; Spiliopoulos, S.; Maroulis, I.; Petsas, T.; Karnabatidis, D. Comparative Effectiveness of Different Transarterial Embolization Therapies Alone or in Combination with Local Ablative or Adjuvant Systemic Treatments for Unresectable Hepatocellular Carcinoma: A Network Meta-Analysis of Randomized Controlled Trials. PLoS ONE 2017, 12, e0184597. [Google Scholar] [CrossRef]
  208. Lewis, S.R.; Pritchard, M.W.; Evans, D.J.; Butler, A.R.; Alderson, P.; Smith, A.F.; Roberts, I. Colloids versus Crystalloids for Fluid Resuscitation in Critically Ill People. Cochrane Database Syst. Rev. 2018, 8, 1465–1858. [Google Scholar] [CrossRef]
  209. Lewis, A.L.; Taylor, R.R.; Hall, B.; Gonzalez, M.V.; Willis, S.L.; Stratford, P.W. Pharmacokinetic and Safety Study of Doxorubicin-Eluting Beads in a Porcine Model of Hepatic Arterial Embolization. J. Vasc. Interv. Radiol. 2006, 17, 1335–1343. [Google Scholar] [CrossRef]
  210. Tang, Y.; Taylor, R.R.; Gonzalez, M.V.; Lewis, A.L.; Stratford, P.W. Evaluation of Irinotecan Drug-Eluting Beads: A New Drug–Device Combination Product for the Chemoembolization of Hepatic Metastases. J. Control. Release 2006, 116, e55–e56. [Google Scholar] [CrossRef]
  211. Ashrafi, K.; Tang, Y.; Britton, H.; Domenge, O.; Blino, D.; Bushby, A.J.; Shuturminska, K.; den Hartog, M.; Radaelli, A.; Negussie, A.H.; et al. Characterization of a Novel Intrinsically Radiopaque Drug-Eluting Bead for Image-Guided Therapy: DC Bead LUMITM. J. Control. Release 2017, 250, 36–47. [Google Scholar] [CrossRef]
  212. Sharma, K.V.; Dreher, M.R.; Tang, Y.; Pritchard, W.; Chiesa, O.A.; Karanian, J.; Peregoy, J.; Orandi, B.; Woods, D.; Donahue, D.; et al. Development of “Imageable” Beads for Transcatheter Embolotherapy. J. Vasc. Interv. Radiol. 2010, 21, 865–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Jayakrishnan, A.; Thanoo, B.C.; Rathinam, K.; Mohanty, M. Preparation and Evaluation of Radiopaque Hydrogel Microspheres Based on PHEMA/Iothalamic Acid and PHEMA/Iopanoic Acid as Particulate Emboli. J. Biomed. Mater. Res. 1990, 24, 993–1004. [Google Scholar] [CrossRef] [PubMed]
  214. Thanoo, B.C.; Jayakrishnan, A. Radiopaque Hydrogel Microspheres. J. Microencapsul. 1989, 6, 233–244. [Google Scholar] [CrossRef]
  215. Horák, D.; Metalová, M.; Svec, F.; Drobník, J.; Kálal, J.; Borovicka, M.; Adamyan, A.A.; Voronkova, O.S.; Gumargalieva, K.Z. Hydrogels in Endovascular Embolization. III. Radiopaque Spherical Particles, Their Preparation and Properties. Biomaterials 1987, 8, 142–145. [Google Scholar] [CrossRef]
  216. Pesapane, F.; Nezami, N.; Patella, F.; Geschwind, J.F. New Concepts in Embolotherapy of HCC. Med. Oncol. 2017, 34, 58. [Google Scholar] [CrossRef] [PubMed]
  217. Duran, R.; Sharma, K.; Dreher, M.R.; Ashrafi, K.; Mirpour, S.; Lin, M.; Schernthaner, R.E.; Schlachter, T.R.; Tacher, V.; Lewis, A.L.; et al. A Novel Inherently Radiopaque Bead for Transarterial Embolization to Treat Liver Cancer—A Pre-Clinical Study. Theranostics 2016, 6, 28–39. [Google Scholar] [CrossRef] [PubMed]
  218. Kehoe, S.; Langman, M.; Werner-Zwanziger, U.; Abraham, R.J.; Boyd, D. Mixture Designs to Assess Composition–Structure–Property Relationships in SiO2–CaO–ZnO–La2O3–TiO2–MgO–SrO–Na2O Glasses: Potential Materials for Embolization. J. Biomater. Appl. 2013, 28, 416–433. [Google Scholar] [CrossRef]
  219. Yamamoto, A.; Chervoneva, I.; Sullivan, K.L.; Eschelman, D.J.; Gonsalves, C.F.; Mastrangelo, M.J.; Berd, D.; Shields, J.A.; Shields, C.L.; Terai, M.; et al. High-Dose Immunoembolization: Survival Benefit in Patients with Hepatic Metastases from Uveal Melanoma. Radiology 2009, 252, 290–298. [Google Scholar] [CrossRef] [Green Version]
  220. Berg, M.; Wingender, G.; Djandji, D.; Hegenbarth, S.; Momburg, F.; Hämmerling, G.; Limmer, A.; Knolle, P. Cross-Presentation of Antigens from Apoptotic Tumor Cells by Liver Sinusoidal Endothelial Cells Leads to Tumor-Specific CD8+ T Cell Tolerance. Eur. J. Immunol. 2006, 36, 2960–2970. [Google Scholar] [CrossRef]
  221. Weiskirch, L.M.; Bar-Dagan, Y.; Mokyr, M.B. Transforming Growth Factor-Beta-Mediated down-Regulation of Antitumor Cytotoxicity of Spleen Cells from MOPC-315 Tumor-Bearing Mice Engaged in Tumor Eradication Following Low-Dose Melphalan Therapy. Cancer Immunol. Immunother. 1994, 38, 215–224. [Google Scholar] [CrossRef]
  222. Kambayashi, T.; Alexander, H.R.; Fong, M.; Strassmann, G. Potential Involvement of IL-10 in Suppressing Tumor-Associated Macrophages. Colon-26-Derived Prostaglandin E2 Inhibits TNF-Alpha Release via a Mechanism Involving IL-10. J. Immunol. 1995, 154, 3383–3390. [Google Scholar] [PubMed]
  223. Bot, F.J.; van Eijk, L.; Schipper, P.; Löwenberg, B. Human Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) Stimulates Immature Marrow Precursors but No CFU-GM, CFU-G, or CFU-M. Exp. Hematol. 1989, 17, 292–295. [Google Scholar] [PubMed]
  224. Albert, M.L.; Sauter, B.; Bhardwaj, N. Dendritic Cells Acquire Antigen from Apoptotic Cells and Induce Class I-Restricted CTLs. Nature 1998, 392, 86–89. [Google Scholar] [CrossRef] [PubMed]
  225. Bowen, D.G.; McCaughan, G.W.; Bertolino, P. Intrahepatic Immunity: A Tale of Two Sites? Trends Immunol. 2005, 26, 512–517. [Google Scholar] [CrossRef] [PubMed]
  226. Hou, X.-Y.; Jiang, G.; Yang, C.-S.; Tang, J.-Q.; Wei, Z.-P.; Liu, Y.-Q. Application of Nanotechnology in the Diagnosis and Therapy of Hepatocellular Carcinoma. PRA 2016, 11, 322–331. [Google Scholar] [CrossRef] [PubMed]
  227. European Society of Radiology (ESR) Medical Imaging in Personalised Medicine: A White Paper of the Research Committee of the European Society of Radiology (ESR). Insights Imaging 2015, 6, 141–155. [CrossRef] [PubMed]
  228. Degrauwe, N.; Hocquelet, A.; Digklia, A.; Schaefer, N.; Denys, A.; Duran, R. Theranostics in Interventional Oncology: Versatile Carriers for Diagnosis and Targeted Image-Guided Minimally Invasive Procedures. Front. Pharmacol. 2019, 10, 450. [Google Scholar] [CrossRef]
  229. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.D.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano Based Drug Delivery Systems: Recent Developments and Future Prospects. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef] [Green Version]
  230. Sechi, M.; Sanna, V.; Pala, N. Targeted Therapy Using Nanotechnology: Focus on Cancer. IJN 2014, 467. [Google Scholar] [CrossRef] [Green Version]
  231. Mohapatra, A.; Uthaman, S.; Park, I.-K. External and Internal Stimuli-Responsive Metallic Nanotherapeutics for Enhanced Anticancer Therapy. Front. Mol. Biosci. 2021, 7, 597634. [Google Scholar] [CrossRef]
  232. Lewandowski, R.J.; Geschwind, J.-F.; Liapi, E.; Salem, R. Transcatheter Intraarterial Therapies: Rationale and Overview. Radiology 2011, 259, 641–657. [Google Scholar] [CrossRef] [PubMed]
  233. Niu, M.; Yi, M.; Li, N.; Wu, K.; Wu, K. Advances of Targeted Therapy for Hepatocellular Carcinoma. Front. Oncol. 2021, 11, 719896. [Google Scholar] [CrossRef]
  234. Ding, X.; Sun, W.; Li, W.; Shen, Y.; Guo, X.; Teng, Y.; Liu, X.; Zheng, L.; Li, W.; Chen, J. Transarterial Chemoembolization plus Lenvatinib versus Transarterial Chemoembolization plus Sorafenib as First-Line Treatment for Hepatocellular Carcinoma with Portal Vein Tumor Thrombus: A Prospective Randomized Study. Cancer 2021, 127, 3782–3793. [Google Scholar] [CrossRef]
  235. Zhang, L.; Ding, J.; Li, H.-Y.; Wang, Z.-H.; Wu, J. Immunotherapy for Advanced Hepatocellular Carcinoma, Where Are We? Biochim. Et Biophys. Acta (BBA) Rev. Cancer 2020, 1874, 188441. [Google Scholar] [CrossRef] [PubMed]
  236. Yi, M.; Niu, M.; Xu, L.; Luo, S.; Wu, K. Regulation of PD-L1 Expression in the Tumor Microenvironment. J. Hematol. Oncol. 2021, 14, 10. [Google Scholar] [CrossRef]
  237. Niu, M.; Yi, M.; Li, N.; Luo, S.; Wu, K. Predictive Biomarkers of Anti-PD-1/PD-L1 Therapy in NSCLC. Exp. Hematol. Oncol. 2021, 10, 18. [Google Scholar] [CrossRef] [PubMed]
  238. Bucalau, A.-M.; Tancredi, I.; Verset, G. In the Era of Systemic Therapy for Hepatocellular Carcinoma Is Transarterial Chemoembolization Still a Card to Play? Cancers 2021, 13, 5129. [Google Scholar] [CrossRef]
  239. Fong, Z.V.; Palazzo, F.; Needleman, L.; Brown, D.B.; Eschelman, D.J.; Chojnacki, K.A.; Yeo, C.J.; Rosato, E.L. Combined Hepatic Arterial Embolization and Hepatic Ablation for Unresectable Colorectal Metastases to the Liver. Am. Surg. 2012, 78, 1243–1248. [Google Scholar] [CrossRef]
  240. Dawoud, M.A.; Mohamed, R.E.; El Waraki, M.S.; Gabr, A.M. Single-Session Combined Radiofrequency Ablation and Transarterial Chemoembolization in the Treatment of Hepatocellular Carcinoma. Egypt J. Radiol. Nucl. Med. 2017, 48, 935–946. [Google Scholar] [CrossRef]
  241. Yamashita, Y.-I.; Takeishi, K.; Tsuijita, E.; Yoshiya, S.; Morita, K.; Kayashima, H.; Iguchi, T.; Taketomi, A.; Shirabe, K.; Maehara, Y. Beneficial Effects of Preoperative Lipiodolization for Resectable Large Hepatocellular Carcinoma (≥5 Cm in Diameter). J. Surg. Oncol. 2012, 106, 498–503. [Google Scholar] [CrossRef]
  242. Li, C.; Wang, M.-D.; Lu, L.; Wu, H.; Yu, J.-J.; Zhang, W.-G.; Pawlik, T.M.; Zhang, Y.-M.; Zhou, Y.-H.; Gu, W.-M.; et al. Preoperative Transcatheter Arterial Chemoembolization for Surgical Resection of Huge Hepatocellular Carcinoma (≥ 10 Cm): A Multicenter Propensity Matching Analysis. Hepatol. Int. 2019, 13, 736–747. [Google Scholar] [CrossRef] [PubMed]
  243. Choi, J.-H.; Hwang, S.; Lee, Y.-J.; Kim, K.-H.; Ko, G.-Y.; Gwon, D.I.; Ahn, C.-S.; Moon, D.-B.; Ha, T.-Y.; Song, G.-W.; et al. Prognostic Effect of Preoperative Sequential Transcatheter Arterial Chemoembolization and Portal Vein Embolization for Right Hepatectomy in Patients with Solitary Hepatocellular Carcinoma. Korean J. Hepatobiliary Pancreat. Surg. 2015, 19, 59–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Zhou, W.-P.; Lai, E.C.H.; Li, A.-J.; Fu, S.-Y.; Zhou, J.-P.; Pan, Z.-Y.; Lau, W.Y.; Wu, M.-C. A Prospective, Randomized, Controlled Trial of Preoperative Transarterial Chemoembolization for Resectable Large Hepatocellular Carcinoma. Ann. Surg. 2009, 249, 195–202. [Google Scholar] [CrossRef] [PubMed]
  245. Yamasaki, S.; Hasegawa, H.; Kinoshita, H.; Furukawa, M.; Imaoka, S.; Takasaki, K.; Kakumoto, Y.; Saitsu, H.; Yamada, R.; Oosaki, Y.; et al. A Prospective Randomized Trial of the Preventive Effect of Pre-Operative Transcatheter Arterial Embolization against Recurrence of Hepatocellular Carcinoma. Jpn J. Cancer Res. 1996, 87, 206–211. [Google Scholar] [CrossRef] [PubMed]
  246. Zhang, Y.-F.; Guo, R.-P.; Zou, R.-H.; Shen, J.-X.; Wei, W.; Li, S.-H.; OuYang, H.-Y.; Zhu, H.-B.; Xu, L.; Lao, X.-M.; et al. Efficacy and Safety of Preoperative Chemoembolization for Resectable Hepatocellular Carcinoma with Portal Vein Invasion: A Prospective Comparative Study. Eur. Radiol. 2016, 26, 2078–2088. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Transarterial therapies. (A) Bland embolization (BE). (B) Conventional transarterial chemoembolization (cTACE). (C) Drug-eluting beads–transarterial chemoembolization (DEB–TACE). (D) Radioembolization.
Figure 1. Transarterial therapies. (A) Bland embolization (BE). (B) Conventional transarterial chemoembolization (cTACE). (C) Drug-eluting beads–transarterial chemoembolization (DEB–TACE). (D) Radioembolization.
Cancers 14 03351 g001
Figure 2. Indications for the use of transarterial therapies in patients with hepatocellular carcinoma (HCC).
Figure 2. Indications for the use of transarterial therapies in patients with hepatocellular carcinoma (HCC).
Cancers 14 03351 g002
Figure 3. Patient with right lobe HCC with extension into the hepatic vein causing thrombosis. Pre-procedural investigation showed 30% shunt fraction, and therefore the patient was treated with cTACE. (A) Pre-procedural coronal CT shows right lobe HCC lesion with hepatic vein thrombosis. (B) A 99mTc-MAA SPECT/CT shows a lung shunt with a lung shunt fraction of 30%. (C) Intraoperative CT showing good lipiodol uptake in the treated lesion areas. (D) Follow-up MRI showing reduced non-enhancing HCC and hepatic vein thrombus due to good response after treatment.
Figure 3. Patient with right lobe HCC with extension into the hepatic vein causing thrombosis. Pre-procedural investigation showed 30% shunt fraction, and therefore the patient was treated with cTACE. (A) Pre-procedural coronal CT shows right lobe HCC lesion with hepatic vein thrombosis. (B) A 99mTc-MAA SPECT/CT shows a lung shunt with a lung shunt fraction of 30%. (C) Intraoperative CT showing good lipiodol uptake in the treated lesion areas. (D) Follow-up MRI showing reduced non-enhancing HCC and hepatic vein thrombus due to good response after treatment.
Cancers 14 03351 g003
Figure 4. A patient with multifocal HCC who underwent DEB–TACE for its management. CT showing multifocal HCC involving the right (A) and left lobe (B). Common hepatic artery angiogram showing tumor blush (C), which completely disappeared after DEB–TACE (D). Follow-up CT showing good tumor response in the right (E) and left (F) lobe of the liver.
Figure 4. A patient with multifocal HCC who underwent DEB–TACE for its management. CT showing multifocal HCC involving the right (A) and left lobe (B). Common hepatic artery angiogram showing tumor blush (C), which completely disappeared after DEB–TACE (D). Follow-up CT showing good tumor response in the right (E) and left (F) lobe of the liver.
Cancers 14 03351 g004aCancers 14 03351 g004b
Figure 5. Radiation segmentectomy in a patient with HCC with a past surgical history of right hepatectomy. (A) CT showing a large lesion in the left lobe of the liver. (B) Left hepatic artery angiogram showing vessels supplying the tumor. (C) SPECT-CT imaging after left segment-2 sub-segmentectomy, showing good dose delivery. (D) Follow-up CT showing good response in the treated area.
Figure 5. Radiation segmentectomy in a patient with HCC with a past surgical history of right hepatectomy. (A) CT showing a large lesion in the left lobe of the liver. (B) Left hepatic artery angiogram showing vessels supplying the tumor. (C) SPECT-CT imaging after left segment-2 sub-segmentectomy, showing good dose delivery. (D) Follow-up CT showing good response in the treated area.
Cancers 14 03351 g005
Table 1. Barcelona clinic liver cancer classification for the prognosis and treatment of hepatocellular carcinoma [5].
Table 1. Barcelona clinic liver cancer classification for the prognosis and treatment of hepatocellular carcinoma [5].
StagesCharacteristics
Very early stage (0)Single lesion ≤2 cm
Preserved liver function
Performance status: 0 (fully active)
Early stage (A)Single lesion or ≤3 nodules each ≤3 cm in size
Preserved liver function
Performance status: 0
Intermediate stage (B)Multinodular
Preserved liver function
Performance status: 0
Advanced stage (C)Portal invasion and/or extrahepatic spread
Preserved liver function
Performance status: 1 (cannot do heavy physical work) or 2 (up and about more than half the day, can look after self but cannot work)
Terminal stage (D)Any tumor burden
End-stage liver function
Performance status: 3 (in bed or a chair for more than half the day and need help to look after self) or 4 (in bed or chair all the time needing complete care)
Table 2. Eastern Cooperative Oncology Group Performance Status Scale.
Table 2. Eastern Cooperative Oncology Group Performance Status Scale.
GradeECOG Performance Status
0Fully active
1Cannot do heavy physical work
2Up and about more than half the day, can look after self but cannot work
3In bed or a chair for more than half the day and need help to look after self
4In bed or chair all the time needing complete care
5Dead
6Fully active
Table 3. Child–Pugh classification.
Table 3. Child–Pugh classification.
ParameterPoints Assigned
123
AscitesAbsentSlightModerate
Serum bilirubin<2 mg dL−1 (<34.2 micromol L−1)2 to 3 mg dL−1 (34.2 to 51.3 micromol L−1)>3 mg dL−1 (>51.3 micromol L−1)
Serum albumin>3.5 mg dL−1 (35 g L−1)2.8 to 3.5 g dL−1 (28 to 35 g L−1)<2.8 g dL−1 (<28 g L−1)
Prothrombin time or INR<4 or <1.74 to 6 or 1.7 to 2.3>6 or >2.3
EncephalopathyNoneGrade 1 to 2Grade 3 to 4
Table 4. Albumin–bilirubin (ALBI) score.
Table 4. Albumin–bilirubin (ALBI) score.
ALBI GradeScore
Grade 1≤2.6
Grade 2>2.6 to 1.39
Grade 3>1.39
Table 5. Platelet–ALBI (pALBI) score.
Table 5. Platelet–ALBI (pALBI) score.
pALBI GradeScore
Grade 1≤−2.53
Grade 2>−2.53 and ≤−2.09
Grade 3>−2.09
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Garg, T.; Shrigiriwar, A.; Habibollahi, P.; Cristescu, M.; Liddell, R.P.; Chapiro, J.; Inglis, P.; Camacho, J.C.; Nezami, N. Intraarterial Therapies for the Management of Hepatocellular Carcinoma. Cancers 2022, 14, 3351. https://doi.org/10.3390/cancers14143351

AMA Style

Garg T, Shrigiriwar A, Habibollahi P, Cristescu M, Liddell RP, Chapiro J, Inglis P, Camacho JC, Nezami N. Intraarterial Therapies for the Management of Hepatocellular Carcinoma. Cancers. 2022; 14(14):3351. https://doi.org/10.3390/cancers14143351

Chicago/Turabian Style

Garg, Tushar, Apurva Shrigiriwar, Peiman Habibollahi, Mircea Cristescu, Robert P. Liddell, Julius Chapiro, Peter Inglis, Juan C. Camacho, and Nariman Nezami. 2022. "Intraarterial Therapies for the Management of Hepatocellular Carcinoma" Cancers 14, no. 14: 3351. https://doi.org/10.3390/cancers14143351

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