Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Aug 19.
Published in final edited form as: Dig Dis Interv. 2020 May 25;4(2):134–147. doi: 10.1055/s-0040-1712146

Embolotherapy for Hepatic Oncology: Current Perspectives and Future Directions

Raazi Bajwa 1, David C Madoff 2, Sirish A Kishore 3
PMCID: PMC7437993  NIHMSID: NIHMS1616620  PMID: 32832829

Introduction

Primary and secondary liver cancers represent a significant cause of cancer-related death worldwide. Primary liver cancer, including hepatocellular carcinoma and cholangiocarcinoma, is the sixth most common cancer worldwide and the fourth most common cause of cancer death1. Secondary, or metastatic, liver cancer is more common than primary liver cancer, with colorectal liver metastases being the most common2. Other common metastases are from gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs), breast, lung, and melanoma.

While surgical intervention is often considered “curative,” only a minority of patients with primary and secondary liver tumors are surgical candidates36. Thus, trans-arterial embolotherapeutic techniques represent an important therapeutic option in hepatic oncology. Approximately 75% of the blood supply to normal liver parenchyma is supplied by the portal vein, and the remaining 25% by the hepatic artery. Liver tumors, on the other hand, are almost exclusively supplied by the hepatic artery. This pathophysiologic phenomenon is the underlying principle exploited by transarterial hepatic embolotherapy7,8. Embolotherapy is a transarterial catheter-based locoregional therapy whereby any of a variety of therapeutic embolic agents are delivered via the hepatic artery, interrupting the tumoral arterial blood supply while delivering the therapeutic agent preferentially to the areas of tumor involvement. The tumor becomes hypoxic, ischemic, and subsequently undergoes coagulative necrosis. Embolotherapy plays an important role in the management of both primary and secondary liver tumors, ranging from curative as a standalone therapy, an adjunct to additional therapies, or as a salvage/palliative option. The objectives of this review article are to explain the concepts behind hepatic embolotherapy for primary and secondary liver cancer, and to summarize the most current available literature reporting the indications, safety, and efficacy of embolotherapy in hepatic oncology.

Technical Overview and Patient Selection

Technical Overview

Hepatic embolotherapy consists of three primary modalities: transarterial embolization (TAE), transarterial chemoembolization (TACE), and transarterial radioembolization (TARE) or selective internal radiotherapy (SIRT). The technical aspects of each modality are detailed in Table 1. These transarterial therapies are performed after accessing either common femoral artery or the left radial artery. After placement of a vascular sheath in the site of entry, hepatic angiography is then performed, often with the assistance of intra-procedural cone-beam computed tomography (CT) and post-processing software to delineate tumor-feeding vessels of interest. Once the artery supplying tumor is catheterized, known as selective or super-selective catheterization depending on the location within the hepatic arterial system, embolotherapeutic agents are delivered through the catheter, interrupting the tumoral blood supply to the desired angiographic endpoint. The ideal catheter position depends in large part by the transarterial therapeutic modality, but is generally recommended to be as selective as possible without compromising access to tumor supplying arteries, while minimizing exposure to normal hepatic parenchyma.

Table 1.

Overview of Hepatic Embolotherapeutic Techniques

Embolotherapy technique Mechanism Advantages Disadvantages
Transarterial embolisation (TAE) or ‘bland embolization’ Permanent occlusion of the tumor vascular bed - require small particles (50 – 120 μn), such as polyvinyl alcohol (PVA) particles or microspheres, made of trisacryl gelatin, are used 910. Relatively inexpensive Marked post-embolization syndrome, potentially necessitating hospital admission
Readily available “off the shelf”
No chemotherapy induced hepatocellular toxicity
Small particles may shunt to the lungs, causing pulmonary infarction
Conventional transarterial chemoembolization or (cTACE) Two-step procedure:
  1. Embolic material loaded with chemotherapy drug delivered in lipiodol emulsion. Typical chemotherapy agents used are doxorubicin, cisplatin, mitomycin C, irinotecan or gemcitabine, alone or in combination 11-13.

  2. Bland embohzation with temporary agent e.g. Gelfoam®14

Deliver higher doses of cytotoxic chemotherapy and potentiate therapeutic action, while decreasing the systemic effects of the chemotherapy agent15,16 Post-embolization syndrome
Highest level of evidence in treatment of HCC and incorporated in BCLC* staging system
Chemotherapy drug still reaches systemic circulation
No standardized protocol
Drug-eluting bead transarterial chemoembolization (DEB-TACE) Specialized microspheres are loaded with antitumor drugs, most commonly doxorubicin or irinotecan, which are then delivered transarterially to the tumor. Drug delivery more predictable and sustained, with higher intra-tumoral concentrations and lower peak systemic doses of chemotherapy compared to cTACE 17,18. Post-embolization syndrome
More expensive than cTACE
DEB irinotecan TACE more effective in treating colorectal metastases than cTACE19 Potential biliary necrosis/risk of biloma formation in treating neuroendocrine tumors 20
Selective internal radiation therapy (SIRT) Type of intra-arterial brachytherapy: Microspheres containing the radioactive element yttrium-90 (Y-90) cause DNA-induced damage and radiation necrosis. Two FDA approved devices:
  1. Glass microspheres (Therasphere): FDA approved as a palliative option for advanced HCC

  2. Resin microspheres: FDA approved for progressive colorectal liver metastases (CLM)

Deliver high doses of radiation to tumor and minimize dose to radiation sensitive liver Most expensive of all techniques
Requires institutional development of service, with coordination between multiple departments and nuclear regulatory commission (NRC)
Radiation adverse events to patient; such as radiation-induced liver disease (REILD), biliary toxicity, cirrhosis, and small particles may shunt to the lungs, causing pulmonary fibrosis
Role in all stages of HCC; curative, downstage to transplant eligibility, local control and possibly safer in advanced disease (portal vein invasion)
Two types of specialized dosimetric techniques over standard delivery:
  1. Radiation segmentectomy – ‘curative’ therapy by treating ≤ two hepatic segments using a lobar dose

  2. Radiation lobectomy – local tumor control and contralateral lobar hypertrophy

Currently, the optimal endpoint of TACE is not formally established by current evidence, and embolization is continued until achievement of the chosen fluoroscopic endpoint: either stasis or sub-stasis (sub-stasis is defined as contrast clearance within 2 – 5 heartbeats)21. Lewandowski et al. defined a four-point scale in the treatment of HCC known as the subjective angiographic chemoembolization endpoint (SACE) scale [Table 2]22. The authors reported that a sub-stasis endpoint (SACE levels 2 and 3) during TACE improved survival compared to embolization to a more complete stasis endpoint (SACE level 4)23. Post procedural CT is helpful in determining lipiodol retention patterns, another important treatment endpoint, and a potential surrogate for treatment response in HCC24. On the other hand, incomplete embolization of HCC increases plasma levels of angiogenic factors; vascular endothelial growth factor receptor (VEGFR) and insulin-like growth factor receptor 2 (IGFR-2), which have been associated with the development of metastasis after embolization25. If near stasis is not achieved by the end of TACE, embolization can be continued with bland particles until the desired endpoint is achieved, the procedure can be combined with ablation, or a repeat procedure can be scheduled.

Table 2.

Subjective angiographic chemoembolization endpoint (SACE) scale22

Grade Antegrade arterial flow Tumor blush
I Normal Normal / Reduced
II Reduced Reduced
III Reduced None
IV None None

Technical endpoints for liver metastases are more variable, with some texts advising embolization to stasis for bland embolization, while others recommend embolization to sub-stasis in TACE12,26. Following embolotherapy, cross-sectional imaging is usually performed one-month post procedure, though this practice varies by operator and center. Tumor response is typically radiologically assessed by the modified Response Evaluation Criteria in Solid Tumors (mRECIST) or European Association for the study the Liver (EASL) criteria27.

Recently, there has been increased interest in embolotherapy using flow / pressure modulation in parallel with the development of anti-reflux catheters. New anti-reflux catheter devices may improve therapeutic response from both safety and efficacy perspective. Two strategies have been developed to improve delivery of embolic agents to the tumor; balloon-occlusion (B-TACE) and micro-valve catheters (MVI-TACE). Balloon occlusion of the artery supplying the tumor preferentially re-routes blood flow to the tumor. These hemodynamic alterations allow increased embolic agent delivery and is based on fluid mechanic principles whereby blood flows from high to low pressure2830. It has been shown usage of balloon-occlusion catheters in the treatment of HCC can result in much denser lipiodol uptake in tumors, correlating with treatment response31,32.

A micro-valve catheter has an expandable tip that collapses during forward flow and then dynamically seals off the vessel with reversal of flow, analogous to a valve. The expandable tip will collapse except if the pressure in the tip is greater than systolic blood pressure, in which case the tip will expand and continue to seal the vessel33. TACE using micro-valve catheters has demonstrated a higher pathologic HCC tumor response compared to standard end-hole catheters34.

Patient Selection

Evaluation of patients with liver cancer includes a detailed history and physical exam, as well as laboratory and imaging tests to assess generalized performance status, hepatic function, tumor burden, and vascular anatomy. Patients with an incompetent or recently cannulated Sphincter of Oddi are often given pre-procedure and post-procedure antibiotics. While controversial, many standard contraindications are considered relative depending on the operator, degree of planned treatment, presence and extent of comorbidities, and overall clinical scenario. While dependent on therapeutic modality and operator, general contraindications to embolotherapy are detailed in Table 33537.

Table 3.

General Contraindications to Hepatic Embolotherapy

Patient factors
  1. Performance status (Eastern Cooperative Oncology Group > 2)

  2. Renal failure (creatinine ≥2.0 mg/dL)

  3. Unfavorable anatomy (inability to prevent arteriovenous shunting or non-target embolization)

Liver factors
  1. Decompensated liver disease (Child Pugh B ≥ 8)

  2. Alterations to portal vein flow (transjugular intrahepatic portosystemic shunt, thrombosis, hepatofugal flow)

Tumor factors
  1. Extensive tumor with complete replacement of both lobes (> 75%)

Applications of Embolotherapy by Liver Tumor Type

Primary Liver Cancer

Hepatocellular carcinoma (HCC)

HCC is the most common primary liver tumor, accounting for eighty percent1. Its incidence and prevalence are increasing worldwide, likely as a result of increasing alcohol use and obesity rates, in spite of improvements in treatments for infectious hepatitis38. The staging of HCC is multifaceted and though multiple staging systems exist, the Barcelona Clinic Liver Cancer (BCLC) Staging and Treatment Strategy is one of the most common and widely accepted staging paradigms39. Within the BCLC system, HCC amenable to treatment an be categorized as very early stage, early stage, intermediate stage, and advanced. Embolotherapy serves an important role in the management of HCC across all stages of disease.

Very Early Stage (0) or Early Stage (A)

In early and very early stage patients, where curative surgery or transplantation is not an immediate option, embolotherapy can serve the following roles: increase future liver reserve, provide a bridge therapy to transplant, or serve as a curative treatment in unsuitable surgical candidates.

Increase future liver remnant (FLR)

Embolotherapy—either alone or in combination with portal vein embolization (PVE)—can induce future liver remnant (FLR) hypertrophy while simultaneously providing local disease control40,41. A systematic review and meta-analysis demonstrated an increased surgical resection rate in sequential cTACE and PVE than PVE alone for HCC > 5 cm (90 % vs. 75 %; P < 0.001)41. Furthermore, combined cTACE and PVE was associated with better overall survival and recurrence-free survival than PVE alone for patients waiting to undergo major hepatectomy,42,43.

Radiation lobectomy, can also induce FLR hypertrophy and provide local tumor control with volumetric changes comparable to portal vein embolization, albeit slightly slower44. A systematic review, constituting a sub-cohort of 215 patients with HCC (out of 312 patients), found rates of contralateral liver lobe hypertrophy following radiation lobectomy ranging from 26 – 47% at a range of time to hypertrophy from 44 days to 9 months. One of the included studies compared PVE to radiation lobectomy directly, describing significantly greater hypertrophy in the PVE group—61.5% versus 29.0% (P<0.001)—with a shorter median time to hypertrophy of 33 days (range 24–56 days) versus 46 days (range 27–79 days)45. Resection following radiation lobectomy has been found to be safe and feasible, offering “curative” surgery to patients initially thought to have unresectable tumors46.

Bridge to transplant

Embolotherapy can decrease serum alpha-fetoprotein (AFP), reduce organ waitlist drop-off due to tumor progression or death, as well as downstage tumors to transplantable disease. Bridging therapy is estimated to reduce dropout rate to 0 – 10% from up to 19.5% at 12 months47,48. All embolotherapies have shown similar safety and efficacy profiles for patients requiring bridging therapy4951. Although there was no difference in overall survival, a prospective study of patients with either BCLC Stage A or B comparing SIRT to cTACE observed longer times to progression (TTP) in the SIRT group in comparison to the cTACE group – >26 months in the SIRT group versus 6.8 months in the cTACE group, P = 0.012, (HR 0.122, 95% CI, 0.027–0.557, P=.007). The authors concluded that the longer TTP provided by radioembolization could potentially reduce dropout from transplant waitlists. However, there was no significant difference in tumor necrosis and median survival times52.

Pathologic response to locoregional techniques may also have important prognostic consequences: complete pathologic response (CPN) of the treated tumor on explant is associated with reduced HCC recurrence and improved post-transplant survival53,54. The use of specialized catheter delivery tools such as balloon and microvalve catheters may further augment tumor response for appropriate candidates in this setting. CTACE performed with micro-valve catheters prior to transplant for HCC demonstrated significantly higher percentage tumor necrosis on explant, 89.0±2.2%, compared to standard end-hole catheters, 56.1±44.5% (P=0.006)34. Locoregional therapy performed prior to transplantation can provide useful prognostic information. Candidates with an ineffective response or interval progression can be removed from the waiting list based on anticipated long term prognosis55.

Alternative to surgery / ablation

Ablation is recommended for patients with unresectable tumor(s) or contraindications to surgical resection or transplantation for very early and early stage HCC56,57. In cases where ablation is not feasible, embolotherapy offers an alternative locoregional option. A retrospective study achieved similar 5-year overall survival for patients with a single-nodule HCC of 3 cm or smaller, without vascular invasion, if treated by resection, ablation or TACE58.

In cases where TACE is performed as first line therapy, initial objective response (complete response and partial response by mRECIST criteria) after the first session is the most robust predictor of favorable outcome27. In solitary tumors ≤5 cm, an ultraselective approach is preferred. Ultraselective cTACE is defined as cTACE administered at the most distal portion of the subsubsegmental hepatic artery59. This is because well-differentiated tumors or tumors with (extra)capsular and microsatellite tumors are also supplied by the small arterial collaterals and the portal vein60. If not completely treated, recurrent or residual tumor may transform to a more aggressive grade6163. Ultraselective cTACE facilitates maximal lipiodol injection into the tumor sinusoids by physiological blood flow and mechanical blockage of backflow by the microcatheter, which then pervades into the portal system59. Then, subsequent Gelfoam® embolization occludes the hepatic artery and additional arterial collaterals, and prevents washout of the chemoembolic emulsion. Given higher rates of CPN using B-TACE or MVI-TACE as discussed above, efficacy of TACE in such scenarios can potentially be improved for appropriate candidates. The precise mechanism is not known, though may be secondary to flow diversion and delivery via small intra-hepatic collaterals which would not be apparent using standard end-hole techniques, or delivery of higher doses of therapeutic agent deeper into the tumor by overcoming intra-tumoral pressure barriers34,64. Dual occlusion balloon catheter systems may also facilitate embolotherapy to hypovascular or infiltrative tumors that have tiny feeding vessels that cannot be selectively catheterized, such as those arising from a main arterial pedicle, otherwise precluding both non-selective and selective delivery27.

Radiation segmentectomy may be employed as an alternative to percutaneous ablation, particularly in patients with unfavorable anatomic location of the target lesion. Lewandowski et al. performed radiation segmentectomy in 70 patients with solitary HCC, less than or equal to 5 cm, not amenable to surgery or ablation and Child-Pugh class A liver function65. 59% of tumors demonstrated a complete response by European Association for the Study of the Liver (EASL). The median TTP recorded was 2.4 years (95% CI: 2.1 – 5.7 years) and median overall survival was 6.7 years (95% CI: 3.1 – 6.7 years). The authors determined radiation segmentectomy provided response rates and survival outcomes comparable to curative-intent treatments for selected patients with early-stage HCC.

Intermediate Stage (B)

BCLC intermediate stage accounts for 20 % of HCC patients with a 2-year survival of approximately 50 %66. It is important to note the extensive heterogeneity of this patient sub-cohort given the potential range of tumor numbers and sizes as well as range of hepatic function. Patients in the intermediate stage are stratified according to these parameters, with therapeutic indication either as a means of downstaging to potential surgery or as palliative therapy to delay progression and prolong overall survival.

Downstage to Transplant

The Milan criteria is stringent in order to identify the optimal candidates for transplant with the lowest risk of tumor recurrence. However, many patients are ineligible for transplantation due to tumor burden that extends beyond these size and number criteria. Therefore, locoregional therapy is often attempted to downstage patients to transplant eligibility, and to provide a test of tumor biology prior to work up for transplant. Historically, TACE has been the most widely used downstaging therapy, however, SIRT or TAE also provides a viable alternative. A systematic review and meta-analysis was performed to evaluate the success of downstaging to Milan criteria, post-transplant recurrence and overall survival67. In patients with HCC without vascular invasion, a pooled TACE and SIRT success rate of 54% (95% CI: 45% – 63%) was reported, with no difference between rates of successful downstaging between (P = 0.51). The pooled post-transplant recurrence rate was 0.16 (95% CI: 0.11 – 0.23), with no difference between modalities (P = 0.40). However, there was moderate heterogeneity between the included studies, and results were further hampered by inconsistency in patient selection, technique, and outcome assessments. The authors also noted heterogeneous and inconsistently reported wait-times and substantial variations in the time-to-recurrence. Post-transplant recurrence rates and survival data are difficult to objectively examine due to heterogenous patient populations and administered locoregional therapies68. Multicenter, well-controlled studies are recommended though logistically challenging68.

Palliative treatment of multinodular HCC

cTACE is the historically recommended therapy for patients with intermediate stage HCC in the BCLC system. This recommendation stems from seminal work by Llovet et al. which demonstrated increased survival in patients with unresectable HCC, as well as preserved liver function (CP score < 9) and performance status (ECOG 0), treated by cTACE compared to conservative management. Survival probabilities at 1 year and 2 years were 82% and 63% for cTACE, and 63% and 27% for control (P = 0.009). The SPACE clinical trial attempted to build on this by investigating potential improvement in efficacy and safety of DEB-TACE when combined with sorafenib by comparing to DEB-TACE alone in intermediate stage HCC69. The combined arm did not delay time to progression or prolong overall survival in patients, while appearing to increase expected adverse events.

There is increasing use of SIRT for unresectable HCC. A meta-analysis of three randomized controlled trials, including the SIRTACE and PREMIERE trials, found no difference in terms of progression free or overall survival between TACE and SIRT. Of note, however, the PREMIERE trial was one of the first randomized controlled trials to compare SIRT to TACE in BCLC Stage A/B patients, and demonstrated a longer time to progression, 14.5 months (SIRT) versus 6.4 months (TACE) (P = 0.0019) without significant difference in overall survival (23.8 months vs 17.7 months, P = 0.9772)70. On evaluation of the three trials, there were significant study limitations, such as unmet predetermined sample size and inconsistent results, probably due to differences in study populations, BCLC stage, and reported outcomes71. However, in a study examining patients refractory to TACE, 36.7 % patients underwent SIRT and demonstrated a response on imaging, with 10% successfully downstaged received liver transplantation72.

Advanced Stage (C)

Advanced stage HCC carries a dismal prognosis with a median survival of 2 – 4 months [61]. Systemic treatment is the recommended for patients with portal vein invasion73. Sorafenib confers a modest survival increase at the expense of considerable drug-related toxicity, most commonly, diarrhea, weight loss, hand–foot skin reaction, and hypophosphatemia74.

Historically, embolotherapy had been contraindicated in portal vein thrombosis, as TACE administered as lobar and even whole-liver treatments carried the theoretical risk of ischemia, infarction and worsening of liver function75. A systematic review and meta-analysis examined the efficacy of cTACE stratified by main (MPV) and branch (PVB) portal vein invasion76. 1333 patients received an estimated median of 2 (Range 2 – 4) TACE sessions. Response rates, defined by complete or partial response according to the mRECIST criteria, were similar; 14% (9 – 20%) and 16% (8% – 26%) (P = 0.238). However, results demonstrated a 1-year survival rate of 16 % (95% CI: 7% – 28%) and 37% (23 – 51%), for MPV and PVB, respectively (P < 0.001). Pooled liver failure and complication (not otherwise specified) rates were 1% and 18%, respectively. The authors attribute the improved survival of cTACE to improvements in microcatheter technology, facilitating superselective and ultraselective cTACE. By delivering cTACE in such a fashion, arterial blood flow to normal hepatic parenchyma can be preserved, minimizing the risk of the infarction of normal hepatic parenchyma and subsequent liver failure. Additionally, migration of the lipophilic chemoembolic emulsion into the portal system may theoretically provide additional antitumor effects.

It was postulated that administration of sorafenib in combination with TACE might be useful to counter act TACE-induced angiogenic factors and, therefore, improve outcomes from TACE treatment77. This appeared to be confirmed in a systematic review and meta-analysis, demonstrating TACE-sorafenib combination therapy significantly improved time to progression (HR=0.66, 95% CI 0.50–0.81, P=0.002) with a trend to improved overall survival (HR=0.63, 95% CI 0.55–0.71, P=0.058), compared to TACE alone. Also, the disease control rate, defined as a combination of complete response rate, partial response rate and stable disease rate, increased significantly in the combination therapy group (OR=2.93, 95% CI 1.59–5.41, P=0.005)78.

The role of SIRT in advanced HCC remains undetermined in BCLC guidelines79. The SARAH trial failed to show superiority of SIRT to sorafenib, in 467 patients with BCLC Stage A – C, recurrent tumor ineligible for curative surgical therapy or tumors refractory to transarterial chemoembolization, Child-Pugh score A – B and ECOG 0 – 180. Similarly, the SORAMIC trial failed to illustrate a survival benefit of SIRT-sorafenib combined therapy in BCLC B – C patients not eligible for TACE81.

When comparing SIRT to sorafenib in patients with HCC and portal invasion, a study examining 151 patients with HCC and portal vein tumor thrombus found an overall median survival benefit in patients treated with SIRT compared to sorafenib, 26.2 months versus 8.7 months, respectively (HR 0.40 95 % CI: 0.19 – 0.82) (P = 0.013). The difference in overall survival was more pronounced in patients with branch portal vein thrombosis (as opposed to main portal vein thrombosis), with a respective median OS of 25.3 months (95 % CI 13.8– 36.8) vs 7.0 months (95 % CI 5.2–8.9) (p=0.001), than in cases of main PVT, with a respective median OS of 12.0 months (95 % CI 4.6–19.3) vs 6.5 months (95 % CI 4.8–8.3) (p=0.195)82. SIRT has also been shown to preserve health-related quality of life in patients with HCC and portal vein invasion in comparison to systemic therapy83.

Intrahepatic cholangiocarcinoma (ICC)

ICC accounts for 10 – 20% of primary liver tumors, though the incidence is rising84. Surgery is the only curative therapy; however, unlike perihilar or distal bile duct tumors, intrahepatic lesions are often asymptomatic and often present with locally advanced tumours beyond the threshold of surgical resection85. Overall survival is 11.7 months with systemic chemotherapy86. The National Comprehensive Cancer Network (NCCN) guidelines recommend consideration of locoregional therapies in cases of unresectable or metastatic ICC25.

A systematic review examined hepatic embolotherapy, specifically TACE and SIRT, in 929 patients with unresectable ICC87. The majority of the included studies were retrospective with small patient cohorts, highlighting the need for prospective trials in this patient subset. There were variable prior chemotherapy and surgical treatments in the different studies. TACE regimens lacked standardization of technique, from chemotherapeutic agents delivered to number of sessions. Median duration of follow up was 35 months (Ranged 1.8 – 29 months). Complete, partial response, and stable disease rates for TACE were 10%, 20% and 57%, respectively, and for SIRT were 10, 25.5% and 66.5%, respectively. Progression free and overall survival for TACE were 8 months and 13 months, respectively, and for SIRT were 9.8 months and 12.5 months, respectively.

Zhen et al. performed a pooled analysis of 472 patients who received SIRT for unresectable intrahepatic cholangiocarcinoma88. The majority of the included studies were retrospective with small patient cohorts. The overall response rate was 11.5% (Range: 4.8 – 35.3%) The pooled median overall survival was 14.3 months (95% CI: 11.9 – 17.1 months). OS appeared consistent across studies without significant study heterogeneity, I2 = 50.9% (P= 0.068). However, not all studies reported the population and treatment characteristics. There was no difference in OS between glass or resin microspheres. Most common side effects were fatigue (median: 31.7%; range: 0.0–87.5%), abdominal pain (median: 30.0%; range: 0.0–85.0%) and nausea (median: 16.0%; range: 0.0–62.5%). Grade III – IV toxicities were reported in 10 studies, median of 7.8% (range: 0.0–25.0%). 10% of patients in three selected studies were converted to resectable disease based on a systematic review and pooled analysis of unresectable ICC, and is therefore now presented as an option to patients with advanced disease in the latest European Society for Medical Oncology (ESMO) guidelines89. TACE and SIRT are safe and efficacious in the treatment of unresectable ICC, but, their exact role in a therapeutic paradigm remains to be defined.

Colorectal liver metastases

Colorectal cancer is the third most common cancer in the United States90, with 15% of presenting with synchronous hepatic metastases, and 50% of patients with subsequent metastases91. Hepatic metastectomy in the setting of colorectal liver metastasis (CRLM) has been established as a standard of care through comprehensive, retrospective, large population-based studies incorporating surgery and/or ablation demonstrating 5-year survival of 30–50% in patients undergoing surgical resection as compared with 0% 5-year survival in patients that forgo surgery92,93. Yet, only 20 – 30% of patients will have resectable disease, of which 70–80% will have recurrence within 5 years35. For unresectable CRLM, systemic therapy has demonstrated overall survival of 15–20 months, with mortality predominantly attributed to hepatic progression9496.

The NCCN guidelines recommend hepatic resection for resectable liver metastases from colorectal cancer97. Currently, in patients with liver-only / liver-dominant metastases that cannot be surgically resected or ablated, transarterial therapies may be considered, particularly in patients who have progressed on first line therapy. The ESMO guidelines state radioembolization should be considered and chemoembolization may be considered in patients with liver-confined disease refractory to systemic chemotherapy98. However, there may be a role for embolotherapy in the neoadjuvant and adjuvant settings99.

Two randomized controlled trials have been performed, comparing DEB-IRI TACE to different systemic chemotherapy regimens96,100. Martin et al. conducted a phase II clinical trial assessing the efficacy and safety of DEBIRI in combination with systemic modified FOLFOX (folate, fluorouracil and oxaliplatin) ± bevacizumab as compared to FOLFOX ± bevacizumab alone in chemotherapy naïve patients. Seventy patients (40 in the FOLFOXDEBIRI arm and 30 in the control arm) were included in the study. For each DEBIRI treatment, 100 mg of irinotecan-loaded 100–300 μm LC Beads® (BTG, London, UK) were delivered to each lobe. Patients received at least 12 cycles of systemic treatment and two cycles of DEBIRI during chemotherapy off weeks. The median follow-up was 24 months. The overall response rate (CR or PR by mRECIST) was greater in the treatment arm versus the control arm at 2 (78% vs 54%, P=0.02), 4 (95% vs 70%, P=0.03), and 6 months (76% vs 60%, P=0.05). This favorable overall response rate in the treatment arm at 6 months led to more downstaging to resection, 35% versus 16% (P=0.05) and longer median progression-free survival, 15.3 vs 7.6 months.

There was a nonsignificant improvement in median progression-free survival in the control arm, 15 months (Range 10.4 – 20 months) versus the treatment arm, 12 months (Range 9 – 15.4 months) (P=0.18). It should be noted that there were significant baseline differences in disease severity between the study arms in favor of the treatment arm: there was a statistically significantly worse overall Eastern Cooperative Oncology Group performance status, prevalence of extrahepatic disease, and prevalence of KRAS mutant disease in the control arm.

There were more serious adverse events in the FOLFOXDEBIRI arm relative to the FOLFOX alone arm. These were attributed to embolization-related serious adverse events, specifically defined as hospital stay > 23 hours or readmission after DEBIRI treatment, which accounted for 33% of such events. When evaluated only for chemotherapy-associated adverse events, there was no difference between the study arms.

Fiorentini et al. reported their findings in a prospective randomized controlled trial comparing DEBIRI to irinotecan, fluorouracil and leucovorin (FOLFIRI) in patients whom had previously received chemotherapy, but not irinotecan. Seventy-four patients with liver-only metastatic disease were enrolled in the study (36 in the DEBIRI arm and 38 in the FOLFIRI arms), all of whom had received 2–3 lines of prior chemotherapy. The study aimed to deliver 200 mg of irinotecan with 100–300 μm LC beads® two times with one month interval between treatments for the DEBIRI cohort. The FOLFIRI arm received a total of 8 cycles. A significant increase in OS was observed in the DEBIRI group, 22 months, compared to the FOLFIRI group, 15 months (P = 0.031). PFS was also longer in the DEBIRI group, median seven months, versus the FOLFIRI group, 4 months, (P = 0.006). Significantly less neutropenia and mucositis were recorded in the DEBIRI arm compared to FOLFIRI arm, 4% versus 44% (P < 0.0001), and 1% versus 20% (P = 0.00002), respectively.

Selective internal radiation therapy

A phase III clinical study demonstrated a role for SIRT as salvage therapy for patients with chemotherapy-refractory liver-limited metastatic colorectal cancer101. A recent combined analysis of three randomized controlled trials, involving 549 patients, investigated systemic oxaliplatin-based chemotherapy (FOLFOX: leucovorin, fluorouracil, and oxaliplatin) chemotherapy upfront with SIRT or FOLFLOX alone102. There was no difference in overall survival (HR: 1·04, 95% CI 0·90–1·19; P = 0·61). The median survival time in the FOLFOX-SIRT arm was 22.6 months (95% CI 2.0 – 24.5), versus 23.3 months (21.8 – 24.7) in the control arm. Front line use of SIRT with chemotherapy was not recommended and the authors stated the role of SIRT as consolidation therapy after chemotherapy with careful patient selection requires investigation.

In a systematic review and meta-analysis of transarterial therapies in 2306 patients, 1038 TACE and 1268 SIRT, with unresectable CLRM, first line TACE demonstrated a non-statistically significant decreased median OS of 15.2 months compared to SIRT, 29.4 months (P = 0.69)103. For patients failing at ≥ 1 line of systemic therapy; the survival outcomes were 21.3 months (Range 20.6 – 22.4 months) in the TACE group, compared to 10.7 months (Range 9.5 – 12.0 months) in the SIRT group. Chemotherapy regimens were varied amongst the different studies, with DEBIRI accounting for only 53% of studies. TACE had higher rates of conversion to surgically resectable disease, 4% (95% CI: 2 – 7%), compared to SIRT 2% (1–4%). Combined grade 3 – 4 toxicities were seen in 26% (22–30%) of patients who received SIRT, and 17% (13–22%) of patients who underwent TACE. Levy et al conducted a more recent systematic review and meta-analysis of 1583 patients—968 TACE and 615 SIRT—with a different methodology104. Results for TACE were stratified into DEBIRI and cTACE. One-year survival rates were 80.1% (95% CI: 74.2 – 86.0%) for DEBIRI, 69.6% (95% CI: 49.3 – 86.7%) for cTACE and 41% (95% CI: 28 – 54%) for SIRT. Higher 1-year survival rates may be explained by increased radiologic response (CR and PR by RECIST criteria) rates for DEBIRI, 36.2% (95% CI: 0 – 72.5%), compared to cTACE, 23.1% (95% CI: 9.7 – 36.4%). However, pooled median survival were similar 16 months (9.0–23) and 16 months (7.3–25) for DEBIRI and TACE, respectively.

Gastro-entero-pancreatic neuroendocrine tumor (GEP-NET) liver metastases

The prevalence of liver metastases from GEP-NETs vary depending on the site of the tumor. For example, metastases were most common from the pancreas (40.3%) and lung (32.4%), and least common from the rectum (3.4%) and stomach (9.4%)105. The origin, grade (G1-G3) and stage of the tumor are the most important prognostic factors, with presence of metastasis having the highest risk of mortality. Patients with favourable (moderate / well differentiated) pathology and disease burden (regional / localized) can live more than 10 years. For example, only 7.3% of patients with pancreatic NETs were alive at 10 years, compared with 20.5% of those with colon NETs and 27.1% of those with rectal NETs. Treatment strategies are chosen based on three different metastatic patterns: Simple (metastases confined to one liver lobe or limited to two adjacent segments that can be resected by a standard anatomical resection), complex (metastases predominantly in one lobe primarily but with smaller satellites in the contralateral lobe) and diffuse (multifocal metastases). Surgery and/or ablation is generally indicated for simple and complex patterns, with embolotherapy traditionally reserved for diffuse pattern metastases106.

Embolotherapy in the setting of GEP-NET liver metastases is indicated for the control of tumor growth or control of secretory symptoms after the failure / insufficiency of somatostatin analogs, and when hepatic resection is contraindicated106,107. Embolotherapy is recommended early in the disease course if there is significant liver tumor burden or the secretory symptoms are life-threatening or severe in intensity. The European Neuroendcrine Tumor Society (ENETS) recommend embolotherapy only for G1 – G2 tumors, while systemic chemotherapy is typically reserved for neuroendocrine carcinomas (G3).

GEP-NETs are more arterialized than other liver metastases, such as from colorectal cancer108. No specific embolotherapeutic modality is recommended over the other by the NCCN or ENETS, though the choice of embolotherapy should be balanced between effective tumor control and minimizing damage to normal liver. Symptomatic control ranges from 42% – 91% in TAE and 54 – 100% in TACE109. Progression free survival and overall survival ranges from 6 – 36 months and 16 – 69 months in TAE and 5 – 32 months and 15 – 65 months in TACE, respectively. A small randomized trial of 26 patients with progressive unresectable liver metastases from midgut endocrine tumors were randomly assigned bland embolization or chemoembolization and examined for differences in 2-year PFS rate110. A subgroup analysis of one study with 69 patients with G1 NETs and 54 patients with functional islet cell carcinomas showed the former had longer PFS, 22.7 months versus 16.1 months (P = 0.046), and OS, 33.8 months vs. 23.2 months (P = 0.012)111. The authors observed a possible beneficial outcome by delivering chemoembolization in patients with islet cell carcinomas. DEB-TACE is specifically known to cause biliary / liver injury. In a series of 208 patients with either well-differentiated metastatic NET or HCC, treated by cTACE or DEB-TACE, biloma / parenchymal infarct was strongly associated with both DEB-TACE (OR = 9.78, P = 0.002) and NETs (OR = 8.13, P = 0.04)112.

Analogous to HCC, increased levels of angiogenic factors and neoangiogenesis from embolization can drive growth of incompletely treated tumors113,114. A retrospective review examined whether using different size bland microspheres in 160 patients with well to moderately differentiated NET liver metastases115. The hypothesis was whether increased occlusion of end arterioles and subsequent ischemia would lead to improved overall survival. Higher objective response rates in the smaller particle group (< 100 μm) compared to the larger particle group (≥100 μm), 64 versus 42% (P=0.007), respectively, did not translate in to prolonged overall survival. A phase II clinical study administering sunitinib, a tyrosine kinase inhibitor, following embolotherapy in 39 patients with well/moderately differentiated NETs from a range of primary tumor origins (26 patients had small bowel primary tumor) yielded a median PFS of 15.2 months and 1-year and 4-year survival rates of 95% and 59%, respectively116. Another phase II clinical study examined whether everolimus, an mTOR pathway inhibitor, could increase hepatic-PFS at 24 months from 35 to 50 %, when administered after embolotherapy113. The primary endpoint was not reached.

SIRT has can also be employed for the treatment of neuroendocrine tumors, though may potentially manifest undesired sequelae in patients with longer prognoses. The most common early adverse events reported following SIRT are fatigue, abdominal pain, fever and nausea109. Su et al. followed 54 patients treated with SIRT (15 unilobar and 39 whole-liver) for a mean of 4.1 years117. The authors observed imaging cirrhosis-like morphology in 26.7% and 56.4% unilobar and bilobar treatments, respectively. Features of portal hypertension; such as ascites (33% patients) and varices (31% patients), were seen with both more common in whole-liver treatments. Eight patients (20.5%) treated with whole-liver SIRT and exhibited cirrhosis-like morphology showed clinical signs of hepatic decompensation. Tomozawa et al. followed 52 patients for > 1 year treated with SIRT for hepatic metastases from NETs and discovered 15 patients with cirrhosis-like morphology on imaging118. Similar to the study by Su et al., this was more common in bilobar treatments, with significantly elevated liver function tests and thrombocytopenia in this sub-cohort, sustained >4 years after treatment. No significant difference was found in predicting overall survival between unilobar and bilobar treatments (P = 0.676).

A review of the current literature reports successful secretory symptom control rates with embolotherapy ranging from 55% – 100%109. The progression-free survival and the overall survival ranges are 9 – 11 months and 14 – 70 months, respectively109. Factors associated with increased response rate and / or prolonged progression free survival include female gender, radiological hypervascular pattern, well-differentiated tumors and a low Ki67 index119. A small scale prospective study comparing treatment of metastases from GEP-NETs to CRLM by SIRT demonstrated higher objective response rates in the GEP-NET group, 73% versus 25%, and prolonged progression free survival, 1.0 months versus 8.9 months, and overall survival, 6.7 months versus 4.4 months. However, no formal randomized trial has been conducted comparing the efficacy and safety of these therapies.

Other Hepatic Metastases

Liver directed therapy in patients with metastatic disease from non-GI primaries is more controversial, and the evidence more limited. The evidence is perhaps strongest for metastatic breast cancer and melanoma, though treatment has been reported for other primaries such as RCC and lung cancer120123. Approximately 50% of all women with breast cancer develop metastases, of which approximately 50% of patients develop in the liver. Additionally, 5–12% of patients with recurrence develop liver metastases as the primary site recurrence. Generally, embolotherapy is offered to patients with metastatic disease refractory to multiple lines of chemotherapy. A systematic review and pooled analysis of 519 patients with liver metastases from breast cancer treated by TACE (cTACE and DEBIRI) reported median overall survival ranged from 7.3 – 47.0 months and median disease free survival ranged from 2.9 to 17.0 months122. Several retrospective studies have investigated the efficacy of SIRT in patients with liver metastases refractory to systemic treatment, with a median OS of 4 – 13.6 months124.

Uveal melanoma has a distinct propensity to metastasize to the liver, with up to 50% of patients developing hepatic metastases125. Median overall survival of patients with hepatic metastases is reportedly 4 – 5 months, with a 1-year survival of 10%–15%126. Embolotherapies are employed due to the lack of effective systemic chemotherapy12. A review of 649 patients who received TACE, reported that the overall survival ranged from 5 – 29 months123. Chemotherapy regimens varied amongst a combination of retrospective and prospective studies. TACE appeared to be a relatively safe procedure, with eight patients developing grade 3 or 4 complications, three of which were vascular thromboses that precluded further transarterial therapy. Six retrospective studies of 405 patients whom received SIRT reported the median OS ranged from 9 – 24 months123. Currently, a phase I clinical trial evaluating SIRT with sorafenib [NCT01893099] and an early phase I study investigating SIRT with ipilimumab [NCT01730157] are underway.

Future therapies

Precision medicine is a significant innovation in cancer care. One specific area of interest lies in the role of immunotherapy and potential synergy with locoregional therapy. Immune cells and T-cell infiltration correlate with HCC outcome and prognosis, respectively127. Nivolumab and Pembrolizumab, two immune checkpoint inhibitors, have been approved for second line systemic therapy in patients with advanced HCC after failing sorafenib57. Trials have shown that the immune response in HCC elicited after treatment by ablation/embolization can be enhanced by PD-1 inhibition128. A number of trials are being conducted to evaluate potential synergistic therapeutic effects between anti-PD1 inhibitors and transarterial therapy such as SIRT and TACE129. Preliminary studies assessing safety of SIRT-PD1 inhibitor combination therapy has been shown to be safe130.

Another form of precision medicine known as theranostics involves the use of specialized diagnostic and therapeutic agents such as nanoparticles or radiopharmaceuticals to diagnose and treat tumors131. The detection of particular targets in a tumor can help predict whether a patient will benefit from a targeted treatment. Typical uses of theranostics today in hepatic oncology include 177Lu-DOTATATE for metastatic GEP-NETs132. However, the success of theranostic agents are in large part dependent on targeting selectivity and efficiency when delivered systemically while minimizing non-target toxicity, particularly in the kidney, a potential dose limiting organ133,134. Catheter based delivery of theranostic agents may be a way to obviate such limitations. This strategy has the advantage of delivering high doses of nanoparticles locoregionally and minimizing systemic toxicity. In a retrospective analysis of 55 patients with GEP-NETs, intra-arterial injection of peptide receptor radionuclide therapy demonstrated a median PFS and OS were 33.4 and 75.8 months, respectively, in patients with liver-confined tumors135. Real-time treatment response monitoring can also be performed using theranostic agents and positron emission tomography/computed tomography (PET-CT) and magnetic resonance imaging depending on the specific imaging properties136,137.

Artificial intelligence has the potential to revolutionize all aspects of healthcare, particularly image-guided therapy. For example, incomplete HCC treatment via TACE can result in increased levels of angiogenic factors, leading to tumor growth and metastasis138. A novel study conducted by the MD Anderson Cancer Center Group demonstrated machine learning may play a role in predicting patient response to TACE using quantitative imaging features in combination with the BCLC staging system139. Other machine learning applications in interventional oncology include intra-procedural support, such as catheter guidance, and post procedural assessment, such as post-treatment MRI-based texture analysis of tumors140,141.

Conclusion

Embolotherapy is an important tool in the management of liver tumors. The technical advancements and indications for embolotherapy have greatly expanded, providing options to patients ranging from curative treatment in some cancers to prolonging time to progression and overall survival, as well as improved quality of life over systemic therapy. Nonetheless, the quality of evidence and grade of recommendations are limited, which hinder inclusion in to mainstream clinical societal guidelines. Future trials should examine specific and clinically relevant outcomes with strict patient selection criteria. Additionally, parallel advances in immunotherapy and artificial intelligence offer the promise of new multidisciplinary treatment paradigms in the treatment of patients with liver tumors.

Contributor Information

Raazi Bajwa, Memorial Sloan Kettering Cancer Center, Department of Radiology, Division of Interventional Radiology, New York, NY, USA.

David C. Madoff, Yale University School of Medicine, Department of Radiology and Biomedical Imaging, Division of Interventional Radiology, New Haven, CT, USA.

Sirish A. Kishore, Memorial Sloan Kettering Cancer Center, Department of Radiology, Division of Interventional Radiology New York, NY, USA.

References

  • 1.Akinyemiju T, Abera S, Ahmed M, et al. The Burden of Primary Liver Cancer and Underlying Etiologies From 1990 to 2015 at the Global, Regional, and National Level. JAMA Oncol. 2017;3(12):1683. doi: 10.1001/jamaoncol.2017.3055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bosch FX, Ribes J, Díaz M, Cléries R. Primary liver cancer: Worldwide incidence and trends. In: Gastroenterology. Vol 127 W.B. Saunders; 2004. doi: 10.1053/j.gastro.2004.09.011 [DOI] [PubMed] [Google Scholar]
  • 3.Vargas GM, Parmar AD, Sheffield KM, Tamirisa NP, Brown KM, Riall TS. Impact of liver-directed therapy in colorectal cancer liver metastases. J Surg Res. 2014;191(1):42–50. doi: 10.1016/j.jss.2014.05.070 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.John Park MY-JCMW-PLMYFM. The Evolution of Liver-Directed Treatments for Hepatic Colorectal Metastases. 2014. [PubMed]
  • 5.Nanashima A, Takeshita H, Abo T, et al. Recent advances and significance of intra-arterial infusion chemotherapy in non-resectable colorectal liver metastasis. J Gastrointest Oncol. 2013;4(2):164–172. doi: 10.3978/j.issn.2078-6891.2013.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Chapman WC, Klintmalm G, Hemming A, et al. Surgical treatment of hepatocellular carcinoma in North America: can hepatic resection still be justified? J Am Coll Surg. 2015;220(4):628–637. doi: 10.1016/j.jamcollsurg.2014.12.030 [DOI] [PubMed] [Google Scholar]
  • 7.Chegai F, Orlacchio A, Merolla S, Monti S, Mannelli L. Intermediate hepatocellular carcinoma: the role of transarterial therapy. Hepatic Oncol. 2015;2(4):399–408. doi: 10.2217/hep.15.32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.BREEDIS CYOUNG G. The blood supply of neoplasms in the liver. Am J Pathol. 1954;30(5):969–977. [PMC free article] [PubMed] [Google Scholar]
  • 9.Kessel David RI. Interventional Radiology A Survival Guide. Third Edit Elsevier; 2011. [Google Scholar]
  • 10.Vaidya S, Tozer KR, Chen J. An overview of embolic agents. Semin Intervent Radiol. 2008;25(3):204–215. doi: 10.1055/s-0028-1085930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kritzinger J, Klass D, Ho S, et al. Hepatic embolotherapy in interventional oncology: Technology, techniques, and applications. Clin Radiol. 2013;68(1):1–15. doi: 10.1016/j.crad.2012.06.112 [DOI] [PubMed] [Google Scholar]
  • 12.Mahnken AH, Pereira PL, De Baère T. Interventional oncologic approaches to liver metastases. Radiology. 2013;266(2):407–430. doi: 10.1148/radiol.12112544 [DOI] [PubMed] [Google Scholar]
  • 13.Zechlinski JJ, Rilling WS. Transarterial therapies for the treatment of intrahepatic cholangiocarcinoma. Semin Intervent Radiol. 2013;30(1):21–27. doi: 10.1055/s-0033-1333650 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Duran R, Chapiro J, Schernthaner RE, Geschwind J-FH. Systematic review of catheter-based intra-arterial therapies in hepatocellular carcinoma: state of the art and future directions. Br J Radiol. 2015;88(1052):20140564. doi: 10.1259/bjr.20140564 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Rand T, Loewe C, Schoder M, et al. Arterial Embolization of Unresectable Hepatocellular Carcinoma with Use of Microspheres, Lipiodol, and Cyanoacrylate. Cardiovasc Intervent Radiol. 2005;28(3):313–318. doi: 10.1007/s00270-004-0153-y [DOI] [PubMed] [Google Scholar]
  • 16.Maluccio MA, Covey AM, Porat L Ben, et al. Transcatheter arterial embolization with only particles for the treatment of unresectable hepatocellular carcinoma. J Vasc Interv Radiol. 2008;19(6):862–869. doi: 10.1016/j.jvir.2008.02.013 [DOI] [PubMed] [Google Scholar]
  • 17.Varela M, Real MI, Burrel M, et al. Chemoembolization of hepatocellular carcinoma with drug eluting beads: Efficacy and doxorubicin pharmacokinetics. J Hepatol. 2007;46(3):474–481. doi: 10.1016/j.jhep.2006.10.020 [DOI] [PubMed] [Google Scholar]
  • 18.Lee EW, Khan S. Recent advances in transarterial embolotherapies in the treatment of hepatocellular carcinoma. Clin Mol Hepatol. 2017;23(4):265–272. doi: 10.3350/cmh.2017.0111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Liu DM, Thakor AS, Baerlocher M, et al. A review of conventional and drug-eluting chemoembolization in the treatment of colorectal liver metastases: Principles and proof. Futur Oncol. 2015;11(9):1421–1428. doi: 10.2217/fon.15.3 [DOI] [PubMed] [Google Scholar]
  • 20.Bhagat N, Reyes DK, Lin M, et al. Phase II study of chemoembolization with drug-eluting beads in patients with hepatic neuroendocrine metastases: High incidence of biliary injury. Cardiovasc Intervent Radiol. 2013;36(2):449–459. doi: 10.1007/s00270-012-0424-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Brown DB, Geschwind JFH, Soulen MC, Millward SF, Sacks D. Society of Interventional Radiology Position Statement on Chemoembolization of Hepatic Malignancies. J Vasc Interv Radiol. 2009;20(7 SUPPL.). doi: 10.1016/j.jvir.2009.04.015 [DOI] [PubMed] [Google Scholar]
  • 22.Lewandowski RJ, Wang D, Gehl J, et al. A Comparison of Chemoembolization Endpoints Using Angiographic versus Transcatheter Intraarterial Perfusion/MR Imaging Monitoring. J Vasc Interv Radiol. 2007;18(10):1249–1257. doi: 10.1016/j.jvir.2007.06.028 [DOI] [PubMed] [Google Scholar]
  • 23.Jin B, Wang D, Lewandowski RJ, et al. Chemoembolization endpoints: Effect on survival among patients with hepatocellular carcinoma. Am J Roentgenol. 2011;196(4):919–928. doi: 10.2214/AJR.10.4770 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hu J, Maybody M, Cao G, et al. Lipiodol retention pattern assessed by cone beam computed tomography during conventional transarterial chemoembolization of hepatocellular carcinoma: accuracy and correlation with response. Cancer Imaging. 2016;16(1):32. doi: 10.1186/s40644-016-0090-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Benson AB, D’Angelica MI, Abbott DE, et al. NCCN Guidelines Insights: Hepatobiliary Cancers, Version 1.2017. J Natl Compr Cancer Netw. 2017;15(5):563–573. doi: 10.6004/jnccn.2017.0059 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Riaz A, Salem R. Liver Tumors: Metastases In: Transcatheter Embolization and Therapy. Springer London; 2010:197–203. doi: 10.1007/978-1-84800-897-7_20 [DOI] [Google Scholar]
  • 27.Kim BK, Kim SU, Kim KA, et al. Complete response at first chemoembolization is still the most robust predictor for favorable outcome in hepatocellular carcinoma. J Hepatol. 2015;62(6):1304–1310. doi: 10.1016/j.jhep.2015.01.022 [DOI] [PubMed] [Google Scholar]
  • 28.Rose SC, Narsinh KH, Isaacson AJ, Fischman AM, Golzarian J. The beauty and bane of pressure-directed embolotherapy: Hemodynamic principles and preliminary clinical evidence. Am J Roentgenol. 2019;212(3):686–695. doi: 10.2214/AJR.18.19975 [DOI] [PubMed] [Google Scholar]
  • 29.Rose SC, Lim GM, Arellano RS, Easter DB, Roberts AC. Technical innovation. Temporary splenic artery balloon occlusion for protection of nonsplenic vascular beds during splenic embolization. Am J Roentgenol. 1998;170(5):1186–1188. doi: 10.2214/ajr.170.5.9574581 [DOI] [PubMed] [Google Scholar]
  • 30.Rose SC, Halstead GD, Narsinh KH. Pressure-Directed Embolization of Hepatic Arteries in a Porcine Model Using a Temporary Occlusion Balloon Microcatheter: Proof of Concept. Cardiovasc Intervent Radiol. 2017;40(11):1769–1776. doi: 10.1007/s00270-017-1753-7 [DOI] [PubMed] [Google Scholar]
  • 31.Maruyama M, Yoshizako T, Nakamura T, Nakamura M, Yoshida R, Kitagaki H. Initial Experience with Balloon-Occluded Trans-catheter Arterial Chemoembolization (B-TACE) for Hepatocellular Carcinoma. Cardiovasc Intervent Radiol. 2016;39(3):359–366. doi: 10.1007/s00270-015-1237-6 [DOI] [PubMed] [Google Scholar]
  • 32.Newgard BJ, Getrajdman GI, Erinjeri JP, et al. Incidence and Consequence of Nontarget Embolization Following Bland Hepatic Arterial Embolization. Cardiovasc Intervent Radiol. 2019;42(8):1135–1141. doi: 10.1007/s00270-019-02229-2 [DOI] [PubMed] [Google Scholar]
  • 33.Arepally A, Chomas J, Kraitchman D, Hong K. Quantification and reduction of reflux during embolotherapy using an antireflux catheter and tantalum microspheres: Ex vivo analysis. J Vasc Interv Radiol. 2013;24(4):575–580. doi: 10.1016/j.jvir.2012.12.018 [DOI] [PubMed] [Google Scholar]
  • 34.Titano JJ, Fischman AM, Cherian A, et al. End-hole Versus Microvalve Infusion Catheters in Patients Undergoing Drug-Eluting Microspheres-TACE for Solitary Hepatocellular Carcinoma Tumors: A Retrospective Analysis. Cardiovasc Intervent Radiol 2019;42(4):560–568. doi: 10.1007/s00270-018-2150-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Piscaglia F, Ogasawara S. Patient Selection for Transarterial Chemoembolization in Hepatocellular Carcinoma: Importance of Benefit/Risk Assessment. Liver Cancer. 2018;7(1):104–119. doi: 10.1159/000485471 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Raoul JL, Sangro B, Forner A, et al. 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(3):212–220. doi: 10.1016/j.ctrv.2010.07.006 [DOI] [PubMed] [Google Scholar]
  • 37.Sieghart W, Hucke F, Peck-Radosavljevic M. Transarterial chemoembolization: Modalities, indication, and patient selection. J Hepatol. 2015;62(5):1187–1195. doi: 10.1016/j.jhep.2015.02.010 [DOI] [PubMed] [Google Scholar]
  • 38.Lange N, Dufour JF. Changing Epidemiology of HCC: How to Screen and Identify Patients at Risk? Dig Dis Sci. 2019. doi: 10.1007/s10620-019-05515-8 [DOI] [PubMed] [Google Scholar]
  • 39.Bruix J, Reig M, Sherman M. Evidence-Based Diagnosis, Staging, and Treatment of Patients With Hepatocellular Carcinoma. Gastroenterology. 2016;150(4):835–853. doi: 10.1053/j.gastro.2015.12.041 [DOI] [PubMed] [Google Scholar]
  • 40.Kim RD, Kim JS, Watanabe G, Mohuczy D, Behrns KE. Liver regeneration and the atrophy-hypertrophy complex. Semin Intervent Radiol. 2008;25(2):92–103. doi: 10.1055/s-2008-1076679 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Tustumi F, Ernani L, Coelho FF, et al. Preoperative strategies to improve resectability for hepatocellular carcinoma: a systematic review and meta-analysis. HPB. 2018;20(12):1109–1118. doi: 10.1016/j.hpb.2018.06.1798 [DOI] [PubMed] [Google Scholar]
  • 42.Glantzounis GK, Tokidis E, Basourakos S-P, Ntzani EE, Lianos GD, Pentheroudakis G. The role of portal vein embolization in the surgical management of primary hepatobiliary cancers. A systematic review. Eur J Surg Oncol. 2017;43(1):32–41. doi: 10.1016/j.ejso.2016.05.026 [DOI] [PubMed] [Google Scholar]
  • 43.Yoo H, Kim JH, Ko G-Y, et al. Sequential Transcatheter Arterial Chemoembolization and Portal Vein Embolization versus Portal Vein Embolization Only before Major Hepatectomy for Patients with Hepatocellular Carcinoma. Ann Surg Oncol. 2011;18(5):1251–1257. doi: 10.1245/s10434-010-1423-3 [DOI] [PubMed] [Google Scholar]
  • 44.Vouche M, Lewandowski RJ, Atassi R, et al. Radiation lobectomy: Time-dependent analysis of future liver remnant volume in unresectable liver cancer as a bridge to resection. J Hepatol. 2013;59(5):1029–1036. doi: 10.1016/j.jhep.2013.06.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Teo J-Y, Allen JC, Ng DC, et al. A systematic review of contralateral liver lobe hypertrophy after unilobar selective internal radiation therapy with Y90. HPB. 2016;18(1):7–12. doi: 10.1016/J.HPB.2015.07.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Labgaa I, Tabrizian P, Titano J, et al. Feasibility and safety of liver transplantation or resection after transarterial radioembolization with Yttrium-90 for unresectable hepatocellular carcinoma. HPB. 2019;21(11):1497–1504. doi: 10.1016/j.hpb.2019.03.360 [DOI] [PubMed] [Google Scholar]
  • 47.Coletta M, Nicolini D, Benedetti Cacciaguerra A, Mazzocato S, Rossi R, Vivarelli M. Bridging patients with hepatocellular cancer waiting for liver transplant: all the patients are the same? Transl Gastroenterol Hepatol. 2017;2:78. doi: 10.21037/tgh.2017.09.01 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Mehta N, Dodge JL, Goel A, Roberts JP, Hirose R, Yao FY. Identification of liver transplant candidates with hepatocellular carcinoma and a very low dropout risk: Implications for the current organ allocation policy. Liver Transplant. 2013;19(12):1343–1353. doi: 10.1002/lt.23753 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Tohme S, Sukato D, Chen H-W, et al. Yttrium-90 radioembolization as a bridge to liver transplantation: a single-institution experience. J Vasc Interv Radiol. 2013;24(11):1632–1638. doi: 10.1016/j.jvir.2013.07.026 [DOI] [PubMed] [Google Scholar]
  • 50.Pauwels X, Azahaf M, Lassailly G, et al. Drug-Eluting Beads Loaded With Doxorubicin (DEBDOX) Chemoembolisation Before Liver Transplantation for Hepatocellular Carcinoma: An Imaging/Histologic Correlation Study. Cardiovasc Intervent Radiol. 2015;38(3):685–692. doi: 10.1007/s00270-014-0967-1 [DOI] [PubMed] [Google Scholar]
  • 51.Hodavance MS, Vikingstad EM, Griffin AS, et al. Effectiveness of Transarterial Embolization of Hepatocellular Carcinoma as a Bridge to Transplantation. J Vasc Interv Radiol. 2016;27(1):39–45. doi: 10.1016/J.JVIR.2015.08.032 [DOI] [PubMed] [Google Scholar]
  • 52.Salem R, Gordon AC, Mouli S, et al. Y90 Radioembolization Significantly Prolongs Time to Progression Compared With Chemoembolization in Patients With Hepatocellular Carcinoma. Gastroenterology. 2016;151(6):1155–1163.e2. doi: 10.1053/j.gastro.2016.08.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.DiNorcia J, Florman SS, Haydel B, et al. Pathologic Response to Pretransplant Locoregional Therapy is Predictive of Patient Outcome After Liver Transplantation for Hepatocellular Carcinoma. Ann Surg. March 2019:1. doi: 10.1097/SLA.0000000000003253 [DOI] [PubMed] [Google Scholar]
  • 54.Agopian VG, Morshedi MM, McWilliams J, et al. Complete Pathologic Response to Pretransplant Locoregional Therapy for Hepatocellular Carcinoma Defines Cancer Cure After Liver Transplantation. Ann Surg. 2015;262(3):536–545. doi: 10.1097/SLA.0000000000001384 [DOI] [PubMed] [Google Scholar]
  • 55.Clavien P-A, Lesurtel M, Bossuyt PMM, et al. Recommendations for liver transplantation for hepatocellular carcinoma: an international consensus conference report. Lancet Oncol. 2012;13(1):e11–22. doi: 10.1016/S1470-2045(11)70175-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Galle PR, Forner A, Llovet JM, et al. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. J Hepatol. 2018;69(1):182–236. doi: 10.1016/j.jhep.2018.03.019 [DOI] [PubMed] [Google Scholar]
  • 57.Heimbach JK, Kulik LM, Finn RS, et al. AASLD guidelines for the treatment of hepatocellular carcinoma. Hepatology. 2018;67(1):358–380. doi: 10.1002/hep.29086 [DOI] [PubMed] [Google Scholar]
  • 58.Yang H-J, Lee J-H, Lee DH, et al. Small Single-Nodule Hepatocellular Carcinoma: Comparison of Transarterial Chemoembolization, Radiofrequency Ablation, and Hepatic Resection by Using Inverse Probability Weighting. Radiology. 2014;271(3):909–918. doi: 10.1148/radiol.13131760 [DOI] [PubMed] [Google Scholar]
  • 59.Miyayama S Ultraselective conventional transarterial chemoembolization: When and how? Clin Mol Hepatol. 2019;25(4):344–353. doi: 10.3350/cmh.2019.0016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kuroda C, Sakurai M, Monden M, et al. Limitation of transcatheter arterial chemoembolization using iodized oil for small hepatocellular carcinoma. A study in resected cases. Cancer. 1991;67(1):81–86. doi: [DOI] [PubMed] [Google Scholar]
  • 61.Kojiro M, Sugihara S, Kakizoe S, Nakashima O, Kiyomatsu K. Hepatocellular carcinoma with sarcomatous change: a special reference to the relationship with anticancer therapy. Cancer Chemother Pharmacol. 1989;23 Suppl:S4–8. doi: 10.1007/bf00647229 [DOI] [PubMed] [Google Scholar]
  • 62.Wang B, Xu H, Gao ZQ, Ning HF, Sun YQ, Cao GW. Increased expression of vascular endothelial growth factor in hepatocellular carcinoma after transcatheter arterial chemoembolization. Acta radiol. 2008;49(5):523–529. doi: 10.1080/02841850801958890 [DOI] [PubMed] [Google Scholar]
  • 63.Zen C, Zen Y, Mitry RR, et al. Mixed phenotype hepatocellular carcinoma after transarterial chemoembolization and liver transplantation. Liver Transplant. 2011;17(8):943–954. doi: 10.1002/lt.22314 [DOI] [PubMed] [Google Scholar]
  • 64.O’Hara R A novel method for imaging pressure and flow in solid tumors of the liver. Cardiovasc Interv Radiol. 2018;41(Suppl 2):17–49. [Google Scholar]
  • 65.Lewandowski RJ, Gabr A, Abouchaleh N, et al. Radiation Segmentectomy: Potential Curative Therapy for Early Hepatocellular Carcinoma. Radiology. 2018;287(3):1050–1058. doi: 10.1148/radiol.2018171768 [DOI] [PubMed] [Google Scholar]
  • 66.Llovet JM, Ducreux M, Lencioni R, et al. EASL-EORTC Clinical Practice Guidelines: Management of hepatocellular carcinoma. J Hepatol. 2012;56(4):908–943. doi: 10.1016/j.jhep.2011.12.001 [DOI] [PubMed] [Google Scholar]
  • 67.Parikh ND, Waljee AK, Singal AG. Downstaging hepatocellular carcinoma: A systematic review and pooled analysis. Liver Transplant. 2015;21(9):1142–1152. doi: 10.1002/lt.24169 [DOI] [PubMed] [Google Scholar]
  • 68.Bryce K, Tsochatzis EA. Downstaging for hepatocellular cancer: Harm or benefit? Transl Gastroenterol Hepatol. 2017;2(DEC). doi: 10.21037/tgh.2017.11.18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Lencioni R, Llovet JM, Han G, et al. Sorafenib or placebo plus TACE with doxorubicin-eluting beads for intermediate stage HCC: The SPACE trial. J Hepatol. 2016;64(5):1090–1098. doi: 10.1016/j.jhep.2016.01.012 [DOI] [PubMed] [Google Scholar]
  • 70.Gordon A, Lewandowski R, Hickey R, et al. Prospective randomized phase 2 study of chemoembolization versus radioembolization in hepatocellular carcinoma: results from the PREMIERE trial. J Vasc Interv Radiol. 2016;27(3):S61–S62. doi: 10.1016/j.jvir.2015.12.168 [DOI] [Google Scholar]
  • 71.Casadei Gardini A, Tamburini E, Iñarrairaegui M, Frassineti GL, Sangro B. Radioembolization versus chemoembolization for unresectable hepatocellular carcinoma: a meta-analysis of randomized trials. Onco Targets Ther. 2018;11:7315–7321. doi: 10.2147/OTT.S175715 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Klompenhouwer EG, Dresen RC, Verslype C, et al. Safety and Efficacy of Transarterial Radioembolisation in Patients with Intermediate or Advanced Stage Hepatocellular Carcinoma Refractory to Chemoembolisation. Cardiovasc Intervent Radiol. 2017;40(12):1882–1890. doi: 10.1007/s00270-017-1739-5 [DOI] [PubMed] [Google Scholar]
  • 73.Marrero JA, Kulik LM, Sirlin CB, et al. Diagnosis, Staging, and Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology. 2018;68(2):723–750. doi: 10.1002/hep.29913 [DOI] [PubMed] [Google Scholar]
  • 74.Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359(4):378–390. doi: 10.1056/NEJMoa0708857 [DOI] [PubMed] [Google Scholar]
  • 75.Georgiades CS, Hong K, D’Angelo M, Geschwind JFH. Safety and efficacy of transarterial chemoembolization in patients with unresectable hepatocellular carcinoma and portal vein thrombosis. J Vasc Interv Radiol. 2005;16(12):1653–1659. doi: 10.1097/01.RVI.0000182185.47500.7A [DOI] [PubMed] [Google Scholar]
  • 76.Silva JP, Berger NG, Tsai S, et al. Transarterial chemoembolization in hepatocellular carcinoma with portal vein tumor thrombosis: a systematic review and meta-analysis. In: HPB. Vol 19 Elsevier B.V.; 2017:659–666. doi: 10.1016/j.hpb.2017.04.016 [DOI] [PubMed] [Google Scholar]
  • 77.Abdalla EK, Stuart K Overview of treatment approaches for hepatocellular carcinoma - UpToDate. https://www-uptodate-com.proxy.library.rcsi.ie/contents/overview-of-treatment-approaches-for-hepatocellular-carcinoma?search=hcc&topicRef=3599&source=see_link#H4. Accessed November 10, 2019. [Google Scholar]
  • 78.Li L, Zhao W, Wang M, et al. Transarterial chemoembolization plus sorafenib for the management of unresectable hepatocellular carcinoma: a systematic review and meta-analysis. BMC Gastroenterol. 2018;18(1):138. doi: 10.1186/s12876-018-0849-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Abdel-Rahman OM, Elsayed Z. Yttrium-90 microsphere radioembolisation for unresectable hepatocellular carcinoma. Cochrane database Syst Rev 2016;2:CD011313. doi: 10.1002/14651858.CD011313.pub2 [DOI] [PubMed] [Google Scholar]
  • 80.Vilgrain V, Pereira H, Assenat E, 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(12):1624–1636. doi: 10.1016/S1470-2045(17)30683-6 [DOI] [PubMed] [Google Scholar]
  • 81.Ricke J, Klümpen HJ, Amthauer H, et al. Impact of combined selective internal radiation therapy and sorafenib on survival in advanced hepatocellular carcinoma. J Hepatol. 2019;71(6):1164–1174. doi: 10.1016/j.jhep.2019.08.006 [DOI] [PubMed] [Google Scholar]
  • 82.Edeline J, Crouzet L, Campillo-Gimenez B, et al. Selective internal radiation therapy compared with sorafenib for hepatocellular carcinoma with portal vein thrombosis. Eur J Nucl Med Mol Imaging. 2016;43(4):635–643. doi: 10.1007/s00259-015-3210-7 [DOI] [PubMed] [Google Scholar]
  • 83.Xing M, Kokabi N, Camacho JC, Kim HS. Prospective longitudinal quality of life and survival outcomes in patients with advanced infiltrative hepatocellular carcinoma and portal vein thrombosis treated with Yttrium-90 radioembolization. BMC Cancer. 2018;18(1):75. doi: 10.1186/s12885-017-3921-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Gupta A, Dixon E. Epidemiology and risk factors: intrahepatic cholangiocarcinoma. HepatoBiliary Surg Nutr. 2017;6(2):101–104. doi: 10.21037/hbsn.2017.01.02 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Tan JCC, Coburn NG, Baxter NN, Kiss A, Law CHL. Surgical management of intrahepatic cholangiocarcinoma - A population-based study. Ann Surg Oncol. 2008;15(2):600–608. doi: 10.1245/s10434-007-9627-x [DOI] [PubMed] [Google Scholar]
  • 86.Valle J, Wasan H, Palmer DH, et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med. 2010;362(14):1273–1281. doi: 10.1056/NEJMoa0908721 [DOI] [PubMed] [Google Scholar]
  • 87.Yang L, Shan J, Shan L, Saxena A, Bester L, Morris DL. Trans-arterial embolisation therapies for unresectable intrahepatic cholangiocarcinoma: A systematic review. J Gastrointest Oncol. 2015;6(5):570–588. doi: 10.3978/j.issn.2078-6891.2015.055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zhen Y, Liu B, Chang Z, Ren H, Liu Z, Zheng J. A pooled analysis of transarterial radioembolization with yttrium-90 microspheres for the treatment of unresectable intrahepatic cholangiocarcinoma. Onco Targets Ther. 2019;12:4489–4498. doi: 10.2147/OTT.S202875 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Valle JW, Borbath I, Khan SA, et al. Biliary cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2016;27:v28–v37. doi: 10.1093/annonc/mdw324 [DOI] [PubMed] [Google Scholar]
  • 90.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69(1):7–34. doi: 10.3322/caac.21551 [DOI] [PubMed] [Google Scholar]
  • 91.Ghiringhelli F, Hennequin A, Drouillard A, Lepage Cô, Faivre J, Bouvier AM. Epidemiology and prognosis of synchronous and metachronous colon cancer metastases: A French population-based study. Dig Liver Dis. 2014;46(9):854–858. doi: 10.1016/j.dld.2014.05.011 [DOI] [PubMed] [Google Scholar]
  • 92.Figueras J, Ramos E, López-Ben S, et al. Surgical treatment of liver metastases from colorectal carcinoma in elderly patients. When is it worthwhile? Clin Transl Oncol. 2007;9(6):392–400. doi: 10.1007/s12094-007-0072-x [DOI] [PubMed] [Google Scholar]
  • 93.Manfredi S, Lepage C, Hatem C, Coatmeur O, Faivre J, Bouvier AM. Epidemiology and management of liver metastases from colorectal cancer. Ann Surg. 2006;244(2):254–259. doi: 10.1097/01.sla.0000217629.94941.cf [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kelly H, Goldberg RM. Systemic therapy for metastatic colorectal cancer: Current options, current evidence. J Clin Oncol. 2005;23(20):4553–4560. doi: 10.1200/JCO.2005.17.749 [DOI] [PubMed] [Google Scholar]
  • 95.Holen KD, Saltz LB. New therapies, new directions: Advances in the systemic treatment of metastatic colorectal cancer. Lancet Oncol. 2001;2(5):290–297. doi: 10.1016/S1470-2045(00)00324-7 [DOI] [PubMed] [Google Scholar]
  • 96.Fiorentini G, Aliberti C, Tilli M, et al. Intra-arterial infusion of irinotecan-loaded drug-eluting beads (DEBIRI) versus intravenous therapy (FOLFIRI) for hepatic metastases from colorectal cancer: final results of a phase III study. Anticancer Res. 2012;32(4):1387–1395. http://www.ncbi.nlm.nih.gov/pubmed/22493375. Accessed January 21, 2020. [PubMed] [Google Scholar]
  • 97.Benson AB, Venook AP, Al-Hawary MM, et al. NCCN Guidelines ® Insights Colon Cancer, Version 2.2018 Featured Updates to the NCCN Guidelines. JNCCN J Natl Compr Cancer Netw. 2018;16(4):359–369. doi: 10.6004/jnccn.2018.0021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Van Cutsem E, Cervantes A, Adam R, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016;27(8):1386–1422. doi: 10.1093/annonc/mdw235 [DOI] [PubMed] [Google Scholar]
  • 99.Kemeny N, Kurilova I, Li J, Camacho JC, Sofocleous CT. Liver-Directed and Systemic Therapies for Colorectal Cancer Liver Metastases. Cardiovasc Intervent Radiol. 2019;42(9):1240–1254. doi: 10.1007/s00270-019-02284-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Martin RCG, Scoggins CR, Schreeder M, et al. Randomized controlled trial of irinotecan drug-eluting beads with simultaneous FOLFOX and bevacizumab for patients with unresectable colorectal liver-limited metastasis. Cancer. 2015;121(20):3649–3658. doi: 10.1002/cncr.29534 [DOI] [PubMed] [Google Scholar]
  • 101.Hendlisz A, Van Den Eynde M, Peeters M, et al. Phase III trial comparing protracted intravenous fluorouracil infusion alone or with yttrium-90 resin microspheres radioembolization for liver-limited metastatic colorectal cancer refractory to standard chemotherapy. J Clin Oncol. 2010;28(23):3687–3694. doi: 10.1200/JCO.2010.28.5643 [DOI] [PubMed] [Google Scholar]
  • 102.Wasan HS, Gibbs P, Sharma N, et al. First-line selective internal radiotherapy plus chemotherapy versus chemotherapy alone in patients with liver metastases from colorectal cancer (FOXFIRE, SIRFLOX, and FOXFIRE-Global): a combined analysis of three multicentre, randomised, phase 3 trials. Lancet Oncol. 2017;18(9):1159–1171. doi: 10.1016/S1470-2045(17)30457-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Zacharias AJ, Jayakrishnan TT, Rajeev R, et al. Comparative effectiveness of hepatic artery based therapies for unresectable colorectal liver metastases: A meta-analysis. PLoS One. 2015;10(10). doi: 10.1371/journal.pone.0139940 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Levy J, Zuckerman J, Garfinkle R, et al. Intra-arterial therapies for unresectable and chemorefractory colorectal cancer liver metastases: a systematic review and meta-analysis. HPB. 2018;20(10):905–915. doi: 10.1016/j.hpb.2018.04.001 [DOI] [PubMed] [Google Scholar]
  • 105.Man D, Wu J, Shen Z, Zhu X. Prognosis of patients with neuroendocrine tumor: A SEER database analysis. Cancer Manag Res. 2018;10:5629–5638. doi: 10.2147/CMAR.S174907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Pavel M, O’Toole D, Costa F, et al. ENETS consensus guidelines update for the management of distant metastatic disease of intestinal, pancreatic, bronchial neuroendocrine neoplasms (NEN) and NEN of unknown primary site. In: Neuroendocrinology. Vol 103 S. Karger AG; 2016:172–185. doi: 10.1159/000443167 [DOI] [PubMed] [Google Scholar]
  • 107.Shah MH, Goldner WS, Halfdanarson TR, et al. Neuroendocrine and adrenal tumors, version 2.2018 featured updates to the nccn guidelines. JNCCN J Natl Compr Cancer Netw. 2018;16(6):693–702. doi: 10.6004/jnccn.2018.0056 [DOI] [PubMed] [Google Scholar]
  • 108.De Baere T, Deschamps F, Tselikas L, et al. GEP-NETs update: Interventional radiology: Role in the treatment of liver metastases from GEP-NETs. Eur J Endocrinol. 2015;172(4):R151–R166. doi: 10.1530/EJE-14-0630 [DOI] [PubMed] [Google Scholar]
  • 109.Dermine S, Palmieri L-J, Lavolé J, et al. Non-Pharmacological Therapeutic Options for Liver Metastases in Advanced Neuroendocrine Tumors. J Clin Med. 2019;8(11):1907. doi: 10.3390/jcm8111907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Maire F, Lombard-Bohas C, O’Toole D, et al. Hepatic arterial embolization versus chemoembolization in the treatment of liver metastases from well-differentiated midgut endocrine tumors: A prospective randomized study. Neuroendocrinology. 2012;96(4):294–300. doi: 10.1159/000336941 [DOI] [PubMed] [Google Scholar]
  • 111.Gupta S, Johnson MM, Murthy R, et al. Hepatic arterial embolization and chemoembolization for the treatment of patients with metastatic neuroendocrine tumors: Variables affecting response rates and survival. Cancer. 2005;104(8):1590–1602. doi: 10.1002/cncr.21389 [DOI] [PubMed] [Google Scholar]
  • 112.Guiu B, Deschamps F, Aho S, et al. Liver/biliary injuries following chemoembolisation of endocrine tumours and hepatocellular carcinoma: Lipiodol vs. drug-eluting beads. J Hepatol. 2012;56(3):609–617. doi: 10.1016/j.jhep.2011.09.012 [DOI] [PubMed] [Google Scholar]
  • 113.Walter T, Lepage C, Coriat R, et al. Everolimus after hepatic arterial embolisation therapy of metastases from gastrointestinal neuroendocrine tumours: The FFCD 1104-EVACEL-GTE phase II study. Eur J Cancer. 2019;123:92–100. doi: 10.1016/j.ejca.2019.09.021 [DOI] [PubMed] [Google Scholar]
  • 114.Liao XF, Yi JL, Li XR, Deng W, Yang ZF, Tian G. Angiogenesis in rabbit hepatic tumor after transcatheter arterial embolization. World J Gastroenterol. 2004;10(13):1885–1889. doi: 10.3748/wjg.v10.i13.1885 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Zener R, Yoon H, Ziv E, et al. Outcomes After Transarterial Embolization of Neuroendocrine Tumor Liver Metastases Using Spherical Particles of Different Sizes. Cardiovasc Intervent Radiol. 2019;42(4):569–576. doi: 10.1007/s00270-018-02160-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Strosberg JR, Weber JM, Choi J, et al. A phase ii clinical trial of sunitinib following hepatic transarterial embolization for metastatic neuroendocrine tumors. Ann Oncol. 2012;23(9):2335–2341. doi: 10.1093/annonc/mdr614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Su Y-K, Mackey RV, Riaz A, et al. Long-Term Hepatotoxicity of Yttrium-90 Radioembolization as Treatment of Metastatic Neuroendocrine Tumor to the Liver. J Vasc Interv Radiol. 2017;28(11):1520–1526. doi: 10.1016/j.jvir.2017.05.011 [DOI] [PubMed] [Google Scholar]
  • 118.Tomozawa Y, Jahangiri Y, Pathak P, et al. Long-Term Toxicity after Transarterial Radioembolization with Yttrium-90 Using Resin Microspheres for Neuroendocrine Tumor Liver Metastases. J Vasc Interv Radiol. 2018;29(6):858–865. doi: 10.1016/j.jvir.2018.02.002 [DOI] [PubMed] [Google Scholar]
  • 119.de Mestier L, Zappa M, Hentic O, Vilgrain V, Ruszniewski P. Liver transarterial embolizations in metastatic neuroendocrine tumors. Rev Endocr Metab Disord. 2017;18(4):459–471. doi: 10.1007/s11154-017-9431-2 [DOI] [PubMed] [Google Scholar]
  • 120.Gaba RC, Lakhoo J. Yttrium-90 microsphere radioembolization for treatment of lung cancer hepatic metastases. Case Rep Oncol. 2012;5(2):479–486. doi: 10.1159/000342706 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Kis B, Shah J, Choi J, et al. Transarterial Yttrium-90 Radioembolization Treatment of Patients with Liver-Dominant Metastatic Renal Cell Carcinoma. J Vasc Interv Radiol. 2017;28(2):254–259. doi: 10.1016/j.jvir.2016.09.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Wang M, Zhang J, Ji S, et al. Transarterial chemoembolisation for breast cancer with liver metastasis: A systematic review. Breast. 2017;36:25–30. doi: 10.1016/j.breast.2017.09.001 [DOI] [PubMed] [Google Scholar]
  • 123.Rowcroft A, Loveday BPT, Thomson BNJ, Banting S, Knowles B. Systematic review of liver directed therapy for uveal melanoma hepatic metastases. HPB. 2019. doi: 10.1016/j.hpb.2019.11.002 [DOI] [PubMed] [Google Scholar]
  • 124.Bale R, Putzer D, Schullian P. Local treatment of breast cancer liver metastasis. Cancers (Basel). 2019;11(9). doi: 10.3390/cancers11091341 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Kujala E, Mäkitie T, Kivelä T. Very Long-Term Prognosis of Patients with Malignant Uveal Melanoma. Investig Ophthalmol Vis Sci. 2003;44(11):4651–4659. doi: 10.1167/iovs.03-0538 [DOI] [PubMed] [Google Scholar]
  • 126.Singh M, Durairaj P, Yeung J. Uveal Melanoma: A Review of the Literature. Oncol Ther 2018;6(1):87–104. doi: 10.1007/s40487-018-0056-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Gerbes A, Zoulim F, Tilg H, et al. Gut roundtable meeting paper: selected recent advances in hepatocellular carcinoma. Gut. 2018;67(2):380–388. doi: 10.1136/gutjnl-2017-315068 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Duffy AG, Ulahannan SV., Makorova-Rusher O, et al. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J Hepatol. 2017;66(3):545–551. doi: 10.1016/j.jhep.2016.10.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Dendy MS, Ludwig JM, Stein SM, Kim HS. Locoregional Therapy, Immunotherapy and the Combination in Hepatocellular Carcinoma: Future Directions. Liver cancer. 2019;8(5):326–340. doi: 10.1159/000494843 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Zhan C, Ruohoniemi D, Shanbhogue KP, et al. Safety of Combined Yttrium-90 Radioembolization and Immune Checkpoint Inhibitor Immunotherapy for Hepatocellular Carcinoma. J Vasc Interv Radiol. 2020;31(1):25–34. doi: 10.1016/j.jvir.2019.05.023 [DOI] [PubMed] [Google Scholar]
  • 131.Yordanova A, Eppard E, Kürpig S, et al. Theranostics in nuclear medicine practice. Onco Targets Ther. 2017;10:4821–4828. doi: 10.2147/OTT.S140671 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Strosberg J, El-Haddad G, Wolin E, et al. Phase 3 trial of 177lu-dotatate for midgut neuroendocrine tumors. In: New England Journal of Medicine. Vol 376 Massachussetts Medical Society; 2017:125–135. doi: 10.1056/NEJMoa1607427 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Janib SM, Moses AS, MacKay JA. Imaging and drug delivery using theranostic nanoparticles. Adv Drug Deliv Rev. 2010;62(11):1052–1063. doi: 10.1016/j.addr.2010.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Lee ST, Kulkarni HR, Singh A, Baum RP. Theranostics of Neuroendocrine Tumors. Visc Med. 2017;33(5):358–366. doi: 10.1159/000480383 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Singh A, Zhang J, Kulkarni HBR. Intra-arterial PRRT of SSTR-expressing tumors in patients with hepatic only versus extrahepatic tumor: efficacy and safety evaluation. J Nucl Med. 2019;60(Supplement 1):625 http://jnm.snmjournals.org/content/60/supplement_1/625. Accessed January 25, 2020. [Google Scholar]
  • 136.Lee IJ, Ahn C-H, Cha E-J, Chung IJ, Chung JW, Kim Y Il. Improved drug targeting to liver tumors after intra-arterial delivery using superparamagnetic iron oxide and iodized oil: preclinical study in a rabbit model. Invest Radiol. 2013;48(12):826–833. doi: 10.1097/RLI.0b013e31829c13ef [DOI] [PubMed] [Google Scholar]
  • 137.Tian M, Lu W, Zhang R, et al. Tumor uptake of hollow gold nanospheres after intravenous and intra-arterial injection: PET/CT study in a rabbit VX2 liver cancer model. Mol imaging Biol. 2013;15(5):614–624. doi: 10.1007/s11307-013-0635-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Murata S, Mine T, Sugihara F, et al. Interventional treatment for unresectable hepatocellular carcinoma. World J Gastroenterol. 2014;20(37):13453–13465. doi: 10.3748/wjg.v20.i37.13453 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Morshid A, Elsayes KM, Khalaf AM, et al. A machine learning model to predict hepatocellular carcinoma response to transcatheter arterial chemoembolization. Radiol Artif Intell. 2019;1(5). doi: 10.1148/ryai.2019180021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Letzen B, Wang CJ, Chapiro J. The Role of Artificial Intelligence in Interventional Oncology: A Primer. J Vasc Interv Radiol. 2019;30(1):38–41.e1. doi: 10.1016/j.jvir.2018.08.032 [DOI] [PubMed] [Google Scholar]
  • 141.Reimer RP, Reimer P, Mahnken AH. Assessment of Therapy Response to Transarterial Radioembolization for Liver Metastases by Means of Post-treatment MRI-Based Texture Analysis. Cardiovasc Intervent Radiol. 2018;41(10):1545–1556. doi: 10.1007/s00270-018-2004-2 [DOI] [PubMed] [Google Scholar]

RESOURCES