Skip to main content
Erschienen in: European Journal of Nuclear Medicine and Molecular Imaging 5/2022

Open Access 11.02.2022 | Guidelines

EANM procedure guideline for the treatment of liver cancer and liver metastases with intra-arterial radioactive compounds

verfasst von: M. Weber, M. Lam, C. Chiesa, M. Konijnenberg, M. Cremonesi, P. Flamen, S. Gnesin, L. Bodei, T. Kracmerova, M. Luster, E. Garin, K. Herrmann

Erschienen in: European Journal of Nuclear Medicine and Molecular Imaging | Ausgabe 5/2022

Abstract

Primary liver tumours (i.e. hepatocellular carcinoma (HCC) or intrahepatic cholangiocarcinoma (ICC)) are among the most frequent cancers worldwide. However, only 10–20% of patients are amenable to curative treatment, such as resection or transplant. Liver metastases are most frequently caused by colorectal cancer, which accounts for the second most cancer-related deaths in Europe. In both primary and secondary tumours, radioembolization has been shown to be a safe and effective treatment option. The vast potential of personalized dosimetry has also been shown, resulting in markedly increased response rates and overall survival. In a rapidly evolving therapeutic landscape, the role of radioembolization will be subject to changes. Therefore, the decision for radioembolization should be taken by a multidisciplinary tumour board in accordance with the current clinical guidelines. The purpose of this procedure guideline is to assist the nuclear medicine physician in treating and managing patients undergoing radioembolization treatment.

Preamble

The European Association of Nuclear Medicine (EANM) is a professional non-profit medical association that facilitates communication worldwide among individuals pursuing clinical and research excellence in nuclear medicine. The EANM was founded in 1985. These guidelines are intended to assist practitioners in providing appropriate nuclear medicine care for patients. They are not inflexible rules or requirements of practice and are not intended, nor should they be used, to establish a legal standard of care. The ultimate judgment regarding the propriety of any specific procedure or course of action must be made by medical professionals taking into account the unique circumstances of each case. Thus, there is no implication that an approach differing from the guidelines, standing alone, is below the standard of care. To the contrary, a conscientious practitioner may responsibly adopt a course of action different from that set out in the guidelines when, in the reasonable judgment of the practitioner, such course of action is indicated by the condition of the patient, limitations of available resources or advances in knowledge or technology subsequent to publication of the guidelines. The practice of medicine involves not only the science but also the art of dealing with the prevention, diagnosis, alleviation and treatment of disease. The variety and complexity of human conditions make it impossible to always reach the most appropriate diagnosis or to predict with certainty a particular response to treatment. Therefore, it should be recognised that adherence to these guidelines will not ensure an accurate diagnosis or a successful outcome. All that should be expected is that the practitioner will follow a reasonable course of action based on current knowledge, available resources and the needs of the patient to deliver effective and safe medical care. The sole purpose of these guidelines is to assist practitioners in achieving this objective.
Hinweise
The Therapy, Oncology and Dosimetry Committees collaborated with the aim to revise the European Association of Nuclear Medicine (EANM) guidelines on radioembolization.
This article is part of the Topical Collection on Oncology - Digestive tract

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Purpose

The purpose of this guideline is to assist nuclear medicine physicians in:
1.
Patient selection: evaluating patients who might be candidates for treatment using intra-arterial radioactive microspheres for primary or secondary liver cancer.
 
2.
Treatment procedures: providing information on treatment methods.
 
3.
Clinical follow-up: understanding and evaluating efficacy and toxicity.
 
These guidelines represent an update of the 2011 EANM guidelines on the treatment of liver cancer and liver metastases with intra-arterial radioactive compounds. Information still considered to be up to date was retained. Due to the limited use of 131I-lipiodol in Europe, no further update will be provided on this specific treatment modality. In the light of recent trials, the main focus of this update will instead be placed on dosimetric concepts in different treatment scenarios and the use of the newly introduced 166Ho-microspheres.

Background information and definitions

Definitions

Radionuclides

90Y is a beta-emitting radionuclide with a physical half-life of 2.67 days (64.2 h), without emission of gamma photons but with the emission of secondary “bremsstrahlung” photons. 90Y also emits positrons in 32 decays over a million. Despite this scarcity, 90Y PET imaging is routinely performed post-treatment. The maximum and mean beta particle energies are 2.28 and 0.94 MeV, respectively. The maximum and mean ranges in soft tissue are 11 and 4 mm, respectively [1].
166Ho is a beta-emitting radionuclide with a physical half-life of 1.1 days (26.8 h). The beta particle energies include 1.85 MeV (50.0%) and 1.77 MeV (48.7%). The maximum and mean ranges in soft tissues are 8.7 and 2.2 mm, respectively. 166Ho emits gamma photons at 80 keV (yield 6.7%) and 1.4 MeV (yield 0.9%).
99mTc is a metastable nuclear isomer of 99Tc with a half-life of 6.0 h. It emits gamma photons with a photon energy of 140 keV.

Radioactive microspheres (Table 1)

Resin microspheres are made of an acrylic polymer with a median size of 30 μm in diameter, in which 90Y is bound to the carboxylic group on the surface of the polymer after production of microspheres.
Table 1
Radioembolization microspheres characteristics
Characteristics
SIR-Spheres®
TheraSphere®
QuiremSpheres®
Material
Resin
Glass
Poly-L-lactic acid
Particle size and range (μm)
30 (20–60)
25 (20–30)
30 (15–60)
Embolic effect
Moderate
Mild
Moderate
Activity per sphere (Bq)
40–70
4534 *
200–400
Specific gravity (g/dL)
1.6
3.7
1.4
Activity available (GBq)
3#
3–20^ “
Handling for dispensing
Required
Not required
Not required
Multiple dosing from one vial
Possible
Not possible
Not possible
Modified from Salem and Thurston [2], Smits [3] and Westcott [4]
*Direct measure by Pasciak et al. [5] at calibration, the IFU provide a value of 2500 Bq. The value is variable according to physical decay depending on the day and time of treatment
#Prescribed activity should be withdrawn on site. The FLEXdose option allows injection 3 days before calibration, when the vial activity is 10 GBq
^Vials of 3–20 GBq in steps of 0.5 GBq, calibrated at noon on the Sunday before treatment with a shelf-life of 12 days
“Patient-specific activity is calibrated at the day and time of treatment
Glass microspheres are made of glass with a median size of 25 μm, in which 89Y, embedded in the glass matrix, is activated to 90Y in a nuclear reactor.
166Ho-microspheres are made of poly-L-lactic acid with a median size of 30 μm, in which 165Ho, embedded in the matrix, is activated to 166Ho by neutron activation in a nuclear reactor.
99mTc-macroaggregated albumin (MAA) particles mostly have a size between 10 and 100 μm; 99mTc-human serum albumin (HSA) particles mostly have a size between 15 and 50 μm. Both are synthesized by labeling of MAA and HSA-kits with 99mTc, respectively.

Background

The treatment of hepatic malignancies via the hepatic arterial route is based on the existence of arterial tumoural hypervascularization. Tumours bigger than 2 cm in diameter draw more than 80% of their blood supply from the hepatic artery. Normal liver parenchyma draws more than 80% of blood from the portal vein. Highly selective tumour targeting can thus be achieved by delivery of radioactive microspheres into the hepatic artery [68].
Unlike hepatocellular carcinoma (HCC), liver metastases may have variable vascularity, from avascular hepatic cysts to normal hepatic parenchyma, less vascularised metastatic lesions (e.g. colon, pancreas, breast) and finally hypervascular metastases (e.g. renal, neuroendocrine, thyroid).
In 90Y-microspheres radioembolization, pre-treatment intra-arterial 99mTc-labelled albumin macroaggregated albumin1 (99mTc-MAA) scintigraphy2 (see below) is mandatory to quantify potential liver-lung shunting and to exclude reflux to bowel, stomach or pancreas [9, 10]. Note that the use of 99mTc-MAA (although accepted worldwide) is in principle off-label, since its use is indicated only for lung perfusion scintigraphy. In 166Ho-microspheres radioembolization, the administration of a scout dose of 166Ho-microspheres (i.e. 250 MBq) has been shown to be safe and more accurate for the calculation of the lung shunt fraction when compared to 99mTc-MAA [11, 12]. The therapeutic effect of radioembolization is essentially driven by a radiation effect, as opposed to the ischaemia associated with chemoembolization or bland embolization. The radiobiological effect results from beta particle irradiation, which causes delayed death of tumour cells surrounding microvessels containing a high radioactive microspheres concentration [1319].
One of the main differences between the different microspheres is the specific activity of each microsphere at the calibration time, being much higher for glass microspheres (i.e. approximately 4500 Bq/microsphere at calibration), in contrast to approximately 340 Bq/microsphere for 166Ho-microspheres and 50 Bq/microsphere for resin microspheres.
Commercially available vials of glass microspheres contain up to 20 GBq, 166Ho-microspheres up to approximately 15 GBq and resin microspheres 10 GBq fixed. Consequently, at the same prescribed activity, glass microspheres have the least embolic effect, being injected in much lower numbers. Potentially, at the same prescribed activity, the higher number of resin microspheres may provide a more uniform dose distribution, with a higher biological effect (i.e. toxicity and efficacy), with 166Ho-microspheres falling in between. This argument holds if the clustering effect is neglected. Being present in both kinds of 90Y-microspheres [16], the optimal number of microspheres to obtain uniform coverage is still unclear [20]. A theoretical disadvantage of glass microspheres is the often quoted, but never demonstrated influence of gravity on their biodistribution [2124]. Resin microspheres are also available at higher specific activity, the so-called 1-, 2- or 3-day calibration, with 3 GBq activity calibrated at day 0, but supplied days before to increase specific activity.

Indication

Primary or secondary liver tumours. With mounting evidence, the indication with regard to tumour type and specific clinical settings will rapidly change over the years. This falls outside the scope of this procedural guideline.

Contraindications

Absolute
  • Pregnancy, breastfeeding
  • Life expectancy of less than 3 months
  • Clinical liver failure (i.e. ascites, icterus, encephalopathy)
  • Disseminated extrahepatic malignant disease (see section Diagnostic work-up for reference)
  • In case the pre-treatment intra-arterial scout dose scintigraphy (or peri-procedural C-arm CT) shows any extrahepatic activity (or contrast enhancement) in the gastrointestinal tract that cannot be corrected by angiographic techniques (exceptions include the gallbladder, lymph nodes, falciform ligament)
Relative
  • Child-Pugh score higher than B7. A liver decompensation rate as high as 89% after glass administration with standard dosage (i.e. single compartment modelling) has been reported in patients with B7 liver cirrhosis [68]. Caution is warranted in any treatment that is not (bi-)segmental.
  • High intrahepatic tumour burden. Depending on tumour type, more (e.g. neuroendocrine, more indolent, hypervascular, symptomatic disease in need of palliation) or less (hepatocellular carcinoma, underlying liver disease, more aggressive) tumour burden is acceptable. A cut-off of 50–70% is often reported.
  • High extrahepatic tumour burden. Depending on tumour type, more (e.g. neuroendocrine, prognosis depends on liver disease, more indolent) or less (e.g. intrahepatic cholangiocarcinoma (ICC), more aggressive) extrahepatic disease is acceptable. Hilar lymph nodes (up to 2 cm short axis) and lung nodules (up to 1 cm; up to 5) are often accepted.
  • Main portal vein thrombosis (PVT) with poor targeting evidenced by scintigraphy. These patients will have a very poor outcome with little benefit from treatment.
  • Poor targeting of portal vein thrombosis in the main trunk.
  • Acute or severe chronic renal failure (i.e. creatinine clearance <30 ml/min).
  • Contraindications to hepatic artery catheterization (e.g. unmanageable coagulation disorder, renal failure, severe allergy to contrast media, vascular abnormalities).
  • Lung shunting that would lead to a lung dose >30 Gy per session or > 50 Gy cumulatively (determined by pre-treatment planar 99mTc-MAA scintigraphy). Not an absolute contraindication, because lung shunting by planar 99mTc-MAA is overestimated and a reliable safety limit has not been established yet.
Special warnings
  • Inadvertent delivery of microspheres to the gastrointestinal tract or pancreas may cause acute abdominal pain, acute pancreatitis or peptic ulceration.
  • Prior external beam radiation therapy (EBRT) of the liver. Radioembolization appears to be safe for the treatment of hepatic malignancies only in patients who have had limited hepatic exposure to prior EBRT, with the strongest predictor for hepatotoxicity being the liver fraction exposed to ≥30 Gy [25]. While liver regeneration might allow combined treatments, little is known about the cumulative thresholds. Absorbed dose values cannot be simply summed, since a reliable method to combine such values is still lacking.
  • Repeated radioembolization treatments seem less critical than radioembolization after EBRT. Young et al. [26] repeated lobar radioembolization an average of 2.6 times in HCC patients with 17% toxicity incidence. No data is available to strictly demonstrate whether the dose tolerance in a second treatment can be considered the same as for the first treatment. However, liver regeneration might allow this strategy, provided that a sufficient interval between treatments is kept (3 months or longer).
    Radioembolization after peptide radioreceptor therapy (PRRT) of neuroendocrine tumours was reported as safe and effective, with a low incidence (5%) of REILD [27, 28].
  • Markedly abnormal liver function tests (e.g. aspartate aminotransferase or alanine aminotransferase >5 times upper limit of normal or elevated bilirubin). Caution is advised when bilirubin exceeds normal levels, especially in bilobar metastatic disease, in case whole liver treatment is warranted.
  • Excessive radiation to the normal liver parenchyma may result in radiation hepatitis or radioembolization-induced liver disease [29], characterized by the combination of increased bilirubin, low albumin and ascites. The usual onset is 2–6 months after radioembolization [79]. It may co-exist with progressive disease (complicating the clinical presentation), but the diagnosis is definite in the absence of progressive disease.
  • The risk of cholangitis or abscess may be elevated in patients with a history of biliary intervention. Although a direct relation has not been established (in contrast to TACE), caution is advised, and prophylactic antibiotics may be considered.
  • Excessive hepatotoxic systemic therapy prior to radioembolization may increase the risk of post-treatment liver failure. However, in mCRC, it was evidenced in randomized controlled trials that the combination of first- and second-line systemic therapy with radioembolization is safe [3032]. In HCC, the combination of first-line sorafenib and radioembolization proved to be safe [33], but an unacceptable risk of liver failure was identified for ICC patients with underlying cirrhosis treated as first-line with concomitant radioembolization and standard chemotherapy in a multicentre phase II study [34].
  • Non-dosimetric activity calculation methods (single compartment modelling; BSA-method) may lead to under- or overtreatment. Please note that under- and overtreatment may be equally harmful to the patient.

Diagnostic work-up

Clinical and laboratory evaluation

Patients should be accurately staged according to international standards. Clinical history, physical examination, laboratory values and performance status are evaluated. Parameters to assess the indication for radioembolization include determination of tumour load, volume and serum tumour markers (e.g. alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA)). Serum liver enzymes, bilirubin, albumin, cholinesterase, blood cell count, coagulation and creatinine should be monitored and known before the procedure. Selection of patients with adequate hepatic reserve and good functional status will maximize the beneficial therapeutic effect with minimal risk to normal liver parenchyma.

Pre-treatment imaging

Pre-treatment imaging is important to establish feasibility and objectives of treatment, which can extend from palliative treatments to radiation segmentectomy for very limited disease. Imaging techniques include contrast-enhanced CT or MRI performed within 30 days of the procedure for the calculation of the tumour volume and for staging purposes. In addition, (early) arterial phase CT is done to identify hepatic arterial anatomy (e.g. origin of right gastric artery, origin of segment 4 artery). Also, for FDG-avid hepatic malignancies, 18F-fluorodeoxyglucose (FDG) PET is performed for staging purposes, to identify metabolically active liver disease and to rule out prognostically relevant extrahepatic disease. Moreover, metabolic response can be seen 4–6 weeks after radioembolization, whereas response at cross-sectional anatomic imaging may take longer (e.g. 2–3 months) [35, 36]. Both pre- and early post-therapeutic FDG-PET have shown potential to stratify patients with regard to OS and PFS [3740]. Pre-therapeutic hepatobiliary scintigraphy (e.g. using 99mTc-mebrofenin) may be considered, particularly in patients scheduled for partial liver treatment, to measure segmental distribution of liver function and quantify functional liver remnants [41].
There is consensus that radioembolization is generally only indicated in scenarios with no or very limited extrahepatic spread; however, different definitions of limited extrahepatic spread have been employed [30, 4246]. In an analysis of over 1000 patients with primary and metastatic liver neoplasms prospectively included in the observational study ‘CIRSE Registry for SIR-Spheres® Therapy (CIRT)’, low overall survival (OS) was mainly associated with extrahepatic disease extent [46]. In HCC, a sub-analysis of the SORAMIC trial study cohort did not show a negative impact of limited extrahepatic spread, which was defined as involvement of lymph nodes, bones and/or adrenal glands [47]. In mCRC, up to five pulmonary nodules and either lymph nodes belonging to one region [30] or one additional metastatic site amenable to future definitive treatment [43] have been employed as thresholds for limited extrahepatic spread and may be used for reference.
In patients with ICC, lymph node metastases have not shown any negative impact on OS and may therefore not be viewed as exclusion criteria [48]. In the presence of solid organ metastases, caution is warranted, and treatment should be decided on a case-by-case basis depending on whether the intra- or extrahepatic tumour spread is considered life-limiting.

Peri-procedural imaging

Peri-procedural angiography, performed by high-speed multislice CT (angio-CT), cone-beam CT, or digital subtraction angiography, is valuable for procedure planning, in order (1) to assess the hepatic vascular anatomy, (2) to verify the presence or absence of portal venous thrombosis in the portal venous phase, and the possible presence of major arteriovenous malformations or aberrant vasculature, (3) to assess perfused volumes and tumour targeting, (4) to assess intrahepatic interval progression and potential new lesions, and (5) to assess extrahepatic contrast enhancement, precluding safe administration of radioactive microspheres.
99mTc-MAA scintigraphy / 166Ho-microspheres scout dose9
9mTc-MAA is a surrogate for the estimated activity distribution of 90Y- and 166Ho-microspheres. Intrinsic differences in size and rheological properties between MAA [49] and microspheres, together with the operator dependent positioning of the catheter tip in the therapeutic session, are responsible for documented major variations in a minority of patients between MAA predicted and actual therapeutic distribution [50]. Scintigraphy should be performed as soon as possible (preferably within one hour) after administration because of the degradation of 99mTc-MAA (timing of preparation is of less importance); 99mTc-human serum albumin is more resistant to degradation, but also less available [51]. A total of approximately 150 MBq of 99mTc-MAA is administered into the respective branch (or branches) of the hepatic artery.
Planar imaging is used for the first liver-lung shunt calculation. The estimate without attenuation correction gives a large overestimation in comparison with attenuation-corrected evaluations [12, 52]. Scatter correction could also be important, especially for the right lung, strongly influenced by liver photon emission. However, since tolerance doses for the lungs were empirically established using planar imaging, lung doses must be primarily calculated with this approach [5355]. For more accurate calculation, an attenuation and scatter corrected tomographic SPECT/CT covering lung is suggested in cases of substantial lung shunt fraction (> 10%), both pre- and post-treatment.
In the case of multifocal HCC, liver-lung shunting may be assessed before each treatment at the lobar level, because tumours located in different lobes may shunt to varying degrees. The lung shunt fraction is determined by the following equation:
$$\mathrm{Lung}\ \mathrm{shunt}\ \mathrm{fraction}=\frac{ lung\ counts}{lung+ liver\ counts}$$
SPECT (or SPECT/CT) centred on the upper abdomen is advised for the assessment of gastrointestinal shunting,3 appreciation of tumour targeting and better visualization. Moreover, obtained images allow for dosimetric evaluation.
Pre-treatment 99mTc-MAA targeting predicts the 90Y absorbed dose to the non-tumour and (less accurately) to the tumour tissue [56, 57]. It can be used for personalized dosimetry and treatment planning, even if some reports have shown poor prediction of post-treatment 90Y-microsphere distribution by 99mTc-MAA [5860].
Indeed, many studies have demonstrated a clear dose–response relationship based on 99mTc-MAA dosimetry a least for HCC using both glass [6164] or resin microspheres [65, 66].
Most importantly, level 1 evidence of the clinical impact of 99mTc-MAA planning based on personalized dosimetry in large HCC has been provided in a multicentre randomized study comparing standard administration versus 99mTc-MAA-based personalized dosimetry. Statistically significant increases of the response rate and the median OS (26.7 months versus 10.7 months, p = 0.012) were observed in the personalized dosimetry arm [67].
Despite the above-mentioned limitations, 99mTc-MAA is currently the only method available for treatment planning for 90Y-microspheres.
Because the isotope 166Ho also emits gamma rays, the administration of a scout dose (250 MBq divided among injection positions; ±3 million particles) of 166Ho-microspheres is a viable alternative to 99mTc-MAA. It has been shown to be safe [11] and to give a more accurate assessment of the lung shunt fraction [12]. Moreover, a scout dose of 166Ho-microspheres has also been shown to have a superior predictive value in comparison with 99mTc-MAA for the prediction of intrahepatic of 166Ho-microsphere activity distribution [68]. Using the same microspheres for simulation and therapy, at least, the intrinsic problem of particle difference is overcome. Nevertheless, even the 166Ho-microspheres scout dose gives uncertain tumour absorbed dose prediction (95% C.I. of about ±100 Gy), indicating that, with the present available devices, the discrepancy between prediction and treatment is an intrinsic feature of radioembolization.
It is important to emphasize that in order to use 99mTc-MAA or 166Ho-microsphere scout dose for intrahepatic dosimetry, the surrogate used (99mTc-MAA or 166Ho-microspheres) and 90Y/166Ho microspheres should be injected in the same angiographic position, minimizing arterial spasm and the influence of vessel bifurcations, and injected slowly (20–30 s), in order to mimic microsphere infusion [67, 69]. Minimizing arterial spasm includes avoiding (whenever technically possible) coil embolization and favouring (whenever technically possible) the use of floppy catheters [67].
When 99mTc-MAA (or 166Ho-microspheres scout dose) is used to plan the treatment with personalized dosimetry, a minimal pretherapy visual quality control is required evaluating the concordance between the 99mTc-MAA tumour coverage and the CT or MRI tumour vascularity. Large discrepancies mean that the simulation was not accurate (e.g. influence of a bifurcation, spasm) and may be reconsidered.
To optimise the prediction of microsphere distribution, treatment should be performed timely after the diagnostic work up, ideally within 15 days.

Treatment planning

A subset of different clinical scenarios has been described, depending on the tumour burden [69]. The following sections provide definitions of clinical scenarios and general dosimetry recommendations for the respective cohorts.

Radiation segmentectomy

In patients with liver malignancy limited to ≤2 liver segments, higher absorbed doses to the perfused target volume can be administered. This leads to high response rates and long PFS [7072], potentially superior to chemoembolization [73]. The risk is mitigated by the small volume of the irradiated liver. Therefore, it may also entail a viable treatment strategy for patients with a worse liver function. Limited data is available on patients with liver tumours other than HCC. Yet, the available literature suggests that the same principles can be applied [71], but further research is warranted.

Radiation lobectomy

In some patients with unilobar disease, future liver remnants are insufficient to allow for resection. Unilobar treatment allows for higher absorbed doses, since part of the liver remains untreated, aiming for tumour control. Additionally, contralateral lobar hypertrophy can be induced, with the potential of bridging previously ineligible patients to resection with curative intent. Radiation lobectomy is also a feasible strategy in downstaging/bridge-to-transplant settings and includes a biologic test of time which may help identify patients most likely to benefit from resection. Hepatobiliary scintigraphy may be considered to assess future liver remnant function at baseline and during follow-up.

Lobar disease with/without PVT

Patients with unilobar disease, who are not amenable for curative surgery because of portal hypertension, cirrhosis, PVT, extrahepatic disease or clinical performance, may be treated with a palliative intent. It is advised to use dosimetry for activity planning, aiming for a sufficient tumour absorbed dose, while keeping the absorbed dose to functional liver tissue below safety limits. Since only part of the liver is treated, higher absorbed doses to functional liver tissue may be acceptable to optimize the tumour absorbed dose. Hepatobiliary scintigraphy may be used to assess the differential function of the treated and non-treated parts of the liver. Targeting of existing PVT is fundamental for treatment success and should be evaluated prior to treatment [74].

Bilobar disease

Patients in whom extensive disease precludes radiation segmentectomy or lobectomy, activity has to be planned with the aim to achieve high tumour absorbed doses with tolerable absorbed doses to functional liver tissue. High tumour absorbed doses have been shown to be correlated with treatment response, whereas high absorbed doses to functional liver parenchyma have been shown to increase the risk for radioembolization-induced liver disease (i.e. REILD). Sequential treatment of both liver lobes may be considered in patients with a dismal balance between dosimetry (i.e. absorbed dose to functional liver) and patient characteristics (e.g. liver cirrhosis, liver volume < 1.5 l, elevated bilirubin). In these cases, the typical interval ranges from 6 weeks to 3 months. A shorter interval may decrease the chance of interval progression but may increase the chance of cumulative toxicity. A longer interval may increase interval recovery but at the potential cost of interval progression. Treatment strategy should take these considerations into account, on an individual patient basis. Because of the documented variability of predictive dosimetry based on 99mTc-MAA SPECT/CT, a sequential lobar treatment has the advantage that the prescribed activity of the second treatment can be adjusted based on the true 90Y-PET/CT based dosimetry of the first treatment. In this setting, shorter interval between treatments is acceptable. Hepatobiliary scintigraphy may be used to assess overall liver function.

Dosimetry

Dosimetry can be performed using a single compartment model, a multi-compartment model or a method using a voxel-based approach. In the single compartment model, there is no distinction between the tumour and the normal liver parenchyma, and a mean dose is evaluated for the perfused volume. In the multi-compartment model, doses are evaluated separately for the tumour and the normal perfused liver. In voxel-based dosimetry, dosimetry is evaluated for each reconstructed voxel. Optimization of radiotherapeutic exposure, indicated in article 56 of the EU Council Directive 2013/59 and advised by a recent EANM position paper [75], requires a separate evaluation of target and nontarget tissues (multi-compartment dosimetry).

Single versus multi-compartment versus voxel-based dosimetry

An obvious limitation of the single compartment model (as advocated for the standard use of glass 90Y-microspheres and 166Ho-microspheres and which can be called standard dosimetry) is that the actual spatial dose distribution of an individual patient accessible with the present scanner spatial resolution is neglected. In general, these methods seek to prevent overdosing to the functional liver parenchyma (and lungs), minimizing the occurrence of radioembolization-induced liver disease. As a consequence, the resulting prescribed activities are likely curbed by toxicity limitations of the most vulnerable patients and the occurrence of patients with a highly unfavourable dose distribution. This is thought to result in underdosing in some patients and overdosing in others.
In the multi-compartment model, a mean dose is individually evaluated for each compartment (tumour, normal liver and lung tissue). By doing so, it allows for the selection of a prescribed activity that maximizes the dose to the tumour tissue while not exceeding toxicity thresholds for the other two compartments. However, as a limitation, it does not consider the heterogeneity of the dose distribution in each compartment.
The respective compartments are usually segmented on an anatomical imaging modality (e.g. contrast-enhanced CT) and registered to the reconstructed tomographic distribution of a functional modality (e.g. SPECT thresholding). In the partition model [76], the activity distribution over the compartments is described by the tumour-to-normal tissue ratio (T/N ratio), expressed as.
\(\frac{T}{N}\)= \(\frac{A_{tumour}\left[\mathrm{MBq}\right]\ }{{\mathrm{M}}_{tumour}\ \left[\mathrm{kg}\right]}\) / \(\frac{A_{normal\ liver}\ \left[\mathrm{MBq}\right]\ }{M_{normal\ liver}\left[\mathrm{kg}\right]}\),
where A and M indicate the activity in and the mass of the tumour (T) and functional liver tissue (N) compartments.
The T/N ratio lays the foundation to calculate the desired treatment activity:
$$A(GBq)=\frac{D(Gy)\times \left(\left[\frac{T}{N}\times {M}_{tumour}\ \left[ kg\right]\right]+{M}_{liver}\left[ kg\right]\right)}{CF\times \left(1- lung\ shunt\ fraction\right)},$$
with CF being the absorbed dose conversion factor [CF(90Y) = 49.67 J/GBq; CF(166Ho) = 14.85 J/GBq].
Earlier implementations of this model had the limit of evaluating only the absorbed dose averaged over many lesions. A more general method capable of providing individual mean lesion absorbed dose evaluation is available [77].
Voxel-based dosimetry allows for the expression of (estimated) dose gradients and non-homogeneities on a small spatial scale, somewhat similar to external beam radiotherapy (EBRT). This contrasts with multi-compartment models, where dose estimates are averaged over each compartment. By including this spatial dimension, voxel-based dosimetry potentially provides a link to the rich EBRT literature on dose–effect relationships, which can be used for planning and outcome assessment. The usefulness of voxel dosimetry in nuclear medicine therapy is under debate [78]. In radioembolization, no study was able to demonstrate its superiority over the mean dose approach [77, 79, 80]. Many software solutions for dosimetry analyses, such as Simplicity™, Velocity Rapidsphere™, or Qsuite™, are now available [50].

Activity calculation and treatment planning

Methodological details on the various technical aspects of dosimetry are reported in the recent EANM 90Y microspheres dosimetry guideline [81]. These guidelines do not apply to 166Ho microspheres, yet in the absence of dedicated guidelines, some of the underlying concepts may be applicable to treatment with 166Ho microspheres.
Whenever possible, multi-compartment dosimetry should be used. Whenever possible means that tumour segmentation is feasible and the clinical data support tumouricidal doses and maximum tolerated dose for the product used, the tumour histology and the indication (curative/palliative).
When multi-compartment dosimetry is not possible (e.g. infiltrative lesion, no/insufficient clinical dosimetry data available), simple single compartment dosimetry applied to the whole liver is conservative and should be used. Approaches described in the instruction for users of each product (e.g. single compartment dosimetry) can be used from a legal point of view, but they do not optimize treatment.
Although the current data on multi-compartment dosimetry indicate a dose–response relationship, reported dose thresholds vary and are mostly derived from retrospective studies [82]. The following sections and Tables 2, 3 and 4 provide an overview of dose thresholds that can be used for orientation, including the level of evidence supporting them. The reported data will be subject to change. Since most of the data was derived from non-comparative, retrospective analyses of dose–effect relationships, these preliminary data are very heterogeneous. Differences exist in terms of segmentation methods/software used, pre- versus post-treatment analysis, response criteria and reported dose metric (e.g. median/mean of response category, ROC analysis, most sensitive/specific for response). The reported numbers are based on current recommendations and reported literature; it is advised to read this literature before implementing dosimetry-based treatment planning in clinical practice.
Table 2
Absorbed dose recommendations for 90Y glass microspheres and the respective level of evidence (LOE)
 
Single compartment
Multi-compartment
Clinical scenario
Perfused volume dose
LOE
Normal liver dose
Tumour dose
LOE
HCC
Segmentectomy
> 400 [83]
3
Not applicable
Lobectomy
> 150 if whole liver dose <150 [67]
140–150 [84]
1*
3
≥ 88** [85]
< 75 (range: 50/90***)
[86]
≥ 205 [67]
≥ 250–300****
3
Unilobar
> 150 if whole liver dose <150 [67]
80–150 [61, 74]
1*
3
< 120** if HR < 30% [67]
< 75 (range: 50/90***) [86]
≥ 205 [67]
≥ 250–300****
1*
3
Bilobar
80–150**** [13, 69, 87]
1, 4
< 50/90*** [86]
≥ 205 [62]
3
ICC
Segmentectomy
> 400 [60]
4
Not applicable
Lobectomy
140–150
4
< 75 (range: 50/90***)
≥ 260 [88]
3
Unilobar
80–150 [89]
3
< 75 (range: 50/90***)
≥ 260 [88]
3
Bilobar
80–150 [89]
3
< 75 (range: 50/90***)
≥ 260 [88]
3
mCRC
Segmentectomy
> 400 [90]
3
Not applicable
Lobectomy
140–150
4
< 75 (range: 50/90***)
≥ 189 [91]
3
Unilobar
80–150 [92]
3
< 75 (range: 50/90***)
≥ 189 [91]
3
Bilobar
80–150 [92]
3
< 75 (range: 50/90***)
≥ 189 [91]
3
HR, hepatic reserve, i.e. untreated liver fraction
*In patients comparable to the DOSISPHERE-01 [67] study population (Child-Pugh A, large lesions, at least 30% of hepatic reserve)
**Dose to the normal perfused liver, based on the first treatment
***Dose to the whole normal liver. In HCC patients with total bilirubin levels >1.1 mg/dl, an upper threshold of 50 Gy should be used; in patients with total bilirubin levels <1.1 mg/dl, the whole normal liver dose should be kept below 90 Gy. Data are derived from unilobar treatments without prior RE only. Since these thresholds have been established in mostly cirrhotic HCC patients, they can be considered safe for non-HCC patients; however, caution is warranted particularly in ICC patients with underlying cirrhosis and after chemotherapy
****For large lesions [67]
Table 3
Absorbed dose recommendations for 90Y resin microspheres and the respective level of evidence (LOE)
 
Single compartment
Multi-compartment
Clinical scenario
Perfused volume dose
LOE
Normal perfused liver dose
Tumour dose
LOE
HCC
Segmentectomy
> 150 [93]
4
Not applicable
Lobectomy
Not recommended
> 70 [93]*
≥ 100–120 [93]
4
Unilobar
< 40 [93]
≥ 100–120 [65]
3
4
Bilobar
< 30**/40 [93]
≥ 100–120 [65]
3
4
ICC
Segmentectomy
> 150 [93]
4
Not applicable
Lobectomy
Not recommended
> 70 [93]
≥ 100–120 [94]
3
4
Unilobar
< 40 [93]
≥ 100–120 *** [94]
3
4
Bilobar
< 30**/40 [93]
≥ 100–120 *** [94]
3
4
mCRC
Segmentectomy
> 150 [93]
4
Not applicable
Lobectomy
Not recommended
> 70 [93]
> 100 **** [93]
4
Unilobar
< 40 [93]
> 100 **** [95]
3
4
Bilobar
< 30**/40 [93]
> 100 **** [95]
3
4
Modified from Levillain et al. [93]
*Dose to the normal perfused liver with a hepatic reserve of >30%
**In pretreated patients or those with compromised liver function
***Longer OS for patients treated with a partition model-derived mean tumour dose of 86 Gy vs. BSA-derived tumour dose of 38 Gy
****Tumour absorbed doses >100 Gy have been associated with higher rates of metabolic complete response, whereas a lower threshold of >40–60 Gy predicted metabolic partial response
Table 4
Absorbed dose recommendations for 166Ho microspheres and the respective level of evidence (LOE)
 
One-compartment
Multi-compartment
Clinical scenario
Perfused volume dose
LOE
Whole normal liver dose
Tumour dose
LOE
HCC
Segmentectomy
60
3
Not applicable
Lobectomy
60
3
< 60
Not available
4
Unilobar
60
3
< 60
Not available
4
Bilobar
60
3
< 40***
Not available
4
ICC
Segmentectomy
60
3
Not applicable
Lobectomy
60
3
< 60
> 150*
3–4
Unilobar
60
3
< 60
> 150*
3–4
Bilobar
60
3
< 40*** [96]
> 150* [96]
3–4
mCRC
Segmentectomy
60
3
Not applicable
Lobectomy
60
3
< 60
> 90** [97]
3
Unilobar
60
3
< 60
> 90** [97]
3
Bilobar
60
3
< 40***
> 90** [97]
3
LOE, level of evidence
*Based on median tumour absorbed dose for stable disease in a mixed population
**Based on 100% sensitivity for response
***Up to 60 Gy in patients with more favourable liver function
Dosimetric planning driven mainly by tumour dose has two major weak points: first, for 90Y microspheres, the MAA lesion dose prediction may have an unacceptably large discrepancy with the actual therapeutic dose. Second, even a post-therapy 90Y PET dose above the proposed thresholds cannot guarantee response due to the overlap in dose distribution of responding and non-responding lesions. Therefore, in patients in whom level 1 evidence is not available [67], the administration of the maximum tolerable whole normal liver dose with a predicted tumour dose above the efficacy threshold constitutes an alternative driving criterion. Tumour dose is anyhow of outstanding importance in planning, since we do have level 1 evidence [60] that it prolongs median OS. In patients, in whom sufficient tumour doses cannot be reached without exceeding the proposed whole normal liver dose, alternative treatments may be considered.
Despite being outside of the recommendations provided in the IFU, doses >150 Gy to the whole liver have been shown to be safe [86], if the whole normal liver dose was <50/90 Gy. Due to lacking data (especially on OS), a general recommendation cannot be given.

Resin 90Y-microspheres

The body surface area (BSA)-based method, initially advocated for resin 90Y-microspheres, was based on the observation that BSA correlates with liver volume in the healthy population [98]. The lack of personalization of activity prescription according to the true liver and tumour volumes of the patient, together with concerns about efficacy of this prescription method after several negative multicentre trials implementing this method (e.g. SIRFLOX, FOXFIRE, SARAH), induced further research on the absorbed dose–response relationship. The results lead to the recently published recommendation of a multidisciplinary expert panel to use the 3-compartimental partition model (or a voxel-based dosimetry method) and no longer the BSA-method or one-compartment approaches for activity prescription [93].

Glass 90Y-microspheres

For glass 90Y-microspheres, the absorbed dose of a compartment (e.g. lung, tumour, normal perfused liver) is calculated using the simplified MIRD formula:
$$D\left[ Gy\right]=\frac{A\left[ GBq\right]x\ 50\left[\frac{J}{GBq}\right]}{M\left[ kg\right]}$$
where D = absorbed dose in the selected compartment, A = activity contained in the selected compartment and M the mass of this compartment.
The compartment mass may be determined using either CT, MRI, PET or 99mTc-MAA SPECT.
Lung dose is calculated, assuming a lung mass of 1 kg, using the following MIRD formula:
$$DLung\left[ Gy\right]= injected\ Activity\left[ GBq\right]\kern0.5em \times LSF\times 50$$
The maximal tolerated dose is 30 Gy for one treatment and 50 Gy for repeated treatments.
The activity calculation is based on the desired mean absorbed dose to the target mass (tumour, perfused liver, normal liver), following
$$A\left[ GBq\right]= Desired\ dose\left[ Gy\right]\times \frac{Mtarget\left[ kg\right]\ }{50\ \left[J/ GBq\right]}$$
It has to be underlined that reported values for multi-compartment dosimetry (personalized dosimetry) were obtained for the use of glass microspheres on week 1 post-calibration, usually between 2- or 4-day post-calibration. No dosimetric data are available for a use on week 2 post-calibration.

166Ho-microspheres

Currently, activity calculation for 166Ho-microspheres is based on a method comparable to the Medical Internal Radiation Dose (MIRD) method used for glass 90Y-microspheres, as described above. The absorbed dose in Gy delivered by beta rays by 1 GBq in 1 kg tissue is 15.87 Gy for 166Ho, under the assumption of homogenous distribution in the target volume and absorption of all energy within that volume. The absorbed dose from gamma rays is relatively negligible. The formula for the prescribed activity is based on a 60 Gy average absorbed dose to the whole liver, following
$$A\left[ GBq\right]=\frac{Desired\ dose\left[ Gy\right]\times Mtarget\left[ kg\right]\ }{15.87\ \left[J/ GBq\right]}$$
According to current instructions for use, the average absorbed dose to the perfused volume may exceed 60 Gy, as long as the average absorbed to the whole liver does not exceed 60 Gy. Multi-compartment modelling is advised to optimize tumour absorbed doses while keeping absorbed doses to functional liver tissue within safety limits.

Administration

For resin 90Y-microspheres, a typical treatment consists of injecting about 40–80 million 90Y-microspheres. Given the higher embolic load, no blind infusions should be performed. The catheter must be placed well distal (> 3–4 cm) to the gastroduodenal artery and any other artery that is supplying blood to the gastrointestinal system. The microspheres are delivered slowly at a rate of no more than 5 ml/min, as rapid delivery may cause reflux. The use of 5% glucose solution should be preferred over sterile water as it reduces stasis and thereby procedural patient discomfort [99]. During the procedure, the radiologist must repeatedly check the position of the catheter to ensure its position and continued forward flow. This is performed by injecting contrast medium through the left-hand port of the delivery set [100].
For glass 90Y-microspheres, a typical treatment consists of injecting 1.2–8 millions of glass 90Y-microspheres. The volume of saline solution required to infuse a vial is low (typically 20–60 ml). Furthermore, given the low number of microspheres infused, the entire vascular bed is never completely saturated and continuous fluoroscopic guidance during the infusion is not necessary. A complete infusion usually requires 20–60 ml and 5 min and should be performed with slow hand injection under free breathing.
For 166Ho-microspheres, the same administration system is used for both the scout dose and the treatment dose. A typical treatment consists of 20–30 million microspheres. Given the embolic load (albeit less compared with resin 90Y-microspheres), no blind infusions should be performed. The catheter must be placed well distal (> 3–4 cm) to the gastroduodenal artery, and any other artery that is supplying blood to the gastrointestinal system. The microspheres are delivered slowly at a rate of no more than 5 ml/min, as rapid delivery may cause reflux. Instead of 5% glucose, ordinary saline is used. During the procedure, administration of microspheres and contrast agent can be alternated by rotation of the dial between the administration and the contrast position, to check the catheter position and continued forward flow.

Post-treatment imaging

For 90Y-microspheres, routine 90Y PET, planar scintigraphy and bremsstrahlung single-photon emission computed tomography with integrated computed tomography (SPECT/CT) can be considered for post-treatment imaging. Treating physicians should confirm sufficient tumour uptake, acceptable pulmonary absorbed dose and the absence of visceral 90Y concentration, in line with the pre-therapeutic 99mTc-MAA SPECT/CT.
Despite the low amount of positron emission (31.86 ± 0.37 × 10−6), due to its high sensitivity and available time-of-flight (TOF) information, PET/CT quantitative imaging and dosimetry of 90Y-microspheres are feasible [101, 102]. In addition, PET/CT allows for a superior quantification and spatial resolution compared with bremsstrahlung SPECT/CT [5]. In patients with a considerable lung shunt fraction on pre-treatment 99mTc-MAA scintigraphy, image acquisition should comprise the whole liver and lungs. Post-treatment planar imaging covering the lung region is advisable to identify unexpected lung shunt and complications that can be investigated more in detail with 3D modality [103, 104]. In the absence of a clinically relevant lung shunt fraction, coverage of the lungs is not necessary. With present PET/CT technology (3D acquisition and < 600 ps TOF resolution), to account for the low true-coincidence rate, emission times of at least 15 min (preferably more) are recommended [5, 105].
Due to the gamma-emitting properties of 166Ho-microspheres, post-treatment imaging can be performed by virtue of SPECT [106], ideally in conjunction with additional low-dose CT.
Upon decay, the isotope 166Ho emits several gamma photons, most of which are 81 keV (abundance 6.7%), 1379 keV (0.9%) or 1581 keV (0.2%). To quantify 166Ho accurately, the SPECT detector has to be set to register photons with an energy window of 7.5% around the photon peak of 81 keV. However, bremsstrahlung photons and other high-energy photons cause image-degrading effects because of scattering. Although this may limit accurate activity quantification, using a down-scatter window and a medium-energy collimator can reduce these effects. Immediately after administration, when the activity in the scanner is above approximately 1420 MBq (i.e. dependent on the SPECT/CT system), there is an excess of gamma photons that significantly increases detector dead time [106]. Dependent on the amount of administered activity, it is advised to perform post-treatment 166Ho SPECT/CT between 2 and 5 days after treatment.
Because of its paramagnetic properties, 166Ho can also be visualized and quantified using MRI techniques. MRI is independent of administered activity; however, because of artefacts, it is limited to tissue without air and metal. MRI has a higher resolution than SPECT/CT and takes less time. The presence of 166Ho accelerates the decay of the T2 vector of tissue. A linear relationship exists between T2* times and 166Ho concentration. This relationship, called the relaxivity (R2*), depends on the strength of the main magnetic field of the scanner and the 166Ho content.
The feasibility of MRI quantification of 166Ho-microspheres was validated in patients using SPECT/CT as a reference standard [107]. The recovery of 166Ho-microspheres in the liver using MRI proved to be sufficient (i.e. > 95%) for dosimetry purposes in patients without local surgical clips. A downside of the paramagnetic effects of 166Ho-microspheres is that its susceptibility artefacts obscure gadolinium enhancement on follow-up scans, which can potentially mask enhancing viable residual lesions.

Facility and personnel

The facilities required will depend on national legislation for the emission of beta- (90Y-microspheres) and beta- and gamma-emitting (166Ho) therapeutics. If required by law, the patient should be admitted to an approved isolation facility comprising an appropriately shielded room and en suite bathroom facilities. The radiation field at 1 m from the patient’s abdomen immediately after administration is 1.14 μSv/h/GBq for 90Y microspheres [108]. This gives 3 μSv/h for typical 2.6 GBq glass microsphere administration, and 1.8 μSv/h for 1.6 GBq resin microspheres. For 166Ho, external exposure at 1-m spans from 8 to 60 μSv/h at 1 m [109].
The facility in which treatment is administered must have appropriate personnel, radiation safety equipment, procedures available for waste handling and disposal, handling of contamination, monitoring personnel for accidental contamination and controlling contamination spread.
The administration of 90Y- and 166Ho-microspheres should be undertaken by trained medical staff with supporting physics and nursing staff. Clinicians involved in unsealed source therapy must be knowledgeable about and compliant with all applicable national and local legislation and regulations.
To summarize, the development and establishment of an interdisciplinary team (interventional radiology; medical, radiation and surgical oncology; transplant surgery; nuclear medicine; hepatology; medical physics; and radiation safety) is crucial to the success of the treatment.

Staff exposure

The most critical steps of exposure are the microsphere injections and, eventually, the microsphere preparation in the radiopharmacy (activity measuring and aliquoting when required).
A comparative study has evaluated occupational exposure for glass and resin 90Y-microspheres [110]. Reported equivalent doses Hp(10) are less than 2 μSv/GBq for both microspheres for preparation and injection. Reported finger exposure with 90Y glass microspheres is 14.0 ± 7.9 μSv/GBq for the operator injecting microspheres and 13.5 ± 5.2 μSv/GBq for the radiopharmacist measuring the vial activity. With 90Y resin microspheres reported finger exposure are 235.5 ± 156 μSv/GBq for the operator injecting microspheres and 295.2 ± 271.9 μSv/GBq for the radiopharmacist measuring the vial activity and aliquoting the vial.
Another group evaluated equivalent doses Hp(10) and finger doses from 166Ho exposure [111]. Reported whole-body doses were less than 3 μSv/GBq. Maximum finger doses of 2.9 ± 0.2 × 103 μSv/GBq and 2.5 ± 0.3 × 103 μSv/GBq (2.9 ± 0.2 μSv/MBq and 2.5 ± 0.3 μSv/MBq) have been reported for the for preparation and injection of 166Ho microspheres, respectively.
It should be taken into consideration that the limited ring sensitivity to beta emissions from 166Ho and 90Y may lead to a severe underestimation of actual absorbed doses. To account for this, measured dose rates can be complemented by an assessment of theoretical dose rates using dedicated software.

Patient information and instruction

Patients should receive both written and verbal information on the procedure prior to therapy. Depending on the respective country’s legislation, informed written consent from the patient should be obtained. Unless performed in a preoperative setting, patients should be told that this therapy is not likely to cure their disease and is a palliative treatment directed to their liver lesion(s). Patients must be informed of the potential side effects of therapy and alternative treatment options. Patients must be advised to reduce unnecessary radiation exposure and contamination to family members and the public. Written instructions should be provided where required.

Radiation protection

Any significant medical conditions should be noted, and contingency plans made in case radiation precautions must be breached for a medical emergency. Concern about radiation exposure should not interfere with the prompt appropriate medical treatment of the patient. Dose rate to the workers should be monitored during the treatment. Written instructions should be provided where required. The required radiation protection attention is different depending on the used radionuclide. It is minimal with 90Y since the external dose rate is low for all products used. With 166Ho, a significantly higher exposure rate after treatment should be considered. For all three products, substantial skin doses may occur in case of contamination during preparation or administration. After the treatment, patients should avoid pregnancy for at least 4 months. In reality, it is unlikely that women of childbearing age will be eligible for this therapy. Anyway, Gulec et al. [112] calculated that pregnancy shortly after a simulated treatment with 90Y-microspheres does not induce a relevant irradiation to the embryo. Long-lived radioisotope impurities have been observed and may be taken into consideration in the context of waste management [113]. They do not pose a radiation risk in the context of post-interventional surgery/biopsy.

Side effects

90Y- and 166Ho-microspheres, common side effects (> 10% incidence; usually mild to moderate):
  • Fatigue
  • Abdominal pain
  • Nausea
  • Fever/cold chills
  • Transitory elevation of liver enzymes
  • Transitory decline in lymphocytes
90Y- and 166Ho-microspheres, possible severe adverse events (< 5%):
  • Radioembolization-induced liver disease (i.e. hyperbilirubinemia, hypoalbuminemia, ascites, typically occurring 2–6 months after treatment, without evidence of disease progression)
  • Non-target irradiation: radiation gastritis, gastrointestinal ulceration, upper gastrointestinal bleeding, pancreatitis, (radiation pneumonitis)

Repeated treatment

Limited data on the feasibility of retreatment with radioembolization is available [114117]. Based on the published literature, retreatment with radioembolization is feasible, has an acceptable toxicity profile and can be considered, especially in patients who responded to the first radioembolization treatment. Indications and contraindications described for primary treatment should be used for orientation. However, caution is warranted as most studies involved small and/or heterogeneous sample sizes. No published data on repeated treatment with multi-compartment dosimetry have been published yet. The cumulative absorbed doses to non-tumour liver tissue should be assessed at all times [69].

Follow-up

Monitoring of side effects should take pre-treatment liver function (e.g. the presence/absence of cirrhosis) and the treatment protocol (e.g. absorbed dose, fraction of treated liver) into account. Generally speaking, first laboratory tests and clinical evaluations should take place around 2 to 4 weeks after treatment. Follow-up examinations should be scheduled according to the results.
The first follow-up cross-sectional imaging may be performed 3 months after radioembolization [69]. Response criteria purely based on lesion size (such as RECIST) ignore the occurrence of necrosis and decreased perfusion observed after local ablative treatment and have shown poor correlation with outcome parameters in HCC patients [118]. To account for this, modified RECIST (mRECIST) criteria rely on size assessment of the viable tumour instead [119]. Modified RECIST criteria have shown superiority in response assessment in HCC patients undergoing local treatment and should therefore be used [118, 120122]. Less data is available on metastatic neuroendocrine neoplasms. Across studies, different response criteria were used [123]. RECIST response was significantly associated with OS; in hypervascular tumours, response assessment pursuant to mRECIST criteria may be beneficial [124, 125]. For ICC, size changes of the viable part of the tumour (as assessed by EASL, mRECIST or CHOI criteria) are associated with OS, whereas this correlation was not shown for conventional RECIST [126, 127]. In mCRC on the other hand, RECIST was used for response assessment in multiple RCTs [24, 30, 43] and correlated with OS, whereas mRECIST did not [128]. Additionally, in mCRC patients, FDG-PET may allow for earlier and improved response assessment compared with conventional imaging [40].

Future directions 

Main challenges for future studies include defining and validating dose thresholds in different clinical scenarios. Additionally, prior studies using BSA- or uni-compartment-based approaches yielded negative results, the efficacy of radioembolization using multi-compartment dosimetry needs to be assessed in randomised controlled trials in comparison/ addition to standard of care treatment.

Acknowledgements

This guideline was brought to the attention of all other EANM Committees and the National Societies of Nuclear Medicine. The comments and suggestions from the French Society of Nuclear medicine and the British Nuclear Medicine Society are highly appreciated and have been considered for this Guideline

Declarations

Liability statement

This guideline summarises the views of the EANM Dosimetry, Oncology and Theranostic, Radiation Protection Committee. It reflects recommendations for which the EANM cannot be held responsible. The recommendations should be taken into context of good practice of nuclear medicine and do not substitute for national and international legal or regulatory provisions

Ethics approval

This article does not contain any studies with human participants performed by any of the authors.

Conflict of interest

M. Weber holds/held consulting roles for Boston Scientific, Terumo, and Lilly. M. Lam is a consultant and received research support from Terumo and Boston Scientific. UMC Utrecht receives royalty payments from Terumo. C. Chiesa has been a consultant for Boston Scientific and TERUMO for guideline development and congress symposia. He received a research grant in 2018 from Boston Scientific.
P. Flamen reports advisory board and consultancy fees for SIRTEX. L. Bodei reports non-remunerated consultant/speaker roles for AAA-Novartis, ITG, Iba, Clovis Oncology, and MTTI and research grants by AAA-Novartis.
E. Garin is a consultant for Boston Scientific (1 grant for DOSISPHERE trial, personal fees). K. Herrmann reports personal fees from Bayer, personal fees and other from Sofie Biosciences, personal fees from SIRTEX, non-financial support from ABX, personal fees from Adacap, personal fees from Curium, personal fees from Endocyte, grants and personal fees from BTG, personal fees from IPSEN, personal fees from Siemens Healthineers, personal fees from GE Healthcare, personal fees from Amgen, personal fees from Novartis, personal fees from ymabs, personal fees from Aktis Oncology, personal fees from Theragnostics, and personal fees from Pharma15, outside the submitted work.
M. Konijnenberg, M. Lustre, M. Cremonesi, S. Gnesin, and T. Kracmerova have nothing to disclose.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Radiologie

Kombi-Abonnement

Mit e.Med Radiologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Radiologie, den Premium-Inhalten der radiologischen Fachzeitschriften, inklusive einer gedruckten Radiologie-Zeitschrift Ihrer Wahl.

Fußnoten
1
99mTc-labelled human serum albumin (HSA) particles are not widely available but can also be used.
 
2
Planar and, possibly, single-photon emission computed tomography with integrated computed tomography (SPECT/CT)
 
3
Oral administration of sodium perchlorate before the MAA injection can be considered to prevent physiological uptake of free 99mTc-pertechnetate in the stomach that can be mistaken for visceral shunting.
 
Literatur
1.
Zurück zum Zitat Cremonesi M, et al. Dosimetry in peptide radionuclide receptor therapy: a review. J Nucl Med. 2006;47(9):1467–75.PubMed Cremonesi M, et al. Dosimetry in peptide radionuclide receptor therapy: a review. J Nucl Med. 2006;47(9):1467–75.PubMed
2.
Zurück zum Zitat Salem R, Thurston KG. Radioembolization with (90)yttrium microspheres: a state-of-the-art brachytherapy treatment for primary and secondary liver malignancies part 1: technical and methodologic considerations. J Vasc Interv Radiol. 2006;17(8):1251–78.PubMedCrossRef Salem R, Thurston KG. Radioembolization with (90)yttrium microspheres: a state-of-the-art brachytherapy treatment for primary and secondary liver malignancies part 1: technical and methodologic considerations. J Vasc Interv Radiol. 2006;17(8):1251–78.PubMedCrossRef
3.
Zurück zum Zitat Smits MLJ, et al., Holmium-166 radioembolization for the treatment of patients with liver metastases: design of the phase I HEPAR trial. Journal of Experimental & Clinical Cancer Research, 2010. 29. Smits MLJ, et al., Holmium-166 radioembolization for the treatment of patients with liver metastases: design of the phase I HEPAR trial. Journal of Experimental & Clinical Cancer Research, 2010. 29.
4.
Zurück zum Zitat Westcott MA, et al. The development, commercialization, and clinical context of yttrium-90 radiolabeled resin and glass microspheres. Adv Radiat Oncol. 2016;1(4):351–64.PubMedPubMedCentralCrossRef Westcott MA, et al. The development, commercialization, and clinical context of yttrium-90 radiolabeled resin and glass microspheres. Adv Radiat Oncol. 2016;1(4):351–64.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Lau WY, Li AK. Therapeutic aspects of radioisotopes in hepatobiliary malignancy. Br J Surg. 1992;79(7):711.PubMedCrossRef Lau WY, Li AK. Therapeutic aspects of radioisotopes in hepatobiliary malignancy. Br J Surg. 1992;79(7):711.PubMedCrossRef
7.
Zurück zum Zitat Okuda K, et al. Natural history of hepatocellular carcinoma and prognosis in relation to treatment. Study of 850 patients. Cancer. 1985;56(4):918–28.PubMedCrossRef Okuda K, et al. Natural history of hepatocellular carcinoma and prognosis in relation to treatment. Study of 850 patients. Cancer. 1985;56(4):918–28.PubMedCrossRef
9.
Zurück zum Zitat Lambert B, et al. 99mTc-labelled macroaggregated albumin (MAA) scintigraphy for planning treatment with 90Y microspheres. Eur J Nucl Med Mol Imaging. 2010;37(12):2328–33.PubMedCrossRef Lambert B, et al. 99mTc-labelled macroaggregated albumin (MAA) scintigraphy for planning treatment with 90Y microspheres. Eur J Nucl Med Mol Imaging. 2010;37(12):2328–33.PubMedCrossRef
10.
Zurück zum Zitat Leung WT, et al. Measuring lung shunting in hepatocellular carcinoma with intrahepatic-arterial technetium-99m macroaggregated albumin. J Nucl Med. 1994;35(1):70–3.PubMed Leung WT, et al. Measuring lung shunting in hepatocellular carcinoma with intrahepatic-arterial technetium-99m macroaggregated albumin. J Nucl Med. 1994;35(1):70–3.PubMed
11.
Zurück zum Zitat Braat A, et al. Safety analysis of holmium-166 microsphere scout dose imaging during radioembolisation work-up: a cohort study. Eur Radiol. 2018;28(3):920–8.PubMedCrossRef Braat A, et al. Safety analysis of holmium-166 microsphere scout dose imaging during radioembolisation work-up: a cohort study. Eur Radiol. 2018;28(3):920–8.PubMedCrossRef
12.
Zurück zum Zitat Elschot M, et al. ((9)(9)m)Tc-MAA overestimates the absorbed dose to the lungs in radioembolization: a quantitative evaluation in patients treated with (1)(6)(6)Ho-microspheres. Eur J Nucl Med Mol Imaging. 2014;41(10):1965–75.PubMedCrossRef Elschot M, et al. ((9)(9)m)Tc-MAA overestimates the absorbed dose to the lungs in radioembolization: a quantitative evaluation in patients treated with (1)(6)(6)Ho-microspheres. Eur J Nucl Med Mol Imaging. 2014;41(10):1965–75.PubMedCrossRef
13.
Zurück zum Zitat Dancey JE, et al. Treatment of nonresectable hepatocellular carcinoma with intrahepatic 90Y-microspheres. J Nucl Med. 2000;41(10):1673–81.PubMed Dancey JE, et al. Treatment of nonresectable hepatocellular carcinoma with intrahepatic 90Y-microspheres. J Nucl Med. 2000;41(10):1673–81.PubMed
14.
Zurück zum Zitat Geschwind JF, et al. Yttrium-90 microspheres for the treatment of hepatocellular carcinoma. Gastroenterology. 2004;127(5 Suppl 1):S194–205.PubMedCrossRef Geschwind JF, et al. Yttrium-90 microspheres for the treatment of hepatocellular carcinoma. Gastroenterology. 2004;127(5 Suppl 1):S194–205.PubMedCrossRef
15.
Zurück zum Zitat Goin JE, et al. Treatment of unresectable hepatocellular carcinoma with intrahepatic yttrium 90 microspheres: a risk-stratification analysis. J Vasc Interv Radiol. 2005;16(2 Pt 1):195–203.PubMedCrossRef Goin JE, et al. Treatment of unresectable hepatocellular carcinoma with intrahepatic yttrium 90 microspheres: a risk-stratification analysis. J Vasc Interv Radiol. 2005;16(2 Pt 1):195–203.PubMedCrossRef
16.
Zurück zum Zitat Kennedy AS, et al. Pathologic response and microdosimetry of (90)Y microspheres in man: review of four explanted whole livers. Int J Radiat Oncol Biol Phys. 2004;60(5):1552–63.PubMedCrossRef Kennedy AS, et al. Pathologic response and microdosimetry of (90)Y microspheres in man: review of four explanted whole livers. Int J Radiat Oncol Biol Phys. 2004;60(5):1552–63.PubMedCrossRef
17.
Zurück zum Zitat Lau WY, et al. Selective internal radiation therapy for nonresectable hepatocellular carcinoma with intraarterial infusion of 90yttrium microspheres. Int J Radiat Oncol Biol Phys. 1998;40(3):583–92.PubMedCrossRef Lau WY, et al. Selective internal radiation therapy for nonresectable hepatocellular carcinoma with intraarterial infusion of 90yttrium microspheres. Int J Radiat Oncol Biol Phys. 1998;40(3):583–92.PubMedCrossRef
18.
Zurück zum Zitat Sharma RA, et al. Radioembolization of liver metastases from colorectal cancer using yttrium-90 microspheres with concomitant systemic oxaliplatin, fluorouracil, and leucovorin chemotherapy. J Clin Oncol. 2007;25(9):1099–106.PubMedCrossRef Sharma RA, et al. Radioembolization of liver metastases from colorectal cancer using yttrium-90 microspheres with concomitant systemic oxaliplatin, fluorouracil, and leucovorin chemotherapy. J Clin Oncol. 2007;25(9):1099–106.PubMedCrossRef
19.
Zurück zum Zitat Stubbs RS, Cannan RJ, Mitchell AW. Selective internal radiation therapy with 90yttrium microspheres for extensive colorectal liver metastases. J Gastrointest Surg. 2001;5(3):294–302.PubMedCrossRef Stubbs RS, Cannan RJ, Mitchell AW. Selective internal radiation therapy with 90yttrium microspheres for extensive colorectal liver metastases. J Gastrointest Surg. 2001;5(3):294–302.PubMedCrossRef
20.
Zurück zum Zitat Hogberg J, et al. Increased absorbed liver dose in selective internal radiation therapy (SIRT) correlates with increased sphere-cluster frequency and absorbed dose inhomogeneity. EJNMMI Phys. 2015;2(1):10.PubMedPubMedCentralCrossRef Hogberg J, et al. Increased absorbed liver dose in selective internal radiation therapy (SIRT) correlates with increased sphere-cluster frequency and absorbed dose inhomogeneity. EJNMMI Phys. 2015;2(1):10.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Carr BI. Hepatic arterial 90Yttrium glass microspheres (Therasphere) for unresectable hepatocellular carcinoma: interim safety and survival data on 65 patients. Liver Transpl. 2004;10(2 Suppl 1):S107–10.PubMedCrossRef Carr BI. Hepatic arterial 90Yttrium glass microspheres (Therasphere) for unresectable hepatocellular carcinoma: interim safety and survival data on 65 patients. Liver Transpl. 2004;10(2 Suppl 1):S107–10.PubMedCrossRef
22.
Zurück zum Zitat Garin E, et al. First experience of hepatic radioembolization using microspheres labelled with yttrium-90 (TheraSphere): practical aspects concerning its implementation. Eur J Nucl Med Mol Imaging. 2010;37(3):453–61.PubMedCrossRef Garin E, et al. First experience of hepatic radioembolization using microspheres labelled with yttrium-90 (TheraSphere): practical aspects concerning its implementation. Eur J Nucl Med Mol Imaging. 2010;37(3):453–61.PubMedCrossRef
23.
Zurück zum Zitat Sangro B, et al. Radioembolization using 90Y-resin microspheres for patients with advanced hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2006;66(3):792–800.PubMedCrossRef Sangro B, et al. Radioembolization using 90Y-resin microspheres for patients with advanced hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2006;66(3):792–800.PubMedCrossRef
24.
Zurück zum Zitat Van Hazel G, et al. Randomised phase 2 trial of SIR-spheres plus fluorouracil/leucovorin chemotherapy versus fluorouracil/leucovorin chemotherapy alone in advanced colorectal cancer. J Surg Oncol. 2004;88(2):78–85.PubMedCrossRef Van Hazel G, et al. Randomised phase 2 trial of SIR-spheres plus fluorouracil/leucovorin chemotherapy versus fluorouracil/leucovorin chemotherapy alone in advanced colorectal cancer. J Surg Oncol. 2004;88(2):78–85.PubMedCrossRef
25.
Zurück zum Zitat Lam MG, et al. Safety of 90Y radioembolization in patients who have undergone previous external beam radiation therapy. Int J Radiat Oncol Biol Phys. 2013;87(2):323–9.PubMedCrossRef Lam MG, et al. Safety of 90Y radioembolization in patients who have undergone previous external beam radiation therapy. Int J Radiat Oncol Biol Phys. 2013;87(2):323–9.PubMedCrossRef
26.
Zurück zum Zitat Young JY, et al. Radiation dose limits and liver toxicities resulting from multiple yttrium-90 radioembolization treatments for hepatocellular carcinoma. J Vasc Interv Radiol. 2007;18(11):1375–82.PubMedCrossRef Young JY, et al. Radiation dose limits and liver toxicities resulting from multiple yttrium-90 radioembolization treatments for hepatocellular carcinoma. J Vasc Interv Radiol. 2007;18(11):1375–82.PubMedCrossRef
27.
Zurück zum Zitat Ezziddin S, et al. 90Y radioembolization after radiation exposure from peptide receptor radionuclide therapy. J Nucl Med. 2012;53(11):1663–9.PubMedCrossRef Ezziddin S, et al. 90Y radioembolization after radiation exposure from peptide receptor radionuclide therapy. J Nucl Med. 2012;53(11):1663–9.PubMedCrossRef
28.
Zurück zum Zitat Braat A, et al. Radioembolization with (90)Y resin microspheres of neuroendocrine liver metastases after initial peptide receptor radionuclide therapy. Cardiovasc Intervent Radiol. 2020;43(2):246–53.PubMedCrossRef Braat A, et al. Radioembolization with (90)Y resin microspheres of neuroendocrine liver metastases after initial peptide receptor radionuclide therapy. Cardiovasc Intervent Radiol. 2020;43(2):246–53.PubMedCrossRef
29.
Zurück zum Zitat Sangro B, et al. Liver disease induced by radioembolization of liver tumors: description and possible risk factors. Cancer. 2008;112(7):1538–46.PubMedCrossRef Sangro B, et al. Liver disease induced by radioembolization of liver tumors: description and possible risk factors. Cancer. 2008;112(7):1538–46.PubMedCrossRef
30.
Zurück zum Zitat van Hazel GA, et al. SIRFLOX: randomized phase III trial comparing first-line mFOLFOX6 (plus or minus bevacizumab) versus mFOLFOX6 (plus or minus bevacizumab) plus selective internal radiation therapy in patients with metastatic colorectal cancer. J Clin Oncol. 2016;34(15):1723–31.PubMedCrossRef van Hazel GA, et al. SIRFLOX: randomized phase III trial comparing first-line mFOLFOX6 (plus or minus bevacizumab) versus mFOLFOX6 (plus or minus bevacizumab) plus selective internal radiation therapy in patients with metastatic colorectal cancer. J Clin Oncol. 2016;34(15):1723–31.PubMedCrossRef
31.
Zurück zum Zitat Mulcahy, M.F., et al., Radioembolization with chemotherapy for colorectal liver metastases: a randomized, open-label, international, multicenter, phase III trial. J Clin Oncol, 2021: p. JCO2101839. Mulcahy, M.F., et al., Radioembolization with chemotherapy for colorectal liver metastases: a randomized, open-label, international, multicenter, phase III trial. J Clin Oncol, 2021: p. JCO2101839.
32.
Zurück zum Zitat Wasan HS, 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):1159–71.PubMedPubMedCentralCrossRef Wasan HS, 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):1159–71.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Ricke J, et al. Impact of combined selective internal radiation therapy and sorafenib on survival in advanced hepatocellular carcinoma. J Hepatol. 2019;71(6):1164–74.PubMedCrossRef Ricke J, et al. Impact of combined selective internal radiation therapy and sorafenib on survival in advanced hepatocellular carcinoma. J Hepatol. 2019;71(6):1164–74.PubMedCrossRef
34.
Zurück zum Zitat Edeline, J., et al., Radioembolization plus chemotherapy for first-line treatment of locally advanced intrahepatic cholangiocarcinoma: a phase 2 clinical trial. JAMA Oncol, 2019. Edeline, J., et al., Radioembolization plus chemotherapy for first-line treatment of locally advanced intrahepatic cholangiocarcinoma: a phase 2 clinical trial. JAMA Oncol, 2019.
35.
Zurück zum Zitat Shady W, et al. Metabolic tumor volume and total lesion glycolysis on FDG-PET/CT can predict overall survival after (90)Y radioembolization of colorectal liver metastases: a comparison with SUVmax, SUVpeak, and RECIST 1.0. Eur J Radiol. 2016;85(6):1224–31.PubMedPubMedCentralCrossRef Shady W, et al. Metabolic tumor volume and total lesion glycolysis on FDG-PET/CT can predict overall survival after (90)Y radioembolization of colorectal liver metastases: a comparison with SUVmax, SUVpeak, and RECIST 1.0. Eur J Radiol. 2016;85(6):1224–31.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Shady W, et al. Surrogate imaging biomarkers of response of colorectal liver metastases after salvage radioembolization using 90Y-loaded resin microspheres. AJR Am J Roentgenol. 2016;207(3):661–70.PubMedPubMedCentralCrossRef Shady W, et al. Surrogate imaging biomarkers of response of colorectal liver metastases after salvage radioembolization using 90Y-loaded resin microspheres. AJR Am J Roentgenol. 2016;207(3):661–70.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Fendler WP, et al. Validation of several SUV-based parameters derived from 18F-FDG PET for prediction of survival after SIRT of hepatic metastases from colorectal cancer. J Nucl Med. 2013;54(8):1202–8.PubMedCrossRef Fendler WP, et al. Validation of several SUV-based parameters derived from 18F-FDG PET for prediction of survival after SIRT of hepatic metastases from colorectal cancer. J Nucl Med. 2013;54(8):1202–8.PubMedCrossRef
38.
Zurück zum Zitat Gulec SA, et al. The prognostic value of functional tumor volume and total lesion glycolysis in patients with colorectal cancer liver metastases undergoing 90Y selective internal radiation therapy plus chemotherapy. Eur J Nucl Med Mol Imaging. 2011;38(7):1289–95.PubMedCrossRef Gulec SA, et al. The prognostic value of functional tumor volume and total lesion glycolysis in patients with colorectal cancer liver metastases undergoing 90Y selective internal radiation therapy plus chemotherapy. Eur J Nucl Med Mol Imaging. 2011;38(7):1289–95.PubMedCrossRef
39.
Zurück zum Zitat Soydal C, et al. The prognostic value of quantitative parameters of 18F-FDG PET/CT in the evaluation of response to internal radiation therapy with yttrium-90 in patients with liver metastases of colorectal cancer. Nucl Med Commun. 2013;34(5):501–6.PubMedCrossRef Soydal C, et al. The prognostic value of quantitative parameters of 18F-FDG PET/CT in the evaluation of response to internal radiation therapy with yttrium-90 in patients with liver metastases of colorectal cancer. Nucl Med Commun. 2013;34(5):501–6.PubMedCrossRef
40.
Zurück zum Zitat Zerizer I, et al. The role of early (1)(8)F-FDG PET/CT in prediction of progression-free survival after (9)(0)Y radioembolization: comparison with RECIST and tumour density criteria. Eur J Nucl Med Mol Imaging. 2012;39(9):1391–9.PubMedCrossRef Zerizer I, et al. The role of early (1)(8)F-FDG PET/CT in prediction of progression-free survival after (9)(0)Y radioembolization: comparison with RECIST and tumour density criteria. Eur J Nucl Med Mol Imaging. 2012;39(9):1391–9.PubMedCrossRef
41.
42.
Zurück zum Zitat Vilgrain V, 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):1624–36.PubMedCrossRef Vilgrain V, 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):1624–36.PubMedCrossRef
43.
Zurück zum Zitat Dutton SJ, et al. FOXFIRE protocol: an open-label, randomised, phase III trial of 5-fluorouracil, oxaliplatin and folinic acid (OxMdG) with or without interventional selective internal radiation therapy (SIRT) as first-line treatment for patients with unresectable liver-only or liver-dominant metastatic colorectal cancer. BMC Cancer. 2014;14:497.PubMedPubMedCentralCrossRef Dutton SJ, et al. FOXFIRE protocol: an open-label, randomised, phase III trial of 5-fluorouracil, oxaliplatin and folinic acid (OxMdG) with or without interventional selective internal radiation therapy (SIRT) as first-line treatment for patients with unresectable liver-only or liver-dominant metastatic colorectal cancer. BMC Cancer. 2014;14:497.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Gray B, et al. Randomised trial of SIR-spheres plus chemotherapy vs. chemotherapy alone for treating patients with liver metastases from primary large bowel cancer. Ann Oncol. 2001;12(12):1711–20.PubMedCrossRef Gray B, et al. Randomised trial of SIR-spheres plus chemotherapy vs. chemotherapy alone for treating patients with liver metastases from primary large bowel cancer. Ann Oncol. 2001;12(12):1711–20.PubMedCrossRef
45.
Zurück zum Zitat Chow PKH, et al. SIRveNIB: selective internal radiation therapy versus sorafenib in Asia-Pacific patients with hepatocellular carcinoma. J Clin Oncol. 2018;36(19):1913–21.PubMedCrossRef Chow PKH, et al. SIRveNIB: selective internal radiation therapy versus sorafenib in Asia-Pacific patients with hepatocellular carcinoma. J Clin Oncol. 2018;36(19):1913–21.PubMedCrossRef
46.
Zurück zum Zitat Helmberger T, et al. Clinical application of trans-arterial radioembolization in hepatic malignancies in europe: first results from the prospective multicentre observational study CIRSE registry for SIR-spheres therapy (CIRT). Cardiovasc Intervent Radiol. 2021;44(1):21–35.PubMedCrossRef Helmberger T, et al. Clinical application of trans-arterial radioembolization in hepatic malignancies in europe: first results from the prospective multicentre observational study CIRSE registry for SIR-spheres therapy (CIRT). Cardiovasc Intervent Radiol. 2021;44(1):21–35.PubMedCrossRef
47.
Zurück zum Zitat Schutte K, et al. Impact of extrahepatic metastases on overall survival in patients with advanced liver dominant hepatocellular carcinoma: a subanalysis of the SORAMIC trial. Liver Cancer. 2020;9(6):771–86.PubMedPubMedCentralCrossRef Schutte K, et al. Impact of extrahepatic metastases on overall survival in patients with advanced liver dominant hepatocellular carcinoma: a subanalysis of the SORAMIC trial. Liver Cancer. 2020;9(6):771–86.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Kohler M, et al. Prognostic factors for overall survival in advanced intrahepatic cholangiocarcinoma treated with yttrium-90 radioembolization. J Clin Med. 2019;9:1.CrossRef Kohler M, et al. Prognostic factors for overall survival in advanced intrahepatic cholangiocarcinoma treated with yttrium-90 radioembolization. J Clin Med. 2019;9:1.CrossRef
49.
Zurück zum Zitat Hunt AP, et al. Preparation of Tc-99m-macroaggregated albumin from recombinant human albumin for lung perfusion imaging. Eur J Pharm Biopharm. 2006;62(1):26–31.PubMedCrossRef Hunt AP, et al. Preparation of Tc-99m-macroaggregated albumin from recombinant human albumin for lung perfusion imaging. Eur J Pharm Biopharm. 2006;62(1):26–31.PubMedCrossRef
50.
Zurück zum Zitat Chiesa C, Maccauro M. (166)Ho microsphere scout dose for more accurate radioembolization treatment planning. Eur J Nucl Med Mol Imaging. 2020;47(4):744–7.PubMedCrossRef Chiesa C, Maccauro M. (166)Ho microsphere scout dose for more accurate radioembolization treatment planning. Eur J Nucl Med Mol Imaging. 2020;47(4):744–7.PubMedCrossRef
51.
Zurück zum Zitat Grosser OS, et al. Pharmacokinetics of 99mTc-MAA- and 99mTc-HSA-microspheres used in preradioembolization dosimetry: influence on the liver-lung shunt. J Nucl Med. 2016;57(6):925–7.PubMedCrossRef Grosser OS, et al. Pharmacokinetics of 99mTc-MAA- and 99mTc-HSA-microspheres used in preradioembolization dosimetry: influence on the liver-lung shunt. J Nucl Med. 2016;57(6):925–7.PubMedCrossRef
52.
Zurück zum Zitat Lopez B, et al. Calculation of lung mean dose and quantification of error for (90) Y-microsphere radioembolization using (99m) Tc-MAA SPECT/CT and diagnostic chest CT. Med Phys. 2019;46(9):3929–40.PubMedCrossRef Lopez B, et al. Calculation of lung mean dose and quantification of error for (90) Y-microsphere radioembolization using (99m) Tc-MAA SPECT/CT and diagnostic chest CT. Med Phys. 2019;46(9):3929–40.PubMedCrossRef
53.
Zurück zum Zitat Das A, et al. Safety and efficacy of radioembolization with glass microspheres in hepatocellular carcinoma patients with elevated lung shunt fraction: analysis of a 103-patient cohort. Eur J Nucl Med Mol Imaging. 2020;47(4):807–15.PubMedCrossRef Das A, et al. Safety and efficacy of radioembolization with glass microspheres in hepatocellular carcinoma patients with elevated lung shunt fraction: analysis of a 103-patient cohort. Eur J Nucl Med Mol Imaging. 2020;47(4):807–15.PubMedCrossRef
54.
Zurück zum Zitat Ho S, et al. Clinical evaluation of the partition model for estimating radiation doses from yttrium-90 microspheres in the treatment of hepatic cancer. Eur J Nucl Med. 1997;24(3):293–8.PubMed Ho S, et al. Clinical evaluation of the partition model for estimating radiation doses from yttrium-90 microspheres in the treatment of hepatic cancer. Eur J Nucl Med. 1997;24(3):293–8.PubMed
55.
Zurück zum Zitat Salem R, et al. Incidence of radiation pneumonitis after hepatic intra-arterial radiotherapy with yttrium-90 microspheres assuming uniform lung distribution. Am J Clin Oncol. 2008;31(5):431–8.PubMedCrossRef Salem R, et al. Incidence of radiation pneumonitis after hepatic intra-arterial radiotherapy with yttrium-90 microspheres assuming uniform lung distribution. Am J Clin Oncol. 2008;31(5):431–8.PubMedCrossRef
56.
Zurück zum Zitat Jadoul A, et al. Comparative dosimetry between (99m)Tc-MAA SPECT/CT and (90)Y PET/CT in primary and metastatic liver tumors. Eur J Nucl Med Mol Imaging. 2020;47(4):828–37.PubMedCrossRef Jadoul A, et al. Comparative dosimetry between (99m)Tc-MAA SPECT/CT and (90)Y PET/CT in primary and metastatic liver tumors. Eur J Nucl Med Mol Imaging. 2020;47(4):828–37.PubMedCrossRef
57.
Zurück zum Zitat Gnesin S, et al. Partition model based Tc-99m-MAA SPECT/CT predictive dosimetry compared with Y-90 TOF PET/CT posttreatment dosimetry in radioembolization of hepatocellular carcinoma: a quantitative agreement comparison. J Nucl Med. 2016;57(11):1672–8.PubMedCrossRef Gnesin S, et al. Partition model based Tc-99m-MAA SPECT/CT predictive dosimetry compared with Y-90 TOF PET/CT posttreatment dosimetry in radioembolization of hepatocellular carcinoma: a quantitative agreement comparison. J Nucl Med. 2016;57(11):1672–8.PubMedCrossRef
58.
Zurück zum Zitat Ilhan H, et al. Predictive value of 99mTc-MAA SPECT for 90Y-labeled resin microsphere distribution in radioembolization of primary and secondary hepatic tumors. J Nucl Med. 2015;56(11):1654–60.PubMedCrossRef Ilhan H, et al. Predictive value of 99mTc-MAA SPECT for 90Y-labeled resin microsphere distribution in radioembolization of primary and secondary hepatic tumors. J Nucl Med. 2015;56(11):1654–60.PubMedCrossRef
59.
Zurück zum Zitat Wondergem M, et al. 99mTc-macroaggregated albumin poorly predicts the intrahepatic distribution of 90Y resin microspheres in hepatic radioembolization. J Nucl Med. 2013;54(8):1294–301.PubMedCrossRef Wondergem M, et al. 99mTc-macroaggregated albumin poorly predicts the intrahepatic distribution of 90Y resin microspheres in hepatic radioembolization. J Nucl Med. 2013;54(8):1294–301.PubMedCrossRef
60.
Zurück zum Zitat Haste P, et al. Correlation of technetium-99m macroaggregated albumin and yttrium-90 glass microsphere biodistribution in hepatocellular carcinoma: a retrospective review of pretreatment single photon emission ct and posttreatment positron emission tomography/CT. J Vasc Interv Radiol. 2017;28(5):722–30.PubMedCrossRef Haste P, et al. Correlation of technetium-99m macroaggregated albumin and yttrium-90 glass microsphere biodistribution in hepatocellular carcinoma: a retrospective review of pretreatment single photon emission ct and posttreatment positron emission tomography/CT. J Vasc Interv Radiol. 2017;28(5):722–30.PubMedCrossRef
61.
Zurück zum Zitat Garin E, 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(7):1057–68.PubMedPubMedCentralCrossRef Garin E, 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(7):1057–68.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Garin E, et al. Dosimetry based on 99mTc-macroaggregated albumin SPECT/CT accurately predicts tumor response and survival in hepatocellular carcinoma patients treated with 90Y-loaded glass microspheres: preliminary results. J Nucl Med. 2012;53(2):255–63.PubMedCrossRef Garin E, et al. Dosimetry based on 99mTc-macroaggregated albumin SPECT/CT accurately predicts tumor response and survival in hepatocellular carcinoma patients treated with 90Y-loaded glass microspheres: preliminary results. J Nucl Med. 2012;53(2):255–63.PubMedCrossRef
63.
Zurück zum Zitat Chiesa C, et al. Need, feasibility and convenience of dosimetric treatment planning in liver selective internal radiation therapy with (90)Y microspheres: the experience of the National Tumor Institute of Milan. Q J Nucl Med Mol Imaging. 2011;55(2):168–97.PubMed Chiesa C, et al. Need, feasibility and convenience of dosimetric treatment planning in liver selective internal radiation therapy with (90)Y microspheres: the experience of the National Tumor Institute of Milan. Q J Nucl Med Mol Imaging. 2011;55(2):168–97.PubMed
64.
Zurück zum Zitat Ho CL, et al. Radioembolization with (90)Y glass microspheres for hepatocellular carcinoma: significance of pretreatment (11)C-acetate and (18)F-FDG PET/CT and posttreatment (90)Y PET/CT in individualized dose prescription. Eur J Nucl Med Mol Imaging. 2018;45(12):2110–21.PubMedCrossRef Ho CL, et al. Radioembolization with (90)Y glass microspheres for hepatocellular carcinoma: significance of pretreatment (11)C-acetate and (18)F-FDG PET/CT and posttreatment (90)Y PET/CT in individualized dose prescription. Eur J Nucl Med Mol Imaging. 2018;45(12):2110–21.PubMedCrossRef
65.
Zurück zum Zitat Hermann AL, et al. Relationship of tumor radiation-absorbed dose to survival and response in hepatocellular carcinoma treated with transarterial radioembolization with (90)Y in the SARAH study. Radiology. 2020;296(3):673–84.PubMedCrossRef Hermann AL, et al. Relationship of tumor radiation-absorbed dose to survival and response in hepatocellular carcinoma treated with transarterial radioembolization with (90)Y in the SARAH study. Radiology. 2020;296(3):673–84.PubMedCrossRef
66.
Zurück zum Zitat Lau WY, et al. Treatment of inoperable hepatocellular carcinoma with intrahepatic arterial yttrium-90 microspheres: a phase I and II study. Br J Cancer. 1994;70(5):994–9.PubMedPubMedCentralCrossRef Lau WY, et al. Treatment of inoperable hepatocellular carcinoma with intrahepatic arterial yttrium-90 microspheres: a phase I and II study. Br J Cancer. 1994;70(5):994–9.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Garin, E., et al., Personalised versus standard dosimetry approach of selective internal radiation therapy in patients with locally advanced hepatocellular carcinoma (DOSISPHERE-01): a randomised, multicentre, open-label phase 2 trial. Lancet Gastroenterol Hepatol, 2020 Garin, E., et al., Personalised versus standard dosimetry approach of selective internal radiation therapy in patients with locally advanced hepatocellular carcinoma (DOSISPHERE-01): a randomised, multicentre, open-label phase 2 trial. Lancet Gastroenterol Hepatol, 2020
68.
Zurück zum Zitat Smits MLJ, et al. The superior predictive value of (166)Ho-scout compared with (99m)Tc-macroaggregated albumin prior to (166)Ho-microspheres radioembolization in patients with liver metastases. Eur J Nucl Med Mol Imaging. 2020;47(4):798–806.PubMedCrossRef Smits MLJ, et al. The superior predictive value of (166)Ho-scout compared with (99m)Tc-macroaggregated albumin prior to (166)Ho-microspheres radioembolization in patients with liver metastases. Eur J Nucl Med Mol Imaging. 2020;47(4):798–806.PubMedCrossRef
69.
Zurück zum Zitat Salem R, et al. Clinical and dosimetric considerations for Y90: recommendations from an international multidisciplinary working group. Eur J Nucl Med Mol Imaging. 2019;46(8):1695–704.PubMedCrossRef Salem R, et al. Clinical and dosimetric considerations for Y90: recommendations from an international multidisciplinary working group. Eur J Nucl Med Mol Imaging. 2019;46(8):1695–704.PubMedCrossRef
70.
Zurück zum Zitat Lewandowski RJ, et al. Radiation segmentectomy: potential curative therapy for early hepatocellular carcinoma. Radiology. 2018;287(3):1050–8.PubMedCrossRef Lewandowski RJ, et al. Radiation segmentectomy: potential curative therapy for early hepatocellular carcinoma. Radiology. 2018;287(3):1050–8.PubMedCrossRef
71.
Zurück zum Zitat Meiers C, et al. Safety and initial efficacy of radiation segmentectomy for the treatment of hepatic metastases. J Gastrointest Oncol. 2018;9(2):311–5.PubMedPubMedCentralCrossRef Meiers C, et al. Safety and initial efficacy of radiation segmentectomy for the treatment of hepatic metastases. J Gastrointest Oncol. 2018;9(2):311–5.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Padia SA, et al. Superselective yttrium-90 radioembolization for hepatocellular carcinoma yields high response rates with minimal toxicity. J Vasc Interv Radiol. 2014;25(7):1067–73.PubMedCrossRef Padia SA, et al. Superselective yttrium-90 radioembolization for hepatocellular carcinoma yields high response rates with minimal toxicity. J Vasc Interv Radiol. 2014;25(7):1067–73.PubMedCrossRef
73.
Zurück zum Zitat Biederman DM, et al. Radiation segmentectomy versus selective chemoembolization in the treatment of early-stage hepatocellular carcinoma. J Vasc Interv Radiol. 2018;29(1):30–7 e2.PubMedCrossRef Biederman DM, et al. Radiation segmentectomy versus selective chemoembolization in the treatment of early-stage hepatocellular carcinoma. J Vasc Interv Radiol. 2018;29(1):30–7 e2.PubMedCrossRef
74.
Zurück zum Zitat Garin E, et al. Personalized dosimetry with intensification using 90Y-loaded glass microsphere radioembolization induces prolonged overall survival in hepatocellular carcinoma patients with portal vein thrombosis. J Nucl Med. 2015;56(3):339–46.PubMedCrossRef Garin E, et al. Personalized dosimetry with intensification using 90Y-loaded glass microsphere radioembolization induces prolonged overall survival in hepatocellular carcinoma patients with portal vein thrombosis. J Nucl Med. 2015;56(3):339–46.PubMedCrossRef
75.
Zurück zum Zitat Konijnenberg M., et al. EANM position paper on article 56 of the Council Directive 2013/59/Euratom (basic safety standards) for nuclear medicine therapy. Eur J Nucl Med Mol Imaging. 2021:48(1);67–72 Konijnenberg M., et al. EANM position paper on article 56 of the Council Directive 2013/59/Euratom (basic safety standards) for nuclear medicine therapy. Eur J Nucl Med Mol Imaging. 2021:48(1);67–72
76.
Zurück zum Zitat Ho S, et al. Partition model for estimating radiation doses from yttrium-90 microspheres in treating hepatic tumours. Eur J Nucl Med. 1996;23(8):947–52 Ho S, et al. Partition model for estimating radiation doses from yttrium-90 microspheres in treating hepatic tumours. Eur J Nucl Med. 1996;23(8):947–52
77.
Zurück zum Zitat Chiesa C, et al. Radioembolization of hepatocarcinoma with (90)Y glass microspheres: development of an individualized treatment planning strategy based on dosimetry and radiobiology. Eur J Nucl Med Mol Imaging. 2015;42(11):1718–38.PubMedCrossRef Chiesa C, et al. Radioembolization of hepatocarcinoma with (90)Y glass microspheres: development of an individualized treatment planning strategy based on dosimetry and radiobiology. Eur J Nucl Med Mol Imaging. 2015;42(11):1718–38.PubMedCrossRef
78.
Zurück zum Zitat Chiesa C, Bardies M, Zaidi H. Voxel-based dosimetry is superior to mean absorbed dose approach for establishing dose-effect relationship in targeted radionuclide therapy. Med Phys. 2019;46(12):5403–6.PubMedCrossRef Chiesa C, Bardies M, Zaidi H. Voxel-based dosimetry is superior to mean absorbed dose approach for establishing dose-effect relationship in targeted radionuclide therapy. Med Phys. 2019;46(12):5403–6.PubMedCrossRef
79.
Zurück zum Zitat Dewaraja YK, et al. Prediction of tumor control in (90)Y radioembolization by logit models with PET/CT-based dose metrics. J Nucl Med. 2020;61(1):104–11.PubMedPubMedCentralCrossRef Dewaraja YK, et al. Prediction of tumor control in (90)Y radioembolization by logit models with PET/CT-based dose metrics. J Nucl Med. 2020;61(1):104–11.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Kappadath SC, et al. Hepatocellular carcinoma tumor dose response after (90)Y-radioembolization with glass microspheres using (90)Y-SPECT/CT-based voxel dosimetry. Int J Radiat Oncol Biol Phys. 2018;102(2):451–61.PubMedCrossRef Kappadath SC, et al. Hepatocellular carcinoma tumor dose response after (90)Y-radioembolization with glass microspheres using (90)Y-SPECT/CT-based voxel dosimetry. Int J Radiat Oncol Biol Phys. 2018;102(2):451–61.PubMedCrossRef
81.
Zurück zum Zitat Chiesa, C., et al. EANM dosimetry committee series on standard operational procedures: a unified methodology for 99m Tc-MAA pre- and 90 Y peri-therapy dosimetry in liver radioembolization with 90 Y microspheres. EJNMMI Phys. 2021:8(1);77 Chiesa, C., et al. EANM dosimetry committee series on standard operational procedures: a unified methodology for 99m Tc-MAA pre- and 90 Y peri-therapy dosimetry in liver radioembolization with 90 Y microspheres. EJNMMI Phys. 2021:8(1);77
82.
Zurück zum Zitat Roosen, J., et al., To 1000 Gy and back again: a systematic review on dose-response evaluation in selective internal radiation therapy for primary and secondary liver cancer. Eur J Nucl Med Mol Imaging, 2021. Roosen, J., et al., To 1000 Gy and back again: a systematic review on dose-response evaluation in selective internal radiation therapy for primary and secondary liver cancer. Eur J Nucl Med Mol Imaging, 2021.
83.
Zurück zum Zitat Salem R, et al. Yttrium-90 radioembolization for the treatment of solitary, unresectable hepatocellular carcinoma: the LEGACY study. Hepatology. 2021. Salem R, et al. Yttrium-90 radioembolization for the treatment of solitary, unresectable hepatocellular carcinoma: the LEGACY study. Hepatology. 2021.
84.
Zurück zum Zitat Lewandowski RJ, et al. (90) Y radiation lobectomy: outcomes following surgical resection in patients with hepatic tumors and small future liver remnant volumes. J Surg Oncol. 2016;114(1):99–105.PubMedCrossRef Lewandowski RJ, et al. (90) Y radiation lobectomy: outcomes following surgical resection in patients with hepatic tumors and small future liver remnant volumes. J Surg Oncol. 2016;114(1):99–105.PubMedCrossRef
85.
Zurück zum Zitat Palard X, et al. Dosimetric parameters predicting contralateral liver hypertrophy after unilobar radioembolization of hepatocellular carcinoma. Eur J Nucl Med Mol Imaging. 2018;45(3):392–401.PubMedCrossRef Palard X, et al. Dosimetric parameters predicting contralateral liver hypertrophy after unilobar radioembolization of hepatocellular carcinoma. Eur J Nucl Med Mol Imaging. 2018;45(3):392–401.PubMedCrossRef
86.
Zurück zum Zitat Chiesa C, et al. Radioembolization of hepatocarcinoma with (90)Y glass microspheres: treatment optimization using the dose-toxicity relationship. Eur J Nucl Med Mol Imaging. 2020;47(13):3018–32.PubMedCrossRef Chiesa C, et al. Radioembolization of hepatocarcinoma with (90)Y glass microspheres: treatment optimization using the dose-toxicity relationship. Eur J Nucl Med Mol Imaging. 2020;47(13):3018–32.PubMedCrossRef
87.
Zurück zum Zitat Shepherd FA, et al. A phase I dose escalation trial of yttrium-90 microspheres in the treatment of primary hepatocellular carcinoma. Cancer. 1992;70(9):2250–4.PubMedCrossRef Shepherd FA, et al. A phase I dose escalation trial of yttrium-90 microspheres in the treatment of primary hepatocellular carcinoma. Cancer. 1992;70(9):2250–4.PubMedCrossRef
88.
Zurück zum Zitat Bourien H, et al. Yttrium-90 glass microspheres radioembolization (RE) for biliary tract cancer: a large single-center experience. Eur J Nucl Med Mol Imaging. 2019;46(3):669–76.PubMedCrossRef Bourien H, et al. Yttrium-90 glass microspheres radioembolization (RE) for biliary tract cancer: a large single-center experience. Eur J Nucl Med Mol Imaging. 2019;46(3):669–76.PubMedCrossRef
89.
Zurück zum Zitat Mouli S, et al. Yttrium-90 radioembolization for intrahepatic cholangiocarcinoma: safety, response, and survival analysis. J Vasc Interv Radiol. 2013;24(8):1227–34.PubMedPubMedCentralCrossRef Mouli S, et al. Yttrium-90 radioembolization for intrahepatic cholangiocarcinoma: safety, response, and survival analysis. J Vasc Interv Radiol. 2013;24(8):1227–34.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Padia SA, et al. Yttrium-90 radiation segmentectomy for hepatic metastases: a multi-institutional study of safety and efficacy. J Surg Oncol. 2021;123(1):172–8.PubMedCrossRef Padia SA, et al. Yttrium-90 radiation segmentectomy for hepatic metastases: a multi-institutional study of safety and efficacy. J Surg Oncol. 2021;123(1):172–8.PubMedCrossRef
91.
Zurück zum Zitat Alsultan, A.A., et al., Dose-response and dose-toxicity relationships for yttrium-90 glass radioembolization in patients with colorectal cancer liver metastases. J Nucl Med, 2021. Alsultan, A.A., et al., Dose-response and dose-toxicity relationships for yttrium-90 glass radioembolization in patients with colorectal cancer liver metastases. J Nucl Med, 2021.
92.
Zurück zum Zitat Lewandowski RJ, et al. Twelve-year experience of radioembolization for colorectal hepatic metastases in 214 patients: survival by era and chemotherapy. Eur J Nucl Med Mol Imaging. 2014;41(10):1861–9.PubMedCrossRef Lewandowski RJ, et al. Twelve-year experience of radioembolization for colorectal hepatic metastases in 214 patients: survival by era and chemotherapy. Eur J Nucl Med Mol Imaging. 2014;41(10):1861–9.PubMedCrossRef
93.
Zurück zum Zitat Levillain, H., et al., International recommendations for personalised selective internal radiation therapy of primary and metastatic liver diseases with yttrium-90 resin microspheres. Eur J Nucl Med Mol Imaging, 2021 Levillain, H., et al., International recommendations for personalised selective internal radiation therapy of primary and metastatic liver diseases with yttrium-90 resin microspheres. Eur J Nucl Med Mol Imaging, 2021
94.
Zurück zum Zitat Levillain H, et al. Personalised radioembolization improves outcomes in refractory intra-hepatic cholangiocarcinoma: a multicenter study. Eur J Nucl Med Mol Imaging. 2019;46(11):2270–9.PubMedCrossRef Levillain H, et al. Personalised radioembolization improves outcomes in refractory intra-hepatic cholangiocarcinoma: a multicenter study. Eur J Nucl Med Mol Imaging. 2019;46(11):2270–9.PubMedCrossRef
95.
Zurück zum Zitat van den Hoven AF, et al. Insights into the dose-response relationship of radioembolization with resin 90y-microspheres: a prospective cohort study in patients with colorectal cancer liver metastases. J Nucl Med. 2016;57(7):1014–9.PubMedCrossRef van den Hoven AF, et al. Insights into the dose-response relationship of radioembolization with resin 90y-microspheres: a prospective cohort study in patients with colorectal cancer liver metastases. J Nucl Med. 2016;57(7):1014–9.PubMedCrossRef
96.
Zurück zum Zitat Bastiaannet R, et al. First evidence for a dose-response relationship in patients treated with (166)Ho radioembolization: a prospective study. J Nucl Med. 2020;61(4):608–12.PubMedCrossRef Bastiaannet R, et al. First evidence for a dose-response relationship in patients treated with (166)Ho radioembolization: a prospective study. J Nucl Med. 2020;61(4):608–12.PubMedCrossRef
97.
Zurück zum Zitat van Roekel C, et al. Dose-effect relationships of (166)Ho radioembolization in colorectal cancer. J Nucl Med. 2021;62(2):272–9.PubMedCrossRef van Roekel C, et al. Dose-effect relationships of (166)Ho radioembolization in colorectal cancer. J Nucl Med. 2021;62(2):272–9.PubMedCrossRef
98.
Zurück zum Zitat Vauthey JN, et al. Body surface area and body weight predict total liver volume in Western adults. Liver Transpl. 2002;8(3):233–40.PubMedCrossRef Vauthey JN, et al. Body surface area and body weight predict total liver volume in Western adults. Liver Transpl. 2002;8(3):233–40.PubMedCrossRef
99.
Zurück zum Zitat Ahmadzadehfar H, et al. Evaluation of the delivered activity of yttrium-90 resin microspheres using sterile water and 5% glucose during administration. EJNMMI Res. 2015;5(1):54.PubMedPubMedCentralCrossRef Ahmadzadehfar H, et al. Evaluation of the delivered activity of yttrium-90 resin microspheres using sterile water and 5% glucose during administration. EJNMMI Res. 2015;5(1):54.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Giammarile F, et al. EANM procedure guideline for the treatment of liver cancer and liver metastases with intra-arterial radioactive compounds. Eur J Nucl Med Mol Imaging. 2011;38(7):1393–406.PubMedCrossRef Giammarile F, et al. EANM procedure guideline for the treatment of liver cancer and liver metastases with intra-arterial radioactive compounds. Eur J Nucl Med Mol Imaging. 2011;38(7):1393–406.PubMedCrossRef
101.
Zurück zum Zitat Lhommel R, et al. Feasibility of 90Y TOF PET-based dosimetry in liver metastasis therapy using SIR-spheres. Eur J Nucl Med Mol Imaging. 2010;37(9):1654–62.PubMedCrossRef Lhommel R, et al. Feasibility of 90Y TOF PET-based dosimetry in liver metastasis therapy using SIR-spheres. Eur J Nucl Med Mol Imaging. 2010;37(9):1654–62.PubMedCrossRef
102.
Zurück zum Zitat Selwyn RG, et al. A new internal pair production branching ratio of 90Y: the development of a non-destructive assay for 90Y and 90Sr. Appl Radiat Isot. 2007;65(3):318–27.PubMedCrossRef Selwyn RG, et al. A new internal pair production branching ratio of 90Y: the development of a non-destructive assay for 90Y and 90Sr. Appl Radiat Isot. 2007;65(3):318–27.PubMedCrossRef
103.
Zurück zum Zitat Kesim S, et al. Unexpected radiation pneumonitis after SIRT with significant decrease in DLCO with internal radiation exposure: a case report. BMC Med Imaging. 2020;20(1):52.PubMedPubMedCentralCrossRef Kesim S, et al. Unexpected radiation pneumonitis after SIRT with significant decrease in DLCO with internal radiation exposure: a case report. BMC Med Imaging. 2020;20(1):52.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Auditore L, et al. Monte Carlo (90)Y PET/CT dosimetry of unexpected focal radiation-induced lung damage after hepatic radioembolisation. Phys Med Biol. 2020;65(23):235014.PubMedCrossRef Auditore L, et al. Monte Carlo (90)Y PET/CT dosimetry of unexpected focal radiation-induced lung damage after hepatic radioembolisation. Phys Med Biol. 2020;65(23):235014.PubMedCrossRef
105.
Zurück zum Zitat Scott NP, McGowan DR. Optimising quantitative (90)Y PET imaging: an investigation into the effects of scan length and Bayesian penalised likelihood reconstruction. EJNMMI Res. 2019;9(1):40.PubMedPubMedCentralCrossRef Scott NP, McGowan DR. Optimising quantitative (90)Y PET imaging: an investigation into the effects of scan length and Bayesian penalised likelihood reconstruction. EJNMMI Res. 2019;9(1):40.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Elschot M, et al. Quantitative evaluation of scintillation camera imaging characteristics of isotopes used in liver radioembolization. PLoS One. 2011;6(11):e26174.PubMedPubMedCentralCrossRef Elschot M, et al. Quantitative evaluation of scintillation camera imaging characteristics of isotopes used in liver radioembolization. PLoS One. 2011;6(11):e26174.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat van de Maat GH, et al. MRI-based biodistribution assessment of holmium-166 poly(L-lactic acid) microspheres after radioembolisation. Eur Radiol. 2013;23(3):827–35.PubMedCrossRef van de Maat GH, et al. MRI-based biodistribution assessment of holmium-166 poly(L-lactic acid) microspheres after radioembolisation. Eur Radiol. 2013;23(3):827–35.PubMedCrossRef
108.
Zurück zum Zitat John, L., Discharge of patients undergoing treatment with radioactive substances, A.R.P.a.N.S. Agency, Editor. 2002: Australia John, L., Discharge of patients undergoing treatment with radioactive substances, A.R.P.a.N.S. Agency, Editor. 2002: Australia
109.
Zurück zum Zitat Prince JF, et al. Radiation emission from patients treated with holmium-166 radioembolization. J Vasc Interv Radiol. 2014;25(12):1956–1963e1.PubMedCrossRef Prince JF, et al. Radiation emission from patients treated with holmium-166 radioembolization. J Vasc Interv Radiol. 2014;25(12):1956–1963e1.PubMedCrossRef
110.
Zurück zum Zitat Laffont S, et al. Occupational radiation exposure of medical staff performing (9)(0)Y-loaded microsphere radioembolization. Eur J Nucl Med Mol Imaging. 2016;43(5):824–31.PubMedCrossRef Laffont S, et al. Occupational radiation exposure of medical staff performing (9)(0)Y-loaded microsphere radioembolization. Eur J Nucl Med Mol Imaging. 2016;43(5):824–31.PubMedCrossRef
111.
Zurück zum Zitat Taleb J, et al. Radiation dose measurements for staff members involved in holmium-166 preclinical trial. Radiat Meas. 2013;58:75–8.CrossRef Taleb J, et al. Radiation dose measurements for staff members involved in holmium-166 preclinical trial. Radiat Meas. 2013;58:75–8.CrossRef
112.
Zurück zum Zitat Gulec SA, Mesoloras G, Stabin M. Dosimetric techniques in 90Y-microsphere therapy of liver cancer: the MIRD equations for dose calculations. J Nucl Med. 2006;47(7):1209–11.PubMed Gulec SA, Mesoloras G, Stabin M. Dosimetric techniques in 90Y-microsphere therapy of liver cancer: the MIRD equations for dose calculations. J Nucl Med. 2006;47(7):1209–11.PubMed
113.
Zurück zum Zitat Metyko J, et al. Long-lived impurities of 90Y-labeled microspheres, TheraSphere and SIR-spheres, and the impact on patient dose and waste management. Health Phys. 2012;103(5 Suppl 3):S204–8.PubMedCrossRef Metyko J, et al. Long-lived impurities of 90Y-labeled microspheres, TheraSphere and SIR-spheres, and the impact on patient dose and waste management. Health Phys. 2012;103(5 Suppl 3):S204–8.PubMedCrossRef
114.
Zurück zum Zitat Elsayed M, et al. Incidence of radioembolization-induced liver disease and liver toxicity following repeat 90Y-radioembolization: outcomes at a large tertiary care center. Clin Nucl Med. 2020;45(2):100–4.PubMedCrossRef Elsayed M, et al. Incidence of radioembolization-induced liver disease and liver toxicity following repeat 90Y-radioembolization: outcomes at a large tertiary care center. Clin Nucl Med. 2020;45(2):100–4.PubMedCrossRef
115.
Zurück zum Zitat Lam MG, et al. Safety of repeated yttrium-90 radioembolization. Cardiovasc Intervent Radiol. 2013;36(5):1320–8.PubMedCrossRef Lam MG, et al. Safety of repeated yttrium-90 radioembolization. Cardiovasc Intervent Radiol. 2013;36(5):1320–8.PubMedCrossRef
117.
Zurück zum Zitat Zarva A, et al. Safety of repeated radioembolizations in patients with advanced primary and secondary liver tumors and progressive disease after first selective internal radiotherapy. J Nucl Med. 2014;55(3):360–6.PubMedCrossRef Zarva A, et al. Safety of repeated radioembolizations in patients with advanced primary and secondary liver tumors and progressive disease after first selective internal radiotherapy. J Nucl Med. 2014;55(3):360–6.PubMedCrossRef
118.
Zurück zum Zitat Prajapati HJ, et al. mRECIST and EASL responses at early time point by contrast-enhanced dynamic MRI predict survival in patients with unresectable hepatocellular carcinoma (HCC) treated by doxorubicin drug-eluting beads transarterial chemoembolization (DEB TACE). Ann Oncol. 2013;24(4):965–73.PubMedCrossRef Prajapati HJ, et al. mRECIST and EASL responses at early time point by contrast-enhanced dynamic MRI predict survival in patients with unresectable hepatocellular carcinoma (HCC) treated by doxorubicin drug-eluting beads transarterial chemoembolization (DEB TACE). Ann Oncol. 2013;24(4):965–73.PubMedCrossRef
119.
Zurück zum Zitat Lencioni R, Llovet JM. Modified RECIST (mRECIST) assessment for hepatocellular carcinoma. Semin Liver Dis. 2010;30(1):52–60.PubMedCrossRef Lencioni R, Llovet JM. Modified RECIST (mRECIST) assessment for hepatocellular carcinoma. Semin Liver Dis. 2010;30(1):52–60.PubMedCrossRef
120.
Zurück zum Zitat Shim JH, et al. Which response criteria best help predict survival of patients with hepatocellular carcinoma following chemoembolization? A validation study of old and new models. Radiology. 2012;262(2):708–18.PubMedCrossRef Shim JH, et al. Which response criteria best help predict survival of patients with hepatocellular carcinoma following chemoembolization? A validation study of old and new models. Radiology. 2012;262(2):708–18.PubMedCrossRef
121.
Zurück zum Zitat Gillmore R, et al. EASL and mRECIST responses are independent prognostic factors for survival in hepatocellular cancer patients treated with transarterial embolization. J Hepatol. 2011;55(6):1309–16.PubMedCrossRef Gillmore R, et al. EASL and mRECIST responses are independent prognostic factors for survival in hepatocellular cancer patients treated with transarterial embolization. J Hepatol. 2011;55(6):1309–16.PubMedCrossRef
122.
Zurück zum Zitat European Association for the Study of the Liver. Electronic address, e.e.e. and L. European Association for the Study of the, EASL clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol, 2018. 69(1): p. 182–236 European Association for the Study of the Liver. Electronic address, e.e.e. and L. European Association for the Study of the, EASL clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol, 2018. 69(1): p. 182–236
123.
Zurück zum Zitat Pollock RF, et al. Association between objective response rate and overall survival in metastatic neuroendocrine tumors treated with radioembolization: a systematic literature review and regression analysis. Expert Rev Anticancer Ther. 2020;20(11):997–1009.PubMedCrossRef Pollock RF, et al. Association between objective response rate and overall survival in metastatic neuroendocrine tumors treated with radioembolization: a systematic literature review and regression analysis. Expert Rev Anticancer Ther. 2020;20(11):997–1009.PubMedCrossRef
124.
Zurück zum Zitat Braat A, et al. Radioembolization with (90)Y resin microspheres of neuroendocrine liver metastases: international multicenter study on efficacy and toxicity. Cardiovasc Intervent Radiol. 2019;42(3):413–25.PubMedCrossRef Braat A, et al. Radioembolization with (90)Y resin microspheres of neuroendocrine liver metastases: international multicenter study on efficacy and toxicity. Cardiovasc Intervent Radiol. 2019;42(3):413–25.PubMedCrossRef
125.
Zurück zum Zitat Memon K, et al. Radioembolization for neuroendocrine liver metastases: safety, imaging, and long-term outcomes. Int J Radiat Oncol Biol Phys. 2012;83(3):887–94.PubMedCrossRef Memon K, et al. Radioembolization for neuroendocrine liver metastases: safety, imaging, and long-term outcomes. Int J Radiat Oncol Biol Phys. 2012;83(3):887–94.PubMedCrossRef
126.
Zurück zum Zitat Beuzit L, et al. Comparison of Choi criteria and response evaluation criteria in solid tumors (RECIST) for intrahepatic cholangiocarcinoma treated with glass-microspheres yttrium-90 selective internal radiation therapy (SIRT). Eur J Radiol. 2016;85(8):1445–52.PubMedCrossRef Beuzit L, et al. Comparison of Choi criteria and response evaluation criteria in solid tumors (RECIST) for intrahepatic cholangiocarcinoma treated with glass-microspheres yttrium-90 selective internal radiation therapy (SIRT). Eur J Radiol. 2016;85(8):1445–52.PubMedCrossRef
127.
Zurück zum Zitat Camacho JC, et al. Modified response evaluation criteria in solid tumors and European Association for the Study of the liver criteria using delayed-phase imaging at an early time point predict survival in patients with unresectable intrahepatic cholangiocarcinoma following yttrium-90 radioembolization. J Vasc Interv Radiol. 2014;25(2):256–65.PubMedCrossRef Camacho JC, et al. Modified response evaluation criteria in solid tumors and European Association for the Study of the liver criteria using delayed-phase imaging at an early time point predict survival in patients with unresectable intrahepatic cholangiocarcinoma following yttrium-90 radioembolization. J Vasc Interv Radiol. 2014;25(2):256–65.PubMedCrossRef
128.
Zurück zum Zitat Akinwande O, et al. Comparison of tumor response assessment methods in patients with metastatic colorectal cancer after locoregional therapy. J Surg Oncol. 2016;113(4):443–8.PubMedCrossRef Akinwande O, et al. Comparison of tumor response assessment methods in patients with metastatic colorectal cancer after locoregional therapy. J Surg Oncol. 2016;113(4):443–8.PubMedCrossRef
Metadaten
Titel
EANM procedure guideline for the treatment of liver cancer and liver metastases with intra-arterial radioactive compounds
verfasst von
M. Weber
M. Lam
C. Chiesa
M. Konijnenberg
M. Cremonesi
P. Flamen
S. Gnesin
L. Bodei
T. Kracmerova
M. Luster
E. Garin
K. Herrmann
Publikationsdatum
11.02.2022
Verlag
Springer Berlin Heidelberg
Erschienen in
European Journal of Nuclear Medicine and Molecular Imaging / Ausgabe 5/2022
Print ISSN: 1619-7070
Elektronische ISSN: 1619-7089
DOI
https://doi.org/10.1007/s00259-021-05600-z

Weitere Artikel der Ausgabe 5/2022

European Journal of Nuclear Medicine and Molecular Imaging 5/2022 Zur Ausgabe