Tumor > Therapy > Microspheres

Microsphere brachytherapy- Radioembolization

Liver malignancy:

Transarterial radiation therapy with 90Y-microspheres is another potential treatment method for hepatoma patients with low hepatic reserve [1] and in patients with metastatic disease to the liver [5]. The dual blood supply to the liver provides a natural selectivity for tumor therapy [3]. Tumors generally derive their blood supply from the hepatic artery, while the normal liver parenchyma is perfused via the portal venous system [3]. When injected via a branch of the hepatic arterial system, the microspheres preferentially lodge in the periphery around the tumor [3]. The preferential deposition of the microspheres in the tumor, maximizes tumor irradiation while sparing adjacent liver parenchyma [5]. After implantation, the 90Y-microspheres remain permanently in place [3].

For patients with hepatocellular carcinoma, current data suggest that radioembolization is best placed after the failure of TACE in the early intermediate-stage HCC or in patients with diffuse disease (> 4 tumors) or large tumors (>5cm) [26].

Yttrium-90 can be produced via nuclear reactor production or from a 90Sr/90Y generator [4]. Yttrium-90 (half-life 64.1 hours [2.7 days]) decays to stable Zirconium 90 (90Zr) via a 2.28 MeV maximal energy beta-particle and an accompanying antineutrino 99.98% of the time (it is essentially a pure beta emitter) [3,13,18]. The mean energy is 0.94 MeV which corresponds to a maximum range of 11 mm in tissue, a mean path of 2.5 mm, and a X90 (radius of a sphere in which 90% of the energy is deposited) of 5.3 mm (corresponding to 50-200 cell diameters) [3,6,9,18]. When the beta particles interact with liver parenchyma, bremsstrahlung radiation is emitted and can be used for limited indirect imaging [18]. An alternative minor decay route (32 disintegrations per million) is by internal pair production in which 90Y decays to an excited state of  90Zr which then emits a positron (maximum energy 800 keV) [18]. The interaction of the emitted positron produces two 511 keV photons that can be imaged using a PET scanner [18].  90Y microspheres can deliver an intratumoral dose of 100-150 Gy [6]. As a rule of thumb, 1 Gbq (27 mCi) of uniformly dispersed 90Y delivers an absorbed dose of about 50 Gy (5000 rads) [3,18]. 

Two types of microspheres are available for clinical use- resin and glass microspheres [4]. Resin microspheres (SIR-spheres) are non-degradable polymer beads between 20-60 ?m in diameter and are loaded with 90Y at a specific activity of 40-70 Bq (50 Bq [41]) per sphere (the radioisotope is bound to the surface of the resin microsphere [34]) [4,13,18]. Glass microspheres (TheraSpheres) measure between 20-30 ?m in diameter and are loaded with  90Y at a specific activity of 2400-2700 Bq (2500 Bq [41]) per sphere (the radioisotope is incorporated into the glass matrix [34]) [4]. Resin microspheres have a lower specific activity than glass microspheres (the specific activity of the resin microspheres is 50 times lower than that of TheraSpheres- approximately 50 Bq/microsphere versus 2,500 Bq/microsphere [18]) [13,34]. But resin microspheres also have a lower specific gravity and a higher number of particles per treatment and therefore a greater embolic effect for any given desired activity (i.e.- more particles can be delivered which can result in more uniform particle distribution in the tumor- and this may be important for larger or hypervascular tumors [42]) [13,34,42]. Glass microspheres remain fixed in the liver and are not found in any body fluid, whereas trace amounts of 90Y activity may be excreted in the urine for the first 24 hours following treatment with resin microspheres [4]. It has been suggested that because of glass microspheres low embolic load, they are less likely to limit the prescribed activity of 90Y, while the heavy embolic load of resin microspheres can result in arterial stasis, limiting the actual  90Y dose (i.e.- inability to deliver the entire prescribed dose) [36]. Stasis can also result in reflux of particles into off target arteries [42]. It has been reported that early stasis can be seen in approximately 20% of patients that are treated with resin microspheres [42]. Early stasis can be seen even more frequently (up to 38%) in patients who have received multiple prior lines of chemotherapy, including hepatic arterial infusion pump chemotherapy [42]. The likelihood for early stasis can be reduced by delivering the resin microspheres using 5% dextrose or 50% contrast material and 50% saline (note this method is not recommended by the manufacturer, but does also permit fluoroscopic monitoring of the flow rate during delivery), instead of sterile water [42]. Glass microspheres are delivered in saline which precludes angiographic monitoring during infusion [42].

166Ho-microspheres have been approved for clinical use in the European Union [48]. 166Ho (Holmium) has a half-life of 26.8 hours and a beta emission (max 1.85 MeV) [50]. The agent also emits an 81 keV gamma ray which can be used for imaging purposes [49]. There is a tumor absorbed dose-patient response relationship, with higher tumor absorbed doses associated with complete response [48]. In one study, the mean tumor absorbed dose was 232 Gy in complete responders, 147 Gy in patients with stable disease, and 116 Gy in patients with progressive disease [48]. Patients with an objective response have been shown to exhibit significantly higher overall survival than non-responders (19 months versus 7.5 months) [48].

Pre-procedure antibiotics are often given prior to radioembolization (a single dose of cefazolin IV), although infectious complications are rare [42]. However, for patients with a biliary anastomosis or incompetent sphincter, broad spectrum antibiotics are recommended starting before the procedure and continuing for 5 days following the intervention [42]. To reduce the risk of gastric and duodenal ulcers, a proton pump inhibitor can be administered before and for 1-4 weeks after treatment [42]. To reduce post-radioembolization syndrome (nausea, fatigue, and pain), an antiemetic and a steroid can be given before the procedure [42].

The average dose to the tumor is 100-600 Gy and less than 1% of the normal liver receives more than 30 Gy (if the healthy liver absorbs a dose higher than 30 Gy, the risk of irreversible liver damage limits the overall effectiveness of the treatment) [3,6,16]. Other reported dose limits for the liver are 50 Gy to one third or 35 Gy to two-thirds of the whole liver volume [16]. Other authors report that for lobar radioembolization the maximum tolerable normal liver absorbed dose is less than 70 Gy when using resin microspheres and less than 120 Gy when using glass microspheres [25,38]. For safe treatment, the dose to the lungs should be less than 30 Gy (other authors report the dose must not exceed 30 Gy to 20%, or 15 Gy to 30% of the whole lung volume [16]) [15]. Unfortunately, activity planning for radioembolization is inexact and this can contribute to the non-response rate and to hepatotoxicity (which can occur in up to 20% of patients) [24]. For bilobar disease, the left and right lobe are typically treated in separate sessions 4-8 weeks apart [42]. Treatment of the entire liver in a single session is associated with a higher rate of liver failure [42].

For glass microspheres, there is a strong correlation between tumor response and the dose absorbed by the tumor [15]. If the lesion-absorbed dose is too low, the procedure will be ineffective [16]. A response with improved progression free survival can be predicted using a tumoral threshold dose of 205 Gy or more [15]. Other authors report that when an average dose of 120 +/- 20 Gy can be delivered to the liver lobe bearing the tumor lesions, median survival ranges from 7.1-21 months in patients with HCC, and from 6.7 to 17 months in patients with colorectal liver metastases [16].

For resin microspheres, in one study, the 1-year survival for patients whose tumors received a dose of more than 55 Gy was 100%, whereas the survival was 24% if the dose was below 55 Gy [24]. A tumor dose of over 77 Gy was associated with a 2 year survival of 100%, whereas the survival was 10% when the dose was below 77 Gy [24]. In another study using 90Y-microspheres, overall survival was greatest in patients with tumor absorbed doses of 100 Gy or higher [51]. Other authprs have suggested a 120 Gy threshold for target-tumor radiation absorbed dose with resin microspheres [52].

The treatment is well tolerated, does not typically induce significant hepatic or pulmonary toxicity, and can be administered on an outpatient basis because post embolization syndrome is minimal [1]. Also- the treatment results in very little radiation exposure to heath care workers or family members [4].

Eligible patients should be non-surgical candidates with adequate liver function (Child-Pugh score less than or equal to B7) [5,32]. Relative contraindications include main portal vein thrombosis (although patients with PVT can be treated safely and effectively with a meaningful increase in overall survival, particularly for patients with tumors smaller than 5 cm and Child-Pugh class A patients [43]), bile duct abnormalities or stents, a serum bilirubin > 34 umol/L (2mg/dL), a leukocyte count < 200 or a platelet count < 60,000, and a GFR < 35 [32].

Absolute contraindications include extensive and untreated portal hypertension, significant extrahepatic disease, a life expectancy of less than 3 months, active hepatitis, and unacceptable shunting on Tc-MAA pre-treatment imaging (uncorrectable flow to the GI tract observed or pulmonary shunting with more than 30 Gy estimated dose to be delivered to the lungs in a single dose, or more than 50 Gy in cumulative doses) [5,32]. Main portal vein thrombosis is also a contraindication, but treatment may be considered on a case by case basis [34]. Age is not a contraindication to treatment and has not been shown to alter prognosis [32]. Prior surgical liver resection is not a contraindication, but surgical procedures involving the biliary tract may be a risk factor for infectious complications [32]. Other contraindications include poor liver function (bilirubin > 2 mg/dL; albumin < 3 gm/dL; uncontrolled ascites) and poor performance status (Eastern Cooperative Oncology Group performance status  > 2) [42]. Caution is advised in patients who have a bilirubin level of 1.5 mg/dL (unless super-selective embolization is performed) and in patients with limited hepatic reserve [34].

Bevacizumab is typically withheld for at least 2 and ideally 4 weeks before mapping angiogram and radioembolization procedures, although optimal timing is unknown [42]. Bevacizumab interferes with wound healing, may result in hepatic artery dissection, and increases the risk of stasis being reached- resulting in inability to deliver the entire dose, as well as possible reflux and gastroduodenal ulceration [42].

Mapping lung shunting: Due to disorganized angiogenesis within metastatic lesions, particles may potentially pass through intratumoral hepatic shunts and lodge in the capillaries of the lungs [40]. Prior to treatment, vascular mapping and a hepatopulmonary shunt fraction should be determined using Tc-MAA and planar or SPECT-SPECT/CT imaging [6,18]. Vascular mapping is very important because anatomic variants of the hepatic arterial vasculature are common [32]. The normal hepatic arterial supply originates from the celiac trifurcation from which the common hepatic artery arises [32]. The common hepatic artery becomes the proper hepatic artery after the gastroduodenal artery branches off [32]. The proper hepatic artery branches into the right and left hepatic arteries [32].

Prior to injection, the Tc-MAA syringe should be gently tilted to agitate and re-suspend the MAA particles- this will minimize clumping of the particles [18]. The injection should be given slowly to avoid streaming [18]. The typical dose is 5 mCi (185 mBq) suspended in normal saline- this can be divided between the right (3mCi) and left (2 mCi) lobes if whole liver imaging is performed [18]. Tc-MAA can begin to significantly breakdown into free technetium and varied-sized particles within 75 minutes of administration [33]. Scintigraphy should be performed within one hour of MAA injection to prevent false positive extrahepatic activity due to free technetium (free tech usually produces diffuse gastric uptake and can also be seen in the thyroid gland and urinary system, while pathologic uptake is usually focal) [11,18,33]. The shunt fraction to the lungs is calculated by dividing the total lung counts by the sum of the lung and liver counts [6].

For resin microspheres the amount of administered activity is adjusted on the basis of the calculated shunt fraction [41]. For a shunt fraction of < 10%, there is no reduction in dose [41]. A shunt fraction of  > 20% is a contraindication to therapy with resin microspheres [6]. Treatment guidelines recommend a 20% decrease in administered radioactivity for patients with a lung shunt fraction between 10-15%, and a 40% decrease in administered activity for patients with lung shunt fractions between 15-20% [40]. However, reducing the administered activity can result in sub-therapeutic treatment dosing [40]. The degree of liver cirrhosis has been shown to influence the intra-hepatic 90Y distribution [47]. Compared to patients with Child-Pugh A liver disease, those with Child-Pugh B have demonstrated increased rates of non-target 90Y delivery and higher lung shunt functions [47]. This may be related to cirrhosis associated structural changes with portal hypertension, arterioportal, and hepatovenous shunting [47]. A reduced dose delivered to the tumor results in lower response rates [47].

Because glass microspheres contain more activity per microsphere, a shunt fraction of 10% should be used [6]. Other authors suggest that for glass microspheres the upper limit of allowed activity shunted to the lungs is 16.5 mCi, calculated by multiplying the lung fraction shunt percentage by the planned therapeutic activity [41].

The highest tolerable accumulated absorbed dose to the lungs is defined as 30 Gy after a single treatment and up to 50 Gy after repeated treatments [32]. Bevacizumab is an antiangiogenic agent that is being incorporated into many metastatic colorectal cancer treatment regimens and may be expected to decrease the disorderly angiogenesis of tumor growth and result in a lower lung shunt fraction [40].

If other sites of extrahepatic activity are identified on Tc-MAA imaging, coil embolization of the culprit vessel, or a more distal position of the catheter/superselective catheterization (such as placing the microcatheter distal to the cystic artery) can be used during the procedure [32,33]. In the absence of significant extrahepatic activity, the other dosimetric limitation is total absorbed radiation dose in the healty liver parenchyma [32]. A nontumor liver dose of less than 70 Gy (or 50 Gy in cirrhotic livers) has been proposed [32].

SPECT imaging following MAA administration leads to more accurate calculation of lung shunting (the lung shunt absorbed dose is typically overestimated by planar imaging compared to SPECT [32]) and SPECT is more sensitive and able to detect shunting in a larger number of patients [8]. SPECT/CT has an even higher sensitivity, specificity, and accuracy for the detection of abnormal shunting/extrahepatic sites of activity [8,11,18]. In a study comparing planar, SPECT, and SPECT/CT imaging, the sensitivity for detecting extrahepatic shunting was 32%, 41%, and 100%, respectively [11]. SPECT/CT permits better localization of extrahepatic activity- especially for areas such as the gallbladder wall and duodenum that may lie in close proximity to the liver [11]. The therapy plan may be changed in up to 29% of patients based upon the SPECT/CT exam findings [11]. Gastric activity is also more commonly seen on SPECT imaging, but can sometimes occur secondary to free pertechnetate [14]. The administration of sodium perchlorate prior to tracer administration has been suggested as a means to decrease free pertechnetate activity in the stomach [14]. Focal increased MAA activity in the falciform artery, phrenic artery, duodenum, gastric lumen, or anywhere along the GI tract is concerning for extrahepatic shunting due to hepaticoenteric arterial communications [33]. These vessels include the falciform, accessory or left phrenic, right, or accessory gastric arteries (from the left hepatic artery), supraduodenal, retroduodenal, and accessory right hepatic artery feeding segment 6 (from the gastroduodenal artery) [33].

One might expect a higher response to radioembolization of tumors that demonstrate high MAA uptake compared to those with only low MAA activity [30]. Interestingly, in most instances the degree of intra-tumoral uptake of Tc-MAA does not appear to predict the likelihood for Y90 uptake or response to Y90 resin microspheres and treatment should not be withheld from patients with liver tumors or colorectal liver metastases lacking intratumoral Tc-MAA accumulation [19,35]. In one study, more than 60% of lesions with a pre-therapy uptake lower than healthy liver tissue showed uptake following radioembolization [35]. This discrepancy may be related to the number of particles used for the MAA study (approximately 150,000), compared to the number of resin particles used for treatment (23 million - 300 times more) [19]. Therefore, even lesions that appear hypovascular on Tc-MAA imaging, may receive a sufficient number of particles to have a therapeutic effect [19]. However, MAA uptake in hepatocellular carcinoma has been shown to be predictive of response following radioembolization with glass microspheres with responding tumors having almost double the MAA uptake of nonresponding HCC [30]. Personalized dosimetry based on the MAA scan in a select group of patients with HCC and portal vein thrombosis have been shown to result in prolonged overall survival following treatment with glass microspheres [31]. In general, the lesions which generally have the highest MAA activity include HCC, NET, and cholangiocarcinoma [30]. In patients with metastatic colorectal cancer, a tumor-to-normal liver uptake ratio of greater than 1 has been correlated with a good metabolic response [42].

Some authors suggest that MAA distribution does not accurately predict the 90Y distribution (for resin microspheres) [21]. In one study, up to 68% of segments demonstrate a greater than 10% difference between MAA and 90Y activity [21]. These discrepancies may be related to slight differences in catheter positioning, physiologic variance in hepatic blood flow, and morphologic differences between MAA particles and 90Y microspheres [21].

Combined Tc-MAA and Tc-sulfur colloid imaging can provide information regarding treatment distribution and functioning liver tissue that can aid in proper dose determination (this method assumes that the intact reticuloendothelial function defined by the Tc-SC scan also corresponds to regions of intact hepatocellular function) [24].


Shunting to lung: The patient below was referred for Y90-microshere therapy. A pre treatment TcMAA hepatic arteriogram revealed a severe intrahepatic arteriovenous shunt with a shunt fraction of 87%. Note the intense lung activity.
Coronal CtTc Maa ShuntingA V Shunting


Dosimetric assessment and treatment activity determination:

The maximum activity to be injected to the patient is determined using one of three methods for resin microspheres - the body surface area, the empiric model, or the partition model- or the volume-based model for glass microspheres [27,32,44].

The empiric model recommends exclusively three values of activity based on tumor burden in the liver [44,46]. For tumor involvement of more than 50% of the liver, 3.0 GBq of activity is recommended; for 25-50% 2.5 Gbq is recommended; and for less than 25% tumor involvement 2.0 GBq is recommended [46]. This method has been replaced by the body surface area method [46].

The body surface area (BSA) is based on patient surface area (calculated from the patients weight and height) and percentage of liver tumor involvement (the injected activity is adjusted depending on tumor burden and the patients physical characteristics), but neglects the tumor-to-normal liver uptake ratio in [27,44]. Additionally, this simple method does not incorporate tumor mass, a tumor-absorbed dose, and it does it account for inter-individual differences in microsphere distribution and as a result, the achieved tumor-absorbed dose may be suboptimal and impair treatment efficacy [37].

The partition model is a dosimetric model based on the MIRD approach in which limit values on mean absorbed doses to organs at risk (the lungs and non-tumoral liver) are considered [27]. A noncompartmental MIRD model can be used for glass microspheres and a compartmental MIRD model can be used with either glass or resin microspheres [46]. With the MIRD method, the tumor-to-nontumor tissue ratio is used to express the relative distribution of Tc-MAA by determining areas of interest in healthy and tumoral liver tissue at SPECT image acquisition [46]. The aim is to deliver a tumoricidal dose to the tumor while preserving safe limits of radiation to normal liver parenchyma and the lungs [46]. The recommended safe dose limits are 70 Gy for non tumor liver tissue (< 50 Gy in cirrhotic livers) and 30 Gy to the lungs in a single injection, or 50 Gy total in subsequent treatments [46].

Although this method is more accurate and personalized and permits better therapy selectivity, its main drawback is the underlying assumption of homogeneous activity within regions of interest (uniform dose distribution in tumor) [27,44].

It has been shown that metastases with a higher tumor-absorbed dose have a better metabolic response and this is associated with prolonged overall survival [37].

To decrease the risk of stasis and reflux during administration the microspheres should be given using a slow and pulsatile injection technique [33]. Because Y90 is bound to the resin micospheres through an ion exchange mechanism, sterile water (which is nonionic) has traditionally been used for administration [33]. However, sterile water can remove arterial endothelium and cause vessel constriction/spasm [33]. Using glucose 5% solution (a physiologic isotonic nonionic solution) may help to prevent endothelial injury and vasoconstriction and reduce the need for periprocedural pain medication [33].

Immediately following embolization, planar or SPECT imaging should be performed to detect bremsstrahlung radiation (produced by interaction of the emitted beta-particles with adjacent tissue) and confirm intrahepatic/tumor deposition of the microspheres [6]. Although SPECT imaging has the potential to provide the most accurate depiction of tracer activity, the wide range (0-2.3 MeV) and continuous nature of the 90Y bremsstrahlung photon spectrum prohibit the use of simple energy window-based scatter rejection and correction techniques, and require compensation for collimator and detector related imaging-degrading artifacts such as collimator scatter, lead x-rays, septal penetration, and partial energy deposition in the crystal [22]. Monte Carlo-based modeling can substantially improve image quality and quantitative accuracy of 90Y bremsstrahlung SPECT images [22]. Recent studies also suggest that there is a low incidence of positron decays associated with Y90 that can be detected on PET/CT [12], but this requires use of a time-of-flight PET/CT to obtain images with sufficiently high quantitative accuracy if dosimetric evaluation is going to be performed [22].

Results:

Hepatocellular carcinoma:

Up to 79% of patients with HCCa can show a positive tumor response [6] and prolonged time to progression compared to chemoembolization [41]. The greater the amount of radiation delivered to the tumor, the better the response rate [6]. In one study, a mean tumor dose of 215 Gy was noted in responders (partial or complete) vs 167 in non-responders [32].

Metastatic disease:

Approximately 45% of patients with colorectal cancer develop liver metastases [39]. These metastases are synchronous (present at diagnosis) in 25% of patients or metachronous in 20% of patients [39]. The only potential curative treatment is liver resection, which is associated with 5-year survival rates of 20-58% [39]. However, only approximately 15% of patients are eligible to undergo resection [39]. In colorectal cancer patients, modern chemotherapy regimens and biologic agents have significantly prolonged the median overall survival of patients with liver metastases to approximately 29-32 months [36]. However, once hepatic metastases become chemorefractory, survival is poor and typically between 4 to 5 months [36].  90Y radioembolization can be used for treatment in chemorefractory patients.

For patients with metastatic colorectal cancer, microsphere embolization has been associated with better median survival compared to chemotherapy alone (approximately 10.5-10.6 months for both resin and glass microspheres) [6,36]. In one study of patients with unresectable liver metastases, microsphere therapy produced a complete response in 2% of patients and a partial response in 43% [7]. Some decrease in tumor size was noted in 87% of patients [7]. Survival following treatment was best for patients with four or fewer lesions and those with neuroendocrine tumors [7]. In patients with metastatic colorectal adenocarcinoma, a lung shunt fraction of more than 10% has been shown to be an independent predictor of significantly decreased survival following embolization [40]. Other factors associated with shorter survival include a ECOC performance status of greater than or equal to 1, low albumin level, presence of extrahepatic metastases, lymphovascular invasion of the primary tumor, CEA level of greater than 62 ng/mL, KRAS mutant tumors, and greater than 25% tumor involvement of the treated liver volume [42].

For treatment of metastatic neuroendocrine tumor, a meta-analysis found an objective response rate (CR or PR) of 50% and a weighted avergae disease control rate (CR, PR, or stable disease) of 86% [29]. The response rate was overall slightly lower for patients with metastatic pancreatic neuroendocrine tumor (pancreatic neuroendocrine tumors tend to be more aggressive and small bowel primaries have a nearly 2 fold higher 5 year survival rate [29]).

Radioembolization in combination with systemic chemotherapy can be used to increase median survival in patients with metastasis confined to the liver [17]. A literature review found disease control rates (complete response, partial response, and stable disease) ranged from 29-90% for  90Y radioembolization and from 59-100% for radioembolization combined with chemotherapy [23]. Survival at 12 months ranged from 37-59% for Y90 treatment alone, and from 43-74% for Y90 with chemotherapy [23]. The article also noted that there is a large amount of heterogeneity in the patient populations being studied with regards to disease extent, prior therapies, patient performance status, and criteria used to determine tumor response [23].

Standard RECIST criteria can underestimate response following treatment [39]. Due to edema or inflammation, responding tumors may increase in size following treatment (before 30 days) [46] - although the responding lesions should show evidence of necrosis [2] and decreased attenuation [17]. In general- the presence of necrosis (and decreased attenuation) is a better indicator of response [5,10,17]. Some authors recommend a wait of 3 months before assessing tumor response [46]. Ring enhancement about the lesions may also be seen following therapy and often represents granulation tissue or fibrosis rather than neoplastic tissue- the ring enhancement may persist for months [5,6]. This rim of enhancement is usually smooth and less than 5 mm thick and can be seen in about one-third of treated lesions (an enhancing peripheral nodule is more concerning for residual tumor) [46]. Variable areas of necrosis and residual enhancement do not have predictive value if they are present during the early followup period (30 days), however, persistence after 90 days most likely represents residual disease [46]. Hypertrophy of the non-embolized liver lobe may also occur following treatment [5].

Modified RECIST criteria used for therapy response evaluation include- complete response- disappearance of any intra-tumoral enhancement in all target lesions; partial response- > 30% decrease in the sum of the diameters of the viable target lesions; stable disease; and progressive disease - > 20% increase in the sum of the diameters of viable target lesions [46]. The Choi criteria define a partial response as a 10% reduction in size or a 15% reduction in the attenuation of treated lesions during the portal venous phase of imaging [46].

PET imaging is able to better detect lesion response compared to conventional imaging [2,5]. Responding lesions will demonstrate decreased tracer uptake and SUVmax values [10]. However, some authors suggest that changes in the total metabolic volume and total lesion glycolytic rate are better predictors of survival than changes in SUVmax or RECIST 1.1 criteria [20]. Patients demonstrating a response on FDG PET imaging have been shown to have longer survival periods compared to patients with non-responding lesions [17]. FDG PET response is best assessed 12 weeks following radioembolization [17]. However, PET is limited in its ability to detect small tumors [2].

Common side effects include fatigue, self-limited abdominal pain, nausea, fever, anorexia, and diarrhea [7]. The embolic effect of resin microspheres can sometimes lead to acute ischemic pain during injection, however, it has been shown that when 5% glucose is used instead of sterile water for injection, there is less pain, less stasis, and more efficient administration [32].

Complications:

- Post-radioembolization syndrome: Symptoms include nausea, fatigue, pain, and low grade fever that last for 1-2 weeks following treatment [42].

- Radiation cholecystitis: Due to microspheres entering the cystic artery [5]. Radiation cholecystitis can occur in up to 23% of patients and liver edema up to 42% of patients [2]. Most patients are asymptomatic and imaging findings generally improve with conservative treatment, however, cholecystectomy may be required (in one article only 0.8% of patients developed clinically significant radiation-induced cholecystitis [28] and another indicated that fewer than 1% of patients with radiation-induced cholecystitis require surgical intervention [46]) [2,5]. Radiation cholecystitis appears as GB wall thickening, enhancement, and discontinuity on CT imaging [2].

- Radiation hepatitis/ radioembolization induced liver disease (REILD): Radiation induced liver disease can develop 4-8 weeks after radioembolization (although more delayed hepatic toxicity can also occur) in 4-9% of patients, but can be seen in up to 20% of patients, particularly those that have undergone pretreatment with chemotherapeutic agents [42,46]. Risk factors for radioembolization induced liver disease include prior chemotherapy, lower tumor burden, high baseline bilirubin, younger age, low body mass index, whole liver radioembolization, non-HCC pathology, and cirrhotic liver disease [32,42]. Jaundice, elevated LFTs (bilirubin and alkaline phosphatase), and ascites in the absence of tumor progression or bile duct dilatation are the main symptoms of radioembolization induced liver disease [32].

Histology shows venoocclusive disease in severe cases [42]. Treatment for radiation hepatitis/radiation induced liver disease is usually medical (steroids and anti-inflammatory drugs) [5].

Radiologic findings include intraparenchymal edema and hepatomegaly [5].

- Biliary necrosis and biloma: Potential biliary complications such as cholangitis and biloma can be seen [46]. Unlike the liver which has a dual blood supply, the biliary tree has only a single blood supply- the peribiliary plexus [46]. Acute biliary necrosis is usually seen as small cystic structures adjacent to a portal venous branch within the distribution of an embolized artery or in clusters around a treated tumor [46]. Leaking bile can accumulate to form a biloma [46]. Interestingly, compared to non-cirrhotic livers, cirrhotic livers have a lower risk of biliary necrosis after radioembolization due to hypertrophy of the peribiliary pelxus [46].

- Radiation pancreatitis

- GI tract ulceration- when microspheres enter into the GI circulation via the gastroduodenal artery, right gastric artery, or other vessels supplying the stomach and small bowl, local radiation can result in ulceration [5]. 90Y induced ulcers in the stomach or duodenum can be resistant to medical therapy and surgery may be required [11]. Prophylactic embolization of the gastroduodenal artery, right gastric, and other extrahepatic vessels is recommended by some authors because the risks of reflux outweigh the risk of embolization of these vessels [6,11]. Because these vessels and the organs they supply can revascularize quickly, the embolization should be performed in close proximity to the time of the planned microsphere therapy [11].

- Hepatic biloma or abscess (particularly in patients with incompetant ampulla of Vater) [5].

- Radiation pneumonitis- due to tumor-associated arteriovenous shunting [6]. A hepatopulmonary shunt fraction should be determined prior to treatment to prevent pulmonary toxicity [6].

- Infection- infectious complications such as liver abscess and cholangitis following TARE are rare in the setting of an intact ampulla of Vater (approximately 0-2%) [45]. However, the risk for infection has been shown to be much greater in patients with biliary enteric anastomosis or stents and drains spanning across the ampulla of Vater- between 10-48% [45]. Infection risk has been shown to remain elevated despite antibiotic prophylaxis [45] and the risk for infection appears to be increased with the use of glass microspheres [45].

Repeat Radioembolizations:

In patients with advanced liver tumors, repeat radioembolization can be performed safely using a sequential lobar approach with 4 to 6 weeks between treatment sessions and proper pre-treatment patient selection (exclusion of patients with bilirubin levels exceeding 30 umol/L) [26].

REFERENCES:

(1) AJR 2007; Keppke AL, et al. Imaging of hepatocellular carcinoma after treatment with Yttrium-90 microspheres. 188: 768-775

(2) AJR 2007; Miller FH, et al. Response of liver metastases after treatment with Yttrium-90 microsphers: role of size, necrosis, and PET. 188: 776-783

(3) AJR 2007; Welsh JS. Radiographically identified necrosis after 90Y microsphere brachytherapy: a new standard for oncologic response assessment? 188: 765-767

(4) J Nucl Med 2007; Gulec SA, Siegel JA. Posttherapy radiation safety considerations in radiomicrosphere treatment with 90Y microspheres. 48: 2080-2086

(5) Radiographics 2008; Atassi B, et al. Multimodality imaging following 90Y radioembolization: a comprehensive review and pictorial essay. 28: 81-99

(6) Radiographics 2008; Kalva SP, et al. Recent advances in transarterial therapy of primary and secondary liver malignancies. 28: 101-117

(7) Radiology 2008; Sato KT, et al. Unresectable refractory liver metastases: radioembolization with 90Y microspheres - safety, efficacy, and survival. 247: 507-515

(8) J Nucl Med 2009; Hamami ME, et al. SPECT/CT with 99mTc-MAA in radioembolization with 90Y microspheres in patients with hepatocellular cancer. 50: 688-692

(9) J Nucl Med 2010; Gulec SA, et al. Hepatic structural dosimetry in 90Y-microsphere treatment: a monte carlo modeling approach based on lobular microanatomy. 51: 301-310

(10) Radiology 2010; Tochetto SM, et al. Does multidetector CT attenuation change in colon cancer liver metastases treated with 90Y help predict metabolic activity at FDG PET? 255: 164-172

(11) J Nucl Med 2010; Ahmadzadehfar H, et al. The significance of 99mTc-MAA SPECT/CT liver perfusion imaging in treatment planning for 90Y-microsphere selective internal radiation treatment. 51: 1206-1212

(12) J Nucl Med 2011; Gates VL, et al. Internal pair production of 90Y permits hepatic localization of microspheres using routine PET: proof of concept. 52: 72-76

(13) Radiology 2011; Lewandowski RJ, et al. Transcatheter intraarterial therapies: rationale and overview. 259: 641-657

(14) J Nucl Med 2011; Sabet A, et al. Significance of oral administration of sodium percholate in planning liver-directed radioembolization. 52: 1063-1067

(15) J Nucl Med 2012; 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. 53: 255-263

(16) J Nucl Med 2012; Traino AC, et al. Radiodosimetric estimates for radioembolic therapy of liver tumors: challenges and opportunities. 53: 509-511

(17) AJR 2012; Tochetto SM, et al. Colorectal liver metastasis after 90Y radioembolization therapy: pilot study of change in MDCT attenuation as a surrogate marker for future FDG PET response. 198: 1093-1099

(18) J Nucl Med 2012; Uliel L, et al. From the angio suite to the gamma camera: vascular mapping and 99mTc-MAA hepatic perfusion imaging before liver radioembolization- a comprehensive pictorial review. 53: 1736-1747

(19) J Nucl Med 2013; Ulrich G, et al. Predictive value of intratumoral 99mTc-macroaggregated albumin uptake in patients with colorectal liver metatsases scheduled for radioembolization with 90Y-microspheres. 54: 516-522

(20) J Nucl Med 2013; 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. 54: 1202-1208

(21) J Nucl Med 2013; Wondergem M, et al. 99mTc-macroaggregated albumin poorly predicts the intrahepatic distribution of 90Y resin microspheres in hepatic radioembolization. 54: 1294-1301

(22) J Nucl Med 2013; Elschot M, et al. Quantitative Monte Carlo-based 90Y SPECT reconstruction. 54: 1557-1563

(23) J Nucl Med 2013; Rosenbaum C, et al. Radioembolization for treatment of salvage patients with colorectal cancer liver metastases: a systemic review. 54: 1890-1895

(24) J Nucl med 2013; Lam MGEH, et al. Prognostic utility of 90Y radioembolization dosimetry based on fusion of 99mTc-macroaggregated albumin-99mTc-sulfur colloid SPECT. 54: 2055-2061

(25) J Nucl Med 2014; Walrand S, et al. The low hepatic toxicity per gray of 90Y glass microspheres is linked to their transport in the arterial tree favoring a nonuniform trapping as observed in posttherapy PET imaging. 55: 135-140

(26) J Nucl Med 2014; 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. 55: 360-366

(27) J Nucl Med 2014; Petitquillaume A, et al. Three-dimensional personalized Monte Carlo dosimetry in 90Y resin microspheres therapy of hepatic metastases: nontumoral liver and lungs radiation protection considerations and treatment planning optimization. 55: 405-413

(28) J Nucl Med 2014; Sag AA, et al. Yttrium-90 radioembolization of malignant tumors of the liver: gallbladder effects. 202: 1130-1135

(29) J Nucl Med 2014; Devcic Z, et al. The efficacy of hepatic 90Y resin radioembolization for metastatic neuroendocrine tumors: a meta-analysis. 55: 1404-1410

(30) J Nucl Med 2015; Ilhan H, et al. Systemic evaluation of tumoral 99mTc-MAA uptake using SPECT and SPECT/CT in 502 patients before 90Y radioembolization. 56: 333-338

(31) J Nucl Med 2015; 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. 56: 339-346

(32) J Nucl Med 2015; Braat AJ, et al. 90Y hepatic radioembolization: an update on current practice and recent developments. 56: 1079-1087

(33) J Nucl Med 2015; Gates VL, et al. Intraarterial hepatic SpECT/CT imaging using 99mTc-macroaggregated albumin in preparation for radioembolization. 56: 1157-1162

(34) Radiographics 2015; Camacho JC, et al. 90Y radioembolization: multimodality imaging pattern approach with angiographic correlation for optimal target therapy delivery. 35: 1602-1620

(35) J Nucl Med 2015; Ilhan H, et al. Predictive value of 99mTc-MAA SPECT for 90Y-labeled resin microsphere distribution in radioembolization of primary and secondary hepatic tumors. 56: 1654-1660

(36) J Nucl Med 2016; Hickey R, et al. 90Y radioembolization of colorectal hepatic metastases using glass microspheres: safety and survival outcomes from a 531-patient multicenter study. 57: 665-671

(37) J Nucl Med 2016; van den Hoven AF, et al. Insights into the dose-response relationship of radioembolization with 90Y-microspheres: a prospective cohort study in patients with colorectal cancer liver metastases. 57: 1014-1019

(38) J Nucl Med 2016; Pasciak AS, et al. A microdosimetric analysis of absorbed dose to tumor as a function of number of microspheres per unit volume in 90Y radioembolization. 57: 1020-1026

(39) AJR 2016; Shady W, et al. Surrogate imaging biomakers of response of colorectal liver metastases after salvage radioembolization using 90Y-loaded resin microspheres. 207: 661-670

(40) Radiology 2017; Narsinh KH, et al. Hepatopulmonary shunting: a prognostic indicator of survival in patients with metastatic colorectal adenocarinoma treated with 90Y radioembolization. 282: 281-288

(41) AJR 2017; Jadvar H. Targeted radionuclide therapy: an evolution toward precision cancer treatment. 209: 277-288

(42) J Nucl Med 2017; Boas FE, et al. Radioembolization of colorectal liver metastases: indications, technique, and outcomes. 58: 104S-111S

(43) J Nucl Med 2018; Abouchaleh N, et al. 90Y radioembolization for locally advanced hepatocellular carcinoma with portal vein thrombosis: long-term outcomes in a 185-patient cohort. 59: 1042-1048

(44) J Nucl Med 2018; Kafrouni M, et al. Retrospective voxel-based dosimetry for assessing the ability of the body-surface-area model to predict delivered dose and radioembolization outcome. 59: 1289-1295

(45) Radiology 2018; Devulapalli KK, et al. 90Y radioembolization for hepatic malignancy in patients with previous biliary intervention: multicenter analysis of hepatobiliary infections. 288: 774-781

(46) Radiographics 2019; Spina JC, et al. Expected and unexpected imaging findings after 90Y transarterial radioembolization for liver tumors. 39: 578-595

(47) J Nucl Med 2019; Schobert I, et al. Quanitative imaging biomarkers for 90Y distribution on bremsstrahlung SPECT after resin-based radioembolization. 60: 1066-1072

(48) J Nucl Med 2020; Bastiaannet R, et al. First evidence for a dose-response relationship in patients treated with 166Ho radioembolization: a prospective study. 61: 608-612

(49) J Nucl Med 2018;Prince JF, et al. efficacy of radioembolization with 166Ho-microspheres in salvage patients with liver metastases: a phase 2 study. 59: 582-588

(50) J Exp Clin Cancer Res 2010; Smits MLJ, et al. Holmium-166 radioembolization for the treatment of patients with liver metastases: design of the phase I HEPAR trial. 29: 70.

(51) Radiology 2020; Hermann AL, et al. Relationship of tumor radiation- absorbed dose to survival and response in hepatocellular carcinoma treated with transarterial radioembolization with 90Y in the SARAH study. 296: 673-684

(52) Int J Radiat Oncol Biol Phys 2012; Lau WY, et al. Patient selection and activity planning guide for selective internal radiotherapy with yttrium-90 resin microspheres. 82: 401-407

Page 1 of 6
Next Page