Intraprocedural MRI-based dosimetry during transarterial radioembolization of liver tumours with holmium-166 microspheres (EMERITUS-1): a phase I trial towards adaptive, image-controlled treatment delivery

Joey Roosen, Lovisa E L Westlund Gotby, Mark J Arntz, Jurgen J Fütterer, Marcel J R Janssen, Mark W Konijnenberg, Meike W M van Wijk, Christiaan G Overduin, J Frank W Nijsen, Joey Roosen, Lovisa E L Westlund Gotby, Mark J Arntz, Jurgen J Fütterer, Marcel J R Janssen, Mark W Konijnenberg, Meike W M van Wijk, Christiaan G Overduin, J Frank W Nijsen

Abstract

Purpose: Transarterial radioembolization (TARE) is a treatment for liver tumours based on injection of radioactive microspheres in the hepatic arterial system. It is crucial to achieve a maximum tumour dose for an optimal treatment response, while minimizing healthy liver dose to prevent toxicity. There is, however, no intraprocedural feedback on the dose distribution, as nuclear imaging can only be performed after treatment. As holmium-166 (166Ho) microspheres can be quantified with MRI, we investigate the feasibility and safety of performing 166Ho TARE within an MRI scanner and explore the potential of intraprocedural MRI-based dosimetry.

Methods: Six patients were treated with 166Ho TARE in a hybrid operating room. Per injection position, a microcatheter was placed under angiography guidance, after which patients were transported to an adjacent 3-T MRI system. After MRI confirmation of unchanged catheter location, 166Ho microspheres were injected in four fractions, consisting of 10%, 30%, 30% and 30% of the planned activity, alternated with holmium-sensitive MRI acquisition to assess the microsphere distribution. After the procedures, MRI-based dose maps were calculated from each intraprocedural image series using a dedicated dosimetry software package for 166Ho TARE.

Results: Administration of 166Ho microspheres within the MRI scanner was feasible in 9/11 (82%) injection positions. Intraprocedural holmium-sensitive MRI allowed for tumour dosimetry in 18/19 (95%) of treated tumours. Two CTCAE grade 3-4 toxicities were observed, and no adverse events were attributed to treatment in the MRI. Towards the last fraction, 4/18 tumours exhibited signs of saturation, while in 14/18 tumours, the microsphere uptake patterns did not deviate from the linear trend.

Conclusion: This study demonstrated feasibility and preliminary safety of a first in-human application of TARE within a clinical MRI system. Intraprocedural MRI-based dosimetry enabled dynamic insight in the microsphere distribution during TARE. This proof of concept yields unique possibilities to better understand microsphere distribution in vivo and to potentially optimize treatment efficacy through treatment personalization.

Registration: Clinicaltrials.gov, identifier NCT04269499, registered on February 13, 2020 (retrospectively registered).

Keywords: Dosimetry; Holmium; Image-guided; Personalization; SIRT; TARE.

Conflict of interest statement

J. F. W. Nijsen is a co-founder of Quirem Medical which has been acquired by Terumo Europe NV in July 2020. Nijsen has a scientific advisory role and is entitled to certain milestone payments from Terumo which are related to Quirem’s financial, operational and regulatory performance in the future. Furthermore, Nijsen is an inventor on the patents related to radioactive microspheres that are assigned to University Medical Center Utrecht Holding BV, Quirem Medical or BASF Corp. The activities of J. F. W. Nijsen within Quirem Medical are approved and supported by the Board of Directors of the Radboudumc. The other authors declare that they have no conflict of interest.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Workflow during the treatment procedure. Top row represents treatment steps at the MRI scanner, and bottom row represents treatment steps at the cone-beam CT. Microspheres were injected in 4 fractions (10% and three times 30% of the microspheres), with imaging for MRI-based dosimetry in between
Fig. 2
Fig. 2
Relationship between injected activity and mean tumour and liver doses of two patients, in whom different uptake patterns were observed (A and B). Lines are linear trendlines based on the first 3 data points, and the dashed line indicates where the fourth data point is expected based on the linear trend. A A patient with hepatocellular carcinoma (patient 4) with a linear increase in mean dose in all volumes of interest. B A patient with colorectal cancer metastases (patient 5) with an initial linear increase in mean dose in the tumours (up to 70% injected activity), and then with a decrease in the relative additive dose on the fourth fraction compared to the first three fractions
Fig. 3
Fig. 3
99mTc-SPECT-based treatment simulation and MRI-based dose distributions in patient 5 with colorectal carcinoma liver metastases, in two different slices (rows A and B). Column 1: treatment simulation after injection of 99mTc-MAA. The high activity deposition in segment IVa and the gall bladder (arrowheads) and low uptake in the dorsal part of the second tumour (row B) are probably a result of vasospasm during injection. Column 2: T1-weighted MR images with tumours delineated with the dashed line. Columns 3–5: fusion with MRI-based dose maps generated after 40%, 70% and 100% of the activity had been injected. As more activity is administered, the mean tumour dose increases, but the tumour coverage hardly improves
Fig. 4
Fig. 4
Digital subtraction angiography of the superior mesenteric artery (SMA) of a patient with breast cancer liver metastases (patient 1). A The chosen injection position proximal in the SMA. The asterisk indicates a branch of the SMA that is further explored in B. B More distal branch of the SMA, with contrast enhancement of tumour vasculature (T) that could have been opted for during image-guided TARE
Fig. 5
Fig. 5
99mTc-SPECT-based treatment simulation and MRI-based dose distributions in a tumour that received a low mean dose (breast cancer metastasis, patient 1) via the superior mesenteric artery (SMA), after different fractions of microspheres had been injected. The tumour is delineated with the dashed line. The asterisk in the leftmost MR image indicates a large, confluent tumour, which was vascularized through both the SMA and the right hepatic artery. Dmean indicates the mean dose in the delineated tumour
Fig. 6
Fig. 6
A tumour (hepatocellular carcinoma) of patient 4 treated via the right hepatic artery, in which a small part that was vascularized through an aberrant vessel originating from the phrenic artery remained untreated. A MRI-based dose distributions after each of the four fractions of radioactive microspheres had been administered. The tumour is delineated with the dashed line, and the arrow indicates the untreated area. B99mTc-SPECT/CT acquired for treatment simulation. C and D Diffusion-weighted MRI prior to and 3 months after treatment, indicating persistent diffusion restriction in the untreated area (arrow). E Digital subtraction angiography of the aberrant vessel; the asterisk indicates the persistent tumour vasculature. 166Ho-SPECT/CT after re-treatment of the patient via the aberrant vessel, with high uptake in the untreated area (arrow)

References

    1. 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):1624–1636. doi: 10.1016/S1470-2045(17)30683-6.
    1. Radosa CG, et al. Holmium-166 radioembolization in hepatocellular carcinoma: feasibility and safety of a new treatment option in clinical practice. Cardiovasc Intervent Radiol. 2019;42(3):405–412. doi: 10.1007/s00270-018-2133-7.
    1. 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):1159–1171. doi: 10.1016/S1470-2045(17)30457-6.
    1. Prince JF, et al. Efficacy of radioembolization with (166)Ho-microspheres in salvage patients with liver metastases: a phase 2 study. J Nucl Med. 2018;59(4):582–588. doi: 10.2967/jnumed.117.197194.
    1. van Roekel C, et al. Dose-effect relationships of holmium-166 radioembolization in colorectal cancer. J Nucl Med. 2021;62(2):272–9. 10.2967/jnumed.120.243832.
    1. Reimer P, et al. Prognostic factors in overall survival of patients with unresectable intrahepatic cholangiocarcinoma treated by means of yttrium-90 radioembolization: results in therapy-naive patients. Cardiovasc Intervent Radiol. 2018;41(5):744–752. doi: 10.1007/s00270-017-1871-2.
    1. Pieper CC, et al. Yttrium-90 radioembolization of advanced, unresectable breast cancer liver metastases-a single-center experience. J Vasc Interv Radiol. 2016;27(9):1305–1315. doi: 10.1016/j.jvir.2016.05.028.
    1. Breedis C, Young G. The blood supply of neoplasms in the liver. Am J Pathol. 1954;30(5):969–977.
    1. 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;48(12):3776–90. 10.1007/s00259-021-05340-0.
    1. 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. 10.1148/radiol.2020191606.
    1. 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. 2021;6(1):17–29. 10.1016/S2468-1253(20)30290-9.
    1. Smits ML, et al. In vivo dosimetry based on SPECT and MR imaging of 166Ho-microspheres for treatment of liver malignancies. J Nucl Med. 2013;54(12):2093–2100. doi: 10.2967/jnumed.113.119768.
    1. 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):827–835. doi: 10.1007/s00330-012-2648-2.
    1. Smits ML, et al. Holmium-166 radioembolisation in patients with unresectable, chemorefractory liver metastases (HEPAR trial): a phase 1, dose-escalation study. Lancet Oncol. 2012;13(10):1025–1034. doi: 10.1016/S1470-2045(12)70334-0.
    1. Roosen J, et al. Development of an MRI-guided approach to selective internal radiation therapy using holmium-166 microspheres. Cancers. 2021;13(21):5462. doi: 10.3390/cancers13215462.
    1. 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. 10.1007/s00259-019-04460-y.
    1. Eisenhauer EA, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 11) Eur J Cancer. 2009;45(2):228–47. doi: 10.1016/j.ejca.2008.10.026.
    1. Peters SMB, et al. Intra-therapeutic dosimetry of [(177)Lu]Lu-PSMA-617 in low-volume hormone-sensitive metastatic prostate cancer patients and correlation with treatment outcome. Eur J Nucl Med Mol Imaging. 2022;49(2):460–9. 10.1007/s00259-021-05471-4.
    1. Del Prete M, et al. Personalized (177)Lu-octreotate peptide receptor radionuclide therapy of neuroendocrine tumours: initial results from the P-PRRT trial. Eur J Nucl Med Mol Imaging. 2019;46(3):728–742. doi: 10.1007/s00259-018-4209-7.
    1. 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):1695–1704. doi: 10.1007/s00259-019-04340-5.
    1. 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;48(5):1570–84. 10.1007/s00259-020-05163-5.
    1. Vente MA, et al. Clinical effects of transcatheter hepatic arterial embolization with holmium-166 poly(L-lactic acid) microspheres in healthy pigs. Eur J Nucl Med Mol Imaging. 2008;35(7):1259–1271. doi: 10.1007/s00259-008-0747-8.
    1. HEPAR Primary: Holmium-166-radioembolization in hepatocellular carcinoma patients; Identifier: NCT03379844; .
    1. Reinders MTM, et al. Safety and efficacy of holmium-166 radioembolization in hepatocellular carcinoma - the HEPAR Primary study. J Nucl Med. 2022. 10.2967/jnumed.122.263823.
    1. Gutjahr R, et al. Quantitative dual-energy CT material decomposition of holmium microspheres: local concentration determination evaluated in phantoms and a rabbit tumor model. Eur Radiol. 2021;31(1):139–148. doi: 10.1007/s00330-020-07092-1.
    1. Bakker RC, et al. Feasibility of CT quantification of intratumoural (166)Ho-microspheres. Eur Radiol Exp. 2020;4(1):29. doi: 10.1186/s41747-020-00157-2.
    1. Bakker RC, et al. Intratumoral injection of radioactive holmium-166 microspheres in recurrent head and neck squamous cell carcinoma: preliminary results of first use. Nucl Med Commun. 2018;39(3):213–221. doi: 10.1097/MNM.0000000000000792.
    1. Bonne L, et al. Holmium-166 microspheres are visible in target liver lesions on unenhanced computed tomography. J Gastrointestin Liver Dis. 2021;30(2):192. doi: 10.15403/jgld-3478.
    1. Arranja AG, et al. Preparation and characterization of inorganic radioactive holmium-166 microspheres for internal radionuclide therapy. Mater Sci Eng C Mater Biol Appl. 2020;106:110244. doi: 10.1016/j.msec.2019.110244.
    1. Arranja AG, et al. Radioactive holmium phosphate microspheres for cancer treatment. Int J Pharm. 2018;548(1):73–81. doi: 10.1016/j.ijpharm.2018.06.036.
    1. 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):451–461. doi: 10.1016/j.ijrobp.2018.05.062.
    1. 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):2110–2121. doi: 10.1007/s00259-018-4064-6.
    1. Henry EC, et al. Precision dosimetry in yttrium-90 radioembolization through CT imaging of radiopaque microspheres in a rabbit liver model. EJNMMI Phys. 2022;9(1):21. 10.1186/s40658-022-00447-1.
    1. Drescher R, et al. Radioembolization with holmium-166 polylactic acid microspheres: distribution of residual activity in the delivery set and outflow dynamics during planning and treatment procedures. J Endovasc Ther. 2021;28(3):452–462. doi: 10.1177/1526602821996719.

Source: PubMed

3
Se inscrever