Skip to main content
Neuro-Oncology logoLink to Neuro-Oncology
. 2024 May 4;26(Suppl 9):S215–S228. doi: 10.1093/neuonc/noae069

Radioligand therapies in meningioma: Evidence and future directions

Maximilian J Mair 1,2, Emeline Tabouret 3, Derek R Johnson 4, Erik P Sulman 5,6, Patrick Y Wen 7,8, Matthias Preusser 9, Nathalie L Albert 10,
PMCID: PMC11631075  PMID: 38702966

Abstract

Meningiomas are the most common intracranial neoplasms in adults. While most meningiomas are cured by resection, further treatment by radiotherapy may be needed, particularly in WHO grades 2 and 3 tumors which have an increased risk of recurrence, even after conventional therapies. Still, there is an urgent need for novel therapeutic strategies after the exhaustion of local treatment approaches. Radionuclide therapies combine the specificity of tumor-specific antibodies or ligands with the cytotoxic activity of radioactive emitters. Alongside this, integrated molecular imaging allows for a noninvasive assessment of predictive biomarkers as treatment targets. Whereas the concept of “theranostics” has initially evolved in extracranial tumors such as thyroid diseases, neuroendocrine tumors, and prostate cancer, data from retrospective case series and early phase trials underscore the potential of this strategy in meningioma. This review aims to explore the available evidence of radionuclide treatments and ongoing clinical trial initiatives in meningioma. Moreover, we discuss optimal clinical trial design and future perspectives in the field, including compound- and host-specific determinants of the efficacy of “theranostic” treatment approaches.

Keywords: meningioma, nuclear medicine, positron emission tomography, radionuclide treatment, theranostic


Key Points.

  1. Radionuclide treatments are approved in extracranial malignancies such as neuroendocrine tumors and metastatic prostate cancer based on pivotal clinical trial results.

  2. Small, early-phase studies of somatostatin receptor (SSTR)-targeted radionuclide treatments suggest promising activity in meningioma. Well-designed clinical trials are needed to corroborate these findings.

  3. Studies on the activity of different radionuclides as well as different routes of administration will deliver further insights guiding the development of future compounds.

Meningiomas are the most common intracranial neoplasms.1 Deriving from arachnoidal cap cells, the vast majority of meningiomas exhibit benign biological behavior, although some tumors show atypical features, invasive growth patterns and may even metastasize. This highly variable biological behavior is reflected in tumor grading according to the WHO Classification of Central Nervous System Tumors (current version 2021),2 classifying meningiomas into CNS WHO grades 1 (> 80% of all tumors), 2 (atypical, ~5%–20%) and 3 (anaplastic, ~2%).3,4 Whereas tumor grading was historically based on histological features alone, molecular factors are also increasingly considered. For instance, meningeal tumors with CDKN2A/B homozygous deletion are automatically defined as CNS WHO grade 3, and other molecular aberrations with prognostic impact have been described that may be included in future classification frameworks.5 Moreover, DNA methylation and gene expression profiling have resulted in an even more refined prognostic stratification.6,7

According to current guidelines, meningiomas can be cured by resection, and in asymptomatic patients even a watch-and-wait approach is justifiable.8 In cases where a gross total resection cannot be achieved and/or in higher-grade lesions, adjuvant radiotherapy or stereotactic radiosurgery may be necessary. Systemic treatment options may be used after exhaustion of local therapies, but are considered experimental as the underlying evidence is overall weak.9 New therapeutic options for salvage treatment are therefore urgently needed. Patient groups that could especially benefit from effective systemic therapy include those with tumors in locations that limit complete surgical resection (eg, skull base), patients with atypical and anaplastic meningioma due to higher recurrence rates, those with substantial symptomatic burden, and even rare cases of extracranial metastasis resulting in poor prognosis.

The concept of “theranostics” combines a “therapy” that is targeted at a “diagnostic” biomarker assessed by molecular imaging modalities. Initially, this approach was used in imaging studies and the therapy of both benign and well-differentiated malignant diseases of the thyroid gland using radioactive [131I]-iodine.10 More recently, additional compounds including targeted molecules linked to radionuclides have been developed and evaluated for clinical use in prostate cancer and neuroendocrine tumors (NETs), and further trials in other malignant diseases are ongoing.11 In this review, we aim to explore and discuss available data and future outlooks on “theranostics” approaches and radionuclide therapies in meningioma.

Molecular Imaging of Meningiomas

One advantage of “theranostics” treatment strategies is the availability of noninvasive predictive biomarkers based on advanced imaging modalities. In standard imaging procedures such as computed tomography and magnetic resonance imaging (MRI), meningiomas classically present as contrast-enhancing extra-axial masses with a characteristic “dural tail.”12 Generally, these features allow for delineation of meningeal neoplasms with high certainty. However, challenging situations such as anatomical location in the skull base, atypical morphology such as “en plaque”-like growth patterns, distinction of vital tumor tissue from post-therapeutic changes in suspected recurrence, detection of residual tumor after surgery, and response assessment after radiotherapy or systemic treatment complicate the interpretation of meningioma imaging.

To overcome these challenges, molecular imaging techniques relying on the metabolic activity of neoplastic cells are increasingly considered in clinical routine. This includes [18F]fluoro-desoxy-glucose ([18F]FDG) positron emission tomography (PET) for the visualization of various extracranial tumors as well as the use of radiolabeled amino acids such as [18F]fluoroethyltyrosine (FET) or l-[methyl-11C]methionine in gliomas.13 In addition, radiolabeled ligands to specific receptors can be employed as PET tracers. Overall, 60%–100% of meningiomas express the somatostatin receptor type 2 (SSTR-2),14–19 and treatment with somatostatin analogs such as octreotide or pasireotide has been evaluated in small clinical trials.20–22 Tracers targeting SSTR are frequently used in nuclear imaging of meningeal neoplasms and generally show an improved tumor-to-background ratio compared to [18F]FDG and amino acid PET with almost no uptake in adjacent non-tumoral tissue such as brain and bone.23,24 Here, 68Ga-labeled tracers based on chemical modifications of the somatostatin receptor agonist octreotide are most frequently used, although 64Cu-labeled compounds are also applied. These include the widely available tracers [68Ga]gallium-DOTA-Tyr3-octreotide ([68Ga]Ga-DOTATOC), [68Ga]gallium-DOTA-Tyr3-octreotate ([68Ga]Ga-DOTATATE), and [68Ga]gallium-DOTA-1-NaI(3)-octreotide ([68Ga]Ga-DOTANOC).

Specific guidelines for SSTR-targeted PET imaging of meningiomas have jointly been elaborated by the European Association of Nuclear Medicine, the European Association of Neuro-Oncology, the Response Assessment in Neuro-Oncology working group (RANO), and the Society of Nuclear Medicine & Molecular Imaging (SNMMI).25 Comparative studies between used tracers are only available for NETs, where more lesions could be detected using [68Ga]Ga-DOTATOC and [68Ga]Ga-DOTANOC compared to [68Ga]Ga-DOTATATE.26,27 Conversely, a study in meningioma xenografts in mice showed higher uptake with [68Ga]Ga-DOTATATE compared to [68Ga]Ga-DOTATOC and [68Ga]Ga-DOTANOC.28 However, the clinical relevance of these subtle differences is limited given the already exceptional tumor-to-background ratio in meningiomas. Further tracers such as [18F]F-SiTATE based on a silicon fluoride acceptor (SiFA) are currently under investigation and may yield higher resolution and logistic advantages, given the longer half-life of 18F compared to 68Ga which results in lower radiation exposure and the possibility to centralize the production of larger lots of tracers.29

Prior to the advent of somatostatin receptor PET imaging, single photon emission computed tomography (SPECT) employing SSTR-2-targeted tracers such as [111In]In-pentetreotide or [99Tc]Tc -EDDA/HYNIC-TOC was widely used,30,31 but use today is mostly limited to circumstances where PET scanners are not available. Indeed, advantages of PET over SPECT imaging include higher spatial resolution, improved sensitivity, lower radiation dose to the patient, shorter exam duration, and greater capacity for quantitative information.32

For clinical applications, PET imaging of meningiomas is recommended in situations where the extent of the tumor or the distinction of recurrence from treatment-related changes is unclear,8 and SSTR-directed PET may be particularly helpful in the presurgical planning of meningiomas with intraosseus spread.33 In addition, postoperative PET might support the estimation of the extent of resection, as detection rates were improved compared to postoperative MRI.34,35

Evidence for Radionuclide Treatment Efficacy in Extracranial Solid Tumors

In the 1930s, increasing knowledge of the effects of radioactivity on human tissue and iodine physiology of the thyroid gland provided the basis for radioiodine treatment in thyroid disorders.36 Indeed, 131I remains a mainstay in the treatment of autoimmune hyperthyroidism, toxic (multi-)nodular goiter, and differentiated thyroid cancer.37,38 Radioiodine treatment in thyroid disorders represented a blueprint for future “theranostics” approaches, combining the therapeutic scope of 131I treatment with the diagnostic value of pre- and post-therapeutic 131I scans.

In 2022, the US Food and Drug Administration and European Medicines Agency approved the use of [177Lu]Lu-PSMA-617, a 177Lu-labeled ligand of the prostate-specific membrane antigen (PSMA), for use in refractory, metastatic castration-resistant prostate cancer (mCRPC) after failure of anti-hormonal treatment and chemotherapy. The approval was based on the phase 3 VISION trial showing a significant progression-free (PFS) and overall survival (OS) benefit.39 More recent data confirmed the activity of [177Lu]Lu-PSMA-617 in taxane-naïve patients, showing a superiority in radiographic PFS compared to a change of androgen receptor pathway inhibitor as therapeutic strategy.40 The harnessed target PSMA is a transmembrane protein that is abundantly and almost exclusively expressed on both primary and metastatic prostate cancer cells, and predominantly in more aggressive tumors.41,42 This is underlined by the exceptional tumor-to-background ratio and high sensitivity of PSMA-PET, resulting in improved accuracy compared to conventional imaging by computed tomography and bone scan.43

Neuroendocrine tumors (NETs), especially those of gastroenteropancreatic origin, show strong expression of members of the SSTR family.44–46 Current guidelines recommend peptide receptor radionuclide therapy with [177Lu]Lu-DOTATATE based on the results of the NETTER-1 trial showing PFS superiority of [177Lu]Lu-DOTATATE compared to high-dose octreotide in patients with well-differentiated, SSTR-expressing midgut NETs after progression on monotherapy with long-acting octreotide.47 In this trial, [177Lu]Lu-DOTATATE treatment was well tolerated, and only 6% of patients treated with [177Lu]Lu-DOTATATE experienced serious treatment-related adverse events (grade ≥ 3) which mainly included hematological side effects and renal function impairment. Also in pancreatic NETs, [177Lu]Lu-DOTATATE was associated with higher 12-month PFS rates compared to sunitinib in the OCLURANDOM trial.48 Moving to earlier treatment lines, results of NETTER-2 revealed improved PFS of [177Lu]Lu-DOTATATE combined with low-dose octreotide compared to high-dose octreotide alone in newly diagnosed, SSTR-expressing, advanced grades 2 and 3 gastroenteropancreatic NETs.49 Of note, these are the first results in first-line treatment of [177Lu]Lu-DOTATATE in any solid tumor, highlighting the potential of this therapeutic strategy in earlier treatment lines.

Early-Phase Data on Radionuclide Treatments in Brain Tumors

Overall, treatment responses for CNS tumors are highly variable for targeted agents, probably due to intratumoral heterogeneity in target expression and the influence of the blood-brain/blood-tumor barrier (BBB/BTB). This also pertains to radionuclide treatments, especially if agents are injected intravenously.50 Similarly, antibody-drug conjugates, linking a cytotoxic drug to a tumor-specific antibody, show highly heterogeneous response rates across distinct entities of primary and secondary brain tumors, whereas the underlying biological mechanisms remain largely unclear.51 Here, the so-called “bystander” effect might considerably affect intracranial efficacy. Following binding of the antibody-drug conjugate to the target on the cell surface and endocytosis, the cytotoxic payload is released by proteolytic cleavage or due to the acidic milieu in cellular lysosomes. Depending on the lipophilicity, the payload can diffuse to adjacent cells and exert antineoplastic activity on tumor cells with lower target expression. Similarly, radionuclides enable neighboring cells to be targeted in a certain range depending on the emitter used, often referred to as “crossfire” effect (Figure 1).

Figure 1.

Figure 1.

Overall concept of radioligand therapies in meningioma. DOTATATE, DOTA-Tyr3-octreotate; DOTATOC, DOTA-Tyr3-octreotide; SSTR-2, somatostatin receptor type 2. Figure created using BioRender.com.

Evidence for radionuclide treatments in brain tumors mainly stems from preclinical data and small case series. In glioma, targets such as epidermal growth factor receptor, l-type amino transporter 1 (LAT-1) neural cell adhesion molecule, glioma chloride channels, histone H1, neurokinin type 1 receptor, carbonic anhydrase XII, gastrin-releasing peptide receptor as well as the extracellular matrix protein tenascin-C have been investigated in small clinical trials as reviewed extensively elsewhere, with overall conflicting results.52–55 In addition, small case series also suggest therapeutic activity in (rarely occurring) brain metastases of prostate cancer treated with [177Lu]Lu-PSMA-617.56

Meningioma

Evidence for the use of radionuclides in meningeal tumors is mainly based on small case series and early-phase prospective trials (Table 1). In most clinical trials of radioligand treatments in meningioma, patients were treated with [177Lu]Lu-DOTATATE as approved in NETs. In addition, other 177Lu-linked SSTR-2 ligands such as DOTATOC or 90Y-based radioligands were evaluated. Likewise, anecdotal data from multiple case reports are published where patients had been experimentally treated with SSTR-targeting approaches.57–62

Table 1.

Clinical Data on Radionuclide Treatments in Meningioma

Compound Type of study Patient population Number of included patients with meningioma Outcome data (meningioma patients only) Ref.
[177Lu]Lu-DOTATOC Case series Patients with paraganglioma, meningioma, small-cell lung cancer, and melanoma with positive uptake in [111In]-indium-pentetreotide scan after exhaustion of standard therapies n = 5
-WHO grade III: 3
-Grade unknown: 2
(Other entities: 17)
PD in 3/5 (60%) patients, SD in 2/5 (40%) patients (according to routine CT/MRI) van Essen et al. 200663
[90Y]Y-DOTATOC Prospective (no information on phase) Patients with meningioma and positive SSTR-2 expression according to scintigraphy n = 29
-WHO grade I: 14
-WHO grade II: 9
-WHO grade III: 6
SD in 19/29 (66%) patients, PD in 10/29 (34%).
Median time to progression (TTP): 61 months in WHO grade I, 13 months in WHO grade II-III (according to routine MRI)
Median OS: 69 months in WHO grade I, 30.5 months in WHO grade II-III
Bartolomei et al. 200964
[177Lu]Lu-DOTATATE Phase I-II Patients with unresectable or metastatic SSTR-2-positive tumors (including NETs, paragangliomas, and meningiomas) by [111In]In-pentetreotide scintigraphy n = 51
-Grade unknown: 1
(Other entities: 50)
No meningioma-specific outcome data provided Bodei et al. 201165
[177Lu]Lu-DOTATOC or [177Lu]Lu-DOTATATE followed by external beam radiotherapy Prospective pilot trial (no information on phase) Patients with unresectable advanced primary or recurrent meningioma n = 10
-WHO grade I: 7
-WHO grade II: 2
-Grade unknown: 1
CR in 1/10 (10%) patients, PR in 1/10 (10%) patients, SD in 8/10 (80%) patients according to routine MRI
PET-positive volume reduction to median of 81% of pretreatment size; SUVmax increase by a median of 37% (7 evaluable patients)
Long-term outcome data: CR in 1/10 (10%), SD in 6/10 (60%), PD in 3/10 (30%); median PFS 91.1 months; median OS 105 months.
Kreissl et al. 201266
(long-term outcome data in Hartrampf et al. 202067)
[177Lu]Lu-DOTATOC or [177Lu]Lu-DOTATATE Prospective pilot trial (no information on phase) Patients with primary meningioma of WHO grade 2 and 3 or recurrent meningioma of any grade or meningioma ≥ 5 cm diameter with adjacent critical structures and positive SSTR-2 PET n = 11 (no information on individual WHO grades) Correlations between (1) maximum voxel dose and activity retention 1 hour after administration of [177Lu]Lu-DOTATOC or [177Lu]Lu-DOTATATE and (2) SUVmax in pre-therapeutic PET; no information on treatment response Hänscheid et al. 201268
[111In]In-pentetreotide (in some patients combined with [90Y]Y-DOTATOC and [90Y]Y-DOTATATE) Retrospective case series Patients with meningioma or meningiomatosis n = 8
-WHO grade I: 5
-WHO grade II: 3
PR in 2/8 (25%) patients, SD in 5/8 (62.5% patients), PD in 1/8 (12.5%) case Minutoli et al. 201469
[90Y]Y-DOTATOC Phase II Patients with SSTR-2a-positive (by [111In]In-DOTATOC or [111In]In-pentetreotide scintigraphy), recurrent or progressive meningiomas in functionally critical areas or not amenable to surgery due to unfavorable medical risk profile or refusal of surgery n = 15
-WHO grade I: 9
-WHO grade II: 2
-WHO grade III: 1
-Unknown: 3
SD in 13/15 (87%), PD in 2/15 (13%) of patients
Median PFS: 24 months
Gerster-Gilliéron et al. 201570
[177Lu]Lu-DOTATOC or [90Y]Y-DOTATOC Phase II Patients with progressing meningioma and uptake on SSTR-2 scintigraphy n = 37
-WHO grade I: 5
-WHO grade II: 6
-WHO grade III: 3
-Grade unknown: 23
SD in 23/34 (68%), PD in 11/34 (32%)
Mean OS: 8.6 years
Marincek et al. 201571
[177Lu]Lu-DOTATATE or [90Y]Y-DOTATOC, or both Retrospective case series Patients with progressing meningioma and positive SSTR-2 PET or scintigraphy n = 20
-WHO grade I: 5
-WHO grade II: 7
-WHO grade III: 8
(WHO grades for surgery at recurrence)
WHO grade I:
-SD in 5/5 (100%)
-Median PFS: 32.2 months
-Median OS: not reached
WHO grade II:
-SD in 4/7 (57%), PD in 3/7 (43%)
-Median PFS: 7.6 months
-Median OS: not reached
WHO grade III:
-SD in 1/8 (12.5%), PD in 7/8 (87.5%)
-Median PFS: 2.1 months
-Median OS: 17.2 months
Seystahl et al. 201672
[177Lu]Lu-DOTATATE Retrospective case series Patients with advanced/metastatic NET and incidentally diagnosed [68Ga]Ga-DOTATATE-positive lesions with high suspicion of meningioma based on MRI n = 5 (WHO grades unknown) CR in 2/5 (40%), PR in 1/5 (20%), PD in 2/5 (40%).
Median PFS (meningioma-related): 26 months.
Parghane et al. 201973
[177Lu]Lu-DOTATATE Retrospective case series Patients with progressive intracranial meningioma n = 7
-WHO grade I: 2
-WHO grade II: 5
SD in 2/4 (50%) evaluable patients (according to PET)
PFS at 6 months: 42.9%
Müther et al. 202074
[177Lu]Lu-DOTATOC Retrospective case series Patients with confirmed neurofibromatosis type 2 and multiple partially pretreated meningiomas and SSTR-2 expression by [68Ga]Ga-DOTATOC n = 11
-WHO grade I: 4
-WHO grade II: 6
-WHO grade III: 1
SD in 6/11 (55%), PD in 5/11 (45%)
Median PFS: 4 months
Median OS: 50 months
Kertels et al. 202175
[177Lu]Lu-DOTATATE Retrospective case series Patients with progressive meningioma and no further surgical or radiotherapeutic treatment options and SSTR-positive lesions in PET n = 8
-CNS WHO grade 2: 8
SD in 7/8 (87.5%), PD in 1/8 (12.5%) - according to RANO criteria on MRI
PFS at 6 months: 85.7%
Salgues et al. 202276
[177Lu]Lu-DOTATATE Retrospective case series Patients with progressive meningioma not amenable to further surgery or radiotherapy n = 15
-WHO grade I: 3
-WHO grade II: 5
-WHO grade III: 6
-Grade unknown: 1
SD in 6/15 (40%), PD in 8/15 (53%), one death during treatment—according to RANO criteria on MRI
Median PFS: 7.8 months
Minczeles et al. 202377
[177Lu]Lu-DOTATATE Phase II Patients with progressive meningioma of all grades n = 14 on interim report (32 planned)
-WHO grade 1: 2
-WHO grade 2: 11
-WHO grade 3: 1
SD in 9/14 (64%), PD = 4/14 (29%), lost-to-follow-up in 1 patient
PFS at 6 months: 50%
Median PFS = 8.2 months
Median OS = 21.9 months
>25% SUV reduction in [68Ga]Ga-DOTATATE in 5 lesions
Kurz et al. 202478

CR, complete remission; CT, computed tomography; DOTATATE, DOTA-Tyr3-octreotate; DOTATOC, DOTA-Tyr3-octreotide; MRI, magnetic resonance imaging; NET, neuroendocrine tumor; OS, overall survival; PD, progressive disease; PET, positron emission tomography; PFS, progression-free survival; PR, partial remission; RANO, response assessment in neuro-oncology; SD, stable disease; SSTR-2, somatostatin receptor type 2; SUVmax, maximum standardized uptake value; TTP, time to progression; WHO, World Health Organization.

In an early-phase prospective trial in 29 patients with meningioma of all grades, Bartolomei et al. observed that treatment with [90Y]Y-DOTATOC resulted in stable disease (SD) in 66% of included patients, while progressive disease was seen in 34%. Of note, [90Y]Y-DOTATOC treatment did not result in shrinkage of lesions, and stabilization of lesions was more frequently observed in WHO grade 1 meningioma than in higher-grade lesions.64 Similar results were seen in other studies, and complete (CR) or partial responses were overall rarely reported regardless of the compound used.66,69,73 In line with these findings, the largest trial to date in meningiomas of all grades confirmed uptake in SSTR-2 scintigraphy (n = 37) and showed SD in 68% and progressive disease in 32% of patients treated with [177Lu]Lu-DOTATOC or [90Y]Y-DOTATOC.71 A retrospective case series of patients treated with [177Lu]Lu-DOTATATE and/or [90Y]Y-DOTATOC showed SD in 100% of WHO grade 1 meningioma, whereas WHO grade 2 and 3 lesions had SD in 57% and 12.5%, respectively.72 These results were corroborated by 3D volumetry. Analysis of maximum and mean standardized uptake values (SUVmax, SUVmean) in SSTR PET, as well as immunohistochemical SSTR-2 staining, showed better responses in meningiomas with higher SSTR-2 expression, underscoring the predictive value of target expression assessed by both molecular imaging and in tumor tissue. In another recent publication, ≥25% reduction in SUV was observed in 5/13 measurable lesions upon [177Lu]Lu-DOTATATE treatment, which correlated with disease control and therefore highlights the potential of response monitoring by molecular imaging.78

Data on the toxicity of [177Lu]Lu-DOTATATE treatment in meningioma are scarce.72,76 In one retrospective publication, lymphocytopenia was most frequently observed (70% of patients), where lymphocyte counts correlated with the number of previous systemic treatment lines. Other observed side effects included anemia, thrombocytopenia, increase of gamma-glutamyltransferase, fatigue, alopecia, pituitary insufficiency, and wound complications.72

Despite these studies, high-grade evidence for the use of radionuclide therapies in meningioma is lacking. Some studies included SSTR-positive tumors regardless of the underlying entity, including also NETs, paragangliomas, small-cell lung cancer, and melanoma, and meningioma-specific outcome measures were not reported.63,65 While some publications suggest that the antitumoral activity might be more pronounced in more benign and lower-grade tumors, data on WHO grades is frequently missing. Furthermore, tumor grading and classification have undergone considerable changes in the past decades, limiting the applicability of these results to the integrated molecular diagnostic framework currently in use.2 Additionally, response assessment was not uniformly conducted across published data, and older reports use arbitrary cutoffs to define treatment response or adapted MacDonald criteria for response assessment in malignant glioma. Only recently, standardized criteria for MRI-based response assessment in meningioma were proposed by the RANO working group,79 and currently, there is no generally accepted framework for the standardized assessment of SSTR-based PET imaging in meningioma. In addition, meningiomas frequently show slow growth rates, complicating response assessment given the short follow-up in many publications. Here, 3-dimensional volume growth rate (3DVGR) measurements might be helpful as observed in a recent post hoc analysis of EORTC-1320, a prospective trial in higher-grade meningioma.80 These were shown to correlate with 6-month PFS and may provide a more refined response classification compared to modified Macdonald criteria.

Future Outlooks

Choice of Radionuclide Emitter

To date, data on radionuclide treatment in meningioma are almost exclusively based on radioligands with the β- emitters 177Lu or 90Y. However, their linear energy transfer is relatively low compared to ɑ particle emitters such as 225Ac or 223Ra (Table 2). In specific, β- particles predominantly act by forming reactive oxygen species (ROS) which lead to single-strand breaks, whereas ɑ particles elicit double-strand breaks by direct interaction with DNA, causing higher lethality due to limited DNA repair mechanisms. Indeed, there are no known biological intrinsic resistance mechanisms for ɑ particles so far.81 In addition, the limited range and therefore reduced crossfire effect allows sparing of adjacent tissues while specifically targeting high radiation doses to the tumor, which is of particular significance in brain tumors. This is also underscored by the importance of target delineation in external beam radiotherapy of meningiomas as outlined in recent consensus guidelines.82 On the other hand, the short range of ɑ particles might confer less activity in heterogeneous tumors where some cells show lower target expression. Mixed ɑ/β treatments might therefore combine the higher linear energy transfer of ɑ particles with the increased crossfire effect of β particles, but further studies on efficacy and safety are needed.

Table 2.

Overview of Particle-Specific Properties of Therapeutic Radionuclides

ɑ particles β particles Auger electrons
Isotopes [225Ac], [223Ra], [212Pb] [131I], [90Y], [177Lu] [125I], [111In]
Range 10–100 µm 1–10 mm ~0.1 µm
Linear energy transfer 50–300 keV/µm 0.2 keV/µm 4-26 keV/µm
DNA damage DNA double-strand breaks by direct interaction with DNA DNA single-strand breaks by reactive oxygen species Direct and indirect damage to DNA (single-/double-strand breaks), reactive oxygen species, damage of phospholipid bilayer in membranes

Use of the ɑ emitter 223Ra is currently approved in bone-metastatic, mCRPC based on the phase 3 ALSYMPCA trial.83 Similar to the application of iodine isotopes in thyroid disorders, 223Ra is delivered to the target tissue by mimicking calcium atoms which are absorbed in metastatic bone tissue remodeling. Recently, results of a large retrospective cohort of mCRPC patients treated with the ɑ emitter-based radioligand [225Ac]Ac-PSMA were published, showing antitumoral activity at an acceptable side effect profile.84 In NETs previously treated with 177Lu-based agents, [225Ac]Ac-DOTATATE showed promising results in early pilot studies,85,86 but data in meningioma are lacking so far.

Auger electrons have an even lower range than ɑ particles, thereby reducing toxicity to neighboring tissues. Physically, radioactive decay of nuclides leads to transitions in the electron shell of atoms, releasing energy in the form of X-rays or Auger electrons. These cause direct damage to DNA by eliciting double-strand breaks, but also indirect cell damage through the formation of reactive oxygen species and the interaction with the phospholipid bilayer of the cell membrane have been reported.87 While research efforts in the past years mainly focused on ɑ therapies, the clinical activity and safety of Auger electron therapeutics are not well established. Indeed, most data on Auger electron treatment in cancer is based on [111In]In-pentetreotide treatment in NETs, with overall promising treatment responses and mainly transient, mild hematotoxicity.88–90 In meningioma, only one small case series of 8 patients receiving [111In]In-pentetreotide with or without [90Y]Y-DOTATOC has been published so far, and larger prospective trials are needed to substantiate early signs of antitumoral activity.69

More recently, results of a dosimetric study with 64Cu/67Cu-linked to octreotate by a sarcophagine MeCOSar chelator ([64Cu/67Cu]Cu-SARTATE) in patients with unresectable meningiomas have been reported. The β-emitting isotope pair 64Cu/67Cu has reduced half-lives compared to 177Lu, allowing for more frequent administration and easier logistics in terms of radiation protection regulations. Moreover, the positron-emitting 64Cu used for diagnostic imaging may more accurately predict the biodistribution of the therapeutic 67Cu than in theranostic pairs based on different elements such as 68Ga and 90Y or 117Lu. While tolerability was acceptable, efficacy remains to be demonstrated.91

Improving Dosimetry

Classical pharmacotherapy follows either fixed-dose regimens or is based on body weight or body surface according to pharmacokinetic data. In radionuclide treatment, absorbed doses can vary considerably between different individuals, affecting both therapeutic responses in the target tissue as well as potential side effects in organs at risk, mainly kidneys and bone marrow. While effective and maximum tolerated dose thresholds are standardized in conventional radiotherapy, dose–response, and dose-toxicity relationships are less clearly defined in radionuclide therapies.92 Indeed, dose thresholds are frequently extrapolated from external beam radiotherapy, and fixed-dose regimens based on conventional dose escalation trials are currently in use for all approved treatments including [177Lu]Lu-DOTATATE, [177Lu]Lu-PSMA, and 223Ra. For instance, the approved dosing regimen of [177Lu]Lu-DOTATATE in NETs includes 4 courses with 7400 MBq every 8 weeks. Other schedules are being explored, also in brain tumors such as glioblastoma where administration every 4 weeks is being evaluated in a dose-finding study of [177Lu]Lu-DOTATATE (NCT05109728).

Dosimetry enables the collection of further information on absorbed radiation doses in the target tissue and other organs that may be affected by adverse events. As a basic requirement, patient-averaged dosimetry in standardized nuclear medicine treatments is mandatory according to current regulations in the European Union.93 However, this does not extend to organ- or tissue-specific absorbed radiation doses which would open new avenues to introduce personalized dosing schemes to radionuclide treatments. In specific, future approaches could allow to improve on-target efficacy and spare off-target toxicity by systematically collecting data on tissue-specific absorbed radiation doses.

Radionuclides emitting ɣ rays such as 177Lu allow collection of planar as well as 3D SPECT images to monitor the distribution of radioactive activity. In contrast, 90Y does not emit ɣ radiation. While other radionuclides such as 111In may be employed as surrogate radionuclides for dosimetry, 111In dosimetry does not correlate well with the biological effects of 90Y-based treatment.94 In addition, dosimetry protocols are frequently not harmonized between institutions, challenging the overall acceptance in the nuclear medicine community.95 While results of clinical trials indicating a superiority of personalized radionuclide treatment are so far missing, early results in NETs are promising. Indeed, the cumulative maximum tumor dose could be increased by the factor 1.26 in a prospective trial, whereas the occurrence of side effects was similar to fixed-dose regimens.96

In meningioma, dosimetry information has rarely been reported in published data on radionuclide treatment. In the recently published retrospective analysis by Minczeles et al., absorbed doses were highly heterogeneous and ranged from 7 to 404 Gy in 14 target tumors of 8 patients.77 Another trial aimed to predict dosimetric data based on pre-therapeutic [68Ga]Ga-DOTATOC PET imaging in meningioma. Here, correlations between SUVmean-derived values in [68Ga]Ga-DOTATOC PET performed prior to [177Lu]Lu-DOTATATE and tumor-absorbed doses were observed97. However, the high heterogeneity of applied doses as well as the paucity of data underscore the urgent need for systematic collection of data on dosimetry in radionuclide treatment trials of brain tumors and meningiomas.

Route of Administration

Currently approved radionuclide treatments such as [177Lu]Lu-DOTATATE, [177Lu]Lu-PSMA, and 223Ra are generally administered intravenously. In general, systemic administration is complicated by the BBB/BTB, although meningiomas are usually located outside these barriers. In intraaxial tumors, multiple strategies to disrupt and bypass the BBB/BTB have been described, including cellular, molecular, and physical/chemical approaches as extensively reviewed elsewhere.98

For radionuclide treatments, studies on intraventricular and intracavitary applications have been performed. For instance, intraventricular application of [131I]I-omburtamab, an antibody targeting the tumor antigen B7-H3 conjugated to 131I, has been evaluated in recurrent medulloblastoma and ependymoma in a phase 1 trial, with an acceptable side effect profile and signs of antitumoral activity as compared to historical data.99 In glioma, intracavitary administration of a conjugate consisting of 131I and the monoclonal anti-tenascin antibody 81C6 ([131I]I-81C6) has shown an acceptable side effect profile and compared favorably with historical controls, but further development was discontinued.100 Similar results were seen with [211At]At-81C6.101 Moreover, radionuclide treatment with a nano-liposomal formulation of 186Rh has been explored using pressure gradients through intratumoral and intracavitary application in glioma (also known as convection-enhanced delivery). While overall results were promising, the absorbed doses were highly heterogeneous and future studies are needed to substantiate an improvement in treatment outcomes compared to external beam radiotherapy.102

In meningioma, intraarterial delivery may represent a promising concept to maximize intratumoral doses and minimize systemic toxicity. The feasibility of this approach has initially been demonstrated in a diagnostic [68Ga]Ga-DOTATATE PET study, where intraarterial application of the tracer increased SUVmean values by 2.7-fold compared to systemic intravenous administration.103 One case report of [177Lu]Lu-DOTATATE treatment in meningioma showed improved tumor uptake compared to intravenous radionuclide treatment.60 Another investigation in 4 patients with treatment-refractory meningiomas confirmed these findings, and no significant adverse effects were observed.104 Further clinical research is warranted, especially as intraarterial administration showed conflicting results in other tumors such as NETs metastatic to the liver.105,106

Optimal Clinical Trial Design

Clinical trials in rare and heterogeneous entities such as treatment-refractory meningioma remain challenging. Similar to systemic treatment in meningioma, the evidence for radionuclide therapy is based on retrospective data or small prospective pilot trials, and most included patients are heavily pretreated with no further treatment options.9 Indeed, controlled trials comparing radioligand therapy to other therapeutic approaches in a randomized fashion are lacking, but ultimately needed to define efficacy and safety. This also includes well-designed clinical trials of radionuclide treatment strategies in earlier treatment lines. Moreover, response assessment in available data varies considerably, challenging the interpretation of available outcome data and the use of external controls which is increasingly considered in rare oncological entities where the implementation of randomized controlled trials is not always feasible.107

The European Organization for Research and Treatment of Cancer—Brain Tumor Group (EORTC-BTG) has recently formulated considerations for clinical trial design and conduct in the field of radionuclide treatment.108 These include the endorsement for controlled trials with well-defined endpoints such as OS and PFS based on consensus recommendations for response assessment such as those of the RANO consortium.79 Moreover, the standardized collection of patient-reported outcomes is crucial as intracranial tumors including meningiomas considerably impact physical, neurocognitive, emotional, and social functioning.109,110 From a technical point of view, the systematic acquisition of dosimetric data would allow for improvement in dosing schedules, including optimized length of cycles as well as intratumoral absorbed doses while minimizing systemic toxicity. In addition, harmonization of regulatory frameworks and institutional protocols is a prime prerequisite for the conduct of clinical trials and implementation of standardized treatment schemes in clinical routine.25

In addition, translational research efforts within clinical trials are needed to detect and validate predictive biomarkers. Among these lines, recent data have shown that targeted gene expression profiling allows to identify cases who benefit from postoperative radiotherapy.7 Involved genes include members of pathways concerned with cell cycle and mitotic stability, suggesting that these alterations might also confer higher sensitivity to other cytotoxic treatments such as radionuclide therapies. However, further validation in prospective trials is necessary.

Ongoing Clinical Trial Initiatives

Currently, ongoing clinical trials are given in Table 3. The vast majority of trials aim to investigate [177Lu]Lu-DOTATATE and [177Lu]Lu-DOTATOC in meningiomas of different grades. ELUMEN (NCT06126588) aims to evaluate the combination of [177Lu]Lu-DOTATATE with everolimus based on promising data on systemic treatment of combined mammalian target of rapamycin (mTOR) and SSTR-2 inhibition.111 The crossover phase 0/I/II trial PROMENADE (NCT04997317) aims to compare [177Lu]Lu-DOTATOC with [177Lu]Lu-satoreotide in a 2-step trial design. Indeed, [177Lu]Lu-satoreotide has shown higher intratumoral absorbed doses compared to [177Lu]Lu-DOTATATE, probably given the improved binding of the SSTR antagonist satoreotide compared to the agonist DOTATATE.112 First results of the phase 0 arm of PROMENADE have been published recently,113 with higher tumor-to-bone marrow and tumor-to-kidney ratios in patients receiving [177Lu]Lu-satoreotide compared to [177Lu]Lu-DOTATATE. In addition, early signs of efficacy have been observed as the disease control rate was 83%. Further investigation in the phase I/II arm is ongoing.

Table 3.

Overview of Ongoing Clinical Trials on Radionuclide Treatments in Meningioma

Clinical trial identification/trial name Phase Study design Compound Main inclusion criterion Status (effective January 29, 2024)
NCT06126588 (ELUMEN) Phase II Single-arm [177Lu]Lu−DOTATATE + everolimus Meningioma WHO grades 2 and 3 (histologically verified) with progression and not amenable to surgery or RT and positive in [68Ga]Ga-DOTATOC PET Not yet recruiting
NCT04997317 (PROMENADE) Phase 0/I/II Two-step, cross-over open-label phase 0 followed by single-arm open-label phase I/II [177Lu]Lu−DOTATOC, [177Lu]Lu−DOTA-JR11 ([177Lu]Lu−satoreotide) Recurrent or progressive meningioma (either histologically confirmed or high suspicion based on MRI and SSTR imaging) Recruiting
NCT03971461 Phase II Single-arm [177Lu]Lu−DOTATATE Meningioma WHO grade 1 (progressing after surgery and RT or progressive residual tumor after surgery not amenable to RT) or meningioma WHO grades 2 and 3 (progressing after surgery and RT or residual measurable disease after surgery) Recruiting
NCT04082520 Phase II Single-arm [177Lu]Lu−DOTATATE Progressive meningioma with measurable disease after previous surgery and RT (not amenable to further RT) Recruiting
NCT03273712 Phase II Single-arm [90Y]Y−DOTATOC Pediatric and adult patients with SSTR-2-positive tumors according to [68Ga]Ga-DOTATOC/DOTATE PET (including NET, meningioma, neuroblastoma, and medulloblastoma) not amenable to standard treatment after failure of first-line treatment Completed (no results published)
NCT05278208 Phase I/II Two-step, single-arm phase I/II [177Lu]Lu−DOTATATE Pediatric and young adult patients with recurrent/progressive high-grade CNS tumors and meningiomas expressing SSTR-2A and showing uptake in DOTATATE PET Recruiting

DOTATATE, DOTA-Tyr3-octreotate; DOTATOC, DOTA-Tyr3-octreotide; MRI, magnetic resonance imaging; NET, neuroendocrine tumor; PET, positron emission tomography; RT, radiotherapy; SSTR-2, somatostatin receptor type 2; WHO, World Health Organization.

Within EORTC-BTG-2334 (LUMEN-1), intravenous [177Lu]Lu-DOTATATE will be assessed in recurrent or progressing, SSTR-2-expressing meningiomas of all grades after surgery and radiotherapy. For the first time in meningioma, radionuclide treatment will be compared to local standard of care in a randomized fashion with PFS as primary endpoint, and OS, tolerability, and quality of life as secondary endpoints. Within this trial, translational and further patient-reported outcome data will be systematically collected to define efficacy, safety and potential predictive markers impacting therapeutic responses. Of special interest is the planned collection of dosimetry data as this will allow us to study the inter-individual variability of absorbed doses and their correlation with treatment response.

Conclusion

In conclusion, radionuclide therapies represent a promising treatment approach in meningioma. Data from small clinical trials and retrospective case series demonstrate safety and suggest therapeutic activity in progressing tumors after failure of local treatment, but evidence is still limited. In addition, the utility of these agents earlier in the treatment course of meningioma remains to be established. Further innovative strategies in drug design, such as changes in the employed radionuclide or the used ligand, are being explored in ongoing studies. Alternative dosing regimens (such as shorter application intervals) and combination therapies, for instance with radiosensitizers or immunotherapies, may be explored in order to induce synergistic efficacy. Furthermore, alternative administration routes including intraarterial administration as well as further collection of dosimetry data may lead to improved intratumoral doses and reduced systemic exposure, resulting in even higher therapeutic responses and decreased off-target toxicity. Overall, well-designed trials are urgently needed to define the value of radioligand therapies and related novel approaches in meningioma.

Acknowledgments

This Research Project was supported by ESMO (ESMO Translational Research Fellowship awarded to MJM). Any views, opinions, findings, conclusions, or recommendations expressed in this material are those solely of the author(s) and do not necessarily reflect those of ESMO. This project facilitates the distribution of scientific and educational content of Nuclear Medicine and Neuro-Oncology (NMN).

Contributor Information

Maximilian J Mair, Department of Nuclear Medicine, LMU Hospital, LMU Munich, Munich, Germany; Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.

Emeline Tabouret, Aix-Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, GlioME Team, plateforme PETRA, CHU Timone, Service de Neurooncologie, Marseille, France.

Derek R Johnson, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA.

Erik P Sulman, Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, NYU Langone, New York, New York, USA; Department of Radiation Oncology, New York University Grossman School of Medicine, New York, New York, USA.

Patrick Y Wen, Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA.

Matthias Preusser, Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.

Nathalie L Albert, Department of Nuclear Medicine, LMU Hospital, LMU Munich, Munich, Germany.

Funding

This Research Project was supported by the research budget of the Medical University of Vienna and a fellowship grant from the European Society of Medical Oncology (ESMO Translational Research Fellowship awarded to MJM). Any views, opinions, findings, conclusions, or recommendations expressed in this material are those solely of the author(s) and do not necessarily reflect those of ESMO.

Supplement sponsorship

This article appears as part of the supplement “Theranostics in CNS Tumors” sponsored by Novartis/Advanced Accelerator Applications. The sponsor had no influence on the article content.

Conflicts of interest statement

Maximilian J. Mair has received research funding from Bristol-Myers Squibb and travel support from Pierre Fabre. Emeline Tabouret has received research funding from Léo Pharma and honoraria for consultation and advisory board participation from Servier, Gliocure and Novocure.

Derek R. Johnson has received honoraria for consultation or advisory board participation from Novartis and Telix Pharmaceuticals. Erik Sulman has received honoraria for speaking or advisory board participation from Telix Pharmaceuticals and BrainLab; and research funding from Novartis/Advanced Accelerator Applications and Novocure. Patrick Y Wen has received research support from Astra Zeneca, Black Diamond, Bristol Meyers Squibb, Chimerix, Eli Lilly, Erasca, Global Coalition For Adaptive Research, Kazia, MediciNova, Merck, Novartis, Quadriga, Servier, VBI Vaccines and honoraria for consultation or advisory board participation from Anheart, Astra Zeneca, Black Diamond, Celularity, Chimerix, Day One Bio, Genenta, Glaxo Smith Kline, Kintara, Merck, Mundipharma, Novartis, Novocure, Prelude Therapeutics, Sagimet, Sapience, Servier, Symbio, Tango, Telix, and VBI Vaccines. Matthias Preusser has received honoraria for lectures, consultation or advisory board participation from the following for-profit companies: Bayer, Bristol-Myers Squibb, Novartis, Gerson Lehrman Group (GLG), CMC Contrast, GlaxoSmithKline, Mundipharma, Roche, BMJ Journals, MedMedia, Astra Zeneca, AbbVie, Lilly, Medahead, Daiichi Sankyo, Sanofi, Merck Sharp & Dome, Tocagen, Adastra, Gan & Lee Pharmaceuticals, Janssen, Servier, Miltenyi, Böhringer-Ingelheim, Telix, Medscape. Nathalie L. Albert has received honoraria for consultation or advisory board participation from Novartis, Advanced Accelerator Applications, Servier and Telix Pharmaceuticals and research funding from Novocure.

Authorship statement

Conceptualization, project oversight and coordination: M.J.M., M.P., and N.L.A.. Literature search, writing of first draft: M.J.M. and N.L.A.. Manuscript writing and editing: M.J.M., E.T., D.R.J., E.P.S., P.Y.W., M.P., and N.L.A.. All authors read and approved the final manuscript.

References

  • 1. Ostrom QT, Price M, Neff C, et al. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2016—2020. Neuro Oncol. 2023;25(12 suppl 2):iv1–iv99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Louis DN, Perry A, Wesseling P, et al. The 2021 WHO classification of tumors of the central nervous system: A summary. Neuro Oncol. 2021;23(8):1231–1251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Lin D, Lin J, Deng X, et al. Trends in intracranial meningioma incidence in the United States, 2004-2015. Cancer Med. 2019;8(14):6458–6467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Holleczek B, Zampella D, Urbschat S, et al. Incidence, mortality and outcome of meningiomas: A population-based study from Germany. Cancer Epidemiol. 2019;62:101562. [DOI] [PubMed] [Google Scholar]
  • 5. Maas SLN, Sievers P, Weber DC, et al. Independent prognostic impact of DNA methylation class and chromosome 1p loss in WHO grade 2 and 3 meningioma undergoing adjuvant high-dose radiotherapy: Comprehensive molecular analysis of EORTC 22042-26042. Acta Neuropathol. 2023;146(6):837–840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Sahm F, Schrimpf D, Stichel D, et al. DNA methylation-based classification and grading system for meningioma: A multicentre, retrospective analysis. Lancet Oncol. 2017;18(5):682–694. [DOI] [PubMed] [Google Scholar]
  • 7. Chen WC, Choudhury A, Youngblood MW, et al. Targeted gene expression profiling predicts meningioma outcomes and radiotherapy responses. Nat Med. 2023;29(12):3067–3076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Goldbrunner R, Stavrinou P, Jenkinson MD, et al. EANO guideline on the diagnosis and management of meningiomas. Neuro Oncol. 2021;23(11):1821–1834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Mair MJ, Berghoff AS, Brastianos PK, Preusser M.. Emerging systemic treatment options in meningioma. J Neurooncol. 2023;161(2):245–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Hertz B, Watabe T, Baum RP.. Celebrating 80 years anniversary of radioiodine for use in thyroid cancer and perspectives for theranostics. Ann Nucl Med. 2022;36(12):1007–1009. [DOI] [PubMed] [Google Scholar]
  • 11. Arnold C. Theranostics could be big business in precision oncology. Nat Med. 2022;28(4):606–608. [DOI] [PubMed] [Google Scholar]
  • 12. Starr CJ, Cha S.. Meningioma mimics: Five key imaging features to differentiate them from meningiomas. Clin Radiol. 2017;72(9):722–728. [DOI] [PubMed] [Google Scholar]
  • 13. Albert NL, Galldiks N, Ellingson BM, et al. PET-based response assessment criteria for diffuse gliomas (PET RANO 1.0): A report of the RANO group. Lancet Oncol. 2024;25(1):e29–e41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Schulz S, Pauli SU, Schulz S, et al. Immunohistochemical determination of five somatostatin receptors in meningioma reveals frequent overexpression of somatostatin receptor subtype sst2A. Clin Cancer Res. 2000;6(5):1865–1874. [PubMed] [Google Scholar]
  • 15. Barresi V, Alafaci C, Salpietro F, Tuccari G.. Sstr2A immunohistochemical expression in human meningiomas: Is there a correlation with the histological grade, proliferation or microvessel density? Oncol Rep. 2008;20(3):485–492. [PubMed] [Google Scholar]
  • 16. Durand A, Champier J, Jouvet A, et al. Expression of c-Myc, neurofibromatosis Type 2, somatostatin receptor 2 and erb-B2 in human meningiomas: relation to grades or histotypes. Clin Neuropathol. 2008;27(5):334–345. [DOI] [PubMed] [Google Scholar]
  • 17. Agaimy A, Buslei R, Coras R, Rubin BP, Mentzel T.. Comparative study of soft tissue perineurioma and meningioma using a five-marker immunohistochemical panel. Histopathology. 2014;65(1):60–70. [DOI] [PubMed] [Google Scholar]
  • 18. Dijkstra BM, Motekallemi A, den Dunnen WFA, et al. SSTR-2 as a potential tumour-specific marker for fluorescence-guided meningioma surgery. Acta Neurochir (Wien). 2018;160(8):1539–1546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Behling F, Fodi C, Skardelly M, et al. Differences in the expression of SSTR1-5 in meningiomas and its therapeutic potential. Neurosurg Rev. 2022;45(1):467–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Johnson DR, Kimmel DW, Burch PA, et al. Phase II study of subcutaneous octreotide in adults with recurrent or progressive meningioma and meningeal hemangiopericytoma. Neuro Oncol. 2011;13(5):530–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Norden AD, Ligon KL, Hammond SN, et al. Phase II study of monthly pasireotide LAR (SOM230C) for recurrent or progressive meningioma. Neurology. 2015;84(3):280–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Simó M, Argyriou AA, Macià M, et al. Recurrent high-grade meningioma: A phase II trial with somatostatin analogue therapy. Cancer Chemother Pharmacol. 2014;73(5):919–923. [DOI] [PubMed] [Google Scholar]
  • 23. Chung JK, Kim YK, Kim S, et al. Usefulness of 11C-methionine PET in the evaluation of brain lesions that are hypo- or isometabolic on 18F-FDG PET. Eur J Nucl Med Mol Imaging. 2002;29(2):176–182. [DOI] [PubMed] [Google Scholar]
  • 24. Arita H, Kinoshita M, Okita Y, et al. Clinical characteristics of meningiomas assessed by 11C-methionine and 18F-fluorodeoxyglucose positron-emission tomography. J Neurooncol. 2012;107(2):379–386. [DOI] [PubMed] [Google Scholar]
  • 25. Albert NL, Preusser M, Traub-Weidinger T, et al. Joint EANM/EANO/RANO/SNMMI practice guideline/procedure standards for diagnostics and therapy (theranostics) of meningiomas using radiolabeled somatostatin receptor ligands: version 1.0 [published online ahead of print June 20, 2024]. Eur J Nuclear Med Mol Imaging. doi: https://doi.org/ 10.1007/s00259-024-06783-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Poeppel TD, Binse I, Petersenn S, et al. 68Ga-DOTATOC versus 68Ga-DOTATATE PET/CT in functional imaging of neuroendocrine tumors. J Nucl Med. 2011;52(12):1864–1870. [DOI] [PubMed] [Google Scholar]
  • 27. Wild D, Bomanji JB, Benkert P, et al. Comparison of 68Ga-DOTANOC and 68Ga-DOTATATE PET/CT within patients with gastroenteropancreatic neuroendocrine tumors. J Nucl Med. 2013;54(3):364–372. [DOI] [PubMed] [Google Scholar]
  • 28. Soto-Montenegro ML, Peña-Zalbidea S, Mateos-Pérez JM, et al. Meningiomas: A comparative study of 68Ga-DOTATOC, 68Ga-DOTANOC and 68Ga-DOTATATE for molecular imaging in mice. PLoS One. 2014;9(11):e111624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Unterrainer M, Kunte SC, Unterrainer LM, et al. Next-generation PET/CT imaging in meningioma—first clinical experiences using the novel SSTR-targeting peptide [18F]SiTATE. Eur J Nucl Med Mol Imaging. 2023;50(11):3390–3399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Krenning EP, Kwekkeboom DJ, Bakker WH, et al. Somatostatin receptor scintigraphy with [111In-DTPA-D-Phe1]- and [123I-Tyr3]-octreotide: The Rotterdam experience with more than 1000 patients. Eur J Nucl Med. 1993;20(8):716–731. [DOI] [PubMed] [Google Scholar]
  • 31. Wang S, Yang W, Deng J, et al. Correlation between 99mTc-HYNIC-octreotide SPECT/CT somatostatin receptor scintigraphy and pathological grading of meningioma. J Neurooncol. 2013;113(3):519–526. [DOI] [PubMed] [Google Scholar]
  • 32. Buchmann I, Henze M, Engelbrecht S, et al. Comparison of 68Ga-DOTATOC PET and 111In-DTPAOC (Octreoscan) SPECT in patients with neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2007;34(10):1617–1626. [DOI] [PubMed] [Google Scholar]
  • 33. Kunz WG, Jungblut LM, Kazmierczak PM, et al. Improved detection of transosseous meningiomas using 68Ga-DOTATATE PET/CT compared with contrast-enhanced MRI. J Nucl Med. 2017;58(10):1580–1587. [DOI] [PubMed] [Google Scholar]
  • 34. Ueberschaer M, Vettermann FJ, Forbrig R, et al. Simpson grade revisited - intraoperative estimation of the extent of resection in meningiomas versus postoperative somatostatin receptor positron emission tomography/computed tomography and magnetic resonance imaging. Neurosurgery. 2020;88(1):140–146. [DOI] [PubMed] [Google Scholar]
  • 35. Teske N, Biczok A, Quach S, et al. Postoperative [68Ga]Ga-DOTA-TATE PET/CT imaging is prognostic for progression-free survival in meningioma WHO grade 1. Eur J Nucl Med Mol Imaging. 2023;51(1):206–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Hertz S, Roberts A, Means JH, Evans RD.. Radioactive iodine as an indicator in thyroid physiology: Iodine collection by normal and hyperplastic thyroids in rabbits. Am J Physiol-Legacy Content. 1940;128(3):565–576. [Google Scholar]
  • 37. Campennì A, Avram AM, Verburg FA, et al. The EANM guideline on radioiodine therapy of benign thyroid disease. Eur J Nucl Med Mol Imaging. 2023;50(11):3324–3348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Avram AM, Giovanella L, Greenspan B, et al. SNMMI procedure standard/EANM practice guideline for nuclear medicine evaluation and therapy of differentiated thyroid cancer: Abbreviated version. J Nucl Med. 2022;63(6):15N–35N. [PubMed] [Google Scholar]
  • 39. Sartor O, De Bono J, Chi KN, et al. ; VISION Investigators. Lutetium-177–PSMA-617 for metastatic castration-resistant prostate cancer. N Engl J Med. 2021;385(12):1091–1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Sartor O, Castellano Gauna DE, Herrmann K, et al. LBA13 Phase III trial of [177Lu]Lu-PSMA-617 in taxane-naive patients with metastatic castration-resistant prostate cancer (PSMAfore). Ann Oncol. 2023;34(S2):S1324–S1325. [Google Scholar]
  • 41. Wright GL, Haley C, Beckett ML, Schellhammer PF.. Expression of prostate-specific membrane antigen in normal, benign, and malignant prostate tissues. Urol Oncol. 1995;1(1):18–28. [DOI] [PubMed] [Google Scholar]
  • 42. Mannweiler S, Amersdorfer P, Trajanoski S, et al. Heterogeneity of prostate-specific membrane antigen (PSMA) expression in prostate carcinoma with distant metastasis. Pathol Oncol Res. 2009;15(2):167–172. [DOI] [PubMed] [Google Scholar]
  • 43. Hofman MS, Lawrentschuk N, Francis RJ, et al. ; proPSMA Study Group Collaborators. Prostate-specific membrane antigen PET-CT in patients with high-risk prostate cancer before curative-intent surgery or radiotherapy (proPSMA): A prospective, randomised, multicentre study. Lancet. 2020;395(10231):1208–1216. [DOI] [PubMed] [Google Scholar]
  • 44. Papotti M, Bongiovanni M, Volante M, et al. Expression of somatostatin receptor types 1-5 in 81 cases of gastrointestinal and pancreatic endocrine tumors. A correlative immunohistochemical and reverse-transcriptase polymerase chain reaction analysis. Virchows Arch. 2002;440(5):461–475. [DOI] [PubMed] [Google Scholar]
  • 45. Nasir A, Stridsberg M, Strosberg J, et al. Somatostatin receptor profiling in hepatic metastases from small intestinal and pancreatic neuroendocrine neoplasms: Immunohistochemical approach with potential clinical utility. Cancer Control. 2006;13(1):52–60. [DOI] [PubMed] [Google Scholar]
  • 46. Kaemmerer D, Träger T, Hoffmeister M, et al. Inverse expression of somatostatin and CXCR4 chemokine receptors in gastroenteropancreatic neuroendocrine neoplasms of different malignancy. Oncotarget. 2015;6(29):27566–27579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Strosberg J, El-Haddad G, Wolin E, et al. ; NETTER-1 Trial Investigators. Phase 3 Trial of 177 lu-dotatate for midgut neuroendocrine tumors. N Engl J Med. 2017;376(2):125–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Baudin E, Walter TA, Beron A, et al. 887O First multicentric randomized phase II trial investigating the antitumor efficacy of peptide receptor radionucleide therapy with 177Lutetium-Octreotate (OCLU) in unresectable progressive neuroendocrine pancreatic tumor: Results of the OCLURANDOM trial. Ann Oncol. 2022;33(S7):S954. [Google Scholar]
  • 49. Singh S, Halperin DM, Myrehaug S, et al. [177 Lu]Lu-DOTA-TATE in newly diagnosed patients with advanced grade 2 and grade 3, well-differentiated gastroenteropancreatic neuroendocrine tumors: Primary analysis of the phase 3 randomized NETTER-2 study. J Clin Oncol. 2024;42(3_suppl):LBA588–LBA588. [Google Scholar]
  • 50. Steeg PS. The blood-tumour barrier in cancer biology and therapy. Nat Rev Clin Oncol. 2021;18(11):696–714. [DOI] [PubMed] [Google Scholar]
  • 51. Mair MJ, Bartsch R, Le Rhun E, et al. Understanding the activity of antibody–drug conjugates in primary and secondary brain tumours. Nat Rev Clin Oncol. 2023;20(6):372–389. [DOI] [PubMed] [Google Scholar]
  • 52. Li Y, Marcu LG, Hull A, Bezak E.. Radioimmunotherapy of glioblastoma multiforme - Current status and future prospects. Crit Rev Oncol Hematol. 2021;163:103395. [DOI] [PubMed] [Google Scholar]
  • 53. Gholamrezanezhad A, Shooli H, Jokar N, Nemati R, Assadi M.. Radioimmunotherapy (RIT) in brain tumors. Nucl Med Mol Imaging. 2019;53(6):374–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Puttemans J, Lahoutte T, D’Huyvetter M, Devoogdt N.. Beyond the barrier: Targeted radionuclide therapy in brain tumors and metastases. Pharmaceutics. 2019;11(8):376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Cimini A, Ricci M, Chiaravalloti A, Filippi L, Schillaci O.. Theragnostic aspects and radioimmunotherapy in pediatric tumors. Int J Mol Sci . 2020;21(11):3849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Wei X, Schlenkhoff C, Schwarz B, Essler M, Ahmadzadehfar H.. Combination of 177Lu-PSMA-617 and external radiotherapy for the treatment of cerebral metastases in patients with castration-resistant metastatic prostate cancer. Clin Nucl Med. 2017;42(9):704–706. [DOI] [PubMed] [Google Scholar]
  • 57. Sabet A, Ahmadzadehfar H, Herrlinger U, et al. Successful radiopeptide targeting of metastatic anaplastic meningioma: Case report. Radiat Oncol. 2011;6(1):94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Makis W, McCann K, McEwan AJB.. Rhabdoid papillary meningioma treated with 177Lu DOTATATE PRRT. Clin Nucl Med. 2015;40(3):237–240. [DOI] [PubMed] [Google Scholar]
  • 59. Backhaus P, Huss S, Kösek V, Weckesser M, Rahbar K.. Lung metastases of intracranial atypical meningioma diagnosed on posttherapeutic imaging after 177Lu-DOTATATE Therapy. Clin Nucl Med. 2018;43(6):e184–e185. [DOI] [PubMed] [Google Scholar]
  • 60. Braat AJT, Snijders TJ, Seute T, Vonken EPA.. Will 177Lu-DOTATATE treatment become more effective in salvage meningioma patients, when boosting somatostatin receptor saturation? a promising case on intra-arterial administration. Cardiovasc Intervent Radiol. 2019;42(11):1649–1652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Assadi M, Rekabpour SJ, Amini A, et al. Peptide receptor radionuclide therapy with 177Lu-DOTATATE in a case of concurrent neuroendocrine tumors and meningioma: Achieving two things in a single action. Mol Imaging Radionucl Ther. 2021;30(2):107–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Pirisino R, Filippi L, D’Agostini A, Bagni O.. Management of a patient with metastatic gastrointestinal neuroendocrine tumor and meningioma submitted to peptide receptor radionuclide therapy with 177Lu-DOTATATE. Clin Nucl Med. 2022;47(11):e692–e695. [DOI] [PubMed] [Google Scholar]
  • 63. van Essen M, Krenning EP, Kooij PP, et al. Effects of therapy with [177Lu-DOTA0, Tyr3]octreotate in patients with paraganglioma, meningioma, small cell lung carcinoma, and melanoma. J Nucl Med. 2006;47(10):1599–1606. [PubMed] [Google Scholar]
  • 64. Bartolomei M, Bodei L, De Cicco C, et al. Peptide receptor radionuclide therapy with (90)Y-DOTATOC in recurrent meningioma. Eur J Nucl Med Mol Imaging. 2009;36(9):1407–1416. [DOI] [PubMed] [Google Scholar]
  • 65. Bodei L, Cremonesi M, Grana CM, et al. Peptide receptor radionuclide therapy with 177Lu-DOTATATE: The IEO phase I-II study. Eur J Nucl Med Mol Imaging. 2011;38(12):2125–2135. [DOI] [PubMed] [Google Scholar]
  • 66. Kreissl MC, Hänscheid H, Löhr M, et al. Combination of peptide receptor radionuclide therapy with fractionated external beam radiotherapy for treatment of advanced symptomatic meningioma. Radiat Oncol. 2012;7:99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Hartrampf PE, Hänscheid H, Kertels O, et al. Long-term results of multimodal peptide receptor radionuclide therapy and fractionated external beam radiotherapy for treatment of advanced symptomatic meningioma. Clin Transl Radiat Oncol. 2020;22:29–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Hänscheid H, Sweeney RA, Flentje M, et al. PET SUV correlates with radionuclide uptake in peptide receptor therapy in meningioma. Eur J Nucl Med Mol Imaging. 2012;39(8):1284–1288. [DOI] [PubMed] [Google Scholar]
  • 69. Minutoli F, Amato E, Sindoni A, et al. Peptide receptor radionuclide therapy in patients with inoperable meningiomas: Our experience and review of the literature. Cancer Biother Radiopharm. 2014;29(5):193–199. [DOI] [PubMed] [Google Scholar]
  • 70. Gerster-Gilliéron K, Forrer F, Maecke H, et al. 90Y-DOTATOC as a therapeutic option for complex recurrent or progressive meningiomas. J Nucl Med. 2015;56(11):1748–1751. [DOI] [PubMed] [Google Scholar]
  • 71. Marincek N, Radojewski P, Dumont RA, et al. Somatostatin receptor–targeted radiopeptide therapy with 90 Y-DOTATOC and 177 Lu-DOTATOC in Progressive Meningioma: Long-term results of a phase ii clinical trial. J Nucl Med. 2015;56(2):171–176. [DOI] [PubMed] [Google Scholar]
  • 72. Seystahl K, Stoecklein V, Schüller U, et al. Somatostatin receptor-targeted radionuclide therapy for progressive meningioma: Benefit linked to 68Ga-DOTATATE/-TOC uptake. Neuro Oncol. 2016;18(11): 1538–1547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Parghane RV, Talole S, Basu S.. Prevalence of hitherto unknown brain meningioma detected on 68Ga-DOTATATE positron-emission tomography/computed tomography in patients with metastatic neuroendocrine tumor and exploring potential of 177Lu-DOTATATE peptide receptor radionuclide therapy as single-shot treatment approach targeting both tumors. World J Nucl Med. 2019;18(2):160–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Müther M, Roll W, Brokinkel B, et al. Response assessment of somatostatin receptor targeted radioligand therapies for progressive intracranial meningioma. Nuklearmedizin. 2020;59(5):348–355. [DOI] [PubMed] [Google Scholar]
  • 75. Kertels O, Breun M, Hänscheid H, et al. Peptide receptor radionuclide therapy in patients with neurofibromatosis type 2: Initial experience. Clin Nucl Med. 2021;46(6):e312–e316. [DOI] [PubMed] [Google Scholar]
  • 76. Salgues B, Graillon T, Horowitz T, et al. Somatostatin receptor theranostics for refractory meningiomas. Curr Oncol. 2022;29(8):5550–5565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Minczeles NS, Bos EM, de Leeuw RC, et al. Efficacy and safety of peptide receptor radionuclide therapy with [177Lu]Lu-DOTA-TATE in 15 patients with progressive treatment-refractory meningioma. Eur J Nucl Med Mol Imaging. 2023;50(4):1195–1204. [DOI] [PubMed] [Google Scholar]
  • 78. Kurz SC, Zan E, Cordova C, et al. Evaluation of the SSTR2-targeted Radiopharmaceutical 177Lu-DOTATATE and SSTR2-specific 68Ga-DOTATATE PET as Imaging Biomarker in Patients with Intracranial Meningioma. Clin Cancer Res. 2024;30(4):680–686. [DOI] [PubMed] [Google Scholar]
  • 79. Huang RY, Bi WL, Weller M, et al. Proposed response assessment and endpoints for meningioma clinical trials: Report from the Response Assessment in Neuro-Oncology Working Group. Neuro Oncol. 2019;21(1):26–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Tabouret E, Furtner J, Graillon T, et al. 3D volume growth rate evaluation in the EORTC-BTG-1320 clinical trial for recurrent WHO grade 2 and 3 meningiomas. Neuro-Oncology. 2024:noae037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Parker C, Lewington V, Shore N, et al. ; Targeted Alpha Therapy Working Group. Targeted alpha therapy, an emerging class of cancer agents: A review. JAMA Oncol. 2018;4(12):1765–1772. [DOI] [PubMed] [Google Scholar]
  • 82. Martz N, Salleron J, Dhermain F, et al. Target volume delineation for radiotherapy of meningiomas: An ANOCEF consensus guideline. Radiat Oncol. 2023;18(1):113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Parker C, Nilsson S, Heinrich D, et al. ; ALSYMPCA Investigators. Alpha emitter Radium-223 and survival in metastatic prostate cancer. N Engl J Med. 2013;369(3):213–223. [DOI] [PubMed] [Google Scholar]
  • 84. Sathekge MM, Lawal IO, Bal C, et al. Actinium-225-PSMA radioligand therapy of metastatic castration-resistant prostate cancer (WARMTH Act): A multicentre, retrospective study. Lancet Oncol. 2024;25(2):175–183. [DOI] [PubMed] [Google Scholar]
  • 85. Yadav MP, Ballal S, Sahoo RK, Bal C.. Efficacy and safety of 225Ac-DOTATATE targeted alpha therapy in metastatic paragangliomas: A pilot study. Eur J Nucl Med Mol Imaging. 2022;49(5):1595–1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Demirci E, Alan Selçuk N, Beydağı G, et al. Initial findings on the use of [225Ac]Ac-DOTATATE therapy as a theranostic application in patients with neuroendocrine tumors. Mol Imaging Radionucl Ther. 2023;32(3):226–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Ku A, Facca VJ, Cai Z, Reilly RM.. Auger electrons for cancer therapy - A review. EJNMMI Radiopharm Chem. 2019;4(1):27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Krenning EP, de Jong M, Kooij PP, et al. Radiolabelled somatostatin analogue(s) for peptide receptor scintigraphy and radionuclide therapy. Ann Oncol. 1999;10(suppl 2):S23–S29. [DOI] [PubMed] [Google Scholar]
  • 89. Valkema R, De Jong M, Bakker WH, et al. Phase I study of peptide receptor radionuclide therapy with [In-DTPA]octreotide: The Rotterdam experience. Semin Nucl Med. 2002;32(2):110–122. [DOI] [PubMed] [Google Scholar]
  • 90. Limouris GS, Chatziioannou A, Kontogeorgakos D, et al. Selective hepatic arterial infusion of In-111-DTPA-Phe1-octreotide in neuroendocrine liver metastases. Eur J Nucl Med Mol Imaging. 2008;35(10):1827–1837. [DOI] [PubMed] [Google Scholar]
  • 91. Bailey DL, Willowson KP, Harris M, et al. 64Cu treatment planning and 67cu therapy with radiolabeled [64Cu/67Cu]MeCOSar-octreotate in subjects with unresectable multifocal meningioma: Initial results for human imaging, safety, biodistribution, and radiation dosimetry. J Nucl Med. 2023;64(5):704–710. [DOI] [PubMed] [Google Scholar]
  • 92. Haug AR. PRRT of neuroendocrine tumors: Individualized dosimetry or fixed dose scheme? EJNMMI Res. 2020;10(1):35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Konijnenberg M, Herrmann K, Kobe C, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Wiseman GA, Kornmehl E, Leigh B, et al. Radiation dosimetry results and safety correlations from 90Y-ibritumomab tiuxetan radioimmunotherapy for relapsed or refractory non-Hodgkin’s lymphoma: combined data from 4 clinical trials. J Nucl Med. 2003;44(3):465–474. [PubMed] [Google Scholar]
  • 95. Sjögreen Gleisner K, Spezi E, Solny P, et al. Variations in the practice of molecular radiotherapy and implementation of dosimetry: Results from a European survey. EJNMMI Phys. 2017;4(1):28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Del Prete M, Buteau FA, Arsenault F, et al. Personalized 177Lu-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] [PubMed] [Google Scholar]
  • 97. Boursier C, Zaragori T, Bros M, et al. Semi-automated segmentation methods of SSTR PET for dosimetry prediction in refractory meningioma patients treated by SSTR-targeted peptide receptor radionuclide therapy. Eur Radiol. 2023;33(10):7089–7098. [DOI] [PubMed] [Google Scholar]
  • 98. Arvanitis CD, Ferraro GB, Jain RK.. The blood–brain barrier and blood–tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20(1):26–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Kramer K, Pandit-Taskar N, Kushner BH, et al. Phase 1 study of intraventricular 131I-omburtamab targeting B7H3 (CD276)-expressing CNS malignancies. J Hematol Oncol. 2022;15(1):165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Reardon DA, Zalutsky MR, Akabani G, et al. A pilot study: 131I-antitenascin monoclonal antibody 81c6 to deliver a 44-Gy resection cavity boost. Neuro Oncol. 2008;10(2):182–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Zalutsky MR, Reardon DA, Akabani G, et al. Clinical experience with alpha-particle emitting 211At: Treatment of recurrent brain tumor patients with 211At-labeled chimeric antitenascin monoclonal antibody 81C6. J Nucl Med. 2008;49(1):30–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Brenner AJ, Phillips WT, Bao A, et al. 277O The ReSPECT-GBMTM phase I/IIa trial of rhenium-186 nanoliposome (186RNL) in recurrent glioma via convection enhanced delivery (CED) and planned phase IIb trial. Ann Oncol. 2022;33(S7):S666. [Google Scholar]
  • 103. Verburg FA, Wiessmann M, Neuloh G, Mottaghy FM, Brockmann MA.. Intraindividual comparison of selective intraarterial versus systemic intravenous 68Ga-DOTATATE PET/CT in patients with inoperable meningioma. Nuklearmedizin. 2019;58(1):23–27. [DOI] [PubMed] [Google Scholar]
  • 104. Vonken EJPA, Bruijnen RCG, Snijders TJ, et al. Intraarterial administration boosts 177Lu-HA-DOTATATE accumulation in salvage meningioma patients. J Nucl Med. 2022;63(3):406–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Ebbers S, Barentsz M, Braat A, Lam M.. Intra-arterial peptide receptor radionuclide therapy for neuroendocrine tumor liver metastases. Dig Dis Interv. 2019;03(01):081–090. [Google Scholar]
  • 106. Lawhn-Heath C, Fidelman N, Chee B, et al. Intraarterial peptide receptor radionuclide therapy Using 90Y-DOTATOC for hepatic metastases of neuroendocrine tumors. J Nucl Med. 2021;62(2):221–227. [DOI] [PubMed] [Google Scholar]
  • 107. Agrawal S, Arora S, Amiri-Kordestani L, et al. Use of single-arm trials for US Food and drug administration drug approval in oncology, 2002-2021. JAMA Oncol. 2022;9(2):266. [DOI] [PubMed] [Google Scholar]
  • 108. Albert NL, Le Rhun E, Minniti G, et al. Translating the theranostic concept to neuro-oncology: disrupting barriers. Lancet Oncol. 2024;25(9):e441–e451. [DOI] [PubMed] [Google Scholar]
  • 109. Aaronson NK, Ahmedzai S, Bergman B, et al. The European organization for research and treatment of cancer QLQ-C30: A Quality-of-life instrument for use in international clinical trials in oncology. J Natl Cancer Inst. 1993;85(5):365–376. [DOI] [PubMed] [Google Scholar]
  • 110. Osoba D, Aaronson NK, Muller M, et al. The development and psychometric validation of a brain cancer quality-of-life questionnaire for use in combination with general cancer-specific questionnaires. Qual Life Res. 1996;5(1):139–150. [DOI] [PubMed] [Google Scholar]
  • 111. Graillon T, Sanson M, Campello C, et al. Everolimus and octreotide for patients with recurrent meningioma: Results from the phase II CEVOREM trial. Clin Cancer Res. 2020;26(3):552–557. [DOI] [PubMed] [Google Scholar]
  • 112. Wild D, Fani M, Fischer R, et al. Comparison of somatostatin receptor agonist and antagonist for peptide receptor radionuclide therapy: A Pilot Study. J Nucl Med. 2014;55(8):1248–1252. [DOI] [PubMed] [Google Scholar]
  • 113. Eigler C, McDougall L, Bauman A, et al. Radiolabeled somatostatin receptor antagonist versus agonist for peptide receptor radionuclide therapy in patients with therapy-resistant meningioma: PROMENADE Phase 0 Study. J Nucl Med. 2024;65(4):573–579. [DOI] [PubMed] [Google Scholar]

Articles from Neuro-Oncology are provided here courtesy of Society for Neuro-Oncology and Oxford University Press

RESOURCES