Abstract
Introduction
Meningiomas are associated with several gonadal steroid hormone-related risk factors and demonstrate a predominance in females. These associations led to investigations of the role that hormones may have on meningioma growth and development. While it is now accepted that most meningiomas express progesterone and somatostatin receptors, the conclusion for other receptors has been less definitive.
Methods
We performed a review of what is known regarding the relationship between hormones and meningiomas in the published literature. Furthermore, we reviewed clinical trials related to hormonal agents in meningiomas using MEDLINE PubMed, Scopus, and the NIH clinical trials database.
Results
We identify that all steroid-hormone trials lacked receptor identification or positive receptor status in the majority of patients. In contrast, four out of five studies involving somatostatin analogs used positive receptor status as part of the inclusion criteria.
Conclusions
Several clinical trials have recently been completed or are now underway using somatostatin analogs in combination with other therapies that appear promising, but a reevaluation of hormone-based monotherapy is warranted. Synthesizing this evidence, we clarify the remaining questions and present future directions for the study of the biological role and therapeutic potential of hormones in meningioma and discuss how the stratification of patients using features such as grade, receptor status, and somatic mutations, might be used for future trials to select patients most likely to benefit from specific therapies.
Keywords: Meningioma, Hormones, Receptors, Estrogen, Progesterone, Androgen
Introduction
Meningiomas have a female predominance with an overall ratio of 2:1, and assessments of risk factors have implicated gonadal steroid hormones and receptor expression relevant to the growth hormone (GH/IGF-1) signaling axis [1-3]. Most meningiomas are progesterone receptor (PGR) positive, but inconsistent results between studies and incomplete assessment of all relevant hormone receptors have obscured the results for androgen receptors (AR) and estrogen receptors [4, 5]. Scientific interest in the effects of gonadal steroid hormones on meningioma tumorigenesis and treatment has been renewed after evidence that some patients with long-term progestin/antiandrogen [cyproterone acetate (CPA)] exposure share molecular, histological, and anatomical features in addition to an increased risk of developing a meningioma [6-9]. These developments, and still poorly understood contributions of hormones to meningioma growth and development, led us to review what is known about this subject, the clinical trials that have been conducted, and considerations for the future of hormone therapy in meningiomas.
Female predominance in meningiomas
In Harvey Cushing’s 1938 landmark, “Meningiomas”, he wrote, “we have noted for many years a definite predominance in the incidence of meningiomas in females over males, the proportion being 60–40…still more striking is the fact that in our series tumors of certain loci are largely restricted to women… Meningiomas in not a few instances appear to have been incited by unusual activity, or at least to have shown their first recognizable symptoms soon after a recent pregnancy” [10]. Both Cushing’s study and contemporary quantifications show a difference between female and male incidence in meningioma, which changes over the lifespan and reaches the greatest difference of approximately 3:1 among those aged 35–55 (Fig. 1) [10, 11]. Notably, this age range coincides with perimenopause in females, a time known to be metabolically important for many organ systems, including the brain, and a process resulting in dramatic hormonal changes [12]. More recently, advances in the understanding of the genetic drivers of meningiomas has helped elucidate heterogeneity related to patient and tumor features, even within the same grade [13, 14]. Genomic subgroups associate with tumor location, histology, and outcomes [13, 14]. Additionally, female sex is overrepresented in KLF4/TRAF7 and POLR2A genomic subgroups, which are also known to be associated with skull base location [13, 15, 16].
Fig. 1.
Female predominance in meningiomas. A Figure from Wiemels et al. [11] age and sex incidence of meningioma per 100,000 people in United States from 2002 to 2006
In addition to gonadal steroid hormones, the X-chromosome has been hypothesized as a contributing factor to the observed sex predominance [17]. A population-based study in Great Britain of people with Turner Syndrome (45,X0) revealed that among all tumors, Turner Syndrome participants were much more likely to develop meningiomas, SIR = 12.0 CI95%[4.8–24.8], compared to the general population without Turner Syndrome. Females with Turner Syndrome suffer from premature ovarian failure and lack the ability to synthesize gonadal steroid hormones estradiol and progesterone at typical post-pubertal levels and are often supplemented with hormone replacement therapy (HRT). The authors of the study postulate this relationship but report that they did not have access to information regarding patient exposure to HRT [18].
Hormone associated risk factors
Several reviews dedicated to the topic of meningioma hormone associated risk factors have been published, with some well supported risk factors including oral contraceptives in premenopausal women, elevated BMI, uterine fibroids, endometriosis, and a decreased risk among women who breastfed after pregnancy [1, 19]. Interestingly, meningiomas belonging to people with a history of CPA use were found to be more likely to harbor PIK3CA mutations and less likely to have NF2 loss [8, 20]. The importance of this can be exemplified by reports that the same hormone can have different effects on meningiomas depending on the mutation status. One such example is a case series of individuals with multiple meningiomas with different underlying mutations, in which those different tumors responded to the treatment in opposing ways (regression vs progression) despite changing only a single hormone therapy [21]. It is worth noting that while CPA is widely available in many countries, it is not approved by the US FDA and therefore is unlikely to be a significant contributor to meningiomas seen in the United States [9]. A recent study of 120 patients analyzed the growth rate in incidental meningiomas and identified that patients undergoing estrogen-based hormone replacement therapy had significantly smaller tumors compared to age-matched female controls, but there was not a statistical difference in progression free survival (PFS) [22]. Still, there are 44 gonadal-hormone related FDA-approved moieties, 16 of which have been used to treat various cancers, and only 4 that have been clinically evaluated in meningioma (Table 1) [23]. More systematic work is needed to characterize the risk or benefit that hormones and/or hormonal therapies can have on meningiomas.
Table 1.
FDA approved gonadal-hormone modulatory active moieties
| Active moiety | Pharmacological class | Common use (PubChem) |
|---|---|---|
| Danazol | Androgen Receptor Agonist | Endometriosis, fibrocystic breast disease, hereditary angioedema |
| Fluoxymesterone‡ | Androgen receptor agonist | Anabolic steroid that has been used in the treatment of male hypogonadism, delayed puberty in males, breast neoplasms in women, and off label use in anemia |
| Methyltestosterone‡ | Androgen | Anabolic steroid hormone used to treat men with a testosterone deficiency, women with breast cancer, breast pain, swelling due to pregnancy, and with the addition of estrogen it can treat symptoms of menopause |
| Nandrolone | Androgen | Hypogonadal men, HIV-wasting syndrome, and in other conditions in order to increase nitrogen retention and fat-free muscle mass |
| Testosterone | Androgen | Primary hypogonadism and hypogonadotropic hypogonadism |
| Bicalutamide‡ | Androgen receptor inhibitor | Prostate cancer that has metastasized and other cancers in combination with a luteinizing hormone-releasing hormone agonist |
| Clascoterone | Androgen receptor inhibitor | Acne vulgaris (topical) |
| Enzalutamide‡ | Androgen receptor inhibitor | Metastatic castration-resistant prostate cancer who have previously received docetaxel |
| Flutamide‡ | Androgen receptor inhibitor | Prostate cancer that has metastasized and other cancers |
| Nilutamide‡ | Androgen receptor inhibitor | Prostate cancer that has metastasized |
| Estetrol anhydrous | Estrogen | Contraception prevents pregnancy by suppressing ovulation |
| Estradiol* | Estrogen | Vasomotor symptoms from menopause, vaginal atrophy, hypoestrogenism due to hypogonadism, castration, or ovarian failure, palliative breast cancer and other uses |
| Estrogens, conjugated | Estrogen | Vasomotor symptoms from menopause, vaginal atrophy, hypoestrogenism due to hypogonadism, castration, or ovarian failure |
| Ethinyl estradiol | Estrogen | Premenstrual dysphoric disorder, moderate acne, moderate to severe vasomotor symptoms of menopause, prevention of postmenopausal osteoporosis |
| Mestranol | Estrogen | Oral contraceptive |
| Bazedoxifene | Estrogen agonist/antagonist | Treatment of vasomotor symptoms associated with menopause, prevention of postmenopausal osteoporosis |
| Clomiphene | Estrogen agonist/antagonist | Hypogonadotropic hypogonadism in men |
| Ospemifene | Estrogen agonist/antagonist | Treatment of dyspareunia, a symptom of vulvar and vaginal atrophy due to menopause |
| Raloxifene‡ | Estrogen agonist/antagonist | Osteoporosis in postmenopausal women and corticosteroid-induced bone loss. Prevention for women at high risk of developing breast cancer |
| Tamoxifen‡,* | Estrogen agonist/antagonist | HR+breast cancer |
| Toremifene‡ | Estrogen agonist/antagonist | HR+breast cancer |
| Fulvestrant‡ | Estrogen receptor antagonist | HR+metastatic breast cancer |
| Dutasteride | 5-Alpha reductase inhibitor | Benign prostatic hyperplasia (BPH) |
| Finasteride | 5-Alpha reductase inhibitor | Benign prostatic hyperplasia (BPH) |
| Anastrozole‡ | Aromatase inhibitor | Certain women with HR+breast cancer |
| Exemestane‡ | Aromatase inhibitor | Advanced breast cancer in postmenopausal women whose disease has progressed following tamoxifen therapy |
| Letrozole‡ | Aromatase inhibitor | Certain women with HR+breast cancer |
| Testolactone‡ | Aromatase inhibitor | Advanced breast cancer in postmenopausal women |
| Progesterone | Progesterone | Prevention of endometrial hyperplasia in non-hysterectomized postmenopausal women who are receiving conjugated estrogens |
| Ulipristal | Progesterone agonist/antagonist | Emergency contraception |
| Desogestrel | Progestin | Oral contraceptives that contain a mix of estrogen and progestin which inhibit ovulation |
| Dienogest | Progestin | Treatment of endometriosis alone and as a contraceptive in combination with ethinylestradiol |
| Drospirenone | Progestin | Moderate acne vulgaris, premenstrual dysphoric disorder, and menopause-associated symptoms, such as vasomotor symptoms and vulvovaginal atrophy |
| Ethynodiol | Progestin | Oral contraceptive |
| Etonogestrel | Progestin | Subdermal implants as long-acting reversible contraception |
| Hydroxyprogesterone‡ | Progestin | Contraception, uterine bleeding, dysmenorrhea, endometriosis, endometrial, renal cell, breast, and prostate cancers |
| Levonorgestrel | Progestin | Emergency contraception (alone), long-term contraception (with estradiol), menopausal vasomotor symptoms and prevent osteoporosis prevention. Off-label, may be used to treat menorrhagia, endometrial hyperplasia, and endometriosis |
| Medroxyprogesterone‡ | Progestin | Oral contraception, uterine bleeding, fibroids, uterine cancer |
| Megestrol‡,* | Progestin | Advanced breast or endometrial carcinoma |
| Norelgestromin | Progestin | Contraception |
| Norethindrone | progestin | Contraception and hormone replacement therapy in the treatment of postmenopausal osteoporosis and moderate-to-severe vasomotor symptoms arising from menopause |
| Norgestimate | Progestin | Contraception as well as the treatment of moderate acne vulgaris |
| Mifepristone* | Progestin antagonist | Medical termination of intrauterine pregnancy through 49 days’ pregnancy and can be used to control hyperglycemia secondary to hypercortisolism in adults |
| Segesterone | Progestin/estrogen CHC | Contraceptive and endometriosis (implant) |
Trialed in meningioma
Used as a treatment for cancer
Hormone receptors in meningioma
Hormones mediate control of gene regulation by binding to nuclear and trans-membrane receptors resulting in cell signaling cascades and epigenetic modifications. Nuclear hormone receptors relevant to meningioma include PGR, AR, estrogen receptor alpha ESR1 (ERα) and estrogen receptor beta ESR2 (ERβ) [24]. These bind to the DNA and act to influence chromatin configuration by recruitment of different coregulatory complexes responsible for different biological activity [25]. In the case of estrogen receptors, these represent distinct gene products on different chromosomes that lack cross-affinity when detected by antibodies and can have opposing gene regulation [26]. Another class of hormone receptors includes the trans-membrane receptors, many of which are G-coupled membrane receptors, such as somatostatin receptors 1–5 (SSTRs), gonadotropin-releasing hormone receptor (GnRHR or LHRHR), and the G-coupled Protein Estrogen Receptor 1 (GPER1) [27].
The clinical importance of distinguishing between receptors binding to the same ligand can be exemplified by the selective estrogen receptor modulator (SERM) tamoxifen. A SERM is a mixed agonist/antagonist with activity that differs between estrogen receptors; specifically, it is known to be an agonist to ERα but an antagonist to ERβ and GPER1 [25, 28]. The clinical consequence of this mixed activity is demonstrated in tamoxifen’s use in treating breast cancer but its risk for developing endometrial cancer [29]. Our unpublished meta-analysis and comprehensive review of the literature identifies that ERα was the only estrogen receptor assessed in almost all studies of meningioma, with few notable exceptions [24]. Some authors who have detected ERα have reported its association with increased proliferation and high grade meningiomas [30, 31]. Most meningiomas are PGR positive and its expression has been associated with WHO grade I and inversely correlated with chromosome 22 loss, where NF2 is located [31-33].
GNRHR expression is important because luteinizing hormone agonists are sometimes used to treat prostate cancer and meningioma growth is a potential complication of this treatment [34, 35]. Multiple publications have identified SSTR1-5 expression in meningioma at high frequencies [3]. A large study of meningiomas with over 700 tumors identified that SSTRs are associated with different tumor characteristics, such as SSTR2A overrepresentation in skull-base tumors and a negative association with high grade and NF2 mutant meningiomas [3] and reported to be a prognostic marker [2]. While meningioma literature has often evaluated steroid-hormone receptors and SSTRs separately, ERα in particular is known to play a fundamental role in normal physiological regulation of GH, and the relationship between SSTRs, ERα and PGR expression in cancer is reported in prostate and breast cancer cells [36-38]. Additionally, high BMI is associated with meningioma development and it has been found to correlate with leptin receptor (LEPR) expression in meningiomas, a receptor known to communicate with the IGF-1/GH and gonadal steroid hormone pathways [39-41].
Clinical trials
We reviewed clinical trials related to hormonal agents in meningiomas (Table 2) using MEDLINE PubMed, Scopus, and the NIH clinical trials database (clinicaltrials.gov). Among these studies, only one was a randomized controlled trial (NCT03015701, Table 4), which assessed mifepristone in meningiomas, summarized below [42]. Several other studies were conducted but all had thirty-four patients or less with heterogenous patient and tumor inclusion characteristics. Studies defined complete response as no tumor after treatment, partial response (PR) as > 50% tumor reduction, and progressive disease > 25% tumor increase, unless otherwise specified.
Table 2.
All hormone related clinical trials in this review
| First author (year) |
Drug | Dose | Study design | Median treatment duration (months) |
Number of patients |
Sex | Median age (years) |
Primary outcome |
|---|---|---|---|---|---|---|---|---|
| Markwalder (1985) [47] | Tamoxifen | 10 mg orally t.i.d | Prospective cohort; pilot study | 12 | 6‡ | 5 F; 1 M | 71.5 | 4 assessed; 1 non-specific response; 2 stable; 1 progressed |
| Goodwin (1993) [48] | Tamoxifen | 40 mg b.i.d. for 4 days then 10 mg orally b.i.d | Prospective cohort; phase II | 31 | 21‡ | 14 F; 7 M | 58 | 1 partial response; 2 minor response; 6 stable; 10 progressed |
| Grunberg (1990) [50] | Megestrol acetate | 40–80 mg orally q.i.d | Prospective cohort | 5 | 9 | 5 F; 4 M | 40 | 2 stable; 7 progressed |
| Grunberg (2006) [51] | Mifepristone 14-day course of dexamethasone 1 mg | 200 mg q.d. orally | Prospective cohort | 35 | 28 | 19 F; 9 M | 56 | 5 minor response; 3 clinical without radiographic improvement; 2 progressed |
| Touat (2014) [52] | Mifepristone | 200 mg q.d. orally | Case series | 60 | 3 | 3 F | 54 | 100% had > 20% volume reduction |
| Ji (2015) [42] | Mifepristone | 200 mg b.i.d. or placebo orally | Randomized phase III clinical trial | Placebo 11[6-18]; mifepristone 10[7–13]+ | 164 | 116 F; 48 M | 57 | HR 1.02 95%CI[0.72, 1.48] Adjusted P = 0.9 |
Cohort statistics includes patients that were not included in the trial aggregated by publishing author
Failure free survival
Table 4.
Hormone-related NIH registered clinical trials at Clinicaltrials.gov
| NCT number | Start date | Results | Title | Study | |
|---|---|---|---|---|---|
| NCT03015701 | Aug-92 | Southwest Oncology Group∣National Cancer Institute (NCI) | Completed | S9005 mifepristone in meningioma | Drug: Mifepristone∣Other: Placebo |
| NCT00006368 | Jan-98 | Novartis Pharmaceuticals∣Novartis | Completed | Yttrium Y 90 SMT 487 in treating patients with refractory or recurrent cancer | Radiation: yttrium Y 90-edotreotide |
| NCT00049023 | Jan-02 | O’Dorisio, M S∣National Cancer Institute (NCI)∣University of Iowa | Completed | Radiolabeled octreotide in treating children with advanced or refractory Solid tumors | Radiation: 90Y-DOTA-tyr3-OCTREOTIDE |
| NCT00813592 | Nov-08 | University of Utah∣Novartis | Terminated | Phase II study of SOM230 in patients with recurrent or progressive meningioma | Drug: SOM230B |
| NCT00859040 | Mar-09 | Patrick Y. Wen, MD∣Brigham and Women’s Hospital∣Massachusetts General Hospital∣Beth Israel Deaconess Medical Center∣Memorial Sloan Kettering Cancer Center∣Wake Forest University Health Sciences∣Duke University∣Cedars-Sinai Medical Center∣Northwestern University∣Novartis∣Dana-Farber Cancer Institute | Completed | Monthly SOM230C for recurrent or progressive meningioma | Drug: SOM230C |
| NCT02194452 | Sep-13 | Sue O’Dorisio∣National Cancer Institute (NCI)∣Ride for Kids∣University of Iowa | Withdrawn | Efficacy of 68Ga-DOTATOC positron emission tomography (PET) CT in children and young adults with brain tumors | Radiation: gallium Ga 68-edotreotide∣Procedure: positron emission tomography∣Procedure: computed tomography∣Other: laboratory biomarker analysis |
| NCT02333565 | Jan-15 | Assistance Publique Hopitaux De Marseille | Unknown status | Combination of everolimus and octreotide LAR in aggressive recurrent meningiomas | Drug: Everolimus∣Drug: Octreotide |
| NCT03013387 | Jan-17 | Sue O’Dorisio∣National Institutes of Health (NIH)∣National Cancer Institute (NCI)∣University of Iowa | Withdrawn | Dosimetry guided PRRT with 90Y-DOTA-TOC | Radiation: 90Y-DOTA-3-tyr-Octreotide∣Procedure: Positron Emission Tomography (PET) whole body scan∣Drug: Amino Acids |
| NCT03001349 | 16-May-17 | M.D. Anderson Cancer Center∣National Cancer Institute (NCI) | Terminated | 68Ga-DOTA-TOC PET/CT in imaging participants with neuroendocrine tumors | Procedure: Computed Tomography∣Drug: Gallium Ga 68-Edotreotide∣Procedure: Positron Emission Tomography |
| NCT03273712 | 29-Sep-17 | Sue O’Dorisio∣National Institutes of Health (NIH)∣National Cancer Institute (NCI)∣University of Iowa | Unknown status | Dosimetry-guided, peptide receptor radiotherapy (PRRT) With 90Y-DOTA-tyr3-octreotide (90Y-DOTATOC) | Radiation: 90Y-DOTA tyr3-Octreotide∣Diagnostic Test: 68Ga-DOTATOC PET Positron Emission Tomography (PET) whole body scan∣Drug: Amino Acids |
| NCT03936426 | 9-Jul-18 | Clarity Pharmaceuticals Ltd | Completed | Peptide receptor radionuclide therapy administered to participants with meningioma with 67Cu-SARTATE | Drug: Cu-64 SARTATE and Cu-67 SARTATE |
| NCT03583528 | 11-Jul-18 | British Columbia Cancer Agency | Recruiting | DOTATOC PET/CT for imaging NET patients | Diagnostic Test: 68Ga-DOTATOC PET/CT∣Diagnostic Test: 18F-FDG PET/CT |
| NCT03953131 | 10-Jan-19 | M.D. Anderson Cancer Center∣National Cancer Institute (NCI) | Recruiting | Gallium Ga 68-DOTATATE PET/CT in predicting tumor growth in patients with meningiomas | Procedure: Computed Tomography∣Radiation: Gallium Ga 68-DOTATATE∣Procedure: Positron Emission Tomography |
| NCT04109404 | 25-Feb-19 | University Hospital, Brest | Completed | Meningiomas and treatment with CYPROTERONE ACETATE or progestin | Meningiomas and Treatment with CYPROTERONE ACETATE or Progestin |
| NCT03971461 | 15-May-19 | NYU Langone Health | Recruiting | Phase II study of 177Lu-DOTATATE radionuclide in adults with progressive or high-risk meningioma | Drug: Lutathera |
| NCT04081701 | 4-Sep-19 | Weill Medical College of Cornell University∣Novartis Pharmaceuticals | Recruiting | 68-Ga DOTATATE PET/MRI in the diagnosis and management of somatostatin receptor positive CNS tumors | Diagnostic Test: Ga68-DOTATATE-PET/MRI |
| NCT04082520 | 10-Mar-20 | Mayo Clinic∣National Cancer Institute (NCI) | Recruiting | Lutathera for the treatment of inoperable, Progressive meningioma after external beam radiation therapy | Radiation: Gallium Ga 68-DOTATATE∣Drug: Lutetium Lu 177 Dotatate∣Procedure: Magnetic Resonance Imaging∣Procedure: Positron Emission Tomography∣Other: Quality-of-Life Assessment∣Other: Questionnaire Administration |
| NCT04298541 | 18-Mar-21 | Weill Medical College of Cornell University∣Cornell University | Recruiting | Direct comparison of Ga-68-DOTATATE and Ga-68-DOTATOC | Drug: Ga-68-DOTATATE∣Drug: Ga-68-DOTATOC |
| NCT04997317 | 21-Apr-21 | University Hospital, Basel, Switzerland∣Swiss Cancer League | Recruiting | Treatment of recurrent or progressive meningiomas with the radiolabelled somatostatin antagonist 177Lu-satoreotide | Drug: 177Lu-DOTA-JR11 (Phase 0); Cycle 1 and Cycle 2 (cross-over)∣Drug: 177Lu-DOTATOC (Phase 0); Cycle 1 and Cycle 2 (cross-over), Cycle 3 and 4∣Drug: 177Lu-DOTA-JR11 (Phase I/II) |
| NCT04372095 | 6-Jul-21 | Assistance Publique—Hospitaux de Paris | Recruiting | Androcur (cyproterone acetate) and meningioma development: a genotype-environment association study | Procedure: oral smears |
| NCT05278208 | 25-May-22 | Ralph Salloum∣Children’s Hospital Medical Center, Cincinnati∣Nationwide Children’s Hospital | Not yet recruiting | Lutathera for treatment of recurrent or progressive high-grade CNS tumors or meningiomas expressing SST2A2 | Drug: LUTATHERA (lutetium Lu 177 dotatate) |
Tamoxifen
Early in vitro studies of tamoxifen in meningioma revealed a complex interaction with other hormones that could result in the stimulation or inhibition of growth [43, 44]. Some population-based studies have suggested that patients who received tamoxifen for breast cancer treatment may have a decreased risk of meningioma relative to other breast cancer patients, but these results are limited due to the retrospective observational nature of the study design and relatively weak associations [45, 46]. In 1985, Markwalder et al. conducted the first pilot study of tamoxifen for the treatment of meningioma patients and included 6 patients treated for a median duration of 31 months (Table 2) [47]. Four were considered inoperable and method of meningioma confirmation was not specified. Two other tumors were considered non-operative recurrences. Four patients were evaluated at the end of the study, with one patient showing what the author describes as a nonspecific response to tamoxifen, two were stable, and one progressed (Table 2). In 1993, Goodwin et al. conducted a single-arm Phase II clinical trial to assess the efficacy of tamoxifen on progressive meningiomas secondary to resection or radiation treatment failure [48]. There were twenty-one patients enrolled in the study but two were immediately excluded after pathologist review of tissue, though they are still included in aggregate demographics (Table 2). Only one tumor (5%) was tested for estrogen receptor (isoform not specified) and it was negative (Table 3). No information regarding the histology, grade, or location of tumors was provided. In total, one patient achieved partial response (PR) (> 50%), two (11%) had minor response (unspecified definition) of short duration (4–20 months), six (32%) were stable, and ten (53%) demonstrated progression (unspecified definition) with an overall median time to progression of 15.1 months.
Table 3.
Tumor features of meningiomas in included clinical trials
| First author (year) | Drug | Histology* | Location* | Grade* | Receptors tested |
Confirmed receptor positive (% cohort) |
Receptor detection method |
|---|---|---|---|---|---|---|---|
| Markwalder (1985) [47] | Tamoxifen | 1 fibrous | 1 skull base; 1 parasagittal | Not specified | 1 | 0 | Not specified |
| Goodwin (1993) [48] | Tamoxifen | Not specified | Not specified | Not specified | 1 | 0 | Not specified |
| Grunberg (1990) [50] | Megestrol acetate | 6 meningothelial; 2 fibrous; 1 anaplastic | 8 skull base; 1 convexity | 8 WHO I; 1 WHO III | 2 | 0 | Not specified |
| Grunberg (2006) [51] | Mifepristone 14-day course of dexamethasone 1 mg | 13 meningothelial | 22 skull base; 4 spinal; 2 convexity | 13 WHO I;2 WHO III | 0 | 0 | NA |
| Touat (2014) [52] | Mifepristone | 1 transitional | 3 multifocal | 1 WHO I | 3 | 1 (33% cohort; 100% assessed) | IHC‡ |
| Ji (2015) [42] | Mifepristone | 17 atypical | Not specified | 17 WHO II | 84 | 75 (46% cohort; 89% assessed) | IHC‡ |
| Chamberlain (2007) [56] | Octreotide (Sandostatin LAR) | Not specified | 7 skull base; 6 convexity; 3 multifocal | 8 WHO I; 3 WHO II; 5 WHO III | 16 | 16 (100%) | Radioimaging |
| Norden (2015) [57] | Octreotide (pasireotide LAR) | Not specified | Not specified | 18 WHO I; 18 WHO II/III | 34 | 34 (100%) | IHC‡ |
| Hrachova(2020) [58] | Octreotide (sandostatin LAR) | 5 atypical 6 anaplastic | 23 skull base; 15 convexity; 5 mixed | 32 WHO I; 5 WHO II; 6 WHO III | 43 | 43 (100%) | Radioimaging |
Immunohistochemistry
The sum may not add up to total patients as information only given for tumors specified in studies, which frequently did not specify histology, location, and grade for every tumor
Megestrol acetate
Megestrol acetate is a progestin with a similar profile to CPA in terms of antiandrogenic and PGR agonist activity, but is typically used as a hunger stimulant [49]. In 1990, Grunberg et al. conducted a single-arm study on a prospective cohort consisting of nine patients with measurable or evaluable unresectable tissue-confirmed meningiomas with recurrence after prior surgery [50]. They reported that two (22%) meningiomas had PGR status assessed, which were both negative. Seven patients (78%) experienced progression, with four (44%) being removed from therapy within 2.5 months. Two (22%) patients remained stable and stopped treatment due to weight gain without tumor progression. There were no cases where megestrol acetate resulted in regression of the tumor by any observable amount, but recent reports of progestin therapy contributing to skull base tumor growth, and the lack of assessment of grade, proliferation index, and other features, should be considered as explanations for rapid progression in this trial.
Mifepristone
Mifepristone is an antagonist to glucocorticoid receptor (GR) and PGR and a weak androgen/AR agonist [53]. In vitro and in vivo studies revealed effectiveness, but the authors also noted that mifepristone was effective in PGR negative meningiomas, implying its possible action on other receptors that were not evaluated [54]. In 2006, Grunberg et al. conducted the first human prospective study using mifepristone for persistent or recurrent unresectable meningioma [51]. Twenty-four patients (86%) had undergone prior surgery and tissue confirmation of meningioma; whereas the other four patients were not operated on and diagnosed based on clinical and radiological evidence alone. No tumors were tested for PGR, GR, or AR status but radiographic evidence of tumor regression equaling about 5–10% was seen in five (18%) patients and an objective improvement in visual field examination without radiographic improvement was noted in three (11%) patients. In 2014, Touat, et al. published a case-series of three female patients with multiple-meningiomas and evaluated the long-term treatment with mifepristone [52]. One tumor was evaluated for PGR where it was found to be expressed at the protein level. All three patients saw reduction in tumor volume of 22–72% after a period of 5–10 years.
These initial findings prompted the initiation of a phase III randomized clinical trial of mifepristone that included one-hundred and sixty four patients with eighty in the placebo and eighty-four in the mifepristone arms [42]. Patients with pathology confirmed meningiomas demonstrating radiotherapy treatment failure defined by radiographic evidence of recurrence or progression after surgical resection, were eligible for the trial and were randomly assigned to either oral mifepristone 200 mg daily or placebo for 2 years. Notably, this study reported that 32% of patients had non-measurable—but evaluable—meningiomas at baseline, which others have pointed out as being unlikely to see any improvement compared to baseline [55]. While 52% of tumors were tested for PGR, nearly 12% of those tumors were found to be PGR negative and the remaining 48% were not evaluated. Of the 164 patients who began the trial, 30% of the mifepristone arm and 33% of the placebo arm completed the 2 years of treatment as planned. The median failure free survival (FFS) for placebo was 11 months CI95%[6–18] and 10 months CI95%[7–13], (p = 0.9) adjusted for sex, prior radiotherapy and disease status [HR 1.02, CI95%(0.72–1.48)]. One patient each in the placebo and mifepristone trials achieved partial response (PR). Histology (aside from 10% atypical), grade, and location were not specified. The study intended to recruit 200 patients but did not achieve this. In combination with the inclusion of patients unlikely to show radiographic response (non-measurable tumors) and receptor negative meningiomas, along with missing relevant information, the study is likely to be more underpowered than originally calculated with limited interpretation of results. Thus, a future clinical trial with more specific patient inclusion criteria and documented tumor characteristics may be considered.
Somatostatin
In vitro studies of somatostatin showed efficacy, but the author of at least one highly cited study noted that one of the four tumors demonstrating growth inhibition was devoid of any SSTR expression [59]. A recent systematic review with meta-analysis of individual patient data was published that obtained complete datasets for 7 of 8 clinical trials assessing somatostatin as a monotherapy and combined therapy in meningioma [60]. When using somatostatin as a monotherapy for disease control, the meta-analysis found an odds ratio of 1.42 CI95%[1.11–1.81, p = 0.005] for each 100 mg increase in total somatostatin. This effect for disease control was most pronounced in WHO I tumors, with the relative odds ratio for WHO II and WHO III tumors decreasing to 0.31 CI95%[0.09–1.05, p = 0.06] and 0.08 CI95%[0.02–0.33, p < 0.0001]. The authors note that their meta-analysis was compromised by poor quality of evidence due to different treatment regimens and heterogeneity between patients within trials and state they could not conclusively support or disqualify somatostatin as a treatment for meningioma.
Several combination therapies with octreotide have been attempted including with everolimus, an mTOR inhibitor, and use of radiolabeled somatostatin analogs 90Y-DOTATOC, 177Lu-DOTATOC, 177Lu-DOTATATE. Studies generated in vitro suggested that everolimus has a synergistic effect with octreotide [61]. This led to two clinical trials including a total of 34 patients, to study the combination of octreotide and everolimus for disease control [62, 63]. These studies were combined in the aforementioned meta-analysis, which found an odds ratio for disease control of 1.44, CI95%[1.00–2.08, p = 0.05] [60]. Seperately, an individual patient data meta-analysis of radiolabeled somatostatin analogs for radiopeptide therapy synthesized 6 cohort studies including 111 patients and found a PFS6 of 94% for WHO I, 48% for WHO II, and 0% for WHO III [64]. The potential benefit of radiopeptide therapy over conventional radiation therapy is the ability to target tumors more precisely; however, the side effect profile is similar to other forms of radiation treatment.
Nearly all studies required SSTR positive status but showed inconsistent efficacy; however, most studies included only somatostatin-radioligand detection of SSTR activity in the tumor and not a specific assay of the SSTR present (by IHC or another receptor-specific method). Future studies could investigate whether all SSTRs are functional in meningioma, or whether the receptors result in differences in treatment efficacy.
Conclusion
In this review of hormone-related clinical trials in meningioma, it is apparent that a disparity in the assessment of receptors in these tumors indeed exists between trials that employed gonadal steroid hormones versus somatostatin analogs. Understandably, the need for receptor status obtained by tissue confirmation complicates clinical trials that primarily includes unresectable tumors. However, given that the mechanism of action for most hormone therapy requires binding to specific receptors, it is imperative to assess that status before treating patients. Further complicating results in all trials are the small number of patients included and the lack of stratification based on grade and mutation status, which was not known when many of these studies were conducted, but influence recurrence of the tumors even within the same grade [14]. The use of radiolabeled somatostatin greatly enhanced clinicians to assess receptor status non-invasively; however, it does not provide specific receptor status of SSTR1-5, which may have influenced treatment outcome. Almost all current recruiting clinical trials are radiopeptide trials of somatostatin analogs with the exception of an observational study of meningiomas in a group of patients being treated with CPA (NCT04372095, Table 4), but there are no ongoing trials investigating other hormone-based therapy. Combination therapy appears to be a promising avenue, but given meningioma heterogeneity, more work should be done to evaluate what particular features—such as grade, receptor status, and somatic mutations—might predict an efficacious response in monotherapy, which can be used to further employ strategic combination therapy.
Future directions
There is significant heterogeneity between treatment groups included between trials which results in overall poor quality of evidence. Because of this heterogeneity, along with the slow-growing nature of meningiomas, a recent effort has been initiated to strive for an agreed upon set of standard outcomes for studies assessing progression-free survival, referred to as the ‘Core Outcome Sets’ for Meningioma in Clinical Studies (The COSMIC Project). The authors of this project also note the need for global and multi-institutional collaborations for clinical trials in order to increase the participation of patients per study [65].
A major challenge in meningioma research is the relative lack of adequate meningioma models, with multiple reports of tumors losing features important to tumor identity upon cell culture and even in vivo modeling [66]. Considering this limitation, the use of radioligands have been beneficial in the non-invasive identification of tumors that express SSTRs, but that same technique has not been applied to other receptors in meningioma on a large scale. In 1997, a small study including 6 patients with meningioma successfully used PET imaging with Fluoroestradiol (F18-FES), a radioligand of estradiol. They demonstrated acceptable radioligand uptake and a correlation between the ligand uptake and immunohistochemical expression of an unspecified estrogen receptor [67]. As of 2020, F18-FES is FDA approved for metastatic breast cancer. There also exists radioligand progestin 21-[18F]fluorofuranyl-norprogesterone and testosterone derivatives 16β-[18F] fluoro-5α-dihydrotestosterone, both of which are in earlier phases of study in metastatic breast cancer [68, 69]. The use of these agents in studying meningioma could help clarify the role of gonadal steroid hormones in meningiomas and be used for selecting patients for treatment; however, confirmation of ligand uptake is not equivalent to receptor isoform elucidation important for predicting biological effects, which needs further study. Historic work on the matter of gonadal steroid hormones in meningioma should be evaluated in this context of poor receptor detection and with the understanding that the gonadal-steroid hormone and IGF-1/GH signaling axes are not independent features, but rather, parts of a shared regulatory axis. Similarly, the meningioma microenvironment investigating immune cell infiltrate has gained attention and recent years with successful clinical trials targeting PD-L1 [70]. Understanding more about how these pathways intersect and influence the tumor microenvironment has promise for precision medicine and uncovering biological mechanisms of meningioma tumorigenesis. The synthesis of clinical, pharmacological, genomic, and molecular data in meningiomas may help select the patients who will benefit most from various targeted therapies.
Funding
This study was supported by the Gregory M. Kiez and Mehmet Kutman Foundation, Connecticut Brain Tumor Alliance, and Yale School of Medicine funds, NIH/NCI (No.F31CA254426) & NIH-Medical Scientist Training Program (No.T32GM007205) R01 Grants CA109468, CA109461, CA109745, CA109473, CA052689, and CA151933.
Footnotes
Competing interests The authors declare no competing interests.
References
- 1.Claus EB et al. (2013) Exogenous hormone use, reproductive factors, and risk of intracranial meningioma in females: clinical article. J Neurosurg 118(3):649–656 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Fodi C et al. (2021) The immunohistochemical expression of SSTR2A is an independent prognostic factor in meningioma. Neurosurg Rev 45:2671–2679 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Behling F et al. (2022) Differences in the expression of SSTR1–5 in meningiomas and its therapeutic potential. Neurosurg Rev 45(1):467–478 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Donnell MS, Meyer GA, Donegan WL (1979) Estrogen-receptor protein in intracranial meningiomas. J Neurosurg 50(4):499–502 [DOI] [PubMed] [Google Scholar]
- 5.Markwalder T-M et al. (1983) Estrogen and progesterone receptors in meningiomas in relation to clinical and pathologic features. Surg Neurol 20(1):42–47 [DOI] [PubMed] [Google Scholar]
- 6.Gil M et al. (2011) Risk of meningioma among users of high doses of cyproterone acetate as compared with the general population: evidence from a population-based cohort study. Br J Clin Pharmacol 72(6):965–968 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lee KS et al. (2022) A systematic review and meta-analysis of the association between cyproterone acetate and intracranial meningiomas. Sci Rep 12(1):1942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Peyre M et al. (2018) Progestin-associated shift of meningioma mutational landscape. Ann Oncol 29(3):681–686 [DOI] [PubMed] [Google Scholar]
- 9.Samarut E et al. (2021) Meningiomas and cyproterone acetate: a retrospective, monocentric cohort of 388 patients treated by surgery or radiotherapy for intracranial meningioma. J Neuro-Oncol 152(1):115–123 [DOI] [PubMed] [Google Scholar]
- 10.Cushing H (1938) Meningiomas: their classification, regional behavior, life history, and surgical end result. Springfield Charles C Thomas 111:735 [Google Scholar]
- 11.Wiemels J, Wrensch M, Claus EB (2010) Epidemiology and etiology of meningioma. J Neuro-Oncol 99(3):307–314 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Brinton RD et al. (2015) Perimenopause as a neurological transition state. Nat Rev Endocrinol 11(7):393–405 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Youngblood MW et al. (2019) Correlations between genomic subgroup and clinical features in a cohort of more than 3000 meningiomas. J Neurosurg 133(5):1345–1354 [DOI] [PubMed] [Google Scholar]
- 14.Youngblood MW et al. (2021) Associations of meningioma molecular subgroup and tumor recurrence. Neuro-Oncology 23(5):783–794 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Apra C et al. (2020) Female gender and exogenous progesterone exposition as risk factors for spheno-orbital meningiomas. J Neuro-Oncol 149(1):95–101 [DOI] [PubMed] [Google Scholar]
- 16.Meling TR et al. (2019) Meningiomas: skull base versus non-skull base. Neurosurg Rev 42(1):163–173 [DOI] [PubMed] [Google Scholar]
- 17.Pier DB et al. (2014) Turner syndrome and meningioma: support for a possible increased risk of neoplasia in Turner syndrome. Eur J Med Genet 57(6):269–274 [DOI] [PubMed] [Google Scholar]
- 18.Schoemaker MJ et al. (2008) Cancer incidence in women with Turner syndrome in Great Britain: a national cohort study. Lancet Oncol 9(3):239–246 [DOI] [PubMed] [Google Scholar]
- 19.Claus EB et al. (2011) Family and personal medical history and risk of meningioma: clinical article. J Neurosurg 115(6):1072–1077 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Portet S et al. (2019) Histomolecular characterization of intracranial meningiomas developed in patients exposed to high-dose cyproterone acetate: an antiandrogen treatment. Neuro-Oncol Adv. 10.1093/noajnl/vdz003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Passeri T et al. (2022) Atypical evolution of meningiomatosis after discontinuation of cyproterone acetate: clinical cases and histomolecular characterization. Acta Neurochir 164(1):255–263 [DOI] [PubMed] [Google Scholar]
- 22.Dresser L et al. (2020) Estrogen hormone replacement therapy in incidental intracranial meningioma: a growth-rate analysis. Sci Rep 10(1):1–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.FDA (2021) FDA listing of established pharmacologic class text phrases October 2021. https://www.fda.gov/media/156478/download
- 24.Carroll RS, Zhang J, Black PM (1999) Expression of estrogen receptors alpha and beta in human meningiomas. J Neuro-Oncol 42(2):109–116 [DOI] [PubMed] [Google Scholar]
- 25.Martinkovich S et al. (2014) Selective estrogen receptor modulators: tissue specificity and clinical utility. Clin Interv Aging 9:1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Andersson S et al. (2017) Insufficient antibody validation challenges oestrogen receptor beta research. Nat Commun 8(1):1–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Revankar CM et al. (2005) A transmembrane intracellular estrogen receptor mediates rapid cell signaling. Science 307(5715):1625–1630 [DOI] [PubMed] [Google Scholar]
- 28.Prossnitz ER, Arterburn JB (2015) International union of basic and clinical pharmacology. XCVII. G protein-coupled estrogen receptor and its pharmacologic modulators. Pharmacol Rev 67(3):505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.van Leeuwen FE et al. (1994) Risk of endometrial cancer after tamoxifen treatment of breast cancer. Lancet 343(8895):448–452 [DOI] [PubMed] [Google Scholar]
- 30.Hua L et al. (2017) Prognostic value of estrogen receptor in WHO Grade III meningioma: a long-term follow-up study from a single institution. J Neurosurg 128(6):1698–1706 [DOI] [PubMed] [Google Scholar]
- 31.Pravdenkova S et al. (2006) Progesterone and estrogen receptors: opposing prognostic indicators in meningiomas. J Neurosurg 105(2):163–173 [DOI] [PubMed] [Google Scholar]
- 32.Claus EB et al. (2008) Specific genes expressed in association with progesterone receptors in meningioma. Can Res 68(1):314–322 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Korhonen K et al. (2006) Female predominance in meningiomas can not be explained by differences in progesterone, estrogen, or androgen receptor expression. J Neuro-Oncol 80(1):1–7 [DOI] [PubMed] [Google Scholar]
- 34.Li Q et al. (2013) Emerging association between androgen deprivation therapy and male meningioma: significant expression of luteinizing hormone-releasing hormone receptor in male meningioma. Prostate Cancer Prostatic Dis 16(4):387–390 [DOI] [PubMed] [Google Scholar]
- 35.Lee KL, Terris MK (2003) Luteinizing hormone-releasing hormone agonists and meningioma: a treatment dilemma. Urology 62(2):351. [DOI] [PubMed] [Google Scholar]
- 36.Leung K-C et al. (2004) Estrogen regulation of growth hormone action. Endocr Rev 25(5):693–721 [DOI] [PubMed] [Google Scholar]
- 37.Kumar U et al. (2005) Somatostatin receptors in primary human breast cancer: quantitative analysis of mRNA for subtypes 1–5 and correlation with receptor protein expression and tumor pathology. Breast Cancer Res Treat 92(2):175–186 [DOI] [PubMed] [Google Scholar]
- 38.Rossi V et al. (2019) Estrogens modulate somatostatin receptors expression and synergize with the somatostatin analog pasireotide in prostate cells. Front Pharmacol 10:28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Rutkowski R et al. (2016) Correlation of leptin receptor expression with BMI in differential grades of human meningiomas. Oncol Lett 11(4):2515–2519 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Rogol AD (2010) Sex steroids, growth hormone, leptin and the pubertal growth spurt. Pediatr Neuroendocrinol 17:77–85 [DOI] [PubMed] [Google Scholar]
- 41.Di Carlo C, Tommaselli GA, Nappi C (2002) Effects of sex steroid hormones and menopause on serum leptin concentrations. Gynecol Endocrinol 16(6):479–491 [PubMed] [Google Scholar]
- 42.Ji Y et al. (2015) Double-blind phase III randomized trial of the antiprogestin agent mifepristone in the treatment of unresectable meningioma: SWOG S9005. J Clin Oncol 33(34):4093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Jay JR et al. (1985) Modulation of meningioma cell growth by sex steroid hormones in vitro. J Neurosurg 62(5):757–762 [DOI] [PubMed] [Google Scholar]
- 44.Olson JJ et al. (1986) Hormonal manipulation of meningiomas in vitro. J Neurosurg 65(1):99–107 [DOI] [PubMed] [Google Scholar]
- 45.Ji J, Sundquist J, Sundquist K (2016) Association of tamoxifen with meningioma: a population-based study in Sweden. Eur J Cancer Prev 25(1):29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sun L-M et al. (2019) Long-term use of tamoxifen is associated with a decreased subsequent meningioma risk in patients with breast cancer: a nationwide population-based cohort study. Front Pharmacol 10:674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Markwalder T-M, Seiler RW, Zava DT (1985) Antiestrogenic therapy of meningiomas—a pilot study. Surg Neurol 24(3):245–249 [DOI] [PubMed] [Google Scholar]
- 48.Goodwin JW et al. (1993) A phase II evaluation of tamoxifen in unresectable or refractory meningiomas: a Southwest Oncology Group study. J Neuro-Oncol 15(1):75–77 [DOI] [PubMed] [Google Scholar]
- 49.Schindler AE et al. (2008) Reprint of classification and pharmacology of progestins. Maturitas 61(1):171–180 [DOI] [PubMed] [Google Scholar]
- 50.Grunberg SM, Weiss MH (1990) Lack of efficacy of megestrol acetate in the treatment of unresectable meningioma. J Neuro-Oncol 8(1):61–65 [DOI] [PubMed] [Google Scholar]
- 51.Grunberg SM et al. (2006) Long-term administration of mifepristone (RU486): clinical tolerance during extended treatment of meningioma. Cancer Investig 24(8):727–733 [DOI] [PubMed] [Google Scholar]
- 52.Touat M et al. (2014) Successful treatment of multiple intracranial meningiomas with the antiprogesterone receptor agent mifepristone (RU486). Acta Neurochir 156(10):1831–1835 [DOI] [PubMed] [Google Scholar]
- 53.Sitruk-Ware R, Spitz IM (2003) Pharmacological properties of mifepristone: toxicology and safety in animal and human studies. Contraception 68(6):409–420 [DOI] [PubMed] [Google Scholar]
- 54.Matsuda Y et al. (1994) Antitumor effects of antiprogesterones on human meningioma cells in vitro and in vivo: case report. J Neurosurg 80(3):527–534 [DOI] [PubMed] [Google Scholar]
- 55.Chamberlain MC (2016) What lessons are imparted from SWOG S9005 for recurrent meningioma? J Clin Oncol 34(15):1825–1826 [DOI] [PubMed] [Google Scholar]
- 56.Chamberlain MC, Glantz MJ, Fadul CE (2007) Recurrent meningioma: salvage therapy with long-acting somatostatin analogue. Neurology 69(10):969–973 [DOI] [PubMed] [Google Scholar]
- 57.Norden AD et al. (2015) Phase II study of monthly pasireotide LAR (SOM230C) for recurrent or progressive meningioma. Neurology 84(3):280–286 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Hrachova M et al. (2020) A retrospective interventional cohort study to assess the safety and efficacy of sandostatin LAR for treatment of recurrent and/or refractory meningiomas. Front Neurol. 10.3389/fneur.2020.00373 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Arena S et al. (2004) Expression of somatostatin receptor mRNA in human meningiomas and their implication in in vitro antiproliferative activity. J Neuro-Oncol 66(1):155–166 [DOI] [PubMed] [Google Scholar]
- 60.Jensen LR et al. (2022) Somatostatin analogues in treatment-refractory meningioma: a systematic review with meta-analysis of individual patient data. Neurosurg Rev 45(5):3067–3081 [DOI] [PubMed] [Google Scholar]
- 61.Graillon T et al. (2015) Combined treatment by octreotide and everolimus: octreotide enhances inhibitory effect of everolimus in aggressive meningiomas. J Neuro-Oncol 124(1):33–43 [DOI] [PubMed] [Google Scholar]
- 62.Cardona AF et al. (2019) Systemic management of malignant meningiomas: a comparative survival and molecular marker analysis between octreotide in combination with everolimus and sunitinib. PLoS ONE 14(6):e0217340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Graillon T et al. (2020) Everolimus and octreotide for patients with recurrent meningioma: results from the phase II CEVOREM trial. Clin Cancer Res 26(3):552–557 [DOI] [PubMed] [Google Scholar]
- 64.Mirian C et al. (2021) Somatostatin receptor-targeted radiopeptide therapy in treatment-refractory meningioma: individual patient data meta-analysis. J Nucl Med 62(4):507. [DOI] [PubMed] [Google Scholar]
- 65.Millward CP et al. (2022) Development of ‘Core Outcome Sets’ for Meningioma in Clinical Studies (The COSMIC Project): protocol for two systematic literature reviews, eDelphi surveys and online consensus meetings. BMJ Open 12(5):e057384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Choudhury A, Raleigh DR (2020) Chapter 8—preclinical models of meningioma: cell culture and animal systems. In: McDermott MW (ed) Handbook of clinical neurology. Elsevier, Amsterdam, pp 131–136 [DOI] [PubMed] [Google Scholar]
- 67.Moresco RM et al. (1997) Oestrogen receptors in meningiomas: a correlative PET and immunohistochemical study. Nucl Med Commun 18(7):606–615 [DOI] [PubMed] [Google Scholar]
- 68.Dehdashti F et al. (2021) Association of PET-based estradiol-challenge test for breast cancer progesterone receptors with response to endocrine therapy. Nat Commun 12(1):733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Boers J et al. (2021) Serial [(18)F]-FDHT-PET to predict bicalutamide efficacy in patients with androgen receptor positive metastatic breast cancer. Eur J Cancer 144:151–161 [DOI] [PubMed] [Google Scholar]
- 70.Brastianos PK et al. (2022) Phase 2 study of pembrolizumab in patients with recurrent and residual high-grade meningiomas. Nat Commun 13(1):1325. [DOI] [PMC free article] [PubMed] [Google Scholar]

