Skip to main content
Journal of the Endocrine Society logoLink to Journal of the Endocrine Society
. 2022 Dec 31;7(3):bvac197. doi: 10.1210/jendso/bvac197

Nuclear Receptor Imaging In Vivo—Clinical and Research Advances

Ephraim E Parent 1, Amy M Fowler 2,3,4,
PMCID: PMC9838808  PMID: 36655003

Abstract

Nuclear receptors are transcription factors that function in normal physiology and play important roles in diseases such as cancer, inflammation, and diabetes. Noninvasive imaging of nuclear receptors can be achieved using radiolabeled ligands and positron emission tomography (PET). This quantitative imaging approach can be viewed as an in vivo equivalent of the classic radioligand binding assay. A main clinical application of nuclear receptor imaging in oncology is to identify metastatic sites expressing nuclear receptors that are targets for approved drug therapies and are capable of binding ligands to improve treatment decision-making. Research applications of nuclear receptor imaging include novel synthetic ligand and drug development by quantifying target drug engagement with the receptor for optimal therapeutic drug dosing and for fundamental research into nuclear receptor function in cells and animal models. This mini-review provides an overview of PET imaging of nuclear receptors with a focus on radioligands for estrogen receptor, progesterone receptor, and androgen receptor and their use in breast and prostate cancer.

Keywords: estrogen receptor, progesterone receptor, androgen receptor, cancer, positron emission tomography


Nuclear receptors are transcription factors that regulate gene expression in normal development, reproduction, metabolism, and homeostasis. These proteins are also key regulators in cancer, inflammation, diabetes, and metabolic disease [1]. The nuclear receptor superfamily consists of 48 proteins in humans, which share a conserved functional domain organization including a ligand-binding domain, DNA-binding domain, amino-terminal region, and hinge region [2].

Nuclear receptors are important targets for therapy, with approximately 13% of US Food and Drug Administration (FDA)-approved drugs targeting nuclear receptors [3, 4]. In oncology, ligands targeting the estrogen receptor (ER) and androgen receptor (AR) are routinely used in the clinical treatment of breast cancer and prostate cancer, respectively. Additionally, molecular imaging of nuclear receptors can be a noninvasive way to identify whether the target for therapy is present to help guide optimal use of targeted therapies.

Positron emission tomography (PET) radiopharmaceuticals used for imaging nuclear receptors are small molecules labeled with a positron emitting radioisotope such as 18F. Due to the intracellular localization of nuclear receptors, radiolabeled antibodies are not effective. After intravenous injection, the PET radiopharmaceutical distributes throughout the body and accumulates in organs and tumors expressing the targeted nuclear receptor. PET scanners coupled with computed tomography (PET/CT) or magnetic resonance imaging (PET/MRI) provide visual localization and quantitative information regarding receptor binding and occupancy. Steroidal PET radiopharmaceuticals typically demonstrate nanomolar to subnanomolar binding affinity for their respective steroid receptor, produce high-quality images using subpharmacologic mass doses, and thus do not functionally activate or inhibit the targeted nuclear receptor with minimal adverse events compared to conventional intravenous contrast agents used for CT and MRI [5].

Several targeted and selective PET imaging radiopharmaceuticals for nuclear receptors have been developed with various preclinical and clinical applications (Fig. 1). Potential uses span from basic and translational research to clinical use with recent approval by the FDA for an ER PET radiopharmaceutical, 16α-[18F]fluoro-17β-estradiol (FES). This article reviews PET imaging of nuclear receptors with a focus on recent ER, progesterone receptor (PR), and AR imaging clinical trials and a brief review of other less well-studied nuclear receptor targets. The rationale for development and significance for clinical use and for quantitative tools in fundamental research into nuclear receptor function will be discussed. For additional details regarding the development of PET imaging agents for steroid hormone receptors in breast and prostate cancer, readers are referred to this comprehensive review [6]. It should be noted that while FES has received FDA approval for clinical use, all other steroid ligand radiopharmaceuticals mentioned in this mini-review are for investigational purposes only.

Figure 1.

Figure 1.

Development of positron emission tomography (PET) radiopharmaceuticals targeting nuclear receptors from basic preclinical research culminating in US Food and Drug Administration (FDA) approval. 16α-[18F]fluoro-17β-estradiol (FES) has been approved for clinical use in patients with estrogen receptor–positive breast cancer. 21-18F-Fluorofuranylnorprogesterone (FFNP) and 16β-18F-fluoro-5α-dihydrotestosterone (FDHT) have both reached clinical trials for patients with breast cancer and prostate cancer, respectively. 11C-YJH08 is a new radioligand specific to glucocorticoid receptor (not shown) with an ongoing first-in-human study in patients with prostate cancer. Various PET radiopharmaceuticals (not shown) targeting other nuclear receptors (PPARα, PPARγ, VDR, RXR, FXR) have been developed and tested in preclinical research.

Estrogen Receptor

Breast cancer is the most common malignancy in women, excluding nonmelanoma skin cancer. The incidence of breast cancer has risen over the past several decades, primarily due to increasing local-stage and hormone receptor–positive disease with 80% of invasive breast cancers being ER positive (ER+) [7]. Breast cancer is the second leading cause of death for women after lung cancer and is the leading cause of death in Black and Hispanic women [8].

2-Deoxy-2-[18F]fluoro-D-glucose (FDG) is clinically the most commonly employed molecular imaging agent to identify local and metastatic breast cancer, and to evaluate response to therapy of patients with metastatic breast cancer [9]. FDG is a glucose analogue that undergoes transport across the cell membrane by glucose transporters (GLUT) and becomes phosphorylated by hexokinase as the first step in the glycolytic pathway, effectively trapping it within the cell. Many breast cancers have high reliance on glucose metabolism (Warburg effect) resulting in high FDG uptake [10-13]. However, there are limitations for FDG PET in the evaluation of breast cancer, namely that 1) some breast malignancies, such as invasive lobular breast carcinoma, have notoriously low FDG uptake [14] and 2) FDG is unable to identify functional hormone receptor status [15]. FDG uptake is positively associated with tumor grade and Ki-67 proliferation index, and is inversely proportional to ER status [16], affording an opportunity for improved lesion detection and signal-to-noise with ER imaging [17]. A meta-analysis of 7 studies including 171 patients compared the diagnostic accuracy of FES and FDG PET/CT for ER+ breast cancer and found that FES was more sensitive than FDG for lesion-level detection at the time of disease relapse with a trend toward statistical significance [18].

FES was the first nuclear receptor imaging agent approved by the FDA for clinical use in 2020 “with PET imaging for the detection of ER+ lesions as an adjunct to biopsy in patients with recurrent or metastatic breast cancer” [19]. FES was developed in the 1980s at the University of Illinois [20] and was the first receptor-based radiopharmaceutical to be successfully evaluated in humans [6, 21]. There are 2 subtypes of ER, ERα and ERβ, both of which function as ligand-dependent transcription factors. While ERβ is expressed in physiologic breast, ovarian, and prostate tissues, ERα is more commonly overexpressed in ER+ malignancies, with ERβ levels decreasing as the cancer progresses [22]. Although FES has high, nanomolar-range binding affinity both for ERα and ERβ, FES has a 6.3-fold preferential affinity for ERα [23]. ERβ-specific radiopharmaceuticals have been developed but have not yet been evaluated in human studies [23-26]. To the best of our knowledge, there have been no publications investigating whether FES recognizes membrane-bound ER.

FES is an ER PET radiopharmaceutical, and ERα expression has been shown to correlate with clinical response to hormonal treatment in several malignancies including breast cancer and endometrial cancer [27, 28]. FES uptake requires an intact ER ligand-binding pocket and thus detects ER that is available and capable of binding ligand [29]. However, FES uptake is not affected by common clinically important activating mutations in ESR1, the gene coding for the ERα protein [30, 31]. While FES PET can be performed regardless of diet or allergies to iodinated contrast, it is important to note that patients should be imaged before starting therapies with selective ER modulators (tamoxifen) and selective ER downregulators (fulvestrant), which bind to the estrogen-binding pocket of the receptor. Tamoxifen and fulvestrant can block ER for up to 8 weeks and 28 weeks, respectively, and can cause false-negative results.

Following FDA approval, FES has become generally available for integration into clinical practice [17, 32]. According to the recently released Appropriate Use Criteria from the Society of Nuclear Medicine and Molecular Imaging, clinical scenarios for breast cancer for which FES PET imaging is appropriate include (1) detecting ER status when other imaging tests are equivocal or suspicious, (2) assessing ER status in lesions that are difficult to biopsy or when biopsy results are nondiagnostic, (3) after progression of ER+ metastatic disease for considering second-line endocrine therapy, and (4) at the initial diagnosis of ER+ metastatic disease for considering first-line endocrine therapy [33]. FES can be useful as a diagnostic problem-solving tool, for instance when there is clinical suspicion of disease recurrence or progression despite negative conventional imaging to identify occult ER+ metastatic breast cancer (Fig. 2) or to determine the distribution of ER+ disease in patients with a combination of metastatic breast cancer and a separate non–ER-expressing malignancy [34-37]. Although FDG PET can be used to monitor response to therapy, FES PET is not recommended for this clinical scenario [33]. Given the extensive molecular imaging trials of FES dating back to the 1980s, a complete description of the clinical studies using FES in the evaluation of breast cancer is beyond the scope of this mini-review but can be further explored in other recent reviews [38]. Selected points involving recent studies in the past few years are highlighted next.

Figure 2.

Figure 2.

Restaging 16α-[18F]fluoro-17β-estradiol (FES) positron emission tomography/computed tomography (PET/CT) imaging of a woman with estrogen receptor–positive (ER+) breast cancer. A woman with a history of strongly ER+ right breast adenocarcinoma treated with bilateral mastectomy and adjuvant endocrine therapy presented clinically with continually increasing tumor markers and CA 15-3 of 121 U/mL. Images shown from restaging FDG (2-deoxy-2-[18F]fluoro-D-glucose) PET/CT including A, maximum intensity projection (MIP) and selected transaxial fused FDG B, PET/CT; C, CT; and D, FDG PET was negative for local or metastatic disease (arrows), as was bone scintigraphy (not shown). Abdominal and pelvis magnetic resonance imaging (MRI) scan was also interpreted as negative for metastatic disease with an enhancing lesion in the left iliac bone as seen on E, T1+ contrast; F, out of phase; and G, in phase being interpreted as red marrow. FES PET/CT was conducted for definitive staging with H, FES MIP demonstrating several FES-avid osseous lesions (arrows) and transaxial fused FES I, PET/CT; J, CT; and K, FES PET identifying ER+ metastatic disease in the left iliac bone and no CT correlate (arrows). Subsequent biopsy of the left iliac FES-positive lesion was consistent with ER+ breast metastatic disease.

Currently, ER expression in primary and metastatic breast cancer is determined by immunohistochemistry [39]. However, biopsy may lead to sampling errors and may not be feasible because of its invasive nature or the location of the metastatic lesion. Also, heterogeneity in ER expression between the primary and metastatic lesions result in discrepancies in ER expression in 16% to 40% of patients [39, 40], and poor diagnostic and therapeutic outcomes can be seen when treatment-management decisions are based solely on primary lesion immunohistochemistry [41]. A high correlation has been found between FES uptake and immunohistochemistry findings for determination of ER status for metastatic disease [42]. To demonstrate the utility that FES has to provide whole-body, real-time interrogation of ER+ metastatic disease, a recent study combined primary data and a meta-analysis of 556 patients who underwent FES PET to identify ER+ status in metastatic disease and found an excellent positive predictive value and negative predictive value of 93% and 85%, respectively [43].

Additionally, tumor ER expression may change over time, especially in the setting of ER-targeted therapies leading to clinical dilemmas regarding both the correct diagnosis and the best choice of therapy. FES has been shown to accurately estimate ER expression of all tumor lesions and can be reliably used for individualized therapy decision-making [44]. A retrospective study of 56 patients with known ER+ metastatic breast cancer who underwent FES PET/CT before combined endocrine and palbociclib (cyclin-dependent kinase 4/6-inhibitor) therapy found that patients with only FES-positive lesions had a substantially longer progression-free survival compared to those ER+ breast cancer patients who had at least one FES-negative lesion (23.6 months compared to 2.4 months) [45]. Similarly, a retrospective study of 75 patients with ER+ metastatic breast cancer found that patients with ER heterogeneity (both FES-positive and FES-negative lesions) responded better to chemotherapy than endocrine therapy, and did not improve with combined chemotherapy and endocrine therapy [46]. The utility of FES to guide clinical management can be especially useful in patients who have both ER+ breast cancer and at least one other primary malignancy. A retrospective analysis of 83 patients with conventional imaging findings indeterminate for ER+ metastatic disease found that 87% of patients had their clinical dilemma solved by FES PET [34].

In addition to breast cancer, the role of FES PET in other known ER-dependent malignancies, such as endometrial and ovarian cancers, has been evaluated in clinical trials. Endometrial cancers are classified into type I and type II tumors. Type I, including endometrioid adenocarcinomas, are estrogen dependent, and are often preceded by endometrial hyperplasia. Type II tumors, including serous or clear cell carcinomas, are commonly estrogen independent, are less well differentiated, and have poorer prognoses. Several studies have used FES PET in combination with FDG to characterize endometrial carcinomas [47, 48]. A prospective study of 67 patients with endometrial carcinoma demonstrated that low FES uptake in the primary tumor is strongly associated with adverse prognostic factors, and that FES uptake is an independent prognostic factor for progression-free survival [49]. Among other factors, ERα loss is associated with lymphovascular space involvement [28] and metastatic spread in part because angiogenesis and other steps required for metastatic progression are modulated by sex-steroid hormones [50]. There are few studies reporting the efficacy of FES PET in the evaluation of ovarian carcinoma [51]. One interesting study found that, in patients with metastatic disease, FES PET correlated with histology at the time of debulking, but not at primary diagnosis, suggesting some degree of transformation in metastatic disease [52]. As with other molecular imaging techniques, such as with FDG PET, ovarian metastatic disease evaluation with FES PET is limited in cystic lesions and requires a large solid component for accurate quantitative measurement [53].

Progesterone Receptor

Transcription of the gene encoding progesterone receptor (PGR) is controlled by ER with increased expression in response to estrogen stimulation. As with ER, PR protein expression is determined by tissue immunohistochemistry and is routinely used clinically as a tumor biomarker in breast cancer [54-58]. Measuring changes in PR protein expression in metastatic breast cancer can serve as an indicator of ER functional activity and hormonal responsiveness [57, 59, 60]. Additionally, there is growing evidence indicating a more direct role of PR in breast cancer biology and crosstalk with ER [61-63], which has fueled clinical trials testing new antiprogestin therapeutics selectively targeting PR [64-67]. Thus, PR imaging may be useful as an early-response biomarker for ER-targeted endocrine therapy response and as a potential predictive biomarker for PR-targeted therapies.

21-[18F]Fluorofuranylnorprogesterone (FFNP) is the most studied PR-targeted radioligand in preclinical [68-74] and clinical research [75, 76] (Fig. 3). Also developed at the University of Illinois, FFNP has high binding affinity for PR [68, 69]. Furthermore, FFNP has comparable binding both to PR-A and PR-B isoforms, the 2 main isoforms expressed in breast cancer [72, 73, 77]. To the best of our knowledge, there have been no publications investigating whether FFNP recognizes membrane-bound PR.

Figure 3.

Figure 3.

21-18F-Fluorofuranylnorprogesterone (FFNP) positron emission tomography/computed tomography (PET/CT) imaging of a woman with biopsy-proven progesterone receptor–positive (PR+) breast cancer and axillary lymph node metastasis. FFNP PET/CT images (sagittal view) of a postmenopausal woman with estrogen receptor–positive (ER+)/PR+/HER2– invasive lobular carcinoma show radioligand uptake in the biopsy-proven malignancy involving the upper left breast (arrow) and multiple left level 1 axillary lymph nodes (LN arrow). The patient was imaged under a research protocol.

Preclinical studies have shown how FFNP imaging can measure real-time changes in PR expression as an indicator of ER functionality and endocrine therapy sensitivity in hormone receptor–positive breast cancer [70, 71, 73, 74]. The stimulatory effect of estradiol on PR expression in T47D human breast cancer cells and tumor xenografts was evident by increased FFNP uptake within 1 to 2 days of treatment [73]. Conversely, the inhibitory effect of estrogen deprivation via ovariectomy or ER inhibition via fulvestrant treatment on PR expression in STAT1-deficient mouse mammary tumors was also demonstrated by decreased FFNP uptake within 3 to 4 days [70, 71]. This imaging phenotype occurred only for endocrine-sensitive tumors, whereas endocrine-resistant tumors showed no significant changes in FFNP uptake [70, 71]. Furthermore, early assessment of PR expression dynamics using FFNP PET predicted inadequate tumor growth inhibition with endocrine therapy in xenografts expressing activating mutations of the ESR1 gene, another mechanism of endocrine resistance [74]. Together these studies demonstrated that imaging the molecular changes in the expression of a downstream estrogen-regulated target gene can serve as a surrogate measure of endocrine sensitivity within a few days of treatment before anatomic changes in tumor size can be measured.

Results from clinical studies of FFNP PET imaging for patients with breast cancer align with the conclusions drawn from preclinical research and provide important human safety data for potential clinical use. In 2012, the “first-in-human” study of FFNP PET/CT involving 20 patients with breast cancer reported no adverse events or abnormal vital signs with a resulting radiation exposure for the patient comparable to standard clinical PET imaging agents (eg, FDG) [75]. Tumor uptake of FFNP peaked a few minutes after injection and remained stable through 60 minutes with no significant washout [75]. When corrected for normal breast tissue background, tumor FFNP uptake measured with PET/CT correlated with PR expression scores based on immunohistochemistry and was greater in PR+ cancers compared to PR-negative cancers [75]. Subsequent work by this group demonstrated that an increase in tumor FFNP uptake by at least 6.7% after a 1-day stimulation with estradiol predicted endocrine therapy response with 100% specificity and sensitivity in their prospective, single-center phase 2 study of 43 postmenopausal women with advanced and metastatic ER+ breast cancer [76]. Longer overall survival was observed in the responding participants. Thus, FFNP PET has been shown to be a safe method for measuring tumor PR expression, and changes in tumor FFNP uptake after estradiol stimulation are highly predictive of endocrine therapy response and survival in patients with advanced ER+ breast cancer.

PR also plays an important role in benign and malignant gynecologic processes. No studies have been published yet using FFNP PET imaging for gynecologic disease; however, there are 2 clinical trials on ClinicalsTrials.gov. The purpose of one study (NCT05483023) is to evaluate the utility of FFNP PET/MRI to predict response to progestin hormonal therapy for patients with complex atypical hyperplasia and endometrial carcinoma. Another study (NCT05480995) aims to assess the sensitivity and specificity of FFNP PET/MRI for the diagnosis of endometriosis. Thus, the potential clinical utility of PR-targeted imaging could extend beyond breast cancer to also include gynecologic disease.

There are some limitations to FFNP as a PET imaging agent for PR. A current practical challenge that may improve in the future is that FFNP is not yet approved by the FDA or commercially available and thus must be obtained close to the site of use typically by a local cyclotron and radiopharmacy [78, 79]. Based on the timeline and number of clinical trials performed before FDA approval of FES, more studies are needed before FFNP will likely gain approval for use in clinical practice. An inherent limitation of FFNP is that the hepatic metabolism results in high background activity and inhibit detection and quantification of uptake in liver lesions, similar to FES. Nonsteroidal PR-targeted radioligands and delayed time point imaging may improve this limitation [80]. Lastly, FFNP has been shown to also bind glucocorticoid receptor (GR) in vitro, which may be a confounding factor when both PR and GR are highly expressed [80]. However, FFNP uptake in one mouse mammary tumor model system was shown to be blocked by coadministration of the progestin R5020, which binds specifically to PR, but was not blocked with dexamethasone, which is specific to GR, supporting the conclusion that FFNP uptake appropriately reflects PR binding in vivo [71]. Other radiolabeled progestins, including those with very low GR-binding affinity, have been developed and tested in preclinical models, but not yet in humans [80-85].

Androgen Receptor

Prostate cancer is the second most common cancer among men in the United States, with 1 out of 8 men diagnosed during their lifetime [86]. When identified early, patients with prostate cancer can undergo definitive radical prostatectomy or radiotherapy. However, up to 30% of patients with prostate cancer will eventually develop metastatic castration-resistant prostate cancer (mCRPC) [87, 88]. Androgen deprivation therapy remains a mainstay of treatment in mCRPC despite androgen insensitivity and is used in conjunction with bone-modifying agents and chemotherapy [89]. Advances in the field of targeted molecular imaging and radionuclide therapy for mCRPC has led to the widespread adoption of PET amino acids (18F Fluciclovine; Axumin) and prostate-specific membrane antigen (PSMA) ligands (68Ga PSMA-11; Locometz, Illucix) and radionuclide therapy including bone-specific radionuclide agents (223Ra dichloride; Xofigo) and PSMA targeted radiotherapy (177Lu PSMA-617; Pluvicto) [90]. PSMA imaging in particular has eclipsed the ability of androgen receptor (AR) imaging to identify mCRPC lesions Similar to ER and PR expression in breast cancer, AR expression is heterogeneous and expression can change over time, particularly when patients are placed on hormonal therapy. Regardless, noninvasive PET imaging of AR expression has shown some utility in documenting the extent of the disease and can identify patients who may benefit from AR-targeting therapy. 16β-18F-Fluoro-5α-dihydrotestosterone (FDHT) is an AR-specific radiopharmaceutical and analogue of 5α-dihydrotestosterone (DHT) [91-93]. FDHT is rapidly metabolized, and the downstream radiolabeled metabolites have persistent affinity for blood proteins resulting in a relatively high background uptake [94]. Despite this limitation, FDHT demonstrates high repeatability and interobserver reproductivity in men with mCRPC [95].

FHDT was first evaluated in clinical studies in 2004 [96, 97], and while early trials with FDHT concentrated on evaluating metastatic staging and response to antiandrogen therapy, FDHT has limited sensitivity for mCRPC compared to other PET radiopharmaceuticals such as 68Ga- and 18F-labeled prostate-specific membrane antigen (PSMA)-based tracers, 18F-fluciclovine, and 11C-choline. Early studies of FDHT PET identified only 63% to 78% of radiographically proven metastases [96, 97]. As with FES and FFNP, FDHT has greater utility in interrogating the AR bioavailability and likelihood of success in patients being considered for continued AR therapy. A prospective study of 133 men with mCRPC who underwent both FDG PET and FDHT PET found that AR expression (as determined by FDHT PET) and glycolytic activity–Warburg effect (as determined by FDG PET) are independent factors of mCRPC progression and that patients with the most FDHT-negative lesions had the worst outcomes when treated with antiandrogen therapy [98]. Conversely, a retrospective analysis of 38 patients with mCRPC also undergoing evaluation both with FDG PET and FDHT PET found that patients with the most FDHT uptake had the shortest survival [99]. However, it should be noted that this particular study accrued patients between 2008 and 2009, before approval of modern antiandrogen pharmaceuticals such as enzalutamide. Several smaller phase 1 and 2 trials have used FDHT to identify AR-rich disease before and during enzalutamide and apalutamide therapy, but the authors did not correlate FDHT uptake or change in FDHT uptake to outcomes [100, 101].

While AR expression is most commonly associated with prostate cancer, AR is also abundantly expressed in breast cancer with up to 75% to 95% of ER+ and 10% to 35% of triple-negative (ER–, PR–, and HER2–) breast cancers expressing AR [102-104]. Several studies have shown that FDHT can be used to identify AR expression in breast cancers [105], and AR stimulation has been used in clinical trials to inhibit breast cancer tumor growth [106-108]. A prospective study was performed in 10 patients with ER+ breast cancer who underwent steroid hormone receptor interrogation both with FES and FDHT PET to evaluate the interreader visual and quantitative agreement [109]. For the 120 identifiable lesions, the study found a high visual positive and negative interobserver agreement with FES PET (84% and 83%, respectively) but low agreement with FDHT PET (49% and 74%). Conversely, both FES and FDHT PET had good quantification agreement of 0.98 and 0.78, respectively. The authors felt that this was due to the relatively low FDHT uptake thus requiring quantitative analysis of FDHT for complete evaluation in patients with breast cancer. A feasibility study using FDHT PET to predict response to antiandrogen therapy with bicalutamide in 21 patients with AR+ breast cancer patients found a baseline sensitivity of 66% in radiographically identifiable lesions, and a total of 21 new lesions were identified with FDHT PET [110]. However, while there was a decrease in radiotracer uptake after treatment with bicalutamide, FDHT PET could not predict which patients would have a response to antiandrogen therapy. Similarly, a phase 2 clinical trial with a novel oral nonsteroid androgen agonist, GTx-024, used FDHT PET to noninvasively interrogate whole-body AR expression in patients with metastatic ER+ breast cancer during therapy [111]. Nine women underwent FDHT PET, and patients who were found to have the most clinical benefit from AR stimulation had the largest decline in radiotracer uptake on the post FDHT PET. Of note, there was no correlation between baseline FDHT PET and circulating estradiol or testosterone levels. These trials in breast cancer targeting AR with androgen agonists or antagonists are likely constrained by the same limitations in studies with FDHT in patients with prostate cancer, in that AR ligand therapy (agonist or antagonist) will block the bioavailable AR and therapy does not necessarily correspond to a decrease (and thus response) in the AR protein expression itself [97].

Glioblastoma is the most common and aggressive form of primary brain malignancy, and AR is found overexpressed in a majority of glioblastomas [112]. AR antagonists have been shown to induce dose-dependent death of several glioblastoma cell lines and in vivo reduction in tumors. A feasibility study of 12 patients (6 men and 6 women) with high-grade glioma underwent FDHT PET to noninvasively evaluate AR expression [113]. Five of 12 glioblastoma lesions demonstrated significantly higher FDHT accumulation compared to normal brain with a maximum standardized uptake value (SUVmax) tumor/control of 1.6 to 3.4, and lesions with greater FDHT demonstrated a linear correlation with AR protein expression on histopathological analysis. While the SUV of the lesions was relatively low (SUVmax 0.45-2.3), the negligible FDHT uptake in normal brain parenchyma provided a good contrast to the area of AR-expressing glioma. This trial provides initial data for the potential use of FDHT to identify patients who may benefit from AR antagonist therapy.

To date, FDHT is the only positron-labeled AR-targeted radiopharmaceutical to enter clinical trials; however, efforts have been made to improve binding affinity and metabolic stability [114]. For example, enzalutamide is a pure AR antagonist routinely used in hormonal therapy in prostate cancer, and the radiolabeled 18F-enzalutamide has been found to have improved binding affinity and metabolic stability compared to FDHT [115]. Future clinical trials are planned to determine if 18F-enzalutamide will be able to improve on the low signal-to-noise that has hampered FDHT trials.

Other Nuclear Receptors

Compared to ER, PR, and AR, there are fewer published data for PET imaging agents targeting other nuclear receptors. GR and mineralocorticoid receptor are included in the nuclear receptor 3C subfamily along with PR and AR [116]. GR regulates many cellular processes, including catabolism and apoptosis. The transcriptional activity of GR in peripheral tissues is activated by binding to corticosteroids, which are synthesized and secreted by the adrenal cortex [117]. Corticosteroid production is controlled by a negative-feedback endocrine loop termed the hypothalamus-pituitary-adrenocortical axis. In healthy organisms, a pulse of high corticosteroid production and secretion typically occurs transiently after periods of stress, whereupon homeostasis is restored by corticosteroid metabolism in peripheral tissues. Dysregulation of GR signaling is known to stimulate several diseases including endocrine disorders, pulmonary diseases, mood disorders, and even cancers [118, 119]. For instance, hyperactivation of GR in tumor cells overrides the effects of cytotoxic chemotherapy in breast and ovarian cancer [120]. Moreover, high expression of GR in newly diagnosed triple-negative breast cancer appears to result in an especially fatal form of this already aggressive subtype [121]. However, accurate evaluation of the role that GR plays in human biology has been limited by the lack of an in vivo means to measure GR expression [122].

Several studies have focused on developing radioligands for GR with a particular interest in brain imaging [123-132]. Further development of these early GR-targeted radioligands was limited by a variety of technical issues including metabolic instability, defluorination, inability to cross the blood-brain barrier, insufficient brain uptake, and lack of specific binding. Investigators concluded that ligands with improved metabolic stability and higher receptor-binding affinity and selectivity were needed. Promising nonsteroidal radioligands with specific binding to GR have recently emerged from researchers at the University of California San Francisco that appear to overcome the limitations of previous agents [133-136]. Preliminary results from their ongoing first-in-human study (NCT04927663) evaluating 11C-YJH08 PET imaging for detecting GR expression in mCRPC were recently presented [137]. To date, there are no reported positron-labeled mineralocorticoids that have been evaluated in humans.

Radioligands targeting peroxisome proliferator-activated receptor γ (PPARγ) [138-144] and peroxisome proliferator-activated receptor α (PPARα) [145] have been synthesized and studied as potential PET imaging agents using various rodent models. PPARγ has several functions including regulating fat metabolism and is also seen in several pathologies including cancer, neurodegenerative diseases, and inflammation [146, 147]. PPARγ agonists have shown promising results by inducing cell cycle arrest and apoptosis in various malignancies in addition to preventing local invasion and metastasis [148]. Targeted PET radiopharmaceuticals to measure not only the level of PPARγ but also ligand-binding ability would provide a more accurate prediction of the outcome of PPARγ agonist treatment. PPARγ-targeted PET radiopharmaceutical development has been hindered by poor specificity and metabolic instability [139]. No human clinical trials have yet been reported for PPARγ positron-containing ligands.

The vitamin D receptor (VDR) binds the hormone calcitriol, also known as 1,25-dihydroxyvitamin D3, and regulates calcium homeostasis [149]. In addition to the classic role in bone health, the actions of VDR modulates the immune system and can inhibit proliferation, stimulate differentiation, and induce apoptosis in various normal and malignant cells [150]. Thus, VDR-targeted pharmaceuticals and imaging agents may have broad applications in the fields of oncology, endocrinology, immunology, and bone disease. Bonasera et al [151] developed 11C-labeled 1,25(OH)2 D3 with the goal of in vivo measurement of VDR expression and ligand occupancy. Their study showed high affinity in vitro binding to purified VDR. Preclinical PET imaging studies have not yet been published using this radioligand.

The retinoid X receptor (RXR) is an important DNA binding partner for several nuclear receptors, including VDR, PPARs, thyroid receptor, and retinoic acid receptor [152]. RXR is activated by 9-cis retinoic acid and plays a role in many biological processes including development, cellular differentiation, metabolism, and cell death [153, 154]. 18F-Labeled and 11C-labeled radioligands for RXR have been developed and studied in rodents and nonhuman primates as potential imaging biomarkers for treatment of central nervous system diseases [155-160]. However, translation of these RXR radioligands has not yet progressed to first-in-human studies.

The farnesoid X receptor (FXR) is activated by bile acids, controls bile acid synthesis, and modulates lipid and glucose metabolism [161]. FXR also plays a role in cancer and cardiovascular disease [162]. Jia et al [163] developed and evaluated an 18F-labeled bile acid compound as a potential PET imaging agent. The authors demonstrated high in vitro and in vivo metabolic stability of this agent and feasibility through PET/CT imaging of athymic nude mice. They propose potential future application as a PET imaging agent for early detection of FXR-related diseases and that further research is needed.

Conclusion

Considerable work has been achieved in the synthesis and initial preclinical testing of radioligands targeting nuclear receptors. Like therapeutic drug development, many agents do not proceed as far as first-in-human studies. However, the recent FDA approval of FES is a success story and clinical implementation of FES PET imaging for patients with recurrent and metastatic breast cancer is increasing at many institutions. Additional research is needed for FFNP and FDHT to be ready for clinical use.

Search Strategy and Selection Criteria

An electronic literature search was performed using PubMed to identify potential studies published in English until August 2022. The search terms “positron emission tomography” and “estrogen receptor,” “progesterone receptor,” “androgen receptor,” “glucocorticoid receptor,” “vitamin D receptor,” and “retinoid X receptor” were used. Relevant studies were retrieved, and their references were reviewed to identify any additional studies. Articles relevant to the scope of this mini-review were included.

Acknowledgments

The authors thank Kelley Salem, PhD, for assistance in figure preparation. We also acknowledge the University of Wisconsin Carbone Cancer Center Support Grant P30 CA014520 and the Department of Radiology, University of Wisconsin School of Medicine and Public Health for research support.

Abbreviations

AR

androgen receptor

CT

computed tomography

ER

estrogen receptor

FDA

US Food and Drug Administration

FDG

2-deoxy-2-[18F]fluoro-D-glucose

FDHT

16β-18F-fluoro-5α-dihydrotestosterone

FES

16α-[18F]fluoro-17β-estradiol

FFNP

21-18F-fluorofuranylnorprogesterone

FXR

farnesoid X receptor

GR

glucocorticoid receptor

mCRPC

metastatic castration-resistant prostate cancer

MRI

magnetic resonance imaging

PET

positron emission tomography

PPARγ

peroxisome proliferator-activated receptor γ

PR

progesterone receptor

PSMA

prostate-specific membrane antigen

RXR

retinoid X receptor

SUVmax

maximum standardized uptake value

VDR

vitamin D receptor

Contributor Information

Ephraim E Parent, Mayo Clinic Florida, Department of Radiology, Jacksonville, Florida 32224, USA.

Amy M Fowler, Email: afowler@uwhealth.org, Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792-3252, USA; University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53792, USA; Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53705, USA.

Financial Support

This work was supported by the American Cancer Society (No. RSG-22-015-01-CCB to A.M.F.) and the National Institutes of Health/National Cancer Institute (No. 1R01CA272571-01 to A.M.F.).

Disclosures

E.E.P. has nothing to disclose. A.M.F. receives a book chapter royalty from Elsevier, Inc and has served on an advisory board for GE Healthcare. The Department of Radiology at the University of Wisconsin School of Medicine and Public Health receives research support from GE Healthcare.

Data Availability

Data sharing is not applicable to this article as no data sets were generated or analyzed during the present study.

References

  • 1. Zhao L, Zhou S, Gustafsson JÅ. Nuclear receptors: recent drug discovery for cancer therapies. Endocr Rev. 2019;40(5):1207–1249. [DOI] [PubMed] [Google Scholar]
  • 2. Frigo DE, Bondesson M, Williams C. Nuclear receptors: from molecular mechanisms to therapeutics. Essays Biochem. 2021;65(6):847–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Kinch MS, Hoyer D, Patridge E, Plummer M. Target selection for FDA-approved medicines. Drug Discov Today. 2015;20(7):784–789. [DOI] [PubMed] [Google Scholar]
  • 4. Overington JP, Al-Lazikani B, Hopkins AL. How many drug targets are there? Nat Rev Drug Discov. 2006;5(12):993–996. [DOI] [PubMed] [Google Scholar]
  • 5. Allott L, Smith G, Aboagye EO, Carroll L. PET imaging of steroid hormone receptor expression. Mol Imaging. 2015;14(10):534–550. [DOI] [PubMed] [Google Scholar]
  • 6. Katzenellenbogen JA. PET imaging agents (FES, FFNP, and FDHT) for estrogen, androgen, and progesterone receptors to improve management of breast and prostate cancers by functional imaging. Cancers (Basel). 2020;12(8):2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Giaquinto AN, Sung H, Miller KD, et al. Breast cancer statistics, 2022. CA Cancer J Clin. 2022;72(6):524–541. [DOI] [PubMed] [Google Scholar]
  • 8. Giaquinto AN, Miller KD, Tossas KY, Winn RA, Jemal A, Siegel RL. Cancer statistics for African American/Black people 2022. CA Cancer J Clin. 2022;72(3):202–229. [DOI] [PubMed] [Google Scholar]
  • 9. Fowler AM, Cho SY. PET imaging for breast cancer. Radiol Clin North Am. 2021;59(5):725–735. [DOI] [PubMed] [Google Scholar]
  • 10. Liberti MV, Locasale JW. The Warburg effect: how does it benefit cancer cells? Trends Biochem Sci. 2016;41(3):211–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Warburg O. On the origin of cancer cells. Science. 1956;123(3191):309–314. [DOI] [PubMed] [Google Scholar]
  • 12. Warburg O. On respiratory impairment in cancer cells. Science. 1956;124(3215):269–270. [PubMed] [Google Scholar]
  • 13. Wahl RL, Cody RL, Hutchins GD, Mudgett EE. Primary and metastatic breast carcinoma: initial clinical evaluation with PET with the radiolabeled glucose analogue 2-[F-18]-fluoro-2-deoxy-D-glucose. Radiology. 1991;179(3):765–770. [DOI] [PubMed] [Google Scholar]
  • 14. Groheux D, Giacchetti S, Moretti JL, et al. Correlation of high 18F-FDG uptake to clinical, pathological and biological prognostic factors in breast cancer. Eur J Nucl Med Mol Imaging. 2011;38(3):426–435. [DOI] [PubMed] [Google Scholar]
  • 15. Iqbal R, Mammatas LH, Aras T, et al. Diagnostic performance of [18F]FDG PET in staging grade 1-2, estrogen receptor positive breast cancer. Diagnostics (Basel). 2021;11(11):1954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Jung NY, Kim SH, Choi BB, Kim SH, Sung MS. Associations between the standardized uptake value of (18)F-FDG PET/CT and the prognostic factors of invasive lobular carcinoma: in comparison with invasive ductal carcinoma. World J Surg Oncol. 2015;13(1):113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Kurland BF, Wiggins JR, Coche A, et al. Whole-body characterization of estrogen receptor status in metastatic breast cancer with 16α-18F-fluoro-17β-estradiol positron emission tomography: meta-analysis and recommendations for integration into clinical applications. Oncologist. 2020;25(10):835–844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Piccardo A, Fiz F, Treglia G, Bottoni G, Trimboli P. Head-to-head comparison between 18F-FES PET/CT and 18F-FDG PET/CT in oestrogen receptor-positive breast cancer: a systematic review and meta-analysis. J Clin Med. 2022;11(7):1919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. US Food and Drug Administration . Drug Trial Snapshot: CERIANNA. Accessed December 22, 2022. https://www.fda.gov/drugs/drug-approvals-and-databases/drug-trial-snapshot-cerianna
  • 20. Kiesewetter DO, Kilbourn MR, Landvatter SW, Heiman DF, Katzenellenbogen JA, Welch MJ. Preparation of four fluorine- 18-labeled estrogens and their selective uptakes in target tissues of immature rats. J Nucl Med. 1984;25(11):1212–1221. [PubMed] [Google Scholar]
  • 21. Mintun MA, Welch MJ, Siegel BA, et al. Breast cancer: PET imaging of estrogen receptors. Radiology. 1988;169(1):45–48. [DOI] [PubMed] [Google Scholar]
  • 22. Fox EM, Davis RJ, Shupnik MA. ERbeta in breast cancer—onlooker, passive player, or active protector? Steroids. 2008;73(11):1039–1051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Yoo J, Dence CS, Sharp TL, Katzenellenbogen JA, Welch MJ. Synthesis of an estrogen receptor beta-selective radioligand: 5-[18F]fluoro-(2R,3S)-2,3-bis(4-hydroxyphenyl)pentanenitrile and comparison of in vivo distribution with 16alpha-[18F]fluoro-17beta-estradiol. J Med Chem. 2005;48(20):6366–6378. [DOI] [PubMed] [Google Scholar]
  • 24. Moon BS, Carlson KE, Katzenellenbogen JA, et al. Synthesis and evaluation of aryl-substituted diarylpropionitriles, selective ligands for estrogen receptor beta, as positron-emission tomographic imaging agents. Bioorg Med Chem. 2009;17(9):3479–3488. [DOI] [PubMed] [Google Scholar]
  • 25. Lee JH, Peters O, Lehmann L, et al. Synthesis and biological evaluation of two agents for imaging estrogen receptor β by positron emission tomography: challenges in PET imaging of a low abundance target. Nucl Med Biol. 2012;39(8):1105–1116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Antunes IF, van Waarde A, Dierckx RA, de Vries EG, Hospers GA, de Vries EF. Synthesis and evaluation of the estrogen receptor β-selective radioligand 2-18F-fluoro-6-(6-hydroxynaphthalen-2-yl)pyridin-3-ol: comparison with 16α-18F-fluoro-17β-estradiol. J Nucl Med. 2017;58(4):554–559. [DOI] [PubMed] [Google Scholar]
  • 27. Tryfonidis K, Zardavas D, Katzenellenbogen BS, Piccart M. Endocrine treatment in breast cancer: cure, resistance and beyond. Cancer Treat Rev. 2016;50:68–81. [DOI] [PubMed] [Google Scholar]
  • 28. Jongen V, Briët J, de Jong R, et al. Expression of estrogen receptor-alpha and -beta and progesterone receptor-A and -B in a large cohort of patients with endometrioid endometrial cancer. Gynecol Oncol. 2009;112(3):537–542. [DOI] [PubMed] [Google Scholar]
  • 29. Salem K, Kumar M, Powers GL, et al. 18F-16α-17β-Fluoroestradiol binding specificity in estrogen receptor-positive breast cancer. Radiology. 2018;286(3):856–864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Kumar M, Salem K, Michel C, Jeffery JJ, Yan Y, Fowler AM. 18F-fluoroestradiol PET imaging of activating estrogen receptor-α mutations in breast cancer. J Nucl Med. 2019;60(9):1247–1252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Boers J, Venema CM, de Vries EFJ, et al. Molecular imaging to identify patients with metastatic breast cancer who benefit from endocrine treatment combined with cyclin-dependent kinase inhibition. Eur J Cancer. 2020;126:11–20. [DOI] [PubMed] [Google Scholar]
  • 32. Venema CM, Apollonio G, Hospers GAP, et al. Recommendations and technical aspects of 16α-[18F]fluoro-17β-estradiol PET to image the estrogen receptor in vivo: the Groningen experience. Clin Nucl Med. 2016;41(11):844–851. [DOI] [PubMed] [Google Scholar]
  • 33. Ulaner GA, Mankoff DA, Clark AS, et al. Appropriate use criteria for estrogen receptor (ER)-targeted PET imaging with 16α-18F-fluoro-17β-fluoroestradiol (18F-FES). society of nuclear medicine and molecular imaging. Accessed December 22, 2022. https://www.snmmi.org/ClinicalPractice/content.aspx?ItemNumber=42023 [Google Scholar]
  • 34. Boers J, Loudini N, Brunsch CL, et al. Value of 18F-FES PET in solving clinical dilemmas in breast cancer patients: a retrospective study. J Nucl Med. 2021;62(9):1214–1220. [DOI] [PubMed] [Google Scholar]
  • 35. Lee Y, Yoo IR, Ha S. 18F-FES PET/CT for characterization of brain and leptomeningeal metastasis in double primary cancer patient. Clin Nucl Med. 2022;47(8):e554–e556. [DOI] [PubMed] [Google Scholar]
  • 36. Yang Z, Xie Y, Liu C, et al. The clinical value of 18F-fluoroestradiol in assisting individualized treatment decision in dual primary malignancies. Quant Imaging Med Surg. 2021;11(9):3956–3965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. van Kruchten M, Glaudemans AW, de Vries EF, et al. PET Imaging of estrogen receptors as a diagnostic tool for breast cancer patients presenting with a clinical dilemma. J Nucl Med. 2012;53(2):182–190. [DOI] [PubMed] [Google Scholar]
  • 38. Ulaner GA. 16α-18F-Fluoro-17β-fluoroestradiol (FES): clinical applications for patients with breast cancer. Semin Nucl Med. 2022;52(5):574–583. [DOI] [PubMed] [Google Scholar]
  • 39. Harbeck N, Gnant M. Breast cancer. Lancet. 2017;389(10074):1134–1150. [DOI] [PubMed] [Google Scholar]
  • 40. Zardavas D, Irrthum A, Swanton C, Piccart M. Clinical management of breast cancer heterogeneity. Nat Rev Clin Oncol. 2015;12(7):381–394. [DOI] [PubMed] [Google Scholar]
  • 41. Haynes B, Sarma A, Nangia-Makker P, Shekhar MP. Breast cancer complexity: implications of intratumoral heterogeneity in clinical management. Cancer Metastasis Rev. 2017;36(3):547–555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Chae SY, Ahn SH, Kim SB, et al. Diagnostic accuracy and safety of 16α-[18F]fluoro-17β-oestradiol PET-CT for the assessment of oestrogen receptor status in recurrent or metastatic lesions in patients with breast cancer: a prospective cohort study. Lancet Oncol. 2019;20(4):546–555. [DOI] [PubMed] [Google Scholar]
  • 43. van Geel JJL, Boers J, Elias SG, et al. ; IMPACT-Metastatic Breast Consortium . Clinical validity of 16α-[18F]fluoro-17β-estradiol positron emission tomography/computed tomography to assess estrogen receptor status in newly diagnosed metastatic breast cancer. J Clin Oncol. 2022;40(31):3642–3652. [DOI] [PubMed] [Google Scholar]
  • 44. Nienhuis HH, van Kruchten M, Elias SG, et al. 18F-fluoroestradiol tumor uptake is heterogeneous and influenced by site of metastasis in breast cancer patients. J Nucl Med. 2018;59(8):1212–1218. [DOI] [PubMed] [Google Scholar]
  • 45. Liu C, Hu S, Xu X, et al. Evaluation of tumour heterogeneity by 18F-fluoroestradiol PET as a predictive measure in breast cancer patients receiving palbociclib combined with endocrine treatment. Breast Cancer Res. 2022;24(1):57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Xie Y, Du X, Zhao Y, et al. Chemotherapy shows a better efficacy than endocrine therapy in metastatic breast cancer patients with a heterogeneous estrogen receptor expression assessed by 18F-FES PET. Cancers (Basel). 2022;14(14):3531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Tsujikawa T, Yoshida Y, Mori T, et al. Uterine tumors: pathophysiologic imaging with 16alpha-[18F]fluoro-17beta-estradiol and 18F fluorodeoxyglucose PET—initial experience. Radiology. 2008;248(2):599–605. [DOI] [PubMed] [Google Scholar]
  • 48. Tsujikawa T, Yoshida Y, Kiyono Y, et al. Functional oestrogen receptor α imaging in endometrial carcinoma using 16α-[18F]fluoro-17β-oestradiol PET. Eur J Nucl Med Mol Imaging. 2011;38(1):37–45. [DOI] [PubMed] [Google Scholar]
  • 49. Yamada S, Tsuyoshi H, Yamamoto M, et al. Prognostic value of 16α-18F-fluoro-17β-estradiol PET as a predictor of disease outcome in endometrial cancer: a prospective study. J Nucl Med. 2021;62(5):636–642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Trovik J, Wik E, Werner HMJ, et al. Hormone receptor loss in endometrial carcinoma curettage predicts lymph node metastasis and poor outcome in prospective multicentre trial. Eur J Cancer. 2013;49(16):3431–3441. [DOI] [PubMed] [Google Scholar]
  • 51. Yoshida Y, Kurokawa T, Tsujikawa T, Okazawa H, Kotsuji F. Positron emission tomography in ovarian cancer: 18F-deoxy-glucose and 16alpha-18F-fluoro-17beta-estradiol PET. J Ovarian Res. 2009;2(1):7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. van Kruchten M, de Vries EFJ, Arts HJG, et al. Assessment of estrogen receptor expression in epithelial ovarian cancer patients using 16α-18F-fluoro-17β-estradiol PET/CT. J Nucl Med. 2015;56(1):50–55. [DOI] [PubMed] [Google Scholar]
  • 53. Tsujikawa T, Makino A, Mori T, et al. PET imaging of estrogen receptors for gynecological tumors. Clin Nucl Med. 2022;47(7):e481–e488. [DOI] [PubMed] [Google Scholar]
  • 54. Savouret JF, Bailly A, Misrahi M, et al. Characterization of the hormone responsive element involved in the regulation of the progesterone receptor gene. EMBO J. 1991;10(7):1875–1883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Read LD, Snider CE, Miller JS, Greene GL, Katzenellenbogen BS. Ligand-modulated regulation of progesterone receptor messenger ribonucleic acid and protein in human breast cancer cell lines. Mol Endocrinol. 1988;2(3):263–271. [DOI] [PubMed] [Google Scholar]
  • 56. Horwitz KB, Koseki Y, McGuire WL. Estrogen control of progesterone receptor in human breast cancer: role of estradiol and antiestrogen. Endocrinology. 1978;103(5):1742–1751. [DOI] [PubMed] [Google Scholar]
  • 57. Horwitz KB, McGuire WL. Predicting response to endocrine therapy in human breast cancer: a hypothesis. Science. 1975;189(4204):726–727. [DOI] [PubMed] [Google Scholar]
  • 58. Allison KH, Hammond MEH, Dowsett M, et al. Estrogen and progesterone receptor testing in breast cancer: ASCO/CAP guideline update. J Clin Oncol. 2020;38(12):1346–1366. [DOI] [PubMed] [Google Scholar]
  • 59. Howell A, Harland RN, Barnes DM, et al. Endocrine therapy for advanced carcinoma of the breast: relationship between the effect of tamoxifen upon concentrations of progesterone receptor and subsequent response to treatment. Cancer Res. 1987;47(1):300–304. [PubMed] [Google Scholar]
  • 60. Namer M, Lalanne C, Baulieu EE. Increase of progesterone receptor by tamoxifen as a hormonal challenge test in breast cancer. Cancer Res. 1980;40(5):1750–1752. [PubMed] [Google Scholar]
  • 61. Daniel AR, Gaviglio AL, Knutson TP, et al. Progesterone receptor-B enhances estrogen responsiveness of breast cancer cells via scaffolding PELP1- and estrogen receptor-containing transcription complexes. Oncogene. 2015;34(4):506–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Mohammed H, Russell IA, Stark R, et al. Progesterone receptor modulates ERα action in breast cancer. Nature. 2015;523(7560):313–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Singhal H, Greene ME, Tarulli G, et al. Genomic agonism and phenotypic antagonism between estrogen and progesterone receptors in breast cancer. Sci Adv. 2016;2(6):e1501924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Cottu PH, Bonneterre J, Varga A, et al. Phase I study of onapristone, a type I antiprogestin, in female patients with previously treated recurrent or metastatic progesterone receptor-expressing cancers. PLoS One. 2018;13(10):e0204973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Lewis JH, Cottu PH, Lehr M, et al. Onapristone extended release: safety evaluation from phase I-II studies with an emphasis on hepatotoxicity. Drug Saf. 2020;43(10):1045–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Lee O, Sullivan ME, Xu Y, et al. Selective progesterone receptor modulators in early-stage breast cancer: a randomized, placebo-controlled phase II window-of-opportunity trial using telapristone acetate. Clin Cancer Res. 2020;26(1):25–34. [DOI] [PubMed] [Google Scholar]
  • 67. Kamaraju S, Fowler AM, Weil E, et al. Leveraging antiprogestins in the treatment of metastatic breast cancer. Endocrinology. 2021;162(8):bqab060. [DOI] [PubMed] [Google Scholar]
  • 68. Buckman BO, Bonasera TA, Kirschbaum KS, Welch MJ, Katzenellenbogen JA. Fluorine-18-labeled progestin 16 alpha, 17 alpha-dioxolanes: development of high-affinity ligands for the progesterone receptor with high in vivo target site selectivity. J Med Chem. 1995;38(2):328–337. [DOI] [PubMed] [Google Scholar]
  • 69. Vijaykumar D, Mao W, Kirschbaum KS, Katzenellenbogen JA. An efficient route for the preparation of a 21-fluoro progestin-16 alpha,17 alpha-dioxolane, a high-affinity ligand for PET imaging of the progesterone receptor. J Org Chem. 2002;67(14):4904–4910. [DOI] [PubMed] [Google Scholar]
  • 70. Fowler AM, Chan SR, Sharp TL, et al. Small-animal PET of steroid hormone receptors predicts tumor response to endocrine therapy using a preclinical model of breast cancer. J Nucl Med. 2012;53(7):1119–1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Chan SR, Fowler AM, Allen JA, et al. Longitudinal noninvasive imaging of progesterone receptor as a predictive biomarker of tumor responsiveness to estrogen deprivation therapy. Clin Cancer Res. 2015;21(5):1063–1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Salem K, Kumar M, Kloepping KC, Michel CJ, Yan Y, Fowler AM. Determination of binding affinity of molecular imaging agents for steroid hormone receptors in breast cancer. Am J Nucl Med Mol Imaging. 2018;8(2):119–126. [PMC free article] [PubMed] [Google Scholar]
  • 73. Salem K, Kumar M, Yan Y, et al. Sensitivity and isoform specificity of 18F-fluorofuranylnorprogesterone for measuring progesterone receptor protein response to estradiol challenge in breast cancer. J Nucl Med. 2019;60(2):220–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Kumar M, Salem K, Jeffery JJ, Yan Y, Mahajan AM, Fowler AM. Longitudinal molecular imaging of progesterone receptor reveals early differential response to endocrine therapy in breast cancer with an activating ESR1 mutation. J Nucl Med. 2021;62(4):500–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Dehdashti F, Laforest R, Gao F, et al. Assessment of progesterone receptors in breast carcinoma by PET with 21-18F-fluoro-16α,17α-[(R)-(1'-α-furylmethylidene)dioxy]-19-norpregn- 4-ene-3,20-dione. J Nucl Med. 2012;53(3):363–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Dehdashti F, Wu N, Ma CX, Naughton MJ, Katzenellenbogen JA, Siegel BA. Association of PET-based estradiol-challenge test for breast cancer progesterone receptors with response to endocrine therapy. Nat Commun. 2021;12(1):733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Fowler A, Salem K, Henze Bancroft L, et al. Targeting the progesterone receptor in breast cancer using simultaneous FFNP breast PET/MRI: a pilot study. J Nucl Med. 2022;63(Suppl 2):2589–2589. [Google Scholar]
  • 78. Zhou D, Lin M, Yasui N, et al. Optimization of the preparation of fluorine-18-labeled steroid receptor ligands 16alpha-[18F]fluoroestradiol (FES), [18F]fluoro furanyl norprogesterone (FFNP), and 16beta-[18F]fluoro-5alpha-dihydrotestosterone (FDHT) as radiopharmaceuticals. J Labelled Comp Radiopharm. 2014;57(5):371–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Basuli F, Zhang X, Blackman B, et al. Fluorine-18 labeled fluorofuranylnorprogesterone ([18F]FFNP) and dihydrotestosterone ([18F]FDHT) prepared by “fluorination on Sep-Pak” method. Molecules. 2019;24(13):2389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Lee JH, Zhou HB, Dence CS, Carlson KE, Welch MJ, Katzenellenbogen JA. Development of [F-18]fluorine-substituted tanaproget as a progesterone receptor imaging agent for positron emission tomography. Bioconjug Chem. 2010;21(6):1096–1104. [DOI] [PubMed] [Google Scholar]
  • 81. Wu X, You L, Zhang D, et al. Synthesis and preliminary evaluation of a 18F-labeled ethisterone derivative [18F]EAEF for progesterone receptor targeting. Chem Biol Drug Des. 2017;89(4):559–565. [DOI] [PubMed] [Google Scholar]
  • 82. Gao F, Peng C, Zhuang R, et al. 18F-Labeled ethisterone derivative for progesterone receptor targeted PET imaging of breast cancer. Nucl Med Biol. 2019;72-73:62–69. [DOI] [PubMed] [Google Scholar]
  • 83. Zhou HB, Lee JH, Mayne CG, Carlson KE, Katzenellenbogen JA. Imaging progesterone receptor in breast tumors: synthesis and receptor binding affinity of fluoroalkyl-substituted analogues of tanaproget. J Med Chem. 2010;53(8):3349–3360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Allott L, Miranda C, Hayes A, Raynaud F, Cawthorne C, Smith G. Synthesis of a benzoxazinthione derivative of tanaproget and pharmacological evaluation for PET imaging of PR expression. EJNMMI Radiopharm Chem. 2019;4(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Merchant S, Allott L, Carroll L, et al. Synthesis and pre-clinical evaluation of a [18F]fluoromethyl-tanaproget derivative for imaging of progesterone receptor expression. RSC Adv. 2016;6(62):57569–57579. [Google Scholar]
  • 86. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33. [DOI] [PubMed] [Google Scholar]
  • 87. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–249. [DOI] [PubMed] [Google Scholar]
  • 88. Mollica V, Rizzo A, Rosellini M, et al. Bone targeting agents in patients with metastatic prostate cancer: state of the art. Cancers (Basel). 2021;13(3):546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Thompson IM J, Goodman PJ, Tangen CM, et al. Long-term survival of participants in the prostate cancer prevention trial. N Engl J Med. 2013;369(7):603–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Parent EE, Kase AM. A treatment paradigm shift: targeted radionuclide therapies for metastatic castrate resistant prostate cancer. Cancers (Basel). 2022;14(17):4276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Liu A, Carlson KE, Katzenellenbogen JA. Synthesis of high affinity fluorine-substituted ligands for the androgen receptor. Potential agents for imaging prostatic cancer by positron emission tomography. J Med Chem. 1992;35(11):2113–2129. [DOI] [PubMed] [Google Scholar]
  • 92. Liu A, Dence CS, Welch MJ, Katzenellenbogen JA. Fluorine-18-labeled androgens: radiochemical synthesis and tissue distribution studies on six fluorine-substituted androgens, potential imaging agents for prostatic cancer. J Nucl Med. 1992;33(5):724–734. [PubMed] [Google Scholar]
  • 93. Bonasera TA, O’Neil JP, Xu M, et al. Preclinical evaluation of fluorine-18-labeled androgen receptor ligands in baboons. J Nucl Med. 1996;37(6):1009–1015. [PubMed] [Google Scholar]
  • 94. Beattie BJ, Smith-Jones PM, Jhanwar YS, et al. Pharmacokinetic assessment of the uptake of 16beta-18F-fluoro-5alpha-dihydrotestosterone (FDHT) in prostate tumors as measured by PET. J Nucl Med. 2010;51(2):183–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Vargas HA, Kramer GM, Scott AM, et al. Reproducibility and repeatability of semiquantitative 18F-fluorodihydrotestosterone uptake metrics in castration-resistant prostate cancer metastases: a prospective multicenter study. J Nucl Med. 2018;59(10):1516–1523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Larson SM, Morris M, Gunther I, et al. Tumor localization of 16beta-18F-fluoro-5alpha-dihydrotestosterone versus 18F-FDG in patients with progressive, metastatic prostate cancer. J Nucl Med. 2004;45(3):366–373. [PubMed] [Google Scholar]
  • 97. Dehdashti F, Picus J, Michalski JM, et al. Positron tomographic assessment of androgen receptors in prostatic carcinoma. Eur J Nucl Med Mol Imaging. 2005;32(3):344–350. [DOI] [PubMed] [Google Scholar]
  • 98. Fox JJ, Gavane SC, Blanc-Autran E, et al. Positron emission tomography/computed tomography-based assessments of androgen receptor expression and glycolytic activity as a prognostic biomarker for metastatic castration-resistant prostate cancer. JAMA Oncol. 2018;4(2):217–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Vargas HA, Wassberg C, Fox JJ, et al. Bone metastases in castration-resistant prostate cancer: associations between morphologic CT patterns, glycolytic activity, and androgen receptor expression on PET and overall survival. Radiology. 2014;271(1):220–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Scher HI, Beer TM, Higano CS, et al. ; Prostate Cancer Foundation/Department of Defense Prostate Cancer Clinical Trials Consortium . Antitumour activity of MDV3100 in castration-resistant prostate cancer: a phase 1-2 study. Lancet. 2010; 375(9724):1437–1446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Rathkopf DE, Morris MJ, Fox JJ, et al. Phase I study of ARN-509, a novel antiandrogen, in the treatment of castration-resistant prostate cancer. J Clin Oncol. 2013;31(28):3525–3530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Collins LC, Cole KS, Marotti JD, Hu R, Schnitt SJ, Tamimi RM. Androgen receptor expression in breast cancer in relation to molecular phenotype: results from the Nurses’ Health Study. Mod Pathol. 2011;24(7):924–931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Kraby MR, Valla M, Opdahl S, et al. The prognostic value of androgen receptors in breast cancer subtypes. Breast Cancer Res Treat. 2018;172(2):283–296. [DOI] [PubMed] [Google Scholar]
  • 104. Kensler KH, Regan MM, Heng YJ, et al. Prognostic and predictive value of androgen receptor expression in postmenopausal women with estrogen receptor-positive breast cancer: results from the Breast International Group Trial 1-98. Breast Cancer Res. 2019;21(1):30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Venema CM, Mammatas LH, Schroder CP, et al. Androgen and estrogen receptor imaging in metastatic breast cancer patients as a surrogate for tissue biopsies. J Nucl Med. 2017;58(12):1906–1912. [DOI] [PubMed] [Google Scholar]
  • 106. Venema CM, Bense RD, Steenbruggen TG, et al. Consideration of breast cancer subtype in targeting the androgen receptor. Pharmacol Ther. 2019;200:135–147. [DOI] [PubMed] [Google Scholar]
  • 107. Lu Q, Xia W, Lee K, et al. Bicalutamide plus aromatase inhibitor in patients with estrogen receptor-positive/androgen receptor-positive advanced breast cancer. Oncologist. 2020;25(1):21-e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Narayanan R, Dalton JT. Androgen receptor: a complex therapeutic target for breast cancer. Cancers (Basel). 2016;8(12):108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Mammatas LH, Venema CM, Schröder CP, et al. Visual and quantitative evaluation of [18F]FES and [18F]FDHT PET in patients with metastatic breast cancer: an interobserver variability study. EJNMMI Res. 2020;10(1):40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Boers J, Venema CM, de Vries EFJ, et al. Serial [18F]-FDHT-PET to predict bicalutamide efficacy in patients with androgen receptor positive metastatic breast cancer. Eur J Cancer. 2021;144:151–161. [DOI] [PubMed] [Google Scholar]
  • 111. Jacene H, Liu M, Cheng SC, et al. Imaging androgen receptors in breast cancer with 18F-fluoro-5α-dihydrotestosterone PET: a pilot study. J Nucl Med. 2022;63(1):22–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Zalcman N, Canello T, Ovadia H, et al. Androgen receptor: a potential therapeutic target for glioblastoma. Oncotarget. 2018;9(28):19980–19993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Orevi M, Shamni O, Zalcman N, et al. [18F]-FDHT PET/CT as a tool for imaging androgen receptor expression in high-grade glioma. Neurooncol Adv. 2021;3(1):vdab019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Kim TH, Jeong JW, Song JH, et al. Pharmacokinetics of enzalutamide, an anti-prostate cancer drug, in rats. Arch Pharm Res. 2015;38(11):2076–2082. [DOI] [PubMed] [Google Scholar]
  • 115. Antunes IF, Dost RJ, Hoving HD, et al. Synthesis and evaluation of 18F-enzalutamide, a new radioligand for PET imaging of androgen receptors: a comparison with 16β-18F-fluoro-5α-dihydrotestosterone. J Nucl Med. 2021;62(8):1140–1145. [DOI] [PubMed] [Google Scholar]
  • 116. Nuclear Receptors Nomenclature Committee . A unified nomenclature system for the nuclear receptor superfamily. Cell. 1999;97(2):161–163. [DOI] [PubMed] [Google Scholar]
  • 117. Timmermans S, Souffriau J, Libert C. A general introduction to glucocorticoid biology. Front Immunol. 2019;10:1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Oakley RH, Cidlowski JA. The biology of the glucocorticoid receptor: new signaling mechanisms in health and disease. J Allergy Clin Immunol. 2013;132(5):1033–1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Conzen SD. Recent advances in understanding glucocorticoid receptor function in cancer. Clin Adv Hematol Oncol. 2017;15(5):338–340. [PMC free article] [PubMed] [Google Scholar]
  • 120. Chen Z, Lan X, Wu D, et al. Ligand-dependent genomic function of glucocorticoid receptor in triple-negative breast cancer. Nat Commun. 2015;6(1):8323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Pan D, Kocherginsky M, Conzen SD. Activation of the glucocorticoid receptor is associated with poor prognosis in estrogen receptor-negative breast cancer. Cancer Res. 2011;71(20):6360–6370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Sundahl N, Bridelance J, Libert C, De Bosscher K, Beck IM. Selective glucocorticoid receptor modulation: new directions with non-steroidal scaffolds. Pharmacol Ther. 2015;152:28–41. [DOI] [PubMed] [Google Scholar]
  • 123. Feliu AL, Rottenberg DA. Synthesis and evaluation of fluorine-18 21-fluoroprednisone as a potential ligand for neuro-PET studies. J Nucl Med. 1987;28(6):998–1005. [PubMed] [Google Scholar]
  • 124. Pomper MG, Kochanny MJ, Thieme AM, et al. Fluorine-substituted corticosteroids: synthesis and evaluation as potential receptor-based imaging agents for positron emission tomography of the brain. Int J Rad Appl Instrum B. 1992;19(4):461–480. [DOI] [PubMed] [Google Scholar]
  • 125. Dasilva JN, Crouzel C, Stulzaft O, Khalili-Varasteh M, Hantraye P. Synthesis, tissue distribution in rats and PET studies in baboon brain of no-carrier-added [18F]RU 52461: in vivo evaluation as a brain glucocorticoid receptor radioligand. Int J Rad Appl Instrum B. 1992;19(2):167–173. [DOI] [PubMed] [Google Scholar]
  • 126. Visser GM, Krugers HJ, Luurtsema G, et al. Synthesis and organ distribution of [18F]fluoro-Org 6141 in the rat: a potential glucocorticoid receptor ligand for positron emission tomography. Nucl Med Biol. 1995;22(7):915–920. [DOI] [PubMed] [Google Scholar]
  • 127. Hoyte RM, Labaree DC, Fede J-M, Harris C, Hochberg RB. Iodinated and fluorinated steroid 2′-aryl-[3,2-c] pyrazoles as potential glucocorticoid receptor imaging agents. Steroids. 1998;63(11):595–602. [DOI] [PubMed] [Google Scholar]
  • 128. Wüst F, Carlson KE, Katzenellenbogen JA. Synthesis of novel arylpyrazolo corticosteroids as potential ligands for imaging brain glucocorticoid receptors. Steroids. 2003;68(2):177–191. [DOI] [PubMed] [Google Scholar]
  • 129. Steiniger B, Kniess T, Bergmann R, Pietzsch J, Wuest FR. Radiolabeled glucocorticoids as molecular probes for imaging brain glucocorticoid receptors by means of positron emission tomography (PET). Mini Rev Med Chem. 2008;8(7):728–739. [DOI] [PubMed] [Google Scholar]
  • 130. Wuest F, Carlson KE, Katzenellenbogen JA. Expeditious synthesis of steroids containing a 2-methylsulfanyl-acetyl side chain as potential glucocorticoid receptor imaging agents. Steroids. 2008;73(1):69–76. [DOI] [PubMed] [Google Scholar]
  • 131. Wuest F, Kniess T, Bergmann R, Henry B, Pietzsch J. Synthesis and radiopharmacological characterization of [11C]AL-438 as a nonsteroidal ligand for imaging brain glucocorticoid receptors. Bioorg Med Chem Lett. 2007;17(14):4035–4039. [DOI] [PubMed] [Google Scholar]
  • 132. Wuest F, Kniess T, Henry B, et al. Radiosynthesis and radiopharmacological evaluation of [N-methyl-11C]org 34850 as a glucocorticoid receptor (GR)-binding radiotracer. Appl Radiat Isot. 2009;67(2):308–312. [DOI] [PubMed] [Google Scholar]
  • 133. Truillet C, Parker MFL, Huynh LT, et al. Measuring glucocorticoid receptor expression in vivo with PET. Oncotarget. 2018;9(29):20399–20408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Huang Y, Zhao N, Wang YH, et al. A novel radioligand reveals tissue specific pharmacological modulation of glucocorticoid receptor expression with positron emission tomography. ACS Chem Biol. 2020;15(6):1381–1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Huang Y, Zhao N, Wang YH, et al. The synthesis and structural requirements for measuring glucocorticoid receptor expression in vivo with (±)-(11)C-YJH08 PET. J Nucl Med. 2021;62(5):723–731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Kim H, Huang Y, Zhao N, et al. In vivo profiling with 18F-YJH08 reveals diverse tissue patterns of antagonist/glucocorticoid receptor interactions. Mol Pharm. 2022;19(2):704–709. [DOI] [PubMed] [Google Scholar]
  • 137. Evans M, Kim H, Aggarwal R, Behr S, Huang Y. Measuring glucocorticoid receptor expression and drug/GR interactions in vivo with YJH08: from bench to bedside. J Nucl Med. 2022;63(Suppl 2):2274–2274. [Google Scholar]
  • 138. Kim SH, Jonson SD, Welch MJ, Katzenellenbogen JA. Fluorine-substituted ligands for the peroxisome proliferator-activated receptor gamma (PPARgamma): potential imaging agents for metastatic tumors. Bioconjug Chem. 2001;12(3):439–450. [DOI] [PubMed] [Google Scholar]
  • 139. Lee BC, Dence CS, Zhou H, Parent EE, Welch MJ, Katzenellenbogen JA. Fluorine-18 labeling and biodistribution studies on peroxisome proliferator-activated receptor-gamma ligands: potential positron emission tomography imaging agents. Nucl Med Biol. 2009;36(2):147–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Lee H, Chen DL, Rothfuss JM, Welch MJ, Gropler RJ, Mach RH. Synthesis and evaluation of 18F-labeled PPARγ antagonists. Nucl Med Biol. 2012;39(1):77–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Mathews WB, Foss CA, Stoermer D, et al. Synthesis and biodistribution of (11)C-GW7845, a positron-emitting agonist for peroxisome proliferator-activated receptor-{gamma}. J Nucl Med. 2005;46(10):1719–1726. [PubMed] [Google Scholar]
  • 142. Lee BC, Lee KC, Lee H, Mach RH, Katzenellenbogen JA. Synthesis and binding affinity of a fluorine-substituted peroxisome proliferator-activated gamma (PPARgamma) ligand as a potential positron emission tomography (PET) imaging agent. Bioconjug Chem. 2007;18(2):507–513. [DOI] [PubMed] [Google Scholar]
  • 143. Lee BC, Lee KC, Lee H, Mach RH, Katzenellenbogen JA. Strategies for the labeling of halogen-substituted peroxisome proliferator-activated receptor gamma ligands: potential positron emission tomography and single photon emission computed tomography imaging agents. Bioconjug Chem. 2007;18(2):514–523. [DOI] [PubMed] [Google Scholar]
  • 144. Lee H, Finck BN, Jones LA, Welch MJ, Mach RH. Synthesis and evaluation of a bromine-76-labeled PPARgamma antagonist 2-bromo-5-nitro-N-phenylbenzamide. Nucl Med Biol. 2006;33(7):847–854. [DOI] [PubMed] [Google Scholar]
  • 145. Solingapuram Sai KK, Kil KE, Tu Z, et al. Synthesis, radiolabeling and initial in vivo evaluation of [(11)C]KSM-01 for imaging PPAR-α receptors. Bioorg Med Chem Lett. 2012;22(19):6233–6236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Dacic M, Shibu G, Rogatsky I. Physiological convergence and antagonism between GR and PPARγ in inflammation and metabolism. Adv Exp Med Biol. 2022;1390:123–141. [DOI] [PubMed] [Google Scholar]
  • 147. Katoch S, Sharma V, Patial V. Peroxisome proliferator-activated receptor gamma as a therapeutic target for hepatocellular carcinoma: experimental and clinical scenarios. World J Gastroenterol. 2022;28(28):3535–3554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Wu L, Guo C, Wu J. Therapeutic potential of PPARγ natural agonists in liver diseases. J Cell Mol Med. 2020;24(5):2736–2748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Bouillon R, Marcocci C, Carmeliet G, et al. Skeletal and extraskeletal actions of vitamin D: current evidence and outstanding questions. Endocr Rev. 2019;40(4):1109–1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Murray A, Madden SF, Synnott NC, et al. Vitamin D receptor as a target for breast cancer therapy. Endocr Relat Cancer. 2017;24(4):181–195. [DOI] [PubMed] [Google Scholar]
  • 151. Bonasera TA, Grue-Sørensen G, Ortu G, et al. The synthesis of [26,27-11C]dihydroxyvitamin D(3), a tracer for positron emission tomography (PET). Bioorg Med Chem. 2001;9(12):3123–3128. [DOI] [PubMed] [Google Scholar]
  • 152. Evans RM, Mangelsdorf DJ. Nuclear receptors, RXR, and the big bang. Cell. 2014;157(1):255–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Sharma S, Shen T, Chitranshi N, et al. Retinoid X receptor: cellular and biochemical roles of nuclear receptor with a focus on neuropathological involvement. Mol Neurobiol. 2022;59(4):2027–2050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Shao M, Lu L, Wang Q, et al. The multi-faceted role of retinoid X receptor in cardiovascular diseases. Biomed Pharmacother. 2021;137:111264. [DOI] [PubMed] [Google Scholar]
  • 155. Wang M, Davis T, Gao M, Zheng QH. Synthesis of a new fluorine-18-labeled bexarotene analogue for PET imaging of retinoid X receptor. Bioorg Med Chem Lett. 2014;24(7):1742–1747. [DOI] [PubMed] [Google Scholar]
  • 156. Rotstein BH, Hooker JM, Woo J, et al. Synthesis of [(11)C]bexarotene by Cu-mediated [(11)C]carbon dioxide fixation and preliminary PET imaging. ACS Med Chem Lett. 2014;5(6):668–672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Kobayashi T, Furusawa Y, Yamada S, et al. Positron emission tomography to elucidate pharmacokinetic differences of regioisomeric retinoid X receptor agonists. ACS Med Chem Lett. 2015;6(3):334–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Rotstein BH, Placzek MS, Krishnan HS, et al. Preclinical PET neuroimaging of [11C]bexarotene. Mol Imaging. 2016;15:1536012116663054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Shibahara O, Watanabe M, Yamada S, et al. Synthesis of 11C-labeled RXR partial agonist 1-[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydronaphthalen-2-yl)amino]benzotriazole-5-carboxylic acid (CBt-PMN) by direct [11C]carbon dioxide fixation via organolithiation of trialkyltin precursor and PET imaging thereof. J Med Chem. 2017;60(16):7139–7145. [DOI] [PubMed] [Google Scholar]
  • 160. Takamura Y, Shibahara O, Watanabe M, et al. Fluorine-18 (18F)-labeled retinoid X receptor (RXR) partial agonist whose tissue transferability is affected by other RXR ligands. Bioorg Med Chem. 2019;27(14):3128–3134. [DOI] [PubMed] [Google Scholar]
  • 161. Sonne DP. Mechanisms in endocrinology: FXR signalling: a novel target in metabolic diseases. Eur J Endocrinol. 2021;184(5):R193–R205. [DOI] [PubMed] [Google Scholar]
  • 162. Girisa S, Henamayee S, Parama D, Rana V, Dutta U, Kunnumakkara AB. Targeting farnesoid X receptor (FXR) for developing novel therapeutics against cancer. Mol Biomed. 2021;2(1):21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Jia L, Jiang D, Hu P, et al. Synthesis and evaluation of (18)F-labeled bile acid compound: a potential PET imaging agent for FXR-related diseases. Nucl Med Biol. 2014;41(6):495–500. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no data sets were generated or analyzed during the present study.


Articles from Journal of the Endocrine Society are provided here courtesy of The Endocrine Society

RESOURCES