Next Article in Journal
Steroidomic Changes in the Cerebrospinal Fluid of Women with Multiple Sclerosis
Next Article in Special Issue
Modeling the Bone Marrow Niche in Multiple Myeloma: From 2D Cultures to 3D Systems
Previous Article in Journal
Complement System Inhibitors in Nephrology: An Update—Narrative Review
Previous Article in Special Issue
Elucidating DNA Damage-Dependent Immune System Activation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Bridge Too Far? Towards Medical Therapy for Clinically Nonfunctioning Pituitary Tumors

1
UCLA Department of Medicine, Los Angeles, CA 90095, USA
2
UCLA Department of Neurosurgery, Los Angeles, CA 90095, USA
*
Author to whom correspondence should be addressed.
Current address: David Geffen School of Medicine, University of California, Los Angeles, UCLA Medical Center, 757 Westwood Plaza, Los Angeles, CA 90095, USA.
Int. J. Mol. Sci. 2025, 26(12), 5898; https://doi.org/10.3390/ijms26125898
Submission received: 18 May 2025 / Revised: 13 June 2025 / Accepted: 16 June 2025 / Published: 19 June 2025

Abstract

Clinically nonfunctioning pituitary tumors (CNFPTs) typically do not cause hormonal excess, progress insidiously, and are often large and invasive at presentation. Complete resection is frequently not attainable; radiotherapy (RT) may effectively limit growth but carries a significant risk of hypopituitarism. Medical therapy with dopamine D2 receptor agonists and/or somatostatin analogs has been explored in CNFPTs but have yielded inconsistent results, and there is an unmet need for novel efficacious and safe medical therapies. The authors used the PubMed database to identify and review articles published from January 1982 to July 2024, that discussed the medical treatment of CNFPTs. The most commonly studied medical therapies were somatostatin receptor ligands (SRLs) and dopamine D2 receptor agonists. Of 111 patients with CNFPTs treated with SRLs, 31 (28%) exhibited tumor shrinkage. Following dopamine agonist treatment in 355 patients, tumor shrinkage occurred in 113 (32%), tumor stabilization in 182 (51%), and tumor growth in 60 (17%). The efficacy of other less commonly employed therapies such as GnRH analogs, PRRT, and temozolomide was also reviewed. Efficacious and safe medical therapies evaluated in robust randomized placebo-controlled clinical trials are needed to improve the management of CNFPTs.

1. Introduction

Clinically nonfunctioning pituitary tumors (CNFPTs) account for 14–54% of all pituitary tumors, grow insidiously, and are typically found incidentally during cranial imaging or when patients present with visual field defects and/or hypopituitarism [1,2]. CNFPTs can rarely present with pituitary apoplexy (8%) or hyperprolactinemia from stalk displacement [3,4]. The complete surgical resection of CNFPTs is frequently unattainable, and ~65% of patients will harbor remnant tumor after surgery with a >40% risk of regrowth over 5–10 years [4,5,6]. Advances in histopathological classification have provided a more detailed characterization of the cellular origin of CNFPTs but molecular studies have not yet guided choice of medical therapies. Medical therapies that have primarily been used for CNFPT treatment include dopamine agonists and somatostatin receptor (SSTR) ligands.
This review highlights some of the challenges for drug discovery for CNFPTs given their heterogeneity, lack of circulating biomarkers, and assessment of tumor responses. There is a renewed interest in discovering innovative medical therapies for this comparatively common but elusive subset of pituitary tumors, and this review will hopefully spur efforts in this field to identify novel therapeutic options for patients.

2. Materials and Methods

The authors used the PubMed database to identify and review articles published from January, 1982, to July, 2024, using the search terms “CNFPTs,” “clinically nonfunctioning pituitary tumors,” “NFPAs,” and “nonfunctioning pituitary adenomas,” in combination with “dopamine agonist,” “somatostatin ligand,” “GnRH analog,” PRRT,” “peptide receptor radionuclide therapy,” “temozolomide,” “folate receptors,” “mTOR inhibitors,” and “VEGF inhibitors.” This literature review encompasses studies that were conducted worldwide, reflecting its global scope. The studies that were ultimately included were conducted in a variety of countries, including the United States, Portugal, Spain, Japan, and Italy. Additionally, all studies were accessed in English. Statistical or subgroup analyses were not possible, as the studies employed different methods of measurement of tumor size, medications and doses, duration of treatment, and definitions of significant change in tumor size.

3. Results

3.1. CNFPTs Represent a Heterogenous Group of Pituitary Tumors

The availability of immunohistochemical evaluation for pituitary-derived hormones and cell lineage-specific transcription factors in resected pituitary tumors has demonstrated that CNFPTs comprise a range of pituitary tumor subtypes (Table 1) [7,8]. This recognition that they are not a single entity may have implications for the development of medical therapy unless a common therapeutic target is identified.

3.1.1. Gonadotroph Tumors

Gonadotroph tumors are the most common subtype of CNFPTs and the second most common pituitary tumor subtype after lactotroph tumors [9]. They are immunoreactive to the transcription factor steroidogenic factor 1 (SF-1/NR5A1) and/or the gonadotropins follicular stimulating hormone (FSH) and luteinizing hormone (LH) [10]. The vast majority of gonadotroph tumors (~90%) are not associated with a clinical syndrome; but rarely cause ovarian hyperstimulation in women and sexual dysfunction in men due to excessive gonadotropins [11]

3.1.2. Silent Corticotroph Tumors

Silent corticotroph tumors account for ~30% of all corticotroph tumors and 10–20% of silent pituitary tumors and originate from a TBX19/Tpit expressing cell lineage [12]. Some cause elevated circulating adrenocorticotropic hormone (ACTH) levels, though manifestations of hypercortisolism are infrequently seen due to the incomplete processing of the proopiomelanocortin (POMC) peptide [12]. The rare Crooke’s cell adenoma subtype of silent corticotroph tumors exhibits higher proliferative and recurrence rates [11].

3.1.3. Silent Somatotroph Tumors

Silent somatotroph tumors, immunoreactive to POU class 1 homeobox 1 (POU1F1/Pit-1) and growth hormone (GH) at variable levels, are exceptionally rare and account for 2–4% of all types of pituitary tumors [13]. They typically do not cause increased GH and insulin like growth factor-1 (IGF-1) levels or cause acromegalic features [14]. They are often resistant to somatostatin analog treatment due to low somatostatin receptor 2 (SSTR2) expression [15].

3.1.4. Silent Thyrotroph Tumors

Functioning thyrotroph tumors are very rare, accounting for 1 to 2% of all pituitary adenomas, and silent thyrotroph tumors are even less common [16]. These thyrotroph tumors express either the thyroid stimulating hormone (TSH) α- and β-subunits and/or POU1F1/Pit-1 and/or GATA binding protein 2 (GATA2). They typically produce dysfunctional TSH and do not cause hyperthyroxinemia [16].

3.1.5. Silent Lactotroph Tumors

Rarely, silent lactrotroph tumors that express POU1F1/Pit-1 and estrogen receptor alpha (ERα) but not prolactin (PRL) are encountered and can cause a mild elevation of prolactin levels due to stalk displacement [17]. They can occasionally be found as an admixture with GH-expressing cells in mammosomatotroph tumors and Pit-1 + plurihormonal tumors [18,19].

3.1.6. Null Cell Tumors

True null cell tumors account for 1–5% of CNFPTs and express the neuroendocrine marker synaptophysin (SYP), but not any pituitary lineage specific transcription factors (POU1F1, SF1, TBX19, and ERα) and/or hormones (GH, PRL, TSH, FSH-β, LH-β, ACTH, and α-subunit) [20,21].

3.2. Current Management of CNFPTs

Therapy for CNFPTs should include consideration of tumor size, the effect on the optic tract, pituitary gland, or cranial nerves, patient co-morbidities, and patient wishes. In asymptomatic and small CNFPTs, it may be appropriate to monitor endocrine function and radiologic tumor size annually without intervention [22]. Asymptomatic patients with macroadenomas without visual threat or significant comorbidities may also warrant a conservative approach, though imaging may be more frequent (9–12 months) along with the monitoring of visual fields [23]. Fortunately, pituitary surgery in expert centers is a comparatively safe procedure with low complication (<5%) and mortality rates (<1%) [24]. Thus, surgical resection is generally offered to younger patients even with asymptomatic CNFPT macroadenomas as the tumors have a longer time to grow [23]. Surgery is indicated in any patient in whom compressive symptoms on the optic chiasm or cranial nerves can be alleviated by tumor debulking [23]. The role of surgery in reversing established hypopituitarism in these patients is not clear, with one meta-analysis reporting only one third of patients with an improvement in pituitary function following tumor resection, with a 4.9- and 2.5-fold risk of developing new anterior pituitary dysfunction and permanent central arginine vasopressin (AVP)-deficiency, respectively, following the surgery [24].
Surgical gross total resection (GTR) or near-total resection of CNFPTs ranges from 20 to 80% [25,26] but growth of the remnant tumor is seen in 30–50% of patients over five to ten years [4,5,6,25]. Currently, options for recurrence include further surgical debulking or adjunctive radiotherapy (RT). RT effectively stops further tumor growth in 85% to 95% of patients at five to ten years [27]. However, radiation carries a risk of hypopituitarism in up to 40% of patients, though this risk may be less in patients where the radio-target is distant from the pituitary stalk and the normal pituitary gland [28,29].

3.3. Current Role of Medical Therapy in CNFPTs

While the use of immunohistochemical transcription marker analysis and pituitary hormone assessment in CNFPTs has improved our ability to sub-categorize these tumors, there are no circulating or tissue biomarkers that can be monitored in parallel with tumor growth. Additionally, the presence of several distinct CNFPT subtypes may make finding a single efficacious therapy for all elusive. Nonetheless, given the frequent inability to achieve total tumor resection in CNFPTs, their regrowth rates, and risk of hypopituitarism and secondary brain tumors after adjuvant radiation, there is a clear unmet need for medical therapies for these common pituitary tumors. Medical therapy for CNFPTs has thus far been directed at the somatostatin receptors (primarily subtype 2 [SSTR-2]) and dopamine receptors (subtype 2 [DR2)]), with some use of gonadotrophin-releasing hormone (GnRH) analogs.

3.3.1. Somatostatin Receptor Ligands

Somatostatin binds SSTRs to induce cell cycle arrest (SSTR-1, -2, -4, and -5) and apoptosis (SSTR-3) and inhibit vascular endothelial growth factor (VEGF) and α-subunit secretion [30,31]. Studies have reported the expression of SSTR subtypes in CNFPTs, predominantly SSTR-2 and SSTR-3, with a minority reporting SSTR-5 [11,32,33,34,35]. Somatostatin receptor ligand (SRL) therapy with octreotide and lanreotide, (targeting SSTR-2 and -5) and pasireotide (targeting SSTR-1, -3, and -5) has been evaluated in CNFPTs [5,36,37,38]. Table 2 summarizes the results of eight studies from the literature that evaluated the efficacy of somatostatin receptor ligands in treating CNFPTs. In total, 31/111 patients (28%) with CNFPTs treated with SRLs exhibited tumor shrinkage. The number of patients who had tumor stabilization or tumor growth was not possible to calculate as some studies did not carefully distinguish between tumor stabilization and tumor growth.
One review of 11 studies highlighted the challenges in comparing different parameters (diameter, volume, and/or area) to assess tumor size. Overall, this analysis reported that only 12% of cases treated with octreotide exhibited a reduction in tumor size, with the majority exhibiting stable tumor sizes [36]. In another study of eight patients with CNFPTs treated with octreotide 100 mcg TID for 3–6 months, 3/8 patients had tumor diameter reductions (median reduction of 5 mm), 1/8 exhibited a stable tumor, and 4/8 exhibited tumor growth (median increase of 1.5 mm, interquartile range (IQR) [1, 2]) [39]. In one study of 19 patients with CNFPTs treated with octreotide 150–300 mcg daily for 1–12 months, 15/19 patients exhibited a decreased tumor area (median reduction of 12 mm2, IQR [−6–−30]), 0/19 exhibited stable tumors, and 4/19 exhibited tumor growth (median growth of 5 mm2, IQR [2–14]). However, no statistically significant change in the average cross-sectional area was found [40]. In one study of nine patients with CNFPTs treated with octreotide 0.05 mg three times weekly to 0.6 mg daily, 3/9 had reductions in tumor size (maximum reduction of 30%), though the number of patients with stable tumors or growth was not documented [41]. A later review paper from the same authors reported two patients with tumor shrinkage, seven patients with stable tumors, and no patients with tumor growth [36]. In another study of 14 patients treated with octreotide at 300–1500 mcg per day for three months, 2/14 patients had tumor shrinkage (maximum reduction of 96%, though the treatment dosage and duration were not carefully documented) [42] and the number of patients with tumor stabilization or growth was not documented. In a further study of seven patients with CNFPTs treated with octreotide 100 mcg daily to 200 mcg three times daily for up to two months, 3/7 patients exhibited tumor shrinkage by 26–73% (median change, −38%), 3/7 had stable tumors, and 1/7 had tumor growth, by 48% [43]. The GALANT double-blind, placebo-controlled, phase 3 clinical trial compared the efficacy of 72 weeks of treatment with lanreotide 120 mg (22 patients [50%]) versus placebo (22 patients [50%]) administered every 28 days on tumor diameter and volume in 44 patients with DOTATATE-PET-positive residual CNFPTs [44]. The study found no clinically significant change (defined as an increase or decrease of ≥2 mm in diameter or ≥20% in volume) in tumor size in patients treated with lanreotide (mean [SD] change + 1.2 [2.5] mm) versus placebo (+1.3 [1.5] mm) [45]. However, 3/22 (14%), 8/22 (36%), and 11/22 (50%) of patients treated with lanreotide exhibited significant tumor shrinkage, stabilization, and growth, respectively, compared to 0/22, 13/22 (64%), and 8/22 [36%] in placebo-treated patients. The authors concluded that they could not recommend SSA therapy in the treatment of residual CNFPTs.
Table 2. Somatostatin receptor ligand (SRL) efficacy on size of clinically nonfunctional pituitary tumors (CNFTs).
Table 2. Somatostatin receptor ligand (SRL) efficacy on size of clinically nonfunctional pituitary tumors (CNFTs).
First Author (Year) ReferenceNumber of Treated Patients with CNFTs MedicationDoseDuration of Therapy (Mths)Number of Patients (%), [Median Change (IQR)] Exhibiting Tumor Shrinkage; Measurement Method Number of Patients (%) Exhibiting Stable Tumor SizeNumber of Patients (%), [Median Change (IQR)] Exhibiting Tumor Growth; Measurement Method
Katznelson (1992) [31]6Octreotide SC50 µg BID–250 µg TID 22, [insuff];
V (%)
40, [NA];
V (%)
Gasperi (1993) [39]8Octreotide SC100 µg TID3–63, [−5 (insuff)];
D (mm)
14, [1.5 (1–2)];
D (mm)
Merola (1993) [40]19Octreotide SC150–300 µg QD1–1215, [−12 (−6–−30)];
A (mm2)
04, [5 (2–14)];
A (mm2)
Plockinger (1994) [42]14Octreotide SC300–1500 µg QD32, [−76.5 (insuff)];
V (%)
120, [NA];
V (%)
Warnet (1997) [43]7 with imaging before and after treatmentOctreotide SC100 µg QD–200 µg TID23, [−38 (insuff)];
V (%)
13, [48 (insuff)];
V (%)
Colao (1999) [41]9Octreotide SC50 µg TID–600 µg QD6–123, [insuff];
V (%)
insuffinsuff, [insuff];
V (%)
Fusco (2012) [5]26Octreotide LAR20 mg Q1M37 ± 180, [NA];
D (mm)
215, [insuff];
D (mm)
Boertien (2024) [45]Lanreotide (n = 22)
Placebo (n = 22)
Lanreotide LAR SC120 mg Q28D18Lanreotide
3/22 (14%), [insuff];
D (mm), V (mm3)
Placebo
0/22 (0%), [NA];
D (mm), V (mm3)
Lanreotide
8/22 (36%), [insuff];
D (mm), V (mm3)
Placebo
14/22 (64%), [insuff];
D (mm), V (mm3)
Lanreotide
11/22 (50%), [insuff];
D (mm), V (mm3)
Placebo
8/22 (36%), [insuff];
D (mm), V (mm3)
Total
Number (%)
111 31 (28%)insuffinsuff
Legend: SSTR, somatostatin receptor; LAR, long-acting release; mg, milligrams; µg, micrograms; mm, millimeters; BID, twice a day; TID, thrice a day; QD, daily; Q1M, monthly; Mths, months; V, volume; D, dimension; A, area; NA, not applicable; IQR, interquartile range; insuff, insufficient data.
The Passion 1 (NCT01283542) open-label single arm study is assessing the efficacy and safety of pasireotide LAR 60 mg every 28 days for 24 weeks on tumor volume and hormone levels in 20 patients with CNFPTs [46].

3.3.2. Dopamine Agonist Therapy

Dopamine receptors include five subtypes, D1-5. D2 receptor agonists are highly effective in controlling prolactin secretion and tumor size in prolactinomas [47,48], and beneficial actions of D2 agonists have also been reported in GH- and ACTH-secreting pituitary tumors [49,50]. It is reported that 67% of CNFPTs express the D2 receptor.
One study summarized 27 studies between 1979 and 2016 of dopamine agonist therapy (bromocriptine, quinagolide, and/or cabergoline) for CNFPTs, reporting that 41/122 (33.6%) patients exhibited tumor shrinkage, 57/122 (46.72%) exhibited tumor stabilization, and 24/122 (19.67%) experienced tumor growth [51]. Another review of 16 articles by Cooper et al. found that CNFPTs in 30% of patients treated with dopamine agonists exhibited tumor shrinkage, 58% remained stable, and 12% exhibited tumor growth despite treatment [52].
Table 3 summarizes the results of 18 studies including 355 patients with CNFPTs treated with dopamine agonists. A total of 113/355 (32%) patients exhibited tumor shrinkage, 182/355 (51%) exhibited tumor stabilization, and 60/355 (17%) exhibited tumor growth [50].
Older studies examining the effect of bromocriptine on CNFPT size reported between 0 and 81% of patients achieved tumor reduction [53]. Unfortunately, four out of six studies did not report the exact change in tumor area, volume, or diameter [53,54,55,56,57,58]. Two additional studies found that quinagolide induced tumor shrinkage in 20–33% of 16 patients (Table 3) [59,60].
In a study of 13 patients treated with cabergoline 0.25 mg per week, 7/13 had tumor shrinkage (maximum reduction of 18%, median change of −15%, IQR [−12–−18]) [61], 5/13 had stable tumors, and 1/13 had tumor growth (median increase 25%), respectively. One study of 33 patients with visible post-surgical residual CNFPTs divided patients into a “primary prevention” group without documented tumor growth (Group I) and a secondary prevention group who had exhibited tumor growth (Group II). These patients were compared to two control groups including patients with documented tumor growth who declined treatment (Control for Group II) and matched patients who had residual tumor but had not exhibited tumor growth (Control for Group I). In the preventative group, tumor diameters reduced in 9/20 patients (45%, compared to 0% in both control groups), remained stable in 9/20 (45%), and grew in 2/20 (10%). In the secondary prevention group, tumors shrank in 15%, remained stable in 46%, and grew in 39% of patients [62]. This study suggested that early treatment with dopamine agonists may help to mitigate the growth of CNFPTs.
A further study measuring tumor diameter and volume in nine patients with post-surgical residual CNFPTs treated with cabergoline (1 to 3 mg/week) reported that 56% (median change −49.3%, IQR [−38.9–−57.2]), 0%, and 44% of the patients exhibited tumor shrinkage, tumor stabilization, and tumor growth (median change 8.7%, IQR [6.2–29.5]), respectively [50].
In a second larger study of 79 patients with ACTH- and GH-negative CNFPTs, 55 patients (preventative group) were treated after surgery with non-standardized doses of dopamine agonist therapy for residual tumors). In total, 24 patients (the remedial group) were treated with D2 agonists after initially declining therapy. No tumor growth was seen in 30% of the remedial group, 87% of the preventative group, and 47% of the untreated control group [60]. Tumor shrinkage was seen in 7 (29.2%), 21 (38.2%), and 0 (0%) of patients in the remedial, preventative, and control groups, respectively. The authors concluded that early adjunctive medical therapy with D2 agonist therapy reduced the need for additional surgery and/or radiotherapy from 47 to 16%, and tumor response did not appear to be associated with DR2 expression.
An additional randomized open-label two-year clinical trial compared cabergoline (n = 59) to no intervention (n = 57) in 116 patients with residual CNFPTs after transsphenoidal surgery [36,63]. Overall, 28.8%, 66.1%, and 5.1% of patients in the CBG-treated group demonstrated tumor shrinkage ≥ 25%, stabilization < 25%, and enlargement ≥ 25%, respectively, in comparison to (10.5% shrinkage, 74% stability, and 16% growth in the control group [p = 0.01]). Baseline patient characteristics were similar and tumor response did not appear to correlate with DR2 expression [36,59,64].
Table 3. Dopamine agonist (DA) efficacy on size of clinically nonfunctional pituitary tumors (CNFTs).
Table 3. Dopamine agonist (DA) efficacy on size of clinically nonfunctional pituitary tumors (CNFTs).
First Author (Year) ReferenceNumber of Treated Patients with CNFTs MedicationDoseDuration of Therapy (Mths)Number of Patients (%), [Median Change (IQR)] Exhibiting Tumor Shrinkage; Measurement Method Number of Patients (%) Exhibiting Stable Tumor SizeNumber of Patients (%), [Median Change (IQR)] Exhibiting Tumor Growth; Measurement Method
Wollesen (1982) [53]11BCP15–60 mg/d2–339, [−32 (−21.5–−41.5)];
A (%)
02, [14 (insuff)];
A (%)
Barrow (1984) [54]7BCP2.5–7.5 mg/d1.51, [−2.1 (insuff)];
A (cm2)
51, [0.1 (insuff)];
A (cm2)
Pullan (1985) [55]5BCP15–37.5 mg/d3–271, [insuff];
NA
40 [NA];
NA
Verde (1985) [56]20BCP7.5–20 mg/d1–321, [insuff];
NA
190 [NA];
NA
Zarate (1985) [57]7BCP15–22.5 mg/d0.5–120, [NA];
NA
70 [NA];
NA
van Schaarde-nburg (1989) [58]25
(11/25 had imaging)
BCP5–22.5 mg/d1–732, [insuff];
NA
72 [insuff];
NA
Ferone (1998) [60]6QUI300–600 µg/d6–122, [insuff];
NA
40 [NA];
NA
Colao (2000) [65]10QUI
CBG
600 µg/d (QUI)
3 mg/w (CBG)
122, [insuff];
V (%)
80 [NA];
V (%)
Nobels (2000) [59]10QUI300 µg/d36–932, [−1.8 (insuff)];
V (cm3)
[−37.3% (insuff)];
V (%)
08, [2.3 cm3 (1.1–3.5)];
V (cm3)
[31.3% (13.1–54.8)];
V (%)
Lohmann (2001) [61]13CBG0.25–1.0 mg/w127, [−15 (−12–−18)];
V (%)
51, [25 (insuff)];
V (%)
Pivonello (2004) [50]9CBG1–3 mg/w125, [−49.3 (−38.9–−57.2)];
V (%)
04, [8.7 (6.2–29.5)];
V (%)
Greenman (2005) [62]Preventative (I): n = 20
Secondary (II): n = 13
Control I (CI): n = 47
Control II (CII): n = 38
BCP
QUI
CBG
10 mg QD (BCP)
300 mg QD (QUI)
1.5 mg/week (CBG)
40 ± 48I:
9/20 (45%), [insuff];
D (mm)
II:
2/13 (15%), [insuff];
D (mm)
CI:
0 (0%), [NA];
D (mm)
CII:
0 (0%), [NA];
D (mm)
I:
9/20 (45%), [insuff];
D (mm)
II:
6/13 (46%), [insuff];
D (mm)
CI:
18/47 (38%), [insuff];
D (mm)
CII:
18/38 (47%), [insuff];
D (mm)
I:
2/20 (10%), [insuff];
D (mm)
II:
5/13 (39%), [insuff];
D (mm)
CI:
29/47 (62%), [insuff];
D (mm)
CII:
20/38 (53%), [insuff];
D (mm)
Garcia (2013) [64]19CBG2 mg/w69, [−26.7 (−16.7–−43.4);
V (%)
64, [14.8 (12.5–74.4)];
V (%)
Vieira Neto (2015) [66]9CBG3 mg/w68, [−29.2 (−18.9–−39.5)];
V (%) by 3D
[−17.3 (−9.6–−30.9)];
V (%) by Di Chiro and Nelson
01, [2.6 (insuff)];
V (%) by 3D
[0.2 (insuff)];
V (%) by Di Chiro and Nelson
Greenman (2016) [6]Preventative (P) (n = 55)
Remedial (R) (n = 24)
Control (C) (n = 60)
CBG
BCP
10 mg QD (BCP)
2 mg/w (CBG)
105.6 ± 78P:
21/55 (38%), [insuff];
D (mm)
R:
7/24 (29%), [insuff];
D (mm)
C:
0/60 (0%), [NA];
D (mm)
P:
27/55 (49%), [insuff];
D (mm)
R:
7/24 (29%), [insuff];
D (mm)
C:
28/60 (47%), [insuff];
D (mm)
P:
7/55 (13%), [insuff];
D (mm)
R:
10/24 (42%), [insuff];
D (mm)
C:
32/60 (53%), [insuff];
D (mm)
Batista (2019) [63]Medical Therapy (MT) (n = 59)
Control (C) (n = 57)
CBG0.5 mg/d24MT:
17/59 (29%), [insuff];
V (%)
C:
6/57 (11%), [insuff];
V (%)
MT:
39/59 (66%), [insuff];
V (%)
C:
42/57 (74%), [insuff];
V (%)
MT:
3/59 (5%), [insuff];
V (%)
C:
9/57 (16%), [insuff];
V (%)
Iglesias (2022) [67]Medical Therapy (CAB) (n = 22)
Observation (OBS) (n = 40)
CBG0.5–1.5 mg/w13 (10.5–17)CAB:
3/22 (14%), [insuff];
D (mm)
OBS:
2/40 (5%), [insuff];
D (mm)
CAB:
17/22 (77%), [insuff];
D (mm)
OBS:
32/40 (80%), [insuff];
D (mm)
CAB:
2/22 (9%), [insuff];
D (mm)
OBS:
6/40 (15%), [insuff];
D (mm)
Ayalon-Dangur (2024) [68]25CBG≥ 1 mg/w245, [6 (−2–−75)];
D (mm)
128, [4 (2.5–8)];
D (mm)
Total
Number (%)
355 113 (33%)182 (51%)60 (17%)
Legend: QUI, quinagolide; CBG, cabergoline; BCP, bromocriptine; mg, milligrams; µg, micrograms; QD, daily; d, day; w, week; mths, months; V, volume; D, dimension; A, area; NA, not applicable; 3D, 3-dimensional; IQR, interquartile range; insuff, insufficient data.
One study of 36 patients with CNFPTs (9/36 patients received post-operative cabergoline 3 mg/week for at least 6 months), 8/9 (88.9%) patients exhibited a tumor volume reduction using two methods (median change −29.2%, IQR [−18.9–−39.5] by three-dimensional [3D] measurements; and median change −17.3%, IQR [−9.6–−30.9] by Di Chiro and Nelson measurements), 0 patients exhibited tumor stabilization, and 1 patient exhibited tumor growth (2.6% by 3D and 0.2% by Di Chiro and Nelson). Significant tumor volume reduction was arbitrarily defined as a reduction in tumor volume ≥25%, which was observed in six of nine (67%) patients [66].
In another study where 22/62 patients with residual CNFPTs (35.5%) were treated with cabergoline (compared with a placebo in 40/62 (64.5%)) [67], 3/22 (13.6%), 17/22 (77.2%), and 2/22 (9.1%) patients experienced tumor shrinkage, stabilization, and growth, respectively, compared to 2/40 (5%), 32/40 (80%), and 6/40 (15%) in the placebo group [4].
A recent study examined 25 surgery-naive patients with CNFPTs ≥1 cm treated with cabergoline ≥1 mg per week for at least 24 months (range 1–2.5 mg/week, [median 1.5 mg/week]). Change in tumor size was defined as ≥2 mm. In total, 5/25 patients (20%), 12/25 (48%), and 8/25 (32%) exhibited tumor size reduction (median change −6 mm, IQR [−2–−7.5]), tumor stabilization, and tumor growth (median increase 4 mm, IQR [2.5–8]), respectively. A total of 6/7 patients (28%) had elevated prolactin levels (1.5–2.5-times the upper limit of normal) and a further patient had a four-fold elevation in the serum prolactin level [68].
Some studies have examined combination therapy of SRLs with D2 agonists. In one study of 10 patients with CNFPTs treated with octreotide 200 mcg TID and cabergoline 0.5 mg daily, 6/10 patients exhibited a mean tumor shrinkage of 30% ± 4% with a range of 18–46%, tumor stabilization was seen in 3/10, and tumor growth in 1/10 patients [69]. There is an ongoing phase 2 open-label clinical trial (NCT01620138) that will compare tumor responses to combination therapy with cabergoline (3 mg/week) and pasireotide (900 mcg BID) for 6 months in 21 patients with CNFPTs.

3.3.3. GnRH Analog Therapy

As discussed above, the majority of CNFPTs express gonadotropins or their α- or β-subunits, and so GnRH analogs have also been explored as a treatment for CNFPTs. Data on the effect of GnRH analogs on CNFPTs is not well described in the literature, and the existing studies point to an equivocal effect on circulating α-subunit or gonadotrophin levels [30,36,70,71,72,73,74,75,76]. Changes in tumor size were not carefully documented.

3.3.4. Peptide Receptor Radionuclide Therapy and Temozolomide

Peptide receptor radionuclide therapy (PRRT) involves linking the radionuclide lutetium-177 to a targeting somatostatin analog, and it is effective in treating a variety of neuroendocrine tumors [77]. PRRT using lutetium-177 linked to octreotide has been used in six “aggressive” CNFPTs, where three out of six PRRT-treated patients with CNFPTs exhibited stable disease, two had progressive disease, and one patient died [78].
Finally, the alkylating chemotherapeutic agent temozolomide has been used in the treatment of pituitary carcinomas and “aggressive” pituitary tumors of various subtypes including CNFPTs. There is some suggestion that CNFPTs may be less responsive to temozolomide based on one study in 110 patients where only 17% of patients with CNFPTs demonstrated tumor regression compared to 45% of patients with functional pituitary tumors [79]. A second study summarizing eight prior case series of 100 patients with pituitary tumors treated with temozolomide found that of 27 patients with CNFPTs, 7 (22%) demonstrated tumor regression, 13 (48%) had stable disease, and 8 (30%) had tumor growth [80]. One retrospective study of 28 patients with aggressive pituitary tumors treated with temozolomide after surgery, 15 of whom had CNFPTs [81], found that 2/15 (13.3%), 9/15 (60%), and 3/15 (20%) patients exhibited a partial response, tumor stabilization, and tumor growth, respectively. Insufficient data was available in the final patient. In total, 4/15 patients received medical therapy with cabergoline during the course of their treatment.

3.4. Emerging Experimental Novel Treatment Modalities of CNFPT

3.4.1. Immune Checkpoint Inhibitor Therapy

The use of immune checkpoint inhibitors (ICIs) targeting programmed death-ligand (PD-L1) and others has proven highly effective in treating several cancers including lung cancer and melanoma. To date, limited studies have examined the use of ICIs such as PD-1/PD-L1 inhibitors in pituitary neuroendocrine tumors and have been mostly limited to refractory pituitary tumors. A review of 29 studies of patients with pituitary neuroendocrine tumors treated with a variety of ICIs included four patients with CNFPTs, three of whom had metastatic disease. Two of these four patients (50%) responded partially with radiographic improvement in response to therapy. The other two patients (50%) exhibited stable disease without improvement, and none exhibited disease progression [82]. Further studies with larger sample sizes are needed to clarify the efficacy of immunotherapy in CNFPTs.

3.4.2. Folate Receptor-Mediated Drug Targeting

Folate receptors (FR α, β, and γ) are cysteine-rich glycosyl-phosphatidyl-inositol-anchored membrane proteins that bind folic acid and folate conjugates, which can then facilitate de novo nucleotide synthesis, DNA-methylation, and DNA-damage repair [83]. When cognate folic acid binds the FR, the receptor–ligand complex clusters into lipid rafts, initiating a clathrin-independent, caveolae-mediated endocytosis process [84]. The plasma membrane then invaginates to form vesicles (endosomes) containing the FR–folate complex [85]. As the endosome matures, the acidic environment causes folate to dissociate from the receptor, which subsequently is recycled back to the cell surface for another round of uptake [85]. This highly efficient and selective process is frequently exploited in cancer therapy and imaging [85]. FR expression is typically restricted to the placenta, lung, kidneys, and choroid plexus, but is overexpressed in many malignancies [86]. Increased FRα mRNA and protein expression has been reported in CNFPTs compared to hormonally active pituitary tumors [87]. FRα overexpression in murine αT3 gonadotroph tumor cells in vitro increased S-phase cells and cell proliferation, in part by activation of the NOTCH3 pathway [88]. In vivo imaging using a 99 mTc-Folate SPECT/CT demonstrated high FR expression in patients with CNFPTs [89]. FRα-targeted liposomal doxorubicin treatment was also shown to inhibit proliferation and invasion in primary cultures of human CNFPTs [90,91,92], suggesting that FR-mediated tumor targeting could be a feasible novel treatment option for CNFPTs and warrant further investigation (Figure 1) [93].

3.4.3. mTOR Inhibitors

Activation of the AKT/mTOR pathway in human CNFPTs has been identified by transcriptomic and proteomic profiling approaches (Figure 1) [94,95]. Treatment with the mTOR inhibitors everolimus and rapamycin inhibited in vitro proliferation in 35–70% of primary cultures of CNFPTs when used as a monotherapy [96,97], and co-treatment with somatostatin analogs (octreotide and SOM230) appeared to potentiate these mTOR anti-proliferative effects [97,98]. Up to now, isolated case reports have described the use of the mTOR inhibitor everolimus in treating refractory corticotrophs [99,100,101,102], but the efficacy of mTOR inhibition in CNFPTs is still undetermined [103].

3.4.4. Targeting Tumor Angiogenesis

The normal anterior and posterior lobes of the pituitary gland are highly vascular and supplied by the superior and inferior hypophyseal arteries, respectively [103]. The portal hypophyseal veins transport endocrine hormones from the pituitary into the peripheral circulation [103]. The microvascular density of pituitary carcinomas is reportedly higher than in the normal pituitary gland, suggesting that inhibiting angiogenesis could be a way to treat some pituitary tumors [104,105]. Bevacizumab, a vascular endothelial-derived growth factor (VEGF) inhibitor, apatinib, a VEGFR2 inhibitor, and sunitinib, an oral tyrosine kinase inhibitor, are all anti-angiogenic agents that have been used to treat pituitary tumors (Figure 1) [106]. In one study of three patients with CNFPTs previously treated with temozolomide, the progression-free survival following bevacizumab ranged from 9 to 26 months [107].

4. Discussion

We describe the natural history of these tumors, summarize currently available therapeutic options and their efficacy, and discuss how the diversity of CNFPT subtypes potentially complicates the discovery of new effective medical therapies. The interpretation and statistical analyses of many of these studies are challenging due to varying definitions of therapeutic response, differing treatment doses and durations, variable methods of tumor size measurement, and missing data points, all of which contribute to inconsistent findings from these mostly open-label studies.

5. Conclusions

We conclude that robust, blinded, randomized placebo-controlled clinical trials to study the efficacy and safety of SRLs and D2 agonists in CNFPTs are needed with parallel efforts to identify novel agents and potentially study some of the additional targets discussed, such as inhibitors of angiogenic pathways, folate uptake, and/or the mTOR pathway.

Author Contributions

Original draft, data curation, investigation, writing, editing, N.M.; software, writing—original draft, investigation, resources, data curation, D.Z.; primary investigator, corresponding author, writing—review and editing, conceptualization, formal analysis, methodology, project administration, supervision, validation, A.P.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created in the publication of this review.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CNFPTsClinically nonfunctioning pituitary tumors
NFPAsNonfunctioning pituitary adenomas
RTRadiation therapy
SSTRSomatostatin receptor
SF-1Steroidogenic factor 1
FSHFollicular stimulating hormone
LHLuteinizing hormone
ACTHAdrenocorticotropic hormone
POMCProopiomelanocortin
GHGrowth hormone
IGF-1Insulin like growth factor-1 (IGF-1)
TSHThyroid stimulating hormone
GATAGATA binding protein
ERαEstrogen receptor alpha
PRLProlactin
SYPSynaptophysin
GTRGross total resection
DRDopamine receptor
GnRHGonadotrophin-releasing hormone
VEGFVascular endothelial growth factor
SRLSomatostatin receptor ligand
IQRInterquartile range
FRFolate receptors
LARLong-acting release
BIDTwice daily
TIDThrice daily

References

  1. Ntali, G.; Wass, J.A. Epidemiology, clinical presentation and diagnosis of non-functioning pituitary adenomas. Pituitary 2018, 21, 111–118. [Google Scholar] [CrossRef]
  2. Molitch, M.E. Diagnosis and Treatment of Pituitary Adenomas: A Review. JAMA 2017, 317, 516–524. [Google Scholar] [CrossRef] [PubMed]
  3. Chen, L.; White, W.L.; Spetzler, R.F.; Xu, B. A prospective study of nonfunctioning pituitary adenomas: Presentation, management, and clinical outcome. J. Neuro-Oncol. 2011, 102, 129–138. [Google Scholar] [CrossRef] [PubMed]
  4. Ferrante, E.; Ferraroni, M.; Castrignanò, T.; Menicatti, L.; Anagni, M.; Reimondo, G.; Del Monte, P.; Bernasconi, D.; Loli, P.; Faustini-Fustini, M.; et al. Non-functioning pituitary adenoma database: A useful resource to improve the clinical management of pituitary tumors. Eur. J. Endocrinol. 2006, 155, 823–829. [Google Scholar] [CrossRef] [PubMed]
  5. Fusco, A.; Giampietro, A.; Bianchi, A.; Cimino, V.; Lugli, F.; Piacentini, S.; Lorusso, M.; Tofani, A.; Perotti, G.; Lauriola, L.; et al. Treatment with octreotide LAR in clinically non-functioning pituitary adenoma: Results from a case-control study. Pituitary 2012, 15, 571–578. [Google Scholar] [CrossRef]
  6. Greenman, Y.; Cooper, O.; Yaish, I.; Robenshtok, E.; Sagiv, N.; Jonas-Kimchi, T.; Yuan, X.; Gertych, A.; Shimon, I.; Ram, Z.; et al. Treatment of clinically nonfunctioning pituitary adenomas with dopamine agonists. Eur. J. Endocrinol. 2016, 175, 63–72. [Google Scholar] [CrossRef]
  7. Osborn, A.G.; Louis, D.N.; Poussaint, T.Y.; Linscott, L.L.; Salzman, K.L. The 2021 World Health Organization Classification of Tumors of the Central Nervous System: What Neuroradiologists Need to Know. Am. J. Neuroradiol. 2022, 43, 928–937. [Google Scholar] [CrossRef]
  8. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef]
  9. Larkin, S.; Ansorge, O. Pathology and pathogenesis of pituitary adenomas and other sellar lesions. In Endotext; MDText.com, Inc.: South Dartmouth, MA, USA, 2017. [Google Scholar]
  10. Mete, O.; Cintosun, A.; Pressman, I.; Asa, S.L. Epidemiology and biomarker profile of pituitary adenohypophysial tumors. Mod. Pathol. 2018, 31, 900–909. [Google Scholar] [CrossRef]
  11. Nielsen, S.; Mellemkjaer, S.; Rasmussen, L.M.; Ledet, T.; Olsen, N.; Bojsen-Møller, M.; Astrup, J.; Weeke, J.; Jørgensen, J.O. Expression of somatostatin receptors on human pituitary adenomas in vivo and ex vivo. J. Endocrinol. Investig. 2001, 24, 430–437. [Google Scholar] [CrossRef]
  12. Zhang, D.; Hugo, W.; Bergsneider, M.; Wang, M.B.; Kim, W.; Vinters, H.V.; Heaney, A.P. Single-cell RNA sequencing in silent corticotroph tumors confirms impaired POMC processing and provides new insights into their invasive behavior. Eur. J. Endocrinol. 2022, 187, 49–64. [Google Scholar] [CrossRef] [PubMed]
  13. Langlois, F.; Woltjer, R.; Cetas, J.S.; Fleseriu, M. Silent somatotroph pituitary adenomas: An update. Pituitary 2018, 21, 194–202. [Google Scholar] [CrossRef] [PubMed]
  14. Chinezu, L.; Vasiljevic, A.; Trouillas, J.; Lapoirie, M.; Jouanneau, E.; Raverot, G. Silent somatotroph tumour revisited from a study of 80 patients with and without acromegaly and a review of the literature. Eur. J. Endocrinol. 2017, 176, 195–201. [Google Scholar] [CrossRef]
  15. Bano, G. Somatotroph adenomas: Histological subtypes and predicted response to treatment. Int. J. Endocr. Oncol. 2020, 7, IJE29. [Google Scholar] [CrossRef]
  16. Glynn, N.; Hannon, A.M.; Farrell, M.; Brett, F.; Javadpour, M.; Agha, A. Variable Thyroid-Stimulating Hormone Dynamics in ‘Silent’ Thyrotroph Adenomas. AACE Clin. Case Rep. 2016, 2, e155–e160. [Google Scholar] [CrossRef]
  17. Manojlovic-Gacic, E.; Engström, B.E.; Casar-Borota, O. Histopathological classification of non-functioning pituitary neuroendocrine tumors. Pituitary 2018, 21, 119–129. [Google Scholar] [CrossRef]
  18. Kuzu, F.; Bayraktaroğlu, T.; Zor, F.; Gün, B.; Salihoğlu, Y.S.; Kalaycı, M. A thyrotropin-secreting macroadenoma with positive growth hormone and prolactin immunostaining: A case report and literature review. Niger. J. Clin. Pract. 2015, 18, 693–697. [Google Scholar] [CrossRef] [PubMed]
  19. Drummond, J.; Roncaroli, F.; Grossman, A.B.; Korbonits, M. Clinical and Pathological Aspects of Silent Pituitary Adenomas. J. Clin. Endocrinol. Metab. 2019, 104, 2473–2489. [Google Scholar] [CrossRef]
  20. Asa, S.L.; Mete, O.; Perry, A.; Osamura, R.Y. Overview of the 2022 WHO Classification of Pituitary Tumors. Endocr. Pathol. 2022, 33, 6–26. [Google Scholar] [CrossRef]
  21. Lopes, M.B.S. The 2017 World Health Organization classification of tumors of the pituitary gland: A summary. Acta Neuropathol. 2017, 134, 521–535. [Google Scholar] [CrossRef]
  22. Drummond, J.B.; Ribeiro-Oliveira, A., Jr.; Soares, B.S. Non-Functioning Pituitary Adenomas. In Endotext; Feingold, K.R., Anawalt, B., Blackman, M.R., Boyce, A., Chrousos, G., Corpas, E., de Herder, W.W., Dhatariya, K., Dungan, K., Hofland, J., et al., Eds.; MDText.com, Inc.: South Dartmouth, MA, USA, 2000. [Google Scholar]
  23. Huang, W.; Molitch, M.E. Management of nonfunctioning pituitary adenomas (NFAs): Observation. Pituitary 2018, 21, 162–167. [Google Scholar] [CrossRef] [PubMed]
  24. Murad, M.H.; Fernández-Balsells, M.M.; Barwise, A.; Gallegos-Orozco, J.F.; Paul, A.; Lane, M.A.; Lampropulos, J.F.; Natividad, I.; Perestelo-Pérez, L.; Ponce de León-Lovatón, P.G.; et al. Outcomes of surgical treatment for nonfunctioning pituitary adenomas: A systematic review and meta-analysis. Clin. Endocrinol. 2010, 73, 777–791. [Google Scholar] [CrossRef]
  25. Reddy, R.; Cudlip, S.; Byrne, J.V.; Karavitaki, N.; Wass, J.A. Can we ever stop imaging in surgically treated and radiotherapy-naive patients with non-functioning pituitary adenoma? Eur. J. Endocrinol. 2011, 165, 739–744. [Google Scholar] [CrossRef]
  26. Øystese, K.A.; Evang, J.A.; Bollerslev, J. Non-functioning pituitary adenomas: Growth and aggressiveness. Endocrine 2016, 53, 28–34. [Google Scholar] [CrossRef] [PubMed]
  27. Minniti, G.; Clarke, E.; Scaringi, C.; Enrici, R.M. Stereotactic radiotherapy and radiosurgery for non-functioning and secreting pituitary adenomas. Rep. Pr. Pract. Oncol. Radiother. 2016, 21, 370–378. [Google Scholar] [CrossRef] [PubMed]
  28. Dekkers, O.M.; Pereira, A.M.; Romijn, J.A. Treatment and follow-up of clinically nonfunctioning pituitary macroadenomas. J. Clin. Endocrinol. Metab. 2008, 93, 3717–3726. [Google Scholar] [CrossRef]
  29. Lucas, J.W.; Bodach, M.E.; Tumialan, L.M.; Oyesiku, N.M.; Patil, C.G.; Litvack, Z.; Aghi, M.K.; Zada, G. Congress of Neurological Surgeons Systematic Review and Evidence-Based Guideline on Primary Management of Patients with Nonfunctioning Pituitary Adenomas. Neurosurgery 2016, 79, E533–E535. [Google Scholar] [CrossRef]
  30. Zatelli, M.C.; Piccin, D.; Vignali, C.; Tagliati, F.; Ambrosio, M.R.; Bondanelli, M.; Cimino, V.; Bianchi, A.; Schmid, H.A.; Scanarini, M.; et al. Pasireotide, a multiple somatostatin receptor subtypes ligand, reduces cell viability in non-functioning pituitary adenomas by inhibiting vascular endothelial growth factor secretion. Endocr. Relat. Cancer 2007, 14, 91–102. [Google Scholar] [CrossRef]
  31. Katznelson, L.; Oppenheim, D.S.; Coughlin, J.F.; Kliman, B.; Schoenfeld, D.A.; Klibanski, A. Chronic somatostatin analog administration in patients with alpha-subunit-secreting pituitary tumors. J. Clin. Endocrinol. Metab. 1992, 75, 1318–1325. [Google Scholar] [PubMed]
  32. Taboada, G.F.; Luque, R.M.; Bastos, W.; Guimarães, R.F.; Marcondes, J.B.; Chimelli, L.M.; Fontes, R.; Mata, P.J.; Filho, P.N.; Carvalho, D.P.; et al. Quantitative analysis of somatostatin receptor subtype (SSTR1–5) gene expression levels in somatotropinomas and non-functioning pituitary adenomas. Eur. J. Endocrinol. 2007, 156, 65–74. [Google Scholar] [CrossRef]
  33. Gabalec, F.; Drastikova, M.; Cesak, T.; Netuka, D.; Masopust, V.; Machac, J.; Marek, J.; Cap, J.; Beranek, M. Dopamine 2 and somatostatin 1-5 receptors coexpression in clinically non-functioning pituitary adenomas. Physiol. Res. 2015, 64, 369–377. [Google Scholar] [CrossRef] [PubMed]
  34. Neto, L.V.; Machado Ede, O.; Luque, R.M.; Taboada, G.F.; Marcondes, J.B.; Chimelli, L.M.; Quintella, L.P.; Niemeyer, P., Jr.; de Carvalho, D.P.; Kineman, R.D.; et al. Expression analysis of dopamine receptor subtypes in normal human pituitaries, nonfunctioning pituitary adenomas and somatotropinomas, and the association between dopamine and somatostatin receptors with clinical response to octreotide-LAR in acromegaly. J. Clin. Endocrinol. Metab. 2009, 94, 1931–1937. [Google Scholar] [CrossRef]
  35. Tateno, T.; Kato, M.; Tani, Y.; Oyama, K.; Yamada, S.; Hirata, Y. Differential expression of somatostatin and dopamine receptor subtype genes in adrenocorticotropin (ACTH)-secreting pituitary tumors and silent corticotroph adenomas. Endocr. J. 2009, 56, 579–584. [Google Scholar] [CrossRef] [PubMed]
  36. Colao, A.; Di Somma, C.; Pivonello, R.; Faggiano, A.; Lombardi, G.; Savastano, S. Medical therapy for clinically non-functioning pituitary adenomas. Endocr. Relat. Cancer 2008, 15, 905–915. [Google Scholar] [CrossRef] [PubMed]
  37. Snyder, P.J. Gonadotroph cell adenomas of the pituitary. Endocr. Rev. 1985, 6, 552–563. [Google Scholar] [CrossRef]
  38. Bevan, J.S. Clinical review: The antitumoral effects of somatostatin analog therapy in acromegaly. J. Clin. Endocrinol. Metab. 2005, 90, 1856–1863. [Google Scholar] [CrossRef]
  39. Gasperi, M.; Petrini, L.; Pilosu, R.; Nardi, M.; Marcello, A.; Mastio, F.; Bartalena, L.; Martino, E. Octreotide treatment does not affect the size of most non-functioning pituitary adenomas. J. Endocrinol. Investig. 1993, 16, 541–543. [Google Scholar] [CrossRef]
  40. Merola, B.; Colao, A.; Ferone, D.; Selleri, A.; Di Sarno, A.; Marzullo, P.; Biondi, B.; Spaziante, R.; Rossi, E.; Lombardi, G. Effects of a chronic treatment with octreotide in patients with functionless pituitary adenomas. Horm. Res. 1993, 40, 149–155. [Google Scholar] [CrossRef]
  41. Colao, A.; Lastoria, S.; Ferone, D.; Varrella, P.; Marzullo, P.; Pivonello, R.; Cerbone, G.; Acampa, W.; Salvatore, M.; Lombardi, G. The pituitary uptake of 111In-DTPA-D-Phe1-octreotide in the normal pituitary and in pituitary adenomas. J. Endocrinol. Investig. 1999, 22, 176–183. [Google Scholar] [CrossRef]
  42. Plöckinger, U.; Reichel, M.; Fett, U.; Saeger, W.; Quabbe, H.J. Preoperative octreotide treatment of growth hormone-secreting and clinically nonfunctioning pituitary macroadenomas: Effect on tumor volume and lack of correlation with immunohistochemistry and somatostatin receptor scintigraphy. J. Clin. Endocrinol. Metab. 1994, 79, 1416–1423. [Google Scholar]
  43. Warnet, A.; Harris, A.G.; Renard, E.; Martin, D.; James-Deidier, A.; Chaumet-Riffaud, P. A prospective multicenter trial of octreotide in 24 patients with visual defects caused by nonfunctioning and gonadotropin-secreting pituitary adenomas. French Multicenter Octreotide Study Group. Neurosurgery 1997, 41, 786–795. [Google Scholar] [CrossRef] [PubMed]
  44. Boertien, T.M.; Drent, M.L.; Booij, J.; Majoie, C.; Stokkel, M.P.M.; Hoogmoed, J.; Pereira, A.; Biermasz, N.R.; Simsek, S.; Groote Veldman, R.; et al. The GALANT trial: Study protocol of a randomised placebo-controlled trial in patients with a 68Ga-DOTATATE PET-positive, clinically non-functioning pituitary macroadenoma on the effect of lan reotide on tumour size. BMJ Open 2020, 10, e038250. [Google Scholar] [CrossRef] [PubMed]
  45. Boertien, T.M.; Drent, M.L.; Booij, J.; Majoie, C.B.L.M.; Stokkel, M.P.M.; Hoogmoed, J.; Pereira, A.M.; Biermasz, N.R.; Simsek, S.; Veldman, R.G.; et al. Lanreotide versus placebo for tumour reduction in patients with a 68Ga-DOTATATE PET-positive, clinically non-functioning pituitary macroadenoma (GALANT study): A randomised, multicentre, phase 3 trial with blinded outcome assessment. Lancet Reg. Health Eur. 2024, 42, 100923. [Google Scholar] [CrossRef]
  46. Gulde, S.; Wiedemann, T.; Schillmaier, M.; Valença, I.; Lupp, A.; Steiger, K.; Yen, H.Y.; Bäuerle, S.; Notni, J.; Luque, R.; et al. Gender-Specific Efficacy Revealed by Head-to-Head Comparison of Pasireotide and Octreotide in a Representative In Vivo Model of Nonfunctioning Pituitary Tumors. Cancers 2021, 13, 3097. [Google Scholar] [CrossRef] [PubMed]
  47. Colao, A.; Di Sarno, A.; Landi, M.L.; Cirillo, S.; Sarnacchiaro, F.; Facciolli, G.; Pivonello, R.; Cataldi, M.; Merola, B.; Annunziato, L.; et al. Long-term and low-dose treatment with cabergoline induces macroprolactinoma shrinkage. J. Clin. Endocrinol. Metab. 1997, 82, 3574–3579. [Google Scholar] [CrossRef]
  48. Nissim, M.; Ambrosi, B.; Bernasconi, V.; Giannattasio, G.; Giovanelli, M.A.; Bassetti, M.; Vaccari, U.; Moriondo, P.; Spada, A.; Travaglini, P.; et al. Bromocriptine treatment of macroprolactinomas: Studies on the time course of tumor shrinkage and morphology. J. Endocrinol. Investig. 1982, 5, 409–415. [Google Scholar] [CrossRef]
  49. Bression, D.; Brandi, A.M.; Nousbaum, A.; Le Dafniet, M.; Racadot, J.; Peillon, F. Evidence of dopamine receptors in human growth hormone (GH)-secreting adenomas with concomitant study of dopamine inhibition of GH secretion in a perifusion system. J. Clin. Endocrinol. Metab. 1982, 55, 589–593. [Google Scholar] [CrossRef]
  50. Pivonello, R.; Matrone, C.; Filippella, M.; Cavallo, L.M.; Di Somma, C.; Cappabianca, P.; Colao, A.; Annunziato, L.; Lombardi, G. Dopamine receptor expression and function in clinically nonfunctioning pituitary tumors: Comparison with the effectiveness of cabergoline treatment. J. Clin. Endocrinol. Metab. 2004, 89, 1674–1683. [Google Scholar] [CrossRef]
  51. Capatina, C.; Poiana, C. Dopamine Agonists in the Management of Non-Functioning Pituitary Adenomas. Acta Endocrinol. 2021, 17, 377–382. [Google Scholar] [CrossRef]
  52. Cooper, O.; Greenman, Y. Dopamine Agonists for Pituitary Adenomas. Front. Endocrinol. 2018, 9, 469. [Google Scholar] [CrossRef]
  53. Wollesen, F.; Andersen, T.; Karle, A. Size reduction of extrasellar pituitary tumors during bromocriptine treatment. Ann. Intern. Med. 1982, 96, 281–286. [Google Scholar] [CrossRef] [PubMed]
  54. Barrow, D.L.; Tindall, G.T.; Kovacs, K.; Thorner, M.O.; Horvath, E.; Hoffman, J.C., Jr. Clinical and pathological effects of bromocriptine on prolactin-secreting and other pituitary tumors. J. Neurosurg. 1984, 60, 1–7. [Google Scholar] [CrossRef]
  55. Pullan, P.T.; Carroll, W.M.; Chakera, T.M.; Khangure, M.S.; Vaughan, R.J. Management of extra-sellar pituitary tumours with bromocriptine: Comparison of prolactin secreting and non-functioning tumours using half-field visual evoked potentials and computerised tomography. Aust. N. Z. J. Med. 1985, 15, 203–208. [Google Scholar] [CrossRef]
  56. Verde, G.; Oppizzi, G.; Chiodini, P.G.; Dallabonzana, D.; Luccarelli, G.; Liuzzi, A. Effect of chronic bromocriptine administration on tumor size in patients with nonsecreting pituitary adenomas. J. Endocrinol. Investig. 1985, 8, 113–115. [Google Scholar] [CrossRef] [PubMed]
  57. Zárate, A.; Morán, C.; Klériga, E.; Loyo, M.; González-Angulo, A.; Aquilar-Parada, E. Bromocriptine therapy as pre-operative adjunct of non-functional pituitary macroadenomas. Acta Endocrinol. 1985, 108, 445–450. [Google Scholar] [CrossRef]
  58. van Schaardenburg, D.; Roelfsema, F.; van Seters, A.P.; Vielvoye, G.J. Bromocriptine therapy for non-functioning pituitary adenoma. Clin. Endocrinol. 1989, 30, 475–484. [Google Scholar] [CrossRef] [PubMed]
  59. Nobels, F.R.; de Herder, W.W.; van den Brink, W.M.; Kwekkeboom, D.J.; Hofland, L.J.; Zuyderwijk, J.; de Jong, F.H.; Lamberts, S.W. Long-term treatment with the dopamine agonist quinagolide of patients with clinically non-functioning pituitary adenoma. Eur. J. Endocrinol. 2000, 143, 615–621. [Google Scholar] [CrossRef]
  60. Ferone, D.; Lastoria, S.; Colao, A.; Varrella, P.; Cerbone, G.; Acampa, W.; Merola, B.; Salvatore, M.; Lombardi, G. Correlation of scintigraphic results using 123I-methoxybenzamide with hormone levels and tumor size response to quinagolide in patients with pituitary adenomas. J. Clin. Endocrinol. Metab. 1998, 83, 248–252. [Google Scholar] [CrossRef]
  61. Lohmann, T.; Trantakis, C.; Biesold, M.; Prothmann, S.; Guenzel, S.; Schober, R.; Paschke, R. Minor tumour shrinkage in nonfunctioning pituitary adenomas by long-term treatment with the dopamine agonist cabergoline. Pituitary 2001, 4, 173–178. [Google Scholar] [CrossRef]
  62. Greenman, Y.; Tordjman, K.; Osher, E.; Veshchev, I.; Shenkerman, G.; Reider, G., II.; Segev, Y.; Ouaknine, G.; Stern, N. Postoperative treatment of clinically nonfunctioning pituitary adenomas with dopamine agonists decreases tumour remnant growth. Clin. Endocrinol. 2005, 63, 39–44. [Google Scholar] [CrossRef]
  63. Batista, R.L.; Musolino, N.R.C.; Cescato, V.A.S.; da Silva, G.O.; Medeiros, R.S.S.; Herkenhoff, C.G.B.; Trarbach, E.B.; Cunha-Neto, M.B. Cabergoline in the Management of Residual Nonfunctioning Pituitary Adenoma: A Single-Center, Open-Label, 2-Year Randomized Clinical Trial. Am. J. Clin. Oncol. 2019, 42, 221–227. [Google Scholar] [CrossRef] [PubMed]
  64. Garcia, E.C.; Naves, L.A.; Silva, A.O.; de Castro, L.F.; Casulari, L.A.; Azevedo, M.F. Short-term treatment with cabergoline can lead to tumor shrinkage in patients with nonfunctioning pituitary adenomas. Pituitary 2013, 16, 189–194. [Google Scholar] [CrossRef] [PubMed]
  65. Colao, A.; Ferone, D.; Lastoria, S.; Cerbone, G.; Di Sarno, A.; Di Somma, C.; Lucci, R.; Lombardi, G. Hormone levels and tumour size response to quinagolide and cabergoline in patients with prolactin-secreting and clinically non-functioning pituitary adenomas: Predictive value of pituitary scintigraphy with 123I-methoxybenzamide. Clin. Endocrinol. 2000, 52, 437–445. [Google Scholar] [CrossRef] [PubMed]
  66. Vieira Neto, L.; Wildemberg, L.E.; Moraes, A.B.; Colli, L.M.; Kasuki, L.; Marques, N.V.; Gasparetto, E.L.; de Castro, M.; Takiya, C.M.; Gadelha, M.R. Dopamine receptor subtype 2 expression profile in nonfunctioning pituitary adenomas and in vivo response to cabergoline therapy. Clin. Endocrinol. 2015, 82, 739–746. [Google Scholar] [CrossRef] [PubMed]
  67. Iglesias, P.; Biagetti, B.; Araujo-Castro, M.; Alcázar, V.; Guerrero-Pérez, F.; Rivero, N.; Casteràs, A.; Gómez, C.G.; Izquierdo, B.G.; Torres, V.V.; et al. Effect of cabergoline on tumor remnant after surgery in nonfunctioning pituitary adenoma. J. Neuro-Oncol. 2022, 160, 351–359. [Google Scholar] [CrossRef]
  68. Ayalon-Dangur, I.; Turjeman, A.; Hirsch, D.; Robenshtok, E.; Tsvetov, G.; Gorshtein, A.; Masri, H.; Shraga-Slutzky, I.; Manisterski, Y.; Akirov, A.; et al. Cabergoline treatment for surgery-naïve non-functioning pituitary macroadenomas. Pituitary 2024, 27, 52–60. [Google Scholar] [CrossRef]
  69. Andersen, M.; Bjerre, P.; Schrøder, H.D.; Edal, A.; Høilund-Carlsen, P.F.; Pedersen, P.H.; Hagen, C. In vivo secretory potential and the effect of combination therapy with octreotide and cabergoline in patients with clinically non-functioning pituitary adenomas. Clin. Endocrinol. 2001, 54, 23–30. [Google Scholar] [CrossRef]
  70. Roman, S.H.; Goldstein, M.; Kourides, I.A.; Comite, F.; Bardin, C.W.; Krieger, D.T. The luteinizing hormone-releasing hormone (LHRH) agonist [D-Trp6-Pro9-NEt]LHRH increased rather than lowered LH and α-subunit levels in a patient with an LH-secreting pituitary tumor. J. Clin. Endocrinol. Metab. 1984, 58, 313–319. [Google Scholar] [CrossRef]
  71. Chapman, A.J.; MacFarlane, I.A.; Shalet, S.M.; Beardwell, C.G.; Dutton, J.; Sutton, M.L. Discordant serum α-subunit and FSH concentrations in a woman with a pituitary tumour. Clin. Endocrinol. 1984, 21, 123–129. [Google Scholar] [CrossRef]
  72. Colombo, P.; Ambrosi, B.; Saccomanno, K.; Bassetti, M.; Cortelazzi, D.; Faglia, G. Effects of long-term treatment with the gonadotropin-releasing hormone analog nafarelin in patients with non-functioning pituitary adenomas. Eur. J. Endocrinol. 1994, 130, 339–345. [Google Scholar] [CrossRef]
  73. Daneshdoost, L.; Pavlou, S.N.; Molitch, M.E.; Gennarelli, T.A.; Savino, P.J.; Sergott, R.C.; Bosley, T.M.; River, J.E.; Vale, W.W.; Snyder, P.J. Inhibition of follicle-stimulating hormone secretion from gonadotroph adenomas by repetitive administration of a gonadotropin-releasing hormone antagonist. J. Clin. Endocrinol. Metab. 1990, 71, 92–97. [Google Scholar] [CrossRef] [PubMed]
  74. Klibanski, A.; Deutsch, P.J.; Jameson, J.L.; Ridgway, E.C.; Crowley, W.F.; Hsu, D.W.; Habener, J.F.; Black, P.M. Luteinizing hormone-secreting pituitary tumor: Biosynthetic characterization and clinical studies. J. Clin. Endocrinol. Metab. 1987, 64, 536–542. [Google Scholar] [CrossRef] [PubMed]
  75. Klibanski, A.; Jameson, J.L.; Biller, B.M.; Crowley, W.F., Jr.; Zervas, N.T.; Rivier, J.; Vale, W.W.; Bikkal, H. Gonadotropin and alpha-subunit responses to chronic gonadotropin-releasing hormone analog administration in patients with glycoprotein hormone-secreting pituitary tumors. J. Clin. Endocrinol. Metab. 1989, 68, 81–86. [Google Scholar] [CrossRef] [PubMed]
  76. Sassolas, G.; Lejeune, H.; Trouillas, J.; Forest, M.G.; Claustrat, B.; Lahlou, N.; Loras, B. Gonadotropin-releasing hormone agonists are unsuccessful in reducing tumoral gonadotropin secretion in two patients with gonadotropin-secreting pituitary adenomas. J. Clin. Endocrinol. Metab. 1988, 67, 180–185. [Google Scholar] [CrossRef] [PubMed]
  77. Ito, T.; Jensen, R.T. Perspectives on the current pharmacotherapeutic strategies for management of functional neuroendocrine tumor syndromes. Expert. Opin. Pharmacother. 2021, 22, 685–693. [Google Scholar] [CrossRef]
  78. Maclean, J.; Aldridge, M.; Bomanji, J.; Short, S.; Fersht, N. Peptide receptor radionuclide therapy for aggressive atypical pituitary adenoma/carcinoma: Variable clinical response in preliminary evaluation. Pituitary 2014, 17, 530–538. [Google Scholar] [CrossRef]
  79. McCormack, A.; Dekkers, O.M.; Petersenn, S.; Popovic, V.; Trouillas, J.; Raverot, G.; Burman, P. Treatment of aggressive pituitary tumours and carcinomas: Results of a European Society of Endocrinology (ESE) survey 2016. Eur. J. Endocrinol. 2018, 178, 265–276. [Google Scholar] [CrossRef]
  80. Halevy, C.; Whitelaw, B.C. How effective is temozolomide for treating pituitary tumours and when should it be used? Pituitary 2017, 20, 261–266. [Google Scholar] [CrossRef]
  81. Lamas, C.; Cámara, R.; Fajardo, C.; Remon-Ruiz, P.; Biagetti, B.; Guerrero-Pérez, F.; Araujo-Castro, M.; Mora, M.; Hanzu, F.; Iglesias, P.; et al. Efficacy and safety of temozolomide in the treatment of aggressive pituitary neuroendocrine tumours in Spain. Front. Endocrinol. 2023, 14, 1204206. [Google Scholar] [CrossRef]
  82. Lopes-Pinto, M.; Lacerda-Nobre, E.; Silva, A.L.; Marques, P. Therapeutical Usefulness of PD-1/PD-L1 Inhibitors in Aggressive or Metastatic Pituitary Tumours. Cancers 2024, 16, 3033. [Google Scholar] [CrossRef]
  83. Wibowo, A.S.; Singh, M.; Reeder, K.M.; Carter, J.J.; Kovach, A.R.; Meng, W.; Ratnam, M.; Zhang, F.; Dann, C.E. Structures of human folate receptors reveal biological trafficking states and diversity in folate and antifolate recognition. Proc. Natl. Acad. Sci. USA 2013, 110, 15180–15188. [Google Scholar] [CrossRef] [PubMed]
  84. Jones, S.K.; Sarkar, A.; Feldmann, D.P.; Hoffmann, P.; Merkel, O.M. Revisiting the value of competition assays in folate receptor-mediated drug delivery. Biomaterials 2017, 138, 35–45. [Google Scholar] [CrossRef]
  85. Matherly, L.H.; Hou, Z. Structure and function of the reduced folate carrier a paradigm of a major facilitator superfamily mammalian nutrient transporter. Vitam. Horm. 2008, 79, 145–184. [Google Scholar] [PubMed]
  86. Sosnik, A. Chapter 1—From the “magic bullet” to advanced nanomaterials for active targeting in diagnostics and therapeutics. In Biomedical Applications of Functionalized Nanomaterials; Sarmento, B., das Neves, J., Eds.; Elsevier: Amsterdam, The Netherlands, 2018; pp. 1–32. [Google Scholar]
  87. Evans, C.O.; Reddy, P.; Brat, D.J.; O’Neill, E.B.; Craige, B.; Stevens, V.L.; Oyesiku, N.M. Differential expression of folate receptor in pituitary adenomas. Cancer Res. 2003, 63, 4218–4224. [Google Scholar]
  88. Evans, C.O.; Yao, C.; Laborde, D.; Oyesiku, N.M. Folate receptor expression in pituitary adenomas cellular and molecular analysis. Vitam. Horm. 2008, 79, 235–266. [Google Scholar]
  89. Galt, J.R.; Halkar, R.K.; Evans, C.-O.; Osman, N.A.; LaBorde, D.; Fox, T.H.; Faraj, B.A.; Kumar, K.; Wang, H.; Oyesiku, N.M. In Vivo Assay of Folate Receptors in Nonfunctional Pituitary Adenomas with 99mTc-Folate SPECT/CT. J. Nucl. Med. 2010, 51, 1716–1723. [Google Scholar] [CrossRef]
  90. Liu, X.; Ma, S.; Dai, C.; Cai, F.; Yao, Y.; Yang, Y.; Feng, M.; Deng, K.; Li, G.; Ma, W.; et al. Antiproliferative, antiinvasive, and proapoptotic activity of folate receptor α-targeted liposomal doxorubicin in nonfunctional pituitary adenoma cells. Endocrinology 2013, 154, 1414–1423. [Google Scholar] [CrossRef] [PubMed]
  91. Hu, C.; Wang, X.; Li, Y.; Han, X.; Ren, B.; Yin, G. Targeting pituitary adenomas with folate-conjugated multiple drug decorated liposomal formulations for improved antiproliferative anticancer efficacy. J. Exp. Nanosci. 2022, 17, 14–33. [Google Scholar] [CrossRef]
  92. Evans, C.-O.; Halkar, R.K.; Galt, J.R.; Faraj, B.A.; Oyesiku, N.M. Molecular Targeting and Imaging of Non-Functional Pituitary Tumors: Preliminary Results. Jpn. J. Neurosurg. 2006, 15, 437–443. [Google Scholar]
  93. Lee, M.; Pellegata, N.S. Folate Receptor-Mediated Drug Targeting: A Possible Strategy for Nonfunctioning Pituitary Adenomas? Endocrinology 2013, 154, 1387–1389. [Google Scholar] [CrossRef]
  94. Cheng, T.; Wang, Y.; Lu, M.; Zhan, X.; Zhou, T.; Li, B.; Zhan, X. Quantitative Analysis of Proteome in Non-functional Pituitary Adenomas: Clinical Relevance and Potential Benefits for the Patients. Front. Endocrinol. 2019, 10, 854. [Google Scholar] [CrossRef] [PubMed]
  95. Chen, Y.; Gao, H.; Xie, W.; Guo, J.; Fang, Q.; Zhao, P.; Liu, C.; Zhu, H.; Wang, Z.; Wang, J.; et al. Genomic and transcriptomic analysis of pituitary adenomas reveals the impacts of copy number variations on gene expression and clinical prognosis among prolactin-secreting subtype. Aging 2020, 13, 1276–1293. [Google Scholar] [CrossRef] [PubMed]
  96. Mangili, F.; Esposito, E.; Treppiedi, D.; Catalano, R.; Marra, G.; Di Muro, G.; Barbieri, A.M.; Locatelli, M.; Lania, A.G.; Mangone, A.; et al. DRD2 Agonist Cabergoline Abolished the Escape Mechanism Induced by mTOR Inhibitor Everolimus in Tumoral Pituitary Cells. Front. Endocrinol. 2022, 13, 867822. [Google Scholar] [CrossRef] [PubMed]
  97. Zatelli, M.C.; Minoia, M.; Filieri, C.; Tagliati, F.; Buratto, M.; Ambrosio, M.R.; Lapparelli, M.; Scanarini, M.; degli Uberti, E.C. Effect of Everolimus on Cell Viability in Nonfunctioning Pituitary Adenomas. J. Clin. Endocrinol. Metab. 2010, 95, 968–976. [Google Scholar] [CrossRef]
  98. Cerovac, V.; Monteserin-Garcia, J.; Rubinfeld, H.; Buchfelder, M.; Losa, M.; Florio, T.; Paez-Pereda, M.; Stalla, G.K.; Theodoropoulou, M. The Somatostatin Analogue Octreotide Confers Sensitivity to Rapamycin Treatment on Pituitary Tumor Cells. Cancer Res. 2010, 70, 666–674. [Google Scholar] [CrossRef] [PubMed]
  99. Zhang, D.; Way, J.S.; Zhang, X.; Sergey, M.; Bergsneider, M.; Wang, M.B.; Yong, W.H.; Heaney, A.P. Effect of Everolimus in Treatment of Aggressive Prolactin-Secreting Pituitary Adenomas. J. Clin. Endocrinol. Metab. 2019, 104, 1929–1936. [Google Scholar] [CrossRef]
  100. Lin, A.L.; Geer, E.B.; Lala, N.; Page-Wilson, G.; Magge, R.; Young, R.J.; Tabar, V. The treatment of aggressive prolactinomas with everolimus. Pituitary 2023, 26, 474–481. [Google Scholar] [CrossRef]
  101. Jouanneau, E.; Wierinckx, A.; Ducray, F.; Favrel, V.; Borson-Chazot, F.; Honnorat, J.; Trouillas, J.; Raverot, G. New targeted therapies in pituitary carcinoma resistant to temozolomide. Pituitary 2012, 15, 37–43. [Google Scholar] [CrossRef]
  102. Alshaikh, O.M.; Asa, S.L.; Mete, O.; Ezzat, S. An Institutional Experience of Tumor Progression to Pituitary Carcinoma in a 15-Year Cohort of 1055 Consecutive Pituitary Neuroendocrine Tumors. Endocr. Pathol. 2019, 30, 118–127. [Google Scholar] [CrossRef]
  103. Nakano-Tateno, T.; Lau, K.J.; Wang, J.; McMahon, C.; Kawakami, Y.; Tateno, T.; Araki, T. Multimodal Non-Surgical Treatments of Aggressive Pituitary Tumors. Front. Endocrinol. 2021, 12, 118–127. [Google Scholar] [CrossRef]
  104. Turner, H.E.; Nagy, Z.; Gatter, K.C.; Esiri, M.M.; Harris, A.L.; Wass, J.A.H. Angiogenesis in Pituitary Adenomas and the Normal Pituitary Gland. J. Clin. Endocrinol. Metab. 2000, 85, 1159–1162. [Google Scholar] [CrossRef] [PubMed]
  105. Takano, S.; Akutsu, H.; Hara, T.; Yamamoto, T.; Matsumura, A. Correlations of Vascular Architecture and Angiogenesis with Pituitary Adenoma Histotype. Int. J. Endocrinol. 2014, 2014, 989574. [Google Scholar] [CrossRef] [PubMed]
  106. Dai, C.; Liang, S.; Sun, B.; Li, Y.; Kang, J. Anti-VEGF Therapy in Refractory Pituitary Adenomas and Pituitary Carcinomas: A Review. Front. Oncol. 2021, 11, 773905. [Google Scholar] [CrossRef] [PubMed]
  107. Lamb, L.S.; Sim, H.-W.; McCormack, A.I. Exploring the Role of Novel Medical Therapies for Aggressive Pituitary Tumors: A Review of the Literature—“Are We There Yet?”. Cancers 2020, 12, 308. [Google Scholar] [CrossRef]
Figure 1. The summary of current and emerging medicinal treatment for CNFT. Created in BioRender. Zhang, D. (2025) https://BioRender.com/6wpp54q (accessed on 12 June 2025).
Figure 1. The summary of current and emerging medicinal treatment for CNFT. Created in BioRender. Zhang, D. (2025) https://BioRender.com/6wpp54q (accessed on 12 June 2025).
Ijms 26 05898 g001
Table 1. Histological classification of clinically nonfunctioning pituitary tumors.
Table 1. Histological classification of clinically nonfunctioning pituitary tumors.
Tumor TypeSubtypeFrequency (% of All CNFTs)Lineage Specific Transcription FactorHormone Produced
Silent Gonadotroph Tumor 40–79%SF-1 (NR5A1), GATA2, ERaFSHb, LHb, a-subunit
Silent Corticotroph TumorSparsely Granulated Corticotroph Tumor3–6%Tpit (TBX19)ACTH
Crooke’s Cell AdenomaTpit (TBX19)ACTH
Silent Somatotroph TumorSparsely Granulated Somatotroph Tumor2–4%Pit-1 (POU1F1)GH (or PRL)
Mammosomatotroph TumorPit-1 (POU1F1), ERa GH and PRL (same cells)
Mixed GH-PRL TumorPit-1 (POU1F1), ERa GH and PRL (different cells)
Plurihormonal TumorPit-1 (POU1F1), ERa GH, PRL, TSHb
Silent Lactotroph TumorSparsely Granulated Lactotroph Tumor1%Pit-1 (POU1F1), ERa PRL
Mammosomatotroph TumorPit-1 (POU1F1), ERa GH and PRL (same cells)
Mixed GH-PRLTumorPit-1 (POU1F1), ERa GH and PRL (different cells)
Plurihormonal TumorPit-1 (POU1F1), ERa GH, PRL, TSHb
Acidophilic Stem Cell AdenomaPit-1 (POU1F1), ERa GH, PRL
Silent Thyrotroph Tumor 1–2%Pit-1 (POU1F1), GATA2TSHb
Null Cell Tumor 17%NoneNone
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mogar, N.; Zhang, D.; Heaney, A.P. A Bridge Too Far? Towards Medical Therapy for Clinically Nonfunctioning Pituitary Tumors. Int. J. Mol. Sci. 2025, 26, 5898. https://doi.org/10.3390/ijms26125898

AMA Style

Mogar N, Zhang D, Heaney AP. A Bridge Too Far? Towards Medical Therapy for Clinically Nonfunctioning Pituitary Tumors. International Journal of Molecular Sciences. 2025; 26(12):5898. https://doi.org/10.3390/ijms26125898

Chicago/Turabian Style

Mogar, Nikita, Dongyun Zhang, and Anthony P. Heaney. 2025. "A Bridge Too Far? Towards Medical Therapy for Clinically Nonfunctioning Pituitary Tumors" International Journal of Molecular Sciences 26, no. 12: 5898. https://doi.org/10.3390/ijms26125898

APA Style

Mogar, N., Zhang, D., & Heaney, A. P. (2025). A Bridge Too Far? Towards Medical Therapy for Clinically Nonfunctioning Pituitary Tumors. International Journal of Molecular Sciences, 26(12), 5898. https://doi.org/10.3390/ijms26125898

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop