Jump to content

MaryO

~Chief Cushie~
  • Posts

    8,082
  • Joined

  • Last visited

  • Days Won

    555

Posts posted by MaryO

  1. Introduction: Pasireotide (PAS) is a novel somatostatin receptor ligands (SRL), used in controlling hormonal hypersecretion in both acromegaly and Cushing’s Disease (CD). In previous studies and meta-analysis, first-generation SRLs were reported to be able to induce significant tumor shrinkage only in somatotroph adenomas. This systematic review and meta-analysis aim to summarize the effect of PAS on the shrinkage of the pituitary adenomas in patients with acromegaly or CD.

    Materials and methods: We searched the Medline database for original studies in patients with acromegaly or CD receiving PAS as monotherapy, that assessed the proportion of significant tumor shrinkage in their series. After data extraction and analysis, a random-effect model was used to estimate pooled effects. Quality assessment was performed with a modified Joanna Briggs’s Institute tool and the risk of publication bias was addressed through Egger’s regression and the three-parameter selection model.

    Results: The electronic search identified 179 and 122 articles respectively for acromegaly and CD. After study selection, six studies considering patients with acromegaly and three with CD fulfilled the eligibility criteria. Overall, 37.7% (95%CI: [18.7%; 61.5%]) of acromegalic patients and 41.2% (95%CI: [22.9%; 62.3%]) of CD patients achieved significant tumor shrinkage. We identified high heterogeneity, especially in acromegaly (I2 of 90% for acromegaly and 47% for CD), according to the low number of studies included.

    Discussion: PAS treatment is effective in reducing tumor size, especially in acromegalic patients. This result strengthens the role of PAS treatment in pituitary adenomas, particularly in those with an invasive behavior, with progressive growth and/or extrasellar extension, with a low likelihood of surgical gross-total removal, or with large postoperative residual tissue.

    Systematic Review Registration: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022328152, identifier CRD42022328152

     

    Introduction

    Pasireotide (PAS) is a novel somatostatin receptor ligand (SRL) with a high affinity for the somatostatin receptor (SSR) type 5 (1, 2). Somatotroph adenomas are usually responsive to first-generation SRLs (octreotide and lanreotide), as they are able to reduce growth hormone (GH) secretion through SSR type 2 (3). In the flow-chart of acromegaly treatment, PAS is suggested in case of resistance to first-generation SRLs, as SSR type 5 is also abundantly expressed in GH-secreting adenomas (3). A phase III study with PAS long-acting release (LAR) proved its efficacy in first-generation SRLs-resistant acromegalic patients after 6 months (4). In the extension study (Colao A et al.), 37% of patients achieved normalization of insulin-like growth factor 1 (IGF-1) and/or GH levels <1 µg/L, considering both those performing the extension treatment and those crossing over from the first-generation SRL-control group to the PAS LAR group. Nearly two-thirds of responses were achieved after at least 6 months of treatment. Up-titration of the dose from 40 to 60 mg monthly enriched the number of responders, suggesting that the PAS LAR effect may be both time- and dose-dependent (5). Concomitant improvement in signs and symptoms has also been confirmed in other series (69).

    SSR type 5 is the predominant isoform in human corticotroph adenomas, since it is not down-regulated by high cortisol levels, as SSR type 2 does. Therefore, PAS is the only SRL available in patients with Cushing’s Disease (CD) (2). In a phase III study, subcutaneous (s.c.) PAS proved to be effective in normalizing urinary free cortisol (respectively in 13% and 25% of patients taking 600 µg or 900 µg bis-in-die for 12 months) (10), achieving significant clinical improvement (11). In the same clinical setting, PAS LAR showed similar efficacy and safety profiles (12). These benefits could be maintained for up to 5 years in an extension study (13, 14). In a recent meta-analysis, PAS treatment provided disease control in 44% of 522 patients with CD (15). Patients harbouring USP-8 mutations demonstrated an increased SSR type 5 expression in the corticotroph adenoma, increasing the likelihood of a positive response to PAS therapy (16). The safety profile of PAS is similar to that of first-generation SRLs, except for a significant worsening in glucose homeostasis (17).

    Despite the normalization of hormonal excess, another goal of the medical treatment in GH-secreting pituitary adenomas is the reduction of the size of the adenoma (18). First-generation SRLs proved to be effective in achieving tumor shrinkage in acromegaly: Endocrine Society clinical practice guidelines suggested their role as primary therapy in poor surgical candidates and in those with extrasellar extension without chiasmal compression (18). Cozzi et al. reported in a large prospective cohort of acromegalic patients a significant Octreotide-induced tumor shrinkage in 82% of those receiving SRL as first-line treatment; most of them exhibited an early shrinkage with a progressive trend in reduction later on (19). A meta-analysis of 41 studies reported a significant tumor shrinkage in 50% of included patients (20). Data from the primary treatment with once-monthly lanreotide in surgical naïve patients demonstrated its efficacy in reducing tumor volume, achieving significant tumor shrinkage in 63% of them (21). Hypo-intensity on T2-weighted sequences at baseline magnetic resonance imaging (MRI) seems to predict tumor volume reduction during first-generation SRLs treatment (22). Regarding patients with CD, most patients presented a microadenoma, usually not aggressive or invasive: only in selected cases tumor shrinkage is an aim to achieve in patients with corticotropinoma.

    As available data are scarce (or limited to selected studies), and the issue of pituitary adenoma shrinkage is of primary importance in the management of tumors that cannot be addressed through surgery, the aim of this systematic review and meta-analysis is to summarize available data regarding the effect of PAS on tumor size.

    Materials and Methods

    We used the Population-Intervention-Comparison-Outcome (PICO) model to formulate the research questions for the systematic review (23), as summarised in Figure 1. The systematic review and meta-analysis were conducted and are reported according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis of Diagnostic Test Accuracy Studies (PRISMA-DTA) statement (24). We registered the protocol on the International Prospective Register of Systematic Reviews database (PROSPERO, https://www.crd.york.ac.uk/PROSPERO, number CRD42022328152).

    Figure 1
    www.frontiersin.org

    FIGURE 1 PICO (Population-Intervention-Comparison-Outcome) model design to our study.

    Search Strategy

    An extensive Medline search was performed for the research question by two of the authors (F.C. and A.M.) independently, discrepancies were resolved by discussion. The literature search was performed up to January 2022, no language restriction was applied. Research included the following keywords: 1) (“acromegalies” [All Fields] OR “acromegaly”[MeSH Terms] OR “acromegaly”[All Fields]) AND (“pasireotide”[Supplementary Concept] OR “pasireotide”[All Fields]); 2) (“pituitary ACTH hypersecretion”[MeSH Terms] OR (“pituitary”[All Fields] AND “ACTH”[All Fields] AND “hypersecretion”[All Fields]) OR “pituitary ACTH hypersecretion”[All Fields]) AND (“pasireotide”[Supplementary Concept] OR “pasireotide”[All Fields]).

    Inclusion and exclusion criteria were specified in advance and protocol-defined, in order to avoid methodological bias for post-hoc analysis. The searches were designed to select all types of studies (retrospective, observational, controlled, randomized, and non-randomized) conducted in patients with acromegaly or CD treated with PAS as monotherapy; the assessment of the proportion of significant tumor shrinkage was an inclusion criterion. Search terms were linked to Medical Subject Headings when possible. Keywords and free words were used simultaneously. Additional articles were identified with manual searches and included a thorough review of other meta-analyses, review articles, and relevant references. Consolidation of studies was performed with Mendeley Desktop 1.19.8.

    Study Selection

    We included all original research studies conducted in adult patients that underwent PAS treatment used as monotherapy (s.c. bis-in-die and intramuscular once/monthly), that provided sufficient information about tumor size reduction during treatment. In case of overlapping cohorts of patients (as clinical trials with core and extension phases), we included only the extension study, in order to select those patients with measurable tumor shrinkage after long-term treatment. Local reports regarding patients involved in multicenter trials were excluded from the analysis, as they had been already considered in the larger series. Reviewers were not blinded to the authors or journals when screening articles.

    Data Extraction and Quality Assessment

    Two authors (F.C. and A.M.) read the included papers and extracted independently relevant data, any disagreements were resolved by discussion. If data were not clear from the original manuscript, the authors of the primary study were contacted to clarify the doubts.

    Contents of data extraction in the selected paper included: name of the first author, year of publication, setting (referral centre, academic hospital, mono- or multi-centric collection), type of treatment, its dose schedule and duration, pituitary imaging method during follow- up, the endpoint type regarding adenoma size analysis (i.e. primary vs exploratory). When data were reported for each patient or for subgroups, a global percentage of significant tumor shrinkage was calculated considering all subjects involved in the study.

    To assess the risk of bias in the included studies, the critical appraisal tool from Joanna Briggs’s Institute (JBI) was adapted to evaluate those considered in our metanalysis (25). Among the items proposed, we selected the most appropriate to our setting: 1. Were the inclusion criteria clearly identified? 2. Were diagnostic criteria for acromegaly or CD well defined? 3. Were valid methods applied to evaluate tumor shrinkage? 4. Was the inclusion of participants consecutive and complete? 5. Was the reporting of baseline participants’ features (demographic and clinical) complete? 6. Was the report of the outcomes clear? 7. Was the report of demographics of the involved sites complete? 8. Was statistical analysis appropriate? For each aspect we assigned as possible choices of answer: yes, no or unclear.

    Data Synthesis and Analysis

    A qualitative synthesis was performed summarizing the study design and population characteristics (age, male to female ratio, macro- to micro-adenoma ratio, prior treatments).

    A random-effect model was used to estimate pooled effects. Forest plots for percentages of significant tumor size reduction were generated to visualize heterogeneity among the studies. In order to assess publication bias, despite the low number of articles considered, we performed funnel plot and asymmetry analysis adjusted for the low number of studies (an Egger’s regression test and a three-parameter selection model where two tailed p < 0.05 was considered statistically significant). The I2 test was conducted to analyze the heterogeneity between studies: an I2 >50% indicated a between-study heterogeneity.

    Statistical analyses were performed with R: R-4.1.2 for Windows 10 (32/64 bit) released 2021-11-01 and R studio desktop RStudio Desktop 1.4.1717 for Windows 10 64 bit (R Foundation for Statistical Computing, Vienna, Austria, URL https://www.R-project.org/).

    Results

    Study Selection

    The study selection process for acromegaly is depicted in Figure 2. The electronic search revealed 179 articles, with one duplicate (N = 178). After the first screening, 141 articles did not meet the eligibility criteria and were discarded. The full-text examination of the remaining studies excluded additional 31 articles: 27 did not provide adequate data about tumor size, two represented the core phase of an extension study, another one referred to a subset of patients from a larger study, and the last one did not provide sufficient data about the percentage of tumor size reduction. Thus, six studies fulfilling eligibility criteria (reported in Tables 1, 2), were selected for data extraction and analysis.

    Figure 2
    www.frontiersin.org

    FIGURE 2 Search strategy for acromegaly. * Petersenn S, 2010 (PAS sc, phase II) and Colao A, 2014 (PAS LAR). ** Shimon I, 2015 (PAS LAR). *** Tahara S, 2019 (PAS LAR, phase II). PAS, pasireotide, sc, subcutaneous, LAR, long-acting release.

    Table 1
    www.frontiersin.org

    TABLE 1 Studies considered for the metanalysis in acromegaly.

    Table 2
    www.frontiersin.org

    TABLE 2 Studies considered for the metanalysis in acromegaly.

    The study selection process for CD is depicted in Figure 3. The electronic search revealed 122 articles; an additional one had been included post-hoc, through reference analysis of selected articles (N = 123). After the first screening, 91 articles did not meet the eligibility criteria and were discarded. The full-text examination of the remaining studies excluded 29 more articles: 23 did not provide sufficient data on tumor shrinkage, two of them represented the core phase of extension studies, two referred to subsets of patients included in a larger study and two did not provide sufficient data regarding the percentage of tumor size reduction. Thus, three studies fulfilling eligibility criteria (reported in Tables 3, 4) were selected for data extraction and analysis.

    Figure 3
    www.frontiersin.org

    FIGURE 3 Search strategy for Cushing’s Disease. * Lacroix A, 2018 (PAS LAR, phase III) and Lacroix A, 2020 (PAS sc, phase III post-hoc analysis). ** Simeoli C, 2014 (PAS sc) and Colao A 2012 (PAS sc, phase III). *** Daniel E, 2018 (PAS sc and LAR) and Trementino L, 2016 (PAS sc). PAS, pasireotide, sc, subcutaneous, LAR, long acting release.

    Table 3
    www.frontiersin.org

    TABLE 3 Studies considered for the metanalysis in Cushing’s Disease.

    Table 4
    www.frontiersin.org

    TABLE 4 Studies considered for the metanalysis in Cushing’s Disease.

    Study Characteristics

    Four multi- and two mono-centric studies in patients with acromegaly were considered and analyzed, all presenting a prospective design. Tumor size analysis was not one of the primary endpoints in any of the considered studies; from an initial overall recruitment of 358 patients, only 265 were included for tumor size reduction analysis. Most patients had previously undergone different treatments (Table 1). All studies, except one, used PAS LAR, dose titration was allowed in all trials. Median follow-up ranged from 6 to 25 months; MRI was performed to evaluate tumor size reduction and the criteria for considering it significant was mainly based on tumor volume analysis, except for Lasolle H et al. which considered median height reduction (26). Data from the PAOLA study provided separate percentages of significant tumor shrinkage for PAS at 40 mg or 60 mg once monthly; considering that respectively 12 and 7 patients showed a reduction >25%, a significant shrinkage was reported in 19 out of 79 considered cases (24%) (4). Stelmachowska-Banás et al. described one patient with McCune-Albright’s syndrome presenting with pituitary hyperplasia, without a visible adenoma at MRI; as its pituitary volume decreased during treatment, the patient was included in the group with significant tumor shrinkage (27). No study provided information about macro- to micro-adenoma ratio. Data regarding age and male to female ratio are also reported in Table 2.

    Three studies including patients with CD met the eligibility criteria (Tables 3, 4); all of them presented a multicentre prospective design, recruiting 139 patients, most of them assuming PAS as a second-line treatment, after a surgical failure. For tumor shrinkage analysis, a subgroup of 34 patients was considered, taking s.c. PAS bis-in-die in two studies and PAS LAR in the third; in all cases titration was admitted. Tumor size analysis was a secondary endpoint in all three studies. Follow-up ranged from 6 to 60 months; tumor size assessment was performed with pituitary MRI. Only Pivonello et al. evaluated maximum diameter, instead of tumor volume changes (28). The population analyzed for tumor shrinkage mainly presented with a microadenoma. Data regarding age and gender are reported in Table 4. In the trial reported by Petersenn S et al., we arbitrarily fixed the criterion to define a significant tumor volume reduction (at least 25% of the baseline size of the pituitary adenoma), and the proportion of responders was calculated from the supplementary materials accordingly (3/6 = 50%) (13). Pivonello et al. separated patients exhibiting mild-moderate from those with severe hypercortisolism; we considered them together for tumor size analysis obtaining an overall proportion of significant size reduction of 21.4% (3 out of 14 subjects) (28).

    Risk of Bias

    The evaluation of the risk of bias performed with the adapted JBI tool is reported in Table 5. All studies presented clear diagnostic and inclusion criteria, except that of Lasolle H et al. (26). Although all papers reported a valid tool for tumor shrinkage analysis (MRI), two of them did not analyse tumor volume and did not provide a clear definition of significant size reduction (26, 28). Regarding other items, the majority of the considered studies did not appear to present a clear source of bias.

    Table 5
    www.frontiersin.org

    TABLE 5 Evaluation of the risk of bias performed with the adapted Joanna Briggs’s Institute (JBI) tool.

    Meta-Analysis

    In the six studies considered for acromegaly, 37.7% (95%CI: [18.7%; 61.5%]) of patients demonstrated a significant tumor size reduction (Figure 4). As expected, heterogeneity in tumor reduction between studies was high (I2 = 90%). We attempted to address publication bias despite the low-number of studies (Figure 6A😞 Egger’s regression test did not indicate the presence of funnel plot asymmetry (intercept = -3.15 with 95%CI: [-10.17; 3.85], t = -0.883, p = 0.427) and the three-parameter selection model performed for p < 0.05 (and p < 0.1 as a sensitivity analysis) suggested absence of publication bias (28).

    Figure 4
    www.frontiersin.org

    FIGURE 4 Pooled effect for the proportion of responders (i.e. presenting significant tumor shrinkage) in acromegaly. CI, confidence interval.

    In the three studies considered for CD, 41,2% (95%CI: [22.9%; 62.3%]) of patients overall demonstrated a significant tumor size reduction (Figure 5). The heterogeneity in tumor reduction between the studies represented by I2 amounted to 47%. Publication bias analysis (Figure 6B) was performed using Egger’s regression test (intercept = -1.828 with 95%CI: [-14.53; 10.88], t = -0.282, p = 0.825) without evidence of asymmetry. The three-parameter selection model on the contrary could not be performed due to the small number of studies.

    Figure 5
    www.frontiersin.org

    FIGURE 5 Pooled effect for the proportion of responders (i.e. presenting significant tumor shrinkage) in Cushing’s Disease. CI, confidence interval.

    Figure 6
    www.frontiersin.org

    FIGURE 6 (A) Funnel plot assessing publication bias for Acromegaly. (B) Funnel plot assessing publication bias for Cushing’s Disease.

    Discussion

    The biochemical efficacy of medical treatment with PAS in GH- or ACTH-secreting pituitary adenomas has been described in previous metanalyses for acromegaly (29, 30) and CD (15), the latter also exploring the clinical benefit. In addition to these reports, this meta-analysis shows that PAS treatment can induce an additional clinically significant tumor shrinkage in approximately 40% of patients.

    Acromegaly

    Overall, PAS treatment provided tumor shrinkage in 37.7% of the considered patients. A previous metanalysis on octreotide in acromegaly provided a higher percentage of tumor size reduction (over 50%) (20). Nevertheless, since PAS treatment is usually considered as a second- or third-line treatment in the therapeutic flow-chart of acromegaly, the population recruited is mainly composed of patients with first-generation SRL-resistant somatotroph adenomas. This bias in recruited populations of acromegalic patients may explain this difference in the outcome. In a direct comparison, although PAS LAR seemed more effective in achieving biochemical control, both the SRLs, the first- and the second-generation types, achieved similar percentages of tumor shrinkage (6, 7). Moreover, in the crossover extension, the switch from octreotide to PAS was more effective than the reverse schedule, achieving a slightly higher percentage of further significant tumor shrinkage (8). Lasolle et al. reported that the expression of SSR type 5 and the granulation pattern are of limited value for the prediction of PAS responsiveness: 5 out of 9 somatotropinomas in their series were densely granulated (two did not respond to PAS), and the expression of SSR type 5 was modest in one controlled patient (26).

    Other than SRLs, a further therapeutic option targeting the somatotroph adenoma is cabergoline, either as monotherapy in mild cases or as an add-on treatment for resistant adenomas (18). In a previous metanalysis, cabergoline in monotherapy resulted less effective than SRLs, achieving tumor shrinkage in about one third of the enrolled patients (31). It should also be mentioned that some studies reported an escape phenomenon from its treatment efficacy (32).

    Data coming from the combination of PAS LAR and pegvisomant in acromegaly were not considered in our metanalysis, due to inclusion criteria and variable combination therapy of the two drugs (33). Since some cases of adenoma growth had been reported during pegvisomant use (34, 35), this combination therapy represents a rational approach, but tumor volume analysis is less reliable, given the purpose of our study. Despite concerns regarding tumor growth, pegvisomant effectiveness in acromegaly is well documented (18, 29), although the cost of this combination treatment can limit its applicability in real-life practice. Moreover, it is worth mentioning Coopmans and collaborators’ follow-up analysis, suggesting a PAS mediated anti-tumoral effect in acromegaly. During treatment, patients exhibited a significant increase in T2-weighted sequences signal at MRI; moreover, patients exhibiting this MRI characteristic in their adenomas showed a more evident decrease in IGF-1 levels, but not a similar pattern in reduction of pituitary adenoma size (36). This finding may be related to cell degeneration or tumor cell necrosis, without necessarily determining significant tumor size reduction. Further studies, probably with more data coming from histological reports, may be necessary to better understand these findings.

    Cushing’s Disease

    Overall, PAS treatment provided significant tumor shrinkage in 41.2% of CD patients. Regarding pituitary-directed drugs, at this moment available for CD treatment, the efficacy of cabergoline has been proven in vitro studies, but its efficacy in clinical trials is still debated (15, 37). In a previous prospective study, cabergoline induced significant tumor shrinkage (defined as tumor volume reduction >20%) in 4 out of 20 (20%) of the patients recruited after 24 months (38). PAS is the only pituitary-directed treatment for this condition approved by Drug Agencies. Although few studies have been considered in this metanalysis, due to the strict inclusion criteria, PAS appears more effective in tumor size reduction versus cabergoline, resulting in a better choice in CD therapy when aiming to control the pituitary adenoma.

    In contrast to acromegaly, the majority of CD patients present a microadenoma, suggesting that tumor size might be a less relevant issue during medical treatment, even if the “cure” of the disease may forecast the resolution of the adenoma. Besides, up to 30% of CD patients, depending also on MRI accuracy and neuro-radiologist’s expertise, may present with negative imaging that prevents any evaluation of tumor shrinkage (39). In spite of that, endocrinologists, not so infrequently, deal with aggressive corticotroph adenomas, characterized by invasive local growth and/or resistance to conventional therapies. This challenging entity often requires multidisciplinary expertise to suggest different approaches, including PAS treatment (40). It should be mentioned that some non-pituitary targeting drugs, as inhibitors of cortisol synthesis, have been associated with tumor growth, due to cortisol-ACTH negative feedback. In particular, during osilodrostat treatment in a phase III study, four recruited patients discontinued osilodrostat after a significant increase in tumor volume (two with micro- and two with macro-adenomas 41), and this growth had also been described during ketoconazole and mitotane treatments (42). Thus, it may be speculated that PAS could provide a rational approach as an combination treatment with steroidogenesis inhibitors. Moreover, after bilateral adrenalectomy, pituitary adenoma tumor size is of the utmost importance, as patients may be at risk of developing a progression of the adenoma, the so-called Nelson’s syndrome. In a prospective study from Daniel E et al., PAS proved to be also effective in this setting, reducing ACTH levels and stabilizing the residual tumor over a treatment period of 7 months (43). Further studies, with longer treatment observation, may reveal whether PAS may achieve significant tumor shrinkage in these patients, as suggested by previous case reports in literature (44, 45).

    Conclusion

    The main limitation of our study resides in the scarce literature provided up to now (260 patients with acromegaly and 34 with CD), in the different therapy schedules and different criteria for tumor shrinkage in the selected study (largest tumor diameter vs a selected percentage of reduction). Moreover, in none of the study tumor reduction was one of the primary endpoints, and surgery was performed before PAS in most patients (78-88% of CD and 43-96% of acromegaly).

    PAS is a novel compound, with a rising role in the treatment of secreting pituitary adenomas. Thus, this topic might be amplified with more data coming from further clinical studies, as real-life studies, possibly also addressing markers predictive of response to this treatment (e.g., expression of SSR type 2 and type 5 or somatic mutations in USP8 at tissue level of ACTH-secreting adenomas). Nevertheless, we can already state that PAS treatment is effective in reducing tumor size, especially in acromegaly. Our results strengthen the role of PAS treatment in somatotroph and corticotroph adenomas, especially when tumor volume is a relevant issue (i.e. tumor progression, extrasellar invasion) (18, 39), as a neoadjuvant treatment before surgery or as tailored treatment, alone or in combination, in persistent disease or when surgery is not feasible. Future research aiming to characterize markers predictive of response could help to identify optimal candidates for this treatment.

    Data Availability Statement

    The original contributions presented in the study are included in the article. Further inquiries can be directed to the corresponding author.

    Ethics Statement

    Informed consent was obtained from all subjects participating in the studies analyzed.

    Author Contributions

    Authors involved contributed to research as reported: literature search (FC, AM), preparation of original draft (FC, AM, MB, LD), literature review (CS, FC, AM, MF), manuscript editing (CS, FC, AM, MB, LD, MF) and supervision (RM, CS). All authors approved the final version of the paper.

    Conflict of Interest

    The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

    Publisher’s Note

    All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

    References

    1. Chinezu L, Vasiljevic A, Jouanneau E, François P, Borda A, Trouillas J, et al. Expression of Somatostatin Receptors, SSTR2A and SSTR5, in 108 Endocrine Pituitary Tumors Using Immunohistochemical Detection With New Specific Monoclonal Antibodies. Hum Pathol (2014) 45(1):71–7. doi: 10.1016/j.humpath.2013.08.007

    PubMed Abstract | CrossRef Full Text | Google Scholar

    2. Ceccato F, Scaroni C, Boscaro M. Clinical Use of Pasireotide for Cushing's Disease in Adults. Ther Clin Risk Manage (2015) 11:425–34. doi: 10.2147/TCRM.S37314

    CrossRef Full Text | Google Scholar

    3. Melmed S, Colao A, Barkan A, Molitch M, Grossman AB, Kleinberg D, et al. Guidelines for Acromegaly Management: An Update. J Clin Endocrinol Metab (2009) 94(5):1509–17. doi: 10.1210/jc.2008-2421

    PubMed Abstract | CrossRef Full Text | Google Scholar

    4. Gadelha MR, Bronstein MD, Brue T, Coculescu M, Fleseriu M, Guitelman M, et al. Pasireotide Versus Continued Treatment With Octreotide or Lanreotide in Patients With Inadequately Controlled Acromegaly (PAOLA): A Randomised, Phase 3 Trial. Lancet Diabetes Endocrinol (2014) 2(11):875–84. doi: 10.1016/S2213-8587(14)70169-X

    PubMed Abstract | CrossRef Full Text | Google Scholar

    5. Colao A, Bronstein MD, Brue T, De Marinis L, Fleseriu M, Guitelman M, et al. Pasireotide for Acromegaly: Long-Term Outcomes From an Extension to the Phase III PAOLA Study. Eur J Endocrinol (2020) 182(6):583. doi: 10.1530/EJE-19-0762

    PubMed Abstract | CrossRef Full Text | Google Scholar

    6. Colao A, Bronstein MD, Freda P, Gu F, Shen CC, Gadelha M, et al. Pasireotide Versus Octreotide in Acromegaly: A Head-to-Head Superiority Study. J Clin Endocrinol Metab (2014) 99(3):791–9. doi: 10.1210/jc.2013-2480

    PubMed Abstract | CrossRef Full Text | Google Scholar

    7. Sheppard M, Bronstein MD, Freda P, Serri O, De Marinis L, Naves L, et al. Pasireotide LAR Maintains Inhibition of GH and IGF-1 in Patients With Acromegaly for Up to 25 Months: Results From the Blinded Extension Phase of a Randomized, Double-Blind, Multicenter, Phase III Study. [Published Correction Appears in Pituitary]. (2015) 18(3):385–94. doi: 10.1007/s11102-014-0585-6

    CrossRef Full Text | Google Scholar

    8. Bronstein MD, Fleseriu M, Neggers S, Colao A, Sheppard M, Gu F, et al. Switching Patients With Acromegaly From Octreotide to Pasireotide Improves Biochemical Control: Crossover Extension to a Randomized, Double-Blind, Phase III Study. BMC Endocr Disord (2016) 16:16. doi: 10.1186/s12902-016-0096-8

    PubMed Abstract | CrossRef Full Text | Google Scholar

    9. Gadelha M, Bex M, Colao A, Pedroza García EM, Poiana C, Jimenez-Sanchez M, et al. Evaluation of the Efficacy and Safety of Switching to Pasireotide in Patients With Acromegaly Inadequately Controlled With First-Generation Somatostatin Analogs. Front Endocrinol (Lausanne). (2020) 10:931. doi: 10.3389/fendo.2019.00931

    PubMed Abstract | CrossRef Full Text | Google Scholar

    10. Colao A, Petersenn S, Newell-Price J, Findling JW, Gu F, Maldonado M, et al. A 12-Month Phase 3 Study of Pasireotide in Cushing's Disease. N Engl J Med (2012) 366(10):914–24. doi: 10.1056/NEJMoa1105743

    PubMed Abstract | CrossRef Full Text | Google Scholar

    11. Pivonello R, Petersenn S, Newell-Price J, Findling JW, Gu F, Maldonado M, et al. Pasireotide Treatment Significantly Improves Clinical Signs and Symptoms in Patients With Cushing's Disease: Results From a Phase III Study. Clin Endocrinol (Oxf). (2014) 81(3):408–17. doi: 10.1111/cen.12431

    PubMed Abstract | CrossRef Full Text | Google Scholar

    12. Lacroix A, Gu F, Gallardo W, Pivonello R, Yu Y, Witek P, et al. Efficacy and Safety of Once-Monthly Pasireotide in Cushing's Disease: A 12 Month Clinical Trial. Lancet Diabetes Endocrinol (2018) 6(1):17–26. doi: 10.1016/S2213-8587(17)30326-1

    PubMed Abstract | CrossRef Full Text | Google Scholar

    13. Petersenn S, Salgado LR, Schopohl J, Portocarrero-Ortiz L, Arnaldi G, Lacroix A, et al. Long-Term Treatment of Cushing's Disease With Pasireotide: 5-Year Results From an Open-Label Extension Study of a Phase III Trial. Endocrine. (2017) 57(1):156–65. doi: 10.1007/s12020-017-1316-3

    PubMed Abstract | CrossRef Full Text | Google Scholar

    14. Fleseriu M, Petersenn S, Biller BMK, Kadioglu P, De Block C, T'Sjoen G, et al. Long-Term Efficacy and Safety of Once-Monthly Pasireotide in Cushing's Disease: A Phase III Extension Study. Clin Endocrinol (Oxf). (2019) 91(6):776–85. doi: 10.1111/cen.14081

    PubMed Abstract | CrossRef Full Text | Google Scholar

    15. Simões Corrêa Galendi J, Correa Neto ANS, Demetres M, Boguszewski CL, Nogueira VDSN. Effectiveness of Medical Treatment of Cushing's Disease: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne). (2021) 12:732240. doi: 10.3389/fendo.2021.732240

    PubMed Abstract | CrossRef Full Text | Google Scholar

    16. Hayashi K, Inoshita N, Kawaguchi K, Ibrahim Ardisasmita A, Suzuki H, Fukuhara N, et al. The USP8 Mutational Status may Predict Drug Susceptibility in Corticotroph Adenomas of Cushing's Disease. Eur J Endocrinol (2016) 174(2):213–26. doi: 10.1530/EJE-15-0689

    PubMed Abstract | CrossRef Full Text | Google Scholar

    17. Samson SL, Gu F, Feldt-Rasmussen U, Zhang S, Yu Y, Witek P, et al. Managing Pasireotide-Associated Hyperglycemia: A Randomized, Open-Label, Phase IV Study. Pituitary. (2021) 24(6):887–903. doi: 10.1007/s11102-021-01161-4

    PubMed Abstract | CrossRef Full Text | Google Scholar

    18. Katznelson L, Laws ER Jr, Melmed S, Molitch ME, Murad MH, Utz A, et al. Acromegaly: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab (2014) 99(11):3933–51. doi: 10.1210/jc.2014-2700

    PubMed Abstract | CrossRef Full Text | Google Scholar

    19. Cozzi R, Montini M, Attanasio R, Albizzi M, Lasio G, Lodrini S, et al. Primary Treatment of Acromegaly With Octreotide LAR: A Long-Term (Up to Nine Years) Prospective Study of its Efficacy in the Control of Disease Activity and Tumor Shrinkage. J Clin Endocrinol Metab (2006) 91(4):1397–403. doi: 10.1210/jc.2005-2347

    PubMed Abstract | CrossRef Full Text | Google Scholar

    20. Giustina A, Mazziotti G, Torri V, Spinello M, Floriani I, Melmed S. Meta-Analysis on the Effects of Octreotide on Tumor Mass in Acromegaly. PloS One (2012) 7(5):e36411. doi: 10.1371/journal.pone.0036411

    PubMed Abstract | CrossRef Full Text | Google Scholar

    21. Caron PJ, Bevan JS, Petersenn S, Flanagan D, Tabarin A, Prévost G, et al. Tumor Shrinkage With Lanreotide Autogel 120 Mg as Primary Therapy in Acromegaly: Results of a Prospective Multicenter Clinical Trial. J Clin Endocrinol Metab (2014) 99(4):1282–90. doi: 10.1210/jc.2013-3318

    PubMed Abstract | CrossRef Full Text | Google Scholar

    22. Potorac I, Petrossians P, Daly AF, Alexopoulou O, Borot S, Sahnoun-Fathallah M, et al. T2-Weighted MRI Signal Predicts Hormone and Tumor Responses to Somatostatin Analogs in Acromegaly. Endocr Relat Cancer. (2016) 23(11):871–81. doi: 10.1530/ERC-16-0356

    PubMed Abstract | CrossRef Full Text | Google Scholar

    23. Schardt C, Adams MB, Owens T, Keitz S, Fontelo P. Utilization of the PICO Framework to Improve Searching PubMed for Clinical Questions. BMC Med Inform Decis Mak. (2007) 7:16. doi: 10.1186/1472-6947-7-16

    PubMed Abstract | CrossRef Full Text | Google Scholar

    24. McInnes MDF, Moher D, Thombs BD, McGrath TA, Bossuyt PM. Preferred Reporting Items for a Systematic Review and Meta-Analysis of Diagnostic Test Accuracy Studies: The PRISMA-DTA Statement [Published Correction Appears in JAMA. 2019 Nov 26;322(20):2026]. JAMA. (2018) 319(4):388–96. doi: 10.1001/jama.2017.19163

    CrossRef Full Text | Google Scholar

    25. Munn Z, Barker TH, Moola S, Tufanaru C, Stern C, McArthur A, et al. Methodological Quality of Case Series Studies: An Introduction to the JBI Critical Appraisal Tool. JBI Evid Synth. (2020) 18(10):2127–33. doi: 10.11124/JBISRIR-D-19-00099

    PubMed Abstract | CrossRef Full Text | Google Scholar

    26. Lasolle H, Ferriere A, Vasiljevic A, Eimer S, Nunes ML, Tabarin A. Pasireotide-LAR in Acromegaly Patients Treated With a Combination Therapy: A Real-Life Study. Endocr Connect. (2019) 8(10):1383–94. doi: 10.1530/EC-19-0332

    PubMed Abstract | CrossRef Full Text | Google Scholar

    27. Stelmachowska-Banaś M, Czajka-Oraniec I, Tomasik A, Zgliczyński W. Real-World Experience With Pasireotide-LAR in Resistant Acromegaly: A Single Center 1-Year Observation. Pituitary. (2022) 25(1):180–90. doi: 10.1007/s11102-021-01185-w

    PubMed Abstract | CrossRef Full Text | Google Scholar

    28. Pivonello R, Arnaldi G, Scaroni C, Giordano C, Cannavò S, Iacuaniello D, et al. The Medical Treatment With Pasireotide in Cushing's Disease: An Italian Multicentre Experience Based on "Real-World Evidence". Endocrine. (2019) 64(3):657–72. doi: 10.1007/s12020-018-1818-7

    PubMed Abstract | CrossRef Full Text | Google Scholar

    29. Leonart LP, Ferreira VL, Tonin FS, Fernandez-Llimos F, Pontarolo R. Medical Treatments for Acromegaly: A Systematic Review and Network Meta-Analysis. Value Health (2018) 21(7):874–80. doi: 10.1016/j.jval.2017.12.014”

    PubMed Abstract | CrossRef Full Text | Google Scholar

    30. Qiao N, He M, Shen M, Zhang Q, Zhang Z, Shou X, et al. Comparative Efficacy Of Medical Treatment For Acromegaly: A Systematic Review And Network Meta-Analysis Of Integrated Randomized Trials And Observational Studies. Endocr Pract (2020) 26(4):454–62. doi: 10.4158/EP-2019-0528

    PubMed Abstract | CrossRef Full Text | Google Scholar

    31. Sandret L, Maison P, Chanson P. Place of Cabergoline in Acromegaly: A Meta-Analysis. J Clin Endocrinol Metab (2011) 96(5):1327–35. doi: 10.1210/jc.2010-2443

    PubMed Abstract | CrossRef Full Text | Google Scholar

    32. Giraldi EA, Ioachimescu AG. The Role of Dopamine Agonists in Pituitary Adenomas. Endocrinol Metab Clin North Am (2020) 49(3):453–74. doi: 10.1016/j.ecl.2020.05.006

    PubMed Abstract | CrossRef Full Text | Google Scholar

    33. Muhammad A, van der Lely AJ, Delhanty PJD, Dallenga AHG, Haitsma IK, Janssen JAMJL, et al. Efficacy and Safety of Switching to Pasireotide in Patients With Acromegaly Controlled With Pegvisomant and First-Generation Somatostatin Analogues (PAPE Study). J Clin Endocrinol Metab (2018) 103(2):586–95. doi: 10.1210/jc.2017-02017

    PubMed Abstract | CrossRef Full Text | Google Scholar

    34. uhk JH, Jung S, Psychogios MN, Göricke S, Hartz S, Schulz-Heise S, et al. Tumor Volume of Growth Hormone-Secreting Pituitary Adenomas During Treatment With Pegvisomant: A Prospective Multicenter Study. J Clin Endocrinol Metab (2010) 95(2):552–8. doi: 10.1210/jc.2009-1239

    PubMed Abstract | CrossRef Full Text | Google Scholar

    35. Marazuela M, Paniagua AE, Gahete MD, Lucas T, Alvarez-Escolá C, Manzanares R, et al. Somatotroph Tumor Progression During Pegvisomant Therapy: A Clinical and Molecular Study. J Clin Endocrinol Metab (2011) 96(2):E251–9. doi: 10.1210/jc.2010-1742

    PubMed Abstract | CrossRef Full Text | Google Scholar

    36. Coopmans EC, van der Lely AJ, Schneiders JJ, Neggers SJCMM. Potential Antitumour Activity of Pasireotide on Pituitary Tumours in Acromegaly. Lancet Diabetes Endocrinol (2019) 7(6):425–6. doi: 10.1016/S2213-8587(19)30113-5

    PubMed Abstract | CrossRef Full Text | Google Scholar

    37. Ferriere A, Cortet C, Chanson P, Delemer B, Caron P, Chabre O, et al. Cabergoline for Cushing's Disease: A Large Retrospective Multicenter Study. Eur J Endocrinol (2017) 176(3):305–14. doi: 10.1530/EJE-16-0662

    PubMed Abstract | CrossRef Full Text | Google Scholar

    38. Pivonello R, De Martino MC, Cappabianca P, De Leo M, Faggiano A, Lombardi G, et al. The Medical Treatment of Cushing's Disease: Effectiveness of Chronic Treatment With the Dopamine Agonist Cabergoline in Patients Unsuccessfully Treated by Surgery. J Clin Endocrinol Metab (2009) 94(1):223–30. doi: 10.1210/jc.2008-1533

    PubMed Abstract | CrossRef Full Text | Google Scholar

    39. Fleseriu M, Auchus R, Bancos I, Ben-Shlomo A, Bertherat J, Biermasz NR, et al. Consensus on Diagnosis and Management of Cushing's Disease: A Guideline Update. Lancet Diabetes Endocrinol (2021) 9(12):847–75. doi: 10.1016/S2213-8587(21)00235-7

    PubMed Abstract | CrossRef Full Text | Google Scholar

    40. Ceccato F, Lombardi G, Manara R, Emanuelli E, Denaro L, Milanese L, et al. Temozolomide and Pasireotide Treatment for Aggressive Pituitary Adenoma: Expertise at a Tertiary Care Center. J Neurooncol. (2015) 122(1):189–96. doi: 10.1007/s11060-014-1702-0

    PubMed Abstract | CrossRef Full Text | Google Scholar

    41. Pivonello R, Fleseriu M, Newell-Price J, Bertagna X, Findling J, Shimatsu A, et al. Efficacy and Safety of Osilodrostat in Patients With Cushing's Disease (LINC 3): A Multicentre Phase III Study With a Double-Blind, Randomised Withdrawal Phase. Lancet Diabetes Endocrinol (2020) 8(9):748–61. doi: 10.1016/S2213-8587(20)30240-0

    PubMed Abstract | CrossRef Full Text | Google Scholar

    42. Varlamov EV, Han AJ, Fleseriu M. Updates in Adrenal Steroidogenesis Inhibitors for Cushing's Syndrome - A Practical Guide. Best Pract Res Clin Endocrinol Metab (2021) 35(1):101490. doi: 10.1016/j.beem.2021.101490

    PubMed Abstract | CrossRef Full Text | Google Scholar

    43. Daniel E, Debono M, Caunt S, Girio-Fragkoulakis C, Walters SJ, Akker SA, et al. A Prospective Longitudinal Study of Pasireotide in Nelson's Syndrome. Pituitary. (2018) 21(3):247–55. doi: 10.1007/s11102-017-0853-3

    PubMed Abstract | CrossRef Full Text | Google Scholar

    44. Katznelson L. Sustained Improvements in Plasma ACTH and Clinical Status in a Patient With Nelson's Syndrome Treated With Pasireotide LAR, a Multireceptor Somatostatin Analog. J Clin Endocrinol Metab (2013) 98(5):1803–7. doi: 10.1210/jc.2013-1497

    PubMed Abstract | CrossRef Full Text | Google Scholar

    45. He X, Spencer-Segal JL. Rapid Response of Nelson's Syndrome to Pasireotide in Radiotherapy-Naive Patient. Clin Diabetes Endocrinol (2020) 6(1):22. doi: 10.1186/s40842-020-00110-7

    PubMed Abstract | CrossRef Full Text | Google Scholar

     

    Keywords: pasireotide, cushing, acromegaly, tumor volume, tumor size

    Citation: Mondin A, Manara R, Voltan G, Tizianel I, Denaro L, Ferrari M, Barbot M, Scaroni C and Ceccato F (2022) Pasireotide-Induced Shrinkage in GH and ACTH Secreting Pituitary Adenoma: A Systematic Review and Meta-Analysis. Front. Endocrinol. 13:935759. doi: 10.3389/fendo.2022.935759

    Received: 04 May 2022; Accepted: 06 June 2022;
    Published: 01 July 2022.

    Edited by:

    Mohammad E. Khamseh, Iran University of Medical Sciences, Iran

    Reviewed by:

    Rosa Paragliola, Catholic University of the Sacred Heart, Rome, Italy
    Marek Bolanowski, Wroclaw Medical University, Poland
    Adriana G Ioachimescu, Emory University, United States

    Copyright © 2022 Mondin, Manara, Voltan, Tizianel, Denaro, Ferrari, Barbot, Scaroni and Ceccato. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

    *Correspondence: Filippo Ceccato, filippo.ceccato@unipd.it

    ORCID: Alessandro Mondin, orcid.org/0000-0002-6046-5198
    Renzo Manara, orcid.org/0000-0002-5130-3971
    Giacomo Voltan, orcid.org/0000-0002-3628-0492
    Irene Tizianel, orcid.org/0000-0003-4092-5107
    Luca Denaro, orcid.org/0000-0002-2529-6149
    Marco Ferrari, orcid.org/0000-0002-4023-0121
    Mattia Barbot, orcid.org/0000-0002-1081-5727
    Carla Scaroni, orcid.org/0000-0001-9396-3815
    Filippo Ceccato, orcid.org/0000-0003-1456-8716

     

    Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.

    From https://www.frontiersin.org/articles/10.3389/fendo.2022.935759/full

    • Like 1
  2. Abstract

    Although human cultures stimulated with dexamethasone suggest that the glucocorticoid receptor (GR) activates stress erythropoiesis, the effects of GR activation on erythropoiesis in vivo remains poorly understood.

    We characterized the phenotype of a large cohort of patients with Cushing’s Disease, a rare condition associated with elevated cortisol levels. Results from hypercortisolemic patients with active Cushing’s were compared with those obtained from eucortisolemic patients after remission and from non-diseased volunteers. Active Cushing’s patients exhibit erythrocytosis associated with normal hemoglobin F levels. In addition, their blood contained elevated numbers of the GR-induced CD163+ monocytes and a unique class of CD34+ cells expressing CD110, CD36, CD133 and the GR-target gene CXCR4.

    When cultured, these CD34+ cells generated similarly large numbers of immature erythroid cells in the presence and absence of dexamethasone, with raised expression of the GR-target gene GILZ. Of interest, blood from Cushing’s patients in remission maintained high numbers of CD163+ monocytes and, although their CD34+ cells had a normal phenotype, these cells were unresponsive to added dexamethasone.

    Collectively, these results indicate that chronic exposure to excess glucocorticoids in vivo leads to erythrocytosis by generating erythroid progenitor cells with a constitutively active GR.

    Although remission rescues the erythrocytosis and the phenotype of the circulating CD34+ cells, a memory of other prior changes is maintained in remission.

    From https://haematologica.org/article/view/haematol.2021.280542

    • Like 1
  3. First of its kind CME webinar on #CushingsDisease for #endocrinologists and other clinicians treating patients with #Cushings

     

    Aug. 2, 2022 / PRZen / HAZLET, N.J. — In this CME Webinar, #endocrinology experts in the management of #CushingsDisease will describe best practices for the diagnosis and treatment to improve long-term outcomes for patients..

     

    For more information

    SPEAKERS:
    Maria Fleseriu, MD
    Professor of Medicine and Neurological Surgery
    Oregon Health & Science University

    Irina Bancos, MD
    Associate Professor of Medicine
    Mayo Clinic

    Voxmedia LLC gratefully acknowledges the educational donation provided by Recordati Rare Diseases, Inc.

    This educational activity is intended for #endocrinologists and other clinicians who manage patients with cushing’s disease.

    Voxmedia LLC is accredited by the Accreditation Council for Continuing Medical Education (ACCME) to provide continuing medical education for physicians.

    Voxmedia LLC designates this webinar activity for a maximum of 1.00 AMA PRA Category Credit(s)™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.

    Nurse practitioners may participate in this educational activity and earn a certificate of completion as AANP accepts AMA PRA Category 1 Credits™ through its reciprocity agreements.

    The National Commission on Certification of Physician Assistants accepts AMA PRA Category 1 Credits™ from organizations accredited by the ACCME.

    For additional CME activities and online cme courses visit CMEPlanet. #endocrinologist #EndocrinePractice #Cushings #Cushing #ThinkCushings #CushingsAwarenessDay #pituitary #TheEndoSociety #ENDO2022

    Follow the full story here: https://przen.com/pr/33469903

     



    Read more: https://www.digitaljournal.com/pr/cme-webinar-best-practices-for-the-management-of-individuals-with-cushings-disease#ixzz7bN6UUAQa

    • Like 1
  4. Journal of the Endocrine Society, Volume 6, Issue 7, July 2022, bvac080, https://doi.org/10.1210/jendso/bvac080

    Abstract

    Context

    Subclinical pituitary hemorrhage, necrosis, and/or cystic degeneration (SPH) presents mainly in large tumors and prolactinomas. The characteristics of patients with Cushing disease (CD) and SPH are not known.

    Objective

    To determine if SPH affects the presentation and biochemical profile of young patients with CD.

    Methods

    Pediatric and adolescent patients who were diagnosed with CD between 2005 and 2021 and available magnetic resonance imaging images were evaluated for SPH. The clinical and biochemical characteristics of patients with and without SPH were compared.

    Results

    Evidence of possible SPH was present in 12 out of 170 imaging studies (7.1%). Patients with and without SPH had similar age at diagnosis and sex distribution but differed in disease duration (median duration: 1.0 year [1.0-2.0] in the SPH group vs 2.5 years [1.5-3.0] in the non-SPH group, P = .014). When comparing their biochemical evaluation, patients with SPH had higher levels of morning adrenocorticotropin (ACTH) (60.8 pg/mL [43.5-80.3]) compared to patients without SPH (39.4 pg/mL [28.2-53.2], P = .016) and the degree of cortisol reduction after overnight high dose (8 mg or weight-based equivalent) dexamethasone was lower (–58.0% [–85.4 to –49.7]) compared to patients without SPH (85.8 [–90.5 to –76.8], P = .035). The presence of SPH did not affect the odds of remission after surgery or the risk of recurrence after initial remission.

    Conclusion

    SPH in ACTH-secreting pituitary adenomas may affect their biochemical response during endocrine evaluations. They may, for example, fail to suppress to dexamethasone which can complicate diagnosis. Thus, SPH should be mentioned on imaging and taken into consideration in the work up of pediatric patients with CD.

    Acute hemorrhage or necrosis of pituitary adenomas (PAs), defined as pituitary apoplexy, is a rare life-threatening condition that requires emergent neurosurgical evaluation [1]. However, subclinical hemorrhage, necrosis, intratumor cystic degeneration, and/or infarct of PAs, herein all events included in the term subclinical pituitary hemorrhage (SPH) for brevity, may occur in up to 7% to 22% of all pituitary tumors [2-9]. SPH is not associated with significant clinical symptoms and is often discovered at the time of routine diagnostic evaluation [2-9].

    Previous studies suggested that SPH is more common in large tumors, prolactin-secreting or nonfunctioning PAs, while other factors, such as initiation or withdrawal of treatment with dopamine agonists, use of anticoagulants and others, have also been hypothesized to be involved in this process [5, 6]. Overall, adrenocorticotropin (ACTH)-secreting PAs represent small percentage of SPH (0%-3.2% of cases reported) [3, 5, 6, 8]. Although pituitary apoplexy is associated with pituitary hormone deficiencies, SPH has a lower if any effect on the function of the remaining pituitary gland when it occurs in nonfunctioning adenomas [3, 4].

    The diagnosis of Cushing syndrome (CS) involves elaborate and time-dependent tests that are based on cortisol secretion and its regulation by ACTH [10]. Furthermore, the differential diagnosis of ACTH-dependent causes between ACTH-secreting PAs (Cushing disease, CD) and ectopic ACTH secretion is based on several dynamic tests, such as corticotropin-releasing hormone (CRH) stimulation and dexamethasone suppression [11]. The biochemical profile of corticotropinomas with SPH to both baseline and dynamic endocrine tests is not known.

    Materials and Methods

    Participants

    Individuals enrolled under the protocol 97-CH-0076 (ClinicalTrials.gov identifier NCT00001595) at the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), from 2005 to 2020 with confirmed diagnosis of CD, were screened for eligibility in the study. Pediatric and adolescent patients (diagnosis age < 21 years) with imaging studies available before any surgical intervention were included in the analysis. Patients with previous surgery of the pituitary gland or who were evaluated during recurrence were excluded from the study since postoperative changes make imaging findings difficult to distinguish from true SPH, and biochemical presentation at recurrence often differs in severity from initial diagnosis. CS diagnosis was based on criteria defined by the Endocrine Society guidelines and adjusted for the pediatric and adolescent population as needed (abnormal measurements in at least 2 of the following criteria: elevated 24 hour urinary free cortisol [UFC], elevated midnight serum cortisol [> 4.4 mcg/dL in children or > 7.5 mcg/dL for patients age > 18 years], and/or failure to suppress cortisol to 1 mg [or weight-based equivalent dose] overnight dexamethasone suppression test [postdexamethasone cortisol > 1.8 mcg/dL]). CD diagnosis was based on postoperative histologic confirmation of ACTH-secreting PA in most cases, or remission after pituitary surgery even if histologic report failed to identify a PA in the studied material. Remission was defined as postoperative cortisol nadir of less than 2 mcg/dL and/or clinical/biochemical remission during follow-up.

    Informed consent was obtained from parents and assent from patients if developmentally appropriate. Study procedures were approved by the NICHD and/or central National Institutes of Health Institutional Review Board.

    Magnetic Resonance Imaging Scans

    SPH was defined as minimal or no clinical symptoms reported by the patients (apart from those commonly associated with hypercortisolemia) and magnetic resonance imaging (MRI) findings consistent with hemorrhage, intratumor infarction, and/or intratumor cyst formation (suggesting old infarcts) based on the radiologist and the principal investigator’s (C.A.S.) assessment [12, 13]. MRI scans were performed based on standard clinical protocols as previously described [14]. Briefly, MRIs at the National Institutes of Health were performed before and after intravenous administration of gadolinium contrast material, with a gradient echo sequence and thin slices (≤ 1.5 mm). MRIs were performed in either a 1.5 Tesla or 3 Tesla MR machine from various manufacturers over time.

    Clinical and Biochemical Data

    Clinical and biochemical data were extracted from electronic medical records. Tumor size was recorded as the maximum dimension retrieved from the histology report. In cases where histologic report was not available, failed to identify a PA in the studied material, or if the histology report recorded only dimensions on fragments of the tumor, the maximum dimension was retrieved from the MRI images. If the MRI was negative and the histology was negative (but the patient achieved remission after surgery), the tumor size was recorded as missing.

    Serum cortisol and plasma ACTH levels were calculated as the average value of the corresponding levels performed at 23:30 h and 00:00 h (reported as midnight values) and 07:30 h and 08:00 h (reported as morning values). Twenty-four–hour UFC was calculated as the average of the first 2 or 3 samples reported in the electronic medical records. Given the possible differences in the assays and reported reference range for UFC, we calculated the increase of UFC based on the upper limit of normal according to the following formula: UFC fold change = UFC/upper limit of the reference range. Serum cortisol was measured with solid-phase, competitive chemiluminescent enzyme immunoassay on a Siemens Immulite 2500 analyzer. Plasma ACTH was measured with a chemiluminescence immunoassay on a Siemens Immulite 200 XPi analyzer. UFC was measured with a chemiluminescent enzyme immunoassay until 2011 and with high-performance liquid chromatography–tandem mass spectrometry since 2011. High-dose dexamethasone suppression test was performed as previously described. Briefly, oral dexamethasone (120 mcg/kg, max 8 mg) was administered at 23:00 and cortisol was measured before (8 AM the day of administration) and after (9 AM the day after dexamethasone administration). The change of cortisol was calculated as: 100* [(postdexamethasone cortisol – predexamethasone cortisol)/predexamethasone cortisol]. Levels of cortisol lower than the lower limit of detection of the assay (< 1 mcg/dL) were substituted with the intermediate value (0.5) for all analyses. CRH stimulation test was performed as previously described. Briefly, an intravenous catheter was placed in the forearm the night before testing; the patient was fasting and lying in bed, and ovine CRH (oCRH) was administered (1 mcg/kg, max 100 mcg). Samples for cortisol and ACTH were taken at –5, 0, 15, 30, and 45 minutes after the administration of oCRH. The response to the last was expressed as the percentage change from baseline by subtracting the pretest cortisol and ACTH values from the posttest values and dividing by the former. The mean cortisol increase was estimated at 30 and 45 minutes from baseline. For ACTH the mean increase was estimated at 15 and 30 minutes after the administration of oCRH. CRH stimulation test was not performed after the discontinuation of oCRH by the company in November 2020.

    Statistical Analysis

    Categorical data are described as counts (proportions) and were compared between groups using the Fisher exact test. Fisher odds ratio (OR) was used to assess the odds of remission based on the presence of SPH and is presented as OR and 95% CI.

    Continuous data were checked for normality and not normally distributed data are presented as median (first quartile to third quartile) and were compared between 2 groups using Wilcoxon rank-sum test. The Cox proportional hazards test was used to assess the risk of recurrence based on the presence of SPH and is presented as hazard ratio (HR) and 95% CI. Statistical analyses were performed in R.

    Results

    Clinical Data

    Out of 170 patients with available MRI before first surgery, 12 patients had evidence of possible SPH (7.1%) (Table 1). Various MRI findings were noted (Fig. 1), most commonly hyperintense lesions in T1 and T2 precontrast images (Fig. 1A and 1B), while some patients had intratumor heterogeneity suggestive of cystic formation (Fig. 1C-1F). As expected, the tumor size of patients with SPH as noted in histology reports or MRI images was higher than that in patients without SPH (median size: 8.5 mm [7.0-11.25] in the SPH group vs 5.4 mm [4-8] in the non-SPH group; P < .001).

     

           
           
           
           
           
           
           
           
           
           
           
           
           
           
           
           

     

     

     
    Table 1.

    Characteristics of patients with and without subclinical pituitary hemorrhage, necrosis, and/or cystic degeneration

      No SPH (N = 158) SPH (N = 12) P
    Age at diagnosis, y  13.0 (10.6 to 15.4)  12.5 (10.6 to 15.6)  .95 
    Sex       
     Female  89 (56.3%)  5 (41.7%)  .49 
     Male  69 (43.7%)  7 (58.3%)   
    Disease duration, y  2.50 (1.5 to 3.0)
    n = 138 
    1.00 (1.0 to 2.0)
    n = 10 
    .014 
    Morning cortisol, mcg/dL  16.2 (12.6 to 20.4)
    n = 139 
    18.4 (13.7 to 27.5)
    n = 10 
    .28 
    Midnight cortisol, mcg/dL  14.0 (10.7 to 19.7)
    n = 133 
    16.3 (11.0 to 23.0)
    n = 9 
    .52 
    UFC fold change  4.89 (2.51 to 8.50)
    n = 131 
    8.81 (6.86 to 9.42)
    n = 9 
    .18 
    Morning ACTH, pg/mL  39.4 (28.2 to 53.2)
    n = 143 
    60.8 (43.5 to 80.3)
    n = 10 
    .016 
    Cortisol change during CRH stimulation test, %  68.7 (33.3 to 111)
    n = 128 
    46.0 (–7.46 to 90.7)
    n = 8 
    .22 
    ACTH change during CRH stimulation test, %  145 (59.6 to 260)
    n = 127 
    103 (–5.75 to 278)
    n = 8 
    .55 
    Cortisol change during high-dose dexamethasone suppression test, %  –85.8 (–90.5 to –76.8)
    n = 120 
    –58.0 (–85.4 to –49.7)
    n = 9 
    .035 
    Remission       
     Yes  127 (80.4%)  10 (83.3%)  .99 
     No  25 (15.8%)  2 (16.7%)   

    Number in each cell reports the number of patients with available results.

    Abbreviations: ACTH, adrenocorticotropin; CRH, corticotropin-releasing hormone; SPH, subclinical pituitary hemorrhage, necrosis, and/or cystic degeneration; UFC, urinary free cortisol.

    Figure 1.

    Imaging findings in patients with subclinical pituitary hemorrhage, necrosis, and/or cystic degeneration. A, T1, and B, T2 magnetic resonance imaging (MRI) scans of the same patient showing area of high intensity inside the tumor suggesting acute/subacute episode. C, T2, and D, T1 MRI scans of the same patient showing heterogeneity in a large tumor with high-intensity areas suggesting blood-filled cavities and/or necrosis. E, T1, and F, T2 MRI scans of the same patient showing fluid inside the tumor.

     
    Imaging findings in patients with subclinical pituitary hemorrhage, necrosis, and/or cystic degeneration. A, T1, and B, T2 magnetic resonance imaging (MRI) scans of the same patient showing area of high intensity inside the tumor suggesting acute/subacute episode. C, T2, and D, T1 MRI scans of the same patient showing heterogeneity in a large tumor with high-intensity areas suggesting blood-filled cavities and/or necrosis. E, T1, and F, T2 MRI scans of the same patient showing fluid inside the tumor.
     

     

    Patients with and without SPH were similar in age (median age: 12.5 years [10.6-15.6] in the SPH group vs 13.0 years [10.6-15.4] in the non-SPH group; P = .95) and sex distribution (n of female = 5 (41.7%) in the SPH group vs 89 (56.3%) in the non-SPH group; P = .49). Patients with SPH had a shorter duration of disease as noted by changes in their growth chart parameters (median duration: 1.0 [1.0-2.0] year in the SPH group vs 2.5 [1.5-3.0] years in the non-SPH group; P = .014).

    Patients in the 2 groups did not differ on their anthropometric characteristics, including height and body mass index z scores(P > .05). They also had similar blood pressure parameters and did not differ in terms of the frequency of hypertension diagnosis. No patient was on anticoagulation treatment nor had received radiation at the time of the MRI. One patient in the SPH group had a history of lower leg deep vein thrombosis and was previously on low-heparin therapy, but he had stopped treatment at least 6 months before the MRI.

    Biochemical Evaluation of Hypercortisolemia

    Morning and midnight serum cortisol and 24-hour UFC levels were similar in both groups, but patients with SPH had higher levels of morning ACTH (60.8 pg/mL [43.5-80.3]) compared to patients without SPH (39.4 pg/mL [28.2-53.2]; P = .016). Changes in cortisol and ACTH levels during the CRH stimulation test were similar between the 2 groups, but patients with SPH who underwent the overnight high-dose dexamethasone suppression test (n = 8) had lower suppression of cortisol after dexamethasone (–58.0% [–85.4 to –49.7]) compared to patients without SPH (n = 120) (–86.0% [–90.5 to –76.7]; P = .035) (Fig. 2). When the cutoff of suppression of more than 69% was considered, patients with SPH had a lower chance of suppressing more than 69% compared to patients without SPH (OR: 0.18; 95% CI, 0.03-0.95).

    Figure 2.

    Cortisol levels before and after high-dose dexamethasone suppression test in patients with and without subclinical pituitary hemorrhage, necrosis, and/or cystic degeneration (SPH).

     
     Cortisol levels before and after high-dose dexamethasone suppression test in patients with and without subclinical pituitary hemorrhage, necrosis, and/or cystic degeneration (SPH).
     

     

    Remission After Surgical Treatment and Risk for Recurrence

    The chance of immediate postoperative remission after surgery was similar in patients with and without SPH. For patients with initial remission and follow-up of at least 3 months, analysis of the risk of recurrence did not show a difference in recurrence rate between the 2 groups (HR: 1.12; 95% CI, 0.13-9.4, in the SPH group compared to the non-SPH group, adjusting for the neurosurgeon).

    Discussion

    SPH is often an incidental finding in the imaging evaluation of patients with PAs. The frequency of SPH in patients with CD is low (7.1% in our study) but these patients may differ in terms of their history of shorter duration of symptoms and the biochemical evaluation. More specifically, patients with CD and SPH showed higher ACTH levels and lower suppression of cortisol to high-dose dexamethasone. This, however, did not affect their prognosis in terms of immediate postoperative remission and long-term risk of recurrence.

    SPH has been mainly studied in cohorts of patients with various types of PAs. From these studies important conclusions have been made suggesting that the risk of SPH is higher in patients with large nonfunctioning PAs or prolactinomas. Other risk factors for pituitary apoplexy are thought to be size of the adenoma, change in size, initiation, and withdrawal of dopamine agonists, type of dopamine agonist, use of anticoagulants, diabetes mellitus, hypertension, head trauma, radiotherapy, and preceding dynamic endocrine testing.

    Patients with CD often represent a small portion of these cohorts, and to our knowledge there is no study to investigate how these patients respond to stimulation/suppression tests. For that reason, we evaluated these findings in our cohort of only patients with ACTH-secreting adenomas. As most of our referrals involve pediatric patients, we limited our cohort only to patients diagnosed at younger than 21 years to have a more homogeneous group.

    The pathogenesis of pituitary apoplexy has been hypothesized to lie in more friable vessels in PAs, which, while the tumor increases in size, are more susceptible to rupture or cause surrounding feeding vessels to extend and bleed [7, 15]. CS, because of the coexisting hypercortisolemia, leads independently to endothelial dysfunction and coagulation defects, which are often apparent as easy bruising, thrombotic events, and other signs. However, review of the literature and the estimated frequency of SPH in our cohort suggest that patients with CD are not at increased risk for SPH, potentially related to the relatively small adenomas present in these patients [16, 17].

    The main difference of patients with CD and SPH compared to those without lies in their biochemical testing, more characteristically in lower suppression to dexamethasone. The overnight high-dose dexamethasone suppression test was originally designed to differentiate various types of CS [18]. Although originally described as a highly accurate test, in clinical practice, cutoffs of 50% to 80% have shown variable sensitivity and specificity and certain centers opt not to use this test unless all other diagnostic evaluations yield confounding results [19-21]. In previous studies a threshold of suppression of more than 69% showed the highest accuracy (sensitivity: 71%, specificity: 100%), and we have incorporated this in our diagnostic algorithm (acknowledging the limitations of the test) [22, 23]. In our analysis, patients with SPH had lower suppression of cortisol under the effect of high-dose dexamethasone and a higher chance of not passing the aforementioned threshold. The mechanism for the lower suppression to dexamethasone of these tumors may be due to lower vascular circulation of dexamethasone at the level of the tumor, and/or the lower sensitivity of necrotic cells to the negative feedback by circulating glucocorticoids.

    A limitation of this study was that the diagnosis of SPH was based on MRI findings. However, MRI sequences and machines differed between patients and over time. Thus, although large hemorrhagic/necrotic lesions are probably accurately identified, it is possible that smaller lesions are misclassified as negative; the effect of smaller hemorrhagic areas to the biochemical testing may however be smaller as well. Further, the MRIs were not read by a central radiologist, but rather from the radiologist on call at each time point, and this could lead to discrepancies in readings. In addition, our cohort’s data may not be generalized to the pediatric or adult CD population, as often our referrals consist of patients with difficult to treat, small, or otherwise unusual tumors.

    In conclusion, SPH may be incidentally identified in up to 7% of patients with CD. Patients with CD and SPH may differ in terms of their response to endocrine tests, and this finding should be incorporated in their evaluation.

    Abbreviations

               

    • ACTH

      adrenocorticotropin

    • CD

      Cushing disease

    • CRH

      corticotropin-releasing hormone

    • CS

      Cushing syndrome

    • HR

      hazard ratio

    • MRI

      magnetic resonance imaging

    • NICHD

      Eunice Kennedy Shriver National Institute of Child Health and Human Development

    • OR

      odds ratio

    • PA

      pituitary adenoma

    • SPH

      subclinical pituitary hemorrhage

    • UFC

      urinary free cortisol

    Financial Support

    This work was supported by the intramural research program of the Eunice Kennedy Shriver NICHD, NIH, Bethesda, MD 20892, USA.

    Disclosures

    Dr Stratakis holds patents on the function of the PRKAR1A, PDE11A, and GPR101 genes and related issues; his laboratory has also received research funding on the GPR101 gene, and on abnormal growth hormone secretion and its treatment by Pfizer, Inc. He is currently employed by ELPEN, SA and has been consulting for Lundbeck Pharmaceuticals and Sync, SA. The other authors have nothing to disclose.

    Data Availability

    Some or all datasets generated during and/or analyzed during the current study are not publicly available but may be available from the corresponding author on reasonable request.

     

     

     
    Published by Oxford University Press on behalf of the Endocrine Society 2022.
    This work is written by (a) US Government employee(s) and is in the public domain in the US.
    Published by Oxford University Press on behalf of the Endocrine Society 2022.
  5. Introduction: Patients with Cushing’s syndrome (CS) represent a highly sensitive group during corona virus disease 2019 (COVID-19) pandemic. The effect of multiple comorbidities and immune system supression make the clinical picture complicated and treatment challenging.

    Case report: A 70-year-old female was admitted to a covid hospital with a severe form of COVID-19 pneumonia that required oxygen supplementation. Prior to her admission to the hospital she was diagnosed with adrenocorticotropic hormone (ACTH)-dependent CS, and the treatment of hypercortisolism had not been started yet. Since the patient’s condition was quickly deteriorating, and with presumend immmune system supression due to CS, we decided on treatement with intraveonus immunoglobulins (IVIg) that enabled quick onset of immunomodulatory effect. All comorbidities were treated with standard of care. The patient’s condition quickly stabilized with no direct side effects of a given treatment.

    Conclusion: Treatment of COVID-19 in patients with CS faces many challenges due to the complexity of comorbidity effects, immunosupression and potential interactions of available medications both for treatment of COVID-19 and CS. So far, there are no guidelines for treatment of COVID-19 in patients with active CS. It is our opinion that immunomodulating therapies like IVIg might be an effective and safe treatment modality in this particularly fragile group of patients.

     

    Introduction

    Dealing with corona virus disease 2019 (COVID-19) focused medical attention on several sensitive population groups. While the knowledge is still improving, some of the recognized risk factors for severe form of the disease are male sex, older age, obesity, hypertension, diabetes mellitus, and cardio-vascular disease (1). This constellation of morbidities is particularly intriguing from endocrine point of view, since they are all features of patients with Cushing’s syndrome (CS). Another relevant feature of CS is a propensity for infections due to profound immune suppression, with prevalence of 21-51%; even more so, infections are the second cause of death (31%) in CS after disease progression, and are the main cause of death (37%) in patients who died within 90 days of diagnosis (2).

    Immune system alterations in CS lead to depression of both innate and adaptive immune responses, favoring not only commonly acquired but also opportunistic bacterial infections, fungal infections, and severe, disseminated viral infections (3). Susceptibility to infections directly positively correlates with cortisol level, and is more frequent in ectopic ACTH secretion (EAS). Hypercortisolism hampers the first-line response to external agents and consequent activation of the adaptive response (3). Consequently, there is a decrease in total number of T-cells and B-cells, as well as a reduction in T-helper cell activation, which might favor opportunistic and intracellular infections. On the other hand, an increase in pro-inflammatory cytokine secretion, including interleukine-6 (IL-6) and tumor necrosis factor-α (TNF-α) leads to persistent, low-grade inflammation. It is important to note that immune system changes are confirmed both during the active phase and while in remission of CS (3).

    In view of the aforementioned data, a few topics emerge regarding patients with CS and COVID-19. Initial clinical presentation may be altered – low-grade chronic inflammation and poor immune reaction might limit febrile response in the early phase of infection, aggravating timely diagnosis (4). Increased cytokine levels may put patients with CS at increased risk of severe course and progression to acute respiratory distress syndrome (ARDS). On the other hand, the rise in cytokine levels associated with exposure to external agents is significantly hampered, probably because of persistently elevated pro-inflammatory cytokine secretion (4, 5). Patients with CS have a possibility for prolonged duration of viral infections and risk for superinfections leading to sepsis and increased mortality risk; this is especially relevant for hospitalized patients and mandates empirical prophylaxis with broad-spectrum antibiotics (6). Both COVID-19 and CS individually represent disease states of increased thromboembolic (TE) risk, requiring additional care (6).

    Due to very limited data, it is still not possible to address these topics with certainty and make recommendations for optimal management of these patients. Current clinical practice guidance for management of CS during COVID-19 commissioned by the European Society of Endocrinology (ESE) emphasizes prompt and optimal control of hypercortisolism and adequate treatment of all comorbidities (7). Although individual circumstances must always be considered, we need more direct clinical experience, especially regarding the actual treatment of COVID-19 in this sensitive group. So far, there are only five published case studies of patients with CS and COVID-19, with eight patients in total (812). In this study, we present a patient with newly diagnosed ACTH-dependent CS who was diagnosed with COVID-19 before the initiation of specific medical treatment.

    Case Report

    A 70-year-old female was admitted to our Covid hospital due to bilateral interstitial pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Six days before she was discharged from endocrinology department of another hospital where she was hospitalized due to newly diagnosed diabetes mellitus. Her personal history was unremarkable, and she was vaccinated with two doses of inactivated COVID-19 vaccine Sinopharm BBIBP. During this hospitalization Cushingoid features were noted (moon face, centripetal obesity, thin extremities with multiple hematomas, bilateral peripheral edema), as well as diabetes mellitus (HbA1c 8.7%), arterial hypertension (BP 180/100 mmHg), hypokalemia (2.0 mmol/L), mild leukocytosis (WBC 12.9x10e9/L) with neutrophilia, and mildly elevated CRP (12.3 mg/L). Hormonal functional testing confirmed ACTH-dependent Cushing’s syndrome: morning ACTH 92.6 pg/mL (reference range 10-60 pg/mL), morning serum cortisol 1239 nmol/L (reference range 131-642 nmol/L), midnight serum cortisol 1241 nmol/L, lack of cortisol suppression in overnight dexamethasone suppression test (978 nmol/L). Pituitary MRI was unremarkable other than empty sella, and CT scan of thorax normal other than left adrenal hyperplasia. Diabetes mellitus was successfully controlled with metformin, hypertension with ACE-inhibitor, Ca-channel blocker and beta-blocker, and hypokalemia with potassium supplementation along with spironolactone. Steroidogenesis inhibitors were not available in this institution, but before referral to a tertiary care hospital she was tested for SARS-CoV-2, and the test came back positive (sample was obtained by nasopharyngeal swab). Since she was asymptomatic, with normal thoracic CT scan and stabile CRP level (9.1 mg/L), she was discharged with detailed recommendations for conduct in case of progression of COVID symptoms.

    Next day she started feeling malaise with episodes of fever (up to 38.2°C). Symptomatic therapy was advised in an outpatient clinic (no antiviral therapy was recommended), but 5 days later respiratory symptoms ensued. During examination, the patient was weak, with dyspnea and tachypnea (RR 22/min), afebrile (36.9°C) and with oxygen saturation (SO2) of 85% measured by pulse oximeter. Chest X-ray confirmed bilateral interstitial pneumonia with parenchymal consolidation in the right lower lung lobe, so she was referred to the COVID hospital.

    Laboratory analyses upon admission are presented in the Supplementary Table 1. In addition to her previous testing, elevated chromogranin A (CgA) level was verified (538.8 ng/mL, reference range 11-98.1). The patient was treated with supplemental oxygen with maximal flow of 13 l/min. For the reason of previously confirmed severe endogenous hypercortisolism, glucocorticoids were not administered. Due to limited therapeutic options and presumed further clinical deterioration, we decided to treat the patient with intravenous immunoglobulins (IVIg) 30 g iv for 5 days, starting from the 2nd day of hospitalization. We did not observe any side effects of a given treatment. In parallel, the patient received broad-spectrum antibiotics (ceftazidime and levofloxacin), proton pump inhibitor, LMWH in prophylactic dose, oral and parenteral potassium supplementation along with spironolactone. She continued with her previous antihypertensive therapy with good control of blood pressure. While the patient was on oxygen supplementation, glycaemia was controlled with short acting insulin before meals. Following given treatment, we observed clinical, biochemical (Supplementary Table 1.) and radiological improvement (Supplementary Figure 1). Oxygen supplementation was gradually discontinued. With regard to D-dimer levels and risk factors for TE events due to COVID-19 and CS, we performed color Doppler scan of lower extremities veins, and CT pulmonary angiography, but there were no signs of thrombosis. During hospital stay, there were no signs of secondary infection and cotrimoxazole was not added to the current treatment. The patient was discharged with advice to continue her prior medical therapy along with increased dose of spironolactone and initiation of rivaroxaban. She was referred to the tertiary institution for the initiation of steroidogenesis inhibitor and further diagnostics.

    Discussion

    Endogenous Cushing’s syndrome is a rare disease with an incidence of 0.7-2.4 million person-years in European population-based studies (13). Significant morbidity yields a standard mortality ratio of 3.7 (95%CI 2.3–5.3), with the highest mortality during the first year after initial presentation. COVID-19 pandemic imposes additional challenge to this fragile group of patients. Due to lack of solid experience, it is still difficult to define potential clinical course and outcome of patients with CS and COVID-19. In addition, currently there are no guidelines for management of SARS-CoV-2 infection in patients with active CS.

    So far, only two small case series followed patients with Cushing’s disease (CD) in various disease stages (not all were active) during COVID-19 pandemic (9, 12). Small number of SARS-CoV-2 positive cases (3/22 and 2/61) is clearly biased by shortness of analyzed period (one and a half, and three and a half months). Additionally, a small number of patients was actually tested by nasopharyngeal swab for SARS-CoV-2 even in the presence of indicative symptoms, albeit mild. Nevertheless, all these limitations included, it seems that the prevalence of COVID-19 might be greater in patients with CD than in general population (12). This is accordant with studies on patients on exogenous glucocorticoid (GC) treatment. Overall, there is a growing body of evidence that patients on chronic GC therapy are at higher risk for SARS-CoV-2 infection and a severe course of disese, regardless of age and comorbidities (14). In many studies patients on high-dose GC therapy were at particularly high risk for a severe course of disease, so it is reasonable to assume that there is a dose-dependent effect (14).

    All patients except one with endogenous CS and COVID-19 presented in literature were hospitalized, with majority of them requiring oxygen supplementation, which classified them as serious cases of disease (812). Parameters of inflammation (namely CRP) were highly variable (from normal to elevated) and did not seem to reflect severity of COVID-19 consistently. Two patients had fatal outcome; one with postoperative hypocortisolism that required stress doses of hydrocortisone, and with terminal kidney failure as significant comorbidity; the other with suspected EAS who developed ARDS in contrast to normal CRP and absence of fever (9, 12). Based on reported cortisol levels in these patients, it seems that the severity of COVID-19 pneumonia depended on severity of hypercortisolism (812). A patient with probable EAS even developed ARDS, which adds to ongoing controversy regarding the risk of ARDS due to SARS-CoV-2 in patients with CS (3, 15). We ourselves have treated a severely obese female patient with active CD on pasireotide, who developed ARDS despite addition of high doses of methylprednisolone (unpublished data). Additional risk imposed by comorbidities cannot be underestimated (15, 16). This is particularly relevant for obesity, that not only hampers immune system (leading to increased levels of IL-1, IL-6, and TNF-α), but adipocytes represent a reservoir of SARS-CoV-2 thanks to ACE2 receptor, crucial for virus attachment (15).

    Majority of depicted patients with active CS were already medically treated for hypercortisolism but with various compliance (sometimes very poor), and two young patients have just started steroidogenesis inhibitors (metyrapone/ketoconazole). Infection with SARS-CoV-2 was treated by national protocols that were mostly based on supportive care. These protocols changed over time, so a few patients received antiviral therapy (favipiravir), and one young patient with suspected EAS was treated with methylprednisolone along with high doses of ketoconazole (10). Treatment was complicated with adrenal insufficiency (AI) in three patients (8, 11, 12).

    We have presented a patient with CS and rapid development of serious case of COVID-19 pneumonia that required hospital admission and oxygen support. She was febrile and had positive laboratory parameters of inflammation. Her CS was active, with very high cortisol levels, no prior medical treatment and with clinical suspicion of EAS (ACTH-dependent disease of short duration, severe hypercortisolism, hypokalemia, very high CgA, no visible pituitary tumor). With this in mind, and with regard to rapid progression of COVID-19 pneumonia, it was our opinion that the patient required treatment with quick onset and presumable immune system modulation.

    A logical approach to treatment of CS during COVID-19 pandemic includes meticulous therapy for comorbidities (namely antihypertensives, anti-diabetic drugs, low molecular weight heparin, etc.), and steroidogenesis inhibitors for treatment for hypercortisolemia (7). While some of these drugs demonstrate quick onset of action regarding normalization of cortisol level (and hence improve clinical comorbidities), rapid effects on immune system responses are not likely, which might be of great relevance in case of acute infection. Secondly, adrenolytic therapy increases a risk of AI, which can be even more perilous than CS in case of infection or other stress situations (8, 12, 15, 16). A modified “block and replace” approach may be considered, where addition of hydrocortisone could diminish the risk of AI (7). Still, there are a few potential pitfalls with this regimen as well. Some people fail to respond to high doses of adrenal-blocking agents due to genetic differences in the steroidogenic enzymes, since therapeutic responses to metyrapone and ketoconazole in patients with CS are associated with the polymorphism in the CYP17A1 gene (17). Additionally, there are not enough data about possible interactions between adrenolytic drugs (majority of them being metabolized through the CYP450/CYP3A4 pathway) and medications used to treat COVID-19, most of which are only just emerging (18). Special concerns, amplified with similar potential effects of SARS-CoV-2 itself as well as specific therapies are liver dysfunction (metyrapone, ketoconazole), hypokalemia (metyrapone, ketoconazole), QT-interval prolongation (ketoconazole, osilodrostat), gastrointestinal distress (mitotane, osilodrostat, etomidate) (18). Metyrapone may cause accumulation of androgenic precursors secondary to the blockade of cortisol synthesis, that can virtually enhance expression of transmembrane protease serine 2 (TMPRSS2), found to be essential to activate the viral spikes, induce viral spread, and pathogenesis in the infected hosts (19). Another important issue concerns biochemical estimation of disease control (and hence risk for AI), since most commercially available assays can overestimate cortisol level in patients treated with metyrapone due to cross-reactivity with the precursor 11-deoxicortisol (7, 15). Mass spectrometry is a method of choice to overcome this problem, but it is not available in many centers. Some centers advocate titration and/or temporary halting medical therapies in the treatment of patients with CS in the context of COVID-19 infection (20). Treatement was stopped in a few patients with severe COVID-19 symptoms who were then given high dose GC for a few days with no long-term complications, and with full recovery (20).

    There are no data about the effect of anti-viral drugs in patients with CS and COVID-19. A special concern refers to adipose tissuse, as adipose tissue is difficult for antiviral drugs to reach. It cannot be excluded that the constant release of viral replicas from the adipose tissue reservoir may interfere with COVID-19 infection treatment, delaying its resolution and favoring a worse prognosis (15). If antiviral drugs are started, it is suggested that immunocompromised patients may require prolonged therapy (18). However, the timing is difficult in practice and candidates for antivirals are limited.

    Since the clinical course of COVID-19 only initially depends on viral replication, immunomodulatory therapy emerged as a valuable treatment option to control the host immune response. This became apparent ever since RECOVERY trial proved efficacy of glucocortiods (21). But this therapeutic option is fairly inapplicable in patients with active CS, since glucocorticoid treatment in chronic hypercortisolism seems to enhance immune system alterations (22). In parallel with the development of new agents, it is prudent to study the efficacy of existing therapeutic options with acceptable safety profile (20). Beside glucocorticoids, inflammation blockers, intravenous immunoglobulin and convalescent plasma were used in various settings (23).

    Intravenous immunoglobulin (IVIg) is a blood product prepared from the serum pooled from thousands of healthy donors, containing a mixture of polyclonal IgG antibodies, mostly IgG1 and IgG2 subclasses (24, 25). Initial rationale for its use was immunodefficiency due to hypoglobulinemia. Since then it has been shown that IVIg exerts pleiotropic immunomodulating action involving both innate and adaptive immunity and it has been used in a variety of diseases (26). In previous studies on MERS (Middle East Respiratory Syndrome) and SARS (Severe Acute Respiratory Syndrome) using IVIg showed beneficial clinical effects (25). Although pathogenesis of COVID-19 has not be fully elucidated, there is a consensus that immune-mediated inflammation plays an important role in the progression of this disease, just as it did in prior coronavirus infections (27). In this context, the actual role of IVIg in COVID-19 patients might be not to boost the immune system, but through its immunomodulatory effect to suppress a hyperactive immune response that is seen in some patients (28). So far, a limited number of studies, case series and meta-analyses demonstrate a promising potential of IVIg in patients with COVID-19. The effect was demonstrated in terms of mortality, improvement of clinical symptoms, laboratory examinations, imaging and length of hospital stay, especially in patients with moderate/severe form of the disease, and with emphasis on early administration (within 3 days of admission) (24, 25, 2731). A recent double blind, placebo-controlled, phase 3, randomized trial tested hyperimmune intravenous immunoglobulin (hIVIg) to SARS-CoV-2 derived from recovered donors with no demonstrated effect compared with standard of care, but therapy was administered in patients symptomatic up to 12 days (32). Additional clinical trials are underway, hopefully with more guidance for proper selection of patients that might benefit from this type of treatment.

    Conclusion

    To our knowledge, this is the first case of IVIg treatment in a COVID-19 patient with CS. It is our opinion that immune-modulating properties of IVIg might present an attractive treatment option, especially in those CS patients that show rapid clinical progression and positive laboratory parameters of inflammation. While we await for new therapeutic modalities for COVID-19 and while some of the modalities remain not widely available, IVIg is more accessible, safe method, which could be rescuing in carefully selected patients. Of note, we consider our patient’s vaccinal status as an unquestionable positive contributor to the favorable outcome

    Data Availability Statement

    The raw data supporting the conclusions of this article will be made available by the authors, without undue reservation.

    Ethics Statement

    Ethical review and approval was not required for the study on human participants in accordance with the local legislation and institutional requirements. The patients/participants provided their written informed consent to participate in this study. Written informed consent was obtained from the individual(s) for the publication of any potentially identifiable images or data included in this article.

    Author Contributions

    BP, AS, JV, TG, MJ-L, JV, VS, ZG and TA-V analyzed and interpreted the patient data. BP, AP, DI, and DJ were major contributors in writing the manuscript. All authors contributed to the article and approved the submitted version.

    Conflict of Interest

    The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

    Publisher’s Note

    All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

    Supplementary Material

    The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fendo.2022.889928/full#supplementary-material

    References

    1. Hu J, Wang Y. The Clinical Characteristics and Risk Factors of Severe COVID-19. Gerontology (2021) 67(3):255–66. doi: 10.1159/000513400

    PubMed Abstract | CrossRef Full Text | Google Scholar

    2. Valassi E, Tabarin A, Brue T, Feelders RA, Reincke M, Netea-Maier R, et al. High Mortality Within 90 Days of Diagnosis in Patients With Cushing's Syndrome: Results From the ERCUSYN Registry. Eur J Endocrinol (2019) 181(5):461–72. doi: 10.1530/EJE-19-0464

    PubMed Abstract | CrossRef Full Text | Google Scholar

    3. Hasenmajer V, Sbardella E, Sciarra F, Minnetti M, Isidori AM, Venneri MA. The Immune System in Cushing's Syndrome. Trends Endocrinol Metab (2020) 31(9):655–69. doi: 10.1016/j.tem.2020.04.004

    PubMed Abstract | CrossRef Full Text | Google Scholar

    4. Pivonello R, Isidori AM, De Martino MC, Newell-Price J, Biller BM, Colao A. Complications of Cushing's Syndrome: State of the Art. Lancet Diabetes Endocrinol (2016) 4(7):611–29. doi: 10.1016/S2213-8587(16)00086-3

    PubMed Abstract | CrossRef Full Text | Google Scholar

    5. Martins-Filho PR, Tavares CSS, Santos VS. Factors Associated With Mortality in Patients With COVID-19. A Quantitative Evidence Synthesis of Clinical and Laboratory Data. Eur J Intern Med (2020) 76:97–9. doi: 10.1016/j.ejim.2020.04.043

    PubMed Abstract | CrossRef Full Text | Google Scholar

    6. Pivonello R, Ferrigno R, Isidori AM, Biller BMK, Grossman AB, Colao A. COVID-19 and Cushing's Syndrome: Recommendations for a Special Population With Endogenous Glucocorticoid Excess. Lancet Diabetes Endocrinol (2020) 8(8):654–6. doi: 10.1016/S2213-8587(20)30215-1

    PubMed Abstract | CrossRef Full Text | Google Scholar

    7. Newell-Price J, Nieman LK, Reincke M, Tabarin A. ENDOCRINOLOGY IN THE TIME OF COVID-19: Management of Cushing's Syndrome. Eur J Endocrinol (2020) 183(1):G1–7. doi: 10.1530/EJE-20-0352

    PubMed Abstract | CrossRef Full Text | Google Scholar

    8. Beretta F, Dassie F, Parolin M, Boscari F, Barbot M, Busetto L, et al. Practical Considerations for the Management of Cushing's Disease and COVID-19: A Case Report. Front Endocrinol (Lausanne) (2020) 11:554. doi: 10.3389/fendo.2020.00554

    PubMed Abstract | CrossRef Full Text | Google Scholar

    9. Belaya Z, Golounina O, Melnichenko G, Tarbaeva N, Pashkova E, Gorokhov M, et al. Clinical Course and Outcome of Patients With ACTH-Dependent Cushing's Syndrome Infected With Novel Coronavirus Disease-19 (COVID-19): Case Presentations. Endocrine (2021) 72(1):12–9. doi: 10.1007/s12020-021-02674-5

    PubMed Abstract | CrossRef Full Text | Google Scholar

    10. Rehman T. Image of the Month: Diagnostic and Therapeutic Challenges in the Management of Ectopic ACTH Syndrome: A Perfect Storm of Hypercortisolism, Hyperglycaemia and COVID-19. Clin Med (Lond) (2021) 21(3):231–4. doi: 10.7861/clinmed.2021-0005

    PubMed Abstract | CrossRef Full Text | Google Scholar

    11. Yuno A, Kenmotsu Y, Takahashi Y, Nomoto H, Kameda H, Cho KY, et al. Successful Management of a Patient With Active Cushing's Disease Complicated With Coronavirus Disease 2019 (COVID-19) Pneumonia. Endocr J (2021) 68(4):477–84. doi: 10.1507/endocrj.EJ20-0613

    PubMed Abstract | CrossRef Full Text | Google Scholar

    12. Serban AL, Ferrante E, Carosi G, Indirli R, Arosio M, Mantovani G. COVID-19 in Cushing Disease: Experience of a Single Tertiary Centre in Lombardy. J Endocrinol Invest (2021) 44(6):1335–6. doi: 10.1007/s40618-020-01419-x

    PubMed Abstract | CrossRef Full Text | Google Scholar

    13. Sharma ST, Nieman LK, Feelders RA. Cushing's Syndrome: Epidemiology and Developments in Disease Management. Clin Epidemiol (2015) 7:281–93. doi: 10.2147/CLEP.S44336

    PubMed Abstract | CrossRef Full Text | Google Scholar

    14. Vogel F, Reincke M. Endocrine Risk Factors for COVID-19: Endogenous and Exogenous Glucocorticoid Excess. Rev Endocr Metab Disord (2021) 23(2):233–50. doi: 10.1007/s11154-021-09670-0

    PubMed Abstract | CrossRef Full Text | Google Scholar

    15. Guarnotta V, Ferrigno R, Martino M, Barbot M, Isidori AM, Scaroni C, et al. Glucocorticoid Excess and COVID-19 Disease. Rev Endocr Metab Disord (2021) 22(4):703–14. doi: 10.1007/s11154-020-09598-x

    PubMed Abstract | CrossRef Full Text | Google Scholar

    16. Chifu I, Detomas M, Dischinger U, Kimpel O, Megerle F, Hahner S, et al. Management of Patients With Glucocorticoid-Related Diseases and COVID-19. Front Endocrinol (Lausanne) (2021) 12:705214. doi: 10.3389/fendo.2021.705214

    PubMed Abstract | CrossRef Full Text | Google Scholar

    17. Valassi E, Aulinas A, Glad CA, Johannsson G, Ragnarsson O, Webb SM. A Polymorphism in the CYP17A1 Gene Influences the Therapeutic Response to Steroidogenesis Inhibitors in Cushing's Syndrome. Clin Endocrinol (Oxf) (2017) 87(5):433–9. doi: 10.1111/cen.13414

    PubMed Abstract | CrossRef Full Text | Google Scholar

    18. Berlinska A, Swiatkowska-Stodulska R, Sworczak K. Old Problem, New Concerns: Hypercortisolemia in the Time of COVID-19. Front Endocrinol (Lausanne) (2021) 12:711612. doi: 10.3389/fendo.2021.711612

    PubMed Abstract | CrossRef Full Text | Google Scholar

    19. Barbot M, Ceccato F, Scaroni C. Consideration on TMPRSS2 and the Risk of COVID-19 Infection in Cushing's Syndrome. Endocrine (2020) 69(2):235–6. doi: 10.1007/s12020-020-02390-6

    PubMed Abstract | CrossRef Full Text | Google Scholar

    20. Fleseriu M. Pituitary Disorders and COVID-19, Reimagining Care: The Pandemic A Year and Counting. Front Endocrinol (Lausanne) (2021) 12:656025. doi: 10.3389/fendo.2021.656025

    PubMed Abstract | CrossRef Full Text | Google Scholar

    21. Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, Linsell L, et al. Dexamethasone in Hospitalized Patients With Covid-19. N Engl J Med (2021) 384(8):693–704. doi: 10.1056/NEJMoa2021436

    PubMed Abstract | CrossRef Full Text | Google Scholar

    22. Bergquist M, Lindholm C, Strinnholm M, Hedenstierna G, Rylander C. Impairment of Neutrophilic Glucocorticoid Receptor Function in Patients Treated With Steroids for Septic Shock. Intensive Care Med Exp (2015) 3(1):59. doi: 10.1186/s40635-015-0059-9

    PubMed Abstract | CrossRef Full Text | Google Scholar

    23. Cao W, Li T. COVID-19: Towards Understanding of Pathogenesis. Cell Res (2020) 30(5):367–9. doi: 10.1038/s41422-020-0327-4

    PubMed Abstract | CrossRef Full Text | Google Scholar

    24. Tzilas V, Manali E, Papiris S, Bouros D. Intravenous Immunoglobulin for the Treatment of COVID-19: A Promising Tool. Respiration (2020) 99(12):1087–9. doi: 10.1159/000512727

    PubMed Abstract | CrossRef Full Text | Google Scholar

    25. Mohtadi N, Ghaysouri A, Shirazi S, Sara A, Shafiee E, Bastani E, et al. Recovery of Severely Ill COVID-19 Patients by Intravenous Immunoglobulin (IVIG) Treatment: A Case Series. Virology (2020) 548:1–5. doi: 10.1016/j.virol.2020.05.006

    PubMed Abstract | CrossRef Full Text | Google Scholar

    26. Schwab I, Nimmerjahn F. Intravenous Immunoglobulin Therapy: How Does IgG Modulate the Immune System? Nat Rev Immunol (2013) 13(3):176–89. doi: 10.1038/nri3401

    PubMed Abstract | CrossRef Full Text | Google Scholar

    27. Cao W, Liu X, Hong K, Ma Z, Zhang Y, Lin L, et al. High-Dose Intravenous Immunoglobulin in Severe Coronavirus Disease 2019: A Multicenter Retrospective Study in China. Front Immunol (2021) 12:627844. doi: 10.3389/fimmu.2021.627844

    PubMed Abstract | CrossRef Full Text | Google Scholar

    28. Bongomin F, Asio LG, Ssebambulidde K, Baluku JB. Adjunctive Intravenous Immunoglobulins (IVIg) for Moderate-Severe COVID-19: Emerging Therapeutic Roles. Curr Med Res Opin (2021) 37(6):903–5. doi: 10.1080/03007995.2021.1903849

    PubMed Abstract | CrossRef Full Text | Google Scholar

    29. Gharebaghi N, Nejadrahim R, Mousavi SJ, Sadat-Ebrahimi SR, Hajizadeh R. The Use of Intravenous Immunoglobulin Gamma for the Treatment of Severe Coronavirus Disease 2019: A Randomized Placebo-Controlled Double-Blind Clinical Trial. BMC Infect Dis (2020) 20(1):786. doi: 10.1186/s12879-020-05507-4

    PubMed Abstract | CrossRef Full Text | Google Scholar

    30. Ali S, Uddin SM, Shalim E, Sayeed MA, Anjum F, Saleem F, et al. Hyperimmune Anti-COVID-19 IVIG (C-IVIG) Treatment in Severe and Critical COVID-19 Patients: A Phase I/II Randomized Control Trial. EClinicalMedicine (2021) 36:100926. doi: 10.1016/j.eclinm.2021.100926

    PubMed Abstract | CrossRef Full Text | Google Scholar

    31. Xiang HR, Cheng X, Li Y, Luo WW, Zhang QZ, Peng WX. Efficacy of IVIG (Intravenous Immunoglobulin) for Corona Virus Disease 2019 (COVID-19): A Meta-Analysis. Int Immunopharmacol (2021) 96:107732. doi: 10.1016/j.intimp.2021.107732

    PubMed Abstract | CrossRef Full Text | Google Scholar

    32. Polizzotto MN, Nordwall J, Babiker AG, Phillips A, Vock DM, Eriobu N, et al. Hyperimmune Immunoglobulin for Hospitalised Patients With COVID-19 (ITAC): A Double-Blind, Placebo-Controlled, Phase 3, Randomised Trial. Lancet (2022) 399(10324):530–40. doi: 10.1016/S0140-6736(22)00101-5

    PubMed Abstract | CrossRef Full Text | Google Scholar

     

    Keywords: Cushing’s syndrome, COVID-19, IVIg, hypercortisolism, immunomodulation, immunosuppression

    Citation: Popovic B, Radovanovic Spurnic A, Velickovic J, Plavsic A, Jecmenica-Lukic M, Glisic T, Ilic D, Jeremic D, Vratonjic J, Samardzic V, Gluvic Z and Adzic-Vukicevic T (2022) Successful Immunomodulatory Treatment of COVID-19 in a Patient With Severe ACTH-Dependent Cushing’s Syndrome: A Case Report and Review of Literature. Front. Endocrinol. 13:889928. doi: 10.3389/fendo.2022.889928

    Received: 04 March 2022; Accepted: 17 May 2022;
    Published: 22 June 2022.

    Edited by:

    Giuseppe Reimondo, University of Turin, Italy

    Reviewed by:

    Nora Maria Elvira Albiger, Veneto Institute of Oncology (IRCCS), Italy
    Miguel Debono, Royal Hallamshire Hospital, United Kingdom

    Copyright © 2022 Popovic, Radovanovic Spurnic, Velickovic, Plavsic, Jecmenica-Lukic, Glisic, Ilic, Jeremic, Vratonjic, Samardzic, Gluvic and Adzic-Vukicevic. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

    *Correspondence: Bojana Popovic, popbojana@gmail.com

     

    Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.

     

    From https://www.frontiersin.org/articles/10.3389/fendo.2022.889928/full

    • Like 1
  6. Abstract

    Cushing's syndrome (CS) shows diverse signs such as centripetal obesity, moon face, and buffalo hump, which can complicate the diagnosis. Facial features including eyelid edema, as an underrecognized sign, can be diagnostic clues for an excess of corticoids in a CS patient.

     

    A 49-year-old woman presented with amenorrhea and weight gain that had continued for 2 years. Her medical history was dyslipidemia, hypertension, and osteoporosis. Physical examination revealed eyelid edemas (Figure 1A), moon face, buffalo hump, abdominal purple striae, and centripetal obesity (body mass index (BMI), 30.8 kg/m2). Basal plasma adrenocorticotropin was undetectable and serum cortisol level was high (16.9 μg/dl) without circadian rhythms. Free cortisol level in a 24-h urine collection was elevated (158.7 μg/day). Overnight administration of dexamethasone (1 mg) did not reduce serum cortisol level (17.4 μg/dl). Magnetic resonance imaging suggested bilateral adenomas. We made a diagnosis of adrenal Cushing's syndrome (CS). Since 131l-adosterol scintigraphy showed specific uptake in the left adrenal gland, left adrenalectomy was laparoscopically performed. Histopathology of the tumor was compatible with adrenocortical adenoma. Three months after surgery, her BMI decreased to 25.0 kg/m2 and eyelid edemas were ameliorated (Figure 1B).

     

    Details are in the caption following the image

    (A) Bilateral eyelid edemas due to Cushing's syndrome are shown. (B) These findings were improved three months after surgery for left adrenal adenomas

    Eyelid edema, in addition to centripetal obesity, moon face, and buffalo hump, is also a significant sign of CS; however, it has scarcely been reported in countries other than Japan.1, 2 Increased capillary permeability, insufficient venous return due to muscle atrophy, and sodium retention due to mineralocorticoid actions conceivably cause edema in CS.

    AUTHORS’ CONTRIBUTIONS

    KY wrote the first draft and managed all the submission processes. KO and KH contributed to the clinical management of the patient. FO organized the writing the manuscript.

    ACKNOWLEDGMENT

    None.

      CONFLICT OF INTEREST

      The authors declare no conflicts of interest.

      ETHICAL APPROVAL

      Written informed consent was obtained from the patient to publish this case report.

      • 1Lacroix A, Feelders RA, Stratakis CA, Nieman LK. Cushing's syndrome. Lancet. 2015; 386: 913- 927.
      • 2Komiya I, Takasu N, Ohara N, et al. Forty-one cases of Cushing's syndrome: a comparison between Cushing's syndrome (adrenal adenoma) and Cushing's disease (adrenal hyperplasia). Nihon Naibunpi Gakkai Zasshi. 1992; 68: 607- 622.

      https://doi.org/10.1002/ccr3.5940

      From https://onlinelibrary.wiley.com/doi/10.1002/ccr3.5940

       

      • Like 1
    1. Abstract

      Background

      Endoscopic endonasal surgery is the main transsphenoidal approach for pituitary surgery in many centers, however few studies compare the endoscopic and microscopic surgical approach with regard to long-term follow-up. This single-center study aimed to compare the two techniques over 15 years.

      Methods

      Medical records and magnetic resonance images from 40 patients with primary transsphenoidal surgery for Cushing’s disease at Sahlgrenska University Hospital between 2003 and 2018 were reviewed. Fourteen patients who underwent microscopic surgery and 26 patients who underwent endoscopic surgery were included in this study.

      Results

      In the microscopic group, 12 of 14 patients achieved endocrine remission, compared to 19 of 26 patients in the endoscopic group (n. s.). Three patients in each group developed a late recurrence. Complications were seen in 5 patients in the microscopic group and in 8 patients in the endoscopic group (n. s.). No serious complications, such as carotid artery damage, cerebrovascular fluid leakage, epistaxis, or meningitis, occurred in any group. The postoperative hospital stay was shorter in the endoscopic than the microscopic group.

      Conclusion

      Endoscopic endonasal surgery for Cushing’s disease showed no difference in remission, recurrence, and complication rates compared to the microscopic approach. The endoscopic group had a shorter postoperative hospital stay than the microscopic group, which in part may be due to the minimal invasiveness of the endoscopic approach.

       
       
       
       

      References (0)

       

      Cited by (0)

       
       
       

      Conflicts of interest

       

      The authors have no conflicts of interest.

       

      Author statements

       

      Conceptualization: D. Farahmand, E. Backlund, O. Ragnarsson and P. Trimpou

       

      Data curation: Dan Farahmand, Erica Backlund, J. Carlqvist, T. Skoglund, T. Hallén, O. Ragnarsson, P. Trimpou.

       

      Formal Analysis: D. Farahmand, E. Backlund

       

      Funding acquisition: D. Farahmand

       

      Investigation: D. Farahmand, E. Backlund, O. Ragnarsson and P. Trimpou

       

      Methodology: D. Farahmand, E. Backlund, O. Ragnarsson and P. Trimpou

       

      Project administration: D. Farahmand, E. Backlund, O. Ragnarsson and P. Trimpou

       

      Supervision: D. Farahmand

       

      Writing – original draft: Penelope Trimpou

       

      Writing – review & editing: E. Backlund, O. Ragnarsson, T. Skoglund, T. Hallén, G. Gudnadottir, J. Carlqvist and D. Farahmand.

      View full text

      From https://www.sciencedirect.com/science/article/abs/pii/S1878875022009640

    2. Background

      Cushing’s disease (CD) is among the most common etiologies of hypercortisolism. Magnetic resonance imaging (MRI) is often utilized in the diagnosis of CD, however, up to 64% of adrenocorticotropic hormone (ACTH)-producing pituitary microadenomas are undetectable on MRI. We report 15 cases of MRI negative CD who underwent surgical resection utilizing a purely endoscopic endonasal approach.

      Methods

      Endoscopic endonasal transsphenoidal surgery (EETS) was performed on 134 CD cases by a single surgeon. Fifteen cases met inclusion criteria: no conclusive MRI studies and no previous surgical treatment. Data collected included signs/symptoms, pre- and post-operative hormone levels, and complications resulting from surgical or medical management. Data regarding tumor diameter, location, and tumor residue/recurrence was obtained from both pre- and post-operative MRI. Immunohistochemistry was performed to assess for tumor hormone secretion.

      Results

      Aside from a statistically significant difference (P = 0.001) in histopathological results between patients with negative and positive MRI, there were no statistically significant difference between these two groups in any other demographic or clinical data point. Inferior petrosal sinus sampling (IPSS) with desmopressin (DDAVP®) administration was performed on the 15 patients with inconclusive MRIs to identify the origin of ACTH hypersecretion via a central/peripheral (C/P) ratio. IPSS in seven, five and three patients showed right, left, and central side lateralization, respectively. With a mean follow-up of 5.5 years, among MRI-negative patients, 14 (93%) and 12 patients (80%) achieved early and long-term remission, respectively. In the MRI-positive cohort, over a mean follow-up of 4.8 years, 113 patients (94.9%) and 102 patients (85.7%) achieved initial and long-term remission, respectively.

      Conclusions

      Surgical management of MRI-negative/inconclusive Cushing’s disease is challenging scenario requiring a multidisciplinary approach. An experienced neurosurgeon, in collaboration with a dedicated endocrinologist, should identify the most likely location of the adenoma utilizing IPSS findings, followed by careful surgical exploration of the pituitary to identify the adenoma.

      Peer Review reports

      Introduction

      Cushing’s disease (CD) is the most common cause of hypercortisolism [1]. Left untreated, CD can result in multiple complications, most often cardiovascular disease or infection, and has a mortality rate 1.7–4.8-times higher than the general population [2,3,4]. Although MRI is the imaging modality of choice for identifying these tumors, imaging is often inconclusive [5].

      Prior studies have shown that adrenocorticotropic hormone (ACTH)-producing pituitary microadenomas are undetectable on MRI in 36–64% of cases [5]. However, the development and widespread utilization of 3-T MRI (3TMRI) has led to much higher tumor detection rates [6, 7]. With a negative predictive value of approximately 19–94% and variable sensitivity and specificity, anywhere from 4 to 54% of MRIs are incorrectly reported, especially in the setting of ACTH-secreting pituitary adenomas [8, 9]. With such variation in radiographic appearance, reliance on imaging for the management of CD patients can cause significant uncertainty for neurosurgeons and endocrinologists alike.

      The choice approach in the surgical management of these adenomas is via an endoscopic endonasal transsphenoidal surgery (EETS) [2, 10, 11], resulting in overall post-operative remission rates of 64–93% globally and 50–71% for cases without a conclusive MRI [12,13,14,15]. Inconclusive MRIs pose a significant challenge in the surgical management of CD, with the decision to pursue surgery for MRI-negative CD remaining highly controversial [8, 10, 14, 16]. In this study, we report 15 cases of CD without positive MRIs who underwent adenoma resection via EETS.

      Patients, materials and methods

      Patients population

      Between January 2005 and December 2018, EETS was performed in 134 CD cases by a single surgeon at Loghman hakim and Erfan hospitals. Of those patients, 15 cases met inclusion criteria: inconclusive MRI studies and no prior surgical treatment. The population consisted of 12 women (mean age 32.5 years; range 14–65 years) and 3 men (mean age 35 years; range 22–60 years). Data collected included signs/symptoms, pre- and post-operative hormone levels, and complications resulting from surgical or medical management. Data regarding tumor diameter, location, and tumor residue/recurrence was obtained from both pre- and post-operative MRIs. Immunohistochemistry was performed to assess for tumor hormone secretion.

      Ethics approval and consent to participate

      All procedures performed in this study involving human participants were in accordance with the ethical standards and approved by the Shahid Beheshti Medical University (SBMU) Ethical Committee and the 1964 Helsinki Declaration and its later amendments or comparable ethical standards. Also, a written informed consent was obtained from all subjects (or their parent or legal guardian in the case of children under 16).

      Imaging

      All patients underwent pre- and post-operative dynamic pituitary MRI via a superconducting 1.5-T scanner. Prior to gadolinium injection, T1-weighted Spin Echo (SE) and T2-weighted turbo SE images, followed by coronal dynamic acquisition (T1-weighted turbo SE), were obtained in the coronal plane using the following protocol: TR/TE, 400/20 ms; 288 · 192 matrix; two excitations; 18 · 18 cm field of view (FOV); 3 mm in thickness with 0.3-mm intersection gap. Afterwards, with simultaneous gadolinium injection, coronal and sagittal T1-weighted SE images were obtained 2 minutes following injection. All images were independently reviewed by both a radiologist and a neurosurgeon.

      MRIs studies were categorized into direct and indirect signs of CD. Direct signs consisted of any inhomogeneity found in the pituitary, such as a lesions with diminished enhancement. Indirect signs included pituitary stalk deviation and bulging or erosion of the sellar contour. MRI studies were considered negative (normal) if no direct or indirect signs were identified.

      In some cases, small lesions with diameters under 6 mm may be seen on MRI however are not considered indicative of CD due to the high prevalence of incidentalomas in this region. MRIs in which these lesions were present were classified as inconclusive.

      Any uncertainty in interpreting the MRIs by any of the reviewers resulted in exclusion of the image from this study.

      Pre-operative endocrine examination

      All cases were ACTH-dependent Cushing syndrome showing clinical features including weight gain, proximal myopathy, and wide base purple striae. Furthermore, all cases demonstrated laboratory abnormalities consistent with CD, including increased 24-hour urinary free cortisol (UFC) excretion, loss of the cortisol circadian rhythm, high basal ACTH level, failure of low-dose dexamethasone to suppress cortisol secretion in addition to serum suppression or 24-hour UFC after high-dose dexamethasone. Additionally, pre- and post-operative levels of anterior pituitary hormone including prolactin, growth hormone (GH), insulin-like growth factor I (IGF-I), thyroid stimulating hormone (TSH), free/total Triiodothyronine (T3)/ Thyroxine (T4), follicle-stimulating hormone (FSH), Luteinizing hormone (LH), and free/total testosterone (men) or estradiol (premenopausal women) were measured.

      The 15 cases of MRI negative CD were diagnosed and categorized according to their endocrine profile in order to distinguish the ACTH-dependent CD from pseudo-cushing syndrome.

      Bilateral inferior petrosal sinus sampling (BIPSS)

      All 15 cases of MRI-negative ACTH-dependent Cushing’s syndrome underwent bilateral inferior petrosal sinus sampling (BIPSS). To confirm that the elevated ACTH secretion originated from the pituitary, BIPSS was simultaneously performed with central/peripheral (C/P) ACTH gradient measurement, utilizing the calculations described by Oldfield et al. [17].

      No significant complications occurred in performing the procedures. A petrosal to peripheral ACTH ratio ≥ 2.0 in the basal state, a peak ratio ≥ 3.0 after desmopressin (DDAVP®) administration, or a normalized IPS:P ratio > 0.8 were considered diagnostic of CD. Additionally, tumor lateralization was specified when the interpetrosal gradient ratio of ACTH was ≥1.4 [18].

      Endoscopic Endonasal Transsphenoidal surgical approach

      All patients underwent surgery by a single neurosurgeon and otolaryngologist (ENT) with extensive experience in pituitary tumor excision via EETS. Exposure to the sellar floor was provided by an ENT surgeon while drilling of the sella was performed by the neurosurgeon. Extensive drilling of the sellar floor laterally up to the carotid artery bilaterally provided a wide view of the medial wall of the cavernous sinus as well as exposure of the anterior and posterior intercavernous sinuses was performed in all cases. The dura was then opened to expose the pituitary gland. Following tumor identification, adenomectomy was performed with selective removal of a rim of normal pituitary tissue. In cases where a tumor was not visualized on initial exposure of the pituitary, the pituitary gland was explored laterally via a horizontal paramedian incision on the IPSS suggesting side. If a tumor was not visualized at this stage, a vertical paramedian incision was then performed. In some cases, a cream-like substance was drained from the pituitary incision. Although this was suspicious of a tumor and tissue biopsy was obtained, it was not considered a definite tumor diagnosis and thus surgical exploration (EXP) was done in the same manner on the other side of the pituitary. In the scenario where no distinct adenoma was found, both sides of the pituitary gland underwent EXP with emphasis on lateralizing sides distinguished by IPSS. However, we did not rely solely on IPSS lateralization, as whole gland EXP was performed in all cases. Although ACTH secreting pituitary adenomas are the most common cause of Cushing syndrome, pituitary adenomas can also be ectopic, forming outside of the sella turcica with no direct connection to the pituitary gland [19]. After EXP of each side of the gland, ipsilateral periglandular inspection with visualization of the medial wall of the cavernous sinus and diaphragm was performed to identify a potential ectopic microadenoma in the periglandular region. Although the exact origin of ectopic ACTH-producing pituitary adenomas is unclear, they likely emerge from remnants of Rathke’s pouch during its development course [20]. As a result, these tumors can be discovered in the nasopharynx, sphenoid sinus, cavernous sinus, clivus, or suprasellar area [21]. Detecting an adenoma at this stage may prevent further unnecessary EXP of pituitary gland. If a visible tumor was still not detected, a vertical medial incision was made on the pituitary gland adjacent to the pituitary stalk and neurohypophysis. If a tumor could not be reliably identified by extensive EXP of the entire pituitary gland or BIPSS failed to localize a pituitary adenoma, we did not progress to performing incomplete or complete hypophysectomy. Figures 1 and 2, respectively, demonstrate the surgical management algorithm and pituitary incisions for MRI-negative CD.

      Fig. 1
       

      figure 1

      Eight-step MRI negative Cushing’s disease surgical management

      Fig. 2
       

      figure 2

      Schematic illustration of 8 steps in endoscopic endonasal approach to MRI inconclusive Cushing’s disease (Resembling half Georgia flag)

      If an ectopic ACTH-secreting adenoma is not easily found, permanent destructive or ablative surgeries such as bilateral adrenalectomy and hypophysectomy may be required [20]. Despite the danger of Nelson syndrome, bilateral adrenalectomy remains a feasible option in the management of refractory CD [22, 23].

      Histological examination

      All intraoperative tissue specimens obtained underwent histological examination by a pathologist. Pituitary specimens were fixed in buffered 10% formalin and embedded in paraffin wax. All specimens were first examined by Hematoxylin and Eosin (H&E) staining to detect regions which had loss of acinar organization. Additionally, reticulin and periodic Acid-Schiff (PAS) staining was implemented for a more accurate histopathologic diagnosis. Immunohistochemistry staining was used to identify cytokeratin and anterior pituitary hormones, including ACTH, in the case of a pituitary adenoma not being detected by H&E staining. The presence of ACTH-secreting cells was examined via immunocytochemistry using specific anti-ACTH antibodies.

      Post-operative endocrinologic assessment and follow up

      Serum cortisol and ACTH levels were monitored for 2–5 days following surgery. Initial follow-up occurred 2 weeks post-operatively with a subsequent visit occurring 3 months postoperatively, during each visit a complete pituitary hormonal evaluation was performed. This evaluation was repeated every 3 months for up to 2 years and every 6 months after that. An initial postoperative pituitary MRI was typically performed within 3 months after surgery. For patients to be considered to be in initial post-operation remission, a basal plasma cortisol level lower than 140 nmol/L (5 μg/dL) or adequate suppression of plasma cortisol (≤56 nmol/L) (≤1.8 μg/dL) following the 1-mg dexamethasone suppression test was necessary during the first month following surgery. Long term remission was defined as a plasma cortisol lower than 84 nmol/L (3 μg/dL) after a 1-mg dexamethasone suppression test at the final visit. Recurrence was defined as a recurring case of hypercortisolism with insufficient suppression of plasma cortisol (> 140 nmol/L) after a 1-mg dexamethasone suppression test. Clinical criteria for remission included significant symptomatic improvement or resolution without additional therapy (radiotherapy, adrenalectomy). Patients achieving remission had to meet both laboratory and clinical criteria to be classified as such. Glucocorticoids were not given postoperatively except when there was laboratory evidence of hypercortisolism and/or clinical manifestations of glucocorticoid insufficiency. Additionally, 4 to 6 weeks post-operatively, thyroid and gonadal axis function was assessed by measuring free T4, TSH, FSH, and LH levels in addition to end-organ hormones (estradiol in women and testosterone in men).

      Statistical analysis

      SPSS software (version 26, Chicago, IL) was used to analyze the data. For continuous data, we calculated descriptive statistics, mean and standard deviation (SD), and for categorical variables, frequency and percentages were calculated. The chi-square or Fisher’s exact test was used to analyze categorical data, while the student’s t-test or Mann- Whitney U test was used to analyze continuous variables’ means, depending on the distribution’s normality. Statistical significance was defined by a p value of < 0.05.

      Results

      Demographic and clinical data of 134 patients with CD who underwent EETS are shown in Table 1. Fifteen (11.2%) of the 134 CD patients who underwent EETS were MRI-negative and 119 patients (88.8%) were MRI positive. The female/male ratio in the MRI-negative group was four to one while this ratio in the MRI-positive cohort was 2.6. With regards to sex distribution, Fisher’s exact test found no statistically significant difference between these two groups (P = 0.565). All patients had clinical manifestation of Cushing’s syndrome including obesity, hirsutism, glucose intolerance, and hypertension. As shown in Table 1, pre-operative ACTH level was 134.02 ± 21.78 ng/l and 151.76 ± 44.17 ng/l in MRI-negative and MRI-positive patients, respectively, and no statistically significant difference was observed between these two groups (P = 0.781). As demonstrated in Table 1, UFC was 462.3 ± 43.98 μg/24 h and 478.4 ± 73.02 in MRI-negative and MRI-positive patients, respectively, and no statistically significant difference was observed between these two groups (P = 0.832).

      Table 1 Demographic and clinical data

      IPSS with DDAVP® administration was performed on the 15 MRI-negative patients to identify the origin of ACTH hypersecretion via the C/P ratio. Seven patients showed right-sided lateralization and five patients showed left-sided lateralization. In remaining three patients, IPSS did not show an ACTH interpetrosal gradient ratio greater than the cutoff point, which was interpreted as an ACTH hypersecretion with central origin. On EXP, adenomas were found in 2 of the 3 patients, with no adenoma being found in the 3rd. The IPSS results were in concordance with our observations during EXPs in 60% of patients. However, in 13% of patients, no adenoma was detected, and in 26% an adenoma was found on the opposite side of the pituitary where pre-operative IPSS results initially reported a tumor or was suggestive of one being present. In 60% of MRI-negative patients, histological examination demonstrated an adrenocorticotropic pituitary adenoma, but in 40% no adenoma was found after pathological examinations. In MRI-positive patients, positive histology was observed in 112 patients (94.1%), while in 7 patients (5.9%) histopathological studies were negative. Fisher’s exact test revealed that the difference between MRI-negative and MRI-positive patients in terms of histopathological result was statistically significant (P = 0.001).

      In all four patients who had discordant IPSS results as well as the patients who had negative or inconclusive findings on EXP, tissue samples were obtained from suspicious sites during EXP and were sent for histopathological examination. Histopathology demonstrated adrenocorticotropic adenoma tissues in 3 of them on the opposite side of the IPSS suggested region, while in 1 of them the histological results were inconclusive. This patient (case 10) achieved initial remission, however she experienced recurrence after 25 months, and similarly to her initial presentation, MRI findings were negative and IPSS suggested right sided lateralization. She underwent revision surgery, and a distinct adenoma was detected on the right side, which was confirmed by histological examination, after which she went into remission following selective adenectomy (Table 2).

      Table 2 Presents summary of patients’ demographics, IPSS and surgical exploration results

      Among the patients with inconclusive MRI, 14 (93%) achieved initial remission, 12 of which (80%) went on to long term remission with a mean follow up of 5.5 years. Two patients (cases 10 and 11) developed recurrence following initial remission; according to the IPSS suggested side, partial hypophysectomy was performed in both cases however neither was able to achieve remission afterwards. One patient (case 13) was unable to achieve initial remission following the initial surgery and thus required continued medical management. With a mean follow-up of 4.8 years among the 119 patients with positive MRI, 113 patients (94.9%) and 102 patients (85.7%) achieved initial and long-term remission, respectively. There were no statistically significant differences between these two groups in terms of either initial (P = 0.767) or long-term remission (P = 0.457). Among the 102 patients who achieved long-term remission, 12 patients (11.7%) experienced disease recurrence. With regards to recurrence rate, there was no statistically significant difference between patients with either positive or negative MRI (P = 0.542).

      In two patients (cases 2 and 6) the adenoma was not found during EXP, however tissue samples obtained from the IPSS suggested side demonstrated adrenocorticotropic pituitary adenoma in both patients on histopathological examination.

      Diabetes insipidus (DI) was the most frequent complication associated with CD. Transient DI occurred in seven cases with resolution prior to discharge. There was one case of permanent DI diagnosed in follow-up. Additionally, one patient developed symptomatic adrenal insufficiency requiring glucocorticoid replacement. Two patients developed hypothyroidism requiring hormone replacement. Panhypopituitarism was not seen following the initial surgeries however occurred in one case following revision surgery (partial hypophysectomy) which required hormone replacement therapy. Cerebrospinal fluid (CSF) leak resulting in meningitis was seen in one patient, however no other complications occurred during the post-operative period. None of our patients demonstrated clinical or endocrinological signs of gonadal insufficiency in follow-up aside from the aforementioned case of panhypopituitarism following revision partial hypophysectomy. In the MRI-positive cohort, 51 patients showed transients DI (42.8%), with 4 of the patients (3.4%) experiencing DI till last follow-up. Partial anterior pituitary insufficiency and complete anterior pituitary insufficiency was observed in one (0.8%) and two (1.6%) patients, respectively. Syndrome of inappropriate antidiuretic hormone (SIADH) secretion was observed in 3 patients (2.5%).

      Discussion

      In this study we present the outcomes of pure endoscopic endonasal surgical treatment of fifteen patients with MRI-negative Cushing’s disease. Due to the arduous nature of treatment in this patient population, we used a precise method of EXP as described above, resulting in initial remission in 93% of patients post-operatively. Based on the work of Bansal et al., patients with a definite adenoma on MRI who underwent microscopic transsphenoidal surgery had a statistically significant greater rate of early remission and lower rates of persistent disease than those with negative/equivocal findings [24]. However, in terms of late remission and recurrence, there was no statistically significant difference between these two groups [24]. Negative/equivocal MRI results and the incidence of macroadenoma, particularly in patients with cavernous sinus invasion, were found to predict poor remission rates [24]. According to some investigations, MRI-negative CD patients had a poorer remission rate [25, 26]. In other studies, however, there was no statistically significant difference in remission between those who had MRI-negative CD and those who had a MRI-positive CD, which is consistent with our result [14, 27,28,29,30,31,32]. Recurrence occurred in 2 patients, while 12 patients showed no clinical or endocrinological signs of recurrence during the mean follow-up of 5 years, and one patient did not go to remission. Aside from one CSF leak leading to meningitis and one case of permanent DI, there were no major surgery related complications. Pituitary CD is a common and potentially lethal condition that, if left untreated, can lead to sequelae such as morbid obesity, hypertension, and diabetes mellitus. Diagnosis and treatment of CD is more challenging than other functional pituitary adenomas. Currently, trans-sphenoidal pituitary EXP is considered the standard of care for CD [33,34,35]. CD is typically diagnosed by endocrinologist through clinical symptoms, and supported by laboratory tests such as the 8 AM blood or saliva cortisol level, 24 hours urinary free cortisol level, low- and high-dose dexamethasone suppression tests, and the corticotropin-releasing hormone (CRH) stimulating test [36,37,38]. When ACTH-dependent CD is diagnosed, or clinical signs and symptoms are highly suggestive of it, MRI imaging of the pituitary is often the next step to identify the causative agent i.e., a pituitary adenoma. With regards to pituitary lesions, MRI is considered the most sensitive imaging modality, however reported sensitivity varies significantly between studies, with reported rates ranging from 22 to 92% [39,40,41].

      The rate of MRI-negative microadenomas is reported to be between 36 to 63% [5]. Hofmann et al. reported no identified tumor in 49.3% of 270 MRIs [29]. Yamada et al. reported a lower frequency (17%) of MRI-negative CD in their series [42]. In our series, only 15 out of 134 (11.19%) CD patients were MRI-negative. In general, negative-MRIs could be explained by several factors such as field strength, technique (the correct pulse sequence and parameters), radiologist interpretation errors, or tumor size. Identifying tumors smaller than 3 mm in diameter is difficult in MRIs with 2.5- to 3-mm-thick image sections [29]. Dynamic MRI and 3-TMRI can result in a higher sensitivity in identifying ACTH-secreting microadenomas [6, 7, 43]. In addition, spoiled gradient-recalled echo sequence (SPGR) view can help to increase sensitivity [44]. The relatively low number of negative-MRIs in our study can be attributed to the more extensive review of MRI images, utilization of high-field strength MRI (1.5 T), as well as the implementation of SPGR dynamic studies with 1.5- to 2.0-mm-thick sections, in addition to standard methods. Additionally, assessment of images by experienced pituitary neuroradiologists may have reduced the negative-MRI rate in our series. Although small tumor size is a likely factor in MRI-negative CD, prior studies have reported examples of MRI-negative microadenomas 4-6 mm in size, typically large enough to be easily identified on EXP [42].

      If MRI is unable to identify the tumor definitively, the next best step is venous sampling to confirm CD. There are various indication for BIPSS, including patients who have clinical and laboratory findings of CD but normal or inconclusive MRI results [45], cases that do not have a clear hormone test response, or cases where there are inconsistencies between laboratory and imaging results [46]. BIPSS is also recommended by some as standard for any case of confirmed ACTH-dependent Cushing’s syndrome [47, 48]. In our institution, BIPSS is reserved for MRI-negative Cushing’s patients. Newell-Price et al. reviewed 21 studies with 569 total patients, and found that BIPSS with CRH stimulation had a 96% sensitivity and 100% specificity in separating CD from pseudo-Cushing’s states [49]. Most studies report a 90–100% sensitivity and specificity for BIPSS [50,51,52]. In the majority of cases of CD, a pituitary microadenoma can be found eccentric to one side of the pituitary, having venous drainage directly into the ipsilateral inferior petrosal sinus (IPS) [53].

      This phenomenon is the basis for utilizing BIPSS as a means of lateralizing ACTH secreting pituitary tumors. There are many instances where EXP fails to detect a pituitary adenoma, despite conformation of pituitary origin of ACTH secretion via BIPSS. Evidence of lateralization prior to surgery can convince the surgeon to perform a guided hemi hypophysectomy. In our series, the accuracy of BIPSS for lateralizing adenomas was 60%, similar to the reported accuracy in the literature of approximately 70% [17]. Inaccurate lateralization from BIPSS has been attributed to asymmetrical venous drainage with shunting of blood toward the dominant side. Thus, BIPSS appears to be a superior diagnostic tool compared to other means of lateralization, and neurosurgeons should be wary of making operative decisions solely from BIPSS data [49].

      The standard of care for MRI-negative CD is highly disputed. There is evidence suggesting surgical exploration is more problematic than watchful waiting [8], or that it is not indicated in MRI-negative CD [54]. Many advancements have led to the widespread adoption of transsphenoidal approach during the last three decades, especially the endoscope [31]. Regardless of the width or depth of access, the endoscopic approach allows the surgeon to have a large panoramic view. Many cases in the literature have reported successfully treating functional pituitary tumors via endoscopic surgery [27, 31, 55,56,57,58]. The results suggest that they are on par with, if not superior to, traditional microscopic approaches. When patients were operated on utilizing a microscopic technique assisted by a pre-operative ACTH gradient, the overall rate of partial adenomectomy (partial hypophysectomy) was 30%, including 19% in patients with positive MRIs and 40% in those with negative MRIs [28]. However, endoscopic visualization of pituitary adenomas has allowed for the need for partial adenomectomy to be reduced to less than 2%, limiting the damage to the normal pituitary gland during operation [28]. A recently published meta-analysis demonstrated that although there was no statistically significant differences between EETS and microscopic endonasal transsphenoidal surgery in the sub-analysis with regards to recurrence rate, remission rate, and persistence rate, the recurrence rate in the microscopic endonasal transsphenoidal surgery group was almost three times higher than in the EETS group [11]. As a result, EETS appears to be a possible suggested therapeutic method, while more studies are needed to establish the therapeutic method of choice [59].

      In general, pituitary surgery is not advisable in cases of MRI-negative CD where IPSS is not able to prove a central origin of ACTH secretion [42]. However, when IPSS demonstrates central ACTH secretion, surgical intervention has been proposed as a first line treatment in MRI-negative CD [25, 32, 42, 60]. The outcome of surgical intervention in MRI-negative patients is variable in the literature. Some reports indicated lower remission rate in these patients [42, 61], while others have concluded that EXP results in greater complications in this population [8, 15]. Additionally, several studies have shown no significant difference in outcomes of pituitary surgery between MRI-negative and MRI-positive patients [14, 25, 32]. Pivonello et al. found the lack of tumor detection on pre-operative MRI operation to be a negative prognostic factor in surgical management [62]. In the present study, surgery was performed for all MRI-negative Cushing’s patients with positive IPSS results. We achieved 93% initial remission and 80% long term remission rates, comparable to mean remission rates in patients with preoperative identification of tumor, as reported in the literature, ranging from 52.6–100% [62].

      Failure to identify an adenoma on EXP or in histologic examination is not uncommon in the surgical management of CD. Intraoperative detection of the adenoma has been shown to be a factor of favorable prognosis [63,64,65]. Similarly, failure to identify an adenoma on histopathology has been found to be a negative prognostic indicator. Specifically, remission rates were significantly lower in cases where no histological tumor identification could be provided [14, 63, 66]. In our study, two cases revealed no adenoma on EXP, however the tissue samples subsequently obtained from the IPSS suggesting side were consistent with pituitary adenoma on histologic examinations. In six cases, a cream-like substance was identified within the pituitary following incision, however histologic examination failed to demonstrate adrenocorticotropic adenoma in any of them. Nonetheless, 5 of the 6 patients went into remission following surgery, potentially due to the small size of tissue samples obtained which in turn made accurate histopathological assessment more difficult [14, 67].

      In cases where EXP does not result in localization of an adenoma, surgical decision making becomes complicated. Generally, total hypophysectomy is not advisable due to high rates of endocrine complications as well as failing to provide significantly increased remission rates over partial hypophysectomy [62, 68]. In this scenario, multiple studies have recommended partial hypophysectomy based on IPSS lateralization as the next best step in management [63, 69]. Carr et al. suggested the advantage of 2/3 gland resection on remission rate in MRI-negative CD [60], but as previously discussed, IPSS may incorrectly lateralize adenomas, and thus surgeons should be hesitant when making decisions regarding tumor lateralization based solely on BIPSS data [17, 49]. Moreover, both adenomectomy and hypophysectomy are not without risks and potential complications. Surgical aggressiveness is correlated with increased likelihood of pituitary loss-of-function, supported by literature showing that the larger the amount of resection, the higher the rate of hypopituitarism. It has been reported that patients undergoing adenomectomy, hemi-hypophysectomy, and-total hypophysectomy had mean rates of hypopituitarism of 6.6, 20.2, and 80.2%, respectively [63, 70, 71]. As most CD patients are females of reproductive age, preserving child-bearing capacity is an important consideration, one which results in reluctance to perform hemi-hypophysectomy. In our series, we performed selective adenectomy when distinct adenomas were found, and in the cases where no adenoma was detected, meticulous EXP of pituitary gland bilaterally was performed. Subsequently, if EXP was inconclusive, a vertical median incision was made near the pituitary stalk to explore central part of the gland, which is believed to be the nest for adrenocorticotropic cells. Although an important step in localizing the adenoma, this also likely explains the high rate of postoperative DI in our study. No additional hemi-hypophysectomy was performed during the initial surgery in our study. With this technique, we achieved acceptable results with regards to remission rates, and none of our patients experienced panhypopituitarism in postoperative follow-ups. In one patient where CD recurred 2 years post-operatively, inadequate bony exposure and limited visualization of the medial wall of the right cavernous sinus resulted in failure to identify the adenoma during the initial surgery, further supporting the strategy of creating extensive exposure of the operative field in MRI-negative CD. Another possible reason for recurrence in this patient would be growth of a previously undetected microadenoma.

      Conclusion

      Surgical treatment of MRI-negative Cushing’s disease is a demanding scenario necessitating multidisciplinary management. An experienced neurosurgeon working in collaboration with an endocrinologist should specify the most likely region of the tumor via IPSS. Additionally, surgical exploration of the pituitary is an invaluable tool in identifying adenomas while reducing the need for aggressive hypophysectomy, thus decreasing the likelihood of complications. Although MRI-negative Cushing’s disease presents significant challenges to neurosurgeons, surgical management remains essential in achieving remission.

      Availability of data and materials

      The authors confirm that the data supporting the findings of this study are available within the article.

      References

      1. Mehta GU, Lonser RR, Oldfield EH. The history of pituitary surgery for Cushing disease: historical vignette. J Neurosurg. 2012;116(2):261–8.

        PubMed Article Google Scholar 

      2. Hammer GD, Tyrrell JB, Lamborn KR, Applebury CB, Hannegan ET, Bell S, et al. Transsphenoidal microsurgery for Cushing’s disease: initial outcome and long-term results. J Clin Endocrinol Metabol. 2004;89(12):6348–57.

        CAS Article Google Scholar 

      3. Nieman LK. Cushing's syndrome: update on signs, symptoms and biochemical screening. Eur J Endocrinol. 2015;173(4):M33–8.

        CAS PubMed PubMed Central Article Google Scholar 

      4. Swearingen B, Biller BM, Barker FG, Katznelson L, Grinspoon S, Klibanski A, et al. Long-term mortality after transsphenoidal surgery for Cushing disease. Ann Intern Med. 1999;130(10):821–4.

        CAS PubMed Article Google Scholar 

      5. Lüdecke DK, Flitsch J, Knappe UJ, Saeger W. Cushing's disease: a surgical view; this publication is dedicated to our professor emeritus Dr. med. Rudolf Kautzky. J Neuro-Oncol. 2001;54(2):151–66.

        Article Google Scholar 

      6. Bartynski WS, Lin L. Dynamic and conventional spin-echo MR of pituitary microlesions. Am J Neuroradiol. 1997;18(5):965–72.

        CAS PubMed PubMed Central Google Scholar 

      7. Erickson D, Erickson B, Watson R, Patton A, Atkinson J, Meyer F, et al. 3 tesla magnetic resonance imaging with and without corticotropin releasing hormone stimulation for the detection of microadenomas in Cushing’s syndrome. Clin Endocrinol. 2010;72(6):793–9.

        CAS Article Google Scholar 

      8. Semple PL, Vance ML, Findling J, Laws ER Jr. Transsphenoidal surgery for Cushing's disease: outcome in patients with a normal magnetic resonance imaging scan. Neurosurgery. 2000;46(3):553–9.

        CAS PubMed Article Google Scholar 

      9. Tabarin A, Laurent F, Catargi B, Olivier-Puel F, Lescene R, Berge J, et al. Comparative evaluation of conventional and dynamic magnetic resonance imaging of the pituitary gland for the diagnosis of Cushing's disease. Clin Endocrinol. 1998;49(3):293–300.

        CAS Article Google Scholar 

      10. Nieman LK, Biller BM, Findling JW, Murad MH, Newell-Price J, Savage MO, et al. Treatment of Cushing's syndrome: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2015;100(8):2807–31.

        CAS PubMed PubMed Central Article Google Scholar 

      11. Sabahi M, Shahbazi T, Maroufi SF, Vidal K, Recinos PF, Kshettry VR, et al. MRI-negative Cushing's disease: a review on therapeutic management. World Neurosurg. 2022.

      12. Ciric I, Zhao J-C, Du H, Findling JW, Molitch ME, Weiss RE, et al. Transsphenoidal surgery for Cushing disease: experience with 136 patients. Neurosurgery. 2012;70(1):70–81.

        PubMed Article Google Scholar 

      13. Netea-Maier R, Van Lindert E, Den Heijer M, Van der Eerden A, Pieters G, Sweep C, et al. Transsphenoidal pituitary surgery via the endoscopic technique: results in 35 consecutive patients with Cushing’s disease. Eur J Endocrinol. 2006;154(5):675–84.

        CAS PubMed Article Google Scholar 

      14. Salenave S, Gatta B, Pecheur S, Fo S-G, Visot A, Lasjaunias P, et al. Pituitary magnetic resonance imaging findings do not influence surgical outcome in adrenocorticotropin-secreting microadenomas. J Clin Endocrinol Metab. 2004;89(7):3371–6.

        CAS PubMed Article Google Scholar 

      15. Semple PL, Laws ER. Complications in a contemporary series of patients who underwent transsphenoidal surgery for Cushing's disease. J Neurosurg. 1999;91(2):175–9.

        CAS PubMed Article Google Scholar 

      16. Lonser RR, Nieman L, Oldfield EH. Cushing's disease: pathobiology, diagnosis, and management. J Neurosurg. 2017;126(2):404–17.

        PubMed Article Google Scholar 

      17. EH OLDFIELD, ME GIRTON, JL DOPPMAN. Absence of intercavernous venous mixing: evidence supporting lateralization of pituitary microadenomas by venous sampling. J Clin Endocrinol Metab. 1985;61(4):644–7.

        Article Google Scholar 

      18. Mamelak AN, Dowd CF, Tyrrell JB, McDonald JF, Wilson CB. Venous angiography is needed to interpret inferior petrosal sinus and cavernous sinus sampling data for lateralizing adrenocorticotropin-secreting adenomas. J Clin Endocrinol Metab. 1996;81(2):475–81.

        CAS PubMed Google Scholar 

      19. Thompson LD, Seethala RR, Müller S. Ectopic sphenoid sinus pituitary adenoma (ESSPA) with normal anterior pituitary gland: a clinicopathologic and immunophenotypic study of 32 cases with a comprehensive review of the English literature. Head and neck pathology. 2012;6(1):75–100.

        PubMed PubMed Central Article Google Scholar 

      20. Seltzer J, Lucas J, Commins D, Lerner O, Lerner A, Carmichael JD, et al. Ectopic ACTH-secreting pituitary adenoma of the sphenoid sinus: case report of endoscopic endonasal resection and systematic review of the literature. Neurosurg Focus. 2015;38(2):E10.

        PubMed Article Google Scholar 

      21. Langford L, Batsakis JG. Pituitary gland involvement of the sinonasal tract. Ann Otology, Rhinology & Laryngology. 1995;104(2):167–9.

        CAS Article Google Scholar 

      22. Gurazada K, Ihuoma A, Galloway M, Dorward N, Wilhelm T, Khoo B, et al. Nasally located ectopic ACTH-secreting pituitary adenoma (EAPA) causing Nelson’s syndrome: diagnostic challenges. Pituitary. 2014;17(5):423–9.

        CAS PubMed Article Google Scholar 

      23. Pluta RM, Nieman L, Doppman JL, Watson JC, Tresser N, Katz DA, et al. Extrapituitary parasellar microadenoma in Cushing’s disease. J Clin Endocrinol Metabol. 1999;84(8):2912–23.

        CAS Google Scholar 

      24. Bansal P, Lila A, Goroshi M, Jadhav S, Lomte N, Thakkar K, et al. Duration of post-operative hypocortisolism predicts sustained remission after pituitary surgery for Cushing’s disease. Endocrine connections. 2017;6(8):625–36.

        CAS PubMed PubMed Central Article Google Scholar 

      25. Jagannathan J, Sheehan JP, Jane JA. Evaluation and management of Cushing syndrome in cases of negative sellar magnetic resonance imaging. Neurosurg Focus. 2007;23(3):1–7.

        Article Google Scholar 

      26. Pouratian N, Prevedello DM, Jagannathan J, Lopes MB, Vance ML, Laws ER Jr. Outcomes and management of patients with Cushing’s disease without pathological confirmation of tumor resection after transsphenoidal surgery. J Clin Endocrinol Metab. 2007;92(9):3383–8.

        CAS PubMed Article Google Scholar 

      27. Cebula H, Baussart B, Villa C, Assié G, Boulin A, Foubert L, et al. Efficacy of endoscopic endonasal transsphenoidal surgery for Cushing’s disease in 230 patients with positive and negative MRI. Acta Neurochir. 2017;159(7):1227–36.

        PubMed Article Google Scholar 

      28. Cristante J, Lefournier V, Sturm N, Passagia JG, Gauchez AS, Tahon F, et al. Why we should still treat by neurosurgery patients with Cushing disease and a normal or inconclusive pituitary MRI. J Clin Endocrinol Metab. 2019;104(9):4101–13.

        Article Google Scholar 

      29. Hofmann BM, Hlavac M, Martinez R, Buchfelder M, Müller OA, Fahlbusch R. Long-term results after microsurgery for Cushing disease: experience with 426 primary operations over 35 years. J Neurosurg. 2008;108(1):9–18.

        PubMed Article Google Scholar 

      30. Testa R, Albiger N, Occhi G, Sanguin F, Scanarini M, Berlucchi S, et al. The usefulness of combined biochemical tests in the diagnosis of Cushing’s disease with negative pituitary magnetic resonance imaging. Eur J Endocrinol. 2007;156(2):241–8.

        CAS PubMed Article Google Scholar 

      31. Starke RM, Reames DL, Chen C-J, Laws ER, Jane JA Jr. Endoscopic transsphenoidal surgery for Cushing disease: techniques, outcomes, and predictors of remission. Neurosurgery. 2013;72(2):240–7.

        PubMed Article Google Scholar 

      32. Sun Y, Sun Q, Fan C, Shen J, Zhao W, Guo Y, et al. Diagnosis and therapy for Cushing’s disease with negative dynamic MRI finding: a single-Centre experience. Clin Endocrinol. 2012;76(6):868–76.

        Article Google Scholar 

      33. Esposito F, Dusick JR, Cohan P, Moftakhar P, McArthur D, Wang C, et al. Early morning cortisol levels as a predictor of remission after transsphenoidal surgery for Cushing’s disease. J Clin Endocrinol Metab. 2006;91(1):7–13.

        CAS PubMed Article Google Scholar 

      34. Ram Z, Nieman LK, Cutler GB, Chrousos GP, Doppman JL, Oldfield EH. Early repeat surgery for persistent Cushing's disease. J Neurosurg. 1994;80(1):37–45.

        CAS PubMed Article Google Scholar 

      35. Rollin GAFS, Ferreira NP, Junges M, Gross JL, Czepielewski MA. Dynamics of serum cortisol levels after transsphenoidal surgery in a cohort of patients with Cushing’s disease. J Clin Endocrinol Metab. 2004;89(3):1131–9.

        CAS PubMed Article Google Scholar 

      36. Minami I, Tateno T, Yoshimoto T, Doi M, Izumiyama H, Akashi T, et al. Subclinical Cushings disease with amelioration of metabolic comorbidities after removal of pituitary tumor. Intern Med. 2006;45(21):1231–5.

        PubMed Article Google Scholar 

      37. Nakane T, Kuwayama A, Watanabe M, Takahashi T, Kato T, Ichihara K, et al. Long term results of transsphenoidal adenomectomy in patients with Cushing's disease. Neurosurgery. 1987;21(2):218–22.

        CAS PubMed Article Google Scholar 

      38. Buchfelder M, Nistor R, Fahlbusch R, Huk WJ. The accuracy of CT and MR evaluation of the Sella turcica for detection of adrenocorticotropic hormone-secreting adenomas in Cushing disease. Am J Neuroradiol. 1993;14(5):1183–90.

        CAS PubMed PubMed Central Google Scholar 

      39. Nieman LK. Medical therapy of Cushing's disease. Pituitary. 2002;5(2):77–82.

        CAS PubMed Article Google Scholar 

      40. Escourolle H, Abecassis J, Bertagna X, Guilhaume B, Pariente D, Derome P, et al. Comparison of computerized tomography and magnetic resonance imaging for the examination of the pituitary gland in patients with Cushing's disease. Clin Endocrinol. 1993;39(3):307–13.

        CAS Article Google Scholar 

      41. Findling JW, Doppman JL. Biochemical and radiologic diagnosis of Cushing’s syndrome. Endocrinol Metab Clin N Am. 1994;23(3):511–37.

        CAS Article Google Scholar 

      42. Yamada S, Fukuhara N, Nishioka H, Takeshita A, Inoshita N, Ito J, et al. Surgical management and outcomes in patients with Cushing disease with negative pituitary magnetic resonance imaging. World Neurosurg. 2012;77(3–4):525–32.

        PubMed Article Google Scholar 

      43. Portocarrero-Ortiz L, Bonifacio-Delgadillo D, Sotomayor-González A, Garcia-Marquez A, Lopez-Serna R. A modified protocol using half-dose gadolinium in dynamic 3-tesla magnetic resonance imaging for detection of ACTH-secreting pituitary tumors. Pituitary. 2010;13(3):230–5.

        CAS PubMed Article Google Scholar 

      44. Patronas N, Bulakbasi N, Stratakis CA, Lafferty A, Oldfield EH, Doppman J, et al. Spoiled gradient recalled acquisition in the steady state technique is superior to conventional postcontrast spin echo technique for magnetic resonance imaging detection of adrenocorticotropin-secreting pituitary tumors. J Clin Endocrinol Metabol. 2003;88(4):1565–9.

        CAS Article Google Scholar 

      45. Findling JW, Raff H. Cushing’s syndrome: important issues in diagnosis and management. J Clin Endocrinol Metab. 2006;91(10):3746–53.

        CAS PubMed Article Google Scholar 

      46. Arnaldi G, Angeli A, Atkinson A, Bertagna X, Cavagnini F, Chrousos G, et al. Diagnosis and complications of Cushing’s syndrome: a consensus statement. J Clin Endocrinol Metab. 2003;88(12):5593–602.

        CAS PubMed Article Google Scholar 

      47. McCANCE DR, McILRATH E, McNEILL A, DS GORDON, DR HADDEN, KENNEDY L, et al. Bilateral inferior petrosal sinus sampling as a routine procedure in ACTH-dependent Cushing's syndrome. Clin Endocrinol. 1989;30(2):157–66.

        CAS Article Google Scholar 

      48. JW FINDLING, ME KEHOE, JL SHAKER, RAFF H. Routine inferior petrosal sinus sampling in the differential diagnosis of adrenocorticotropin (ACTH)-dependent Cushing’s syndrome: early recognition of the occult ectopic ACTH syndrome. J Clin Endocrinol Metabol. 1991;73(2):408–13.

        Article Google Scholar 

      49. Newell-Price J, Trainer P, Besser M, Grossman A. The diagnosis and differential diagnosis of Cushing’s syndrome and pseudo-Cushing’s states. Endocr Rev. 1998;19(5):647–72.

        CAS PubMed Google Scholar 

      50. Machado MC, De Sa SV, Domenice S, Fragoso MCBV, Puglia P Jr, Pereira MAA, et al. The role of desmopressin in bilateral and simultaneous inferior petrosal sinus sampling for differential diagnosis of ACTH-dependent Cushing's syndrome. Clin Endocrinol. 2007;66(1):136–42.

        CAS Google Scholar 

      51. Hernandez I, Espinosa-de-los-Monteros AL, Mendoza V, Cheng S, Molina M, Sosa E, et al. Ectopic ACTH-secreting syndrome: a single center experience report with a high prevalence of occult tumor. Arch Med Res. 2006;37(8):976–80.

        CAS PubMed Article Google Scholar 

      52. Invitti C, Giraldi FP, Cavagnini F. Endocrinology SGotHPAAotISo: inferior petrosal sinus sampling in patients with Cushing's syndrome and contradictory responses to dynamic testing. Clin Endocrinol. 1999;51(2):255–7.

        CAS Article Google Scholar 

      53. Obuobie K, Davies J, Ogunko A, Scanlon M. Venous thrombo-embolism following inferior petrosal sinus sampling in Cushing’s disease. J Endocrinol Investig. 2000;s23(8):542–4.

        Article Google Scholar 

      54. Barrou Z, Abecassis J, Guilhaume B, Thomopoulos P, Bertagna X, Derome P, et al. Magnetic resonance imaging in Cushing disease. Prediction Surg Results. Presse medicale. 1997;26(1):7.

        CAS Google Scholar 

      55. Guaraldi F, Zoli M, Asioli S, Corona G, Gori D, Friso F, et al. Results and predictors of outcome of endoscopic endonasal surgery in Cushing’s disease: 20-year experience of an Italian referral pituitary center. J Endocrinol Investig. 2020;43(10):1463–71.

        CAS Article Google Scholar 

      56. Shin S, Gardner P, Ng J, Faraji A, Agarwal N, Chivukula S. Endoscopic endonasal approach for ACTH-secreting pituitary adenomas: outcomes and analysis of remission rates and tumor biochemical activity with respect to tumor invasiveness. World Neurosurg. 2015.

      57. Wagenmakers M, Boogaarts H, Roerink S, Timmers H, Stikkelbroeck N, Smit J, et al. Endoscopic transsphenoidal pituitary surgery: a good and safe primary treatment option for Cushing’s disease, even in case of macroadenomas or invasive adenomas. Eur J Endocrinol. 2013;169(3):329–37.

        CAS PubMed Article Google Scholar 

      58. Zhang K, Shen M, Qiao N, Chen Z, He W, Ma Z, et al. Surgical outcomes and multidisciplinary management strategy of Cushing’s disease: a single-center experience in China. Neurosurg Focus. 2020;48(6):E7.

        PubMed Article Google Scholar 

      59. Sabahi M, Shahbazi T, Maroufi SF, Vidal K, Recinos P, Kshettry V, et al. MRI-negative Cushing's disease: a systematic review and Meta-analysis. J Neurological Surg Part B: Skull Base. 2022;83(S 01):A169.

        Google Scholar 

      60. Carr SB, Kleinschmidt-DeMasters BK, Kerr JM, Kiseljak-Vassiliades K, Wierman ME, Lillehei KO. Negative surgical exploration in patients with Cushing’s disease: benefit of two-thirds gland resection on remission rate and a review of the literature. J Neurosurg. 2017;129(5):1260–7.

        Article Google Scholar 

      61. Bochicchio D, Losa M, Buchfelder M. Factors influencing the immediate and late outcome of Cushing's disease treated by transsphenoidal surgery: a retrospective study by the European Cushing's disease survey group. J Clin Endocrinology Metab. 1995;80(11):3114–20.

        CAS Google Scholar 

      62. Pivonello R, De Leo M, Cozzolino A, Colao A. The treatment of Cushing's disease. Endocr Rev. 2015;36(4):385–486.

        CAS PubMed PubMed Central Article Google Scholar 

      63. Prevedello DM, Pouratian N, Sherman J, Jane JA, Vance ML, Lopes MB, et al. Management of Cushing's disease: outcome in patients with microadenoma detected on pituitary magnetic resonance imaging. J Neurosurg. 2008;109(4):751–9.

        PubMed Article Google Scholar 

      64. Jagannathan J, Smith R, DeVroom HL, Vortmeyer AO, Stratakis CA, Nieman LK, et al. Outcome of using the histological pseudocapsule as a surgical capsule in Cushing disease. J Neurosurg. 2009;111(3):531–9.

        PubMed PubMed Central Article Google Scholar 

      65. Burke C, Adams C, Esiri M, Morris C, Bevan J. Transsphenoidal surgery for Cushing's disease: does what is removed determine the endocrine outcome? Clin Endocrinol. 1990;33(4):525–37.

        CAS Article Google Scholar 

      66. Acebes J, Martino J, Masuet C, Montanya E, Soler J. Early post-operative ACTH and cortisol as predictors of remission in Cushing’s disease. Acta Neurochir. 2007;149(5):471–9.

        CAS PubMed Article Google Scholar 

      67. Mampalam TJ, Tyrrell JB, Wilson CB. Transsphenoidal microsurgery for Cushing disease: a report of 216 cases. Ann Intern Med. 1988;109(6):487–93.

        CAS PubMed Article Google Scholar 

      68. Tyrrell JB, Brooks RM, Fitzgerald PA, Cofoid PB, Forsham PH, Wilson CB. Cushing's disease: selective trans-sphenoidal resection of pituitary microadenomas. N Engl J Med. 1978;298(14):753–8.

        CAS PubMed Article Google Scholar 

      69. Fomekong E, Maiter D, Grandin C, Raftopoulos C. Outcome of transsphenoidal surgery for Cushing's disease: a high remission rate in ACTH-secreting macroadenomas. Clin Neurol Neurosurg. 2009;111(5):442–9.

        PubMed Article Google Scholar 

      70. Rees D, Hanna F, Davies J, Mills R, Vafidis J, Scanlon M. Long-term follow-up results of transsphenoidal surgery for Cushing’s disease in a single Centre using strict criteria for remission. Clin Endocrinol. 2002;56(4):541–51.

        CAS Article Google Scholar 

      71. Trainer P, Lawrie H, Verhelst J, Howlett T, Lowe D, Grossman A, et al. Transsphenoidal resection in Cushing's disease: undetectable serum cortisol as the definition of successfuI treatment. Clin Endocrinol. 1993;38(1):73–8.

        CAS Article Google Scholar 

      Download references

      Acknowledgments

      We are grateful to all those who have helped us to accomplish and fulfil this project.

      Funding

      None.

      Author information

      Authors and Affiliations

      1. Department of Neurosurgery, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran

        Guive Sharifi, Amir Arsalan Amin & Seyed Ali Mousavinejad

      2. Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran

        Guive Sharifi, Amir Arsalan Amin, Nader Akbari Dilmaghani & Seyed Ali Mousavinejad

      3. Neurosurgery Research Group (NRG), Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran

        Mohammadmahdi Sabahi

      4. Department of Neurosurgery, Rutgers-New Jersey Medical School, Newark, NJ, USA

        Nikolas B. Echeverry

      5. Department of Otolaryngology, Head and Neck Surgery, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran

        Nader Akbari Dilmaghani

      6. Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran

        Majid Valizadeh

      7. Department of Endocrinology, Loghman Hakim Hospital, Shahid Beheshti Medical University, Tehran, Iran

        Zahra Davoudi

      8. Department of Neurological Surgery, Pauline Braathen Neurological Center, Cleveland Clinic Florida, Weston, Florida, USA

        Badih Adada & Hamid Borghei-Razavi

      9. Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Director of Minimally Invasive Cranial and Pituitary Surgery Program, Research Director of Neuroscience Institute, Cleveland Clinic Florida Region, 2950 Cleveland Clinic Blvd. Weston, Cleveland, FL, 33331, USA

        Hamid Borghei-Razavi

      Contributions

      Guive Sharifi, Mohammadmahdi Sabahi and Amirarsalan Amin have given substantial contributions to the conception and the design of the manuscript, Mohammadmahdi Sabahi, Nikolas B. Echeverry, Nader Akbari Dilmaghani, Ali Mousavi Nejad, and Zahra Davoudi to the acquisition, analysis, and interpretation of the data. All authors have participated in drafting the manuscript. Mohammadmahdi Sabahi, Majid Valizadeh, and Badih Adada revised it critically. Hamid Borghei-Razavi supervised this project. All authors read and approved the final version of the manuscript. All authors contributed equally to the manuscript and read and approved the final version of the manuscript.

      Corresponding author

      Correspondence to Hamid Borghei-Razavi.

      Ethics declarations

      Ethics approval and consent to participate

      All procedures performed in this study involving human participants were in accordance with the ethical standards and approved by the Shahid Beheshti Medical University (SBMU) Ethical Committee and the 1964 Helsinki Declaration and its later amendments or comparable ethical standards. Also, informed consent to participate in this study was obtained from participants included in the (or their parent or legal guardian in the case of children under 16).

      Consent for publication

      Not applicable.

      Competing interests

      All authors certify that they have no affiliations with or involvement in any organization or entity with any financial interest (such as honoraria; educational grants; participation in speakers’ bureaus; membership, employment, consultancies, stock ownership, or other equity interest; and expert testimony or patent-licensing arrangements), or non-financial interest (such as personal or professional relationships, affiliations, knowledge or beliefs) in the subject matter or materials discussed in this manuscript.

      Additional information

      Publisher’s Note

      Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

       

      Cite this article

      Sharifi, G., Amin, A.A., Sabahi, M. et al. MRI-negative Cushing’s Disease: Management Strategy and Outcomes in 15 Cases Utilizing a Pure Endoscopic Endonasal Approach. BMC Endocr Disord 22, 154 (2022). https://doi.org/10.1186/s12902-022-01069-5

      Download citation

      • Like 1
    3. Abstract

      Background

      Cushing’s disease (CD) is rare in pediatric patients. It is characterized by elevated plasma adrenocorticotropic hormone (ACTH) from pituitary adenomas, with damage to multiple systems and development. In recent years, genetic studies have shed light on the etiology and several mutations have been identified in patients with CD.

      Case presentation

      A girl presented at the age of 10 years and 9 months with facial plethora, hirsutism and acne. Her vision and eye movements were impaired. A quick weight gain and slow growth were also observed. Physical examination revealed central obesity, moon face, buffalo hump, supra-clavicular fat pads and bruising. Her plasma ACTH level ranged between 118 and 151 pg/ml, and sella enhanced MRI showed a giant pituitary tumor of 51.8 × 29.3 × 14.0 mm. Transsphenoidal pituitary debulk adenomectomy was performed and immunohistochemical staining confirmed an ACTH-secreting adenoma. Genetic analysis identified a novel germline GPR101 (p.G169R) and a somatic USP8 (p. S719del) mutation. They were hypothesized to impact tumor growth and function, respectively.

      Conclusions

      We reported a rare case of pediatric giant pituitary ACTH adenoma and pointed out that unusual concurrent mutations might contribute to its early onset and large volume.

      Peer Review reports

      Background

      Cushing’s disease (CD) is caused by the overproduction of adrenocorticotropic hormone (ATCH) by pituitary adenomas (PAs). It is rare in children and accounts for approximately 75% of pediatric Cushing’s syndrome from 7 to 17 years of age [1]. Weight gain and facial changes are more common in children than in adults [2]. Growth retardation is also a characteristic of children with hypercortisolemia [3]. Genetic alterations such as somatic USP8, RASD1, TP53 mutations, and germline AIP, MEN1, and CABLES1 mutations have been identified in CD patients [4]. Here we report a case of pediatric invasive pituitary ACTH macroadenoma associated with a novel germline GPR101 (p. G169R) and a somatic USP8 (p. S719del) mutation.

      Case presentation

      The girl was born at full term with a length of 48 cm and a weight of 2900 g. Her neuromotor and cognitive development was comparable to those of children of the same age. At the age of 9 years and 4 months she developed plethora, hirsutism, facial acne, rapid weight gain, and increased abdominal circumference. Her skin darkened, and purple striae appeared on thighs and in the armpits. She became dull and less talkative, as indicated by her parents. At 10 years and 3 months, the patient complained of pain around the left orbit with an intensity of 4–5 points on a numerical rating scale (NRS). Five months later bilateral blepharoptosis appeared, with significantly impaired vision of the left eye. Soon both eyes failed to rotate in all directions.

      On admission the patient was 10 years and 9 months, with a height of 144 cm (90–97th percentile) and a weight of 48 kg (25–50th percentile). Her weight gain was 20 kg, while the height increased by only 2–3 cm in 18 months. Her blood pressure was 115/76mmHg, and her heart rate was 80 bpm. Apart from the signs mentioned above, physical examination revealed central obesity (BMI 23.1 kg/m2), moon face, buffalo hump, supra-clavicular fat pads and bruising at the left fossa cubitalis. Her pupils were 7 mm in diameter and barely reacted to light. There was a fan-shaped visual field defect in the left eye. Her breasts were Tanner stage III and pubic hair was Tanner stage II, although menarche had not yet occurred. The parents and her younger brother at 6 years of age did not have symptoms related to Cushing syndrome, acromegaly or gigantism. There was no family history of pituitary tumor or other endocrine tumors.

      She had increased midnight serum cortisol (24.35 µg/dL, normal range < 1.8 µg/mL) and 24-hour urine free cortisol (24hUFC) (308.0 µg, normal range 12.3–103.5). The plasma ACTH level ranged from 118 to 151 pg/mL (< 46pg/mL). The 24hUFC was not suppressed (79.2 µg) after 48 h low-dose dexamethasone suppression test (LDDST), but suppressed to 32.8 µg (suppression rate 89.4%) after 48 h high-dose dexamethasone. Sella enhanced MRI showed a giant pituitary tumor measured 51.8 × 29.3 × 14.0 mm with heterogeneous density (Fig. 1). The mass compressed the optic chiasma and surrounded the bilateral cavernous sinus (Knosp 4). Therefore, an invasive giant pituitary ACTH adenoma was clinically diagnosed. The morning growth hormone (GH) was 1.0ng/ml (< 2 ng/ml) and insulin-like growth factor 1 416 ng/ml (88–452 ng/ml). The prolactin (PRL), luteinizing hormone (LH), follicle-stimulating hormone (FSH) and thyroid stimulating hormone (TSH) were all in normal ranges, as well as serum sodium, potassium, blood glucose and urine osmolality. Abdominal ultrasonography revealed a fatty liver. Tests concerning type 1 multiple endocrine neoplasia included serum calcium, phosphate, parathyroid hormone, gastrin and glucagon, which were all unremarkable (Table 1).

      Fig. 1
       

      figure 1

      Contrast-enhanced coronal (A) and sagittal (B) T1-weighted MRI on admission. The sellar mass measured 51.8 × 29.3 × 14.0 cm (TD × VD × APD) with a heterogeneous density in the enhanced scan. The diaphragma sellea was dramatically elevated, with optic chiasm compressed. The sellar floor was sunken and bilateral cavernous sinus was surrounded (Knosp 4)

      Table 1 Laboratory data on admission

      Transsphenoidal pituitary debulk adenomectomy was performed immediately due to multiple cranial nerve involvement and the negative results of Sandostatin loading test. A decompression resection was done. The plasma ACTH level declined to 77 pg/ml and serum cortisol 30.2 µg/dl three days after the operation. Vision, pupil dilation, eye movements and blepharoptosis also partially improved. Histopathology and immunohistochemical staining confirmed a densely–granulated corticotroph adenoma (Fig. 2, NanoZoomer S360 digital slide scanner and NDP.view 2.9.25 software, Hamamatsu, Japan). Neither necrosis nor mitotic activity was observed. The immunostaining for somatostatin receptor SSTR2A was positive with a cytoplasmic pattern, while GH, PRL, TSH, FSH, LH and PIT were all negative. The Ki 67 index was found to be 10%. One month after the operation the ACTH level increased to 132 pg/mL again, and the parents agreed to refer their child for radiotherapy to control the residual tumor.

      Fig. 2
       

      figure 2

      Histopathology and immunohistochemistry staining results of the pituitary tumor. By light microscopy, the tumor cells were mostly basophilic and arranged in papillary architecture. Neither necrosis nor mitotic activity was observed (A hematoxylin-eosin, ×200). Immunohistochemistry staining was positive for ACTH (B immunoperoxidase, ×200) and transcription factor T-PIT (C immunoperoxidase, ×200). Cytoplasmic staining of SSTR2A was observed in around 1/3 tumor cells besides the strong staining of endothelial cells (D immunoperoxidase, ×200). The Ki-67 index was 10% (E immunoperoxidase, ×200). Cytokeratin CAM5.2 was diffusely positive in the cytoplasm (F immunoperoxidase, ×200). The positive control for ACTH and T-PIT was the human anterior pituitary gland, and for SSRT2, Ki-67 and CAM5.2 were cerebral cortex, tonsil and colonic mucosa, respectively

      The early onset and invasive behavior of this tumor led to the consideration of whether there was a genetic defect. Genetic studies were recommended for the families and they all agreed and signed the written informed consent forms. Whole exome sequencing (WES) was performed on the patient’s blood sample using an Illumina HiSeq sequencer to an average read depth of at least 90 times per individual. Raw sequence files were mapped to the GRCH37 human reference genome and analyzed using the Sentieon software. The results revealed a germline heterozygous GPR101 gene mutation c.505G > C (p.Gly169Arg), which was subsequently confirmed to be of maternal origin by Sanger sequencing. Meanwhile WES of the tumor tissue identified an additional somatic heterozygous c.2155_2157delTCC (p.S719del) mutation of the USP8 gene .

      Discussion and conclusions

      In this report, we described an extremely giant and invasive pituitary ACTH adenoma in a 10-year-old girl. According to Trouillas et al., invasive and proliferative pituitary tumors have a poor prognosis [5]. CD is rare among children, and the fast-growing and invasive nature of the tumor in this case led to the investigation of genetic causes. The somatic USP8 gene mutation has been recently reported to be associated with the pathogenesis of CD [6, 7]. This gene encodes ubiquitin-specific protease 8 (USP8). S718, S719 and P720 are hotspots in different studies [6,7,8,9,10,11,12,13,14]. They are located at the 14-3-3 binding motif, and the mutations disrupt the binding between USP8 and 14-3-3 protein, which leads to increased deubiquitination and EGFR signaling. High levels of EGFR consequently trigger proopiomelanocortin (POMC) transcription and ACTH secretion [6, 7]. The p.S719del mutation has been previously reported and its pathogenicity has been confirmed [7]. Thus, we speculate the p.S719del mutation plays a role in this patient with CD.

      It is noteworthy that in our case, the pituitary corticotrophin adenoma was extremely giant and bilaterally invasive. USP8 mutations have been found in 31% of pediatric CD patients [10]. It is well known that microadenomas are most common in adult and pediatric CD patients. Previously, the Chinese and Japanese cohorts observed smaller sizes of USP8-mutated PAs than wild-type PAs [7, 9]. The Chinese cohort also reported a lower rate of invasive adenomas in USP8-mutated PAs [7]. This may be explained by the finding that UPS8 mutations did not significantly promote cell proliferation more than the wild-type ones [6]. Other cohorts suggested no difference in tumor size or invasiveness between USP8-mutated and wild-type PAs [8, 10, 12,13,14], which may be partially explained by the differences in sample sizes and ethnic backgrounds. Owing to the lack of evidence of USP8 mutations significantly contributing to tumor growth and invasiveness, additional pathogenesis should be investigated in this case.

      The p.Gly169Arg mutation of the GPR101 gene has not been reported in patients with pituitary tumors. In silico predictions were performed using Polyphen-2, Mutation Taster and PROVEAN, and all of the programs reported it to be pathogenic. The GPR101 gene encodes an orphan G protein-coupled receptor (GPCR) and microduplication encompassing the gene has been proven to be the cause of X-linked acrogigantism (XLAG) [15]. XLAG is characterized by the early onset of pituitary GH-secreting macroadenomas. Point mutations of GPR101 have been found in patients with PAs that are mostly GH-secreting [15,16,17]. Although their prevalence is very low, an in vitro study supported the pathogenic role of p.E308D, the most common mutation of GPR101. This led to increased cell proliferation and GH production in rat pituitary GH3 cells [15]. Rare cases of PRL, ACTH or TSH-secreting PAs with GPR101 variants were also documented [16, 18]. To date, there have been five cases of ACTH-secreting PAs with four different germline GPR101 mutations: two cases of p.E308D, p.I122T, p.T293I and p.G31S, although in silico predictions and in vitro evaluations using AtT-20 cells have respectively determined the latter two mutations to be non-pathogenic [16, 18]. These patients were mainly children and young adults. Unlike pituitary GH-secreting tumors, the role of GPR101 mutations in the pathophysiology of CD is still questionable. Trivellin et al. demonstrated no statistically significant difference in GPR101 expression between corticotropinomas and normal human pituitaries. No significant correlation between GPR101 and POMC expression levels was found neither [18].

      Given the evidences above, we hypothesize that the somatic USP8 mutation is responsible for the overexpression of ACTH in this CD girl while the germline GPR101 mutation contributes to the early onset and fast-growing nature of the tumor. Similarly, a 27-year-old woman with Nelson’s syndrome originally considered to be associated with a germline AIP variant (p.Arg304Gln) was recently reported to have a somatic USP8 mutation. The patient progressed rapidly and underwent multiple transsphenoidal surgeries [19]. Since germline AIP mutations are more commonly seen in GH-secreting PAs [20], the authors proposed that the USP8 mutation might have shifted the tumor towards ACTH-secreting [19]. Further investigations into the pathogenicity of GPR101 p.Gly169Arg and AIP p.Arg304Gln mutations are required to support the hypothesis.

      In summary, we report a novel germline GPR101 and somatic USP8 mutation in a girl with an extremely giant pituitary ACTH adenoma. The concurrent mutations may lead to the growth and function of the tumor, respectively. Further investigations should be carried out to verify the role of the concurrent mutations in the pathogenesis of pediatric CD.

      Availability of data and materials

      The WES data of the blood sample of the patient is available in the NGDC repository (https://ngdc.cncb.ac.cn/gsa-human/) and the accession number is HRA002396. Any additional information is available from the authors upon reasonable request.

      Abbreviations

      CD:

      Cushing’s disease

      ACTH:

      adrenocorticotropic hormone

      PA:

      pituitary adenoma

      NRS:

      numerical rating scale

      24hUFC:

      24-hour urine free cortisol

      LDDST:

      low-dose dexamethasone suppression test

      USP8:

      ubiquitin-specific protease 8

      POMC:

      proopiomelanocortin

      GPCR:

      G protein-coupled receptor

      XLAG:

      X-linked acrogigantism

      References

      1. Weber A, Trainer PJ, Grossman AB, Afshar F, Medbak S, Perry LA, et al. Investigation, management and therapeutic outcome in 12 cases of childhood and adolescent Cushing’s syndrome. Clin Endocrinol (Oxf). 1995;43(1):19–28.

        CAS Article Google Scholar 

      2. Storr HL, Alexandraki KI, Martin L, Isidori AM, Kaltsas GA, Monson JP, et al. Comparisons in the epidemiology, diagnostic features and cure rate by transsphenoidal surgery between paediatric and adult-onset Cushing’s disease. Eur J Endocrinol. 2011;164(5):667–74.

        CAS Article Google Scholar 

      3. Magiakou MA, Mastorakos G, Oldfield EH, Gomez MT, Doppman JL, Cutler GB Jr, et al. Cushing’s syndrome in children and adolescents. Presentation, diagnosis, and therapy. N Engl J Med. 1994;331(10):629–36.

        CAS Article Google Scholar 

      4. Hernández-Ramírez LC, Stratakis CA. Genetics of Cushing’s Syndrome. Endocrinol Metab Clin North Am. 2018;47(2):275–97.

        Article Google Scholar 

      5. Trouillas J, Roy P, Sturm N, Dantony E, Cortet-Rudelli C, Viennet G, et al. A new prognostic clinicopathological classification of pituitary adenomas: a multicentric case-control study of 410 patients with 8 years post-operative follow-up. Acta Neuropathol. 2013;126(1):123–35.

        Article Google Scholar 

      6. Reincke M, Sbiera S, Hayakawa A, Theodoropoulou M, Osswald A, Beuschlein F, et al. Mutations in the deubiquitinase gene USP8 cause Cushing’s disease. Nat Genet. 2015;47(1):31–8.

        CAS Article Google Scholar 

      7. Ma Z-Y, Song Z-J, Chen J-H, Wang Y-F, Li S-Q, Zhou L-F, et al. Recurrent gain-of-function USP8 mutations in Cushing’s disease. Cell Res. 2015;25(3):306–17.

        CAS Article Google Scholar 

      8. Perez-Rivas LG, Theodoropoulou M, Ferrau F, Nusser C, Kawaguchi K, Stratakis CA, et al. The Gene of the Ubiquitin-Specific Protease 8 Is Frequently Mutated in Adenomas Causing Cushing’s Disease. J Clin Endocrinol Metab. 2015;100(7):E997–1004.

        CAS Article Google Scholar 

      9. Hayashi K, Inoshita N, Kawaguchi K, Ibrahim Ardisasmita A, Suzuki H, Fukuhara N, et al. The USP8 mutational status may predict drug susceptibility in corticotroph adenomas of Cushing’s disease. Eur J Endocrinol. 2016;174(2):213–26.

        CAS Article Google Scholar 

      10. Faucz FR, Tirosh A, Tatsi C, Berthon A, Hernandez-Ramirez LC, Settas N, et al. Somatic USP8 Gene Mutations Are a Common Cause of Pediatric Cushing Disease. J Clin Endocrinol Metab. 2017;102(8):2836–43.

        Article Google Scholar 

      11. Albani A, Perez-Rivas LG, Dimopoulou C, Zopp S, Colon-Bolea P, Roeber S, et al. The USP8 mutational status may predict long-term remission in patients with Cushing’s disease. Clin Endocrinol (Oxf). 2018;89:454–8.

        CAS Article Google Scholar 

      12. Ballmann C, Thiel A, Korah HE, Reis AC, Saeger W, Stepanow S, et al. USP8 Mutations in Pituitary Cushing Adenomas-Targeted Analysis by Next-Generation Sequencing. J Endocr Soc. 2018;2(3):266–78.

        CAS Article Google Scholar 

      13. Losa M, Mortini P, Pagnano A, Detomas M, Cassarino MF, Pecori Giraldi F. Clinical characteristics and surgical outcome in USP8-mutated human adrenocorticotropic hormone-secreting pituitary adenomas. Endocrine. 2019;63(2):240–6.

        CAS Article Google Scholar 

      14. Weigand I, Knobloch L, Flitsch J, Saeger W, Monoranu CM, Hofner K, et al. Impact of USP8 Gene Mutations on Protein Deregulation in Cushing Disease. J Clin Endocrinol Metab. 2019;104(7):2535–46.

        Article Google Scholar 

      15. Trivellin G, Daly AF, Faucz FR, Yuan B, Rostomyan L, Larco DO, et al. Gigantism and acromegaly due to Xq26 microduplications and GPR101 mutation. N Engl J Med. 2014;371(25):2363–74.

        CAS Article Google Scholar 

      16. Lecoq AL, Bouligand J, Hage M, Cazabat L, Salenave S, Linglart A, et al. Very low frequency of germline GPR101 genetic variation and no biallelic defects with AIP in a large cohort of patients with sporadic pituitary adenomas. Eur J Endocrinol. 2016;174(4):523–30.

        CAS Article Google Scholar 

      17. Iacovazzo D, Caswell R, Bunce B, Jose S, Yuan B, Hernández-Ramírez LC, et al. Germline or somatic GPR101 duplication leads to X-linked acrogigantism: a clinico-pathological and genetic study. Acta Neuropathol Commun. 2016;4(1):56.

        Article Google Scholar 

      18. Trivellin G, Correa RR, Batsis M, Faucz FR, Chittiboina P, Bjelobaba I, et al. Screening for GPR101 defects in pediatric pituitary corticotropinomas. Endocr Relat Cancer. 2016;23(5):357–65.

        CAS Article Google Scholar 

      19. Perez-Rivas LG, Theodoropoulou M, Puar TH, Fazel J, Stieg MR, Ferrau F, et al. Somatic USP8 mutations are frequent events in corticotroph tumor progression causing Nelson’s tumor. Eur J Endocrinol. 2018;178(1):57–63.

        CAS Article Google Scholar 

      20. Tatsi C, Stratakis CA. The Genetics of Pituitary Adenomas. J Clin Med. 2019;9(1).

      Download references

      Acknowledgements

      We thanked Dr. Xiaohua Shi and Dr. Yu Xiao from the Department of Pathology, Peking Union Medical College Hospital for their expertise in pituitary pathology and critical help in accomplishment of our manuscript.

      Funding

      This research was supported by “The National Key Research and Development Program of China” (No. 2016YFC0901501), “CAMS Innovation Fund for Medical Science” (CAMS-2017-I2M–1–011). They mainly covered the fees for genetic analysis and publications.

      Author information

      Authors and Affiliations

      1. Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China

        Xu-dong Bao

      2. Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China

        Lin Lu, Hui-juan Zhu, Xiao Zhai, Yong Fu, Feng-ying Gong & Zhao-lin Lu

      3. Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China

        Yong Yao, Ming Feng & Ren-zhi Wang

      Contributions

      XB and LL contributed to the study design and manuscript writing. HZ and FG performed genetic analysis. XZ and YF collected the clinical data. YY, MF and RW provided the tumor tissue and histopathology data. ZL revised the manuscript. All authors have read and approved the final manuscript.

      Corresponding author

      Correspondence to Lin Lu.

      Ethics declarations

      Ethics approval and consent to participate

      This study was approved by the Ethics Committee of Peking Union Medical College Hospital. The parents of the patient provided written informed consent for research participation.

      Consent for publication

      The parents of the patient provided written informed consent for the publication of indirectly identifiable data in this research.

      Competing interests

      The authors declare that they have no competing interests.

      Additional information

      Publisher’s note

      Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

      • Like 1
    4. Recordati Rare Diseases, a US biopharma that forms part of the wider Italian group, has presented multiple positive data sets on Isturisa (osilodrostat) at the annual ENDO 2022 meeting in Atlanta, Georgia.

      Isturisa is a cortisol synthesis inhibitor indicated for the treatment of adult patients with Cushing’s disease for whom pituitary surgery is not an option or has not been curative.

      Among the data presented, the Phase III LINC 4 study demonstrated that Isturisa maintained normal mean urinary free cortisol long-term in patients with Cushing’s disease while the Phase III LINC 3 study found adrenal hormone levels changed during early treatment with the drug while stabilizing during long-term treatment.

      The ILLUSTRATE study also showed patients treated with a prolonged titration interval tended to have greater persistence with therapy.

      Mohamed Ladha, president and general manager for North America, Recordati Rare Diseases, said: “The data from these studies reinforces the efficacy and safety of Isturisa as a treatment for patients with Cushing’s disease.

      “We are pleased to share these data with the endocrine community and are excited to provide patients with a much-needed step forward in the management of this rare, debilitating, and potentially life-threatening condition.”

      Cushing’s disease is a rare, serious illness caused by a pituitary tumor that leads to overproduction of cortisol by the adrenal glands. Excess cortisol can contribute to an increased risk of morbidity and mortality. Treatment for the condition seeks to lower cortisol levels to a normal range.

      Isturisa, which was approved by the US Food and Drug Administration in March 2020, works by inhibiting 11-beta-hydroxylase, an enzyme responsible for the final step of cortisol biosynthesis in the adrenal gland.

      From https://www.thepharmaletter.com/article/results-reinforce-efficacy-of-recordati-s-isturisa-in-cushing-s-disease

      • Like 1
    5. MaryO'Note:  I found this article very simplistic.  What do you think?

       

      This article is for informational purposes only and is not a substitute for professional medical advice, diagnosis or treatment. Contact a qualified medical professional before engaging in any physical activity, or making any changes to your diet, medication or lifestyle.

      Imagine the heart-pounding rush of adrenaline you’d get while bungee jumping or zip lining — that’s what Angela Yawn felt all the time before receiving her diagnosis.

      In a span of six years, the 49-year-old gained 52 kg (115 lbs) and suffered from joint swelling, headaches, skin redness and a racing heart.

      “I would put my hand on my chest because it made me feel like that’s what I needed to do to hold my heart in,” Yawn, who lives in Griffin, U.S., told Today. “I noticed it during the day, but at night when I was trying to lie down and sleep, it was worse because I could do nothing but hear it beat, feel it thump.”

      Yawn recalled being the most frustrated with the weight gain, as she’d put on 1 kg (2 lbs) a day while only eating 600 calories. “I was going crazy,” she said.

      After dozens of doctors couldn’t piece together her seemingly unrelated symptoms, Yawn sought out the help of an endocrinologist in February 2021.

      Blood tests and an MRI confirmed that Yawn had a tumour in her pituitary gland — a small, pea-sized organ at the base of the brain — that caused the gland to release excess adrenocorticotropic hormones. As a result, she became inundated with cortisol, a steroid the body releases in response to danger or stress. This combination of factors led to her diagnosis — Cushing’s disease.

      Read on to learn more about Cushing’s disease, signs and symptoms as well as how it can be prevented.

      What is Cushing’s disease?

      “Cushing’s disease is a rare but serious condition that is caused by a pituitary tumour," a specialist from the University of California, Los Angeles (UCLA) pituitary team tells Yahoo Canada. "The gland releases excessive adrenocorticotropic hormones and cortisol into the blood over a long period of time. It’s a hormonal disorder that is sometimes called hypercortisolism, and you will need to see an endocrinologist or someone who specializes in hormonal-related diseases to confirm your diagnosis and to help you receive proper care.”

      Cushing’s disease is not the same as Cushing’s syndrome, which refers to elevated levels of cortisol in the blood and is much more common than Cushing’s disease. Unlike the disease, Cushing’s syndrome can be caused by taking medications that have cortisol such as prednisone, asthma inhalers and joint steroid injections.

      Who is at risk for Cushing’s disease?

      Cushing’s disease is incredibly rare, resulting in only 10 to 15 new cases per million people in the United States each year, according to UCLA Health.

      “It’s most commonly found in people between the ages of 20 and 50, and affects about three times more women than men,” the UCLA source, who asked not to be named, says. “However, you might be more at risk if you have high blood pressure, if you’re overweight or if you have type 2 diabetes.”

      What are the signs and symptoms of Cushing’s disease?

      Although each person may have a unique combination of symptoms, patients typically experience changes to their physical appearance, according to Mayo Clinic.

      “It’s very common to see rapid weight gain, red cheeks and bruising of the skin,” the UCLA source says. “I’ve also seen patients with generalized fatigue, depression, high blood pressure, a rapid heartbeat and loss of vision.”

      “The symptoms can seem random or unrelated, which is why it can be so hard to diagnose,” they add.

      To establish if you have the disease, your doctor will conduct a physical exam and ask you about your symptoms and medical history. Generally, the first step in diagnosing Cushing's disease is determining the state of excess cortisol in the blood. Afterwards, an MRI will determine if a pituitary tumour is visible.

      If you have symptoms of Cushing’s disease, you should make an appointment to see a doctor or endocrinologist.

      How is Cushing’s disease treated?

      In the last decade, treatment options have changed thanks to several breakthroughs in pituitary science.

      “Surgery to remove the tumour is normally the first treatment option. It’s minimally invasive, has a fairly high success rate and it’s the only long-term cure for Cushing’s disease at the moment,” explains the UCLA source.

      If surgery isn’t an option or doesn’t solve the problem, medication and radiation therapy are other ways to treat the disease.

      “No matter the stage of the disease at the time of diagnosis, treating it requires an experienced specialist or team of doctors familiar with pituitary tumours,” the UCLA source adds.

      How can I prevent Cushing’s disease?

      “There’s no tried and true method of preventing the condition,” the source explains. “But if you’re at risk or if you think you have the disease, I always recommend having a doctor monitor your cortisol levels on a regular basis.”

      The UCLA source also recommends implementing healthy lifestyle changes that can help prevent high blood pressure. Examples include reducing stress, getting adequate sleep, exercising regularly and eating a healthy diet that's rich in fruits, vegetables and whole grains.

      Adapted from https://ca.news.yahoo.com/what-is-cushings-disease-experts-warn-rare-serious-condition-120015725.html

      cushings-basics.png

      • Like 1
    6. Abstract

       

      Cushing's disease causes numerous metabolic disorders, cognitive decline, and sarcopenia, leading to deterioration of the general health in older individuals. Cushing's disease can be treated with transsphenoidal surgery, but thus far, surgery has often been avoided in older patients. We herein report an older woman with Cushing's disease whose cognitive impairment and sarcopenia improved after transsphenoidal surgery. Although cognitive impairment and sarcopenia in most older patients show resistance to treatment, our case indicates that normalization of the cortisol level by transsphenoidal surgery can be effective in improving the cognitive impairment and muscle mass loss caused by Cushing's disease.

       

       
       
      References (27)

      From https://www.jstage.jst.go.jp/article/internalmedicine/advpub/0/advpub_8326-21/_article

      • Like 1
    7. RareCare®

      Since 1987, NORD has provided assistance programs to help patients obtain life-saving or life-sustaining medication they could not otherwise afford. These programs provide medication, financial assistance with insurance premiums and co-pays, diagnostic testing assistance, and travel assistance for clinical trials or consultation with disease specialists.

      Read NORD’s Coronavirus: Business Continuity Response for RareCare® patient assistance programs.

      Medical Assistance
      Co-Pay Assistance
      Premium Assistance
      Contact: 1-855-864-4018
      Email: cushings@rarediseases.org
      Fax: 1-203-517-0978

       

      Other diseases:https://rarediseases.org/for-patients-and-families/help-access-medications/patient-assistance-programs-2

      • Like 1
    8. Abstract

      Cushing syndrome is a rare disease that rarely presents as acute psychosis. In this case, the patient presented with acute psychosis and agitation as the first manifestations of the disease which led to the admission of the patient to a psychiatry hospital for one month, as it was difficult to restrain her sufficiently for performing appropriate diagnostic tests due to disturbing behavior. She responded well to treatment with olanzapine and lorazepam to treat the patient’s agitation, and successfully complete her evaluation. Thereafter, she was diagnosed with a pituitary tumor and underwent pituitary lesion resection via a microscopic transsphenoidal as needed. Two months after surgery, her cortisol levels returned to baseline, and she became calmer and decreased the tensity of her psychosis; however, it was only five months after surgery that her psychotic symptoms and disturbed behavior ceased.

      Introduction

      Cushing syndrome is comprised of a group of symptoms induced by prolonged exposure to high blood cortisol levels [1]. It is a rare disease, occurring in approximately 2.4 per million individuals per year [2]. Psychiatric and cognitive manifestations of Cushing syndrome occur in 70%-85% of patients, with irritability, emotional lability, and depression occurring most commonly. Rarer symptoms include mania, panic attacks, anxiety, suicidal ideation, and acute psychosis [3-5]. In this article, we describe a patient with Cushing syndrome who developed psychosis with agitation as the first manifestation of Cushing syndrome. The patient was difficult to manage since her agitation and refusal to undergo evaluation prevented her from receiving outpatient care.

      Case Presentation

      A 22-year-old woman with a three-month history of an increase in appetite, binge eating, and weight gain. After two weeks of her initial symptoms, she started to have grandiose and persecutory delusions, auditory hallucinations, decreased need for sleep, agitation, irritability, and aggression for which she went to a private psychiatry clinic and was given 10 mg olanzapine oral at night. After a month of starting oral olanzapine, she was not improving and was admitted to the psychiatry ward for evaluation. During her admission period, she started to have cognitive symptoms including worsened memory, attention, and orientation. After one month of admission with no improvement on medication, she was noted to have moon face and high blood pressure, and her laboratory investigation showed mild hypokalemia, high cortisol level, and adrenocorticotropic hormone (ACTH), elevated liver enzymes, and mild hypertriglyceridemia. A magnetic resonance imaging (MRI) scan of the brain revealed a 6 × 2-mm hyperintense lesion in the anterior pituitary on a T2-weighted image; therefore, she was transferred to our hospital for further work up and management as we have the endocrine facility. She had no past psychiatric history or family history of psychiatric illnesses, nor a history of substance abuse. She also had no past medical history and was not on any medication prior to this presentation.

      The patient was admitted to the endocrine department to evaluate the possibility of Cushing syndrome. Her blood pressure (150/98), heart rate (128 BPM), and respiratory rate (30 BPM) were elevated. She was treated with losartan, amlodipine, and spironolactone. Basic labs were done (Table 1). Therefore, insulin therapy was initiated. The evaluation of the patient’s condition was difficult as she was aggressive and uncooperative due to a lack of insight. Her primary team planned for sedation with anesthesia to facilitate a clinical evaluation; however, no intensive care unit bed was available.

      Lab test Patient result Reference values
      cortisol levels 1549 nmol/L 140 to 690 nmol/L
      ACTH (Adrenocorticotropic Hormone) 54 pg/mL 10 to 50 pg/mL
      ALT (Alanine transaminase) 305 U/L 7 to 56 U/L
      AST (Aspartate aminotransferase) 112 U/L 8 to 33 U/L
      Alkaline phosphatase 141 IU/L 44 to 147 IU/L
      Hemoglobin A1c 7.3% 5.7% to 6.4%
      Table 1: Lab results for the patient when she first came to our hospital
       
       

      Psychiatry was consulted to manage agitation. We started her on 5 mg olanzapine oral twice daily, and 2 mg lorazepam three times daily intravenous when oral was not possible. Maximum dosage of 5 mg olanzapine and 2 mg lorazepam every four hours were administered as required to manage agitation. Her ECG showed a QTC of 464. One-to-one nurse observation was initiated to detect risky behaviors. The patient slept well and became calmer and more cooperative throughout evaluations when receiving medication. One-to-one nurse observation was discontinued after five days, and lorazepam administration was reduced to two times daily. She remained easily provoked with grandiose and persecutory delusions, auditory hallucinations, and confusion. As the patient calmed, the primary team continued clinical evaluations. A contrast-enhanced MRI showed a focal non-deforming and hypo-enhancing lesion, measuring 7 mm (AP) x 6 mm (TV) x 6 mm (CC), in the anterior pituitary (Figures 1, 2). A minimal leftward deviated pituitary stalk with normal thickness was also identified. An 8 mg dexamethasone suppression test revealed cortisol levels had decreased from 1,500 to 900 nmol/L. The 24-hour cortisol level was not determined, as the patient was easily provoked. Inferior petrosal sinus sampling was performed under general anesthesia. These results are consistent with central Cushing disease.

      Coronal-T1-weighted-MRI-of-the-pituitary-gland-with-contrast-showed-a-hypoenhancing-nodular-lesion-at-the-midline-of-the-anterior-pituitary,-with-mild-eccentric-to-the-right
      Figure 1: Coronal T1-weighted MRI of the pituitary gland with contrast showed a hypoenhancing nodular lesion at the midline of the anterior pituitary, with mild eccentric to the right
       
       
      Brain-MRI-sagittal-view-showing-focal-anterior-pituitary-hypoenhancing-lesion-at-the-midline-and-eccentric-to-the-right
      Figure 2: Brain MRI sagittal view showing focal anterior pituitary hypoenhancing lesion at the midline and eccentric to the right
       
       

      Treatment with 250 mg metyrapone twice daily was initiated and the patient was scheduled for pituitary lesion resection via a microscopic transsphenoidal approach by neurosurgery. Her blood tests began normalizing post-surgery except for low cortisol (Table 2), and her vital signs were within normal range. Medications regulating blood pressure and glucose levels were decreased to monotherapy and discontinued thereafter. And 40 and 20 mg doses of hydrocortisone administered in the morning and night, respectively, were tapered to 5 mg twice daily over a period of two months after the surgery, and cortisol levels were regulated reaching 167 nmol/L. Agitation and irritability, grandiose and persecutory delusion and auditory hallucination tensity were reduced, with intact cognitive and memory function. Therefore, medication dosages were gradually reduced, starting with lorazepam.

      Lab Test Patient result Reference values
      cortisol levels 68 nmol/L 140 to 690 nmol/L
      ACTH (Adrenocorticotropic Hormone) 25 pg/ml 10 to 50 pg/mL
      ALT (Alanine transaminase) 17.2 U/L 7 to 56 U/L
      AST (Aspartate aminotransferase) 19.2 U/L 8 to 33 U/L
      Alkaline phosphatase 121 IU/L 44 to 147 IU/L
      TSH (Thyroid Stimulating Hormone) 1.8 mIU/L 0.5 to 5.0 mIU/L
      Table 2: Lab results after the surgery.
       
       

      Before discharge, the patient’s psychotropic medications were withheld by the primary team for two days due to oversedation. Upon discharge, due to the side effects of olanzapine, the patient was switched to oral risperidone 1 mg at night, with 0.5 mg oral clonazepam twice daily as needed for agitation and psychosis. Throughout follow-up, the patient experienced ongoing psychosis with disturbed behavior even though she is using received clonazepam twice daily. Therefore, her dosage of risperidone was increased to 2 mg orally at night, and oral clonazepam (0.5 to 1 mg) was administered three times daily as needed to manage agitation. After three months of discharge (five months from surgical intervention), her levels of agitation and irritability decreased, delusions and auditory hallucinations ceased, and she returned to baseline, and clonazepam was discontinued and risperidone dosage was tapered to 0.5 mg with observation and follow up in the clinic, and no symptom relapse was observed. The complete discontinuation of her medications is planned next visit while monitoring the patient for signs of relapse. 

      Discussion

      Cushing syndrome may initially present as psychosis, which may be misdiagnosis as a primary psychotic disorder, delaying the proper diagnosis and management. Our patient presented to a psychiatry hospital before being referred to us because she resisted psychosis treatment, the resistance to treatment of primary illness due to psychiatric manifestation is not uncommon, as Fujii et al. [6] reported the management of a patient who resisted schizophrenia treatment for 10 years before being diagnosed with Cushing syndrome.

      Agitation with psychosis is likely the main obstacle for properly evaluating, diagnosing, and treating patients with Cushing syndrome. In our patient, we aimed to reduce her agitation to facilitate clinical evaluation. The organic cause of psychosis often responds poorly to antipsychotic medication and exhibits a challenge in managing agitation which necessitate the utilization of highly sedating medications, to facilitate further clinical evaluation. Shah et al. [7] reported similar difficulty treating a patient with agitation despite prescribing lorazepam and 1 mg haloperidol twice daily, agitation was poorly controlled. In our case, the patient responds to a high dose of Olanzapine with lorazepam in a better way than the case report that was managed with haloperidol with lorazepam.

      Psychiatric symptoms secondary to medical conditions usually occur transiently and they resolve after treatment of the primary cause, however, the duration for complete resolution of symptoms is unknown. In our case, the patient gradually improved for three months prior to achieving remission, whereas a patient reported by Wu et al. [8] went into complete remission one-month post-cortisol level correction.

      Conclusions

      Cushing syndrome, like many other endocrine diseases, can present as treatment-resistant psychiatric symptoms, which may be missed and treated as a primary psychiatric illness due to the lack of proper assessment and management. In this study, we tried to correlate the psychiatric symptoms with Cushing syndrome, the challenges we faced, and the response to the treatment. Our case report gives an insight into possible rare secondary causes of psychosis and advice a thorough evaluation of patients.


      References

      1. Your bibliography. (2021). Accessed: March 27, 2021: https://www.ncbi.nlm.nih.gov/books/NBK470218/.
      2. Etxabe J, Vazquez JA: Morbidity and mortality in Cushing's disease: an epidemiological approach. Clin Endocrinol (Oxf). 1994, 40:479-84. 10.1111/j.1365-2265.1994.tb02486.x
      3. Starkman MN, Schteingart DE: Neuropsychiatric manifestations of patients with Cushing’s syndrome. Relationship to cortisol and adrenocorticotropic hormone levels. Arch Intern Med. 1981, 215:9. 10.1001/archinte.1981.00340020077021
      4. Dorn LD, Burgess ES, Dubbert B, et al.: Psychopathology in patients with endogenous Cushing's syndrome: 'atypical' or melancholic features. Clin Endocrinol (Oxf). 1995, 43:433-42. 10.1111/j.1365-2265.1995.tb02614.x
      5. Sharma ST, Nieman LK, Feelders RA: Cushing's syndrome: epidemiology and developments in disease management. Clin Epidemiol. 2015, 7:281-93. 10.2147/CLEP.S44336
      6. Fujii Y, Mizoguchi Y, Masuoka J, et al.: Cushing’s syndrome and psychosis: a case report and literature review. Prim Care Companion CNS Disord. 2018, 20:18br02279. 10.4088/PCC.18br02279
      7. Shah K, Mann I, Reddy K, John G: A case of severe psychosis due to Cushing’s syndrome secondary to primary bilateral Macronodular adrenal hyperplasia. Cureus. 2019, 11:e6162. 10.7759/cureus.6162
      8. Wu Y, Chen J, Ma Y, Chen Z: Case report of Cushing’s syndrome with an acute psychotic presentation. Shanghai Arch Psychiatry. 2016, 28:169-72. 10.11919/j.issn.1002-0829.215126

      From https://www.cureus.com/articles/98986-cushings-syndrome-with-acute-psychosis-a-case-report

      • Like 1
    9. Dr. Friedman uses several medications to treat Cushing’s syndrome that are summarized in this table. Dr. Friedman especially recommends ketoconazole. An in-depth article on ketoconazole can be found on goodhormonehealth.com. 

       Drug  How it works  Dosing  Side effects 
      Ketoconazole  (Generic, not FDA approved in US)  blocks several steps in cortisol biosynthesis  Start 200 mg at 8 and 10 PM, can up titrate to 1200 mg/day 
      • Transient increase in LFTs 
      • Decreased testosterone levels 
      • Adrenal insufficiency 
      Levoketoconazole (Recorlev)  L-isomer of Ketoconazole  Start at 150 mg at 8 and 10 PM, can uptitrate up to 1200 mg  nausea, vomiting, increased blood pressure, low potassium, fatigue, headache, abdominal pain, and unusual bleeding 
      Isturisa (osilodrostat)  blocks 11-hydroxylase  2 mg at bedtime, then go up to 2 mg at 8 and 10 pm, can go up to 30 mg  Dr. Friedman often gives with spironolactone or ketoconazole. 
      • high testosterone (extra facial hair, acne, hair loss, irregular periods)  • low potassium 
      • hypertension 
      Cabergoline (generic, not FDA approved)  D2-receptor agonist  0.5 to 7 mg 
      • nausea,  • headache  • dizziness 
      Korlym (Mifepristone)  glucocorticoid receptor antagonist  300-1200 mg per day 
      • cortisol insufficiency (fatigue, nausea, vomiting, arthralgias, and headache) 
      • increased mineralocorticoid effects (hypertension, hypokalemia, and edema 
      • antiprogesterone effects (endometrial thickening) 
      Pasireotide (Signafor)  somatostatin receptor ligand  600 μg or 900 μg twice a day  Diabetes, hyperglycemia, gallbladder issues 

      For more information or to schedule an appointment with Dr. Friedman, go to goodhormonehealth.com

      • Like 1
    10. Abstract

      Introduction

      Stress-related brain disorders can be associated with glucocorticoid disturbance and hippocampal alteration. However, it remains largely unknown how cortisol affects the structure and function of hippocampus. Cushing's disease (CD) provides a unique “hyperexpression model” to explore the effects of excessive cortisol on hippocampus as well as the relation between these effects and neuropsychological deficits.

      Methods

      We acquired high-resolution T1-weighted and resting-state functional magnetic resonance imaging in 47 CD patients and 53 healthy controls. We obtained the volume and functional connectivity of the hippocampal rostral and caudal subregions in both groups. Relationships between hippocampal alterations, neuroendocrine, and neuropsychological assessments were identified.

      Results

      Relative to control subjects, the CD patients had smaller volumes of all four hippocampal subregions. Furthermore, whole brain resting-state functional connectivity analyses with these four different hippocampal regions as seeds revealed altered hippocampal functional connectivity with high-order networks, involving the DMN, frontoparietal, and limbic networks in CD patients. The intrinsic hippocampal functional connectivity was associated with the quality of life of the CD patients.

      Conclusions

      Our findings elucidate the cumulative effect of excess cortisol on the morphology and function of hippocampus and reinforce the need for effective interventions in stress-related brain disease to halt potential hippocampal damage.

       

      1 INTRODUCTION

      Converging evidence has pointed to a strong linkage between the cortisol and human brain and stress-related neuropsychiatry disorders, such as major depression disorder and posttraumatic stress disorder (de Kloet et al., 2005). However, it remains to be established how this stress hormone influences specific brain structures and functions, particularly in humans, which is of particular importance for both treatment of stress-related disorders and research on cortisol effects in the brain.

      Cushing's disease (CD) is caused by an adrenocorticotropic hormone pituitary adenoma and characterized by chronic hypercortisolism. This condition is therefore a unique and natural “hyperexpression model” to investigate the chronic effects of cortisol on brain physiology and cognition (Zhang et al., 2021). By applying multimodal neuroimaging techniques to CD patients, previous studies have observed that chronic hypercortisolism could cause a number of abnormalities in various brain phenotypes. Among these neural changes of CD patients, hippocampal anomalies are the most replicated findings. Studies on CD patients report hippocampal changes that converge with morphologic alterations such as reduction in volume (Burkhardt et al., 2015; Toffanin et al., 2011). Moreover, abnormal cerebral blood flow and glucose metabolism in hippocampus have also been found in CD patients. Both structural and functional alterations in the hippocampus might contribute to the psychotic symptoms in CD patients (Frimodt-Møller et al., 2019). However, it is well established that psychosis is better described as a brain connectional diaschisis rather than isolated regional dysfunctions (Matthews & Hampshire, 2016). These current hippocampus-related findings were mainly obtained by voxel-based or regional analyses of brain volume or metabolism properties, and researchers have not determined whether the organizational patterns of hippocampal functional connectivity are disrupted in CD patients.

      The hippocampus is easily targeted by long-term hypercortisolism because this area is a part of the stress response system and is abundant in mineralocorticoid receptors and glucocorticoid receptors (McEwen et al., 2016). Also recently, studies on macaques and humans have observed that hippocampus is an anatomically and functionally heterogeneous region along the rostral/caudal-dorsal/ventral axis (Schultz & Engelhardt, 2014). Specifically, the rostral hippocampus has connections with prefrontal regions and relates to stress, emotion, and affect. In contrast, the caudal hippocampus mainly connects to sensory cortical areas and performs primarily cognitive functions (Fanselow & Dong, 2010). Therefore, the hippocampus should be studied in a set of separate structures with rostral and caudal hippocampus. Whether the hippocampal subregions exhibit differentially altered connectivity patterns responding to chronic hypercortisolism remains largely unknown.

      The present study further extends this work by examining the relationship between hippocampal subregions and resting-state functional connectivity in large-scale brain networks, as measured by resting-state fMRI (rs-fMRI) (Park & Friston, 2013). We focus on default mode network (DMN), frontoparietal, and limbic networks, given their involvement in stress related psychiatric illnesses. The first is the DMN, which supports self-related cognitive functions. Complementing the DMN is the frontoparietal network, which supports the cognitive regulation of behavior and emotion. Finally, the limbic networks play a key role in emotion regulation.

      In this study, first, to explore the structural changes of hippocampal subregions in CD patients, we performed a volumetric MRI analysis of the four subregions (left rostral hippocampus, left caudal hippocampus, right rostral hippocampus, and right caudal hippocampus). Given the known direct neurotoxic effects of cortisol on hippocampus, we predicted that chronic hypercortisolism caused smaller hippocampal volumes in CD patients. Second, we used these four subregions as seed regions separately and mapped whole-brain functional connectivity patterns associated with each subregion to examine alterations in hippocampal functional connectivity in CD patients. Considering the psychiatric symptoms in CD patients, it is reasonable to expect the presence of altered hippocampal functional connectivity with high-order networks.

      2 MATERIAL AND METHODS

      2.1 Participants

      A total of 47 participants with a diagnosis of CD and 53 healthy control (HC) subjects were included in this study. The CD patients underwent transsphenoidal surgery at the Department of Neurosurgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital between May 2017 and November 2019. According to the clinical practice guideline (Nieman et al., 2015), CD was diagnosed by experienced endocrinologists and confirmed by postsurgical pathology. The detailed preoperative assessments of diagnostic criteria have been reported in our previous study. HCs were recruited from the local community and were controlled for any history of psychopathology abnormalities. All participants were right-handed and had normal vision and auditory sensation. The study was approved by the local ethics committee of the Chinese PLA General Hospital and written informed consent was obtained from each participant. The data of these 47 CD and 53 HC subjects have been partially used in our previous studies (Wang et al., 2019; Zhang et al., 2021).

      2.2 Neuroendocrine and neuropsychological assessment

      All participants underwent biochemical evaluation to assess their cortisol level. We quantified the levels of 24-h urinary free cortisol (24hUFC, nmol/24h); serum cortisol (nmol/L) at 0:00, 8:00, and 16:00. Cortisol was detected with an ADVIA Centaur Analyzer (Siemens Healthcare Diagnostics, Tarrytown, NY, USA). Cortisol levels at 8:00 as well as 24hUFC were also measured in 51 HC subjects.

      All participants underwent a comprehensive neuropsychological assessment with an expert psychiatrist, including Self-Rating Depression Scale (SDS), Self-Rating Anxiety Scale (SAS), Mini-mental State Examination (MMSE), and Montreal Cognitive Assessment (MoCA). Moreover, health-related quality of life and neuropsychiatric symptoms of CD patients were evaluated with the Cushing's Quality-of-Life (CushingQoL) questionnaire (Nelson et al., 2013) and Chinese version of the neuropsychiatric inventory (CNPI) (Leung et al., 2001), respectively.

      2.3 Image acquisition

      Structural and functional MRI data were acquired on a 3.0-Tesla MR system (Discovery MR750, General Electric) with an 8-channel head coil. High-resolution structural 3D T1-weighted images were conducted using a sagittal Fast Spoiled Gradient-Echo (FSPGR) sequence with the following parameters: repetition time = 6.7 ms, echo time = 2.9 ms, flip angle = 7°, field of view = 250 × 250 mm2, number of slices = 192, voxel size = 1 × 1 × 1 mm3 with no gap. The functional images were acquired using an echo-planar imaging (EPI) sequence with repetition time = 2000 ms, echo time = 30 ms, flip angle = 90°, thickness/gap = 3.5 mm/0.5 mm, slices = 36, field of view = 224 × 224 mm2, voxel size = 3.5 × 3.5 × 3.5 mm3, number of total volumes = 240. Soft earplugs were used to attenuate scanner noise and head motion was restrained with foam padding. During functional scanning, all participants were requested to keep their eyes closed and stay awake.

      2.4 rs-fMRI data preprocessing

      Preprocessing of the rs-fMRI images was conducted using SPM12 and Data Processing Assistant for Resting-State fMRI (DPABI, http://www.restfmri.net/forum/DPARSF). The first 10 volume of the functional images were removed to avoid initial steady-state problems. Then functional images were spatially realigned to the first image for motion correction, and reslicing for acquisition temporal delay. The head motion of all participants in this study had no more than 2-mm translation or 2° rotation in any direction. Next, functional images were coregistered to each participant's segmented gray matter T1 image, and then spatially normalized to the MNI space, resampled to 3-mm isotropic voxels. Subsequently, the global signal, white matter signal, cerebrospinal fluid signal and 24-motion vectors were regressed from the data. Finally, linear detrending and bandpass filter (0.01−0.08 Hz) were carried out to reduce the effects of low-frequency drift and high-frequency physiological noise.

      2.5 Hippocampal functional connectivity

      The hippocampus has been functionally parcellated into four subregions (left rostral hippocampus, left caudal hippocampus, right rostral hippocampus, and right caudal hippocampus) based on Human Brainnetome Atlas (Fan et al., 2016). On each hippocampal subregion, we performed seed-based functional connectivity analysis. Briefly, hippocampal functional connectivity maps were obtained by computing the Pearson correlation coefficient for each voxel's time course with the average time course inside the region of interest. Notably, the computation was constrained within a gray-matter mask which was generated by thresholding (a threshold of 0.2) a prior gray-matter probability map in SPM12. The resulting correlation coefficients were further converted to z scores using Fisher's r-to-z transform to improve normality. For each subject, we obtained 4 z-score maps indicative of the intrinsic functional connectivity patterns of the four hippocampal subregions. To exclude the possible confounding effect of hippocampal volume in CD patients, we performed a voxel-based morphometry analysis on structural MRI images and took the volume of hippocampal subregions as a covariate in the functional connectivity statistical analyses.

      2.6 Statistical analysis

      All demographic and clinical variables including neuroendocrine and neuropsychological scores were compared by two-sample t-tests. Sex composition of the two groups was compared using a Pearson's chi-square test (two-tailed). To explore differences in hippocampal functional connectivity between CD patients and HCs, general linear models were performed in a voxel-wise fashion. To exclude the possible confounding effects of age, gender, education level, and volume of hippocampal subregions, we used these measures as covariates in the general linear models. Multiple comparison correction was performed using a FDR of 0.05 within the grey matter mask.

      In CD patients group, a linear regression analysis was further performed to explore the relationship between functional connectivity of the clusters showing significant group differences and neuropsychological scores as well as the endocrinological indicators (cortisol and 24hUFC). Multiple comparisons were also corrected using the FDR method with a corrected threshold of q < 0.05.

      3 RESULTS

      3.1 Demographic, endocrinological, and neuropsychological results

      Table 1 shows the demographic characteristics of the CD patients and the HCs. There were no significant differences in terms of age, sex distribution, and years of education between groups. Compared with HCs, CD patients had significantly lower MoCA scores and higher SDS and SAS scores (Table 1). As expected, the CD patients had significantly higher levels of serum cortisol and 24hUFC (p < .001). Moreover, we calculated the volumes of the four hippocampal subregions and found smaller volumes of all four hippocampal subregions in the CD patients.

      TABLE 1. Participant characteristics
        CDs (n = 47) HCs (n = 53) p Value
      Age (years) 37.38 ± 10.61 (20–59) 34.79 ± 10.72 (21–63) .113
      Gender (male/female) 4/43 4/49 .859
      Education (years) 11.00 ± 4.11 11.74 ± 3.10 .311
      Illness duration (months) 41.62 ± 53.71
      Neuropsychological tests      
      MoCA 22.47 ± 3.98 (n = 45) 27.72 ± 2.00 <.001
      SDS 40.18 ± 9.96 (n = 45) 27.13 ± 4.42 <.001
      SAS 38.27 ± 7.90 (n = 45) 26.98 ± 4.47 <.001
      CNPI 11.93 ± 9.68 (n = 45)
      Cushing QOL 37.76 ± 8.29 (n = 45)
      Endocrinological tests      
      Serum cortisol (nmol/L)      
      0:00 am 633.81 ± 237.59 (n = 46)
      8:00 am 735.34 ± 279.44 (n = 47) 358.51 ± 107.43 (n = 51) <.001
      16:00 pm 671.05 ± 273.56 (n = 47)
      24hUFC (nmol/24 h) 2381.59 ± 1653.16 (n = 41) 252.03 ± 119.47 (n = 47) <.001
      Volume of hippocampal subregions (mm3)      
      Left rostral hippocampus 343.75 ± 39.15 (257.18–423.27) 365.69 ± 27.19 (313.21–442.06) .001
      Left caudal hippocampus 272.69 ± 32.74 (206.63–339.04) 296.39 ± 23.13 (249.62–347.61) <.001
      Right rostral hippocampus 305.10 ± 33.71 (229.67–396.89) 336.76 ± 25.98 (274.95–415.16) <.001
      Right caudal hippocampus 320.42 ± 32.60 (238.16–396.58) 347.87 ± 27.16 (294.00–415.80) <.001
      • Abbreviations: 24hUFC, 24-h urinary free cortisol.; CDs, Cushing's disease patients; CNPI, Chinese version of neuropsychiatric inventory; Cushing QOL, Cushing Quality of Life Scale; HCs, healthy controls; MoCA, Montreal Cognitive Assessment; SAS, Self-Rating Anxiety Scale; SDS, Self-Rating Depression Scale.
      • Note: All values are expressed as mean ± SD. Group differences in sex between CDs and HCs were examined using chi-square test. Group differences in the other demographic and clinical characteristics between CDs and HCs were examined using two-sample t-tests (two-tailed).
       

      3.2 Spatial distribution of hippocampal functional connectivity

      The hippocampal functional connectivity maps of both CD patients and HCs are presented in Figure 1. Visually, the spatial distributions of hippocampal functional connectivity were highly similar between groups, in spite of some differences in strength. We observed that the brain regions significantly positively connecting to hippocampus were primarily distributed in several limbic network regions (the orbital frontal cortex, bilateral medial temporal regions, and temporal pole) and DMN regions (bilateral medial frontal cortex, posterior cingulate gyrus/precuneus, and anterior cingulate cortex). Brain regions with negative connectivity to hippocampus were chiefly distributed in the frontoparietal network regions (dorsolateral prefrontal cortex, supramarginal gyrus, and angular gyrus).

       

      Details are in the caption following the image

      Between-group differences in functional connectivity of the hippocampal subregions. The first column shows the hippocampal functional connectivity subregions. The second and third columns show the hippocampal functional connectivity maps within CD and HC groups, respectively. Further between-group comparisons showed that CD patients had significantly altered hippocampal functional connectivities relative to HCs, with a corrected statistical threshold of < .05. ROI1, left rostral hippocampus; ROI2, left caudal hippocampus; ROI3, right rostral hippocampus; ROI4, right caudal hippocampus; ROI, region of interest; CD, Cushing's disease; HC, healthy control

      3.3 Altered hippocampal functional connectivity in CD patients

      The significant differences in functional connectivity with each hippocampal subregion between the CD patients and HCs groups are illustrated in third column of Figure 1. Both the right and left rostral hippocampus exhibited significantly decreased functional connectivity with the superior parietal lobe (SPL), a component of the frontoparietal network. Moreover, right rostral hippocampus exhibited additional increased functional connectivity with right inferior frontal gyrus, a component of DMN. For the left caudal hippocampus, significantly altered functional connectivity was found to the DMN regions, including (bilateral medial frontal cortex, angular gyrus, anterior, and posterior cingulate cortex). We also observed decreased functional connectivity between the right caudal hippocampus and anterior cingulate cortex. Additionally, the right caudal hippocampus exhibited increased functional connectivity with some limbic regions including the right orbital frontal cortex and temporal pole (Table 2).

      TABLE 2. Brain regions showing changed RSFC between CDs and HCs groups
              Peak MNI coordinate  
        Brain regions BA Cluster size (voxels) x y z Peak T
      ROI-based RSFC              
      ROI1 R IFG 48 219 57 21 —3 4.598
      L angular 39 423 −27 −72 51 −5.530
      RIO2 R thalamus 114 9 −6 3 −5.905
      L angular 39 195 −27 −72 54 −4.830
      R angular 39 384 36 −66 48 −5.607
      ROI3 R MTG 20 633 39 6 −21 4.410
      L angular 39 195 −27 −72 54 −4.830
      R angular 39 384 36 −66 48 −5.607
      MFG/ACC 10/32 572 −3 42 −3 −4.033
      PCC/PreCUN 26/23 709 12 −45 27 −4.502
      ROI4 MFG/ACC 32 465 3 48 6 −4.670
      R MTG/OFC 48/21 747 30 3 −21 4.208
      • Note: Statistical threshold was set at p < .05, corrected.
      • Abbreviations: CDs, Cushing's disease patients; HCs, healthy controls; ROI, regions of interest; BA, Brodmann areas; MNI, Montreal Neurological Institute; RSFC, resting-state functional connectivity; SFG, superior frontal gyrus; MFG, middle frontal gyrus; dMFG, dorsal medial frontal gyrus; IPL, inferior parietal lobule; AG, angular gyrus; ROL, rolandic operculum; Ins, insular; PrCG, precentral gyrus; L, left; R, right; ROI1, left rostral hippocampus; ROI2, left caudal hippocampus; ROI3, right rostral hippocampus; ROI4, right caudal hippocampus.
       

      3.4 Brain–behavior relationships in the CD patients

      In the correlation analyses of CD patients, the mean values of the functional connectivity between the left caudal hippocampus and anterior cingulate cortex correlated positively with the Cushing's QoL scores (r = .327, p < .05) (Figure 2). No other correlations were found for volumes and functional connectivity of the four hippocampal subregions with neuroendocrine and neuropsychological assessment in the CD patients.

       

      Details are in the caption following the image

      Significant correlations between left hippocampal functional connectivity and the quality of life in CD patients. CD, Cushing's disease; Hip, hippocampus; ACC, anterior cingulate cortex

      4 DISCUSSION

      Using a cohort of CD patients and HCs, the present study performed a comprehensive investigation to reveal how the chronic hypercortisolism affects the morphology and connectivity of hippocampal subregions and their relationships with neuroendocrine and neuropsychological assessment. Compared with the HCs, the CD patients had smaller volumes of all four hippocampal subregions. Furthermore, CD patients exhibited differential patterns of altered hippocampal functional connectivity with high-order networks, involving the DMN, frontoparietal, and limbic networks. The intrinsic hippocampal functional connectivity was associated with the quality of life of the CD patients. Together, these findings elucidate the cumulative effect of cortisol on the morphology and function of hippocampus and provide important information to further understand the role of hippocampus in stress-related brain disease.

      Cortisol, the end product of the hypothalamic–pituitary–adrenal axis, plays a critical role in the body's response to stress and maintenance of homeostasis (Sapolsky et al., 2000); however, chronic hypercortisolism is known to impair neurons in the hippocampus. CD patients naturally demonstrate chronic excessive amounts of cortisol; therefore these patients serve as a natural “hyperexpression model” to investigate the chronic effects of cortisol on human hippocampus. Importantly, we showed the CD patients are associated with smaller hippocampal volumes in all four subregions. In line with our study, previous structural imaging studies have shown hippocampal volume decreases in CD patients (Frimodt-Møller et al., 2019; Toffanin et al., 2011). Furthermore, Brown et al. found that healthy volunteers were associated with a significant reduction in hippocampal volume following only 3-day stress doses of corticosteroid administration, strongly suggesting the effects of cortisol on hippocampal size. It is important to note that chronic hypercortisolism can affect the hippocampus in at least two ways: by direct neurotoxic effects on the hippocampus (Lupien et al., 2018; Uno et al., 1994) and by reduction in hippocampal neurogenesis (Saaltink & Vreugdenhil, 2014). Moreover, cortisol stimulates the release of excitatory amino acids glutamate on hippocampal cells (de Kloet et al., 2005). On the other hand, chronic elevations of cortisol also reduce neurotrophic factors that includes nerve growth factor and brain-derived neurotrophic factor (McEwen et al., 2015).

      The different patterns of functional connectivity in rostral hippocampus versus caudal hippocampus might be associated to the specific cytoarchitecture along the rostral/caudal hippocampus. Accumulated evidence from both animal and human studies suggests that different parts of the hippocampus display distinctive gene expression and anatomical projections patterns (Fanselow & Dong, 2010). In detail, gene expression in the rostral hippocampus correlates with regions involved in emotion and stress (amygdala and hypothalamus). Moreover, the rostral hippocampus has connections with prefrontal regions, exerts strong regulatory control of the hypothalamic–pituitary–adrenal axis with a negative feedback (Toffanin et al., 2011). Accordingly, as demonstrated in this study, chronic hypercortisolism predominantly disrupted the functional connectivity in rostral hippocampus.

      Another major finding in this study was altered hippocampal functional connectivity with DMN, frontoparietal, and limbic networks in CD individuals relative to that in HCs. Emerging evidence proposes that interactions within and between these large-scale brain networks play important roles on brain functions and may be affected in multiple psychiatric disorders (Menon, 2011; Sha et al., 2019). Among these brain networks, the DMN is anchored in the medial prefrontal cortex and posterior cingulate cortex and is implicated in internally directed attention and self-referential processing (Raichle, 2015), while the frontoparietal and limbic networks support the cognitive regulation of emotion, attention, and behavior (Buhle et al., 2014; Kohn et al., 2014). The engagement of these high-level functional networks may suggest the linkage of abnormal stress hormone cortisol to cognitive deficits in CD patients. In line with our study, previous studies have shown stress-induced cortisol increase was associated with altered connectivity within the major brain networks (Zhang et al., 2019, 2020, 2020). Meanwhile, structural and functional alterations in these brain systems are also found in CD patients. For example, many functional imaging studies have consistently demonstrated altered brain activities and functional connectivity involving in DMN, frontoparietal, and limbic networks (Jiang et al., 2017; Wang et al., 2019; Zhang et al., 2021), even in the patients with long-term remission of CD (van der Werff et al., 2015). Importantly, previous studies have shown that the CD patients had widespread reductions of white matter integrity, which provide further evidence for the structural substrate for the persistence of these functional deficits (Pires et al., 2015; van der Werff et al., 2014). Here, we propose that by altering hippocampal processes via the abundant glucocorticoid and mineralocorticoid receptors, exposure to hypercortisolism disrupts the interactions with DMN, frontoparietal, and limbic networks in CD patients, thus engender vulnerability for emotional and cognitive problems. In line with this view is evidence that altered hippocampal functional connectivity is associated with the quality of life in CD patients. Because impaired quality of life is a persistent complaint from CD patients (Webb et al., 2018), it is important to accurately assess which aspects of QoL are affected in order to better understand the severity of hypercortisolism on patients and the potential efficacy of treatment. CushingQoL questionnaire has proven to be a valuable resource for assessing health-related QoL in CD patients, based on the combination of psychosocial issues and physical problems (Nelson et al., 2013). A better understanding of the neuroplasticity and continuing quality of life change may in turn facilitate advances in management and intervention.

      Several issues need to be addressed further. First, although the sample size of this study was relatively large, the findings still need to be further replicated in an independent sample. Second, the cross-sectional, observational nature of our study design precludes any causal conclusions. Therefore, studies tracking dynamic changes in hippocampal functional connectivity following the remission of hypercortisolism are needed. We are currently following up participants as part of a longitudinal study. Finally, a combined analysis of multimodal imaging including structural and metabolic data would provide integrated information on the effect of cortisol excess on human brain.

      In short, we demonstrate that CD patients present atypical morphology and functional connectivity of hippocampus. Here we observed the chronic hypercortisolism caused smaller volumes of all hippocampal subregions. This volume change was in line with the preclinical research that excess cortisol cause dendritic shrinkage and loss of spines in the hippocampus. Functionally, CD patients demonstrated altered hippocampal connectivity whose nodes include key components of the DMN, frontoparietal, and limbic networks. These multimodal results reinforce the need for effective therapeutic interventions in stress-related brain disease to halt possible hippocampal damage.

      ACKNOWLEDGMENTS

      This study was supported by the National Natural Science Foundation of China (No. 82001798 and No. 81871087), Military Young Scholar Medical Research Fund of Chinese PLA General Hospital (No. QNF19071), and Medical Big Data and Artificial Intelligence Development Fund of Chinese PLA general Hospital (No. 2019MBD-039).

        CONFLICT OF INTEREST

        The authors report no biomedical financial interests or potential conflicts of interest.

        Read more, including references, at https://onlinelibrary.wiley.com/doi/10.1002/brb3.2507

        • Like 1
      1. list_frequency_early_symptoms.png?update
        list_frequency_symptoms.png?updated=2022

        Let’s keep the ball rolling: the more members in the community, the smarter the insights. We're about 1,468 contributors away from being able to tell which treatments were reported to work better for different subgroups with common aspects. You can help get there by simply inviting others! Every invite posted adds about 10 new members to this community. Together, we have the power to build the largest real-world knowledge-base for Cushing's syndrome

        Take the survey: https://stuff.health/s/uyZ91Lzz 

        • Like 1
      2. Clinical trial analyses focus on the human body’s homeostatic response to potent HSD-1 inhibition by SPI-62

        Results highlight that urinary free cortisol is distinct from intracellular cortisol that causes symptoms in patients with Cushing’s syndrome or autonomous cortisol secretion

        PORTLAND, Ore.--()--Sparrow Pharmaceuticals, an emerging, clinical-stage biopharmaceutical company developing novel, targeted therapies for disorders of glucocorticoid excess, today presented new pharmacological data during a poster session and a Rapid Communications session titled, “HPA axis modulation by a potent inhibitor indicates 11β-hydroxysteroid dehydrogenase type 1 (HSD-1) is a main source of cortisol that can bind intracellular receptors” at the 24th European Congress of Endocrinology (ECE 2022). Sparrow scientists examined the steroid hormone changes after administration of its lead therapeutic candidate, SPI-62, an HSD-1 inhibitor, to healthy adults.

         

        “Normalized urinary free cortisol, or UFC, is a standard therapeutic target for patients with Cushing’s syndrome,” said David A. Katz, Ph.D., CSO at Sparrow Pharmaceuticals, “But that biomarker doesn’t measure the cortisol that can access intracellular receptors and cause symptoms. UFC normalization has been shown not to correlate with clinical endpoints in patients with Cushing’s syndrome. Many patients with autonomous cortisol secretion have normal UFC, yet substantial cortisol morbidity. As we conduct clinical trials for patients with those diseases, we’re in search of better ways to measure the cortisol that makes patients ill.”

        The study analyzed historical clinical trial data to better characterize how SPI-62 impacts cortisol levels and the body’s homeostatic response to those changes.

        Conclusions of the study include:

        • Half of hepatocellular cortisol with access to intracellular receptors is generated in healthy adults by HSD-1.
        • ACTH increase compensates for the effect of HSD-1 inhibition on systemic cortisol levels.
        • Secondary increases of androgen levels have not been associated to date with clinical consequences.
        • Large changes of the amount of cortisol that can bind intracellular receptors, and thus cause cortisol-related morbidity, can occur independently of urinary free cortisol levels.

        HSD-1 converts cortisone to cortisol in tissues in which cortisol excess is associated with morbidity including liver, adipose, bone, and brain. SPI-62 is a potent HSD-1 inhibitor in clinical development for treatment of Cushing’s syndrome and autonomous cortisol secretion, and as adjunctive therapy to prednisolone in polymyalgia rheumatica. In Phase 1 clinical trials SPI-62 was generally well tolerated and associated with maximal liver and brain HSD-1 inhibition.

        To register and view the abstracts, visit ECE’s website here.

        From https://www.businesswire.com/news/home/20220524005465/en/Sparrow-Pharmaceuticals-Presents-New-Clinical-Trial-Data-Analyses-on-HSD-1-Inhibitor-SPI-62-at-the-24th-European-Congress-of-Endocrinology

         

        • Like 1
      3. Data presented at AACE 2022 detail levoketoconazole-specific effects observed among patients with endogenous Cushing's syndrome from the phase 3 LOGICS trial.

        New research presented at the American Academy of Clinical Endocrinology (AACE) annual meeting provides insight into the effects of treatment with levoketoconazole (Osilodrostat) among patients with endogenous Cushing’s syndrome.

        An analysis of data from a double-blind, placebo-controlled, randomized withdrawal study, results of the study demonstrate levoketoconazole provided benefits across a range of etiologies and provide evidence of levoketoconazole-specific effects through the withdrawal and reintroduction of therapy during the trial.

        “This LOGICS study showed that treatment with levoketoconazole benefitted patients with Cushing’s syndrome of different etiologies and a wide range in UFC elevations at baseline by frequent normalization of mUFC and concurrent improvements in serum cholesterol,” said Maria Fleseriu, MD, professor of medicine and neurological surgery and director of the Northwest Pituitary Center at Oregon Health and Science University, during her presentation. “The benefits observed were established as levoketoconazole-specific via the loss of therapeutic effect upon withdrawal to placebo and restoration upon reintroduction of levoketoconazole.”

        An orally administered cortisol synthesis inhibitor approved by the US FDA for treatment of endogenous hypercortisolemia in adult patients with Cushing’s syndrome considered ineligible for surgery, levoketoconazole received approval based on results of the phase 3 open-label SONICS trial, which demonstrated . Launched on the heels of SONICS, the current trial, LOGICS, was designed as phase 3, double-blind, placebo-controlled, randomized withdrawal study aimed at assessing the drug-specificity of cortisol normalization in adult patients with Cushing’s syndrome through a comparison of the effects of withdrawing levoketoconazole to placebo against continuing treatment.

        The trial began with an open-label titration maintenance phase followed by a double-blind randomized withdrawal phase and a subsequent restoration phase, with the randomized withdrawal and restoration phase both lasting 8 weeks. A total of 89 patients with Cushing’s syndrome received levoketoconazole to normalize mUFC. Of these, 39 patients on a stable dose for 4 weeks or more were included in the randomized withdrawal stage of the study. These 39, along with 5 completers of the SONICS trial, were randomized in a 1:1 ratio to continue therapy with levoketoconazole or placebo therapy, with 22 patients randomized to each arm.

        The primary outcome of interest in the study was the proportion of patients with loss of mean urinary free cortisol response during the randomized withdrawal phase of the study, which was defined as an mUFC 1.5 times the upper limit of normal or greater or an mUFC 40% or more above baseline. Secondary outcomes of interest included mUFC normalization at the end of the randomized withdrawal phase of the study and changes in comorbidity biomarkers.

        Overall, 21 of the 22 patients randomized to placebo during the withdrawal stage met the primary endpoint of loss of mUFC compared to just 9 of 22 among the levoketoconazole arm of the trial (treatment difference: -54.5% [95% CI, -75.7 to -27.4]; P=.0002). Additionally, at the conclusion of the randomization phase, mUFC normalization was observed among 11 patients in the levoketoconazole arm of the trial compared to 1 patient receiving placebo (treatment difference: 45.5% [95% CI, 19.2 to 67.9]; P=.0015).

        Further analysis indicated the restoration of levoketoconazole therapy was associated with a. Reversal of loss of contrail control in most patients who had been randomized to placebo. Investigators pointed out the mean change from randomized withdrawal baseline to the end of the randomized withdrawal period in total cholesterol was -0.04 mmol/L for levoketoconazole and 0.9 mmol/L for placebo (P=.0004) and the mean change in LDL-C was -0.006 mmol/L and 0.6 mmol/L, respectively (P=0.0056), with the mean increases in cholesterol observed among the placebo arm reversed during the restoration phase.

        In safety analyses, results suggest the most commonly reported adverse events seen with levoketoconazole treatment, during all study phases combined were nausea and hypokalemia, which occurred among 29% and 26% of patients, respectively. Investigators also pointed out liver-related events, QT interval prolongation, and adrenal insufficiency, which were respecified adverse events of special interest occurred among 10.7%, 10.7%, and 9.5% of patients receiving levoketoconazole, respectively.

        This study, “Levoketoconazole in the Treatment of Endogenous Cushing’s Syndrome: A Double-Blind, Placebo-Controlled, Randomized Withdrawal Study,” was presented at AACE 2022.

         
         
         
        • Like 1
      4. Osilodrostat is associated with improvements in physical manifestations of hypercortisolism and reductions in mean body weight and BMI in adults with Cushing’s syndrome, according to a speaker.

        As Healio previously reported, in findings from the LINC 4 phase 3 trial, osilodrostat (Isturisa, Recordati) normalized mean urinary free cortisol level at 12 weeks in more than 75% of adults with Cushing’s disease. In new findings presented at the AACE Annual Scientific and Clinical Conference, most adults with Cushing’s syndrome participating in the LINC 3 phase 3 trial had improvements in physical manifestations of hypercortisolism 72 weeks after initiating osilodrostat, with more than 50% having no dorsal fat pad, supraclavicular fat pad, facial rubor, proximal muscle atrophy, striae, ecchymoses and hirsutism for women at 72 weeks.

        Adrenal transparent _Adobe
        Source: Adobe Stock

        “Many patients with Cushing’s syndrome suffer from clinical manifestations related to hypercortisolism,” Albert M. Pedroncelli, MD, PhD, head of clinical development and medical affairs for Recordati AG in Basel, Switzerland, told Healio. “The treatment with osilodrostat induced a rapid normalization of cortisol secretion, and improvements in physical manifestations associated with hypercortisolism were observed soon after initiation of osilodrostat and were sustained throughout the study.”

        pedroncelli_alberto_2022_web.jpg
        Albert M. Pedroncelli

        Pedroncelli and colleagues analyzed changes in the physical manifestations of hypercortisolism in 137 adults with Cushing’s syndrome (median age, 40 years; 77.4% women) assigned osilodrostat. Dose titration took place from baseline to 12 weeks, and therapeutic doses were administered from 12 to 48 weeks, with some participants randomly assigned to withdrawal between 26 and 34 weeks. An extension phase of the trial took place from 48 to 72 weeks. Investigators subjectively rated physical manifestations of hypercortisolism in participants as none, mild, moderate or severe. Participants were evaluated at baseline and 12, 24, 34, 48 and 72 weeks.

        At baseline, the majority of the study cohort had mild, moderate or severe physical manifestations of hypercortisolism in most individual categories, including dorsal fat pad, central obesity, supraclavicular fat pad, facial rubor, hirsutism in women and striae. Central obesity was the most frequent physical manifestation rated as severe.

        The percentage of participants with improvements in physical manifestations of hypercortisolism increased from week 12 on for all individual manifestations evaluated in the study, and improvements were maintained through week 72. At 72 weeks, the percentage of participants who had no individual physical manifestations was higher than 50% for each category except central obesity, where 30.6% of participants had no physical manifestations.

        In addition to improvement in physical manifestations, the study cohort had decreases in body weight, BMI and waist circumference at weeks 48 and 72 compared with baseline.

        “The main goal of treating patients with Cushing’s syndrome is to normalize cortisol secretion,” Pedroncelli said. “The rapid reduction and normalization of cortisol levels is accompanied by improvement in the associated clinical manifestations. This represents an important objective for patients.”

        From https://www.healio.com/news/endocrinology/20220512/osilodrostat-improves-physical-manifestations-of-hypercortisolism-for-most-adults

        • Like 1
      5. Crinetics Pharmaceuticals, Inc. (Nasdaq: CRNX) today announced positive results from the multiple-ascending dose (MAD) portion of a first-in-human Phase 1 clinical study of CRN04894, the company's first-in-class, investigational, oral, nonpeptide adrenocorticotropic hormone (ACTH) antagonist that is being developed for the treatment of Cushing’s disease, congenital adrenal hyperplasia (CAH) and other conditions of excess ACTH. Following administration of CRN04894, results showed serum cortisol below normal levels and a marked reduction in 24-hour urine free cortisol excretion in the presence of sustained, disease-like ACTH concentrations.

        “The design of our Phase 1 healthy volunteer study allowed us to demonstrate CRN04894’s potent pharmacologic activity in the presence of ACTH levels that were in similar range to those seen in CAH and Cushing’s disease patients,” said Alan Krasner, M.D., Crinetics’ chief medical officer. “The observation of dose-dependent reductions in serum cortisol levels to below the normal range even in the presence of high ACTH indicates that CRN04894 was effective in blocking the key receptor responsible for regulating cortisol secretion. We believe this is an important finding that may be predictive of CRN04894’s efficacy in patients.”

        ACTH is the key regulator of the hypothalamic-pituitary adrenal (HPA) axis controlling adrenal activation. It is regulated by cortisol via a negative feedback loop that acts to inhibit ACTH secretion. This feedback loop is dysregulated in diseases of excess ACTH. In Cushing’s disease, a benign pituitary tumor drives excess ACTH secretion even in the presence of excess cortisol. While in CAH, an enzyme deficiency results in excess androgen synthesis without normal cortisol synthesis, allowing unchecked ACTH production and requiring lifelong glucocorticoid use. In both diseases, excess ACTH drives over-stimulation of the adrenal gland and leads to a host of symptoms including infertility, adrenal rest tumors, and metabolic complications in CAH and, in Cushing’s disease, symptoms include hypertension, central obesity, neuropsychiatric disorders and metabolic complications. To our knowledge, no other ACTH antagonists are currently in clinical development for diseases of ACTH excess such as Cushing’s disease or CAH.

        The 49 healthy adults evaluated in the multiple ascending dose portion of the Phase 1 study were administered 40, 60 or 80 mg doses of CRN04894, or placebo, daily for 10 days. After 10 days of dosing was complete, evaluable participants were administered an ACTH challenge to stimulate adrenal activation to disease relevant levels. Safety and pharmacokinetic data were consistent with expectations from the single-ascending dose cohorts in the Phase 1 study. There were no discontinuations due to treatment-related adverse events and no serious adverse events reported. Glucocorticoid deficiency was the most common treatment-related adverse event in the MAD cohorts. This was an expected extension of pharmacology given the mechanism of action of CRN04894. CRN04894 showed consistent oral bioavailability in the MAD cohorts with a half-life of approximately 24 hours, which is anticipated to support once-daily dosing.

        Participants in the MAD cohorts who were administered once nightly CRN04894 experienced a dose-dependent suppression of adrenal function as measured by suppression of serum cortisol production of 17%, 29% and 37% on average from baseline over 24 hours for the 40, 60 or 80 mg dosing groups respectively, (despite requirement for glucocorticoid supplementation in some of these subjects to prevent clinical adrenal insufficiency), compared to an average 2% increase in serum cortisol for individuals receiving placebo. The strong, dose-dependent suppression of serum and urine free cortisol was achieved despite ACTH levels in subjects in the 60 and 80 mg cohorts similar to those typically seen in patients with CAH and Cushing’s disease. Even when an additional exogenous ACTH challenge was administered on top of the already increased ACTH levels, cortisol levels remained below the normal range in subjects receiving CRN04894, indicating clinically significant suppression of adrenal activity.

        “Due to its central position in HPA axis, ACTH is the obvious target for inhibiting excessive stimulation of the adrenal in diseases of ACTH excess. Even though the field of endocrinology has known about its clinical significance for more than 100 years, we are not aware of any other ACTH antagonist that has entered clinical development. This is an important milestone for endocrinology and for our company.” said Scott Struthers, Ph.D., founder and chief executive officer of Crinetics. “We are very excited to initiate patient studies in Cushing’s disease and CAH with CRN04894, which will be our third home-grown NCE to demonstrate pharmacologic proof-of-concept and enter patient trials.”

        Crinetics plans to present additional details of safety, efficacy, and biomarker results from the CRN04894 Phase 1 study at an endocrinology-focused medical meeting in 2022.

        Data Review Conference Call Crinetics will hold a conference call and live audio webcast today, May 25, 2022, at 8:00 a.m. Eastern Time to discuss results from the MAD cohorts of the Phase 1 study of CRN04894. To participate, please dial 1-877-407-0789 (domestic) or 1-201-689-8562 (international) and refer to conference ID 13730000. To access the webcast, click here. Following the live event, a replay will be available on the Events page of the Company’s website.

        About the CRN04894 Phase 1 Study Crinetics has completed enrollment of the 88 healthy volunteers in this double-blind, randomized, placebo-controlled Phase 1 study. Participants were divided into multiple cohorts in the single ascending dose (n=39) and multiple ascending dose (n=49) portions of the study. In both the SAD and MAD portions of the study, safety and pharmacokinetics were assessed. In addition, pharmacodynamic responses were evaluated before and after challenges with injected synthetic ACTH to assess pharmacologic effects resulting from exposure to CRN04894.

         

        From https://www.streetinsider.com/Corporate+News/Crinetics+Pharmaceuticals+(CRNX)+Reports+Positive+Top-line+Results+Including+Strong+Adrenal+Suppression+from+CRN04894+Phase+1+Study/20126484.html

        • Like 1
      6. Published: May 15, 2022 (see history)

        DOI: 10.7759/cureus.25015

        Cite this article as: Iturregui J, Shi G (May 15, 2022) Recurrent Metatarsal Fractures in a Patient With Cushing Disease: A Case Report. Cureus 14(5): e25015. doi:10.7759/cureus.25015

        Abstract

        Cushing syndrome (CS) can result from excess exposure to exogenous or endogenous glucocorticoids. The most common endogenous cause of CS is an adrenocorticotropic hormone (ACTH)-secreting pituitary adenoma, known as Cushing disease (CD). Patients typically present with characteristics including truncal obesity, moon facies, facial plethora, proximal muscle weakness, easy bruising, and striae. Insufficiency fractures of the metatarsals are a rare presentation for CS. A 39-year-old premenopausal woman presented to the orthopedic outpatient clinic with recurrent metatarsal fractures and no history of trauma. A metabolic bone disease was suspected, and after further evaluation by endocrinology services, the CD was diagnosed. Surgical resection was performed, and pathology confirmed the presence of a pituitary adenoma. Multiple, recurrent, non-traumatic metatarsal fractures can be the initial presentation of CD in a premenopausal woman.

        Introduction

        Cushing syndrome (CS) is a rare clinical and metabolic disorder caused by excessive exposure to glucocorticoids. In the United States, an estimated 10 to 15 people per million population are affected by CS each year, while studies in Europe report an incidence of 0.7 to 2.4 per million people affected annually [1,2]. Furthermore, CS more commonly affects women [2]. Common characteristics of CS include truncal obesity, moon facies, proximal muscle weakness, fatigue, facial plethora, supraclavicular fullness, peripheral edema, weight gain, striae, easy bruising, acne, hirsutism, amenorrhea, dorsocervical "buffalo" hump, depression, hypertension, impaired glucose tolerance, and osteoporosis [1,3,4].

        The most common cause of CS is exogenous glucocorticoid therapy. Meanwhile, endogenous cortisol hypersecretion commonly results from either an adrenocorticotropic hormone (ACTH)-secreting pituitary adenoma or a cortisol-secreting adrenal tumor. When CS is caused by a pituitary adenoma, this is referred to as Cushing disease (CD). CD is the most common endogenous cause of CS, accounting for 80-85% of cases [1,5].

        Whether a patient’s CS is caused by exogenous or endogenous sources, excessive exposure to steroids can have deleterious effects on the bones, resulting in secondary osteoporosis. The decrease in bone mass and microarchitectural changes increase the risk of fragility fractures, with reported rates as high as 30-67% [6]. The most commonly reported fracture site in CS patients is the vertebrae; however, other reported fracture sites include the ribs, sternum, wrist, elbow, shoulder, pelvis, hip, femoral condyles, tibia, fibula, calcaneus, metatarsals, and phalanges [4,6-16]. There are reports of metatarsal fractures occurring in patients diagnosed with endogenous CS [3,6,7,16-19]. However, to the best of our knowledge, there are no reports of multiple, recurrent, bilateral metatarsal fractures as the initial presentation in a pre-menopausal woman with CD. Here, we present a case of a premenopausal woman with recurrent metatarsal stress fractures who was diagnosed with CD after further evaluation.

        Case Presentation

        A 39-year-old premenopausal woman was evaluated by her primary care physician due to right foot pain after feeling a pop while walking. She reported swelling and some bruising along the lateral aspect of her foot. Her exercise regimen consisted of walking twice a week for 30 minutes at each session. She did not report any traumatic injuries to her foot. Imaging revealed a fifth metatarsal fracture (Figure 1). The patient was placed in a cast walker boot and referred to orthopedics for further evaluation. Orthopedic management included no weight bearing on her right foot and continuing using the cast walker boot or a postop shoe, with reevaluation in four weeks.

        Oblique-radiograph-of-the-right-foot-demonstrating-a-mildly-displaced-transverse-fracture-of-the-proximal-fifth-metatarsal-(arrow).
        Figure 1: Oblique radiograph of the right foot demonstrating a mildly displaced transverse fracture of the proximal fifth metatarsal (arrow).
         
         

        At the time of evaluation, the patient was 161.5 cm tall, weighed 101 kg, and had a BMI of 38.86 kg/m2. Her medical history included hypertension, hyperglycemia, hyperlipidemia, hypothyroidism, obesity, anxiety, obstructive sleep apnea, and colon polyps. The patient reported a history of metatarsal fractures in her left foot in 2008, which healed slowly and without surgical intervention. She also underwent bunion and bunionette surgery on her left foot. Her medications included alprazolam, levothyroxine, lisinopril, bimatoprost, ergocalciferol, meloxicam, and ondansetron. She was a former smoker (2007-2010), a daily wine drinker, and had an active job working as a nurse. Her family history included lung cancer and alcohol abuse in her father; hypertension, hypothyroidism, and alcohol abuse in her mother; and osteoporosis and end-stage renal disease secondary to polycystic kidney disease in her sister.

        At the three-month follow-up visit, the fracture line remained clearly visible, and minimal callus had formed at the fracture site. Surgical fixation was recommended and performed four months after the fracture occurred. Six months after her right foot's fifth metatarsal fracture, she developed new-onset swelling and tenderness over the middle metatarsals dorsally in her right foot with no history of trauma. Radiographs demonstrated new second and third metatarsal neck fractures (Figure 2). Conservative management with a postop shoe for six weeks and re-evaluation was recommended. In the interim between her initial right foot fifth metatarsal fracture and the new right foot second and third metatarsal fractures, the patient was diagnosed with diabetes mellitus type II, treated with a plant-based diet, hospitalized for urolithiasis, and diagnosed with depression. She was started on bupropion. 

        Anteroposterior-radiograph-of-bilateral-feet-demonstrating-second-and-third-metatarsal-neck-fractures-of-the-right-foot-(arrows).
        Figure 2: Anteroposterior radiograph of bilateral feet demonstrating second and third metatarsal neck fractures of the right foot (arrows).
         
         

        Due to the recurrent metatarsal stress fractures with no associated trauma, the patient was referred to endocrinology for workup of metabolic bone disease. Her physical exam revealed no abnormalities, and her overall workup was negative. Bone mineral density results demonstrated osteopenia in the lumbar spine (T-score: -1.8) and left femoral neck (T-score: -1.0), and normal bone density in the left total hip (T-score: -0.80).

        Six months following her right foot's second and third metatarsal fractures, the patient developed right great toe and second toe swelling and bruising. Two months later, after trying supportive tennis shoes and reducing weightbearing on her right foot, she did not notice any improvement and sought orthopedic care. Radiographs revealed a new subacute fracture of the right second proximal phalanx (Figure 3). A magnetic resonance imaging (MRI) scan was ordered, which revealed a first metatarsal shaft stress fracture as well (Figure 4). She underwent conservative management with a Cam walker boot and was referred to endocrinology for re-evaluation for suspected endocrinopathy.

        AP-radiograph-of-bilateral-feet-demonstrating-a-subacute-fracture-of-the-second-proximal-phalanx-of-the-right-foot-(arrow).
        Figure 3: AP radiograph of bilateral feet demonstrating a subacute fracture of the second proximal phalanx of the right foot (arrow).
         
         
        T1-weighted-sagittal-MRI-of-the-right-foot-demonstrating-a-first-metatarsal-shaft-stress-fracture-(arrow).
        Figure 4: T1-weighted sagittal MRI of the right foot demonstrating a first metatarsal shaft stress fracture (arrow).
         
         

        At her endocrinology visit, a physical exam revealed some facial hair, frontal hair loss, and a significant dorsocervical and anterior cervical fat pad. A Cushingoid face shape, facial redness, acne, oligomenorrhea, incremental weight gain over the last decade, centripetal adiposity, easy bruising, and lower leg swelling were also reported. Bone mineral density results reported spine and hip Z-scores within the expected range for age, indicating no osteoporosis. Since she had features of hypercortisolism, labs to evaluate for Cushing syndrome were ordered. The 11:00 pm salivary cortisol levels were elevated to 173 ng/dL and 168 ng/dL in two samples. The 1 mg dexamethasone suppression test failed to suppress her cortisol levels, with an elevated cortisol value of 29 mcg/dL. The 24-hour urine-free cortisol level was elevated at 135 mcg/24 hours. These lab results confirmed a diagnosis of Cushing syndrome. Her ACTH was elevated at 86 pg/mL, which indicated an ACTH-dependent CS. Pituitary MRI demonstrated a 1.1 cm × 1.5 cm × 1.1 cm pituitary lesion, representing a pituitary macroadenoma (Figure 5). The patient underwent endoscopic endonasal transsphenoidal pituitary tumor resection with the goal of treating her Cushing disease and preventing further fragility fractures. Pathology evaluation confirmed a pituitary adenoma.

        T1-weighted-coronal-MRI-of-the-pituitary-demonstrating-a-1.1-cm-×-1.5-cm-×-1.1-cm-cystic-sellar-mass-which-represents-a-pituitary-macroadenoma-(arrow).
        Figure 5: T1-weighted coronal MRI of the pituitary demonstrating a 1.1 cm × 1.5 cm × 1.1 cm cystic sellar mass which represents a pituitary macroadenoma (arrow).
         
         

        Discussion

        This is a case of a 39-year-old woman who presented with recurrent metatarsal fractures with no history of trauma, raising suspicion of a metabolic bone disease. The patient also developed centripetal weight gain, glucose intolerance, kidney stones, depression/anxiety, and Cushingoid features. A laboratory workup performed by endocrinology services confirmed a diagnosis of ACTH-dependent CS. An MRI revealed a pituitary lesion which represented a pituitary macroadenoma, for which surgical resection was performed. Pathology confirmed a pituitary adenoma. The association of multiple, non-traumatic metatarsal fractures occurring in premenopausal women with endogenous CS has been reported in the literature [3,7,19]. However, to the best of our knowledge, this is the first report presenting a premenopausal woman with multiple, recurrent metatarsal fractures as the initial manifestation of CD.

        Several mechanisms play a role in glucocorticoid-induced bone loss, which is more prominent in trabecular bone compared to cortical bone [3,4,6,8]. Normally, trabecular bone has a greater bone turnover rate than cortical bone. In the presence of excess glucocorticoids, trabecular bone has greater sensitivity to glucocorticoids and undergoes slower bone turnover. The most significant effects of excess glucocorticoids on bones are decreased osteoblast function and quantity, which explain the reduced trabecular bone turnover rate [4,10]. The proposed mechanisms for this are glucocorticoid-induced inhibition of osteoblast proliferation and genesis, as well as induction of osteoblast and osteocyte apoptosis [4,10,11]. Furthermore, glucocorticoids decrease bone protein synthesis (e.g., osteocalcin), type I collagen formation, and alkaline phosphatase activity [4]. Additional effects include greater bone resorption, inhibition of intestinal calcium absorption, inhibition of renal calcium reabsorption, and decreased secretion of gonadal steroids and growth hormones [8]. Glucocorticoids also induce protein catabolism, which can result in muscle weakness, decreased bone stimulation from weakened muscle contraction, and further bone loss and debility [4].

        Multiple fragility fractures in the foot with no history of trauma or overuse are uncommon. When evaluating a patient with this presentation, secondary causes for these fractures need to be investigated. Differential diagnoses include osteoporosis, Charcot foot, multiple myeloma, celiac disease, avascular necrosis, and endocrine disorders such as hyperthyroidism, primary hyperparathyroidism, or CS, among others [3,6,7].

        There is a high rate of fragility fractures due to secondary osteoporosis in CS patients, with the vertebrae being most commonly affected [6]. LiYeung and Lui [7] and Albon et al. [19] each reported a case of a pre-menopausal woman who initially presented with multiple metatarsal fractures secondary to an adrenal adenoma causing CS. In each case, the patient’s densitometry indicated osteoporosis. However, in our case and the case reported by Molnar et al. [3] of a pre-menopausal woman with multiple fractures due to CD (recurrent fractures were not reported), the bone densitometries performed did not indicate osteoporosis.

        The patients reported by LiYeung and Lui [7], Albon et al. [19], and Molnar et al. [3] did not demonstrate marked clinical characteristics of CS. In comparison to our patient, she did have multiple Cushingoid features upon her second evaluation by endocrinology. Furthermore, in all our cases, the patients were first evaluated for metatarsal fractures as the initial presentation, which resulted in a diagnosis of endogenous CS after further evaluation.

        Finally, early recognition and treatment of CS are important, as there is an increased risk of morbidity and mortality as the condition progresses [20]. In addition, the treatment of CS can reverse the bone loss that occurs with excess glucocorticoid exposure [4,10]. This case also highlights the importance of collaboration between physicians in the different branches of medicine.

        Conclusions

        Excess glucocorticoid exposure can have deleterious effects on the bones, increasing the risk for secondary osteoporosis and fragility fractures. There needs to be an index of suspicion for metabolic bone disease, including endogenous CS caused by CD, as the underlying etiology of multiple, recurrent, atraumatic metatarsal fractures in pre-menopausal women. Early diagnosis and management of CD can lower the risk of morbidity and mortality as well as reverse bone loss.


        References

        1. Guaraldi F, Salvatori R: Cushing syndrome: maybe not so uncommon of an endocrine disease. J Am Board Fam Med. 2012, 25:199-208. 10.3122/jabfm.2012.02.110227
        2. Valassi E, Santos A, Yaneva M, et al.: The European Registry on Cushing's syndrome: 2-year experience. Baseline demographic and clinical characteristics. Eur J Endocrinol. 2011, 165:383-92. 10.1530/EJE-11-0272
        3. Molnar V, Zekan P, Dušek T, Ivković A: Multiple metatarsal fractures: the first manifestation of Cushing’s disease—a case report. J Am Podiatr Med Assoc. 2021, 111:10.7547/19-024
        4. Han JY, Lee J, Kim GE, et al.: A case of cushing syndrome diagnosed by recurrent pathologic fractures in a young woman. J Bone Metab. 2012, 19:153-8. 10.11005/jbm.2012.19.2.153
        5. Barahona MJ, Sucunza N, Resmini E, et al.: Deleterious effects of glucocorticoid replacement on bone in women after long-term remission of Cushing's syndrome. J Bone Miner Res. 2009, 24:1841-6. 10.1359/jbmr.090505
        6. Papadakis G, Uebelhart B, Goumaz M, Zawadynski S, Rizzoli R: An unusual case of hypercortisolism with multiple weight-bearing bone fractures. Clin Cases Miner Bone Metab. 2014, 10:213-7.
        7. LiYeung LL, Lui TH: Bilateral adrenal adenoma presented as multiple metatarsal and phalangeal fractures. J Orthop Case Rep. 2015, 5:77-8. 10.13107/jocr.2250-0685.353
        8. Trementino L, Appolloni G, Ceccoli L, Marcelli G, Concettoni C, Boscaro M, Arnaldi G: Bone complications in patients with Cushing's syndrome: looking for clinical, biochemical, and genetic determinants. Osteoporos Int. 2014, 25:913-21. 10.1007/s00198-013-2520-5
        9. Abdel-Kader N, Cardiel MH, Navarro Compan V, Piedra Priego J, González A: Cushing's disease as a cause of severe osteoporosis: a clinical challenge. Reumatol Clin. 2012, 8:278-9. 10.1016/j.reuma.2011.11.004
        10. Lee HJ, Je JH, Seo JH, Na YJ, Yoo HJ: Multiple spontaneous rib fractures in patient with Cushing’s syndrome. J Bone Metab. 2014, 21:277-82. 10.11005/jbm.2014.21.4.277
        11. Poonuru S, Findling JW, Shaker JL: Lower extremity insufficiency fractures: an underappreciated manifestation of endogenous Cushing's syndrome. Osteoporos Int. 2016, 27:3645-9. 10.1007/s00198-016-3712-6
        12. Belaya ZE, Hans D, Rozhinskaya LY, et al.: The risk factors for fractures and trabecular bone-score value in patients with endogenous Cushing's syndrome. Arch Osteoporos. 2015, 10:44. 10.1007/s11657-015-0244-1
        13. Tajika T, Shinozaki T, Watanabe H, Yangawa T, Takagishi K: Case report of a Cushing's syndrome patient with multiple pathologic fractures during pregnancy. J Orthop Sci. 2002, 7:498-500. 10.1007/s007760200087
        14. Baron E, Sheinfeld M, Migdal EA, Hardoff R: Multiple pathologic fractures mimicking bone metastases in a patient with Cushing's syndrome. Clin Nucl Med. 1996, 21:506-8. 10.1097/00003072-199606000-00027
        15. Bosch S, Bogaerts S: Pituitary adenoma presenting with bilateral calcaneal stress fracture: a case report. JOSPT Cases. 2021, 1:109-111.
        16. Kostoglou-Athanassiou I, Spiliotis G, Athanassiou L, Myriokefalitakis I: Cushing’s syndrome in a patient with systemic lupus erythematosus. Endocrine Abstracts. 2018, 56:106. 10.1530/endoabs.56.P106
        17. Kaur K, Findling JW: Cushing’s disease. A Case-Based Guide to Clinical Endocrinology. Davies TF (ed): Humana Press, Totowa; 2008. 27-33. 10.1007/978-1-60327-103-5_3
        18. Ontell FK, Shelton DK: Multiple stress fractures. An unusual presentation of Cushing's disease. West J Med. 1995, 162:364-6.
        19. Albon L, Rippin J, Franklyn J: “My feet are killing me!” An unusual presentation of Cushing’s syndrome. Endocrine Abstracts. 2003, 5:26.
        20. Nieman LK: Recent updates on the diagnosis and management of Cushing’s syndrome. Endocrinol Metab (Seoul). 2018, 33:139-46. 10.3803/EnM.2018.33.2.139

        From https://www.cureus.com/articles/91295-recurrent-metatarsal-fractures-in-a-patient-with-cushing-disease-a-case-report

        • Like 1
      7. — More than half of patients saw physical manifestations fully resolve by week 72

        by Kristen Monaco, Staff Writer, MedPage Today 

         

        SAN DIEGO -- Osilodrostat (Isturisa) improved many physical features associated with Cushing's disease, according to additional findings from the phase III LINC-3 study.

        Among 137 adults with Cushing's disease, a 39.5% improvement in central obesity scores was observed from baseline to week 72 with osilodrostat, reported Alberto Pedroncelli, MD, PhD, of Recordati AG in Basel, Switzerland.

        Not only was central obesity the most common physical manifestation associated with hypercortisolism among these Cushing's disease patients, but it was also more frequently rated as severe at baseline, Pedroncelli explained during the American Association of Clinical Endocrinology (AACE) annual meeting.

        Osilodrostat treatment also led to a 34.9% improvement in proximal muscle atrophy at week 72, along with a 34.4% improvement in hirsutism scores.

        By week 72, nearly all physical manifestations of hypercortisolism saw significant improvement -- marked by more than 50% of patients scoring these physical traits as nonexistent:

        • Dorsal fat pat: 50.6%
        • Central obesity: 30.6%
        • Supraclavicular fat pad: 51.8%
        • Facial rubor: 64.7%
        • Hirsutism in women: 53.1%
        • Proximal muscle atrophy: 61.2%
        • Striae: 63.5%
        • Ecchymoses: 87.1%

        Most of these physical manifestation improvements were notable soon after treatment initiation with osilodrostat, Pedroncelli pointed out.

        When stratified according to testosterone levels, hirsutism scores remained either stable or improved in the majority of patients who had normal or above normal testosterone levels. More women with normal testosterone levels over time experienced improvements in hirsutism versus those with levels above the upper limit of normal, who mostly remained stable.

        Osilodrostat is an oral agent that was first FDA approved in March 2020 for adults with Cushing's disease who either cannot undergo pituitary gland surgery or have undergone the surgery but still have the disease. Available in 1 mg, 5 mg, and 10 mg film-coated tablets, the drug acts as a potent oral 11-beta-hydroxylase inhibitor -- the enzyme involved in the last step of cortisol synthesis.

        Osilodrostat is taken orally twice daily, once in the morning and once in the evening.

        Approval was based upon findings from the LINC-3 and LINC-4 trials, which found osilodrostat was able to normalize cortisol levels in 53% of patients, based on mean 24-hour urinary free cortisol (UFC) concentrations. During an initial 10-week randomization phase, 86% of patients maintained their complete cortisol response if they remained on osilodrostat versus only 29% of those who were switched to placebo.

        As expected, 77.4% of the 137 adults included in the trial were women. The median participant age was 40 and about 47 months had passed since their initial diagnosis. A total of 87.6% underwent previous pituitary surgery and 16.1% underwent previous pituitary irradiation. At baseline, median and mean 24-hour UFC levels were 3.5 nmol and 7.3 nmol, respectively, based on two or three urine samples.

        Participants had an average body weight of 176.4 lb, body mass index (BMI) of 30, and 41 in waist circumference at baseline. Throughout the trial, all measures dropped, reaching the nadir at week 72: body weight of 165 lb, BMI of 27, and 37.8 in waist circumference.

        The most common side effects reported with the agent include adrenal insufficiency, fatigue, nausea, headache, and edema.

         
        • author['full_name']

          Kristen Monaco is a staff writer, focusing on endocrinology, psychiatry, and nephrology news. Based out of the New York City office, she’s worked at the company since 2015.

        Disclosures

        The study was supported by Recordati AG.

        Pedroncelli reported employment with Recordati.

        • Like 1
      8. Published: May 15, 2022 (see history)

        DOI: 10.7759/cureus.25017

        Cite this article as: Fernandez C, Bhatia S, Rucker A, et al. (May 15, 2022) Intermittent Blurry Vision: An Unexpected Presentation of Cushing’s Syndrome Due to Primary Bilateral Macronodular Adrenal Hyperplasia (PBMAH). Cureus 14(5): e25017. doi:10.7759/cureus.25017


        Abstract

        Cushing’s syndrome (CS) is an uncommon endocrine disorder resulting from prolonged exposure to elevated glucocorticoids, with 10-15 million annual cases per the American Association of Neurological Surgeons. Exogenous and endogenous causes can further be divided into adrenocorticotropic hormone (ACTH) dependent (i.e Cushing’s Disease) or ACTH independent. ACTH-independent CS can be caused by primary bilateral macronodular adrenal hyperplasia (PBMAH) representing less than 1% cases of CS. We report a case of a woman presenting with chronic resistant hypertension, episodic blurry vision, weight gain and wasting of extremities. She was diagnosed with Cushing’s syndrome due to PBMAH.

        Our patient’s presentation was unusual as she presented at 40 years old, 10 years earlier than expected for PBMAH; and primarily with complaints of episodic blurry vision. Her symptoms also progressed rapidly as signs and symptoms largely presented over the course of 12 months, however responded well to surgical resection. 

        Introduction

        Cushing’s syndrome (CS) is an uncommon endocrine disorder caused by prolonged exposure to elevated glucocorticoids [1]. There are exogenous or endogenous causes. The National Institute of Health’s (NIH) Genetic and Rare Diseases Information Center (GARD) estimated the prevalence of endogenous CS to be 1 in 26,000 [2]. According to a large study, the annual incidence of CS in individuals less than 65 years old was nearly 49 cases per million [3]. Cushing’s disease (CD), which is defined as Cushing’s syndrome caused by an adrenocorticotropic hormone (ACTH)-secreting pituitary tumor, accounts for approximately 80% of patients with CS; whereas ACTH-independent CS accounts for the remaining 20% [4]. Among the causes of pituitary ACTH-independent CS is bilateral macronodular adrenal hyperplasia which is rare, comprising less than 1% of patients with CS [5]. Herein is a case of rapid onset Cushing’s syndrome due to PBMAH initially presenting as episodes of bilateral blurry vision.

        Case Presentation

        The patient is a 40-year-old female with a past medical history of resistant hypertension (on four agents), and recently diagnosed type 2 diabetes mellitus (started on insulin regimen). Patient was recently seen by her primary care provider, with complaints of intermittent episodes of blurry vision going on for months. 

        As part of evaluation in December 2020, the patient underwent a renal ultrasound as part of evaluation by the primary physician for uncontrolled hypertension. The doppler incidentally showed an indeterminate hypoechoic mass on the right kidney and presumably located within the right adrenal gland, measuring 3.4 x 5.4 cm, without sonographic evidence of renal artery stenosis. The left kidney appeared normal. She was recommended to have further evaluation with contrast enhanced MR or CT with adrenal protocol.

        In January 2021, the patient was sent from her PCP’s office to the ED as the patient was having blurred vision. She had a plain CT scan of the brain that was unremarkable. The patient's systolic blood pressure was in the 160s-170s mm Hg upon arrival to ED compliance with home medications of 5mg of amlodipine daily, 25mg of metoprolol succinate daily, 100mg of losartan daily, and 25mg of hydrochlorothiazide daily. Physical exam reported obesity without evidence of abdominal striae. Blood work in the ED showed elevated blood glucose level over 600 (mg/dL) despite being on a regimen of lantus 60 units, metformin 1000mg twice a day, and semaglutide SQ weekly. Hemoglobin A1c was greater than 15.5%, and vitamin D was low (15.6 ng/mL). The morning ACTH was low (<5pg/mL) (nAM levels: 7.2 - 63.3 pg/mL), AM cortisol was high at 26.1 ug/ml (normal: 5.0 - 23.0 ug/mL), plasma aldosterone was normal at 4.2 ng/dL with a normal plasma renin at 1.96 (0.25 - 5.82 ng/mL/h). 24-hour urine free cortisol (UFC) was high at 1299.5 (4.0-50.0 mcg/24h). CT of the abdomen/pelvis with and without contrast showed low-attenuation masses (less than 5 Hounsfield units) present in both adrenal glands measuring 6.9 x 5.3 cm on the right and 4.5 x 3.9 cm on the left, and did not demonstrate significant arterial enhancement (Figure 1). MR imaging of the abdomen without and with contrast was also obtained and showed the same masses of the bilateral adrenal glands, with largest on the left measured 3.6 cm and largest on the right measured 3.7 cm, as well as mild fatty infiltration of the liver. General surgery and hematology/oncology were consulted and recommendations were made for outpatient follow-up with PCP and endocrinology. 

        CT-of-the-abdomen/pelvis-with-contrast-showing-low-attenuation-masses-present-in-both-adrenal-glands-measuring-6.9-x-5.3-cm-on-the-right-(dark-gray-arrow)-and-4.5-x-3.9-cm-on-the-left-(light-gray-arrow)
        Figure 1: CT of the abdomen/pelvis with contrast showing low-attenuation masses present in both adrenal glands measuring 6.9 x 5.3 cm on the right (dark gray arrow) and 4.5 x 3.9 cm on the left (light gray arrow)
         
         

        In early February 2021, the patient again presented to the ED complaining of recurrent episodes of bilateral blurry vision. Examination was unremarkable, including an ophthalmological exam with slit lamp exam. Blurred vision was suspected to be due to osmotic swelling in the setting of severe hyperglycemia as the patient had persistently uncontrolled blood sugars. Recommendations were for tighter control of blood glucose, and follow-up with primary care and ophthalmology. 

        Patient followed up with the endocrinologist in mid-February to which the patient reported first noticing a difference in her energy and changes to her weight around one year prior. She communicated a weight gain of 30 to 40 lbs over the past year. Patient had a reported history of gestational hypertension diagnosed five years ago when she gave birth to her daughter, which was steadily worsening over the past year. She reported intermittent myalgias and easy bruising. Patient had no family history or any apparent features to suggest multiple endocrine neoplasia (MEN) syndrome. Blood work revealed ACTH less than 1.5 pg/mL, AM cortisol was high at 24.5 mcg/dL, and normal aldosterone at 3.6 ng/dL, with normal renin and metanephrine levels. Physical examination revealed truncal obesity as well as a round face, cushingoid in appearance, and relatively thin extremities and abdominal striae.

        She was then referred to a surgical specialist, and it was decided that she would undergo laparoscopic bilateral adrenalectomy due to severe Cushing’s syndrome. The surgical pathology report revealed macro-nodular cortical hyperplasia of both left and right adrenal gland masses with random endocrine atypia. The largest nodule on the left measured 4.5 cm and the largest nodule on the right measured 6.6 cm. Post-operatively she was started on hydrocortisone 20 mg every morning and 10 mg every evening, and fludrocortisone 0.1 mg twice a day as part of her steroid replacement regimen. Eventually she changed to hydrocortisone 10 mg three times a day and fludrocortisone 0.1 mg once a day. For her diabetes, her insulin glargine decreased from 60 units to 20 units. Amlodipine and hydrochlorothiazide were discontinued from her antihypertensive medications; she continued losartan and metoprolol. Follow up blood work showed stable electrolytes with potassium 4.2 mmol/L (3.5-5.2 mmol/L), sodium 137 mmol/L (134-144mmol/L), chloride 100 mmol/L (96-106 mmol/L), and carbon dioxide 23 mmol/L (20-29mmol/L).

        Discussion

        ACTH-independent Cushing’s syndrome due to bilateral cortisol-secreting nodules is rare, accounting for 2% of CS cases. The majority of causes include primary bilateral macronodular adrenal hyperplasia (PBMAH), primary pigmented nodular adrenocortical disease (PPNAD), and bilateral adrenocortical adenomas (BAA). In PBMAH, typically patients are diagnosed within the fifth or sixth decade of life [4]. The usual age of onset for PPNAD is within the first to third decade of life, with median age in the pediatric population at age 15 years [6]. BAA is such a rare entity that there exists little epidemiological data with less than 40 reported cases until 2019 [7]. A small subset of patients present with overt clinical symptoms of CS, as hypercortisolism often follows an insidious course that can delay diagnosis from years to decades, with one series reporting a diagnostic delay of approximately eight years [8]. Serum and urine hormone screening in the right clinical setting can provide clues to these endocrine disorders, however diagnosis of ACTH-independent CS often occurs incidentally wherein a radiographic study was done for reasons other than to identify adrenal disease [9]. CT or MRI alone are not able to differentiate these disease entities, requiring pathological examination for final determination [7]. Adrenal venous sampling (AVS) and I-6B-iodomethyl-19-norcholesterol (I-NP-59) can aid in identifying hormone-secreting status of each adrenal lesion, however usefulness is debated among experts [10-12].

        In all cases the end goal is to normalize adrenocortical hormones, and PBMAH primarily involves surgical resection with exogenous hormone replacement. Bilateral adrenalectomy is generally the treatment of choice with overt Cushing syndrome regardless of cortisol level. These patients require lifelong steroid administration [9,13]. Another approach is unilateral adrenalectomy of the larger or more metabolically active gland, which can be identified after AVS or I-NP-59 testing. This has been proposed in order to preserve some autonomous hormonal production and prevent adrenal crisis, however remission rates of Cushing syndrome as high as 84% have been reported with eventual need for bilateral adrenalectomy [7,8,14]. Steroid enzyme inhibition to control cortisol secretion has been used as an adjunct before surgery. In some patients with identified aberrant adrenal hormone receptors, targeted pharmacological inhibition remains an alternative medical approach [8]. Despite these alternatives to surgery, surgical resection remains the optimal approach [1].

        Conclusions

        ACTH-independent Cushing’s syndrome due to PBMAH usually presents as an indolent course, with typical diagnosis in the fifth to sixth decade. As the use of imaging for other non-endocrine related investigations becomes more utilized, PBMAH being less of a rare entity. Clinical presentation usually dictates the timing of and type of surgical intervention. Although there are some reports of unilateral resection resulting in a cure, many of these cases eventually proceed to staged bilateral resection. Our patient’s presentation as her primary complaint was recurrent episodes of blurry vision that were suspected to be due to osmotic swelling because of her uncontrolled hyperglycemia. Her case was also unusual as she presented at 40 years old, an average of 10 years earlier than is typically diagnosed for PBMAH. Her symptoms also progressed rapidly over the course of 12 months with development of resistant hypertension and insulin-dependent diabetes requiring high basal insulin. Following surgical resection, her antihypertensive regimen was de-escalated and had significant reduction in insulin requirements, and was maintained on adrenocorticoid therapy.


        References

        1. Nieman LK: Recent updates on the diagnosis and management of Cushing's syndrome. Endocrinol Metab (Seoul). 2018, 33:139-46. 10.3803/EnM.2018.33.2.139
        2. Rare Disease Database: Cushing Syndrome. (2021). Accessed: 12/17/2021: https://rarediseases.org/rare-diseases/cushing-syndrome/.
        3. Broder MS, Neary MP, Chang E, Cherepanov D, Ludlam WH: Incidence of Cushing's syndrome and Cushing's disease in commercially-insured patients <65 years old in the United States. Pituitary. 2015, 18:283-9. 10.1007/s11102-014-0569-6
        4. Lacroix A, Feelders RA, Stratakis CA, Nieman LK: Cushing’s syndrome. Lancet. 2015, 386:913-27. 10.1016/S0140-6736(14)61375-1
        5. Tokumoto M, Onoda N, Tauchi Y, et al.: A case of adrenocoricotrophic hormone -independent bilateral adrenocortical macronodular hyperplasia concomitant with primary aldosteronism. BMC Surg. 2017, 17:97. 10.1186/s12893-017-0293-z
        6. Stratakis CA: Cushing syndrome caused by adrenocortical tumors and hyperplasias (corticotropin- independent Cushing syndrome). Endocr Dev. 2008, 13:117-32. 10.1159/000134829
        7. Gu YL, Gu WJ, Dou JT, et al.: Bilateral adrenocortical adenomas causing adrenocorticotropic hormone-independent Cushing's syndrome: a case report and review of the literature. World J Clin Cases. 2019, 7:961-71. 10.12998/wjcc.v7.i8.961
        8. Lacroix A: ACTH-independent macronodular adrenal hyperplasia. Best Pract Res Clin Endocrinol Metab. 2009, 23:245-59. 10.1016/j.beem.2008.10.011
        9. Sweeney AT, Srivoleti P, Blake MA: Management of the patient with incidental bilateral adrenal nodules. J Clin Transl Endocrinol Case Rep. 2021, 20:100082. 10.1016/j.jecr.2021.100082
        10. Lumachi F, Zucchetta P, Marzola MC, Bui F, Casarrubea G, Angelini F, Favia G: Usefulness of CT scan, MRI and radiocholesterol scintigraphy for adrenal imaging in Cushing's syndrome. Nucl Med Commun. 2002, 23:469-73. 10.1097/00006231-200205000-00007
        11. Builes-Montaño CE, Villa-Franco CA, Román-Gonzalez A, Velez-Hoyos A, Echeverri-Isaza S: Adrenal venous sampling in a patient with adrenal Cushing syndrome. Colomb Med (Cali). 2015, 46:84-7.
        12. Guo YW, Hwu CM, Won JG, Chu CH, Lin LY: A case of adrenal Cushing's syndrome with bilateral adrenal masses. Endocrinol Diabetes Metab Case Rep. 2016, 2016:150118. 10.1530/EDM-15-0118
        13. Wei J, Li S, Liu Q, et al.: ACTH-independent Cushing's syndrome with bilateral cortisol-secreting adrenal adenomas: a case report and review of literatures. BMC Endocr Disord. 2018, 18:22. 10.1186/s12902-018-0250-6
        14. Osswald A, Quinkler M, Di Dalmazi G, et al.: Long-term outcome of primary bilateral macronodular adrenocortical hyperplasia after unilateral adrenalectomy. J Clin Endocrinol Metab. 2019, 104:2985-93. 10.1210/jc.2018-02204

        From https://www.cureus.com/articles/90069-intermittent-blurry-vision-an-unexpected-presentation-of-cushings-syndrome-due-to-primary-bilateral-macronodular-adrenal-hyperplasia-pbmah

        • Like 1
      ×
      ×
      • Create New...