Jump to content

Leaderboard

Popular Content

Showing content with the highest reputation since 04/28/2023 in all areas

  1. And today, we talk about pink jeeps and ziplines... How in the world did we get here in a Cushing's Challenge? I'm sliding these in because earlier I linked (possibly!) my growth hormone use as a cause of my cancer - and I took the GH due to Cushing's issues. Clear? LOL http://cushieblog.files.wordpress.com/2012/04/pink-jeep.jpg?w=300&h=225 I had found out that I had my kidney cancer on Friday, April 28, 2006 and my surgery on May 9, 2006. I was supposed to go on a Cushie Cruise to Bermuda on May 14, 2006. My surgeon said that there was no way I could go on that cruise and I could not postpone my surgery until after that cruise. I got out of the hospital on the day that the other Cushies left for the cruise and realized that I wouldn't have been much (ANY!) fun and I wouldn't have had any. An especially amusing thread from that cruise is The Adventures of Penelopee Cruise (on the Cushing's Help message boards). Someone had brought a UFC jug and decorated her and had her pose around the ship. The beginning text reads: Penelopee had a lovely time on Explorer of the Seas which was a five day cruise to Bermuda. She needed something to cheer her up since her brother, Tom, went off the deep end, but that's another story! Penelopee wanted to take in all of the sights and sounds of this lovely vessel. Every day she needed to do at least one special thing. Being a Cushie, she didn't have enough spoons to do too much every day. On the first day, she went sunning on the Libido deck......she didn't last too long, only about 10 minutes. Goodness, look at her color! Do you think maybe her ACTH is too high? Although I missed this trip, I was feeling well enough to go to Sedona, Arizona in August 2006. I convinced everyone that I was well enough to go off-road in a pink jeep, DH wanted to report me to my surgeon but I survived without too much pain and posed for the header image. In 2009, I figured I had “extra years” since I survived cancer and I wanted to do something kinda scary, yet fun. So, somehow, I decided on ziplining. Tom wouldn’t go with me but Michael would so I set this up almost as soon as we booked a Caribbean cruise to replace the Cushie Cruise to Bermuda. Each person had a harness around their legs with attached pulleys and carabiners. Women had them on their chests as well. In addition, we had leather construction gloves and hard hats. We climbed to the top of the first platform and were given brief instructions and off we went. Because of the heavy gloves, I couldn’t get any pictures. I had thought that they would take some of us on the hardest line to sell to us later but they didn't. They also didn’t have cave pictures or T-Shirts. What a missed opportunity! This was so cool, so much fun. I thought I might be afraid at first but I wasn’t. I just followed instructions and went. Sometimes they told us to brake. We did that with the right hand, which was always on the upper cable. After the second line, I must have braked too soon because I stopped before I got to the platform. Michael was headed toward me. The guide on the end of the platform wanted me to do some hand over hand maneuver but I couldn’t figure out what he was saying so he came and got me by wrapping his legs around me and pulling me to the platform. After that, no more problems with braking! The next platform was very high – over 70 feet in the air – and the climb up was difficult. It was very hot and the rocks were very uneven. I don’t know that I would have gotten to the next platform if Michael hadn’t cheered me on all the way. We zipped down the next six lines up to 250-feet between platforms and 85-feet high in the trees, at canopy level. It seemed like it was all over too soon. But, I did it! No fear, just fun. Enough of adventures - fun ones like these, and scary ones like transsphenoidal surgery and radical nephrectomy!
    1 point
  2. ABSTRACT Objective Onset and exacerbation of autoimmune, inflammatory or steroid-responsive conditions have been reported following the remission of Cushing syndrome, leading to challenges in distinguishing a new condition versus expected symptomatology following remission. We describe a case of a 42-year-old man presenting with new-onset sarcoidosis diagnosed 12 months following the surgical cure of Cushing syndrome and synthesise existing literature reporting on de novo conditions presenting after Cushing syndrome remission. Methods A scoping review was conducted in Medline, Epub, Ovid and PubMed. Case reports and case series detailing adult patients presenting with new-onset conditions following Cushing syndrome remission were included. Results In total, 1641 articles were screened, 138 full-text studies were assessed for eligibility, and 43 studies were included, of which 84 cases (including our case) were identified. Most patients were female (85.7%), and the median reported age was 39.5 years old (IQR = 13). Thyroid diseases were the most commonly reported conditions (48.8%), followed by sarcoidosis (15.5%). Psoriasis, lymphocytic hypophysitis, idiopathic intracranial hypertension, multiple sclerosis, rheumatoid arthritis, lupus and seronegative arthritis were reported in more than one case. The median duration between Cushing remission and de novo condition diagnosis was 4.1 months (IQR = 3.75). Of those patients, 59.5% were receiving corticosteroid therapy at the time of onset. Conclusion Our scoping review identified several cases of de novo conditions emerging following the remission of Cushing syndrome. They occurred mostly in women and within the year following remission. Clinicians should remain aware that new symptoms, particularly in the first year following the treatment of Cushing syndrome, may be manifestations of a wide range of conditions aside from adrenal insufficiency or glucocorticoid withdrawal syndrome. 1 Introduction Cushing syndrome (CS) is caused by chronic exposure to excessive glucocorticoids, from either endogenous or exogenous sources [1]. Endogenous Cushing syndrome can be classified as either adrenocorticotropic hormone (ACTH) dependent or independent. ACTH-dependent causes comprise 80% of cases, most of which are pituitary corticotroph adenomas. Unilateral adrenal adenomas are the most common ACTH-independent cause, comprising 20% of total cases [2]. Treatment focuses on controlling tissue exposure to cortisol and treating the source of cortisol overproduction, which can be achieved through surgical resection, radiation or medical therapy depending on the underlying aetiology [2]. Following the biochemical remission of Cushing syndrome, patients commonly feel unwell due to adrenal insufficiency (AI) and/or glucocorticoid withdrawal syndrome (GWS). AI is an expected consequence of remission due to the chronic suppression of the hypothalamic-pituitary-adrenal (HPA) axis from glucocorticoid excess and can manifest with heterogeneous symptoms including myalgias, muscle weakness, fatigue, hypersomnolence, anorexia, nausea and abdominal discomfort [3-5]. GWS is due to the dependence on supraphysiologic glucocorticoid levels and has overlapping symptoms with AI, but occurs even with physiologic or supraphysiologic glucocorticoid replacement [5]. Both AI and GWS can persist for 1 year or longer following the remission of Cushing syndrome [5]. Due to immunosuppression induced by glucocorticoid excess [1, 6, 7], the remission of Cushing syndrome has the potential to unmask or aggravate an underlying autoimmune, inflammatory or steroid-responsive condition. Reports of such conditions include thyroiditis, psoriasis, sarcoidosis and systemic lupus erythematosus (SLE) [8-11]. Therefore, persisting symptoms following the remission of Cushing syndrome can be due to AI, GWS or presentation of a new condition. The latter situation may evade timely diagnosis since AI and GWS are expected consequences in this clinical setting. We report a case of a 42-year-old patient with Cushing syndrome secondary to an adrenal adenoma with first presentation of sarcoidosis 12 months after adrenalectomy. We performed a scoping review to synthesise previous reports of de novo autoimmune, inflammatory or steroid-responsive conditions following the remission of Cushing syndrome. Our aim was to characterise these presentations to provide guidance to clinicians in making this diagnosis challenging. 2 Case Report A 42-year-old white man was referred to endocrinology with a 1-year history of insomnia and rapid weight gain of 18 kg. Past medical history was significant for a pituitary lesion presumed to be a Rathke's cleft cyst, which had been stable on neuroimaging for over two decades. He was otherwise healthy with no prescribed medications. On physical examination, blood pressure was 159/99 mmHg. Pertinent findings included facial plethora, dorsal and supraclavicular fat pads, reduced skin thickness and multiple violaceous striae on the abdomen. Biochemistry showed elevated 24-h urine cortisol on two occasions (3067.5 nmol/day, 2704.0 nmol/day; reference range, 100.0–380.0 nmol/day) and elevated late-night salivary cortisol (54.2 nmol/L; reference range, ≤ 3.6 nmol/L). Plasma ACTH level was suppressed (< 1.1 pmol/L; reference range, 2.0–11.5 pmol/L). Serum-free thyroxine (fT4), thyroid-stimulating hormone (TSH), follicle-stimulating hormone (FSH), luteinising hormone (LH) and free testosterone were all within normal limits. Serum random glucose level was normal (4.6 mmol/L; reference range, 3.3–11.0 mmol/L), and haemoglobin A1c (HbA1c) was within the pre-diabetes range at 6.2% (6.0%–6.4%). His serum complete blood count, sodium, potassium and creatinine levels were all within normal limits. His body surface area was 2.53 m2. The patient was diagnosed with ACTH-independent Cushing syndrome. Computed tomography of the abdomen and pelvis revealed a 4.8-cm mass in the left adrenal gland. The patient was referred to endocrine surgery, and in the interim, medical treatment with ketoconazole 200 mg p.o. twice daily and spironolactone 50 mg p.o. daily was initiated, which resulted in normalisation of his 24-h urine cortisol. Shortly after initiating these medications, the patient noticed paraesthesia in his extremities. There was no objective evidence of neuropathy on physical examination, and laboratory investigations including vitamin B12 (329 pmol/L; reference range, 155–700 pmol/L), TSH (2.14 mIU/L) and follow-up HbA1c (5.7%) were within normal range. Three months following his initial presentation, the patient underwent left adrenalectomy. Postoperatively, supraphysiologic glucocorticoids were initiated and he was discharged home on oral hydrocortisone 40 mg in the morning and 20 mg in the afternoon. Pathology was consistent with an adrenal cortical adenoma with Ki-67 < 1%. The patient was highly motivated to wean his glucocorticoid doses to ameliorate symptoms of cortisol excess. He tapered his hydrocortisone to 20 mg in the morning and 10 mg in the late afternoon within 2 weeks postoperatively. He developed significant muscle stiffness to his shoulders, with diffuse myalgias and arthralgias, along with worsening of his pre-existing paraesthesia. Four months after the surgery, he had further reduced his hydrocortisone to 15 mg in the morning and 5 mg in the late afternoon with improvement in his Cushingoid features (reduced supraclavicular fullness, reduced abdominal adiposity, fading of abdominal striae and seven-kilogram weight loss). He was assessed by neurology for his paraesthesia, but no organic cause was identified. Twelve months after surgery, he had weaned off his hydrocortisone to 5 mg twice daily and continued to feel unwell with headaches, muscle weakness and morning stiffness. Morning cortisol after withholding glucocorticoids for 24 h was 35 nmol/L (170–500 nmol/L), demonstrating ongoing HPA axis suppression. The patient's family physician ordered a chest X-ray for a prominent sternoclavicular joint, and the patient was incidentally found to have bilateral hilar lymphadenopathy. The patient was referred to respirology and underwent bronchoscopic sampling of his mediastinal lymph nodes (see Figure 1), which demonstrated well-formed non-necrotising granulomas from lymph node Stations 7 and 11L. Cultures for fungi, AFB and flow cytometry were all negative, confirming Stage 2 pulmonary sarcoidosis. There was no indication for sarcoidosis-specific treatment with glucocorticoids, cytotoxic agents or biologics based on his normal pulmonary function testing and lack of active extra-pulmonary sarcoidosis. However, given the ongoing HPA axis suppression, hydrocortisone was empirically increased to 20 mg total daily dose, which led to improvement in the patient's symptoms. FIGURE 1 Open in figure viewerPowerPoint Enhanced CT scan of the chest demonstrating bilateral hilar and mediastinal lymphadenopathy (indicated by arrows). Due to the ongoing symptoms of headaches and known pituitary lesion potentially concerning for neurosarcoidosis, the patient was referred to neuroimmunology. MRI brain, and cervical, thoracic and lumbar spine showed a reduction in the size of the known cystic pituitary lesion, with no findings suggestive of intracranial or spinal sarcoidosis, and no abnormal leptomeningeal enhancement. Electromyography demonstrated normal nerve conduction studies. Two years following adrenalectomy, the patient has weaned off all glucocorticoid replacement with resolution of his symptoms of adrenal insufficiency. His sarcoidosis remains in remission. 3 Methods A scoping review protocol was developed using the Joanna Briggs Institute methodology [12]. We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) extension for Scoping Reviews guidelines in reporting our protocol and results [13]. 3.1 Systematic Literature Search A preliminary search strategy was developed with the aid of a medical librarian. The full search strategy and terms are presented in Appendix 1. Ovid MEDLINE and Epub Ahead of Print, In-Process, In-Data-Review & Other Non-Indexed Citations and Daily and PubMed databases were searched from inception to 8 September 2022. Additional articles were identified by searching the reference lists of all included articles. 3.2 Eligibility Criteria We considered descriptive observational studies including case series and case reviews, as well as systematic reviews. Articles from all years and locations were included; however, articles written in another language than in English or French were excluded given the limitations in conducting review and data extraction from these sources. Full inclusion and exclusion criteria are shown in Table 1. We included the reports of adults ≥ 18 years of age with endogenous Cushing syndrome with a de novo presentation of an autoimmune, inflammatory or steroid-responsive condition following remission, which could be induced by surgery, radiotherapy, medical therapy or a combination of these treatments. Cases of Cushing syndrome secondary to exogenous corticosteroids were excluded due to the high likelihood of pre-existing steroid-responsive conditions in this population. Flares or recurrences of previously diagnosed inflammatory, autoimmune or steroid-responsive conditions were also excluded. Patients with Cushing syndrome secondary to metastatic cancer (i.e. metastatic corticotroph adenoma or metastatic adrenocortical carcinoma) were excluded. Remission was defined as clinical and/or biochemical evidence of AI following treatment of CS by any modality. TABLE 1. Scoping review inclusion and exclusion criteria. Inclusion criteria Exclusion criteria Studies published in any year and location Studies published in English and French Studies published in another language than in English or French All adults ≥ 18 years old at the time of Cushing syndrome cure Children < 18 years old Endogenous Cushing syndrome Exogenous Cushing syndrome De novo conditions post-remission Pre-existing conditions with flare post-remission Cushing syndrome caused by metastatic cancer 3.3 Study Selection All identified studies were uploaded to Covidence, and duplicate articles were removed. Titles and abstracts were screened for eligibility by one reviewer, and articles without abstracts were screened in totality for eligibility. Selected articles underwent a full-text review by two reviewers for inclusion. Disagreements about eligibility of an article were resolved by a third reviewer. 3.4 Data Extraction Two members of the study team created a data extraction tool to collect patient characteristics from the studies that met eligibility criteria following a full-text review. The data extraction tool was piloted with all study team members, and adjustments were made as needed. Patients' age, gender, aetiology of Cushing syndrome, treatment modality and de novo condition were recorded. Characteristics of de novo conditions were collected including clinical presentation, timing of onset, presence of exogenous steroids at the time of presentation and resolution. Data from all included studies were extracted independently by two study team members and reconciled. Any discrepancies were resolved by referring to the primary article. 3.5 Statistical Analysis In this descriptive study, categorical variables are expressed as percentages and non-normally distributed continuous variables as median and interquartile range (IQR). Median and IQR were preferred over mean and standard deviation given the small sample size. 4 Results The search strategy identified 3123 total citations: 3099 abstracts from database searching and 24 from hand-searching (Figure 2). There were 1641 citations remaining after duplicates were removed. After title and abstract screening, 138 studies underwent full-text review, and 43 studies were included in data extraction and analysis (see Appendix 1 for a full list of included citations). FIGURE 2 Open in figure viewerPowerPoint PRISMA flow diagram of included studies. All included studies were either case reports (n = 34) or case series (n = 9). Five articles [8, 9, 14-16] also included a literature review and four [8, 10, 11, 17] included cohort studies in addition to the case report/series. Included articles were published from 1981 to 2021 inclusively. These 43 studies identified 83 unique patient cases of new-onset conditions following the remission of Cushing syndrome (see Table 2 for full patient characteristics). In addition to our case, this review includes 84 cases. Most patients were female (n = 72, 85.7%), and the median reported age was 39.5 years old (IQR = 13 years old, range, 16–80 years old). TABLE 2. Patients' characteristics. Total cases (n = 84) (% [n]) Age (median [IQR]), years 39.5 (13) Sex Women 85.7 (72) Men 14.3 (12) Aetiology of Cushing syndrome ACTH dependant 71.4 (60) Pituitary source 70.2 (59) Ectopic source 1.2% (1) ACTH independent 28.6 (24) Adrenal adenoma 23.8 (20) Adrenal hyperplasia 4.8 (4) Treatment of Cushing syndromea Surgical resection 97.6 (82) Medical therapy 19.0 (16) Radiation therapy 8.3 (7) Biochemical remission reported 79.8 (67) a Adds up to more than 100% as multiple reasons could be documented. The most common aetiology of CS was pituitary adenoma (n = 59), followed by adrenal adenoma (n = 20) and adrenal hyperplasia (n = 4). One patient had a pulmonary neuroendocrine tumour secreting ACTH [8]. All patients but two underwent surgical resection for definitive management of CS. One patient underwent medical management alone with pasireotide [18], and the other had resolution of CS secondary to an adrenal adenoma following adrenal haemorrhage after a motorcycle collision [14]. All patients included in our analysis had documented clinical remission of hypercortisolism, and biochemical remission was reported in 67 cases (79.8%). The most commonly reported de novo conditions following CS remission were thyroid disorders (n = 41, 48.8%), including 34 cases of thyroiditis [9-11, 17-23] and seven cases of Graves disease [8, 9, 21, 24-26]. Rheumatological disorders were the second most commonly reported conditions (n = 22, 26.2%) with cases of sarcoidosis (n = 13) [8, 14, 27-35], systemic lupus erythematosus (n = 2) [9, 36], rheumatoid arthritis (n = 2) [37, 38], seronegative arthritis (n = 2) [37, 39], polymyalgia rheumatica (n = 1) [40], giant cell arteritis (n = 1) [9] and retinal vasculitis (n = 1) [39] (see Figure 3 and Table 3). Further characterisation of thyroid disorders and sarcoidosis is detailed below. FIGURE 3 Open in figure viewerPowerPoint De novo conditions, by system. TABLE 3. Characteristics of de novo conditions. De novo conditions, by system (n = 84) (% [n]) Thyroid disorder 48.8 (41) Silent thyroiditis 23.8 (20) Hashimoto thyroiditis 13.1 (11) Graves disease 8.3 (7) De Quervain thyroiditis 3.6 (3) Rheumatologic disorder 26.2 (22) Sarcoidosis 15.5 (13) Systemic lupus erythematous 2.4 (2) Rheumatoid arthritis 2.4 (2) Seronegative arthritis 2.4 (2) Polymyalgia rheumatica 1.2 (1) Giant cell arteritis 1.2 (1) Retinal vasculitis 1.2 (1) Neurological disorder 13.1 (11) Idiopathic intracranial hypertension 6.0 (5) Multiple sclerosis 2.4 (2) Lymphocytic hypophysitis 2.4 (2) Myasthenia gravis 1.2 (1) Acute disseminated encephalitis 1.2 (1) Dermatological disorder 8.3 (7) Psoriasis 3.6 (3) Rash 3.6 (3) Generalised rash 1.2 (1) Rosacea-like rash 1.2 (1) Eczematous rash 1.2 (1) Angioedema 1.2 (1) Gastrointestinal disorder 3.6 (3) Celiac disease 1.2 (1) Primary biliary cirrhosis 1.2 (1) Sclerosing pancreatocholangitis 1.2 (1) We identified 11 cases of neurological disorders, including idiopathic intracranial hypertension (IIH) (n = 5) [15, 16, 41-43], multiple sclerosis (n = 2) [44, 45], lymphocytic hypophysitis (n = 2) [46, 47], acute disseminated encephalomyelitis (n = 1) [48] and myasthenia gravis (n = 1) [49]. IIH has been associated with both primary adrenal insufficiency and steroid withdrawal [15]. Glucocorticoids are not routinely used as first-line treatment of IIH (due to the risk of rebound intracranial hypertension upon withdrawal); however, three of the five cases included in this review were successfully treated with higher doses of steroids [15, 16, 41]. Given this association, IIH was considered a steroid-responsive condition for the purpose of this review. Acute disseminated encephalomyelitis is a rare autoimmune disease, causing widespread inflammation of the brain and spinal cord, often associated with preceding viral infection or vaccination. However, as first-line treatment for this condition is high dose corticosteroids, we considered it a steroid-responsive condition and was included in this review [50]. Seven dermatological cases were identified in our scoping review including psoriasis (n = 3) [8, 9], rash (n = 3) [8] and angioedema (n = 1) [51]. Gastrointestinal conditions were the least reported (n = 3) with one case of celiac disease [52], one case of primary biliary cirrhosis [8] and one case of sclerosing pancreatocholangitis [53]. The median reported time between the treatment of CS and the onset of symptoms of de novo condition was 4.1 months (IQR = 3.75 months, range, 10 days to 27 years). Most patients (n = 50, 59.5%) were receiving corticosteroids at the time of onset. Only 22 cases (26.2%) explicitly reported a timeline from discontinuation (n = 6) or tapering (n = 16) of corticosteroid dose to the onset of symptoms, with a median time of 1.75 months (IQR = 3 months, range 7 days-7 months). Thirty-nine patients (46.4%) were subsequently treated with corticosteroids (either re-initiated or at an increased dose). Remission or clinical stability of the de novo condition was reported in 66 cases (78.6%), while seven cases (8.3%) remained uncontrolled, and in 11 cases (13.1%), the outcome was not reported. Of the 44 cases where time to remission was reported, the median time was 3 months (IQR = 4.2 months, range 1–24 months). 4.1 Thyroid Disorder Cases Amongst the seven cases of Graves disease, six patients were women and the median age at onset was 44 years old (IQR = 10.5 years old, range, 33–58 years old). Four patients had a pituitary adenoma, two had an adrenal adenoma and one had unilateral adrenal hyperplasia. They all presented with classical signs and symptoms of this condition such as weight loss, tachycardia, goitre and/or orbitopathy. The median time to onset was 5 months (IQR = 3.55 months, range 2–27 months). The majority (5/7) were not on steroids at the onset of Graves disease, and six required additional treatment with antithyroid medications. Of the 34 cases of thyroiditis, 30 patients were women and the median age at onset was 35.5 years old (IQR = 15.5 years old, range 16–80 years old). Twenty-three patients had a pituitary adenoma, eight had an adrenal nodule and three had adrenal hyperplasia. Twenty patients presented with silent thyroiditis, 11 patients presented with Hashimoto thyroiditis and three patients presented with De Quervain (subacute) thyroiditis with fever, neck pain and malaise. Time to onset ranged from 1 to 9 months, with a median of 4.85 months (IQR = 3 months). Twenty-three patients were on steroids at the time of onset, and all patients with De Quervain thyroiditis (n = 3) and most patients with transient thyrotoxicosis (n = 13) were managed with increased corticosteroid doses. 4.2 Sarcoidosis Cases Amongst the 13 identified sarcoidosis cases, 10 patients were women and the median age at onset was 41 years old (IQR = 9, range 27–45 years old). Eight patients had Cushing disease while five had an adrenal adenoma, and all had undergone surgical resection, except for the patient with adrenal haemorrhage. The time between CS remission and onset ranged from 2 weeks to 17 months, with a median time of 3 months (IQR = 3). Twelve patients had skin manifestations with either painless subcutaneous nodules or erythema nodosum, while our case did not have any skin manifestations. Twelve patients had pulmonary involvement with bilateral mediastinal and/or hilar lymphadenopathy (n = 11) or abnormal pulmonary function test (n = 1). Eleven patients were on corticosteroids at the time of onset, of which four required increased doses, while the other seven patients did not require additional steroids. The remaining two patients who were not receiving corticosteroids were started on them for the management of sarcoidosis. 5 Discussion Our scoping review identified 20 conditions following the remission of CS, suggesting that the resolution of glucocorticoid excess and its associated immunosuppressive effect can unmask these diseases. The majority of cases were female, which is in keeping with the epidemiology of Cushing syndrome [2] as well as of autoimmune disease in the general population [54, 55]. Thyroiditis, sarcoidosis and Graves disease were the most commonly reported conditions. The prevalence of de novo thyroid disorders in our review may reflect that autoimmune and inflammatory thyroid diseases are common in the general population [55-57]. However, detection and publication bias may also play a role, as we presume endocrinologists are more likely to diagnose and report thyroid disorders versus non-endocrine conditions. Though most de novo conditions presented within 1 year of CS remission, the reported timing of onset was variable, ranging from 10 days to 27 years. This may reflect differences in post-remission glucocorticoid doses, weaning schedules and responsiveness of various conditions to glucocorticoids. We emphasise that we cannot prove a causative link between CS remission and the emergence of the de novo condition in our case or the other reported cases. Due to the heterogeneity in glucocorticoid requirements and tapering schedules post-CS remission [58, 59], as well as our aim characterising this clinical presentation, we chose not to specify the timing of the onset of de novo conditions in our inclusion criteria. However, we suggest that the emergence of a condition further out from the withdrawal of supraphysiologic glucocorticoids is less likely to be related to the previous state of hypercortisolism. We are dubious about one case in particular [46] that reported a patient with the onset of lymphocytic hypophysitis 27 years post subtotal adrenalectomy for CS, despite tapering off glucocorticoids within a month of surgery. The second case of lymphocytic hypophysitis occurred 7 years after the remission of Cushing disease, but there is no mention of whether the patient was still on exogenous glucocorticoids at the time of onset [47]. With the exclusion of these two cases, the onset of de novo conditions ranged from 10 days to 60 months, the latter case [8] being the emergence of psoriasis following the delayed normalisation of hypercortisolism with medical therapy and radiotherapy, which is more clinically plausible. Our case highlights the challenge of diagnosing a new systemic disorder when features of AI and/or GWS are concurrently present. To avoid diagnostic delay in this setting, we emphasise that clinicians should have a low threshold to investigate symptoms atypical for AI or GWS including (but not limited to) skin changes, neurological symptoms, pulmonary symptoms and symptoms of thyroid disease, particularly if symptoms present or worsen as supraphysiologic glucocorticoids are weaned. 5.1 Strengths and Limitations To our knowledge, this is the first scoping review to synthesise the existing literature on autoimmune, inflammatory and steroid-responsive conditions following Cushing syndrome remission. We adhered to PRISMA scoping review methodology and developed a comprehensive literature search strategy. However, we limited our review to publications in English and French, which resulted in the exclusion of 17 articles. The reported cases are subject to diagnostic and publication bias; therefore, our review may not encompass all de novo conditions that can present in this setting. As outlined above, we cannot establish a causative link between the remission of CS and the emergence of the reported de novo conditions. 6 Conclusion Our scoping review identified several cases of distinct autoimmune, inflammatory or steroid-responsive conditions emerging following the remission of Cushing syndrome, amongst which thyroid disorders and sarcoidosis were the most commonly reported. Delineating such conditions from the expected clinical course of GWS and/or AI can be a challenge; therefore, clinicians should have a low threshold to investigate any atypical symptoms following the remission of Cushing syndrome. Author Contributions Noémie Desgagnés: Conceptualization (equal); data curation (equal); formal analysis (equal); investigation (equal); methodology (equal); project administration (equal); visualization (equal); writing – original draft (equal). Laura Senior: Data curation (equal); formal analysis (equal); investigation (equal); writing – original draft (equal). Daniel Vis: Writing – review and editing (equal). Katayoun Alikhani: Writing – review and editing (equal). Kirstie Lithgow: Conceptualization (equal); data curation (equal); investigation (equal); methodology (equal); project administration (equal); supervision (lead); writing – review and editing (lead). Acknowledgements The authors thank Dr. Kevin Baird for his feedback and contributions to the manuscript. Conflicts of Interest The authors have declared no conflicts of interest. Appendix 1: MEDLINE search strategy # Query Results 1 exp Cushing Syndrome/ 12,803 2 cushing* syndrome.tw,kf. 10,669 3 cushing* disease.tw,kf. 5341 4 1 or 2 or 3 18,042 5 (de novo adj2 steroid*).tw,kf. 160 6 exp Sarcoidosis/ 26,609 7 sarcoid*.tw,kf. 29,517 8 exp Polymyalgia Rheumatica/ 2725 9 PMR.tw,kf. 3328 10 polymyalgia rheumatica.tw,kf. 2890 11 exp Multiple Sclerosis/ 67,479 12 MS.tw,kf. 395,579 13 multiple sclerosis.tw,kf. 87,101 14 exp Autoimmune diseases/ 527,961 15 autoimmun*.tw,kf. 199,531 16 exp Systemic lupus erythematosus/ 65,187 17 SLE.tw,kf. 38,508 18 systemic lupus erythematosus.tw,kf. 56,501 19 exp Rheumatoid arthritis/ 122,521 20 RA.tw,kf. 86,402 21 rheumatoid arthritis.tw,kf. 117,236 22 arthritis*.tw,kf. 201,619 23 exp Sjogren syndrome/ 14,144 24 sjogren syndrome.tw,kf. 3325 25 exp celiac disease/ 21,506 26 celiac disease.tw,kf. 13,896 27 exp myasthenia gravis/ 16,576 28 myasthenia gravis.tw,kf. 16,140 29 exp Crohn disease/ 43,066 30 crohn disease.tw,kf. 5001 31 crohn*.tw,kf. 54,352 32 exp Ulcerative colitis/ 39,053 33 ulcerative colitis.tw,kf. 46,316 34 UC.tw,kf. 25,420 35 colitis*.tw,kf. 77,427 36 exp dermatitis/ 112,602 37 dermatitis.tw,kf. 68,667 38 exp vasculitis/ 102,088 39 vasculitis.kw,kf. 6283 40 exp myositis/ 21,858 41 exp thyroiditis/ 15,345 42 thyroid*.tw,kf. 216,663 43 exp IgG4/ 154,671 44 igg4.tw,kf. 10,905 45 exp encephalopathy/ 1,361,632 46 encephalopathy.tw,kf. 53,011 47 steroid responsive.tw,kf. 1576 48 5 or 6 or 7 or 8 or 9 or 10 or 11 or 12 or 13 or 14 or 15 or 16 or 17 or 18 or 19 or 20 or 21 or 22 or 23 or 24 or 25 or 26 or 27 or 28 or 29 or 30 or 31 or 32 or 33 or 34 or 35 or 36 or 37 or 38 or 39 or 40 or 41 or 42 or 43 or 44 or 45 or 46 or 47 3,122,540 49 4 and 48 5865 50 exp Case Reports/ 2,289,770 51 case report*.tw,kf. 466,902 52 exp Observational Study/ 132,022 53 observational stud*.tw,kf. 147,088 54 case series.tw,kf. 96,054 55 50 or 51 or 52 or 53 or 54 2,676,537 56 4 and 48 and 55 1479 From https://onlinelibrary.wiley.com/doi/10.1002/edm2.476
    1 point
  3. Over the years, we went on several Windjammer Barefoot Cruises. We liked them because they were small, casual and were fairly easy on the wallet. They sailed around the Caribbean to a variety of islands, although they sometimes changed itineraries depending on weather, crew, whatever. One trip we were supposed to go to Saba but couldn't make port. A lot of people got off at the next port and flew home. The captains were prone to "Bedtime Stories" which were often more fiction than true but they added to the appeal of the trip. We didn't care if we missed islands or not - we were just there to sail over the waves and enjoy the ride. The last trip we took with them was about two years before I started having Cushing's problems. (You wondered how I was going to tie this together, right?) The cruise was uneventful, other than the usual mishaps like hitting docks, missing islands and so on. Until it was a particularly rough sea one day. I was walking somewhere on deck and suddenly a wave came up over the deck making it very slippery. I fell and cracked the back of my head on the curved edge of a table in the dining area. I had the next-to-the-worse headache I have ever had, the worst being after my pituitary surgery. At least after the surgery, I got some morphine. We asked several doctors later if that hit could have contributed to my Cushing's but doctors didn't want to get involved in that at all. The Windjammer folks didn't fare much better, either. In October 1998, Hurricane Mitch was responsible for the loss of the s/v Fantome (the last one we were on). All 31 crew members aboard perished; passengers and other crew members had earlier been offloaded in Belize. The story was recorded in the book The Ship and the Storm: Hurricane Mitch and the Loss of the Fantome by Jim Carrier. The ship, which was sailing in the center of the hurricane, experienced up to 50-foot (15 m) waves and over 100 mph (160 km/h) winds, causing the Fantome to founder off the coast of Honduras. "In October 1998, the majestic schooner Fantome came face-to-face with one of the most savage storms in Atlantic history. The last days of the Fantome are reconstructed in vivid and heartbreaking detail through Jim Carrier's extensive research and hundreds of personal interviews. What emerges is a story of courage, hubris, the agony of command, the weight of lives versus wealth, and the advances of science versus the terrible power and unpredictability of nature." This event was similar to the Perfect Storm in that the weather people were more interested in watching the hurricane change directions than they were in people who were dealing with its effects. I read this book and I was really moved by the plight of those crew members. I'll never know if that hit on my head contributed to my Cushing's but I have seen several people mention on the message boards that they had a traumatic head injury of some type in their earlier lives.
    1 point
  4. In 2021, I had the bestest ever day... and, surpassing that:
    1 point
  5. Abstract Cushing syndrome (CS) is a rare endocrinological disorder resulting from chronic exposure to excessive cortisol. The term Cushing disease is used specifically when this is caused by excessive secretion of adrenocorticotropic hormone (ACTH) by a pituitary tumor, usually an adenoma. This disease is associated with a poor prognosis, and if left untreated, it has an estimated 5-year survival rate of 50%. We present the case of a 66-year-old female patient who received a referral to endocrinology for an evaluation of obesity due to right knee arthropathy. Taking into consideration her age, she was screened for osteoporosis, with results that showed diminished bone density. Considering this, combined with other clinical features of the patient, suspicion turned toward hypercortisolism. Laboratory findings suggested that the CS was ACTH-dependent and originated in the pituitary gland. After a second look at the magnetic resonance imaging results, a 4-mm lesion was identified on the pituitary gland, prompting a transsphenoidal resection of the pituitary adenoma. case report, pituitary ACTH hypersecretion, Cushing syndrome, obesity Issue Section: Case Report Introduction Chronic excessive exposure to glucocorticoids leads to the diverse clinical manifestations of Cushing syndrome (CS), which has an annual incidence ranging from 1.8 to 3.2 cases per million individuals [1]. The syndrome's signs and symptoms are not pathognomonic, and some of its primary manifestations, such as obesity, hypertension, and glucose metabolism alterations, are prevalent in the general population [2], making diagnosis challenging. Endogenous CS falls into 2 categories: adrenocorticotropic hormone (ACTH)-dependent (80%-85% of cases), mostly due to a pituitary adenoma, or ACTH-independent (15%-20% of cases), typically caused by adrenal adenomas or hyperplasia [3]. Cushing disease (CD) represents a specific form of CS, characterized by the presence of an ACTH-secreting pituitary tumor [1]. Untreated CD is associated with high morbidity and mortality compared to the general population [1], with a 50% survival rate at 5 years [2]. However, surgical removal of a pituitary adenoma can result in complete remission, with mortality rates similar to those of the general population [2]. This article aims to highlight the challenges of suspecting and diagnosing CD and to discuss the current management options for this rare condition. Case Presentation A 66-year-old woman received a referral to endocrinology for an evaluation of obesity due to right knee arthropathy. During physical examination, she exhibited a body mass index of 34.3 kg/m2, blood pressure of 180/100, a history of non-insulin-requiring type 2 diabetes mellitus with glycated hemoglobin (HbA1c) of 6.9% (nondiabetic: < 5.7%; prediabetic: 5.7% to 6.4%; diabetic: ≥ 6.5%) and hypertension. Additionally, the patient complained of proximal weakness in all 4 limbs. Diagnostic Assessment Upon admission, densitometry revealed osteoporosis with T scores of −2.7 in the lumbar spine and −2.8 in the femoral neck. Hypercortisolism was suspected due to concomitant arterial hypertension, central obesity, muscle weakness, and osteoporosis. Physical examination did not reveal characteristic signs of hypercortisolism, such as skin bruises, flushing, or reddish-purple striae. Late-night salivary cortisol (LNSC) screening yielded a value of 8.98 nmol/L (0.3255 mcg/dL) (reference value [RV] 0.8-2.7 nmol/L [0.029-0.101 mcg/dL]) and ACTH of 38.1 pg/mL (8.4 pmol/L) (RV 2-11 pmol/L [9-52 pg/mL]). A low-dose dexamethasone suppression test (LDDST) was performed (cutoff value 1.8 mcg/dL [49 nmol/L]), with cortisol levels of 7.98 mcg/dL (220 nmol/L) at 24 hours and 20.31 mcg/dL (560 nmol/L) at 48 hours. Subsequently, a high-dose dexamethasone suppression test (HDDST) was conducted using a dose of 2 mg every 6 hours for 2 days, for a total dose of 16 mg, revealing cortisol levels of 0.0220 nmol/L (0.08 ng/mL) at 24 hours and 0.0560 nmol/L (0.0203 ng/mL) at 48 hours, alongside 24-hour urine cortisol of 0.8745 nmol/L (0.317 ng/mL) (RV 30-145 nmol/24 hours [approximately 11-53 μg/24 hours]) [4]. These findings indicated the presence of endogenous ACTH-dependent hypercortisolism of pituitary origin. Consequently, magnetic resonance imaging (MRI) was requested, but the results showed no abnormalities. Considering ectopic ACTH production often occurs in the lung, a high-resolution chest computed tomography scan was performed, revealing no lesions. Treatment Upon reassessment, the MRI revealed a 4-mm adenoma, prompting the decision to proceed with transsphenoidal resection of the pituitary adenoma. Outcome and Follow-Up The histological analysis revealed positive staining for CAM5.2, chromogranin, synaptophysin, and ACTH, with Ki67 staining at 1%. At the 1-month follow-up assessment, ACTH levels were 3.8 pmol/L (17.2 pg/mL) and morning cortisol was 115.8621 nmol/L (4.2 mcg/dL) (RV 5-25 mcg/dL or 140-690 nmol/L). Somatomedin C was measured at 85 ng/mL (RV 70-267 ng/mL) and prolactin at 3.5 ng/mL (RV 4-25 ng/mL). At the 1-year follow-up, the patient exhibited a satisfactory postoperative recovery. However, she developed diabetes insipidus and secondary hypothyroidism. Arterial hypertension persisted. Recent laboratory results indicated a glycated hemoglobin (HbA1c) level of 5.4%. Medications at the time of follow-up included prednisolone 5 milligrams a day, desmopressin 60 to 120 micrograms every 12 hours, losartan potassium 50 milligrams every 12 hours, and levothyroxine 88 micrograms a day. Discussion CD is associated with high mortality, primarily attributable to cardiovascular outcomes and comorbidities such as metabolic and skeletal disorders, infections, and psychiatric disorders [1]. The low incidence of CD in the context of the high prevalence of chronic noncommunicable diseases makes early diagnosis a challenge [2]. This case is relevant for reviewing the diagnostic approach process and highlighting the impact of the availability bias, which tends to prioritize more common diagnoses over rare diseases. Despite the absence of typical symptoms, a timely diagnosis was achieved. Once exogenous CS is ruled out, laboratory testing must focus on detecting endogenous hypercortisolism to prevent misdiagnosis and inappropriate treatment [5]. Screening methods include 24-hour urinary free cortisol (UFC) for total cortisol load, while circadian rhythm and hypothalamic-pituitary-adrenal (HPA) axis function may be evaluated using midnight serum cortisol and LNSC [5]. An early hallmark of endogenous CS is the disruption of physiological circadian cortisol patterns, characterized by a constant cortisol level throughout the day or no significant decrease [2]. Measuring LNSC has proven to be useful in identifying these patients. The LNSC performed on the patient yielded a high result. To assess HPA axis suppressibility, tests such as the overnight and the standard 2-day LDDST [5] use dexamethasone, a potent synthetic corticosteroid with high glucocorticoid receptor affinity and prolonged action, with minimal interference with cortisol measurement [6]. In a normal HPA axis, cortisol exerts negative feedback, inhibiting the secretion of corticotropin-releasing-hormone (CRH) and ACTH. Exogenous corticosteroids suppress CRH and ACTH secretion, resulting in decreased synthesis and secretion of cortisol. In pathological hypercortisolism, the HPA axis becomes partially or entirely resistant to feedback inhibition by exogenous steroids [5, 6]. The LDDST involves the administration of 0.5 mg of dexamethasone orally every 6 hours for 2 days, with a total dose of 4 mg. A blood sample is drawn 6 hours after the last administered dose [6]. Following the LDDST, the patient did not demonstrate suppression of endogenous corticosteroid production. After diagnosing CS, the next step in the diagnostic pathway involves categorizing it as ACTH-independent vs ACTH-dependent. ACTH-independent cases exhibit low or undetectable ACTH levels, pointing to adrenal origin. The underlying principle is that excess ACTH production in CD can be partially or completely suppressed by high doses of dexamethasone, a response not observed in ectopic tumors [6]. In this case, the patient presented with an ACTH of 38.1 pg/mL (8.4 pmol/L), indicative of ACTH-dependent CD. Traditionally, measuring cortisol levels and conducting pituitary imaging are standard practices for diagnosis. Recent advances propose alternative diagnostic methods such as positron emission tomography (PET) scans and corticotropin-releasing factor (CRF) tests [7]. PET scans, utilizing radioactive tracers, offer a view of metabolic activity in the adrenal glands and pituitary region, aiding in the identification of abnormalities associated with CD. Unfortunately, the availability of the aforementioned tests in the country is limited. Once ACTH-dependent hypercortisolism is confirmed, identifying the source becomes crucial. A HDDST is instrumental in distinguishing between a pituitary and an ectopic source of ACTH overproduction [2, 6]. The HDDST involves administering 8 mg of dexamethasone either overnight or as a 2-day test. In this case, the patient received 2 mg of dexamethasone orally every 6 hours for 2 days, totaling a dose of 16 mg. Simultaneously, a urine sample for UFC is collected during dexamethasone administration. The HDDST suppressed endogenous cortisol production in the patient, suggesting a pituitary origin. In ACTH-dependent hypercortisolism, CD is the predominant cause, followed by ectopic ACTH syndrome and, less frequently, an ectopic CRH-secreting tumor [3, 5]. With the pretest probability for pituitary origin exceeding 80%, the next diagnostic step is typically an MRI of the pituitary region. However, the visualization of microadenomas on MRI ranges from 50% to 70%, requiring further testing if results are negative or inconclusive [5]. Initial testing of our patient revealed no pituitary lesions. Following a pituitary location, ACTH-secreting tumors may be found in the lungs. Thus, a high-resolution chest computed tomography scan was performed, which yielded negative findings. Healthcare professionals must keep these detection rates in mind. In instances of high clinical suspicion, repeating or reassessing tests and imaging may be warranted [3], as in our case, ultimately leading to the discovery of a 4-mm pituitary adenoma. It is fundamental to mention that the Endocrine Society Clinical Practice Guideline on Treatment of CS recommends that, when possible, all patients presenting with ACTH-dependent CS and lacking an evident causal neoplasm should be directed to an experienced center capable of conducting inferior petrosal sinus sampling to differentiate between pituitary and nonpituitary or ectopic cause [8]. However, in this instance, such a referral was regrettably hindered by logistical constraints. Regarding patient outcomes and monitoring in CD, there is no consensus on defining remission criteria following tumor resection. Prolonged hypercortisolism results in suppression of corticotropes, resulting in low levels of ACTH and cortisol after surgical intervention. Typically, remission is identified by morning serum cortisol values below 5 µg/dL (138 nmol/L) or UFC levels between 28 and 56 nmol/d (10-20 µg/d) within 7 days after surgical intervention. In our case, the patient's morning serum cortisol was 115.8621 nmol/L (4.2 µg/dL), indicating remission. Remission rates in adults are reported at 73% to 76% in selectively resected microadenomas and at 43% in macroadenomas [8], highlighting the need for regular follow-up visits to detect recurrence. Following the surgery, the patient experienced diabetes insipidus, a relatively common postoperative occurrence, albeit usually transient [8]. It is recommended to monitor serum sodium levels during the first 5 to 14 days postsurgery for early detection and management. Additionally, pituitary deficiencies may manifest following surgery. In this patient, prolactin levels were compromised, potentially impacting sexual response. However, postoperative somatomedin levels were normal, and gonadotropins were not measured due to the patient's age group, as no additional clinical decisions were anticipated based on those results. Secondary hypothyroidism was diagnosed postoperatively. Moving forward, it is important to emphasize certain clinical signs and symptoms for diagnosing CD. The combination of low bone mineral density (Likelihood Ratio [LR] +21.33), central obesity (LR +3.10), and arterial hypertension (LR + 2.29) [9] has a higher positive LR than some symptoms considered “characteristic,” such as reddish-purple striae, plethora, proximal muscle weakness, and unexplained bruising [2, 10]. It is essential to give relevance to the signs the patient may present, emphasizing signs that have been proven to have an increased odds ratio (OR) such as osteoporosis (OR 3.8), myopathies (OR 6.0), metabolic syndrome (OR 2.7) and adrenal adenoma (OR 2.4) [9‐11]. The simultaneous development and worsening of these conditions should raise suspicion for underlying issues. Understanding the evolving nature of CD signs highlights the importance of vigilance during medical examinations, prioritizing the diagnostic focus, and enabling prompt initiation of treatment. Recognizing the overlap of certain clinical features in CS is fundamental to achieving a timely diagnosis. Learning Points CS diagnosis is challenging due to the absence of pathognomonic signs and symptoms and the overlap of features present in many pathologies, such as metabolic syndrome. Early detection of CS is crucial, given its association with high morbidity and mortality resulting from chronic exposure to glucocorticoids. Recognizing the combination of low bone mineral density, obesity, hypertension, and diabetes as valuable clinical indicators is key in identifying CS. Interdisciplinary collaboration is essential to achieve a comprehensive diagnostic approach. Acknowledgments We extend our gratitude to Pontificia Universidad Javeriana in Bogotá for providing essential resources and facilities that contributed to the successful completion of this case report. Special acknowledgment is reserved for the anonymous reviewers, whose insightful feedback significantly enhanced the quality of this manuscript during the peer-review process. Their contributions are sincerely appreciated. Contributors All authors made individual contributions to authorship. A.B.O. was involved in the diagnosis and management of this patient. M.A.G., J.M.H., and A.B.O. were involved in manuscript drafting and editing. All authors reviewed and approved the final draft. Funding This research received no public or commercial funding. Disclosures The authors declare that they have no conflicts of interest related to the current study. Informed Patient Consent for Publication Signed informed consent could not be obtained from the patient or a proxy but has been approved by the treating institution. Data Availability Statement Restrictions apply to the availability of some or all data generated or analyzed during this study to preserve patient confidentiality or because they were used under license. The corresponding author will on request detail the restrictions and any conditions under which access to some data may be provided. References 1 Hakami OA , Ahmed S , Karavitaki N . Epidemiology and mortality of Cushing's syndrome . Best Pract Res Clin Endocrinol Metab . 2021 ; 35 ( 1 😞 101521 . Google Scholar Crossref PubMed WorldCat 2 Nieman LK , Biller BMK , Findling JW , et al. The diagnosis of Cushing's syndrome: an endocrine society clinical practice guideline . J Clin Endocrinol Metab . 2008 ; 93 ( 5 😞 1526 ‐ 1540 . Google Scholar Crossref PubMed WorldCat 3 Gutiérrez Restrepo J , Latorre Sierra G , Campuzano Maya G . Síndrome de cushing . Med Lab . 2009 ; 15 : 411 ‐ 430 . Google Scholar WorldCat 4 Petersenn S , Newell-Price J , Findling JW , et al. High variability in baseline urinary free cortisol values in patients with Cushing's disease . Clin Endocrinol (Oxf) . 2014 ; 80 ( 2 😞 261 ‐ 269 . Google Scholar Crossref PubMed WorldCat 5 Lila AR , Sarathi V , Jagtap VS , Bandgar T , Menon P , Shah NS . Cushing's syndrome: stepwise approach to diagnosis . Indian J Endocrinol Metab . 2011 ; 15 ( Suppl4 😞 S317 ‐ S321 . Google Scholar PubMed WorldCat 6 Dogra P , Vijayashankar NP . Dexamethasone suppression test. In: StatPearls StatPearls Publishing; 2024 . Accessed January 29, 2024. http://www.ncbi.nlm.nih.gov/books/NBK542317/ 7 Müller OA , Dörr HG , Hagen B , Stalla GK , von Werder K . Corticotropin releasing factor (CRF)-stimulation test in normal controls and patients with disturbances of the hypothalamo-pituitary-adrenal axis . Klin Wochenschr . 1982 ; 60 ( 24 😞 1485 ‐ 1491 . Google Scholar Crossref PubMed WorldCat 8 Nieman LK , Biller BMK , Findling JW , et al. Treatment of Cushing's syndrome: an endocrine society clinical practice guideline . J Clin Endocrinol Metab . 2015 ; 100 ( 8 😞 2807 ‐ 2831 . Google Scholar Crossref PubMed WorldCat 9 Aron DC . Cushing's syndrome: why is diagnosis so difficult? Rev Endocr Metab Disord . 2010 ; 11 ( 2 😞 105 ‐ 116 . Google Scholar Crossref PubMed WorldCat 10 Braun LT , Vogel F , Zopp S , et al. Whom should we screen for cushing syndrome? the Endocrine Society practice guideline recommendations 2008 revisited . J Clin Endocrinol Metab . 2022 ; 107 ( 9 😞 e3723 ‐ e3730 . Google Scholar Crossref PubMed WorldCat 11 Schneider HJ , Dimopoulou C , Stalla GK , Reincke M , Schopohl J . Discriminatory value of signs and symptoms in Cushing's syndrome revisited: what has changed in 30 years? Clin Endocrinol (Oxf) . 2013 ; 78 ( 1 😞 153 ‐ 154 . Google Scholar Crossref PubMed WorldCat Abbreviations ACTH adrenocorticotropic hormone CD Cushing disease CRH corticotropin-releasing hormone CS Cushing syndrome HDDST high-dose dexamethasone suppression test HPA hypothalamic-pituitary-adrenal LDDST low-dose dexamethasone suppression test LNSC late-night salivary cortisol MRI magnetic resonance imaging OR odds ratio RV reference value UFC urinary free cortisol © The Author(s) 2024. Published by Oxford University Press on behalf of the Endocrine Society. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited. From https://academic.oup.com/jcemcr/article/2/4/luae048/7643486?login=false
    1 point
  6. Another picture that weirdly turned into a link. Here's the pink jeep: http://cushieblog.files.wordpress.com/2012/04/pink-jeep.jpg?w=300&h=225
    1 point
  7. Abstract Background: Cushing’s disease (CD) is associated with a specific form of metabolic syndrome that includes visceral obesity, which may affect cardiovascular hemodynamics by stimulating hypercortisolism-related metabolic activity. The purpose of this study was to evaluate the relationship between obesity and the hemodynamic profile of patients with CD. Methods: This prospective clinical study involved a hemodynamic status assessment of 54 patients newly diagnosed with CD with no significant comorbidities (mean age of 41 years). The assessments included impedance cardiography (ICG) to assess such parameters as stroke index (SI), cardiac index (CI), velocity index (VI), acceleration index (ACI), Heather index (HI), systemic vascular resistance index (SVRI), and total arterial compliance index (TACI) as well as applanation tonometry to assess such parameters as central pulse pressure (CPP) and augmentation index (AI). These assessments were complemented by echocardiography to assess cardiac structure and function. Results: Compared with CD patients without obesity, individuals with CD and obesity (defined as a body mass index ≥ 30 kg/m2) exhibited significantly lower values of ICG parameters characterizing the pumping function of the heart (VI: 37.0 ± 9.5 vs. 47.2 ± 14.3 × 1*1000−1*s−1, p = 0.006; ACI: 58.7 ± 23.5 vs. 76.0 ± 23.5 × 1/100/s2, p = 0.005; HI: 11.1 ± 3.5 vs. 14.6 ± 5.5 × Ohm/s2, p = 0.01), whereas echocardiography in obese patients showed larger heart chamber sizes and a higher left ventricular mass index. No significant intergroup differences in blood pressure, heart rate, LVEF, GLS, TACI, CPP, or AI were noted. Conclusions: Hemodynamic changes associated with obesity already occur at an early stage of CD and manifest via significantly lower values of the ICG parameters illustrating the heart’s function as a pump, despite the normal function of the left ventricle in echocardiography. Keywords: Cushing’s disease; global longitudinal strain; impedance cardiography; metabolic syndrome; obesity; hemodynamic profile; cardiovascular complications Graphical Abstract 1. Introduction Cushing’s disease (CD), caused by a pituitary neuroendocrine tumor, leads to a specific type of metabolic syndrome that includes hypertension, obesity, impaired glucose metabolism, and dyslipidemia [1,2,3]. Chronic hypercortisolemia in patients with CD results in the excessive accumulation of visceral fat due to abnormal adipokine production [4]. Visceral obesity plays an important role in hypercortisolism-induced metabolic abnormalities and increased activity of the renin–angiotensin–aldosterone system activity in patients with CD [1,2,3,4,5]. Visceral obesity in patients with CD not only contributes to metabolic syndrome, but it is also an independent risk factor for cardiovascular disease [1,3,6,7]. Importantly, the structure and function of adipose tissue in patients with CD differ from those of healthy individuals [1,8,9]. The various hypercortisolism-induced metabolic abnormalities occurring in obese patients with CD may affect cardiovascular hemodynamics. There are no data on the effect of obesity on the hemodynamic profile of patients with CD and also few data are known on the association between obesity and hemodynamic disturbances in people without CD [10,11]. It was shown that the hemodynamic profile of a person with obesity is characterized by increased cardiac output and thoracic fluid content and decreased vascular resistance in comparison with these parameters in healthy individuals [12]. More studies are needed to enhance our understanding of the pathophysiology of CD-related obesity as a modifiable cardiovascular risk factor, in order to develop effective preventive and therapeutic strategies. Unfortunately, subclinical consequences of hypercortisolism in newly diagnosed patients with early CD, particularly with comorbid obesity, may be undetectable with standard methods. Therefore, novel and easy-to-use diagnostic methods would be of additive value to the standard methods of assessing cardiovascular structure and function in patients with CD. A detailed evaluation of the nature of obesity in patients with CD by innovative noninvasive diagnostic methods, such as impedance cardiography (ICG), applanation tonometry (AT), and echocardiographic assessment of global longitudinal strain (GLS), may provide additional data on cardiovascular hemodynamics, particularly the heart’s pumping function, preload, and afterload [13,14,15,16,17,18]. Our previous studies demonstrated the usefulness of ICG in identifying subclinical cardiovascular complications in patients with CD [19,20]. The purpose of this analysis was to assess the relationship between obesity and the hemodynamic profile of patients newly diagnosed with CD with no significant comorbidities. 2. Materials and Methods 2.1. Study Population This was a prospective observational cohort study involving a comprehensive assessment of 54 patients (mean age of 41 years) newly diagnosed with CD with no significant comorbidities (although 64.8% were diagnosed with hypertension). These patients were admitted to the Military Institute of Medicine—National Research Institute between 2016 and 2021 in order to undergo a thorough cardiovascular assessment prior to transsphenoidal pituitary neuroendocrine tumor resection surgery. This study was approved by the ethics committee at the Military Institute of Medicine—National Research Institute (approval No. 76/WIM/2016) and compliant with the Declaration of Helsinki and Good Clinical Practice guidelines. Each patient received detailed information on the purpose of this study and signed an informed consent form. This study was financed by the Polish Ministry of Research and Higher Education/Military Institute of Medicine—National Research Institute in Warsaw (grant No. 453/WIM). 2.2. Inclusion Criteria The diagnosis of CD was established based on the presence of the typical (clinical and hormonal) evidence of hypercortisolism with no adrenocorticotropic hormone (ACTH) response to corticotropin-releasing hormone (CRH) stimulation, which meets the current guidelines for the diagnosis and treatment of CD [21,22,23]. Physical examination findings consistent with the signs and symptoms of CD, including central obesity with the characteristic altered body fat distribution (a moon face and a short, thick neck); muscle atrophy in the torso and limbs; purplish stretch marks on the abdomen, hips, and thighs; thinned skin; ecchymoses; signs and symptoms of hyperandrogenism; bone pain; frequent infections; erectile dysfunction in men; and secondary amenorrhea and infertility in women. Hormone test results included elevated 24 h urinary free cortisol levels, increased morning serum cortisol levels, altered circadian rhythmicity of ACTH and cortisol secretion, elevated or detectable morning serum ACTH, and a lack of overnight serum cortisol suppression to <1.8 mg/dL during a low-dose dexamethasone suppression test (1 mg or 2 mg of dexamethasone administered at midnight). In order to ensure a pituitary etiology of CD, all patients underwent a two-day high-dose (2 mg every 6 h = a total of 8 mg) dexamethasone suppression test (HDDST), which was expected to show low serum cortisol or a >50% decrease in urinary-free cortisol levels. Moreover, each patient was shown to have no ACTH secretion response to a CRH stimulation test (with 100 μg intravenous CRH), and the presence of a pituitary neuroendocrine tumor was confirmed via contrast magnetic resonance imaging of the pituitary. Patients with inconclusive hormone tests or imaging studies additionally underwent bilateral inferior petrosal sinus sampling (used to determine ACTH levels in the venous blood before and after CRH stimulation) [21,22,23]. 2.3. Exclusion Criteria The following comorbidities, which might considerably affect hemodynamic profiles, constituted our study exclusion criteria: (1) heart failure with mildly reduced or reduced left ventricular ejection fraction (LVEF) (i.e., LVEF of <50%); (2) cardiomyopathy; (3) clinically significant valvular heart disease or arrhythmia; (4) coronary artery disease, including a history of acute coronary syndrome; (5) a poor acoustic window on echocardiography; (6) a history of pulmonary embolism; (7) a history of a stroke or transient ischemic attack; (8) renal failure (estimated glomerular filtration rate < 60 mL/min/1.73 m2); (9) peripheral vascular disease and polyneuropathy; (10) chronic obstructive pulmonary disease; (11) respiratory failure (decreased partial pressure of arterial oxygen [PaO2] < 60 mmHg and/or increased partial pressure of carbon dioxide [PaCO2] > 45 mmHg); (12) a history of head trauma; (13) pregnancy; (14) age < 18 years; (15) no written informed consent. 2.4. Additional Hormone Tests Due to the fact that hypercortisolemia inhibits gonadotropin release, hormone testing was expanded to include follicle-stimulating hormone and luteinizing hormone levels. The patients also had their serum thyroid-stimulating hormone levels tested to determine possible hypothyroidism, associated with reduced CRH and thyroid-stimulating hormone secretion and hypercortisolism-induced alterations in thyroid function. The patients with CD included in this study were not receiving any medications affecting the hypothalamus–pituitary–adrenal axis. None of the female patients with CD were pregnant at the time of the study or had given birth within the previous five years. 2.5. Laboratory Tests In order to detect possible metabolic conditions, such as impaired fasting glucose, type 2 diabetes mellitus, or dyslipidemia, all patients underwent fasting blood tests from venous blood samples collected in the morning (at 6:00 a.m.). The tests evaluated the levels of fasting glucose, creatinine, eGFR, total cholesterol, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, and triglycerides, as well as a complete blood count. 2.6. Anamnesis and Physical Examination The patients were thoroughly evaluated for cardiovascular risk factors, cardiovascular signs and symptoms, a family history of cardiovascular disease, comorbidities, prescription medications and other drugs, and smoking. The body mass index (BMI) was calculated, and obesity was determined based on the International Diabetes Federation and European Society of Cardiology guidelines, which define it as a BMI of ≥30 kg/m2 [24,25]. In the study, patients were divided into two groups: patients with CD and obesity (defined as high body mass index ≥ 30 kg/m2) and patients with CD without obesity (defined as normal BMI < 30 kg/m2). Physical examination included the resting heart rate (HR), systolic and diastolic blood pressure, and anthropometric parameters. Office blood pressure measurements were taken by a trained nurse in seated patients in the morning, after a 5 min rest. The blood pressure monitor used was Omron M4 Plus (Omron Healthcare Co. Ltd., Kyoto, Japan), which meets the European Society of Cardiology criteria [26]. 2.7. Echocardiography Two-dimensional echocardiography included standard parasternal, apical, and subcostal views with a 2.5 MHz transducer (VIVID E95, GE Medical System, Wauwatosa, WI, USA) in accordance with the American Society of Echocardiography (ASE) and the European Association of Cardiovascular Imaging (EACVI) guidelines [27]. The parasternal long-axis view was used to measure the left ventricular end-diastolic diameter (LVEDd), right ventricular end-diastolic diameter (RVEDd), interventricular septal thickness, and left atrial (LA) diameter. Linear 2-dimensional left ventricular measurements were used to calculate the left ventricular mass index (LVMI), which is the left ventricular mass divided by the body surface area (LVMI cut-off values of >115 g/m2 for men and >95 g/m2 for women meet ASE and EACVI criteria for the diagnosis of left ventricular hypertrophy). The LVEF was calculated with the biplane Simpson method, based on 2-dimensional views of the left ventricle during systole and diastole in four- and two-chamber apical views. The ascending aortic diameter, valvular structure and function, and pericardium were assessed. The patients were assessed for left ventricular diastolic dysfunction according to current guidelines. Pulse wave Doppler in an apical four-chamber view aligned with mitral valve tips was used to visualize mitral inflow, including the early passive blood inflow (E) and the later atrial (A) contribution to the mitral inflow, E/A ratio, and early mitral inflow deceleration time. Apical four-chamber views were used to determine the septal and lateral early diastolic mitral annular velocities (e′ avg), and the E/e′ avg ratio was calculated [27,28]. Global longitudinal strain (GLS) was assessed via electrocardiography-gated automated function imaging in two-, three-, and four-chamber views. The rates of >60 frames per second were used for optimal speckle-tracking strain assessment. Patients with a poor acoustic window were excluded from the study. Semiautomated endocardial border detection was initiated by manually selecting two points identifying the mitral annulus and one point at the apex. Segmental and whole-chamber strain was assessed. The results have been presented in the form of a “bull’s eye” graph. The data were analyzed for four-, three-, and two-chamber views, and average GLS was calculated [29]. 2.8. Impedance Cardiography Based on the phenomenon of impedance variability in individual body segments associated with regional arterial blood flow, ICG is a noninvasive tool for assessing cardiovascular hemodynamics. ICG assessments were conducted by a trained nurse with a Niccomo device (Medis, Ilmenau, Germany) in patients who had been resting for 10 min in a supine position. ICG data were recorded during a 10 min assessment and processed with dedicated software (Niccomo Software, Medis). We analyzed the mean values of the following hemodynamic parameters reflecting the pumping function of the heart: (1) stroke volume (SV [mL]) and stroke index (SI [mL/m2]), based on the following formula: SV = VEPT × (dZmax/Z0) × LVET, where VEPT is tissue volume calculated from body weight, height, and patient sex, Z0 is the initial thoracic impedance, dZmax is the maximum change in thoracic impedance, and LVET is the left ventricular ejection time; (2) cardiac output (CO [mL] = SV × HR), and cardiac index (CI [mL*m−2*min−1]); (3) velocity index (VI [1*1000−1*s−1]); (4) acceleration index (ACI [1/100/s2], which is the peak acceleration of blood flow in the aorta; and (5) Heather index (HI [Ohm/s2] = dZmax × TRC, where TRC the time interval between the R-peak in the electrocardiogram and the C-point on the impedance wave). We also conducted a detailed analysis of the following afterload parameters: (1) systemic vascular resistance (SVR [dyn*s*cm−5]) together with SVR index (SVRI [dyn*s*cm−5*m2]) and (2) total arterial compliance (TAC) and TAC index (TACI [mL/mmHg] = SV/pulse pressure [mL/mmHg*m2]). Preload was assessed based on thoracic fluid content (TFC [1/kOhm], based on the formula TFC = 1000/Z0, where Z0 is the initial thoracic impedance [30,31,32]. 2.9. Applanation Tonometry Applanation tonometry is a novel method of indirectly illustrating arterial pressure waveform in the aorta and arterial stiffness, which reflect left ventricular afterload. AT parameters were assessed noninvasively with a SphygmoCor system (AtCor Medical, Sydney, NSW, Australia). The measurements were taken in supine patients by a qualified nurse immediately after ICG. Radial artery pressure curves were recorded via AT with a micromanometer (Millar Instruments, Houston, TX, USA) strapped onto the left wrist. We selected high-quality recordings for our analysis. Radial pulse was calibrated against the latest brachial systolic and diastolic blood pressure measurement with an oscillometric module of the Niccomo device. SphygmoCor software (version 9.0; AtCor Medical Inc. Pty Ltd., Sydney, NSW, Australia) was used to process the arterial waveform and generate an appropriate aortic blood pressure curve from the radial pulse curve. The analyzed waveforms were composed of the pulse wave generated by the aorta and were augmented by an overlapping reflected wave. Our analyses yielded the following parameters: central systolic blood pressure; central diastolic blood pressure; central pulse pressure (CPP); augmentation pressure, which is the absolute increase in aortic systolic pressure (directly generated by left ventricular contraction) resulting from the reflection wave; and the augmentation index, calculated as AP × 100/CPP, which is a quotient of the augmentation pressure and the blood pressure in the aorta [33]. 2.10. Statistical Analysis For the statistical analysis of the results, we used MS Office Excel 2023 and Statistica 12.0 (StatSof Inc., Tulsa, OK, USA). Data distribution and normality were assessed visually on histograms and with the use of the Kolmogorov–Smirnov test. Continuous variables were expressed as mean ± standard deviation (SD) or median (interquartile range, IQR), and categorical variables were expressed as absolute and relative (percentage) values. In order to evaluate differences between the subgroups of CD patients with and without comorbid obesity, we used Student’s t-test for normally distributed data, and the Mann–Whitney U test for non-normally distributed data. A comparative analysis with the use of the Mann–Whitney U test was conducted on the data from patients stratified into two subgroups: patients with CD and obesity (BMI ≥ 30 kg/m2, n = 22) and patients with CD without obesity (BMI < 30 kg/m2, n = 32). The relationship between selected indices of cardiovascular function and obesity (represented as BMI) was analyzed separately for each one in a multivariable regression model, adjusting for age and hypertension as potential covariates related to hemodynamics. The threshold of statistical significance was adopted at p < 0.05. 3. Results 3.1. Baseline Characteristics Nearly half of the patients with CD were found to be obese (n = 22, 40.7%). Overall, 20 of the 54 patients with Cushing’s disease (37%) were diagnosed with type 2 diabetes mellitus, 5 (9.3%) had prediabetes, and 29 (46.3%) had normal glucose tolerance. Of the patients with Cushing’s disease and type 2 diabetes, 14 received metformin, 5 received metformin with insulin, and 1 received insulin. The mean age, HR, hemoglobin, creatinine, and sex distribution were similar in the subgroup with and without obesity (Table 1). Table 1. Clinical, echocardiographic, hemodynamic, and applanation tonometry variables in patients with Cushing’s disease (CD) and with or without obesity. 3.2. Echocardiographic Assessment Patients with CD and obesity (BMI ≥ 30 kg/m2) showed larger dimensions of heart chambers and ascending aorta (RVEDd, p < 0.001; LVEDd, p = 0.028; LA diameter, p < 0.001; aortic arch, p = 0.005) and higher rates of left ventricular mass index (LVMI, p = 0.028). We observed no significant differences between the subgroups in terms of the systolic (LVEF or GLS) or diastolic function of the left ventricle (Table 1). 3.3. ICG and AT Assessment The most noticeable differences in ICG were observed for parameters of the left ventricular function as a pump. In obese individuals, VI (p = 0.006), ACI (p = 0.005), and HI (p = 0.012) were lower, whereas the systolic time ratio (STR) was higher (p = 0.038) than those in non-obese individuals, with SI and CI comparable in both subgroups. We observed no significant differences in afterload (TACI, SVRI, CPP, or augmentation index) or preload (TFC) parameters (Table 1). 3.4. Correlation Analysis Analyzing the relationships between BMI and ICG hemodynamic parameters, we observed significant correlations, independent of sex and hypertension, between BMI and CI (R = 0.46; p < 0.001), SI (R = 0.29; p = 0.043), SVRI (−0.31; 0.028), and VI (R = −0.37; p = 0.0006)—see Table 2. Table 2. Correlations between hemodynamic parameters assessed with impedance cardiography and body mass index, adjusted for sex and hypertension in multivariable regression models. 4. Discussion The results of our study revealed a relationship between obesity and hemodynamic profile assessed via ICG in patients newly diagnosed with active CD. The use of novel diagnostic modalities demonstrated that excessive fat accumulation in young and middle-aged patients with CD, already at the early stages of the disease, is associated with some hemodynamic changes in the cardiovascular system, which—at that stage—may still be undetectable in routine assessments. These findings support the need for the early detection of subclinical heart dysfunction in patients with CD to enable early treatment and help prevent cardiovascular complications [1,34,35,36]. Occurring in 25%–100% of patients with CD, visceral obesity is one of the most common components of metabolic syndrome, often being the first sign of the disease. The duration of hypercortisolism correlates with obesity development [1,7,37,38], with chronic excessive cortisol levels being responsible for the abnormal distribution of adipose tissue [39]. The mechanisms behind this phenomenon may be due to the tissue overexpression of the 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), which affects the pattern of excessive fat distribution in the torso, face, and neck [1,6]. Visceral obesity found in patients with CD is not only a component of metabolic syndrome but is in itself associated with increased metabolic activity, which makes it an independent cardiovascular risk factor, leading to the development of cardiovascular disease [1,4,9]. The tendency to accumulate visceral fat in patients with CD is also associated with abnormal adipokine production [4,6,40,41]. Our study included patients newly diagnosed with active CD with no clinically significant cardiovascular disease. Males were underrepresented in both subgroups. The proportion of patients with hypertension was 64.8%, which is comparable with that reported by other authors [38,42,43,44] and similarly distributed between subgroups. However, the patients in our study presented well-controlled hypertension (mean blood pressure was 126/83 mmHg), usually with one or two medications. Considering both sex and hypertension as potential confounders, these variables were included in regression models evaluating correlations between hemodynamics and BMI. Similar to reports by other authors, our study showed higher SV and CO values in obese patients with CD; however, the respective indexed values (SI and CI) were comparable in obese and non-obese patients [12,45]. A more detailed ICG assessment demonstrated significant impairment of the pumping function of the heart as evidenced by lower HI, VI, and ACI values, and a higher STR value. The analysis of correlations revealed the independence of age and sex interrelation between some hemodynamic indices (CI, SI, SVRI, VI) and BMI. The paradox of the positive relation of obesity with volume indices of left ventricular function (CI and SI), which is negative with the marker of both its outflow and myocardial contractility (VI) encourages further studies investigating the (patho)physiological background of this phenomenon. These findings were detected despite the lack of echocardiographic evidence of left ventricular systolic or diastolic dysfunction. Moreover, our study showed larger heart chamber diameters and significantly higher LVMI in patients with CD and obesity, which is consistent with numerous earlier reports by other authors [46,47,48]. Nonetheless, it seems that in this case, increased heart chamber size and left ventricular hypertrophy should not be considered as only secondary to an increase in body weight. Hypercortisolism in patients with CD worsens the structural and functional condition of the heart muscle and may lead to myocardial fibrosis [48]. This results in myocardial remodeling associated with concentric left ventricular hypertrophy, which may impair left ventricular hemodynamic function, subsequently leading to myocardial dysfunction and symptomatic heart failure [49,50,51]. The effective treatment of patients with CD has been shown to normalize their serum cortisol levels and ultimately stop myocardial remodeling [47]. Therefore, the ICG-evidenced impaired pumping function of the heart may result from myocardial remodeling associated with complex metabolic and neuroendocrine changes in obese patients with CD [52]. These findings are consistent with previous reports on the adverse effect of obesity on left ventricular contractility [53,54,55,56]. The potential mechanisms underlying the results of our study remain to be elucidated. An interesting perspective is represented by the cross-talk between glucocorticoid (GR) and mineralocorticoid receptors (MR) and their impact on metabolic syndrome. Excessive activation of the MR in extra-renal tissues by aldosterone or glucocorticoids depending on the expression of 11beta-hydroxysteroid dehydrogenase type 2 has been shown to be associated with the development of vascular dysfunction and metabolic abnormalities, leading to obesity and metabolic syndrome. High concentrations of aldosterone may also activate the transcriptional function of the GR. These mechanisms result in an interaction between GR and MR in the regulation of adipogenesis [57]. The novelty of our approach is due to the use of noninvasive tools (ICG, AT) for hemodynamic assessment of the cardiovascular system in patients with CD to detect subclinical changes associated with obesity. On the one hand, our findings support earlier observations in other patient groups; on the other hand, they cast a new light on the relationship between obesity and an impaired hemodynamic profile in CD, which may result in the early development of cardiovascular complications. 4.1. Clinical Implications We determined that a dysfunctional pumping action of the heart is the key marker of impaired cardiovascular hemodynamics in obese patients newly diagnosed with CD. The use of noninvasive diagnostic methods in this study revealed a complex relationship between obesity-related hemodynamic changes and the efficiency of left ventricular contractions. An early assessment of a patient’s hemodynamic profile may help detect subclinical cardiovascular dysfunction. Such a personalized approach may facilitate early therapeutic intervention and monitoring of treatment effectiveness focused on preventing myocardial remodeling and heart dysfunction. 4.2. Limitations One limitation of our study was the small sample size. This was a result of the relatively low incidence of pituitary neuroendocrine tumors secreting ACTH. The exclusion of patients with clinically significant comorbidities further diminished the study population. However, this helped to eliminate the effect of additional factors on hemodynamic profiles. The patients assessed in our study were mostly young and middle-aged individuals with CD; therefore, our conclusions should not be extrapolated to older subjects. Although we conducted neither cardiac stress tests nor coronary angiography to exclude asymptomatic ischemic heart disease, other thorough assessments showed no physical, electrocardiographic, or echocardiographic evidence suggesting myocardial ischemia. Another potential limitation of our study is the fact that some patients had hypertension; however, it was well controlled with medications. The hemodynamic assessments involved the use of noninvasive methods as an alternative to the more expensive and less readily available invasive techniques. Nonetheless, we acknowledge the fact that noninvasive measurements can only provide indirect measurements and depend on the patient’s condition, which may vary over time. 5. Conclusions The results of our study support the usefulness of ICG in diagnosing early heart dysfunction associated with obesity in patients with CD. Asymptomatic impairment of the heart’s pumping function seems to be the earliest clinical sign of cardiovascular hemodynamic abnormalities, which at this stage are still undetectable with standard echocardiography. Individual hemodynamic profile assessment with novel noninvasive diagnostic methods encourages further studies on cardiovascular system function in obese individuals with CD and on the use of personalized therapies, which aim at preventing adverse cardiovascular events. Author Contributions Conceptualization, A.J. and P.K.; methodology, A.J., P.K., G.G., B.U.-Ż., P.W. and G.Z.; software, P.K.; validation, A.J., P.K., B.U.-Ż., P.W. and G.Z.; formal analysis, P.K., P.W., G.G. and G.Z.; investigation, A.J., P.K., B.U.-Ż., P.W. and G.Z.; resources, A.J., P.K., B.U.-Ż., P.W. and G.Z.; data curation, A.J., P.K., B.U.-Ż., P.W., G.Z., A.K., R.W. and M.B.; writing—original draft preparation, A.J. and P.K.; writing—review and editing, G.G., B.U.-Ż., P.W. and G.Z.; visualization, A.J.; supervision, G.G. and G.Z.; project administration, G.Z.; funding acquisition, G.Z. All authors have read and agreed to the published version of the manuscript. Funding This research was funded by the Polish Ministry of Research and Higher Education/Military Institute of Medicine—National Research Institute in Warsaw (grant No. 453/WIM). Institutional Review Board Statement The study was conducted in accordance with the Declaration of Helsinki and Good Clinical Practice guidelines and approved by the Bioethics Committee at the Military Institute of Medicine—National Research Institute in Warsaw, Poland (approval No. 76/WIM/2016; 21 December 2016). Informed Consent Statement Informed consent was obtained from all subjects involved in the study. Data Availability Statement The data presented in this study are available upon request from the corresponding author. The data are not publicly available due to privacy or ethical restrictions. Acknowledgments We would like to thank the medical personnel of the Military Institute of Medicine—National Research Institute in Warsaw for the provided patient care. Conflicts of Interest The authors declare no conflicts of interest. References Pivonello, R.; Isidori, A.M.; De Martino, M.C.; Newell-Price, J.; Biller, B.M.; Colao, A. Complications of Cushing’s syndrome: State of the art. Lancet Diabetes Endocrinol. 2016, 4, 611–629. [Google Scholar] [CrossRef] Dekkers, O.M.; Horváth-Puhó, E.; Jørgensen, J.O.; Cannegieter, S.C.; Ehrenstein, V.; Vandenbroucke, J.P.; Pereira, A.M.; Sørensen, H.T. Multisystem morbidity and mortality in Cushing’s syndrome: A cohort study. J. Clin. Endocrinol. Metab. 2013, 98, 2277–2284. [Google Scholar] [CrossRef] [PubMed] Pivonello, R.; Faggiano, A.; Lombardi, G.; Colao, A. The metabolic syndrome and cardiovascular risk in Cushing’s syndrome. Endocrinol. Metab. Clin. N. Am. 2005, 34, 327–339. [Google Scholar] [CrossRef] [PubMed] Pivonello, R.; De Leo, M.; Vitale, P.; Cozzolino, A.; Simeoli, C.; De Martino, M.C.; Lombardi, G.; Colao, A. Pathophysiology of diabetes mellitus in Cushing’s syndrome. Neuroendocrinology 2010, 1, 77–81. [Google Scholar] [CrossRef] Davy, K.P.; Hall, J.E. Obesity and hypertension: Two epidemics or one? Am. J. Physiol. Regul. Integr. Comp. Physiol. 2004, 286, R803–R813. [Google Scholar] [CrossRef] Lee, M.J.; Pramyothin, P.; Karastergiou, K.; Fried, S.K. Deconstructing the roles of glucocorticoids in adipose tissue biology and the development of central obesity. Biochim. Biophys. Acta 2014, 1842, 473–481. [Google Scholar] [CrossRef] [PubMed] Mancini, T.; Kola, B.; Mantero, F.; Boscaro, M.; Arnaldi, G. High cardiovascular risk in patients with Cushing’s syndrome according to 1999 WHO/ISH guidelines. Clin. Endocrinol. 2004, 61, 768–777. [Google Scholar] [CrossRef] Bujalska, I.J.; Kumar, S.; Stewart, P.M. Does central obesity reflect “Cushing’s disease of the omentum”? Lancet 1997, 349, 1210–1213. [Google Scholar] [CrossRef] Galton, D.J.; Wilson, J.P. Lipogenesis in adipose tissue of patients with obesity and Cushing’s disease. Clin. Sci. 1972, 43, 17P. [Google Scholar] [CrossRef] Koch, R.; Sharma, A.M. Obesity and cardiovascular hemodynamic function. Curr. Hypertens. Rep. 1999, 1, 127–130. [Google Scholar] [CrossRef] Raison, J. Conséquences cardiovasculaires de l’obésité associée à l’hypertension artérielle [Cardiovascular consequences of obesity associated with arterial hypertension]. Presse Med. 1992, 21, 1522–1525. [Google Scholar] de Simone, G.; Devereux, R.B.; Kizer, J.R.; Chinali, M.; Bella, J.N.; Oberman, A.; Kitzman, D.W.; Hopkins, P.N.; Rao, D.C.; Arnett, D.K. Body composition and fat distribution influence systemic hemodynamics in the absence of obesity: The HyperGEN Study. Am. J. Clin. Nutr. 2005, 81, 757–761. [Google Scholar] [CrossRef] [PubMed] Krzesiński, P.; Gielerak, G.; Kowal, J. Kardiografia impedancyjna—Nowoczesne narzedzie terapii monitorowanej chorób układu krazenia [Impedance cardiography—A modern tool for monitoring therapy of cardiovascular diseases]. Kardiol. Pol. 2009, 67, 65–71. (In Polish) [Google Scholar] El-Dawlatly, A.; Mansour, E.; Al-Shaer, A.A.; Al-Dohayan, A.; Samarkandi, A.; Abdulkarim, A.; Alshehri, H.; Faden, A. Impedance cardiography: Noninvasive assessment of hemodynamics and thoracic fluid content during bariatric surgery. Obes. Surg. 2005, 15, 655–658. [Google Scholar] [CrossRef] [PubMed] Eikås, J.G.; Gerdts, E.; Halland, H.; Midtbø, H.; Cramariuc, D.; Kringeland, E. Arterial Stiffness in Overweight and Obesity: Association with Sex, Age, and Blood Pressure. High Blood Press Cardiovasc. Prev. 2023, 30, 435–443. [Google Scholar] [CrossRef] [PubMed] Abomandour, H.G.; Elnagar, A.M.; Aboleineen, M.W.; Shehata, I.E. Subclinical Impairment of Left Ventricular Function assessed by Speckle Tracking in Type 2 Diabetic Obese and Non-Obese Patients: Case Control Study. J. Cardiovasc. Echogr. 2022, 32, 95–106. [Google Scholar] [CrossRef] Galderisi, M.; Lomoriello, V.S.; Santoro, A.; Esposito, R.; Olibet, M.; Raia, R.; Di Minno, M.N.; Guerra, G.; Mele, D.; Lombardi, G. Differences of myocardial systolic deformation and correlates of diastolic function in competitive rowers and young hypertensives: A speckle-tracking echocardiography study. J. Am. Soc. Echocardiogr. 2010, 23, 1190–1198. [Google Scholar] [CrossRef] Kalam, K.; Otahal, P.; Marwick, T.H. Prognostic implications of global LV dysfunction: A systematic review and meta-analysis of global longitudinal strain and ejection fraction. Heart 2014, 100, 1673–1680. [Google Scholar] [CrossRef] Jurek, A.; Krzesiński, P.; Gielerak, G.; Witek, P.; Zieliński, G.; Kazimierczak, A.; Wierzbowski, R.; Banak, M.; Uziębło-Życzkowska, B. Cushing’s Disease: Assessment of Early Cardiovascular Hemodynamic Dysfunction With Impedance Cardiography. Front. Endocrinol. 2021, 12, 751743. [Google Scholar] [CrossRef] Jurek, A.; Krzesiński, P.; Uziębło-Życzkowska, B.; Witek, P.; Zieliński, G.; Kazimierczak, A.; Wierzbowski, R.; Banak, M.; Gielerak, G. The patient’s sex determines the hemodynamic profile in patients with Cushing disease. Front. Endocrinol. 2023, 14, 1270455. [Google Scholar] [CrossRef] Fleseriu, M.; Auchus, R.; Bancos, I.; Ben-Shlomo, A.; Bertherat, J.; Biermasz, N.R.; Boguszewski, C.L.; Bronstein, M.D.; Buchfelder, M.; Carmichael, J.D.; et al. Consensus on diagnosis and management of Cushing’s disease: A guideline update. Lancet Diabetes Endocrinol. 2021, 9, 847–875. [Google Scholar] [CrossRef] [PubMed] Nieman, L.K.; Biller, B.M.; Findling, J.W.; Newell-Price, J.; Savage, M.O.; Stewart, P.M.; Montori, V.M. The diagnosis of Cushing’s syndrome: An Endocrine Society Clinical Practice Guideline. J. Clin. Endocrinol. Metab. 2008, 93, 1526–1540. [Google Scholar] [CrossRef] Ceccato, F.; Boscaro, M. Cushing’s syndrome: Screening and diagnosis. High. Blood Press. Cardiovasc. Prev. 2016, 23, 209–215. [Google Scholar] [CrossRef] Alberti, K.G.; Eckel, R.H.; Grundy, S.M.; Zimmet, P.Z.; Cleeman, J.I.; Donato, K.A.; Fruchart, J.C.; James, W.P.; Loria, C.M.; Smith, S.C., Jr.; et al. Harmonizing the metabolic syndrome: A joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation 2009, 120, 1640–1645. [Google Scholar] [CrossRef] Visseren, F.L.J.; Mach, F.; Smulders, Y.M.; Carballo, D.; Koskinas, K.C.; Bäck, M.; Benetos, A.; Biffi, A.; Boavida, J.M.; Capodanno, D.; et al. 2021 ESC Guidelines on cardiovascular disease prevention in clinical practice. Eur. Heart J. 2021, 42, 3227–3337. [Google Scholar] [CrossRef] [PubMed] Williams, B.; Mancia, G.; Spiering, W.; Agabiti Rosei, E.; Azizi, M.; Burnier, M.; Clement, D.L.; Coca, A.; de Simone, G.; Dominiczak, A.; et al. 2018 ESC/ESH Guidelines for the management of arterial hypertension. Eur. Heart J. 2018, 39, 3021–3104. [Google Scholar] [CrossRef] Lang, R.M.; Badano, L.P.; Mor-Avi, V.; Afilalo, J.; Armstrong, A.; Ernande, L.; Flachskampf, F.A.; Foster, E.; Goldstein, S.A.; Kuznetsova, T.; et al. Recommendations for cardiac chamber quantification by echocardiography in adults: An update from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. J. Am. Soc. Echocardiogr. 2015, 28, 1–39.e14. [Google Scholar] [CrossRef] Nagueh, S.F.; Smiseth, O.A.; Appleton, C.P.; Byrd, B.F.; Dokainish, H.; Edvardsen, T.; Flachskampf, F.A.; Gillebert, T.C.; Klein, A.L.; Lancellotti, P.; et al. Recommendations for the evaluation of left ventricular diastolic function by echocardiography: An update from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. J. Am. Soc. Echocardiogr. 2016, 29, 277–314. [Google Scholar] [CrossRef] Voigt, J.U.; Pedrizzetti, G.; Lysyansky, P.; Marwick, T.H.; Houle, H.; Baumann, R.; Pedri, S.; Ito, Y.; Abe, Y.; Metz, S.; et al. Definitions for a common standard for 2D speckle tracking echocardiography: Consensus document of the EACVI/ASE/Industry Task Force to standardize deformation imaging. J. Am. Soc. Echocardiogr. 2015, 28, 183–193. [Google Scholar] [CrossRef] [PubMed] Bhalla, V.; Isakson, S.; Bhalla, M.A.; Lin, J.P.; Clopton, P.; Gardetto, N.; Maisel, A.S. Diagnostic Ability of B-Type Natriuretic Peptide and Impedance Cardiography: Testing to Identify Left Ventricular Dysfunction in Hypertensive Patients. Am. J. Hypertens. 2005, 18, 73S–81S. [Google Scholar] [CrossRef] Parrott, C.W.; Burnham, K.M.; Quale, C.; Lewis, D.L. Comparison of Changes in Ejection Fraction to Changes in Impedance Cardiography Cardiac Index and Systolic Time Ratio. Congest. Heart Fail. 2004, 10, 11–13. [Google Scholar] [CrossRef] Packer, M.; Abraham, W.T.; Mehra, M.R.; Yancy, C.W.; Lawless, C.E.; Mitchell, J.E.; Smart, F.W.; Bijou, R.; O’Connor, C.M.; Massie, B.M.; et al. Utility of Impedance Cardiography for the Identification of Short-Term Risk of Clinical Decompensation in Stable Patients with Chronic Heart Failure. J. Am. Coll. Cardiol. 2006, 47, 2245–2252. [Google Scholar] [CrossRef] Pauca, A.L.; O’Rourke, M.F.; Kon, N.D. Prospective evaluation of a method for estimating ascending aortic pressure from the radial artery pressure waveform. Hypertension 2001, 38, 932–937. [Google Scholar] [CrossRef] [PubMed] Coulden, A.; Hamblin, R.; Wass, J.; Karavitaki, N. Cardiovascular health and mortality in Cushing’s disease. Pituitary 2022, 25, 750–753. [Google Scholar] [CrossRef] [PubMed] Clayton, R.N. Cardiovascular complications of Cushings syndrome: Impact on morbidity and mortality. J. Neuroendocrinol. 2022, 34, e13175. [Google Scholar] [CrossRef] [PubMed] De Leo, M.; Pivonello, R.; Auriemma, R.S.; Cozzolino, A.; Vitale, P.; Simeoli, C.; De Martino, M.C.; Lombardi, G.; Colao, A. Cardiovascular disease in Cushing’s syndrome: Heart versus vasculature. Neuroendocrinology 2010, 1, 50–54. [Google Scholar] [CrossRef] Faggiano, A.; Pivonello, R.; Spiezia, S.; De Martino, M.C.; Filippella, M.; Di Somma, C.; Lombardi, G.; Colao, A. Cardiovascular risk factors and common carotid artery caliber and stiff ness in patients with Cushing’s disease during active disease and 1 year after disease remission. J. Clin. Endocrinol. Metab. 2003, 88, 2527–2533. [Google Scholar] [CrossRef] [PubMed] Witek, P.; Zieliński, G.; Szamotulska, K.; Witek, J.; Zgliczyński, W. Complications of Cushing’s disease—Prospective evaluation and clinical characteristics. Do they affect the efficacy of surgical treatment? Endokrynol. Pol. 2012, 63, 277–285. [Google Scholar] [PubMed] Geer, E.B.; Shen, W.; Gallagher, D.; Punyanitya, M.; Looker, H.C.; Post, K.D.; Freda, P.U. MRI assessment of lean and adipose tissue distribution in female patients with Cushing’s disease. Clin. Endocrinol. 2010, 73, 469–475. [Google Scholar] [CrossRef] [PubMed] Wang, M. The role of glucocorticoid action in the pathophysiology of the metabolic syndrome. Nutr. Metab. 2005, 2, 3. [Google Scholar] [CrossRef] Tataranni, P.A.; Larson, D.E.; Snitker, S.; Young, J.B.; Flatt, J.P.; Ravussin, E. Eff ects of glucocorticoids on energy metabolism and food intake in humans. Am. J. Physiol. 1996, 271, E317–E325. [Google Scholar] [PubMed] Isidori, A.M.; Graziadio, C.; Paragliola, R.M.; Cozzolino, A.; Ambrogio, A.G.; Colao, A.; Corsello, S.M.; Pivonello, R. ABC Study Group. The hypertension of Cushing’s syndrome: Controversies in the pathophysiology and focus on cardiovascular complications. J. Hypertens. 2015, 33, 44–60. [Google Scholar] [CrossRef] Dekkers, O.M.; Biermasz, N.R.; Pereira, A.M.; Roelfsema, F.; van Aken, M.O.; Voormolen, J.H.; Romijn, J.A. Mortality in patients treated for Cushing’s disease is increased, compared with patients treated for nonfunctioning pituitary macroadenoma. J. Clin. Endocrinol. Metab. 2007, 92, 976–981. [Google Scholar] [CrossRef] [PubMed] Lambert, J.K.; Goldberg, L.; Fayngold, S.; Kostadinov, J.; Post, K.D.; Geer, E.B. Predictors of mortality and long-term outcomes in treated Cushing’s disease: A study of 346 patients. J. Clin. Endocrinol. Metab. 2013, 98, 1022–1030. [Google Scholar] [CrossRef] Abel, E.D.; Litwin, S.E.; Sweeney, G. Cardiac remodeling in obesity. Physiol. Rev. 2008, 88, 389–419. [Google Scholar] [CrossRef] Hey, T.M.; Dahl, J.S.; Brix, T.H.; Søndergaard, E.V. Biventricular hypertrophy and heart failure as initial presentation of Cushing’s disease. BMJ Case Rep. 2013, bcr2013201307. [Google Scholar] [CrossRef] [PubMed] Toja, P.M.; Branzi, G.; Ciambellotti, F.; Radaelli, P.; De Martin, M.; Lonati, L.M.; Scacchi, M.; Parati, G.; Cavagnini, F.; Cavagnini, F. Clinical relevance of cardiac structure and function abnormalities in patients with Cushing’s syndrome before and after cure. Clin. Endocrinol. 2012, 76, 332–338. [Google Scholar] [CrossRef] You, K.H.; Marsan, N.A.; Delgado, V.; Biermasz, N.R.; Holman, E.R.; Smit, J.W.; Feelders, R.A.; Bax, J.J.; Pereira, A.M. Increased myocardial fibrosis and left ventricular dysfunction in Cushing’s syndrome. Eur. J. Endocrinol. 2012, 166, 27–34. [Google Scholar] [CrossRef] Baykan, M.; Erem, C.; Gedikli, O.; Hacihasanoglu, A.; Erdogan, T.; Kocak, M.; Kaplan, S.; Kiriş, A.; Orem, C.; Celik, S. Assessment of left ventricular diastolic function and Tei index by tissue Doppler imaging in patients with Cushing’s Syndrome. Echocardiography 2008, 25, 182–190. [Google Scholar] [CrossRef] Ainscough, J.F.; Drinkhill, M.J.; Sedo, A.; Turner, N.A.; Brooke, D.A.; Balmforth, A.J.; Ball, S.G. Angiotensin II type-1 receptor activation in the adult heart causes blood pressure-independent hypertrophy and cardiac dysfunction. Cardiovasc. Res. 2009, 81, 592–600. [Google Scholar] [CrossRef] Uziębło-Życzkowska, B.; Krzesinński, P.; Witek, P.; Zielinński, G.; Jurek, A.; Gielerak, G.; Skrobowski, A. Cushing’s Disease: Subclinical Left Ventricular Systolic and Diastolic Dysfunction Revealed by Speckle Tracking Echocardiography and Tissue Doppler Imaging. Front. Endocrinol. 2017, 8, 222. [Google Scholar] [CrossRef] Eschalier, R.; Rossignol, P.; Kearney-Schwartz, A.; Adamopoulos, C.; Karatzidou, K.; Fay, R.; Mandry, D.; Marie, P.Y.; Zannad, F. Features of cardiac remodeling, associated with blood pressure and fibrosis biomarkers, are frequent in subjects with abdominal obesity. Hypertension 2014, 63, 740–746. [Google Scholar] [CrossRef] Krzesiński, P.; Stańczyk, A.; Piotrowicz, K.; Gielerak, G.; Uziębło-Zyczkowska, B.; Skrobowski, A. Abdominal obesity and hypertension: A double burden to the heart. Hypertens. Res. 2016, 39, 349–355. [Google Scholar] [CrossRef] [PubMed] Wong, C.Y.; O’Moore-Sullivan, T.; Leano, R.; Byrne, N.; Beller, E.; Marwick, T.H. Alterations of left ventricular myocardial characteristics associated with obesity. Circulation 2004, 110, 3081–3087. [Google Scholar] [CrossRef] [PubMed] Parrinello, G.; Licata, A.; Colomba, D.; Di Chiara, T.; Argano, C.; Bologna, P.; Corrao, S.; Avellone, G.; Scaglione, R.; Licata, G. Left ventricular filling abnormalities and obesity-associated hypertension: Relationship with overproduction of circulating transforming growth factor beta1. J. Hum. Hypertens. 2005, 19, 543–550. [Google Scholar] [CrossRef] [PubMed] Wang, Q.; Gao, Y.; Tan, K.; Li, P. Subclinical impairment of left ventricular function in diabetic patients with or without obesity: A study based on three-dimensional speckle tracking echocardiography. Herz 2014, 40, 260–268. [Google Scholar] [CrossRef] Feraco, A.; Marzolla, V.; Scuteri, A.; Armani, A.; Caprio, M. Mineralocorticoid Receptors in Metabolic Syndrome: From Physiology to Disease. Trends Endocrinol. Metab. 2020, 31, 205–217. [Google Scholar] [CrossRef] Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. © 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/). From https://www.mdpi.com/2077-0383/13/6/1658
    1 point
  8. Abstract In Cushing syndrome (CS), prolonged exposure to high cortisol levels results in a wide range of devastating effects causing multisystem morbidity. Despite the efficacy of treatment leading to disease remission and clinical improvement, hypercortisolism-induced complications may persist. Since glucocorticoids use the epigenetic machinery as a mechanism of action to modulate gene expression, the persistence of some comorbidities may be mediated by hypercortisolism-induced long-lasting epigenetic changes. Additionally, glucocorticoids influence microRNA expression, which is an important epigenetic regulator as it modulates gene expression without changing the DNA sequence. Evidence suggests that chronically elevated glucocorticoid levels may induce aberrant microRNA expression which may impact several cellular processes resulting in cardiometabolic disorders. The present article reviews the evidence on epigenetic changes induced by (long-term) glucocorticoid exposure. Key aspects of some glucocorticoid-target genes and their implications in the context of CS are described. Lastly, the effects of epigenetic drugs influencing glucocorticoid effects are discussed for their ability to be potentially used as adjunctive therapy in CS. epigenetic, glucocorticoids, Cushing syndrome Issue Section: Mini-review In Cushing syndrome (CS), adrenocorticotropic hormone (ACTH) hypersecretion by a pituitary adenoma or an ectopic source, or autonomous cortisol hypersecretion by an adrenal tumor, induces chronic endogenous hypercortisolism with loss of the cortisol circadian rhythm (1). CS is more prevalent in women than men and frequently occurs in the fourth to sixth decades of life (2). Glucocorticoids (GC) have extensive physiological actions and regulate up to 20% of the expressed genome, mainly related to the immune system, metabolic homeostasis, and cognition. Therefore, the prolonged exposure to high cortisol levels results in a wide range of devastating effects, including major changes in body composition (obesity, muscle atrophy, osteoporosis), neuropsychiatric disturbances (impaired cognition, depression, sleep disturbances), the metabolic syndrome (obesity, hypertension, insulin resistance, and dyslipidemia), hypercoagulability, and immune suppression (3, 4). The consequences of hypercortisolism lead to compromised quality of life and increased mortality rate (5). The mortality rate in patients with CS is 4 times higher than the healthy control population (6). Risk factors such as obesity, diabetes, and hypertension contribute to the increased risk of myocardial infarction, stroke, and cardiac insufficiency. As a result, cardiovascular disease is the leading cause of the premature death in CS (5). Infectious disease is also an important cause of death in CS (5). Therefore, prompt treatment to control hypercortisolism is imperative to prevent complications and an increased mortality rate. Despite the efficacy of treatment leading to disease remission, the clinical burden of CS improves, but does not completely revert, in every patient (7). Indeed, obesity, neuropsychiatric disturbances, hypertension, diabetes, and osteoporosis persist in a substantial number of biochemically cured patients. For instance, in a study involving 118 CS patients in remission for about 7.8 years (median), resolution of comorbidities such as diabetes occurred in only 36% of cases, hypertension in 23% of cases, and depression in 52% of the cases (8). It has been proposed that epigenetic changes as a consequence of hypercortisolism is a mechanism of the persistence of some comorbidities (9-12). Epigenetics is a reversible process that modifies gene expression without any alterations in DNA sequence; frequently it is mediated by histone modification and DNA methylation together with microRNAs (13-15). GCs use the epigenetic machinery as a mechanism of action to regulate gene expression in physiological circumstances, such as metabolic actions and stress response. Its networks involve DNA and histone modifying enzymes, such as DNA methyltransferases (DNMTs), histone acetyltransferases (HATs), and histone deacetylases (HDACs) (16). (Fig. 1) The DNA methylation process catalyzed by DNMTs is usually associated with downregulation of gene expression (17). Histone modifications catalyzed by HAT enzymes induce gene transcription, while those by HDAC enzymes induce transcriptional repression (17). Drugs interfering with these enzymes (so-called epigenetic drugs) may affect the GC genomic actions confirming the interaction between GC and the epigenetic system (18, 19). Furthermore, GC can modulate HDAC and DNMT expression and activity (16, 19, 20). Based on these data it might be speculated that in CS, epigenetic modifications induced by long-term GC exposure plays a role in the development of the disease-specific morbidity (9, 10). Figure 1. Open in new tabDownload slide Glucocorticoid (GC) and its epigenetic machinery. GC through its receptor interacts with DNA and histone modifying enzymes, such as DNA methyltransferases (DNMTs), histone acetyl transferases (HATs), and histone deacetylases (HDAC) to modulate gene expression. In this review we provide an overview of epigenetic aspects of GC action in physiological conditions and in the context of CS. We start with a detailed characterization of how GC, using the epigenetic system, can change chromatin structure in order to activate or silence gene expression. (Fig. 2) Subsequently, we describe the role of epigenetic mechanisms in the regulation of expression of several GC-target genes related to CS. Finally, we present the current evidence of epigenetic changes caused by the long-term of GC exposure and the potential use of epidrugs influencing GC actions. Figure 2. Open in new tabDownload slide Epigenetic mechanisms of the glucocorticoid action to regulate gene expression. The GR is located in cytoplasm in a multi-protein complex; after GC binding, GR dissociates from the multi-protein complex, crosses the nuclear membrane, dimerizes, and binds to the GRE of the target gene. One of the mechanisms of action of GC is through the recruitment of co-regulators together with epigenetic enzymes, such as HAT, to change the chromatin structure, resulting in activation of gene transcription. Also, GR decreases gene expression by tethering other transcriptional factors and recruiting HDAC2, causing histone deacetylation, which leads to a repressed chromatin. GC can cause hypomethylation through downregulation in the expression of DNMT1. Abbreviations: Ac, acetylation; DNMT1, DNA methyltransferase 1; GC, glucocorticoid; GR, glucocorticoid receptor; GRE, glucocorticoid responsive elements; HAT, histone acetyltransferase; HDAC, histone deacetylases; Me: methylation. Search Strategy A search of the PubMed database was conducted using the advanced search builder tool for articles in the English language on the following terms “glucocorticoids,” “glucocorticoid receptor,” “Cushing,” “hypercortisolism,” “epigenetic,” “DNA methylation,” “histone deacetylase,” “histone acetyltransferase,” “microRNA” “fkbp5,” “clock genes,” and “POMC.” Moreover, references were identified directly from the articles included in this manuscript. The articles were selected by the authors after being carefully analyzed regarding their importance and impact. Epigenetic Aspects of Genomic Action of Glucocorticoids GCs regulate gene expression positively or negatively. GC-responsive genes include genes encoding for proteins associated with inflammation, metabolic processes, blood pressure and fluid homeostasis, apoptosis, cell cycle progression, circadian rhythm, and intracellular signaling (21). The GC actions are cell type–specific (22). For instance, in an in vitro study, the comparison of GC-expressed genes between 2 cell lines, corticotroph (AtT20) and mammary (3134) cell lines, showed a different set of GC-regulated genes, revealing the cell type–specific nature of GC effects (23). GC function depends on the accessibility of glucocorticoid receptor (GR)-binding sites in the DNA of the target tissue, which in turn is mostly established during cell differentiation. Therefore, different chromatin organization explains the distinct GR-binding sites among different tissues (22, 24, 25). The chromatin accessibility is determined by histone modifications such as acetylation, methylation, phosphorylation, and/or DNA methylation, processes that are both dynamic and reversible (26). Furthermore, gene expression is regulated in a GC-concentration-dependent manner which is tissue-specific. Only a few genes can be upregulated or downregulated at low concentrations of GC. For example, a dose of dexamethasone (Dex) as low as 0.5 nM selectively activated PER1 (period 1, transcription factor related to circadian rhythm) expression in lung cancer (A549) cells (21, 27). Additionally, continuous GC exposure or pulsed GC (cortisol fluctuation during circadian rhythm) may cause different responses with respect to gene expression (26, 28). For example, constant treatment with corticosterone induced higher levels of PER1 clock gene mRNA expression compared with pulsatile treatment, as demonstrated in an in vitro study using 3134 cell line (28). The time course for gene expression in response to Dex is fast, with repression occurring slightly slower compared to activation. Half of activated and repressed genes are detected within, respectively, about 40 minutes and 53 minutes following Dex exposure (21). In short, the transcriptional output in response to GC depends on cell type, as well as on the duration and intensity of GC exposure (21, 24, 26, 27). GCs act as a transcriptional regulatory factor resulting in activating or repressing the expression of genes. The GC exerts its function through binding to corticosteroid receptors, specifically, the mineralocorticoid receptor and the GR, members of the nuclear receptor superfamily (29, 30). Glucocorticoid Receptor The GR is located in the cytoplasm in a chaperone complex which includes heat-shock proteins (70 and 90) and immunophilins (such as FK506 binding protein [FKBP5]). Cortisol diffuses across the cell membrane and binds with high affinity to the GR. The activated GR bound to GC dissociates of the multi-protein complex and is transferred to the nucleus, where it ultimately regulates gene expression (26, 31). GR is a transcription factor encoded by nuclear receptor subfamily 3, group C member 1 (NR3C1) gene, located in chromosome 5, and consisting of 9 exons. It is composed of 3 major functional domains, namely a DNA binding domain (DBD), the C-terminal ligand-binding domain (LBD) and the N-terminal domain (NTB). The LBD recognizes and joins the GC. NTB contains an activation function-1 (AF1) which connects with co-regulators and the members of the general transcription machinery to activate target genes. The DBD comprises 2 zinc fingers motifs that are able to identify and bind to glucocorticoid responsive elements (GREs) (32, 33). GRα is the most expressed and functionally active GR. GRβ is another isoform which is the result of an alternative splicing in exon 9 of the GR transcript. The difference between the 2 isoforms is the distinct ligand-binding domain in GRβ. This variance prevents the GRβ from binding to GC. In fact, the GRβ counteracts GRα function by interfering with its binding to a GRE in the target gene, and GRβ expression is associated with GC resistance (32). In addition, GRβ has its own transcriptional activity which is independent and distinct from GRα (34). Another splice variant of human GR, GRγ, is associated with GC resistance in lung cell carcinoma and childhood acute lymphoblastic leukemia (33, 35). There is an additional amino acid (arginine) in the DBD of the GRγ that reduces, by about half, the capacity to activate or suppress the transcription of the target gene, as compared with GRα (32). One study identified GRγ in a small series of corticotroph adenomas (36). Glucocorticoid Mechanism of Action The GR-GC complex induces or represses gene expression directly by binding to DNA, indirectly by tethering other transcription factors or yet in a composite manner that consists in binding DNA in association with binding to other co-regulators (35, 37). The GR has the ability to reorganize the chromatin structure to become more or less accessible to the transcriptional machinery. In the classical mechanism of direct induction of gene expression, the GR dimerizes and binds to a GRE in DNA. The receptor recruits co-regulators, such as CREB binding protein, which has intrinsic histone acetyltransferase (HAT) activity that modifies the chromatin structure from an inactive to an active state. This model, called transactivation, upregulates the expression of some genes related to glucose, protein, and fat metabolism. Gene repression, on the other hand, is accomplished by GR binding to a negative GRE (nGRE) leading to the formation of a chromatin remodeling complex composed by co-repressor factors, such as NCOR1 and SMRT, and histone deacetylases (HDACs), that ultimately turn chromatin less accessible and suppress gene transcription. The gene repression through direct binding events occurs less frequently when compared to gene induction (25, 35, 38). Another mechanism of GC action is through binding to other transcription factors (tethering). In case of switching off inflammatory genes, GR binds to transcriptional co-activator molecules, such as CREB binding protein with intrinsic HAT activity, and subsequently recruits HDAC2 to reverse histone acetylation, thus resulting in a suppression of the activated inflammatory gene (39). In the same model, GC interacts with other cofactors, such as the STAT family, to induce chromatin modifications resulting in increased gene expression (26). Furthermore, the transcriptional dynamics of some genes follow a composite manner. In this model, GR, in conjunction with binding to GRE, also interacts with cofactors in order to enhance or reduce gene expression (35). GCs can also modulate gene expression by influencing the transcription of epigenetic modifiers. An experimental study demonstrated that GC mediated the upregulation of HDAC2 in rats exposed to chronic stress, which in turn decreased the transcription of histone methyltransferase (Ehmt2) that ultimately upregulated the expression of Nedd4. Nedd4 is a ubiquitin ligase, expression of which has been related to cognitive impairment (40). Additionally, GC was found to interact with another epigenetic eraser, namely JMJD3, a histone demethylase, suppressing its transcription in endothelial cells treated with TNFα that led to decreased expression of other genes related to the blood-brain barrier (41). GCs have the ability to induce (de)methylation changes in DNA, ultimately affecting gene expression. The DNA methylation process triggered by GC involves the family of DNA methyltransferases (DNMT) and ten-eleven translocation (TET) protein (20, 42-44). The DNMT, DNMT1, DNMT3A, and DNMT3B are able to transfer a methyl group to a cytosine residue in DNA, forming 5-methylcytosine (5mC), which negatively impacts gene expression. In contrast, TET protein chemically modifies the 5mC to form 5-hydroxymethylcytosine (5hmC), which ultimately leads to unmethylated cytosine, positively influencing gene expression (45). Glucocorticoids mainly induce loss of methylation events rather than gain of methylation across the genome (11, 46). The DNA demethylation process can be either active or passive. The active mechanism is linked to the upregulation of TET enzyme expression that follows GC treatment, which was described in retinal and osteocyte cell line model studies (42, 43). The passive demethylation event involves the downregulation (Fig. 2) or dysfunction of DNMT1. DNMT1 is responsible for maintaining the methylation process in dividing cells (45). In case of GC exposure, GC can cause hypomethylation through downregulation in the expression of DNMT1, a process described in the AtT20 corticotroph tumor cell model, or through GC hindering DNMT activity, particularly DNMT1, as demonstrated in the retinal cell (RPE) line (20, 42, 44). Glucocorticoid-Induced Epigenetic Changes There are several molecular mechanisms connecting GR activation and epigenetic modifications ultimately affecting gene expression (Fig. 2). As described above, GC uses epigenetic machinery, such as DNA and histone modifying enzymes, to restructure the chromatin in order to induce or silence gene transcription (16, 47). In an in vitro study using murine AtT20 corticotroph tumor and neuronal cell lines, after chronic GC exposure followed by a recovery period in the absence of GC, the cells retained an “epigenetic memory” with persistence of loss of methylation content in FKBP5 gene but with no increased gene expression at baseline. The functionality of this “epigenetic memory” only became evident in a second exposure to GC, when the cells responded sharply with a more robust expression of FKBP5 gene compared to the cells without previous exposure to GC (44). Another in vitro study, using a human fetal hippocampal cell line, confirmed long-lasting DNA methylation changes induced by GC. The cells were treated for 10 days with dexamethasone, during the proliferative and cell differentiation phases of the cell line, followed by 20 days without any treatment. The second exposure to GC resulted in an enhanced gene expression of a subset of GC-target genes (48). Additionally, using an animal model subjected to chronic stress, a distinct gene expression profile was demonstrated in response to acute GC challenge compared to those without chronic stress history. The proposed mechanism was that chronic stress resulted in GC-induced enduring epigenetic changes in target genes, altering the responsiveness to a subsequent GC exposure (49). In general, it seems that the majority of differential methylation regions (DMRs) induced by GC are loss of methylation rather than gain of methylation. In an experimental study, an association between hypomethylation and GC exposure was demonstrated in mice previously exposed to high levels of GC. Further analysis demonstrated that the genes linked with DMR were mostly related to metabolism, the immune system, and neurodevelopment (11). Human studies have also shown that excess of cortisol can induce modifications in DNA methylation. DNA methylation data obtained from whole blood samples from patients with chronic obstructive pulmonary disease (COPD) treated with GC revealed DMR at specific CpG dinucleotides across the genome. These DMR were confirmed by pyrosequencing and annotated to genes, such as SCNN1A, encoding the α subunit of the epithelial sodium channel, GPR97, encoding G protein coupled receptor 97, and LRP3, encoding low-density lipoprotein receptor-related protein 3 (50). Furthermore, it has been proposed that the negative impact of chronic GC exposure on the immune system, which increases the risk of opportunistically infections, may be epigenetically mediated (51). In a clinical study, using whole blood samples, an analysis of genome-wide DNA methylation was performed on patients before and after exposure to GC (51). Long-term GC exposure disrupts, through a persistent modification of the cytosine methylation pattern, the mTORC1 pathway which affects CD4+ T cell biology (51). Taken together, these data clearly show the interplay between GC signaling and methylation and histone modifications processes suggesting that GC interferes in the epigenetic landscape modulating gene expression. It is possible that most of these GC-induced epigenetic events are dynamic and temporary, while others may persist leading to long-lasting disorders. Further research to provide insight into what makes some events reversible is warranted. Epigenetic Changes as a Consequence of Long-Term Glucocorticoid Exposure in Cushing Syndrome The comorbidities associated with CS are associated with increased mortality mainly due to cardiovascular events (52). GC-induced comorbidities in CS may be at least in part epigenetically mediated. Previous study using whole blood methylation profile demonstrated that specific hypomethylated CpG sites induced by GC were associated with Cushing comorbidities, such as hypertension and osteoporosis (46). The study identified a methylator predictor of GC excess which could be used as a biomarker to monitor GC status (46). The long-term exposure to high cortisol levels may be crucial for the persistence of some morbidities in CS through epigenetic changes. Hypercortisolism-induced persistent changes in visceral adipose tissue gene expression through epigenetic modifications was investigated in a translational study (12). This study combined data from patients with active CS and data from an animal model of CS in active and remitted phase. Interestingly, the study demonstrated long-lasting changes in the transcriptome of adipose tissue that were associated with histone modifications induced by GC. Therefore, these epigenetic fingerprints observed even after the resolution of hypercortisolism may elucidate the mechanism of persistent modifications in gene expression in the visceral adipose tissue (12). With regard to the persistence of GC-induced DMR, a genome-wide DNA methylation analysis showed a lower average of DNA methylation in patients in remission of CS compared to controls. Interestingly, the most common biologically relevant affected genes were retinoic acid receptors, thyroid hormone receptors, or hormone/nuclear receptors, important genes related to intracellular pathways and regulators of gene expression (9). In summary, this large body of evidence supports the concept that prolonged GC exposure modulates the epigenetic landscape across the genome by inducing DMR and histone modifications. Some epigenetic modifications are persistent, and this may partially explain the incomplete reversibility of some of CS features following clinical remission. Glucocorticoid-Target Genes in Cushing Syndrome A detailed identification and characterization of GC-target genes may shed light in the understanding of the pathophysiology and treatment response in patients with CS. For instance, the GC regulation of pro-opiomelanocortin (POMC) expression as part of the physiologic GC negative feedback may be impaired in Cushing disease (CD), which is an important mechanism for the maintenance of high GC levels (53). Another example is the interaction between GC and clock genes, which may interfere in the loss of the GC circadian rhythm and may contribute to metabolic disorders in CS (54). Furthermore, the suppressive action of GC on drug targets, such as the somatostatin receptor (subtype 2), may influence the efficacy of first-generation somatostatin receptor ligands in normalizing cortisol levels in CD (55). Here we describe how GCs using epigenetic machinery influence the expression of important target genes and their implications in CS. FKBP5 FK506 binding protein (FKBP5) plays an important role in the regulation of hypothalamic-pituitary-adrenal (HPA) system (56). As part of the GC negative feedback loop, GC binds to hypothalamic and pituitary GR. In the cytoplasm, GR is bound to a multi-protein complex including FKBP5. FKBP5 modulates GR action by decreasing GR binding affinity to GC and by preventing GR translocation from cytoplasm to nucleus (57, 58). In other words, an increase of FKBP5 expression is inversely correlated with GR activity and results in GC resistance leading to an impaired negative feedback regulation in the HPA axis (59). FKBP5 is a GC-responsive gene; its upregulation by GC is part of an intracellular negative short-feedback loop (60). The mechanism by which GC regulates FKBP5 expression was shown to include inhibition of DNA methylation (44). In a model for CS, mice treated with corticosterone for 4 weeks had a reduced level of DNA methylation of FKBP5 in DNA extracted from whole blood, which was strongly correlated in a negative manner with GC concentration. Interestingly, a negative correlation was also observed between the degree of FKBP5 gene methylation measured at 4 weeks of GC exposure and the percentage of mice visceral fat (61). Accordingly, previous studies have provided compelling evidence of decreased methylation in the FKBP5 gene in patients with active CS compared to healthy control (10, 46). Even in patients with CS in remission, previous data have suggested a small decrease in FKBP5 methylation levels compared to healthy controls (9, 10). In an in vitro study, it was demonstrated that, by decreasing DNMT1 expression, GC is able to reduce FKBP5 methylation levels and, therefore, increase its expression (44). Likewise, FKBP5 mRNA is also sensitive to GC exposure. A time-dependent increase in blood FKBP5 mRNA after single-dose prednisone administration has been demonstrated in healthy humans (62). Accordingly, patients with ACTH-dependent CS had higher blood FKBP5 mRNA levels compared with healthy controls, and after a successful surgery, FKBP5 mRNA returned to baseline levels (63). Furthermore, in another study, blood FKBP5 mRNA was inversely correlated with FKBP5 promoter methylation and positively correlated with 24-hour urine free cortisol (UFC) levels in patients with CS (46). Taken together, this fine-tuning of FKBP5 DNA methylation and mRNA according to the level of GC suggests that FKBP5 can be used as a biomarker to infer the magnitude of GC exposure. POMC and Corticotropin-Releasing Hormone The partial resistance of the corticotroph adenoma to GC negative feedback is a hallmark of CD. Indeed, the lack of this inhibitory effect constitutes a method to diagnose CD, that is, with the dexamethasone suppression test. One of the mechanisms related to the insensitivity to GC can be attributed to GR mutations which are, however, rarely found in corticotrophinomas (64). Another mechanism that was uncovered in corticotroph adenomas is an overexpression of the HSP90 chaperone resulting in reduced affinity of GR to its ligand and consequently GR resistance (53, 65). In addition, the loss of protein expression of either Brg1, ATPase component of the SWI/SNF chromatin remodeling complex, or HDAC2 has been linked to GC resistance in about 50% of some adenomas (66). The trans-repression process on POMC transcription achieved by GC involves both the histone deacetylation enzyme and Brg1. One mechanism of corticotropin-releasing hormone (CRH)-induced POMC expression is through an orphan nuclear receptor (NR) related to NGFI-B (Nur77). NGFI-B binds to the NurRE sequence in the promoter region of POMC gene and recruits a co-activator to mediate its transcription. In a tethering mechanism, the GR directly interacts with NGFI-B to form a trans-repression complex, which contains the GR itself, Brg1, the nuclear receptor, and HDAC2; the latter being essential to block the gene expression through chromatin remodeling process (53, 66). In CD, hypercortisolism exerts a negative feedback at CRH secretion from the hypothalamus (67). The mechanism involved in GR-induced suppression of CRH expression is through direct binding to a nGRE in the promoter region of CRH gene and subsequent recruitment of repressor complexes. In a rat hypothalamic cell line, it was demonstrated that Dex-induced CRH repression occurs through coordinated actions of corepressors involving Methyl-CpG-binding protein 2 (MeCP2), HDAC1, and DNA methyltransferase 3B (DNMT3B). Possibly, GR bound to nGRE recruits DNMT3B to the promoter in order to methylate a specific region, subsequently binding MeCP2 on these methylated sites followed by the recruitment of chromatin modify corepressor HDAC1, ultimately resulting in CRH suppression. Another possibility is that 2 independent complexes, one consisting of GR with DNMT3 for the methylation and the other the MeCP2, bound to methylated region, interact with HDAC1 to induce repression (68). Clock Genes The clock system and the HPA axis are interconnected regulatory systems. Cortisol circadian rhythm is modulated by the interaction between a central pacemaker, located in the hypothalamic suprachiasmatic nuclei, and the HPA axis (69). At the molecular level, mediators of the clock system and cortisol also communicate with each other, both acting as transcription factors of many genes to influence cellular functions. In CS, the impact of chronic GC exposure on clock genes expression was recently evaluated using peripheral blood samples from patients with active disease compared with healthy subjects. The circadian rhythm of peripheral clock gene expression (CLOCK, BMAL, PER1-3, and CRY1) was abolished as a result of hypercortisolism, and that may contribute to metabolic disorders observed in Cushing patients (70). Another study, which investigated persistent changes induced by hypercortisolism in visceral adipose tissue, found that the expression of clock genes, such as PER1, remained altered in association with persistent epigenetic changes in both H3K4me3 and H3K27ac induced by hypercortisolism even after the resolution of hypercortisolism (12). This suggests that chronic exposure to GC may induce sustained epigenetic changes that can influence clock genes expression. Nevertheless, further studies are warranted to better elucidate how long-term exposure to GC impacts clock genes expression using the epigenetic machinery. Glucocorticoid Effects on MicroRNAs Along with histone modification and DNA methylation, microRNAs (miRNAs) have emerged as an epigenetic mechanism capable of impacting gene expression without changing DNA sequence (15). Interestingly, miRNA expression itself is also under the influence of epigenetic modifications through promoter methylation like any other protein-encoding genes (71). MicroRNAs are small (about 20-25 nucleotides in length) non-coding RNAs that are important in transcriptional silencing of messenger RNA (mRNA). By partially pairing with mRNA, miRNAs can either induce mRNA degradation or inhibit mRNA translation to protein. MiRNAs regulate the translation of about 50% of the transcriptome, allowing them to play an important role in a wide range of biological functions, such as cell differentiation, proliferation, metabolism, and apoptosis under normal physiological and pathological situations. Some miRNAs can be classified as oncogenes or tumor suppressing genes, and aberrant expression of miRNAs may be implicated in tumor pathogenesis (71-73). Insight into the regulation of miRNA expression is, therefore, crucial for a better understanding of tumor development and other human diseases, including cardiac, metabolic, and neurological disorders (73, 74). There are different regulatory mechanisms involved in miRNA expression, including transcriptional factors such as GR-GC. GC may modulate miRNA expression through direct binding to GRE in the promoter region of the host gene, as observed in hemopoietic tumor cells (75). In addition to transcriptional activation, in vascular smooth muscle cells, Dex treatment induces downregulation of DNMT1 and DNMT3a protein levels and reduces the methylation of miRNA-29c promoter, resulting in an increased expression of miRNA-29c (76). Interestingly, it was demonstrated that the increased expression of miRNA-29 family (miRNA-29a, -29b, and -29c) associates with metabolic dysfunction, such as obesity and insulin resistance, which pertains to CS (77, 78). With regard to metabolic dysfunction, miRNA-379 expression was shown to be upregulated by GC and its overexpression in the liver resulted in elevated levels of serum triglycerides associated with very low-density lipoprotein (VLDL) fraction in mice (79). In obese patients, the level of hepatic miRNA-379 expression was higher compared to nonobese patients and positively correlated with serum cortisol and triglycerides (79). Hence, GC-responsive miRNA may be, at least in part, a mediator to GC-driven metabolic conditions in CS. In pathological conditions, such as seen in CS, prolonged exposure to an elevated cortisol level results in a wide range of comorbidities. It can be hypothesized that the chronic and excessive glucocorticoid levels may induce an aberrant miRNA expression that might impact several cellular processes related to bone and cardiometabolic disorders. A recent study addressed the impact of hypercortisolism on bone miRNA of patients with active CD compared to patients with nonfunctional pituitary adenomas. Significant changes in bone miRNA expression levels were observed, suggesting that the disruption of miRNA may be partially responsible for reduced bone formation and osteoblastogenesis (80). Similarly, altered expression levels of selected miRNAs related to endothelial biology in patients with CS may point to a contribution to a high incidence of cardiovascular disorders in Cushing patients (81). Therefore, dysregulated miRNAs as a consequence of high cortisol levels may underpin the development and progression of comorbidities related to CS. To the best of our knowledge, it is currently not clear whether miRNA dysregulation persists after resolution of hypercortisolism, thus contributing to the persistence of some comorbidities. This hypothesis needs to be further investigated. MicroRNA can also be used as a diagnostic tool in CS. A study was performed to identify circulating miRNA as a biomarker to differentiate patients with CS from patients with suspected CS who had failed diagnostic tests (the control group) (82). It was observed that miRNA182-5p was differentially expressed in the CS cohort compared to the control group; therefore, it may be used as a biomarker (82). However, a large cohort is necessary to validate this finding (82). In corticotroph tumors, downregulation of miRNA 16-1 expression was observed relative to normal pituitary tissue (83). In contrast, the plasma level of miRNA16-5p was found to be significantly higher in CD compared to ectopic Cushing (EAS) and healthy controls (84). This finding suggests that miRNA16-5p may be a biomarker capable to differentiate the 2 forms of ACTH-dependent Cushing (84). Epidrugs and Glucocorticoid Action in Cushing's Syndrome The interest in understanding the epigenetic mechanism of GC action in the context of CS is based on reversibility of epi-marks, such as DNA methylation and histone modifications, using epidrugs (85, 86). The biological characteristics of epigenetic drugs and their target have been extensively explored. Their effectiveness as antitumor drugs have been tested on corticotroph tumors using in vitro studies (87-89). However, a limited number of studies have explored the role of epidrugs as a therapeutic tool in reversing the genomic action of GC in CS, particularly in comorbidities induced by hypercortisolism (90, 91). The use of histone deacetylase inhibitors (HDACi) may reduce the genomic action of GC (90-92). It has been demonstrated that the use of the HDAC inhibitor valproic acid increases the acetylation level of GR, consequently attenuating the genomic action of GC. In an experimental Cushing model in rats, the use of valproic acid decreased expression of genes related to lipogenesis, gluconeogenesis, and ion regulators in the kidney that ultimately reduces hepatic steatosis, hyperglycemia, and hypertension in ACTH-infused rats (90, 91). More studies evaluating the effects of epidrugs influencing the GC actions are warranted to further elucidate the underlying mechanisms and to explore potential treatment modalities to reverse long-lasting consequences of chronic corticoid exposure. Conclusions In physiologic conditions, GC are secreted in pulses following a circadian rhythm pattern, as opposed to a constant, chronic, and high GC exposure in CS. This pathological pattern may account for numerous devastating effects observed in CS (7). Yet, the expressed genome in response to chronic GC exposure may potentially be abnormal, leading to dysregulation in clock genes, among other effects. GC levels may return to a normal circadian pattern in response to a successful treatment, but with incomplete reversibility of some CS features, which may in part be explained by epigenetic changes. The epigenetic machinery is used by GC to induce dynamic changes in chromatin to modulate gene expression. (Fig. 2) It seems that most of chromatin modifications are reversible, but some may persist resulting in long-term epigenetic changes. (Table 1) Table 1. Evidence of interaction between glucocorticoid and epigenetic machinery Epigenetic changes/epigenetic enzymes Action Histone acetylation (HAT) Glucocorticoid receptors (GR) recruit co-regulators, such as CREB binding protein (CBP), which has intrinsic histone acetyltransferase (HAT) activity that modifies the chromatin structure from an inactive to an active state (25, 33, 35). Histone deacetylation (HDAC) GR recruit histone deacetylases (HDACs) to turn chromatin less accessible and suppress gene transcription (25, 35). The trans-repression process on POMC transcription achieved by glucocorticoids (GC) involves the histone deacetylation enzyme (HDAC2). GC mediates the upregulation of HDAC2 in rats exposed to chronic stress (40). Histone demethylase (JMJD3) GC suppress transcription of JMJD3 in endothelial cells treated with TNFα (41). Histone modifications Using ChIP-seq, a study in mice treated for 5 weeks with corticosterone showed higher levels of histone modifications (H3K4me3, H3K27ac) compared to control mice. In mice after a 10-week washout period, persistence of this epigenetic fingerprint was observed, which was associated with long-lasting changes in gene expression (12). DNA methylation (DNMT3B) and histone deacetylation (HDAC1) GC mediates CRH downregulation through DNMT3B to the promoter in order to methylate a specific region and recruitment of chromatin modify corepressor HDAC (68). DNA hypomethylation GC induces downregulation of DNMT1 in AtT20 (mouse corticotroph adenoma cell line) (20). GC induces upregulation of TET enzyme expression which was described in retinal and osteocyte cell line model (42, 43). An experimental study in mice previously exposed to high levels of GC showed differentially methylated regions (DMR) induced by GC treatment, of which the majority was loss of the methylation (11). Reduced DNA methylation in FKBP5 gene was found in patients in active disease and also in remission state of Cushing syndrome (CS) as compared to a healthy control group (10). A genome-wide DNA methylation analysis showed a lower average of DNA methylation in patients in remission of CS compared to controls (9). A study using whole blood methylation profile demonstrated an association between cortisol excess and DNA hypomethylation in patients with CS (46). Open in new tab Further studies are needed to elucidate how chronic exposure to GC leads to incomplete reversibility of CS morbidities via sustained modulation of the epigenetic machinery and possibly other mechanisms. Subsequent identification of therapeutic targets may offer new perspective for treatments, for example, with epidrugs, aiming to reverse hypercortisolism-related comorbidities. Funding The authors received no financial support for this manuscript. Disclosures T.P., R.A.F., and L.J.H. have nothing to declare. Data Availability Data sharing is not applicable to this article, as no datasets were generated or analyzed during the current study. From https://academic.oup.com/jcem/advance-article/doi/10.1210/clinem/dgae151/7633538?searchresult=1
    1 point
  9. Wow - a lot has happened since I first shared this post. If you look at the timestamp, I'm writing this at 4:20 am and I've been awake for an hour, even though I'm exhausted. I have been back on Growth Hormone although it doesn't seem to do me any good. I also had my knee replaced last March and I shared more about that in Bee’s Knees: TKR, Finally! I plan to get the other one done, presumably after next summer.
    1 point
  10. Comment from the blog: Thank you Mary. I had surgery to remove pituitary tumor caused by Cushings in 2013. I am just starting to see some returning symptoms. Chronic stiff neck bloating belly gummy eyes. It feels like I am wearing a bras that is 5 times too small for me but I don’t have a bras on. A tightness across my back. 4 days ago one red spot showed up that I know is bleeding under the skin but I never knew the name of it. Now I know it is purpura because of this diagram. I know enough now that anything that looks strange on my body I take pictures of to show the doctor. Having your site is reassuring. I am still learning from it.
    1 point
  11. Even though this was from two years ago, I don't think that there have been any significant changes in the many symptoms of Cushing's. Today's News Item proclaimed loftily But will enough doctors actually allow patients to get that far or will they still blow them off?
    1 point
  12. Please join in a Virtual Town Hall Meeting on Cushing's Awareness Day! Mark your calendars for Monday, April 8, 2024, from 7 - 8 pm EST as we shed light on Cushing's syndrome with two incredible people who are living with this condition. Gain valuable insights, hear personal stories, and learn more about Cushing's syndrome from those who understand it firsthand. Don't miss this opportunity to connect, learn and show your support. Register now to secure your spot: https://www.eventbrite.com/.../cushings-awareness-day.... Let's come together to raise awareness and foster understanding. #CushingsAwareness
    1 point
  13. Somehow, the fact that April 1 was fast approaching slipped my mind this year but here we are. Every month seems to be Cushing’s Awareness Month as more and more people are learning about Cushing's. It doesn’t seem like the diagnostic process is getting any easier, though. I read posts on here, on Facebook, in my inbox and people still aren’t getting the answers they need from doctors. I’ve had my license tags (CUSH1E) for many years and just last week someone mentioned them - and I could share about Cushing’s once again. They make is so easy to start a conversation. While American actress Amy Schumer is spreading the word about Cushing’s in general, I think that in some ways she is hurting things for everyone else. CBS News reports that “she says she now feels ‘reborn’ after her diagnosis, and while some forms of Cushing can be fatal, she has a type that ‘will just work itself out.’” How many others will try to get diagnosed and only be told not to worry, that it “will just work itself out”?
    1 point
  14. YOU’RE INVITED! Webinar on Dr. Theodore Friedman’s update on medical treatment for Cushing’s disease In this informative webinar, Dr. Friedman will discuss What medicines to use to treat Cushing’s disease Side effects and timing of the medicines The use of ketoconazole for a medication trial before surgery Longer-term treatment for Cushing’s How to determine when a patient should go to surgery Sunday • March 31 • 6 PM PDT here to join the meeting or https://us02web.zoom.us/j/4209687343?pwd=amw4UzJLRDhBRXk1cS9ITU02V1pEQT09&omn=88672684111 OR +16699006833,,4209687343#,,,,*111116# US (San Jose) OR Join on Facebook Live https://www.facebook.com/goodhormonehealth Slides will be available on the day of the talk here. There will be plenty of time for questions using the chat button. For more information, email us at mail@goodhormonehealth.com
    1 point
  15. This article costs $70 to buy https://doi.org/10.1136/bcr-2024-259687 Doctors should suspect Cushing’s syndrome when they see patients with purple stretch marks and metabolic conditions such as diabetes, even if those symptoms aren’t the reasons for a medical visit, physicians in Japan wrote in a case study describing how they reached that diagnosis for a woman in her early 30s.
    1 point
  16. Abstract Pituitary surgery, a critical intervention for various pituitary disorders, has sparked ongoing debates regarding the preference between endoscopic and microscopic transsphenoidal approaches. This systematic review delves into the outcomes associated with these techniques, taking into account the recent advancements in neurosurgery. The minimally invasive nature of endoscopy, providing improved visualization and reduced morbidity, stands in contrast to the well-established track record of the conventional microscopic method. Examining outcomes for disorders such as Cushing's disease and acromegaly, the review synthesizes evidence from Denmark, Bulgaria, and China. Noteworthy advantages of endoscopy encompass higher resection rates, shorter surgery durations, and fewer complications, endorsing its effectiveness in pituitary surgery. While emphasizing the necessity for prospective trials, the review concludes that endoscopic approaches consistently showcase favorable outcomes, influencing the ongoing discourse on the optimal surgical strategies for pituitary disorders. Introduction & Background Pituitary surgery is a critical intervention for various pituitary disorders, and the choice between endoscopic and microscopic transsphenoidal approaches has been a subject of ongoing debate within the medical community. This systematic review aims to explore and analyze the outcomes associated with endoscopic and microscopic transsphenoidal pituitary surgery. As advancements in surgical techniques continue to shape the field of neurosurgery, understanding the comparative effectiveness of these two approaches becomes imperative. The endoscopic approach, characterized by its minimally invasive nature, has gained popularity for pituitary surgery in recent years [1]. Proponents argue that it provides enhanced visualization, improved maneuverability, and reduced patient morbidity. On the other hand, traditional microscopic transsphenoidal surgery has been the conventional method for decades, known for its familiarity among surgeons and established track record [2]. Several studies have investigated the outcomes of these approaches in treating pituitary disorders, including but not limited to Cushing's disease, pituitary adenomas, and other tumors. For instance, a systematic review and meta-analysis by Chen et al. compared endoscopic and microscopic transsphenoidal surgery specifically for Cushing's disease, shedding light on the effectiveness of these approaches in managing this specific condition [3]. Moreover, Møller et al. reported promising results for endoscopic pituitary surgery based on the experiences of experienced microscopic pituitary surgeons, indicating a potential shift towards the adoption of the endoscopic technique [1]. Guo et al. conducted a meta-analysis comparing the effectiveness of microscopic and endoscopic surgery for treating pituitary disorders, contributing valuable insights into the overall efficacy of these approaches [4]. This review aims to contribute to the ongoing discourse on pituitary surgery by providing a comprehensive analysis of the outcomes associated with endoscopic versus microscopic transsphenoidal approaches. By synthesizing the existing evidence, we strive to offer valuable insights that can guide both clinicians and researchers in making informed decisions regarding the optimal surgical approach for pituitary disorders. Review Materials and methods This systematic review strictly adheres to the established Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, employing a comprehensive approach to investigate the outcomes of endoscopic versus microscopic transsphenoidal pituitary surgery. The subsequent sections delineate the criteria for study inclusion, the search strategy utilized, and the methodology employed for data synthesis. Search Strategy We conducted a meticulous search across prominent electronic databases, including PubMed, Embase, and the Cochrane Library, to identify pertinent articles. Our search strategy comprised a combination of Medical Subject Headings (MeSH) terms and keywords related to pituitary surgery, encompassing both endoscopic and microscopic approaches. Boolean operators (AND, OR) were strategically employed to refine the search and identify studies meeting our predetermined inclusion criteria. The search string used for PubMed was ("Outcomes" OR "Treatment Outcome" OR "Surgical Outcome") AND ("Endoscopic Transsphenoidal Pituitary Surgery" OR "Endoscopic Pituitary Surgery" OR "Endoscopic Hypophysectomy") AND ("Microscopic Transsphenoidal Pituitary Surgery" OR "Microscopic Pituitary Surgery" OR "Microscopic Hypophysectomy" OR "Endoscopy"[Mesh] OR "Endoscopy, Surgical"[Mesh] OR "Transsphenoidal Hypophysectomy"[Mesh] OR "Microsurgery"[Mesh] OR "Microscopic Hypophysectomy"[Mesh]). Eligibility Criteria Stringent inclusion criteria were predefined to ensure the selection of high-quality and relevant studies. The included studies focused on investigating the outcomes of endoscopic versus microscopic transsphenoidal pituitary surgery. Only articles published in peer-reviewed journals within the timeframe from the inception of relevant databases until October 2023 were considered. Exclusion criteria encompassed studies on other interventions, those lacking sufficient data on surgical outcomes, and studies solely involving animal cells. Additionally, only studies in the English language with full-text availability were included, and gray literature was not considered eligible. Data Extraction and Synthesis Two independent reviewers meticulously screened titles and abstracts to identify potentially eligible studies. Subsequently, full-text articles were retrieved and evaluated for adherence to inclusion criteria. Discrepancies between reviewers were resolved through discussion and consultation with a third reviewer. Relevant data, including study design, patient characteristics, interventions, and surgical outcomes, were systematically extracted using a predefined data extraction form. Data Analysis A narrative synthesis approach was employed to summarize findings from included studies due to anticipated heterogeneity in study designs and outcome measures. Key themes and patterns related to the outcomes of endoscopic versus microscopic transsphenoidal pituitary surgery were identified and presented. Results Study Selection Process Following four database searches, 97 articles were initially identified. After eliminating eight duplicates, the titles and abstracts of the remaining 89 publications were evaluated. Subsequently, 17 potential studies underwent eligibility verification through a thorough examination of their full texts. Ultimately, three articles satisfied the inclusion criteria. No additional studies meeting the eligibility criteria were found during the examination of references in the selected articles. The entire process is visually depicted in the PRISMA flowchart (Figure 1). Figure 1: PRISMA flow diagram of the selection of studies for inclusion in the systematic review. PRISMA: Preferred Reporting Items for Systematic Reviews and Meta-Analyses Characteristics of Selected Studies Overall, three papers met the inclusion criteria. Two studies were randomized controlled trials (RCTs), one each from Bulgaria and China. One study was an observational study from Denmark. The main findings and characteristics of the included studies are mentioned in the following tables (Table 1 and Table 2). Author Year Country Study type Sample size No. of participants in the endoscopic group No. of participants in the microscopic group Main findings Møller et al. [1] 2020 Denmark Observational study 240 45 195 The study comparing endoscopic and microscopic transsphenoidal pituitary surgery revealed that the endoscopic technique exhibited advantages, achieving a higher rate of gross total resection (39% vs. 22%) and shorter surgery duration (86 minutes vs. 106 minutes). Complications within 30 days were lower with the endoscope (17% vs. 27%), and grade II complications or higher were significantly reduced (4% vs. 20%) compared to the microscopic approach. Pituitary function outcomes favored the endoscope, with fewer new deficiencies in the HPA axis (3% vs. 34%) and TSH-dependent deficiencies (15% vs. 38%). The HPG axis also showed better normalization in the endoscopic group (32% vs. 19%). Visual field impairment and postoperative improvement did not significantly differ between the two techniques. Overall, the findings suggest that endoscopic transsphenoidal pituitary surgery may offer superior outcomes compared to the microscopic approach, particularly in terms of resection rates and complication profiles. Vassilyeva et al. [5] 2023 Bulgaria RCT 83 43 40 The study compared endoscopic and microscopic transsphenoidal pituitary surgery in acromegaly patients, revealing comparable demographic profiles between the groups. Endoscopic surgery demonstrated advantages with shorter anesthesia and surgery times, as well as a reduced postoperative hospital stay. Complete tumor removal was more frequent with endoscopic adenomectomy, while microscopic surgery showed a higher rate of sub-total removal. Both techniques led to a tendency for somatic improvement, with more pronounced visual function improvement in the endoscopic group. Complications, such as liquorrhea and endocrine disorders, were generally low, with endoscopic surgery showing mainly mild complications. Disease remission rates were similar between the groups at various follow-up intervals. In conclusion, while both techniques proved effective in achieving remission, endoscopic surgery exhibited favorable outcomes in terms of efficiency and some aspects of complication profiles. Zhang et al. [6] 2021 China RCT 46 23 23 Endoscopic transsphenoidal pituitary surgery for the treatment of Cushing's disease showed comparable efficacy to microscopic transseptal pituitary surgery but with the added benefits of shorter operative time, reduced estimated blood loss, shorter hospital stays, and fewer complications. Table 1: Summary of the studies included in this systematic review. RCT: randomized controlled trial; HPA: hypothalamic-pituitary-adrenal; TSH: thyroid-stimulating hormone; HPG: hypothalamic-pituitary-gonadal Technique Møller et al. [1] Vassilyeva et al. [5] Zhang et al. [6] Male-to-female ratio (endoscopic) 25:20 17:26 13:10 Male-to-female ratio (microscopic) 107:88 16:24 12:11 Mean age in years (endoscopic) 61 43.26 55.6 Mean age in years (microscopic) 58 44.12 53.2 Functional tumors (endoscopic) 15 All functional All functional Non-functional tumors (endoscopic) 29 - - Functional tumors (microscopic) 69 All functional All functional Non-functional tumors (microscopic) 115 - - Microadenoma size (mm) (endoscopic) - 4 19 Macroadenoma size (mm) (endoscopic) - 39 4 Microadenoma size (mm) (microscopic) - 3 18 Macroadenoma size (mm) (microscopic) - 37 5 Mean operative time (min) (endoscopic) 86 142 108 Mean operative time (min) (microscopic) 106 176 174 Mean hospital stay (days) (endoscopic) - 5 2.8 Mean hospital stay (days) (microscopic) - 7 5.1 Postoperative complications (endoscopic) 2 15 3 Postoperative complications (microscopic) 39 10 8 Table 2: Summary of demographics, tumor characteristics, and postoperative outcomes of the studies included in this systematic review. The quality assessment of the selected studies was done using the Newcastle-Ottawa Quality Assessment Scale. All three studies included in this study turned out to be of high quality with a rating of 9/9 stars (Table 3). Author Selection Comparability Outcome Total stars Møller et al. [1] ★★★★ ★★ ★★★ ★★★★★★★★★ Vassilyeva et al. [5] ★★★★ ★★ ★★★ ★★★★★★★★★ Zhang et al. [6] ★★★★ ★★ ★★★ ★★★★★★★★★ Table 3: Quality assessment of the included studies using the Newcastle-Ottawa Quality Assessment Scale. Discussion This systematic review thoroughly assesses the effectiveness and results of endoscopic transsphenoidal pituitary surgery in comparison to microscopic transsphenoidal surgery, with a specific focus on pituitary adenomas, including Cushing's disease and acromegaly. The results contribute significant insights into the evolving landscape of pituitary surgery, highlighting the benefits and limitations of both surgical techniques. The selected studies offer valuable insights into the comparative outcomes. Møller et al.'s observational study in Denmark suggests that endoscopic surgery exhibits superior outcomes with higher gross total resection rates, shorter surgery duration, and fewer complications [1]. Vassilyeva et al.'s RCT in Bulgaria, focusing on acromegaly patients, indicates endoscopic advantages such as shorter anesthesia and surgery times, reduced postoperative stay, and comparable remission rates [5]. Zhang et al.'s RCT in China, specifically for Cushing's disease, suggests comparable efficacy with added benefits favoring endoscopy [6]. The endoscopic approach has been advocated for its panoramic visualization and superior mobility, which are crucial in resecting tumors while preserving normal structures [7,8]. Studies have shown a higher remission rate in endoscopic procedures for endocrine-active tumors, like growth hormone or adrenocorticotropic hormone (ACTH)-secreting adenomas, compared to the microscopic approach [9,10]. Patient comfort and recovery play a significant role in evaluating surgical methods. The endoscopic technique, by avoiding submucosal excision of nasal tissues, typically results in less postoperative pain and rhinological dysfunction. Studies, including ours, have reported shorter operative times and hospital stays with endoscopic surgery, attributed to fewer intraoperative and postoperative complications and a reduced need for wound management [11-13]. Safety is paramount to any surgical intervention. The endoscopic method has shown a decrease in common complications such as cerebrospinal fluid (CSF) leak, pituitary hormone dysfunction, and diabetes insipidus. Additionally, the endoscopic procedure exhibited fewer complications, which could be attributed to the enhanced visualization allowing for more precise tumor excision and preservation of vital structures [14-16]. In the context of acromegaly patients, the endoscopic technique has demonstrated increased radicality in tumor removal. Our review aligns with these findings, showing a higher rate of total tumor resection in endoscopic patients compared to those undergoing microscopic surgery. This improved outcome is likely due to better illumination and a wider angle of vision provided by endoscopic operations [5,17]. The endoscopic technique has shown a statistically significant improvement in visual function post surgery compared to the microscopic method. However, the frequency of certain postoperative complications, such as intraoperative liquorrhea, was higher in microscopic surgery. These differences underline the importance of the surgical technique in influencing the outcomes and complications of pituitary surgery [5,18]. Despite these findings, it is important to recognize the limitations inherent in these studies. Factors such as tumor size, density, and localization significantly influence surgical outcomes and procedure times. Additionally, the retrospective nature of many studies introduces potential biases, underscoring the need for more prospective, randomized trials for a comprehensive understanding of the long-term outcomes of these techniques. Conclusions This systematic review comparing endoscopic and microscopic transsphenoidal pituitary surgery outcomes indicates consistent evidence favoring the endoscopic approach. Notable studies from Denmark, Bulgaria, and China reveal superior results with endoscopic surgery, demonstrating higher resection rates, shorter surgery duration, and fewer complications. Endoscopy's benefits extend to patient comfort, as evidenced by shorter operative times and hospital stays. Safety considerations also support endoscopy, showing a decrease in common complications such as CSF leaks and hormonal dysfunction. Despite these strengths, the review underscores the need for prospective, randomized trials to address limitations and provide a comprehensive understanding of long-term outcomes. References Møller MW, Andersen MS, Glintborg D, Pedersen CB, Halle B, Kristensen BW, Poulsen FR: Endoscopic vs. microscopic transsphenoidal pituitary surgery: a single centre study. Sci Rep. 2020, 10:21942. 10.1038/s41598-020-78823-z Gao Y, Zhong C, Wang Y, et al.: Endoscopic versus microscopic transsphenoidal pituitary adenoma surgery: a meta-analysis. World J Surg Oncol. 2014, 12:94. 10.1186/1477-7819-12-94 Chen J, Liu H, Man S, et al.: Endoscopic vs. microscopic transsphenoidal surgery for the treatment of pituitary adenoma: a meta-analysis. Front Surg. 2022, 8:806855. 10.3389/fsurg.2021.806855 Guo S, Wang Z, Kang X, Xin W, Li X: A meta-analysis of endoscopic vs. microscopic transsphenoidal surgery for non-functioning and functioning pituitary adenomas: comparisons of efficacy and safety. Front Neurol. 2021, 12:614382. 10.3389/fneur.2021.614382 Vassilyeva N, Mena N, Kirov K, Diatlova E: Comparative effectiveness of endoscopic and microscopic adenoma removal in acromegaly. Front Endocrinol (Lausanne). 2023, 14:1128345. 10.3389/fendo.2023.1128345 Zhang T, Zhang B, Yuan L, Song Y, Wang F: Superiority of endoscopic transsphenoidal pituitary surgery to microscopic transseptal pituitary surgery for treatment of Cushing's disease. Rev Assoc Med Bras (1992). 2021, 67:1687-91. 10.1590/1806-9282.20210732 Yadav Y, Sachdev S, Parihar V, Namdev H, Bhatele P: Endoscopic endonasal trans-sphenoid surgery of pituitary adenoma. J Neurosci Rural Pract. 2012, 3:328-37. 10.4103/0976-3147.102615 Louis RG, Eisenberg A, Barkhoudarian G, Griffiths C, Kelly DF: Evolution of minimally invasive approaches to the sella and parasellar region. Int Arch Otorhinolaryngol. 2014, 18:S136-48. 10.1055/s-0034-1395265 Broersen LH, Biermasz NR, van Furth WR, de Vries F, Verstegen MJ, Dekkers OM, Pereira AM: Endoscopic vs. microscopic transsphenoidal surgery for Cushing's disease: a systematic review and meta-analysis. Pituitary. 2018, 21:524-34. 10.1007/s11102-018-0893-3 Torales J, Halperin I, Hanzu F, et al.: Endoscopic endonasal surgery for pituitary tumors. Results in a series of 121 patients operated at the same center and by the same neurosurgeon. Endocrinol Nutr. 2014, 61:410-6. 10.1016/j.endoen.2014.07.002 Zubair A, M Das J: Transsphenoidal hypophysectomy. StatPearls [Internet]. StatPearls Publishing, Treasure Island (FL); 2023. Pan X, Ma Y, Fang M, Jiang J, Shen J, Zhan R: Improvement in the quality of early postoperative course after endoscopic transsphenoidal pituitary surgery: description of surgical technique and outcome. Front Neurol. 2020, 11:527323. 10.3389/fneur.2020.527323 Aiyer RG, Upreti G: Endoscopic endo-nasal trans-sphenoidal approach for pituitary adenomas: a prospective study. Indian J Otolaryngol Head Neck Surg. 2020, 72:36-43. 10.1007/s12070-019-01725-8 Oertel J, Gaab MR, Linsler S: The endoscopic endonasal transsphenoidal approach to sellar lesions allows a high radicality: the benefit of angled optics. Clin Neurol Neurosurg. 2016, 146:29-34. 10.1016/j.clineuro.2016.04.016 Hanson M, Li H, Geer E, Karimi S, Tabar V, Cohen MA: Perioperative management of endoscopic transsphenoidal pituitary surgery. World J Otorhinolaryngol Head Neck Surg. 2020, 6:84-93. 10.1016/j.wjorl.2020.01.005 Qiao N: Endocrine outcomes of endoscopic versus transcranial resection of craniopharyngiomas: a system review and meta-analysis. Clin Neurol Neurosurg. 2018, 169:107-15. 10.1016/j.clineuro.2018.04.009 Nie D, Fang Q, Wong W, Gui S, Zhao P, Li C, Zhang Y: The effect of endoscopic transsphenoidal somatotroph tumors resection on pituitary hormones: systematic review and meta-analysis. World J Surg Oncol. 2023, 21:71. 10.1186/s12957-023-02958-2 Butenschoen VM, Schwendinger N, von Werder A, Bette S, Wienke M, Meyer B, Gempt J: Visual acuity and its postoperative outcome after transsphenoidal adenoma resection. Neurosurg Rev. 2021, 44:2245-51. 10.1007/s10143-020-01408-x From https://www.cureus.com/articles/213241-navigating-the-surgical-landscape-a-comprehensive-analysis-of-endoscopic-vs-microscopic-transsphenoidal-pituitary-surgery-outcomes#!/
    1 point
  17. Abstract We present a patient with Cushing syndrome secondary to accidental intake of corticosteroid tablets—a 66-year-old woman with a history of well-controlled hypertension, who over the course of a few weeks developed full-blown Cushing syndrome with uncontrolled blood pressure, typical central fat accumulation, and easy bruising. The clinical features further worsened upon increase of the dosage of her antihypertensive medication because of rising blood pressure. Biochemical analyses showed low cortisol and ACTH concentrations. Inspection of the patient's medications revealed that she had accidentally been taking corticosteroids tablets, prescribed for her husband, instead of antihypertensives, ie, dexamethasone 4 mg and then 8 mg, instead of candesartan at the same dose. This case highlights the necessity of a thorough review of the medications taken by patients suspected to have exogenous Cushing syndrome, including inspection of the original packaging, and not just relying on information from the patient and electronic health records. This case also highlights the need of special labeling on the packaging for the easy identification of corticosteroid-containing medications given their widespread availability. Cushing syndrome, accidental intake, exogenous Cushing syndrome, differential diagnosis Issue Section: Case Report Introduction Cushing syndrome (CS) is a disorder caused by prolonged and excessive exposure to glucocorticoids. The most common cause of CS is exogenous or iatrogenic, ie, CS caused by administration of glucocorticoids due to inflammatory, autoimmune, or neoplastic diseases. Endogenous CS is a rare condition, caused by either hypersecretion of ACTH from the pituitary gland, ectopic ACTH production, or hypersecretion of cortisol from the adrenal glands. It is of great importance to exclude exogenous CS in all patients who present with signs and symptoms compatible with the syndrome. The following case highlights the need to rule out exogenous CS via a face-to-face review of the medications taken by a patient with CS, rather than only relying on the patient's history and electronic health record. Case Presentation A 66-year-old woman was referred to our department for investigation of suspected CS. She was diagnosed with essential hypertension a couple of years earlier and was prescribed tablet candesartan 4 mg daily. Apart from an otherwise well-controlled hypertension, the patient had a history of bilateral hip replacement, the first performed in 2020 and the second 2 years later. During the 6 weeks prior to our evaluation, the patient had noticed an increasing fat accumulation around her abdomen, upper back, neck, and over the collar bones, despite minimal increase of her body weight. Moreover, the patient had developed a rounded face and increased growth of facial hair, especially on the chin, as well as thin and fragile skin that bruised easily. About 1.5 weeks before she was referred to our clinic, the dose of candesartan was increased by her general practitioner from 4 to 8 mg daily because of rapidly worsening hypertension, confirmed by monitoring 24-hour ambulatory blood pressure. Diagnostic Assessment The physical examination of the patient revealed central obesity and multiple bruises that the patient could not recall. Increased growth of fine hairs on the chin and facial plethora was present. Blood pressure was 165/88 mmHg. The patient did not have any signs of abdominal stretch marks, nor did she have any obvious muscle wasting in the arms and legs (Fig. 1). When comparing to photographs taken about 6 months prior to the examination, the differences were obvious (Fig. 2). Figure 1. Open in new tabDownload slide The patient few weeks prior to admission for evaluation of Cushing syndrome. Figure 2. Open in new tabDownload slide The patient many months before the onset of Cushing syndrome. Biochemical evaluation revealed unmeasurable plasma cortisol at 12:00 PM, 4:00 PM, and 6:00 AM (<28 nmol/L, reference 102-535 nmol/L; <1.01 μg/dL, reference 3.69-19.39 μg/dL). Serum ACTH was also undetectable (<0.2 pmol/L, reference 1.6-13.9 pmol/L; <0.91 pg/mL, reference 2.27-63.18 pg/mL), which raised suspicion of exogenous CS. The patient firmly denied any intake of anything other than her candesartan tablets. She even stated that she avoided any analgesics after the hip replacement previously the same year, nor had she received any intra-articular cortisone injection. The patient gave a very trustworthy and consistent impression, which inevitably led us to proceed to further investigation of the adrenal glands and the pituitary gland to exclude rarer forms of CS, such as cyclic CS and/or pituitary apoplexy of an ACTH-producing pituitary adenoma. The magnetic resonance imaging of the pituitary and the computed tomography of the adrenal glands were normal. Except for the low cortisol and ACTH levels, endocrine workup was unremarkable (Table 1). Table 1. Biochemical evaluation of the patient with Cushing syndrome at baseline, ie, at admission Hormone tested Value Normal Range Plasma cortisol at 08:00 AM <1.01 mcg/dL (<28 nmol/L) 3.70-19.39 mcg/dL (102-535 nmol/L) ACTH <0.91 pg/mL (<0.2 pmol/L) 7.27-63.18 pg/mL (1.6-13.9 pmol/L) TSH 1.0 mIU/L (1.0 mIU/L) 0.4-3.7 mIU/L (0.4-3.7 mIU/L) Free T4 1.01 ng/dL (13 pmol/L) 0.76-1.32 ng/dL (9.8-17 pmol/L) IGF-1 142 ng/mL (18.60 nmol/L) 38-162 ng/mL (4.98-21.22 nmol/L) Prolactin 374 mIU/L (17.58 mcg/L) 63-561 mIU/L (2.96-26.37 mcg/L) FSH 90 mIU/mL (90 IU/L) 27-133 mIU/mL (post-menopausal) (27-133 IU/L) LH 16 mIU/mL (16 IU/L) 5.2-62 mIU/mL (post-menopausal) (5.2-62 IU/L) SHBG 6.07 mcg/mL (54 nmol/L) 2.25-17.42 mcg/mL (20-155 nmol/L) Testosterone 8.65 ng/dL (0.30 nmol/L) 11.53-34.58 ng/dL (0.4-1.2 nmol/L) Estradiol <19.07 pg/mL (<70 pmol/L) <28.06 pg/mL (<103 pmol/L) (post-menopausal with no hormone substitute) Aldosterone 9.05 ng/dL 0.251 pmol/L <23.61 ng/dL (recumbent position) <655 nmol/L Renin 8.25 mIU/L 2.8-40 mIU/L (recumbent position) DHEAS 14.81 mcg/dL (0.4 µmol/L) 29.63-181.48 mcg/dL (0.8-4.9 µmol/L) HbA1c 45 mmol/mol (6.3 %) 31-46 mmol/mol (5-6.4 %) Abnormal values are shown in bold font. Values in parenthesis are International System of Units (SI). Abbreviations: ACTH, adrenocorticotropic hormone; TSH, thyroid-stimulating hormone; T4, thyroxine; IGF-1, insulin-like growth factor 1; FSH, follicle-stimulating hormone; LH, luteinizing hormone; SHBG, sex hormone binding globulin; DHEAS, dehydroepiandrosterone sulfate; HbA1c, glycated hemoglobin. Open in new tab On day 3 after admission, we noted that plasma cortisol at 8:00 AM was measurable, though still low, at 134 nmol/L (4.86 μg/dL), which reinforced our first suspicion of exogenous CS and prompted a more thorough review of the patient's medication. At this time, we asked the patient to show us the tablets that she had been taking at home and that she still carried in her purse. To the patient's frank surprise, it turned out that she was indeed carrying tablets containing 4 mg dexamethasone in the belief that they were candesartan 4 mg tablets. The dexamethasone 4 mg tablet the patient had (generic) was white, scored with a diameter of 6 mm (Fig. 3A). The candesartan 4 mg tablet the patient had been dispensed (generic) was also white, scored and with a diameter of 7 mm (Fig. 3B). Figure 3. Open in new tabDownload slide A. Tablet Dexamethasone 4 mg. White, scored, diameter 6 × 6 mm. B. Tablet Candesartan 4 mg. White, scored, diameter 7 × 7 mm. Treatment The patient was discharged with the same antihypertensive medications as prior to the deterioration and referred to her general practitioner for follow-up of blood pressure. Upon clinical evaluation 5 months after discharge, she showed no signs or symptoms of CS (Fig. 4). Figure 4. Open in new tabDownload slide The patient 5 months after the resolution of Cushing syndrome. Outcome and Follow-up Thus, the patient had accidentally been taking her husband's medication, with which the patient had been aiding her husband, and developed a surreptitious iatrogenic CS. In hindsight, the severity of the clinical features had been worsening and resulted in rapid deterioration alongside the increase of the dosage of the antihypertensives from 4 to 8 mg because of the rising blood pressure. By day 5 after admission, the patient's plasma cortisol and ACTH concentrations had normalized, as had her blood pressure. Discussion Exogenous hypercortisolism is the most common cause of CS, though seldomly published in the literature, and is mainly iatrogenic because of prolonged use of high doses of synthetic glucocorticoids prescribed for the treatment of nonendocrine diseases (1). A recent study has shown that as many as every seventh resident in western Sweden received a glucocorticoid prescription between 2007 and 2014 (2). The rising use of generic medications during the past decade has resulted in corticosteroids being available in different forms, shapes, and packages that make them less easily recognizable. In many countries, corticosteroids are available over-the-counter in almost any form, whereas a variety of agents such as herbal preparations, tonics, and skin-bleaching creams may also contain corticosteroids to the unawareness of the people using them (3, 4). There are no large studies regarding how common the unintentional use of medicines or products that contain corticosteroids. However, studies on traditional Chinese medicine have shown that illegally impure herbs and medicines containing corticosteroids are widely used, suggesting that the accidental intake of corticosteroids is more frequent than we may think (3, 5). Many cases of factitious CS have been reported as a cause of exogenous CS, which makes the diagnosis even more challenging (6-8). The Endocrine Society Clinical Practice Guidelines for the diagnosis of CS recommend that exogenous CS be always excluded before starting the investigation of endogenous CS (9). However, a specific and definitive approach for diagnosing, respectively excluding, exogenous CS is currently lacking. In a recent review, the authors recommend that in addition to asking the patient which medicines they take, the physician should review the electronic health record and ask particularly for medications that are administered via nonoral routes, as well as over-the-counter agents as mentioned earlier (10). If not confirmed by history, the physician is advised to proceed to the measurement of ACTH and/or dehydroepiandrosterone sulfate as well as screening for synthetic glucocorticoids (10). The results usually show low ACTH, dehydroepiandrosterone sulfate, and cortisol levels even though the clinical picture suggests CS. The cross-reactivity of hydrocortisone or cortisone, which is similar to endogenous steroids, in immunoassay-based measurements of plasma and urinary cortisol may show variable levels of cortisol. These measurements combined with low ACTH can make the diagnostic workup much more complex (7). Screening for exogenous substances with the help of high-performance liquid chromatography is usually positive and constructive (7). It is increasingly clear that the risk of accidental ingestion of potent medicines can have deleterious effects on health. This leads us to conclude that thorough face-to-face review of the packaging of medications taken by the patient is mandatory and can spare both physicians and patients from a series of unnecessary investigations. Given the high availability, easy access, and catastrophic adverse effects of the unintentional use of corticosteroids, we therefore propose that all corticosteroid-including medications and agents be marked with a recognizable label. Learning Points Exogenous CS should be always excluded before starting investigation of endogenous CS. Concerning exogenous CS, practitioners should always think broadly and ask for use of herbal preparations, skin-bleaching creams, and any over-the-counter products. Unintentional use of corticosteroids can still be the case even after a thorough review of the electronic records; practitioners should always inspect the medicines the patient has taken. Contributors All authors (K.K., O.R., P.T.) made equal contributions to authorship. K.K., O.R., and P.T. were involved in the diagnosis and management of this patient, as well as in manuscript submission. K.K. and P.T. authored the manuscript draft. All authors (K.K., O.R., P.T.) reviewed and approved the final draft. Funding No public or commercial funding. Disclosures None declared. Informed Patient Consent for Publication Signed informed consent was obtained directly from the patient. © The Author(s) 2024. Published by Oxford University Press on behalf of the Endocrine Society. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited. From https://academic.oup.com/jcemcr/article/2/1/luad160/7504969?login=false
    1 point
  18. Here, we report the first adult case of pancreatic yolk sac tumor with ectopic adrenocorticotropic hormone (ACTH) syndrome. The patient was a 27-year-old woman presenting with abdominal distension, Cushingoid features, and hyperpigmentation. Endogenous Cushing’s syndrome was biochemically confirmed. The ACTH level was in the normal range, which raised the suspicion of ACTH precursor-dependent disease. Elevated ACTH precursors were detected, supporting the diagnosis of ectopic ACTH syndrome. Functional imaging followed by tissue sampling revealed a pancreatic yolk sac tumor. The final diagnosis was Cushing’s syndrome due to a yolk sac tumor. The patient received a steroidogenesis inhibitor and subsequent bilateral adrenalectomy for control of hypercortisolism. Her yolk sac tumor was treated with chemotherapy and targeted therapy. Cushing’s syndrome secondary to a yolk sac tumor is extremely rare. This case illustrated the utility of ACTH precursor measurement in confirming an ACTH-related pathology and distinguishing an ectopic from a pituitary source for Cushing’s syndrome. Introduction Ectopic adrenocorticotrophic hormone (ACTH) syndrome, also termed paraneoplastic Cushing’s syndrome, can be caused by the secretion of ACTH and/or ACTH precursors from ectopic tumors. The tumors concerned secrete ACTH precursors, including unprocessed proopiomelanocortin (POMC) and POMC-derived peptides, owing to the altered post-translational processing of POMC (1). These tumors are associated with intense hypercortisolism and various complications, such as hypertension, hyperglycemia, osteoporosis, infection risks, and thrombotic tendencies (2). Distinguishing ectopic from pituitary-dependent Cushing’s syndrome is often challenging. The two conditions are classically distinguished by their variable responses to dynamic endocrine tests, including the high-dose dexamethasone suppression test, the corticotrophin-releasing-factor (CRF) test, and the desmopressin test (3). Pituitary imaging may sometimes provide a diagnosis if a pituitary macroadenoma is identified at this juncture. The gold standard for diagnosing pituitary Cushing’s is a positive inferior petrosal sinus sampling (IPSS) result. The measurement of ACTH precursors is reported to have diagnostic value in this scenario (4). The most common source of ectopic ACTH is intrathoracic tumors, including bronchial carcinoid and small cell lung cancers. Other possible sources include gut neuroendocrine tumors and medullary thyroid cancer. Recognizing the potential causes of ectopic ACTH syndrome is essential as this provides guidance in locating the causative tumor and allows tumor-directed therapies. A yolk sac tumor as a cause of ectopic ACTH syndrome has only been reported in a 2-year-old child but not in adults (5). Here, we present a case of a 27-year-old Chinese woman who had Cushing’s syndrome due to ectopic ACTH precursor production from a pancreatic yolk sac tumor. Case description A 27-year-old Chinese woman, who had unremarkable past health and family history, presented with right upper quadrant abdominal pain and nausea in early 2020. Abdominal ultrasonography was unrevealing. A few months later, she developed Cushingoid features and oligomenorrhea. At presentation, her blood pressure was 160/95 mmHg, body weight was 65.6 kg, and body mass index was 23.2 kg/m2. She had a moon face, hirsutism, proximal myopathy, bruising, thinning of the skin, and acne. She also had hyperpigmentation on the nails and knuckles of both hands (Figure 1). Figure 1 Figure 1. Cushingoid features at presentation include moon face, acne, thin skin, and easy bruising. Hyperpigmentation on the nails and knuckles was also noted. Diagnostic assessments Her 9 am and 9 pm cortisol were both >1,700 nmol/L. Her 24-h urine-free cortisol was beyond the upper measurable limit at >1,500 nmol/L. Her serum cortisol was 759 nmol/L after a 1 mg overnight-dexamethasone suppression test, confirming endogenous Cushing’s syndrome. The morning ACTH was 35 pg/mL (upper limit of normal is 46 pg/mL). After excluding a high dose-hook effect, her blood sample was concomitantly sent for ACTH measurement using two different platforms to eliminate possible interference, which might cause a falsely low ACTH reading. ACTH was 19 pg/mL (upper limit of normal is 46 pg/mL) using an IMMULITE 2000 XPI, Siemens Healthineers, Erlangen, Germany, and 17 pg/mL (reference range: 7–63 pg/mL) using a Cobas e-801, Roche Diagnostics, Indianapolis, IN, United States, therefore verifying the ACTH measurement. In view of this being ACTH-dependent Cushing’s syndrome, a high-dose-dexamethasone suppression test (HDDST) was performed, and her cortisol was not suppressed at 890 nmol/L, with ACTH 42 pg/mL. The serum cortisol day profile showed a mean cortisol level of >1,700 nmol/L (i.e., higher than the upper measurable limit of the assay) and an ACTH of 17 pg/mL. A CRF test using 100 μg of corticorelin showed less than a 50% rise in ACTH and no rise in cortisol levels (Supplementary Table S1). She suffered from multiple complications of hypercortisolism, including thoracic vertebral collapse with back pain, diabetes mellitus (HbA1c 6.7% and fasting glucose 7.6 mmol/L), and hypokalemic hypertension, with a lowest potassium level of 2.3 mmol/L. The rapid onset of intense hypercortisolism and refractory hypokalemia, as well as the responses in the HDDST and CRF tests raised the suspicion of ectopic ACTH syndrome. Tumor markers were measured. Alpha-fetoprotein (AFP) was markedly raised at 33,357 ng/mL (reference range: <9 ng/mL). Beta-human chorionic gonadotropin (beta-hCG) was not elevated. Carcinoembryonic antigen (CEA) was 4.0 ng/mL (reference range: <3 ng/mL) and CA 19–9 was 57 U/mL (reference range: <37 U/mL). The marked hyperpigmentation in the context of normal ACTH levels pointed to the presence of an underlying tumor producing circulating ACTH precursors. Hence, magnetic resonance imaging (MRI) of the pituitary gland was not performed at this juncture. ACTH precursors were measured using a specialized immunoenzymatic assay (IEMA) employing in-house monoclonal antibodies against the ACTH region and the gamma MSH region. Both monoclonal antibodies have to bind to these regions in POMC and pro-ACTH to create a signal. The patient had a level of 4,855 pmol/L (upper limit of normal is 40 pmol/L) (6). This supported Cushing’s syndrome from an ectopic source secondary to an excess in ACTH precursors. Localization studies were arranged to identify the source of ectopic ACTH precursors. Computed tomography (CT) of the thorax did not show any significant intrathoracic lesion but incidentally revealed a pancreatic mass. Dedicated CT of the abdomen confirmed the presence of a 7.9 × 5.6 cm lobulated mass in the pancreatic body; the adrenal glands were unremarkable. 18-FDG and 68Ga-DOTATATE dual-tracer positron-emission tomography-computed tomography (PET-CT) showed that the pancreatic mass was moderately FDG-avid and non-avid for DOTATATE (Supplementary Figure S1). Multiple FDG-avid nodal metastases were also present, including left supraclavicular fossa lymph nodes. Fine needle aspiration of the left supraclavicular fossa lymph node yielded tumor cells featuring occasional conspicuous nucleoli, granular coarse chromatin, irregular nuclei, and a high nuclear-to-cytoplasmic ratio. Mitotic figures were infrequent. On immunostaining, the tumor cells were positive for cytokeratin 7 and negative for cytokeratin 20. Focal expression of CDX-2, chromogranin, and synaptophysin was noted. They were negative for TTF-1, GCDPF, Gata 3, Pax-8, CD56, ACTH, inhibin, and S-100 protein. Further immunostaining was performed in view of highly elevated AFP. The tumor cells expressed AFP, Sall4, and MNF-116. They were negative for c-kit, calretinin, Melan A and SF-1. Placental ALP (PLAP) was weak and equivocal. The features were in keeping with a yolk sac tumor. Therapeutic intervention and outcome The patient had significant hypokalemic hypertension requiring losartan 100 mg daily, spironolactone 100 mg daily, and a potassium supplement of 129 mmol/day. Co-trimoxazole was given for prophylaxis against Pneumocystis jirovecii pneumonia. Metyrapone was started and up-titrated to 1 gram three times per day. However, in view of persistent hypercortisolism, with urinary free cortisol persistently above the upper measurable limit of the assay, bilateral adrenalectomy was performed. The tumor was mainly in the periadrenal soft tissue, with vascular invasion. The tumor formed cords, nests, and ill-defined lumen (Figure 2). The tumor cells were polygonal and contained pale to eosinophilic cytoplasm and pleomorphic nuclei, some with large nucleoli. Mitosis was present while tumor necrosis was not obvious. The stroma was composed of vascular fibrous tissue, with minimal inflammatory reaction. Immunohistochemical study showed that the tumor was positive for cytokeratin 7, MNF-116, AFP, and glypican-3, and also positive for Sall4 and HNF1β. The tumor cells were negative for cytokeratin 20, PLAP, CD30, negative for neuroendocrine markers including S100 protein, synaptophysin, chromogranin, and also negative for Melan-A, inhibin, and ACTH. Histochemical study for Periodic acid–Schiff–diastase (PAS/D) showed no cytoplasmic zymogen granules like those of acinar cell tumor. The features were compatible with yolk sac tumor. She was put on glucocorticoid and mineralocorticoid replacements post-operatively. Figure 2 Figure 2. Histology and immunohistochemical staining pattern of tumor specimen. (A) HE stain x 40 showing tumor cells in the soft tissue and peritoneum. (B) HE × 400 showing that the tumor forms cords, nests, and ill-formed lumen in the vascular stroma. The tumor cells are polygonal with pale cytoplasm and pleomorphic nuclei. (C) PAS/D stain showing no cytoplasmic zymogen granules. (D) Tumor is diffusely positive for cytokeratin 7. (E) Tumor is positive for AFP. (F) Tumor is positive for glypican-3. (G) Tumor is diffusely positive for HNF1β. (H) Tumor is diffusely positive for SALL4. Regarding her oncological management, she received multiple lines of chemotherapy, but the response was poor. Due to limited access to the ACTH precursor assay, serial measurement was unavailable. Treatment response was monitored by repeated imaging and monitoring of AFP. Figure 3 shows a timeline indicating the key events of the disease, showing the trends of the AFP and cortisol levels. Apart from (i) bleomycin, etoposide, and platinum, she was sequentially treated with (ii) etoposide, ifosfamide with cisplatin, and (iii) palliative gemcitabine with oxaliplatin. Next-generation sequencing showed a BRAF V600E mutation, for which (iv) dabrafenib and trametinib were given. Unfortunately, the disease progressed, and the patient succumbed approximately one year after the disease was diagnosed. Figure 3 Figure 3. Timeline with serial cortisol and alpha-fetoprotein levels from diagnosis to patient death. Discussion This case demonstrates the diagnostic value of ACTH precursor measurement in the diagnosis of ectopic Cushing’s syndrome. ACTH precursors are raised in all ectopic tumors responsible for Cushing’s syndrome and could be useful in distinguishing ectopic from pituitary Cushing’s syndrome (4). Moreover, Cushing’s syndrome due to a yolk sac tumor has been reported only once in a pediatric case, and this is the first adult case reported in the literature (5). POMC is sequentially cleaved in the anterior pituitary into pro-ACTH and then into ACTH, which is released into the circulation and binds to ACTH receptors in the adrenal cortex, leading to glucocorticoid synthesis (5, 7). Due to incomplete processing, ACTH precursors are found in normal subjects at a concentration of 5–40 pmol/L (6). Pituitary tumors are traditionally well-differentiated and can also relatively efficiently process ACTH precursors. However, this processing is less efficient in ectopic tumors that cause Cushing’s syndrome (8). Some less differentiated pituitary macroadenomas can secrete ACTH precursors into the circulation; however, these tumors are diagnosed by imaging and so do not, in general, cause problems with differential diagnosis (9). Measurement of ACTH precursors by immunoradiometric assay (IRMA) was first described by Crosby et al. (10). The assay utilized monoclonal antibodies specific for ACTH and the other binding gamma-MSH. The assay only detects peptides expressing both epitopes and therefore measures POMC and pro-ACTH. The assay does not cross-react with other POMC-derived peptides such as beta-lipotropin, ACTH, and N-POMC. Oliver et al. demonstrated that, compared to the pituitary adenomas in Cushing’s disease, all ectopic tumors responsible for Cushing’s syndrome in their study produce excessive POMC and pro-ACTH (4). The excessive production of ACTH precursors may reflect neoplasm-induced modification and amplification of POMC production. It is suggested that POMC binds to and activates the ACTH receptor because it contains the ACTH amino-acid sequence, or it is cleaved to ACTH in the adrenal glands to cause hypercortisolism (5) (Figure 4). Moreover, cleavage of POMC may produce peptides that exert mitogenic actions on adrenal cells and lead to adrenocortical growth. Outside the adrenal tissue, excessive ACTH precursors in Cushing’s syndrome caused by ectopic tumors can lead to marked hyperpigmentation. Both hypercortisolism and hyperpigmentation were observed in the reported case. Figure 4 Figure 4. Postulated pathological mechanism of ectopic ACTH precursors. In patients with ACTH-dependent Cushing’s syndrome, ectopic tumors should be distinguished from pituitary tumors. The HDDST, at a cut-off of 50% cortisol suppression, gives a sensitivity of 81% and a specificity of 67% for pituitary dependent Cushing’s syndrome (11). The CRF test provides 82% sensitivity and 75% specificity for pituitary disease (8). IPSS is the gold standard in distinguishing pituitary from ectopic tumors in Cushing’s syndrome. Utilization of CRF-stimulated IPSS provides 93% sensitivity and 100% specificity for pituitary disease. It also allows correct lateralization in 78% of patients with pituitary tumors. However, it is only available in specialized centers. In a retrospective cohort, the ACTH precursor level distinguished well between Cushing’s disease and ectopic ACTH syndrome (4). With a cut-off of 100 pmol/L, the test achieved 100% sensitivity and specificity for ectopic ACTH syndrome. More recently, this assay has been used to diagnose patients with occult ectopic ACTH syndrome, with ACTH precursors above 36 pmol/L (8). Unfortunately, the immunoassay for ACTH precursor measurement utilizes in-house monoclonal antibodies, which are not widely available. Cross-reactivity of POMC in commercially available ACTH assays ranges from 1.6% to 4.7% (12). In cases of ectopic tumors causing Cushing’s syndrome with markedly raised ACTH-precursors and intense hypercortisolism, the cross-reactivity would give significantly high ‘ACTH’ measurements to suggest an ACTH-related pathology. The degree of cross-reactivity, which is variable, should ideally be provided by the assay manufacturer as it affects result interpretation. Lower levels of ACTH precursor production might not be detected, especially by assays with low precursor cross-reactivity. Clinical vigilance is crucial in reaching the correct diagnosis. In patients with marked hypercortisolism and a normal ACTH concentration, like in this case, the measurement of ACTH precursors would allow the accurate diagnosis of Cushing’s syndrome caused by ACTH precursors. Ectopic tumors causing Cushing’s syndrome are associated with more intense hypercortisolism than Cushing’s disease (11). However, due to variable cross-reactivity, commercial ACTH assays might not accurately detect the excessive ACTH precursors responsible for the clinical syndrome. For this reason, ACTH measurements in these two conditions can significantly overlap and may not differentiate between ectopic and pituitary diseases (4). On the other hand, the more specific POMC assay described in 1996, which does not cross-react with pro-ACTH, has a low sensitivity of 80% for ectopic Cushing’s syndrome and is not now available (13). Hence, the ACTH precursor assay used in this reported case, which detects POMC and pro-ACTH, appears to provide the best diagnostic accuracy from the available literature. Serial measurement of ACTH precursors may play a role in monitoring the treatment response in an ACTH precursor secreting tumor. In the case of ectopic ACTH secretion, the corticotropic axis is slowed down and ACTH is almost exclusively of paraneoplastic origin. Immunotherapy is known to alter the functioning of the hypothalamic–pituitary corticotropic axis; however, its effect on ectopic secretions is not known. More data is required before the role of ACTH precursor measurement for disease monitoring in these scenarios can be ascertained. The incidence of endogenous Cushing’s syndrome is reported to be 2 to 4 per million people per year (14). Ectopic sources of Cushing’s syndrome are responsible for 9 to 18% of these cases. Typical sources of these ectopic tumors include bronchial carcinoid tumors, small-cell lung cancer, and gut neuroendocrine tumors. Notably, germ cell tumors, including teratomas, ovarian epithelial tumors, and ovarian endometrial tumors, are also possible ectopic sources of Cushing’s syndrome. The histological diagnosis of germ cell tumor in a non-genital site is challenging, especially for the poorly differentiated, or with somatic differentiation. Immunostaining, chromosomal, or genetic study are very important in confirming the diagnosis. AFP elevation in our case limited the differential diagnoses to germ cell tumors/yolk sac tumors, hepatocellular carcinoma, and rare pancreatic tumors. The specimen was biopsied from the retroperitoneum, and the morphology was a dominant trabecular pattern or a hepatoid pattern. It showed diffuse positive immunostaining for cytokeratin, AFP, and glypican-3. It was also diffusely and strongly positive for HNF1β and SALL4, supporting the diagnosis of yolk sac tumor. Both HNF1β and SALL4, being related with the expression of genes associated with stem cells or progenitor cells, are used as sensitive and specific markers for germ cell tumors/yolk sac tumors (15, 16). Staining related to pancreatic acinar cell carcinoma and neuroendocrine tumor were performed. PAS/D staining showed a lack of zymogen granules. A lack of nuclear β-catenin positivity was shown. Staining for neuroendocrine markers, including chromogranin and synaptophysin, was negative. Bcl-10 and trypsin were not available in the local setting. Cushing’s syndrome due to a yolk sac tumor was reported only once, in a 2-year-old child (5). The abdominal yolk sac tumor was resistant to cisplatin, with rapid disease progression, and the patient succumbed 1.5 years after initial presentation. Yolk sac tumor in the pancreas is also rare, with only 4 cases reported so far. The first case was reported in a 57-year-old woman with an incidentally detected abdominal mass (17). The tumor stained positive for AFP, PLAP, and CEA. The second case was a 70-year-old asymptomatic woman with histology showing a group of tumor cells with features of a yolk sac tumor, and another group showing features of pancreatic ductal adenocarcinoma with mucin production, suggesting a yolk sac tumor derived from pancreatic ductal adenocarcinoma (18). The tumor showed partial positivity for AFP, Sall4, glypican-3, and cytokeratin 7, as found in our case, while MNF-116 and PLAP staining results were not described. The third was in a 33-year-old man with a solitary pancreatic head mass with obstructive jaundice (19). The patient had undergone Whipple’s procedure followed by cisplatin-based chemotherapy, resulting in at least 5 years of disease remission. The latest reported case was in a 32-year-old man presenting with abdominal pain (20). Notably, initial imaging showed diffuse enlargement of the pancreas and increased FDG uptake without a distinct mass. Reassessment imaging 11 months later showed a 13 cm pancreatic mass. The initial imaging findings suggested initial intraductal growth of the tumor, as reported in some subtypes of pancreatic carcinoma. None of the reported cases of adult pancreatic yolk sac tumors were associated with abnormal hormone secretion. We reported the first adult case of pancreatic yolk sac tumor with ectopic ACTH syndrome. The case represents an overlap of two rarities. It demonstrates that pancreatic yolk sac tumor is a possible cause of ectopic ACTH syndrome. Conclusion ACTH precursor measurement helps to distinguish ectopic ACTH syndrome from Cushing’s disease. The test has superior diagnostic performance and is less invasive than IPSS. Nonetheless, the limited availability of the assay may restrict its broader use in patient management. We describe the first adult case of pancreatic yolk sac tumor with ACTH precursor secretion resulting in Cushing’s syndrome. This adds to the list of origins of ectopic ACTH syndrome in adults. Data availability statement The original contributions presented in the study are included in the article/Supplementary material, further inquiries can be directed to the corresponding author. Ethics statement Written informed consent was obtained from the individual to publish any potentially identifiable images or data in this article. Author contributions JC wrote the manuscript. JC, CW, WC, AW, KW, and PT researched the data. WC, AL, EL, YW, KT, KL, and CL critically reviewed and edited the manuscript. DL initiated and conceptualized this case report and is the guarantor of this work. All authors contributed to the article and approved the submitted version. Funding The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article. 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/fmed.2023.1246796/full#supplementary-material References 1. Stewar, PM, Gibson, S, Crosby, SR, Pennt, R, Holder, R, Ferry, D, et al. ACTH precursors characterize the ectopic ACTH syndrome. Clin Endocrinol. (1994) 40:199–204. doi: 10.1111/j.1365-2265.1994.tb02468.x PubMed Abstract | CrossRef Full Text | Google Scholar 2. Young, J, Haissaguerre, M, Viera-Pinto, O, Chabre, O, Baudin, E, and Tabarin, A. MANAGEMENT OF ENDOCRINE DISEASE: Cushing's syndrome due to ectopic ACTH secretion: an expert operational opinion. Eur J Endocrinol. (2020) 182:R29–58. doi: 10.1530/EJE-19-0877 PubMed Abstract | CrossRef Full Text | Google Scholar 3. 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) 9:847–75. doi: 10.1016/S2213-8587(21)00235-7 PubMed Abstract | CrossRef Full Text | Google Scholar 4. Oliver, RL, Davis, JR, and White, A. Characterisation of ACTH related peptides in ectopic Cushing's syndrome. Pituitary. (2003) 6:119–26. doi: 10.1023/B:PITU.0000011172.26649.df PubMed Abstract | CrossRef Full Text | Google Scholar 5. Gevers, EF, Meredith, S, Shah, P, Torpiano, J, Peters, C, Sebire, NJ, et al. Cushing syndrome in a child due to pro-opiomelanocortin (POMC) secretion from a yolk sac tumor. Eur J Endocrinol. (2017) 176:K1–7. doi: 10.1530/EJE-16-0776 PubMed Abstract | CrossRef Full Text | Google Scholar 6. Gibson, S, Crosby, SR, Stewart, MF, Jennings, AM, McCall, E, and White, A. Differential release of proopiomelanocortin-derived peptides from the human pituitary: evidence from a panel of two-site immunoradiometric assays. J Clin Endocrinol Metab. (1994) 78:835–41. PubMed Abstract | Google Scholar 7. Harno, E, Gali Ramamoorthy, T, Coll, AP, and White, A. POMC: the physiological power of hormone processing. Physiol Rev. (2018) 98:2381–430. doi: 10.1152/physrev.00024.2017 CrossRef Full Text | Google Scholar 8. Page-Wilson, G, Freda, PU, Jacobs, TP, Khandji, AG, Bruce, JN, Foo, ST, et al. Clinical utility of plasma POMC and AgRP measurements in the differential diagnosis of ACTH-dependent Cushing's syndrome. J Clin Endocrinol Metab. (2014) 99:E1838–45. doi: 10.1210/jc.2014-1448 PubMed Abstract | CrossRef Full Text | Google Scholar 9. Gibson, S, Ray, DW, Crosby, SR, Dornan, TL, Jennings, AM, Bevan, JS, et al. Impaired processing of proopiomelanocortin in corticotroph macroadenomas. J Clin Endocrinol Metab. (1996) 81:497–502. PubMed Abstract | Google Scholar 10. Crosby, SR, Stewart, MF, Ratcliffe, JG, and White, A. Direct measurement of the precursors of adrenocorticotropin in human plasma by two-site immunoradiometric assay. J Clin Endocrinol Metab. (1988) 67:1272–7. doi: 10.1210/jcem-67-6-1272 PubMed Abstract | CrossRef Full Text | Google Scholar 11. Aron, DC, Raff, H, and Findling, JW. Effectiveness versus efficacy: the limited value in clinical practice of high dose dexamethasone suppression testing in the differential diagnosis of adrenocorticotropin-dependent Cushing's syndrome. J Clin Endocrinol Metab. (1997) 82:1780–5. doi: 10.1210/jc.82.6.1780 PubMed Abstract | CrossRef Full Text | Google Scholar 12. Monaghan, PJ, Kyriacou, A, Sturgeon, C, Davies, A, Trainer, PJ, White, A, et al. Proopiomelanocortin interference in the measurement of adrenocorticotrophic hormone: a United Kingdom National External Quality Assessment Service study. Clin Endocrinol. (2016) 85:569–74. doi: 10.1111/cen.13118 CrossRef Full Text | Google Scholar 13. Raffin-Sanson, ML, Massias, JF, Dumont, C, Raux-Demay, MC, Proeschel, MF, Luton, JP, et al. High plasma proopiomelanocortin in aggressive adrenocorticotropin-secreting tumors. J Clin Endocrinol Metab. (1996) 81:4272–7. PubMed Abstract | Google Scholar 14. Beuschlein, F, and Hammer, GD. Ectopic pro-opiomelanocortin syndrome. Endocrinol Metab Clin N Am. (2002) 31:191–234. doi: 10.1016/S0889-8529(01)00025-1 CrossRef Full Text | Google Scholar 15. Yu, DD, Guo, SW, Jing, YY, Dong, YL, and Wei, LX. A review on hepatocyte nuclear factor-1beta and tumor. Cell Biosci. (2015) 5:58. doi: 10.1186/s13578-015-0049-3 PubMed Abstract | CrossRef Full Text | Google Scholar 16. Miettinen, M, Wang, Z, McCue, PA, Sarlomo-Rikala, M, Rys, J, Biernat, W, et al. SALL4 expression in germ cell and non-germ cell tumors: a systematic immunohistochemical study of 3215 cases. Am J Surg Pathol. (2014) 38:410–20. doi: 10.1097/PAS.0000000000000116 PubMed Abstract | CrossRef Full Text | Google Scholar 17. Zhang, B, Gao, S, Chen, Y, and Wu, Y. Primary yolk sac tumor arising in the pancreas with hepatic metastasis: a case report. Korean J Radiol. (2010) 11:472–5. doi: 10.3348/kjr.2010.11.4.472 PubMed Abstract | CrossRef Full Text | Google Scholar 18. Yonemaru, J, Takahashi, M, Nara, S, Ichikawa, H, Ishigamori, R, Imai, T, et al. A yolk sac tumor of the pancreas and derived xenograft model effectively responded to VIP chemotherapy. Pancreatology. (2020) 20:551–7. doi: 10.1016/j.pan.2019.12.021 PubMed Abstract | CrossRef Full Text | Google Scholar 19. Galanis, I, Floros, G, Simou, M, Kyriakopoulos, G, and Stylianidis, G. An extremely rare case of a primary pancreatic yolk sac tumor. Cureus. (2022) 14:e26007. doi: 10.7759/cureus.26007 PubMed Abstract | CrossRef Full Text | Google Scholar 20. Sui, H, Zhu, Z, Li, Z, and Luo, Y. Primary pancreatic yolk sac tumor presenting as diffusely enlarged pancreas in initial 18F-FDG PET/CT. Clin Nucl Med. (2020) 45:483–6. doi: 10.1097/RLU.0000000000003038 PubMed Abstract | CrossRef Full Text | Google Scholar Keywords: Cushing’s syndrome, ectopic ACTH syndrome, yolk sac tumor, pancreatic tumor, ACTH precursor Citation: Chang JYC, Woo CSL, Chow WS, White A, Wong KC, Tsui P, Lee ACH, Leung EKH, Woo YC, Tan KCB, Lam KSL, Lee CH and Lui DTW (2023) Cushing’s syndrome caused by ACTH precursors secreted from a pancreatic yolk sac tumor in an adult—a case report and literature review. Front. Med. 10:1246796. doi: 10.3389/fmed.2023.1246796 Received: 18 July 2023; Accepted: 20 November 2023; Published: 05 December 2023. Edited by: Alessandro Vanoli, University of Pavia, Italy Reviewed by: Petar Brlek, St. Catherine Specialty Hospital, Croatia Wafa Alaya, Hospital University Tahar Sfar, Tunisia Copyright © 2023 Chang, Woo, Chow, White, Wong, Tsui, Lee, Leung, Woo, Tan, Lam, Lee and Lui. 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: David Tak Wai Lui, dtwlui@hku.hk 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/fmed.2023.1246796/full
    1 point
  19. Key takeaways: Crinecerfont was granted FDA breakthrough therapy designation for the treatment of congenital adrenal hyperplasia. The medication met primary and secondary endpoints in a pair of phase 3 trials. The FDA granted breakthrough therapy designation for an oral non-glucocorticoid medication for the treatment of congenital adrenal hyperplasia, according to an industry press release. Crinecerfont (Neurocrine Biosciences) is a selective corticotropin-releasing factor type 1 receptor antagonist under development to lower excess adrenal androgens for people with congenital adrenal hyperplasia due to 21-hyroxylase deficiency. The medication met its primary and secondary endpoints in two phase 3 CAHtalyst trials, one assessing use of crinecerfont by children and the other by adults. In the pediatric trial, children and adolescents receiving crinecerfont had a decrease in serum androstenedione from baseline to 4 weeks. Participants receiving the medication also had a greater reduction in daily glucocorticoid at 28 weeks than placebo. As Healio previously reported, in the adult trial, crinecerfont was associated with a greater reduction in daily glucocorticoid while maintaining androgen control compared with placebo. The most common adverse events in the pediatric study were headache, fever, vomiting, upper respiratory tract infection and nasopharyngitis. Among adults, the most common adverse events were fatigue, headache and COVID-19 infection. No serious adverse events related to crinecerfont were reported. Breakthrough therapy is the latest designation granted to crinecerfont by the FDA. The medication was previously granted fast track and rare pediatric disease designations. "We are very pleased that the FDA granted breakthrough therapy designation for crinecerfont, thus recognizing both the seriousness of congenital adrenal hyperplasia and the significant unmet need currently faced by patients and families living with this condition,” Eiry W. Roberts, MD, Chief Medical Officer for Neurocrine Biosciences, said in a press release. "The outstanding safety and efficacy results from the phase 3 CAHtalyst studies in pediatric and adult patients suggest that crinecerfont has the potential to represent a substantial improvement over current standard of care in congenital adrenal hyperplasia by controlling androgen levels and allowing for reduced steroid doses. We remain on track to submit the new drug application in 2024." From https://www.healio.com/news/endocrinology/20231206/fda-grants-breakthrough-therapy-designation-for-oral-congenital-adrenal-hyperplasia-drug?utm_source=selligent&utm_medium=email&utm_campaign=news&fbclid=IwAR2WXDd3ajhKG0s2h0XD9ZQAstUkSotJYl1KLicH3gmxEPF6hvg6sZu2dCU
    1 point
  20. In this study, we will investigate the possible side effects of psoriasis patients using long-term topical corticosteroids (TCS) such as adrenal insufficiency, Cushing’s Syndrome (CS) and osteoporosis and determine how these side effects develop. Forty-nine patients were included in the study. The patients were divided into two groups based on the potency of the topical steroid they took and the patients’ ACTH, cortisol and bone densitometer values were evaluated. There was no significant difference between the two groups regarding the development of surrenal insufficiency, CS and osteoporosis. One patient in group 1 and 4 patients in group 2 were evaluated as iatrogenic CS. ACTH stimulation tests of these patients in group 2 showed consistent results with adrenal insufficiency, while no adrenal insufficiency was detected in the patient in Group 1. Patients who used more than 50g of superpotent topical steroids per week compared to patients who used 50g of superpotent topical steroids per week. It was identified that patients who used more than 50g of superpotent topical steroids had significantly lower cortisol levels, with a negatively significant correlation between cortisol level and the amount of topical steroid use ( < .01).Osteoporosis was detected in 3 patients in group 1 and 8 patients in Group 2. Because of the low number of patients between two groups, statistical analysis could not be performed to determine the risk factors. Our study is the first study that we know of that investigated these three side effects. We have shown that the development of CS, adrenal insufficiency and osteoporosis in patients who use topical steroids for a long time depends on the weekly TCS dosage and the risk increases when it exceeds the threshold of 50 grams per week. therefore, our recommendation would be to avoid long-term use of superpotent steroids and to choose from the medium-potent group if it is to be used. ABOUT THE CONTRIBUTORS Betul Erdem Department of Dermatology, Van Training and Research Hospital, Van, Turkey. Muzeyyen Gonul Department of Dermatology, Ministry of Health, Ankara Etlik City Hospital, Ankara, Turkey. Ilknur Ozturk Unsal Department of Endocrine and Metabolic Disease, Ministry of Health, Ankara Etlik City Hospital, Ankara, Turkey. Seyda Ozdemir Sahingoz Department of Biochemistry, Ministry of Health, Ankara Etlik City Hospital, Ankara, Turkey. From https://www.physiciansweekly.com/evaluation-of-psoriasis-patients-with-long-term-topical-corticosteroids-for-their-risk-of-developing-adrenal-insufficiency-cushings-syndrome-and-osteoporosis/
    1 point
  21. Abstract Background There is an increasing number of cases of aldosterone- and cortisol-producing adenomas (A/CPAs) reported in the context of primary aldosteronism (PA). Most of these patients have PA complicated with subclinical Cushing's syndrome; cases of apparent Cushing's syndrome (CS) complicated with aldosteronism are less reported. However, Co-secretory tumors were present in the right adrenal gland, a cortisol-secreting adenoma and an aldosterone-producing nodule (APN) were present in the left adrenal gland, and aldosterone-producing micronodules (APMs) were present in both adrenal glands, which has not been reported. Here, we report such a case, offering profound insight into the diversity of clinical and pathological features of this disease. Case presentation The case was a 45-year-old female from the adrenal disease diagnosis and treatment centre in West China Hospital of Sichuan University. The patient presented with hypertension, moon-shaped face, central obesity, fat accumulation on the back of the neck, disappearance of cortisol circadian rhythm, ACTH < 5 ng/L, failed elevated cortisol inhibition by dexamethasone, orthostatic aldosterone/renin activity > 30 (ng/dL)/(ng/mL/h), and plasma aldosterone concentration > 10 ng/dL after saline infusion testing. Based on the above, she was diagnosed with non-ACTH-dependent CS complicated with PA. Adrenal vein sampling showed no lateralization for cortisol and aldosterone secretion in the bilateral adrenal glands. The left adrenocortical adenoma was removed by robot-assisted laparoscopic resection. However, hypertension, fatigue and weight gain were not alleviated after surgery; additionally, purple striae appeared in the lower abdomen, groin area and inner thigh, accompanied by systemic joint pain. One month later, the right adrenocortical adenoma was also removed. CYP11B1 were expressed in the bilateral adrenocortical adenomas, and CYP11B2 was also expressed in the right adrenocortical adenomas. APN existed in the left adrenal gland and APMs in the adrenal cortex adjacent to bilateral adrenocortical adenomas. After another surgery, her serum cortisol and plasma aldosterone returned to normal ranges, except for slightly higher ACTH. Conclusions This case suggests that it is necessary to assess the presence of PA, even in CS with apparent symptoms. As patients with CS and PA may have more complicated adrenal lesions, more data are required for diagnosis. Peer Review reports Background Because both adrenal Cushing's syndrome and primary aldosteronism (PA) can manifest as adrenocortical adenomas, it is difficult to distinguish between them on the sole basis of adrenal computed tomography (CT). There may also be multiple adenomas with different functions in the same adrenal gland [1], which also leads to the difficulty in the interpretation of adrenal vein blood collection results. With the increased reports on cases of PA complicated with subclinical Cushing's syndrome in clinical practice, increasing attention is being given to the screening of PA complicated with subclinical Cushing's syndrome. However, PA screening may be ignored in the diagnosis and treatment of adrenal Cushing's syndrome. Although it has been reported that PA with a diameter > 2 cm may be complicated with aldosterone- and cortisol-producing adenomas (A/CPAs) [2], cases of apparent Cushing's syndrome complicated with PA are less well known. Recently, Y. Fushimi et al. [3] reported a case of apparent Cushing's syndrome complicated with PA. The cortisol-producing enzyme cytochrome P450 (CYP) 11B1 was diffusely expressed in the adenoma, but based on staining, the aldosterone synthase CYP11B2 was significantly expressed in the adjacent adrenal cortex. This finding indicated that aldosterone-producing micronodules (APMs) in the adjacent adrenal cortex may be the pathological basis of PA. Here, a case of bilateral co-secretory lesions presenting with coexisting Cushing syndrome and primary aldosteronism detected by AVS and confirmed by immunohistochemical analysis after surgical resection is reported. Moreover, APMs were found in the adrenal cortex adjacent to bilateral adrenocortical adenomas; an aldosterone-producing nodule was detected adjacent to the unilateral adenoma. Case presentation A 45-year-old female patient was admitted to the adrenal disease diagnosis and treatment centre in West China Hospital of Sichuan University due to "increased blood pressure, weight gain for one year and facial oedema for half a year". After nifedipine controlled-release tablets 30 mg daily and terazosin 2 mg daily were applied, the blood pressure of this patient was still as high as 179/113 mmHg. She had no family history of endocrine disease or malignant tumour. Her body mass index (BMI) was 25.6 kg/m2 at admission, with a moon-shaped face, fat accumulation on the back of the neck and thin skin. Hormonal, glucose, renal function, lipid, and blood electrolyte tests were completed, and the physiological rhythm of cortisol had disappeared. Aldosterone-renin-angiotensin system (RAAS) results showed a significant decrease in renin activity and a significantly higher aldosterone/renin ratio (ARR) (as provided in Table 1). Dynamic testing for hormones was conducted, and the results were as follows: (i) in terms of the saline infusion test (SIT) in supine position, the before and after aldosterone level was 17.03 ng/dL and 15.45 ng/dL, respectively; (ii) in terms of the captopril challenge test (CCT), the before and after aldosterone level was 18.49 ng/dl and 15.25 ng/mL, respectively, with an inhibition rate of 17.52%; (iii) in terms of the standard low-dose dexamethasone suppression test, the before and after serum cortisol level was 467.9 nmol/L and 786.3 nmol/L, respectively; the before and after 24-h urine free cortisol (24-h UFC) level was 332.3 µg/24 and 480.4 µg/24, respectively. An enhanced CT scan revealed adenoma lesions in both adrenal glands (Fig. 1a and b). Bone mineral density measurement with dual-energy X-ray absorptiometry indicated osteoporosis. Chest CT showed old fractures of the 9th rib on the left side and the 2nd rib on the right side. Table 1 Peripheral blood laboratory data for this case Full size table Fig. 1 Adrenal CT of the patient: A nodule with a size of approximately 1.6 × 1.5 cm was found in the left adrenal gland, and a nodule with a size of approximately 2.2 × 1.8 cm was found in the right adrenal gland. Irregular mild to moderate enhancement was on enhanced CT, and the surrounding fat gap was clear Full size image Based on the above clinical features, the patient was diagnosed with "non-ACTH-dependent Cushing's syndrome complicated with PA". To assess lateralization, adrenal vein sampling (AVS) stimulated by ACTH was performed after obtaining informed consent. The results showed no lateralization of cortisol and aldosterone secretion (Table 2). Table 2 Results of AVS Full size table After communicating with the patient, the left adrenocortical adenoma was first removed by robot-assisted laparoscopic resection; the thickened adrenal cortex near the left adrenocortical adenoma was also resected during the surgery. The pathological report revealed adrenocortical adenoma, the Weiss score was 1, and immunohistochemistry showed weak CYP11B1 expression in the adenoma and positive CYP11B2 expression in an adjacent nodule. Hypertension was not alleviated after surgery. One month later, purple lines appeared on both sides of the lower abdomen, groin area and inner thigh, accompanied by weight gain, apparent systemic joint pain and fatigue in both lower limbs. The patient was readmitted to the hospital, and examination revealed orthostatic ALD at 11.99 ng/dL, PRA at 0.08 ng/mL/h, angiotensin II at 39.38 ng/L (reference range: 55.3–115.3 ng/L) and ARR at 149.88 (ng/dL)/(ng/mL/h). In addition, ACTH was 2.37 ng/L, serum cortisol was 352.30–353.50–283.90 nmol/L at 8 h-16 h-24 h, 24-h UFC was 112.8 µg, and serum cortisol was 342.10 nmol/L in the morning after the 1 mg dexamethasone suppression test. Enhanced CT of the kidneys and adrenal glands showed no solid nodules or masses in the left adrenal gland, though a nodule with a size of approximately 2.2*1.8 cm was detected in the right adrenal gland. Enhanced CT showed irregular mild to moderate enhancement. Therefore, the diagnosis was still "non-ACTH-dependent Cushing's syndrome complicated with PA". Subsequently, the right adrenocortical adenoma and the thickened adrenal cortex near the right adrenocortical adenoma were removed by robot-assisted laparoscopic resection. The pathological report indicated adrenocortical adenoma, and immunohistochemistry showed diffuse homogeneous expression of CYP11B1 and CYP11B2. Antibodies against CYP11B1 (MABS502) and CYP11B1 (MABS1251) were purchased from the Millipore Corporation. There were APMs in the adrenal cortex adjacent to the bilateral cortical adenomas. The fluorescence staining image of the left cortical adenoma is shown in Fig. 2. The immunohistochemistry image of the left adrenal gland is given in Fig. 3 and that of the right adrenal gland in Fig. 4. The immunofluorescence method used in this study was indirect immunofluorescence double staining procedure. Paraffin-embedded human adrenal tissues were prepared using heat-induced epitope retrieval after deparaffinization. Tissue sections were blocked with 5% goat serum in PBS, pH 7.4, containing 0.5% SDS, for 1 h. The slides were incubated with individual primary antibodies at 4℃ overnight, followed by incubation with Alexa Fluor 488-, and Alexa Fluor 647-conjugated secondary antibodies specific to the species of the primary antibodies with DAPI for immunofluorescence staining. Antibodies used included anti-CYP11B1 (Millipore, Cat. No. MABS502, 1:100), anti-CYP11B2(Millipore, Cat. No. MABS1251, 1:100), Alexa Fluor 488-conjugated anti-rat IgG secondary antibody (CYP11B1; Green) and Alexa Fluor 647-conjugated anti-mouse IgG secondary antibody (CYP11B2; Red). Nuclei were stained with DAPI. Fig. 2 Routine hematoxylin and eosin (H&E) staining and immunofluorescence of the left adrenocortical adenoma (green represents expression of CYP11B1 and red that of CYP11B2). This adrenocortical adenoma and the surrounding cortex was cut into three parts. A and C show the overall appearance of the resected portion, with a nodule adjacent to the adenoma. B shows a neoplastic lesion formed by clear cells (aldosterone-producing cell) within nodules, lacking a fibrous envelope. C clearly shows the weak and diffuse expression of CYP11B1 in adrenocortical adenoma and CYP11B2 expression in a nodule in the cortex adjacent to the adenoma. D shows local enlargement of the aldosterone-producing nodule and three aldosterone-producing micronodules adjacent to it Full size image Fig. 3 Resected adrenocortical adenoma and part of the adrenal cortex on the left side. A shows expression of Aldosterone-producing micronodule CYP11B2 in the cortex adjacent to the adenoma. B shows an aldosterone-producing nodule with a diameter of approximately 2 mm. C shows weak positive expression of CYP11B1 in the adenoma and D negative expression of CYP11B1 in the aldosterone-producing nodule Full size image Fig. 4 Resected adrenocortical adenoma and part of the adrenal cortex on the right side. A and B show several Aldosterone-producing micronodules (positive expression of CYP11B2) in the cortex adjacent to the adenoma. C shows diffuse expression of CYP11B1 in the adenoma. D shows diffuse expression of CYP11B2 in the adenoma Full size image The Cushing's syndrome in this patient disappeared after surgery, and glucocorticoids were discontinued after 15 months according to medical advice. Follow-up was conducted for half a year after drug discontinuance, and the patient had no fatigue or dizziness; she was satisfied with the outcomes. Her systolic and diastolic blood pressure remained at 100–120 mmHg and 70–80 mmHg, respectively. During the most recent re-examination, the following results were obtained: (1) orthostatic ALD of 19.1 ng/dL and orthostatic renin concentration of 12.59 µIU/mL, with an aldosterone/renin ratio (ARR) of 1.52; (2) PTC at 8 AM of 247 nmol/L, ACTH of 93.55 ng/L and 24-h UFC of 26.8 µg; (3) parathyroid hormone of 3.86 pmol/L; (4) 25-OH-VitD of 119.5 nmol/L; (5) serum creatinine of 60 µmol/L; (6) serum sodium of 140.4 nmol/L, serum potassium of 3.87 mmol/L and serum calcium of 2.27 mmol/L. Discussion and conclusions Adrenal Cushing's syndrome is caused by excessive autonomic secretion of cortisol induced by adrenal cortical tumours or adrenal cortical hyperplasia; primary aldosteronism (PA) is caused by excessive autonomic secretion of aldosterone induced by adrenal cortical tumours or adrenal cortical hyperplasia. More adverse symptoms occur if aldosterone and cortisol-producing adenomas are present. Specifically, (1) it is more difficult to control hypertension; (2) the incidence of major adverse cardiovascular and cerebrovascular events would increase [4]; (3) glucose intolerance and other metabolic complications would be aggravated [5, 6]; (4) patients would be prone towards osteoporosis [7, 8]; (5) adrenal vein sampling results may be misinterpreted [9]; and (6) adrenal insufficiency may occur after surgery. Therefore, it is of great clinical significance to avoid missed diagnosis of A/CPAs. Despite many reports on A/CPAs, the majority of these patients may have subclinical Cushing's syndrome (SCS), and cases of apparent Cushing's syndrome complicated with PA are rarely reported. In the present case, the clinical manifestation of Cushing's syndrome were more apparent, and it would be appropriate to call it cortisol-aldosterone cosecretoma. Naoyoshi Onoda et al. [10] reported a case of Cushing's syndrome caused by a left adrenocortical adenoma (30 mm in diameter) and PA caused by a right adrenocortical adenoma (20 mm in diameter), and Fushimi et al. [3] reported a case of right A/CPA (25 mm*22 mm in size). Interestingly, in the present report, the patient had bilateral A/CPAs, and the clinical manifestations of Cushing's syndrome became more apparent after unilateral resection was performed. Similar to the above two cases, APMs were found in the adrenal cortex adjacent to the A/CPAs, but aldosterone-producing nodules were found near the cortisol-producing adenoma on the left side. The biochemical phenotype of APM-inducing autonomic aldosterone secretion has not been clarified. APMs can also be found in the adrenal tissue of 30% of individuals with normal blood pressure [11] and surrounding areas of APA [12, 13]. APMs do not express CYP11B1 or CYP17A1, which are necessary for the generation of cortisol [12, 14]. In our patient, the aldosterone-producing nodule in the left adrenal gland may have developed from APM. More than one-third of APMs carry known mutations in CACNA1D and ATP1A1, promoting the generation of aldosterone [14, 15]. Unfortunately, we did not perform whole-exome sequencing on the DNA of the peripheral blood and adenoma tissues of this patient. Due to the existence of APMs adjacent to the adenoma, it remains unclear whether there is a risk of the relapse of PA in these cases after resection of adrenal the adenoma. Therefore, it was necessary to conduct medical follow-up for this patient. Remi Goupil et al. performed AVS on 8 patients with cortisol-producing adenoma (CPA), and the results showed that cortisol on the CPA side was higher than that on the contralateral side (median, 6.7 times [range: 2.4–27.2]); P = 0.012]) [16]. There was no significant difference in bilateral cortisol and aldosterone concentrations after AVS in this patient, which is consistent with bilateral A/CPA. Although immunohistochemical results revealed weak expression of CYP11B1 for the first time, expression of cortisol in bilateral adrenal venous blood samples increased significantly after ACTH stimulation. Hence, cortisol was over-synthesized on both sides, and bilateral A/CPAs was definitively diagnosed. In summary, this case highlights the need for A/CPA screening. The complicated pathological features of these cases impose challenges to our understanding of this disease. Due to the presence of APMs in the adrenal cortex near bilateral adrenocortical adenomas, more clinical data are required to identify whether the disease might relapse after simple resection of the adenoma in these patients. Therefore, further medical follow-up of these patient is needed. Availability of data and materials Not applicable. Abbreviations CS: Cushing's syndrome PA: Primary aldosteronism ACTH: Adrenocorticotropic hormone UFC: Urinary free cortisol AVS: Adrenal vein sampling A/CPA: Aldosterone-and cortisol producing adenoma APN: Aldosterone-producing nodules APM: Aldosterone-producing micronodule CYP: Cytochrome P450 CT: Computed tomography PAC: Plasma aldosterone concentration PRA: Plasma renin activity ARR: Aldosterone /renin ratio References Stenman A, Shabo I, Ramström A, Zedenius J, Juhlin CC: Synchronous aldosterone- and cortisol-producing adrenocortical adenomas diagnosed using CYP11B immunohistochemistry. SAGE open medical case reports. 2019, 7:2050313x19883770. Hiraishi K, Yoshimoto T, Tsuchiya K, Minami I, Doi M, Izumiyama H, Sasano H, Hirata YJ. Clinicopathological features of primary aldosteronism associated with subclinical Cushing’s syndrome. Endocr J. 2011;58(7):543–51. Article CAS PubMed Google Scholar Fushimi Y, Tatsumi F, Sanada J, Shimoda M, Kamei S, Nakanishi S, Kaku K, Mune T, Kaneto H. Concurrence of overt Cushing’s syndrome and primary aldosteronism accompanied by aldosterone-producing cell cluster in adjacent adrenal cortex: case report. BMC Endocr Disord. 2021;21(1):163. Article PubMed PubMed Central Google Scholar Araujo-Castro M, BengoaRojano N, FernándezArgüeso M, Pascual-Corrales E, Jiménez Mendiguchía L. García Cano AMCardiometabolic risk in patients with primary aldosteronism and autonomous cortisol secretion. Case-control study. Med Clin (Barc). 2021;157(10):473–9. Article CAS PubMed Google Scholar Petramala L, Olmati F, Concistrè A, Russo R, Mezzadri M, Soldini M, De Vincentis G, Iannucci G, De Toma G, Letizia C. Cardiovascular and metabolic risk factors in patients with subclinical Cushing. Endocrine. 2020;70(1):150–63. Article CAS PubMed Google Scholar Akehi Y, Yanase T, Motonaga R, Umakoshi H, Tsuiki M, Takeda Y, Yoneda T, Kurihara I, Itoh H, Katabami T, et al. High Prevalence of Diabetes in Patients With Primary Aldosteronism (PA) Associated With Subclinical Hypercortisolism and Prediabetes More Prevalent in Bilateral Than Unilateral PA: A Large Multicenter Cohort Study in Japan. Diabetes Care. 2019;42(5):938–45. Article CAS PubMed Google Scholar Shi S, Lu C, Tian H, Ren Y, Chen T. Primary Aldosteronism and Bone Metabolism: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne). 2020;11:574151. Article PubMed Google Scholar Petramala L, Zinnamosca L, Settevendemmie A, Marinelli C, Nardi M, Concistrè A, Corpaci F, Tonnarini G, De Toma G, Letizia C. Bone and mineral metabolism in patients with primary aldosteronism. Int J Endocrinol. 2014;2014:836529. Article PubMed PubMed Central Google Scholar Späth M, Korovkin S, Antke C, Anlauf M, Willenberg HS. Aldosterone- and cortisol-co-secreting adrenal tumors: the lost subtype of primary aldosteronism. Eur J Endocrinol. 2011;164(4):447–55. Article PubMed Google Scholar Onoda N, Ishikawa T, Nishio K, Tahara H, Inaba M, Wakasa K, Sumi T, Yamazaki T, Shigematsu K, Hirakawa K. Cushing’s syndrome by left adrenocortical adenoma synchronously associated with primary aldosteronism by right adrenocortical adenoma: report of a case. Endocr J. 2009;56(3):495–502. Article CAS PubMed Google Scholar Williams T, Gomez-Sanchez C, Rainey W, Giordano T, Lam A, Marker A, Mete O, Yamazaki Y, Zerbini M, Beuschlein F, et al. International Histopathology Consensus for Unilateral Primary Aldosteronism. J Clin Endocrinol Metab. 2021;106(1):42–54. Article PubMed Google Scholar Nishimoto K, Tomlins SA, Kuick R, Cani AK, Giordano TJ, Hovelson DH, Liu CJ, Sanjanwala AR, Edwards MA, Gomez-Sanchez CE, et al. Aldosterone-stimulating somatic gene mutations are common in normal adrenal glands. Proc Natl Acad Sci U S A. 2015;112(33):E4591-4599. Article CAS PubMed PubMed Central Google Scholar Omata K, Anand SK, Hovelson DH, Liu CJ, Yamazaki Y, Nakamura Y, Ito S, Satoh F, Sasano H, Rainey WE, et al. Aldosterone-producing cell clusters frequently harbor somatic mutations and accumulate with age in normal adrenals. J Endocr Soc. 2017;1(7):787–99. Article CAS PubMed PubMed Central Google Scholar Omata K, Tomlins SA, Rainey WE. Aldosterone-producing cell clusters in normal and pathological States. Horm Metab Res. 2017;49(12):951–6. Article CAS PubMed PubMed Central Google Scholar Lalli E, Barhanin J, Zennaro MC, Warth R. Local Control of Aldosterone Production and Primary Aldosteronism. Trends Endocrinol Metab. 2016;27(3):123–31. Article CAS PubMed Google Scholar Goupil R, Wolley M, Ahmed AH, Gordon RD, Stowasser M. Does concomitant autonomous adrenal cortisol overproduction have the potential to confound the interpretation of adrenal venous sampling in primary aldosteronism? Clin Endocrinol (Oxf). 2015;83(4):456–61. Article CAS PubMed Google Scholar Download references Acknowledgements Not applicable Funding This study was supported by the Discipline Excellence Development 1.3.5 Project of West China Hospital, Sichuan University (No. ZYGD18022). Author information Authors and Affiliations Department of Endocrinology and Metabolism, Adrenal Center, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China Hongjiao Gao, Yan Ren, Tao Chen & Haoming Tian Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China Hongjiao Gao Institute of Clinical Pathology, West China Hospital of Sichuan University, Chengdu, Sichuan, China Li Li & Fei Chen Contributions HG, TC researched data and/or wrote the manuscript. LL, FC contributed to immumohistochemical staining. HT, TC, YR contributed to discussion. All authors have read and approved the manuscript. Corresponding authors Correspondence to Tao Chen or Haoming Tian. Ethics declarations Ethics approval and consent to participate Not applicable. Consent for publication Written informed consent was obtained from the patient for publication of this Case report and any accompanying images. A copy of the written consent is available for review by the Editor of this journal. Competing interests We do not have any potential conflicts of interest relevant to this article. Additional information Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Rights and permissions Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data. Reprints and Permissions Cite this article Gao, H., Li, L., Chen, F. et al. Bilateral co-secretory lesions presenting with coexisting Cushing syndrome and primary aldosteronism: a case report. BMC Endocr Disord 23, 263 (2023). https://doi.org/10.1186/s12902-023-01454-8 Download citation Received08 April 2022 Accepted24 August 2023 Published29 November 2023 DOIhttps://doi.org/10.1186/s12902-023-01454-8 Share this article Anyone you share the following link with will be able to read this content: Get shareable link Provided by the Springer Nature SharedIt content-sharing initiative Keywords Cushing’s syndrome Primary aldosteronism Adrenal vein sampling Immunohistochemistry Aldosterone-producing cell cluster Download PDF Sections Figures References Abstract Background Case presentation Discussion and conclusions Availability of data and materials Abbreviations References Acknowledgements Funding Author information Ethics declarations Additional information Rights and permissions About this article From https://bmcendocrdisord.biomedcentral.com/articles/10.1186/s12902-023-01454-8
    1 point
  22. ABSTRACT Objective To determine whether accurate inferior petrosal sinus sampling (IPSS) tumor lateralization is associated with improved clinical outcomes following the surgical treatment of Cushing’s disease. Methods The presented study was performed in accordance with PRISMA guidelines. Data regarding patient demographics, IPSS tumor lateralization, and postoperative endocrinologic outcomes were abstracted and pooled with random effects meta-analysis models. Additional meta-regression models were used to examine the association between the accuracy of IPSS tumor lateralization and postoperative outcomes (recurrence/persistence or remission/cure). Statistical analyses were performed using the Comprehensive Meta-Analysis software (significance of P<0.05). Results Seventeen eligible articles were identified, yielding data on 461 patients. Within average follow-up duration (∼59 months), the rate of correct IPSS tumor lateralization was 69% [95% Confidence Interval: 61%, 76%], and the rate of postoperative remission/cure was 78% [67%, 86%]. Preoperative IPSS tumor lateralization was concordant with MRI lateralization for 53% of patients [40%, 66%]. There was no significant association between the rate of correct IPSS tumor lateralization and postoperative remission/cure among study-level data (P=0.735). Additionally, there was no association among subgroup analyses for studies using stimulatory agents during IPSS (corticotropin-releasing hormone or desmopressin, P=0.635), nor among subgroup analyses for adult (P=0.363) and pediatric (P=0.931) patients. Conclusions Limited data suggest that the rate of correct IPSS tumor lateralization may not be positively associated with postoperative remission or cure in patients with Cushing’s disease. These findings bring into question the utility of IPSS tumor lateralization in the context of preoperative planning and surgical approach rather than confirming a pituitary source. From https://www.sciencedirect.com/science/article/abs/pii/S187887502301745X
    1 point
  23. Abstract The occurrence of a second neoplasm possibly constitutes an adverse and uncommon complication after radiotherapy. The incidence of a second pituitary tumor in patients irradiated for adrenocorticotropic hormone secreting pituitary adenoma is rare. We report a case of a 40-year-old female with Cushing disease who underwent surgical management followed by radiotherapy. After 5 years of initial treatment, an increase in tumor size was evident at the same location, with a significant interval growth of the parasellar component of the lesion. Histology revealed an undifferentiated highly malignant sarcoma. In the span of next 2 years, the patient was followed with 2 repeat decompression surgeries and radiotherapy because of significant recurrent compressive symptoms by locally invasive malignant tumor. Despite the best efforts, the patient remained unresponsive to multiple treatment strategies (eg, surgical resections and radiotherapy) and succumbed to death. radiotherapy, second malignancy, Cushing disease Issue Section: Case Report Introduction Radiation therapy is a commonly used modality for primary or adjuvant treatment of pituitary adenoma. It is also used as an adjuvant therapy for Cushing disease with persistent or aggressive tumor growth or recurrent disease after surgery. The immediate sequelae of radiotherapy for pituitary tumors include nausea, fatigue, diminished taste and olfaction, and hair loss [1]. One frequent long-term side effect is hypopituitarism. The incidence rate of new-onset hypopituitarism after conventional radiotherapy is approximately 30% to 100% after a follow-up of 10 years, whereas after stereotactic radiosurgery or fractionated radiotherapy, the incidence is approximately 10% to 40% at 5 years [2]. The occurrence of a second neoplasm after cranial radiotherapy constitutes possibly one of the most adverse complications. Tumors such as meningioma, glioma, and sarcoma are the most frequently reported secondary neoplasms after pituitary irradiation [3]. The cumulative probability of a second brain tumor in patients irradiated for pituitary adenoma and craniopharyngioma is approximately 4% [4]. We report 1 such case with detailed clinical, histopathological, and radiological characteristics because of its rarity and associated high mortality of radiation-induced sarcoma. Case Presentation The patient first presented at 40 years of age with complaints of weight gain, new-onset diabetes mellitus, hypertension, and cushingoid features in 2014. She was diagnosed with Cushing disease (24-hour urinary cortisol 1384 mcg/24 hours [3819 nmol/24 hours; reference >2 upper limit of normal], low-dose dexamethasone suppression test serum cortisol 16.6 mcg/dL [457.9 nmol/L], ACTH 85 pg/mL [18.7 pmol/L; reference range, <46 pg/mL, <10 pmol/L]) caused by invasive adrenocorticotropic hormone-secreting giant adenoma. The initial imaging revealed a homogenously enhanced pituitary macroadenoma with a size of 42 × 37 × 35 mm with suprasellar extension and encasing both the internal carotid arteries with mass effect on optic chiasma and sellar erosion. The patient underwent tumor excision by endoscopic transsphenoidal transnasal approach. Partial excision of the tumor was achieved because of cavernous sinus invasion. Histopathology and immunohistochemical stains demonstrated a corticotrophin-secreting (ACTH-staining positive) pituitary adenoma with MIB labeling index of 1% to 2%. Because biochemical remission was not achieved (urinary cortisol 794 mcg/24 hours [2191 nmol/24 hours]; ACTH 66 pg/mL [14.5 pmol/L; reference range, <46 pg/mL, <10 pmol/L]), the patient was started on ketoconazole and was received fractionated radiotherapy with a dose of 5040 cGy in 28 fractions. Diagnostic Assessment For the next 5 years, at yearly follow-up, 400 mg ketoconazole was continued in view of insufficient control of ACTH secretion. During follow-up, the size of the tumor was stable at approximately 23 × 16 × 33 mm after radiotherapy with no significant clinical and biochemical changes. Five years after surgery and radiotherapy, the patient developed cerebrospinal fluid rhinorrhea; imaging revealed a cystic transformation of the suprasellar component and increase in the size of the tumor to 39 × 22 × 26 mm, which included visualization of a parasellar component of size 29 × 19 × 15 mm. The patient continued on ketoconazole. The patient was also advised to undergo hypofractionated radiotherapy but did not return for follow-up. Treatment In 2021, 1.5 years after the last visit, the patient developed severe headache, altered sensorium, ptosis, focal seizures, and left-sided hemiparesis. During this episode, the patient had an ACTH of 66 pg/mL (14.53 pmol/L; reference range, <46 pg/mL [<10 pmol/L]) and baseline cortisol of 25 mcg/dL (689 nmol/L; reference range, 4-18 mcg/dL [110-496 nmol/L]). Repeat imaging revealed a significant decrease in the suprasellar cystic component but an increase in the size of the parasellar component to 38 × 21 × 25 mm from 29 × 19 × 15 mm, which was isointense on T1 and T2 with heterogeneous enhancement. Significant brain stem compression and perilesional edema was also visible. The patient underwent urgent frontotemporal craniotomy and decompression of the tumor. On pathological examination, the tumor tissue was composed of small pleomorphic round cells arranged in sheets and cords separated by delicate fibrocollagenous stroma. Cells had a round to oval hyperchromatic nucleus with scanty cytoplasm. Areas of hemorrhage, necrosis, and a few apoptotic bodies were seen. The tumor tissue had very high mitotic activity of >10/10 hpf and MIB labeling index of 70%. Immunohistochemistry demonstrated positivity for vimentin, CD99, and TLE-1. Dot-like positivity was present for HMB 45, synaptophysin. INI-1 loss was present in some cells. Ten percent patchy positivity was present for p53. The tumor cells, however, consistently failed to express smooth muscle actin, CD34, Myf-4, epithelial membrane antigen, desmin, LCA, SADD4, CD138, and S-100 protein. ACTH and staining for other hormones was negative. Based on the immunological and histochemical patterns, a diagnosis of high-grade poorly differentiated malignant tumor with a probability of undifferentiated sarcoma was made. Because of the invasion of surrounding structures and surgical inaccessibility, repeat fractionated radiotherapy was given with a dose of 4500 cGy over 25 fractions at 1.8 Gy daily to the planned target volume via image-guided fractionated radiotherapy. During the next 1.5 years, patient improved clinically with no significant increase in the size of tumor (Fig. 1). The patient was gradually tapered from ketoconazole and developed hypopituitarism requiring levothyroxine and glucocorticoid replacement. There was a significant improvement in the power of the left side and ptosis. Figure 1. Open in new tabDownload slide Contrast-enhanced T1 magnetic resonance imaging dynamic pituitary scan (A, sagittal; B, axial; C, coronal sections) reveals postoperative changes with residual enhancing tumor in the right lateral sella cavity with extension into the right cavernous sinus and parasellar region encasing the cavernous and inferiorly extends through the foramen ovale below the skull base up to approximately 1.5 cm. Anteriorly, it extends up to the right orbital apex and posteriorly extends along the right dorsal surface of clivus. Outcome and Follow-up After 1.5 years of reradiation in 2022, the patient again developed palsies of the abducens, trigeminal, oculomotor, and trochlear cranial nerve on the right side and left-sided hemiparesis. A significant increase in tumor size to 50 × 54 × 45 mm with anterior, parasellar, and infratentorial extension was seen (Fig. 2). Again, repeat decompression surgery was done. Two months after surgery, there was no improvement in clinical features and repeat imaging suggested an increased size of the tumor by 30%, to approximately 86 × 68 × 75 mm. Nine years after initial presentation, the patient had an episode of aspiration pneumonia and died. Figure 2. Open in new tabDownload slide Contrast-enhanced T1 magnetic resonance imaging dynamic pituitary images (A, sagittal; B, axial; C, coronal sections) after 1.5 years of a second session of radiotherapy reveal a significant interval increase in size of heterogeneously enhancing irregular soft tissue in sellar cavity with extension into the right cavernous sinus and parasellar region when compared with previous imaging. Superiorly, it extends in the suprasellar region, causing mass effect on the optic chiasma with encasement of the right prechiasmatic optic nerve and right-sided optic chiasma. Inferiorly, the lesion extends into the sphenoid sinus. Posteriorly, there is interval increase in the lesion involving the clivus and extending into the prepontine and interpeduncular cistern. Anteriorly, mass has reached up to the right orbital apex optic nerve canal, which shows mild interval increase. Discussion Radiation-induced tumors were initially described by Cahan et al in 1948. They also described the prerequisites for a tumor to be classified as a radiation-induced sarcoma [5]. The modified Cahan criteria state that (1) the presence of nonmalignancy or malignancy of a different histological type before irradiation, (2) development of sarcoma within or adjacent to the area of the radiation beam, (3) a latent period of at least 3 years between irradiation and diagnosis of secondary tumor, and (4) histological diagnosis of sarcoma, can be classified as radiation-induced sarcoma [5]. Our patient fulfilled the criteria for a radiation-induced sarcoma with a highly malignant tumor on histopathology. Radiation-induced sarcomas after functional pituitary tumors, especially Cushing disease, are rarely reported. One of the case reports revealed a high-grade osteoblastic osteosarcoma 30 years after treatment for Cushing disease with transsphenoidal resection and external beam radiotherapy [6]. In our case, there was a lag period of approximately 5 years before the appearance of a second highly undifferentiated, malignant, histologically distinct tumor. The cellular origin of this relatively undifferentiated tumor cannot be determined with certainty. However, the interlacing sarcomatous and adenomatous components resulting from distinct positive immunohistochemistry may indicate that the sarcomatous component may be derived from the preexisting pituitary adenoma. A hormonally functional pituitary tumor is not itself expected to be associated with an increased risk of secondary malignancy, except in the case of GH-secreting tumors and those with a hereditary cancer syndrome. Although not proven, immunosuppression from hypercortisolism in Cushing disease has been proposed as a contributor to secondary tumor development [7]. Other mechanisms causing increased risk of secondary malignancy can be double-stranded DNA damage and genomic instability caused by ionizing radiation and germline mutations in tumor suppressor genes such as TP53 and Rb [7]. Radiation-induced intracranial tumors were studied in a multicenter, retrospective cohort of 4292 patients with pituitary adenoma or craniopharyngioma. Radiotherapy exposure was associated with an increased risk of a second brain tumor with a rate ratio of 2.18 (95% CI, 1.31-3.62, P < .0001). The cumulative probability of a second brain tumor was 4% for the irradiated patients and 2.1% for the controls at 20 years [7]. In another study including 426 patients irradiated for pituitary adenoma between 1962 and 1994, the cumulative risk of second brain tumors was 2.0% (CI, 0.9-4.4) at 10 years and 2.4% (95% CI, 1.2-5.0) at 20 years. The relative risk of a second brain tumor compared with the incidence in the normal population is 10.5 (95% CI, 4.3-16.7) [8]. The incidence of radiation-induced sarcomas has been estimated at 0.03% to 0.3% of patients who have undergone radiation therapy. The risk of radiation-induced sarcomas increases with field size and dose. In a systemic review and analysis of 180 cases of radiation-induced intracranial sarcomas, the average dose of radiation delivered was 51.4 ± 18.6 Gy and latent period of sarcoma onset was 12.4 ± 8.6 years. A total of 49 cases were developed after radiation treatment of pituitary adenomas (27.2%). The median overall survival time for all patients with sarcoma was 11 months, with a 5-year survival rate of 14.3% [9]. Our patient received approximately 50 Gy twice through fractionated radiotherapy, resulting in larger field size and significantly higher dose than one would expect with a modern stereotactic treatment. Such a high dose of radiation is indeed a risk factor for secondary malignancy. In our patient, in a period of 2 months, there was already >30% tumor growth after recent repeat decompression surgery. The risk of secondary malignancy is thought to be much lower with stereotactic radiosurgery than conventional external beam radiation therapy, with an estimated cumulative incidence of 0.045% over 10 years (95% CI, 0.00-0.34) [10]. However, long-term follow-up data for patients receiving stereotactic radiation therapy are shorter and thus definitive conclusions cannot be made at this stage. Our case highlights a rare but devastating long-term complication of pituitary tumor irradiation after Cushing disease. The limited response to various available treatment options defines the aggressive nature of radiation-induced malignancy. Learning Points The occurrence of a second neoplasm constitutes possibly one of the most adverse and rare complication after radiotherapy. The incidence of radiation-induced sarcomas has been estimated at 0.03% to 0.3% of patients, but cases after Cushing disease are rarely reported. Patients often present with advanced disease unresponsive to various treatment modalities because of aggressive clinical course. New modalities with stereotactic radiosurgery and proton beam therapy are to be reviewed closely for risk assessment of secondary tumor. Acknowledgments The authors acknowledge Dr. Ishani Mohapatra for her support with histopathology and interpretation. Contributors All authors made individual contributions to authorship. G.B., S.K.M., and V.A.R. were involved in diagnosis and management of the patient. G.B. was involved in the writing of this manuscript and submission. V.P.S. was responsible for patient surgeries. All authors reviewed and approved the final draft. Funding The authors received no financial support for the research, authorship, and/or publication of this article. Disclosures The authors have nothing to disclose. Informed Patient Consent for Publication Signed informed consent could not be obtained from the patient or a proxy but was approved by the treating institute. Data Availability Statement Data sharing is not applicable to this article as no data sets were generated or analyzed during the current study. © The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited. From https://academic.oup.com/jcemcr/article/1/6/luad119/7343968?login=false
    1 point
  24. Abstract Introduction: Chronic exposure to excessive endogenous cortisol leads to brain changes in Cushing’s disease (CD). However, it remains unclear how CD affects large-scale functional networks (FNs) and whether these effects are reversible after treatment. This study aimed to investigate functional network changes of CD patients and their reversibility in a longitudinal cohort. Methods: Active CD patients (N = 37) were treated by transsphenoidal pituitary surgery and reexamined 3 months later. FNs were computed from resting-state fMRI data of the CD patients and matched normal controls (NCs, N = 37). A pattern classifier was built on the FNs to distinguish active CD patients from controls and applied to FNs of the CD patients at the 3-month follow-up. Two subgroups of endocrine-remitted CD patients were identified according to their classification scores, referred to as image-based phenotypically (IBP) recovered and unrecovered CD patients, respectively. The informative FNs identified by the classification model were compared between NCs, active CD patients, and endocrine-remitted patients as well as between IBP recovered and unrecovered CD patients to explore their functional network reversibility. Results: All 37 CD patients reached endocrine remission after treatment. The classification model identified three informative FNs, including cerebellar network (CerebN), fronto-parietal network (FPN), and default mode network. Among them, CerebN and FPN partially recovered toward normal at 3 months after treatment. Moreover, the informative FNs were correlated with 24-h urinary-free cortisol and emotion scales in CD patients. Conclusion: These findings suggest that CD patients have aberrant FNs that are partially reversible toward normal after treatment. Journal Section: Research Article Keywords: Cushing’s disease, Reversibility, Functional networks, Cortisol, Emotion Introduction Cushing’s disease (CD) is characterized by chronic exposure to excessive endogenous glucocorticoid most commonly caused by an adrenocorticotropic hormone (ACTH) pituitary adenoma [1, 2]. The CD is accompanied by multiple physical manifestations such as hypertension and osteoporosis, as well as various neuropsychiatric symptoms including memory lapses, attention deficits, executive function decline, emotional dysfunction, visual-spatial disability, and language defects [3‒14]. These neuropsychiatric symptoms are indicative of the effects of CD on the brain anatomy and function. Therefore, CD provides a unique and naturalistic model for investigating both the effects of hypercortisolism on the human brain and the reversibility of these effects after resolution of hypercortisolism. Recent studies have documented brain structural and metabolic abnormalities in CD patients with a variety of neuroimaging techniques, including structural magnetic resonance imaging (sMRI) [11, 12, 15‒24], diffusion tensor imaging [10, 25‒27], proton magnetic resonance spectroscopy [21, 28‒30], positron emission topography [21, 31], and arterial spin labeling [32]. These studies have shown that brain structural and metabolic abnormalities in CD patients can be partially restored after resolution of hypercortisolism [16, 18, 20‒22, 24, 32‒34], typically after transsphenoidal pituitary surgery (TSS), a safe and effective first-line treatment with a high endocrine remission rate [35, 36]. Several functional magnetic resonance imaging (fMRI) studies have also documented brain functional abnormalities in CD patients [37‒42]. Particularly, aberrant functional connectivity between the anterior cingulate cortex and the limbic network, as well as the lateral occipital cortex and the default mode network (DMN) was observed in endocrine-remitted CD patients after TSS treatment in a cross-sectional resting-state fMRI (rs-fMRI) study [40]. However, the causal effects of hypercortisolism on brain functional connectivity cannot be well investigated in CD patients only through the cross-sectional study. Additionally, the large-scale functional networks (FNs) of CD patients were not well investigated through univariate analyses in previous studies, which only examined one or few FNs in CD patients independently [37‒42]. The present study aims to jointly investigate a number of whole-brain large-scale intrinsic FNs and their reversibility due to hypercortisolism in CD patients based on rs-fMRI with a longitudinal design through multivariate analysis. Particularly, intrinsic FNs altered by CD were identified using a multivariate pattern classification model optimized by selecting intrinsic FNs informative for distinguishing CD patients from matched normal controls (NCs). The changes in these informative FNs of endocrine-remitted CD patients after TSS treatment were quantified at the 3-month follow-up with the established pattern classification model. Furthermore, changes in clinical measures, including serum cortisol, 24-h urinary-free cortisol (24hUFC), ACTH, self-rating depression scale (SDS), and self-rating anxiety scale (SAS), were detected between active and endocrine-remitted CD patients using pseudo paired t tests. Finally, the association between aberrant FNs and clinical measures was investigated in CD patients. Materials and Methods Participants In this study, 50 CD patients undergoing TSS, and 38 NCs with no history of glucocorticoid treatment were recruited at the Department of Neurosurgery, Peking Union Medical College Hospital. All these participants were assessed for depression and anxiety measured by the SDS and SAS, respectively [43]. The inclusion criteria for NCs were no past or present heart history of disease, atherosclerosis, hyperlipidemia, diabetes, neurological/psychiatric disorders, and claustrophobia. The exclusion criteria for both CD patients and NCs were past or present brain trauma, other neurological disorders, history of radiotherapy, or contraindications to MRI. Besides the inclusion and exclusion criteria, the quality of the imaging data was controlled as follows. No participant had head motion exceeding 2.0 mm translation in any of the three directions or exceeding 2.0o maximum rotation around any of the axes during rs-fMRI scanning [44]. Additionally, no participant had root-mean-square value of maximum frame-wise displacement greater than 0.3 mm [45]. After quality control of the imaging data, 37 CD patients and 37 sex-, age-, and education level-matched NCs were included in the study. The diagnosis of active CD was confirmed by experienced endocrinologists along with dynamic enhanced pituitary MRI, low- and high-dose dexamethasone suppression tests, and/or inferior petrosal sinus sampling in accordance with the latest clinical practice guidelines [46]. The 37 active CD patients were treated with TSS rather than radiotherapy. All of the 37 CD patients reached endocrine remission after treatment, which was confirmed by their normal serum cortisol (<5 µg/dL within 7 days of surgery) [46]. These patients were asked to revisit the hospital for reexamination 3 months after surgery, and all of them had no recurrence at the 3-month follow-up. Serum cortisol, 24hUFC, and ACTH were measured by direct chemiluminescence immunoassays in CD patients before surgery and at the 3-month follow-up (Siemens Healthcare Diagnostics Inc., USA). This study was approved by the Medical Ethics Committee of Peking Union Medical College Hospital, and written informed consent was obtained from all participants after explaining to them the nature of the study. Imaging Data Acquisition The MRI data were scanned by using an 8-channel phase-array head coil with a 3.0-Tesla MR scanner (Discovery MR750, General Electric) for all participants, including NCs, active CD patients, and endocrine-remitted CD patients without recurrence at the 3-month follow-up. The rs-fMRI data were acquired axially by using a gradient echo-planar imaging sequence, and the scanning parameters were 200 whole-brain volumes, 36 transverse slices with a thickness of 4 mm, in-plane resolution = 3.75 × 3.75 mm2, field of view = 240 × 240 mm2, flip angle = 90°, repetition time = 2,000 ms, and echo time = 30 ms. The extra high-resolution sagittal 3D T1-weighted data were acquired by using a brain volume sequence, and the scanning parameters were 172 slices with a thickness of 1.0 mm, in-place matrix = 512 × 512, field of view = 256 × 256 mm2, voxel size = 0.5 × 0.5 × 1.0 mm3, flip angle = 12°, repetition time = 7.2 ms, echo time = 3.2 ms, and inversion time = 400 ms. Imaging Data Preprocessing The rs-fMRI data were preprocessed as follows: (1) discarding the first four volumes of the fMRI data; (2) correction for slice timing; (3) 3D rigid-body correction for head motion to the middle frame of the data; (4) global 4D intensity scaling of the fMRI data to yield a mean value of 10,000; (5) nonlinear registration of the fMRI data to the MNI template with the deformation field obtained from its co-registered T1-weighted data using DARTEL within statistical parametric mapping (SPM12) software, with a resampled resolution of 3×3×3 mm3; (6) spatial smoothing with a 6-mm full-width at half maximum Gaussian kernel; (7) motion artifacts removal from fMRI data with ICA-AROMA; (8) regressing out averaged signals of white matter, cerebrospinal fluid, and whole brain; (9) temporal band-pass filtering (0.009–0.08 Hz). The preprocessing procedures were performed by using SPM12 software (https://www.fil.ion.ucl.ac.uk/spm/software/spm12/). Identification of Informative FNs in Active CD Patients The flowchart for identifying informative FNs in active CD patients is shown in Figure 1. First, group information-guided independent component analysis was applied to rs-fMRI data of each participant from NCs, active CD patients, and endocrine-remitted CD patients at 3 months after treatment to extract subject-specific independent components (ICs), referred to as intrinsic FNs [47] (Fig. 1a). Specifically, group-level ICs were computed based on all participants from NC, active CD, and endocrine-remitted CD groups, by using the multivariate exploratory linear optimized decomposition into independent components (MELODIC) toolbox in FSL software (https://fsl.fmrib.ox.ac.uk/fsl/fslwiki/melodic). These group-level ICs were used as guidance information to compute subject-specific ICs of all individuals [47]. The number of ICs was empirically set to be 25, and therefore each individual was characterized by 25 FNs. Particularly, these FNs were restricted to gray matter in order to minimize the partial volume effects of cerebrospinal fluid and confounding effects on the estimated components, and to improve the sensitivity to the changes of blood-oxygen-level-dependent signals. Fig. 1. VIEW LARGEDOWNLOAD SLIDE Flowchart of the multivariate pattern classification method for distinguishing active CD patients from NCs, including data preparation (a), classification modeling (b), as well as identifying CD-associated ICs (c). CD, Cushing’s disease; active CD patients, CD patients before treatment; NCs, normal controls; rs-fMRI, resting-state functional magnetic resonance imaging; ICs, independent components; GIG-ICA, group information-guided ICA; SVM, support vector machine; LOOCV, leave-one-out cross-validation. Subsequently, a multivariate pattern classification method based on support vector machine (SVM) was applied to identify cross-sectional informative FNs, which were most discriminative in distinguishing active patients from NCs [48] (Fig. 1b). Specifically, sigmoid kernel SVM classifiers were built upon a subset of 25 FNs obtained via a forward selection technique to optimize the classification performance for differentiating active patients from NCs. The similarity between subjects in SVM classification was defined as the Riemannian distance of the subset of FNs on the Grassmann manifold [48, 49]. Initially, the forward component selection procedure built a classifier on each individual FN, and the performance of the classifier was estimated using leave-one-out cross-validation (LOOCV) so that each FN could be evaluated for its classification performance. The accuracy rate was chosen as the main metric for evaluating the classification performance. The FN with the best performance was selected to be included in the subsequent classification. Through combining the first selected FN and any one of the remaining FNs, classifiers were built upon all paired FNs which were evaluated based on the training data during the current outer round using an inner LOOCV procedure. The paired FNs with the best performance were selected to be included in the classification. The procedure was repeated to add more FNs in the classification one by one until a single classifier was built upon all available FNs. Accordingly, a subset of FNs with the best performance was deemed to be the final selected components in the classification, hereafter referred to as informative FNs. To avoid potential classification biases, a nested LOOCV procedure was applied to optimize the parameters of the sigmoid kernel SVM classifiers to improve the classification performance during the forward component selection procedure [48, 49]. Since different FNs might be selected in each training runs or each testing run during the nested LOOCV procedure, the informative FNs were selected as the best performing ones with higher frequency (selection frequency>0.5). Based on these informative FNs of 74 subjects (including 37 NCs and 37 active CD patients), the LOOCV classification model yielded 74 aggregated SVM classifiers with the nested LOOCV classifiers, respectively. Each aggregated classifier generated a classification score from its corresponding nested classifiers with a positive value indicating CD state and a negative value indicating healthy state. Finally, the classification performance was evaluated with metrics including classification accuracy, specificity, sensitivity, and the area under the receiver operating characteristic curve (AUROC) (Fig. 1c). Non-parametric permutation tests were adopted to examine the statistical significance of the classification performance. The classification rate for the null distribution was estimated by building sigmoid kernel SVM models upon cross-sectional informative FNs of all active CD patients and NCs with subject labels randomly permuted by using the LOOCV strategy. This procedure was repeated for 10,000 times. Finally, the null distribution of the classification rate based on permuted samples was obtained. Longitudinal Analyses of Informative FNs and Emotion Scales from Active to Endocrine-Remitted CD Patients To investigate the longitudinal functional connectivity changes, pseudo paired t tests between active and endocrine-remitted CD patients (10,000 permutations) were applied voxel-wisely to each of the informative FNs using statistical non-parametric mapping (SnPM) software (http://warwick.ac.uk/snpm). Brain regions with statistical significance within each informative FN were identified at a voxel-wise threshold of p < 0.01 and an extent threshold of 40 adjacent voxels (AlphaSim-corrected p < 0.01). Additionally, statistical analyses were performed to compare the IC’s z scores of FNs as well as emotion scales between any pair of NC, active CD, and endocrine-remitted CD groups to further examine the longitudinal brain functional connectivity changes. Particularly, a pseudo paired t test was applied to all IC’s z scores within each informative FN as well as SDS scores and SAS scores between active and endocrine-remitted CD patients (10,000 permutations). While a pseudo two-sample t test with age, sex, and years of school education as covariates was applied to all IC’s z scores within each informative FN as well as SDS scores and SAS scores between NCs and active CD patients and endocrine-remitted CD patients. Significant differences were determined at a false discovery rate (FDR) threshold of p < 0.05 after adjusting for multiple comparisons. Statistical Analyses of Informative FNs in Endocrine-Remitted CD Patients The established pattern classification model was applied to the FNs of the follow-up endocrine-remitted CD patients. Thus, each endocrine-remitted CD patient had a classification score that reflected the likelihood of the endocrine-remitted CD patient to be active CD or healthy state (a positive value indicating active CD state and a negative value indicating healthy state). Based on the follow-up classification scores, endocrine-remitted CD patients who were correctly classified as active CD patients before treatment were further stratified into two subgroups: subjects with negative classification scores, referred to as image-based phenotypically (IBP) recovered CD patients, and those with positive classification scores, referred to as IBP unrecovered CD patients. Additionally, statistical differences in the IC’s z scores within each of the informative FNs between the IBP recovered and unrecovered CD patients, were assessed to elucidate these endocrine-remitted CD patients’ brain recoveries in these informative FNs at 3 months after treatment. Specifically, a pseudo two-sample t test with age, sex, years of school education, and years of disease duration as covariates was applied to all IC’s z scores of each FN between IBP recovered and unrecovered CD patients, and significant differences were determined at an FDR threshold of p < 0.05 (10,000 permutations) after adjusting for multiple comparisons. Correlation Analyses between Informative FNs and Clinical Measures Correlation analyses were performed to investigate the relationship between informative FNs and clinical measures in all 37 CD patients. The clinical measures of interest were serum cortisol, 24hUFC, ACTH, SDS, and SAS. Specifically, the correlation between each clinical measure and the averaged IC’s z score of each informative FN of CD patients before treatment was computed using a general linear model with age, sex, years of school education, and years of disease duration as covariates. Significant correlations were determined at a threshold of p < 0.05 using FDR corrected for multiple comparisons. Additionally, the correlation between the changes of each clinical measure and the averaged IC’s z score of each informative FN for endocrine-remitted CD patients before and after treatment was computed by using a general linear model with age, sex, years of school education, years of disease duration, and this clinical measure before treatment as covariates. The change of the averaged IC’s z score of each informative FN for each CD patient was calculated as the value after treatment minus the value before treatment divided by the value before treatment, and the change of each clinical measure for each CD patient was calculated similarly. Significant correlations were identified at a threshold of p < 0.05 using FDR corrected for multiple comparisons. Results Demographics and Clinical Characteristics The demographic and clinical data, including age, sex, years of school education, hormones, and emotion scales, are summarized in Table 1. There were no significant differences in age, sex, and years of school education between NCs and CD patients before treatment or at the 3-month follow-up (p > 0.05). The hormone levels, including ACTH, 24hUFC, and serum cortisol, were significantly restored (lower to be precise) in endocrine-remitted CD patients at the 3-month follow-up compared to their pre-treatment levels (FDR-corrected p < 0.05). These CD patients reached endocrine remission confirmed by their normal serum cortisol (<5 µg/dL) within 7 days of surgery. The emotion scales, including SDS scores and SAS scores, were significantly improved (smaller to be precise) in endocrine-remitted CD patients at 3 months after treatment compared to their rating scales in active phase (FDR-corrected p < 0.05), and the SDS scores and SAS scores for these endocrine-remitted CD patients were comparable to those of NCs. There was also significant difference in SDS scores between endocrine-remitted CD patients and NCs (FDR-corrected p < 0.05), while no significant difference was found in SAS scores between endocrine-remitted CD patients and NCs (p = 0.70). These psychometric comparison results suggest that depressive symptoms were partially recovered in endocrine-remitted CD patients, while their anxiety symptoms were also not completely recovered. Table 1. Demographic and clinical data of the participants Characteristics NCs (N = 37) Active CDs (N = 37) Endocrine-remitted CDs (N = 37) p value Age, years 38.46±11.85 33.92±8.57 33.92±8.57 0.062a Sex (M/F) 10/27 8/29 8/29 0.83a Years of school education 12.84±3.53 13.27±3.11 13.27±3.11 0.55a ACTH, pg/mL - 75.70 (45.55, 103.25) 23 (10.33, 30.70) <0.01**b 24hUFC, μg/day - 582.34 (351.30, 991.56) 47.77 (14.41, 186.54) <0.01**b Serum cortisol, μg/dL - 26.58 (20.98, 31.84) 5.49 (1.75, 13.69) <0.01**b Depression (SDS) 38.72±7.45 53.99±9.20 45.54±10.24 <0.01**c <0.01**d Anxiety (SAS) 33.34±5.46 45.27±11.92 34.46±9.78 <0.01**c 0.70d Values for characteristics are presented as mean ± SD or median (25th percentiles, 75th percentiles) unless otherwise indicated. Group differences in age, years of school education, SDS, and SAS between NCs and CD patients before or at the 3-month follow-up were examined using pseudo two-sample t tests. Group differences in sex between NCs and the CD patients before treatment or at the 3-month follow-up were examined using a χ2 test. Group differences in ACTH, 24hUFC, serum cortisol, SDS, and SAS between CD patients before treatment and at the 3-month follow-up were examined using pseudo paired t tests. NCs, normal controls; CDs, patients with Cushing’s disease; ACTH, adrenocorticotropic hormone; 24hUFC, 24-h urinary-free cortisol; SDS, self-rating depression scale; SAS, self-rating anxiety scale; M, male; F, female; SD, standard deviations. **p < 0.01. aNCs versus active or endocrine-remitted CDs. bActive CDs versus endocrine-remitted CDs. cActive CDs versus NCs or endocrine-remitted CDs. dNCs versus endocrine-remitted CDs. Informative FNs in Active CD Patients Active CD patients were mostly different from the NCs in 3 out of 25 FNs (selection frequency>0.5), including cerebellar network (CerebN), fronto-parietal network (FPN), and DMN, as shown in Figure 2a and b. The classification models built upon these three informative FNs yielded an accuracy of 72% (sensitivity: 68%, specificity: 76%, AUROC: 0.81), as shown in Figure 2c. Non-parametric permutation tests demonstrated that the classification accuracy was promising and significant (p < 1.0e−04), as suggested by the histogram of permuted classification rates shown in Figure 2d. Particularly, 25 out of 37 (67%) CD patients were correctly classified as active CD patients before treatment. Fig. 2. VIEW LARGEDOWNLOAD SLIDE Three informative functional brain networks identified by the multivariate pattern classification method and the classification performance. a Three highly selected functional brain networks, including CerebN, FPN, and DMN, for differentiating active CD patients from NCs. b The frequency of the functional brain networks selected in the nested LOOCV experiments. c The receiver operating characteristic (ROC) curve (area under the ROC curve [AUROC] = 0.81) of the classification model built upon the selected most discriminative FNs. d The histogram of the classification rates of the permutation tests and the real classification rate. In panel (a), brain regions with significant functional connectivity were obtained by applying voxel-wise one-sample t tests to the IC’s z scores for each of the FNs across all active CD patients and NCs (p < 0.05, FWE corrected for multiple comparisons, and cluster size >400 voxels). CerebN, cerebellar network; FPN, fronto-parietal network; DMN, default mode network; CD, Cushing’s disease; Pres, CD patients before treatment (i.e., active CD patients); NCs, normal controls; FNs, functional networks; ICs, independent components; FWE, family-wise error; L, left; R, right. Changes in Informative FNs from Active to Endocrine-Remitted CD Patients Two out of the three informative FNs, i.e., CerebN and FPN other than DMN, exhibited significant functional connectivity changes in CD patients between active and endocrine-remitted states (Fig. 3a). Compared with their active state, the endocrine-remitted CD patients had significantly improved (increased to be precise) functional connectivity measured by IC’s z scores in both CerebN and FPN circuits at 3 months after treatment. These results indicate that the FNs of the endocrine-remitted CD patients partially recovered toward the NCs at 3 months after treatment (Fig. 3b). Fig. 3. VIEW LARGEDOWNLOAD SLIDE Two informative functional brain networks as well as emotion scales with significant longitudinal changes in CD patients before treatment and at the 3-month follow-up. a Brain regions with significant longitudinal changes in functional connectivity within circuits of CerebN and FPN for CD patients, identified using non-parametric permutation tests (AlphaSim-corrected p < 0.01). b, c Significantly different functional connectivity measured by IC’s z scores across voxels within circuits of CerebN and FPN as well as emotion scales measured by the self-rating depression scale (SDS) and self-rating anxiety scale (SAS) between any two of NCs, CD patients before the treatment (i.e., active CD patients), and endocrine-remitted CD patients at 3-month follow-up (FDR-corrected p < 0.05). A pseudo paired t test with age, sex, and years of school education as covariates was conducted to compare all IC’s z scores within each functional network as well as the SDS scores and SAS scores between CD patients before treatment and at the 3-month follow-up. While a pseudo two-sample t test with age, sex, and years of school education as covariates was conducted to compare IC’s z scores within each functional network as well as SDS scores and SAS scores between NCs and CD patients before treatment, and endocrine-remitted CD patients at 3-month follow-up. CerebN, cerebellar network; FPN, fronto-parietal network; CD, Cushing’s disease; Pres, CD patients before treatment; Posts, endocrine-remitted CD patients at 3-month follow-up; NCs, normal controls; ICs, independent components; FDR, false discovery rate. Changes in Informative FNs of Endocrine-Remitted CD Patients Among the endocrine-remitted CD patients who were correctly classified as active CD patient before treatment, 14 participants were classified as IBP-recovered patients, while 11 participants were classified as IBP-unrecovered patients. The IBP-recovered and -unrecovered CD patients were determined by using the established pattern classification model according to the opposite signs in their classification scores based on their follow-up rs-fMRI data at 3 months after treatment (Fig. 4b). The IBP recovered patients had better recovery of the impaired functional connectivity within the circuits of CerebN and FPN than the IBP-unrecovered patients, as shown in Figure 4a. Fig. 4. VIEW LARGEDOWNLOAD SLIDE Differences in functional connectivity measured by IC’s z scores across voxels within circuits of CerebN and FPN as well as classification scores between image-based phenotypically (IBP)-recovered and -unrecovered CD patients after treatment. In panel (a), statistical comparisons were performed using pseudo two-sample t tests with age, sex, years of school education, and years of disease duration as covariates (FDR-corrected p < 0.05). In panel (b), violin plots showed opposite signs in classification scores between IBP-recovered and -unrecovered CD patients. CerebN, cerebellar network; FPN, fronto-parietal network; CD, Cushing’s disease; CDs, patients with Cushing’s disease; ICs, independent components; FDR, false discovery rate. Relationship between Informative FNs and Clinical Measures Changes of 24hUFC for endocrine-remitted CD patients before and after treatment were negatively correlated with their changes of averaged IC’s z scores within the FPN circuits with statistical significance (r = −0.37, p = 0.020), as shown in Figure 5a. The emotion scales, including SDS and SAS, were significantly negatively correlated with the averaged IC’s z scores within the CerebN circuits in the active CD patients (r = −0.31, p < 0.042), as shown in Figure 5c and d. There was no significant correlation for other clinical measures. Fig. 5. VIEW LARGEDOWNLOAD SLIDE Correlations between clinical measures and averaged IC’s z scores of informative FNs in CD patients (FDR-corrected p < 0.05). a Scatter plot for the significantly negative correlation between changes in the averaged IC’s z scores of the FPN circuits and 24hUFC of these 37 endocrine-remitted CD patients. b Multi-slice view of the FPN circuits whose changes in the averaged z scores were significantly correlated with changes in 24hUFC for all 37 endocrine-remitted CD patients before and after treatment. c, d Scatter plots for the significantly negative correlations between the averaged IC’s z scores of the CerebN circuits, and the SDS scores and SAS scores in these 37 endocrine-remitted CD patients before treatment. In panel (a), the changes in the averaged IC’s z scores of the FPN circuits were adjusted by regressing out covariates including age, sex, years of school education, years of disease duration, and the pre-treatment 24hUFC. In panels (c) and (d), the averaged IC’s z scores of the CerebN circuits were adjusted by regressing out covariates including age, sex, years of school education, and years of disease duration. 24hUFC, 24-h urinary-free cortisol; SDS, self-rating depression scale; SAS, self-rating anxiety scale; FPN, fronto-parietal network; CerebN, cerebellar network; CD, Cushing’s disease; Pres, CD patients before treatment; Posts, endocrine-remitted CD patients at 3-month follow-up; ICs, independent components; FDR, false discovery rate. Discussion The present study investigated the large-scale FNs of CD patients before and after treatment based on longitudinal rs-fMRI data. To the best of our knowledge, this is the first study to characterize longitudinal large-scale functional brain network changes due to hypercortisolism in CD patients using multivariate analysis. Particularly, the active CD patients had aberrant functional connectivity within circuits of CerebN, FPN, and DMN, respectively. More importantly, the impaired functional connectivity within the circuits of the CerebN and FPN was partially recovered in the endocrine-remitted CD patients, respectively. The changes in 24hUFC of CD patients before and after treatment were correlated with their changes in the functional connectivity of the FPN circuits. In addition, the emotion scales, including SDS and SAS, were also correlated with the functional connectivity of the CerebN circuits in CD patients before treatment. Aberrant FNs in Active CD Patients The informative FNs identified by the multivariate method were able to distinguish active CD patients from NCs with an accuracy of 72% (sensitivity: 68%, specificity: 76%, AUROC: 0.81). The non-parametric permutation tests also suggested that the multivariate method performed well in differentiating active CD patients from NCs. The most frequently selected FNs (Fig. 2b), i.e., informative FNs, were CerebN, FPN, and DMN. The cross-sectional multivariate analyses have revealed that the active CD patients were mostly different from the NCs in the functional connectivity within 3 FNs out of 25 FNs, as shown in Figure 2a. The aforementioned cross-sectional results provided new insights into large-scale functional brain network abnormalities due to hypercortisolism in CD patients. Particularly, our study revealed that active CD patients had significantly disrupted functional connectivity within the cerebellum (Fig. 2a, 3b), and their emotional dysfunctions observed by the SDS and SAS were associated with the impaired functional connectivity within the cerebellum (Fig. 5c, d). Therefore, it was reasonable to speculate that the cognitive or emotional dysfunctions for active CD patients, documented in this study as well as numerous previous studies [3‒5, 7‒9, 11‒14, 50], might be closely related to the observed functional connectivity abnormalities in the cerebellum. Additionally, our study found that the functional connectivity within the FPN circuits was significantly reduced in active CD patients (Fig. 2a, 3b). It was postulated that cognitive impairments in active CD patients, reported in several early studies [6, 14, 51], might be associated with the observed functional connectivity abnormalities in the FPN circuits. While several recent studies reported that active CD patients had structural or metabolic abnormalities in two brain regions of the FPN, namely, the middle frontal gyrus and inferior parietal lobule [17, 21, 32] These local morphological or metabolic abnormalities might exacerbate the observed functional network (FPN) alterations in active CD patients. Moreover, our study found that the functional connectivity within the DMN circuits was vulnerable to the detrimental effects of hypercortisolism in active CD patients. Besides our finding, recent studies reported that active CD patients showed structural, metabolic, or spontaneous activity abnormalities in several brain regions of DMN, including the posterior cingulate cortex, precuneus, parahippocampal gyrus, ventral medial prefrontal cortex, superior frontal gyrus, inferior temporal gyrus, and lateral parietal cortex [21, 27, 30‒32, 38, 52] These local morphological, metabolic or activity abnormalities might exacerbate the newly discovered DMN impairments in active CD patients. Essentially, the functional, morphological, and metabolic abnormalities in regions of the DMN might be directly related to the adverse expressions of glucocorticoid receptor genes within these brain regions caused by excessive exposure to endogenous cortisol [53]. Reversible Impaired FNs in Endocrine-Remitted CD Patients after Treatment The longitudinal statistical analysis has revealed that the endocrine-remitted CD patients’ hormones, including ACTH, 24hUFC, and serum cortisol, maintained near-normal levels at 3 months after treatment, suggesting that these patients did not relapse according to the endocrine hormone levels (Table 1). Meanwhile, their functional connectivity within circuits of the FPN and CerebN was partially restored at the 3-month follow-up after resolution of hypercortisolism (Fig. 3b). Particularly, our combined longitudinal and cross-sectional study found that the functional connectivity within the FPN circuits in endocrine-remitted CD patients was partially restored after treatment. While a cross-sectional sMRI study reported that endocrine-remitted CD patients still had structural abnormalities in the FPN-related region, namely, the middle frontal gyrus [17]. Our study also found that the functional connectivity of the cerebellum in endocrine-remitted CD patients was partially restored after treatment (Fig. 3b). Besides this finding, two other cross-sectional sMRI studies reported that the structural abnormalities of the cerebellum in endocrine-remitted patients were present as well [16, 20]. Taken together, it was postulated that the reversibility of the observed functional connectivity impairments within circuits of the FPN and CerebN might be directly influenced by their local morphological abnormalities in endocrine-remitted CD patients. Moreover, our study uncovered that the IBP-recovered patients exhibited better recovery of the functional connectivity within circuits of the FPN and CerebN than the IBP-unrecovered ones, as shown in Figure 4a. This result demonstrated that different endocrine-remitted CD patients had different recovery levels for the impaired functional connectivity within circuits of these brain FNs. More importantly, our study further found that the recovered 24hUFC was associated with the improved functional connectivity within FPN circuits in endocrine-remitted CD patients at the 3-month follow-up after treatment (Fig. 5a). This finding indicated that chronic endogenous hypercortisolism in CD patients might be directly related to their FPN impairments. Strengths of This Study The combined longitudinal and cross-sectional analyses have confirmed that the brain functional network abnormalities in CD patients were partially reversible at 3 months after resolution of the hypercortisolism. Since the brain structural abnormalities in endocrine-remitted CD patients were not completely recovered [16], it merits further investigation how the brain structural and functional network recoveries couple with each other in a longitudinal design. The present study provided complementary information to existing neuroimaging studies of CD patients. The existing neuroimaging studies have reported that CD patients had brain volume loss in cortical and cerebellar regions, hippocampus, and amygdala, as well as enlarged ventricles. These structural abnormalities were partially recovered for endocrine-remitted CD patients after treatment [11, 15, 16, 18‒22, 24] or after resolution of the hypercortisolism [12, 18, 24]. CD patients also had reduced cortical thickness in many brain regions including superior frontal cortex, caudal middle frontal cortex, precentral gyrus, insula, precuneus, cuneus, caudal/rostral anterior cingulate gyrus, and posterior cingulate gyrus [17, 54]. In addition, disrupted white matter integrity was observed in CD patients throughout the brain including frontal lobe, temporal lobe, hippocampus, parahippocampal gyrus, cingulate cingulum, corpus callosum, uncinate fasciculus, and cerebellum [10, 25‒27]. Furthermore, metabolic abnormalities in CD patients have been reported in widely distributed brain regions [21, 28‒32], which could be almost completely restored after resolution of hypercortisolism. Besides aforementioned structural and metabolic abnormalities, functional abnormalities have also been reported in CD patients using fMRI [37‒42]. Particularly, abnormal functional activations in CD patients have been observed in the prefrontal cortex, superior/middle/inferior frontal gyrus, superior parietal lobule, superior/middle temporal gyrus, inferior occipital gyrus, rostral/dorsal anterior cingulate gyrus, anterior/middle/posterior hippocampus, amygdala, precuneus, cuneus, lingual gyrus, caudate body, pulvinar/lateral posterior nuclei of the thalamus, and substantia nigra using task fMRI [37‒39, 41]. Abnormal spontaneous functional activities measured by both the amplitude of low-frequency fluctuation and regional homogeneity for CD patients have been observed in the prefrontal cortex, occipital lobe, postcentral gyrus, posterior cingulate gyrus, precuneus, thalamus, and cerebellum [20]. The dysregulation of functional connectivity density of CD patients has been found primarily in the prefrontal cortex, lateral parietal cortex, anterior/posterior cingulate gyrus, and parahippocampal gyrus [55]. The abnormal functional connectivity for CD patients has also been observed between the prefrontal cortex and medial temporal lobe, ventromedial prefrontal cortex and posterior cingulate cortex, anterior cingulate gyrus and limbic network, and lateral occipital cortex and DMN using task fMRI or rs-fMRI [39, 40]. Limitations and Future Work This study has several limitations. First, the longitudinal sample size is not large enough due to the rarity of CD, which might lead to relatively low statistical power and potential biases. Second, our study mainly investigated the brain functional network reversibility of the CD. Studies of the CD’s structural reversibility may provide complementary information to the current study. Third, our study investigated the short-term (3 months) effects of hypercortisolism on large-scale functional brain networks in CD patients. Nevertheless, the long-term effects of hypercortisolism on large-scale functional brain networks remain unclear and merit further investigation. In future work, long-term follow-up data of the CD patients recruited in the current study will be collected to investigate the long-term dynamic changes of their impaired large-scale functional brain networks. Conclusion This is the first study to investigate large-scale functional brain networks and their reversibility in a longitudinal CD cohort by using multivariate analysis. The large-scale functional brain networks, including the CerebN, FPN, and DMN, were impaired due to elevated cortisol levels in active CD patients. More importantly, the impaired functional brain networks of these CD patients were partially restored when their hormone levels returned to normal at 3 months after treatment. The changes of the functional connectivity within the impaired FPN were correlated with changes of the 24hUFC in endocrine-remitted CD patients, while the functional connectivity within the impaired CerebN was closely associated with emotion dysfunctions in active CD patients. These findings suggest that pattern recognition techniques could help identify informative functional brain networks in CD patients, which may help open up novel avenues for their postoperative interventions and assessments after endocrine remission. Statement of Ethics This study confirmed to the Declaration of Helsinki and was approved by the Medical Ethics Committee of Peking Union Medical College Hospital (approval number S-424). Written informed consent was obtained from all participants. Conflict of Interest Statement All authors reported no financial interests or potential conflicts of interest. Funding Sources This study was supported in part by the China Postdoctoral Science Foundation (2020T130070, 2019M650567), and the Clinical Application Research of Capital Characteristic Fund from the Beijing Municipal Science and Technology Commission (Z151100004015099). Author Contributions Bing Xing, Feng Feng, and Yong Fan were involved in study conception and design. Bo Hou, Xiaopeng Guo, Yong Yao, and Ming Feng collected clinical and imaging data. Hewei Cheng, Lu Gao, and Rixing Jing performed data preparation and statistical analysis. Hewei Cheng, Lu Gao, Rixing Jing, Bing Xing, Feng Feng, and Yong Fan were involved in data interpretation. Hewei Cheng, Lu Gao, and Rixing Jing wrote the first draft of the manuscript. Hewei Cheng, Lu Gao, Rixing Jing, Bo Hou, Xiaopeng Guo, Zihao Wang, Ming Feng, Bing Xing, Feng Feng, and Yong Fan provided critical editing and revision of the manuscript for important intellectual content. All authors approved the final version of the manuscript. Additional Information Hewei Cheng and Lu Gao contributed equally to this work. Data Availability Statement All data generated or analyzed during this study are included in this article. Further inquiries can be directed to the corresponding author. References 1. Newell-Price J, Bertagna X, Grossman AB, Nieman LK. Cushing’s syndrome. Lancet. 2006;367(9522):1605–17. 2. Pivonello R, De Martino MC, De Leo M, Simeoli C, Colao A. Cushing’s disease: the burden of illness. Endocrine. 2017;56(1):10–8. 3. Whelan TB, Schteingart DE, Starkman MN, Smith A. Neuropsychological deficits in Cushing’s syndrome. J Nerv Ment Dis. 1980;168(12):753–7. 4. Forget H, Lacroix A, Somma M, Cohen H. Cognitive decline in patients with Cushing’s syndrome. J Int Neuropsychol Soc. 2000;6(1):20–9. 5. Starkman MN, Giordani B, Berent S, Schork MA, Schteingart DE. Elevated cortisol levels in Cushing’s disease are associated with cognitive decrements. Psychosom Med. 2001;63(6):985–93. 6. Tiemensma J, Kokshoorn NE, Biermasz NR, Keijser B-JSA, Wassenaar MJE, Middelkoop HAM, et al. Subtle cognitive impairments in patients with long-term cure of Cushing’s disease. J Clin Endocrinol Metab. 2010;95(6):2699–714. 7. Pivonello R, Simeoli C, De Martino MC, Cozzolino A, De Leo M, Iacuaniello D, et al. Neuropsychiatric disorders in Cushing’s syndrome. Front Neurosci. 2015;9:129. 8. Pires P, Santos A, Vives-Gilabert Y, Webb SM, Sainz-Ruiz A, Resmini E, et al. White matter involvement on DTI-MRI in Cushing’s syndrome relates to mood disturbances and processing speed: a case-control study. Pituitary. 2017;20(3):340–8. 9. Santos A, Resmini E, Pascual JC, Crespo I, Webb SM. Psychiatric symptoms in patients with Cushing’s syndrome: prevalence, diagnosis and management. Drugs. 2017;77(8):829–42. 10. Valassi E, Crespo I, Keevil BG, Aulinas A, Urgell E, Santos A, et al. Affective alterations in patients with Cushing’s syndrome in remission are associated with decreased BDNF and cortisone levels. Eur J Endocrinol. 2017;176(2):221–31. 11. Frimodt-Møller KE, Møllegaard Jepsen JR, Feldt-Rasmussen U, Krogh J. Hippocampal volume, cognitive functions, depression, anxiety, and quality of life in patients with Cushing syndrome. J Clin Endocrinol Metab. 2019;104(10):4563–77. 12. Kumar N, Jarial KDS, Bhansali A, Nehra R, Vyas S, Walia R. Neurocognitive functions and brain volume in patients with endogenous cushing’s syndrome before and after curative surgery. Indian J Endocrinol Metab. 2020;24(5):396–401. 13. Lin TY, Hanna J, Ishak WW. Psychiatric symptoms in Cushing’s syndrome: a systematic review. Innov Clin Neurosci. 2020;17(1–3):30–5. 14. Na S, Fernandes MA, Ioachimescu AG, Penna S. Neuropsychological and emotional functioning in patients with Cushing’s syndrome. Behav Neurol. 2020;2020:1–10. 15. Starkman MN, Giordani B, Gebarski SS, Berent S, Schork MA, Schteingart DE. Decrease in cortisol reverses human hippocampal atrophy following treatment of Cushing’s disease. Biol Psychiatry. 1999;46(12):1595–602. 16. Andela CD, van der Werff SJ, Pannekoek JN, van den Berg SM, Meijer OC, van Buchem MA, et al. Smaller grey matter volumes in the anterior cingulate cortex and greater cerebellar volumes in patients with long-term remission of Cushing’s disease: a case–control study. Eur J Endocrinol. 2013;169(6):811–9. 17. Crespo I, Esther G-M, Santos A, Valassi E, Yolanda V-G, De Juan-Delago M, et al. Impaired decision-making and selective cortical frontal thinning in Cushing’s syndrome. Clin Endocrinol. 2014;81(6):826–33. 18. Andela CD, van Haalen FM, Ragnarsson O, Papakokkinou E, Johannsson G, Santos A, et al. Mechanisms in endocrinology: cushing’s syndrome causes irreversible effects on the human brain: a systematic review of structural and functional magnetic resonance imaging studies. Eur J Endocrinol. 2015;173(1):R1–4. 19. Burkhardt T, Lüdecke D, Spies L, Wittmann L, Westphal M, Flitsch J. Hippocampal and cerebellar atrophy in patients with Cushing’s disease. Neurosurg Focus. 2015;39(5):E5–7. 20. Jiang H, Ren J, He NY, Liu C, Sun YH, Jian FF, et al. Volumetric magnetic resonance imaging analysis in patients with short-term remission of Cushing’s disease. Clin Endocrinol. 2017;87(4):367–74. 21. Bauduin SEEC, van der Wee NJA, van der Werff SJA. Structural brain abnormalities in Cushing’s syndrome. Curr Opin Endocrinol Diabetes Obes. 2018;25(4):285–9. 22. Jiang H, Liu C, Pan S-J, Ren J, He N-Y, Sun Y-H, et al. Reversible and the irreversible structural alterations on brain after resolution of hypercortisolism in Cushing’s disease. Steroids. 2019;151:108457. 23. Bauduin SEEC, Pal Z, Pereira AM, Meijer OC, Giltay EJ, Wee NJA, et al. Cortical thickness abnormalities in long-term remitted Cushing’s disease. Transl Psychiatry. 2020;10(1):239. 24. Hou B, Gao L, Shi L, Luo Y, Guo X, Young GS, et al. Reversibility of impaired brain structures after transsphenoidal surgery in Cushing’s disease: a longitudinal study based on an artificial intelligence-assisted tool. J Neurosurg. 2020;134(2):1–10. 25. van der Werff SJ, Andela CD, Nienke Pannekoek J, Meijer OC, van Buchem MA, Rombouts SA, et al. Widespread reductions of white matter integrity in patients with long-term remission of Cushing’s disease. Neuroimage Clin. 2014;4:659–67. 26. Pires P, Santos A, Vives-Gilabert Y, Webb S, Sainz-Ruiz A, Resmini E, et al. White matter alterations in the brains of patients with active, remitted, and cured Cushing syndrome: a DTI study. Am J Neuroradiol. 2015;36(6):1043–8. 27. Jiang H, He N-Y, Sun Y-H, Jian F-F, Bian L-G, Shen J-K, et al. Altered gray and white matter microstructure in Cushing’s disease: a diffusional kurtosis imaging study. Brain Res. 2017;1665:80–7. 28. Khiat A, Bard C, Lacroix A, Boulanger Y. Recovery of the brain choline level in treated Cushing’s patients as monitored by proton magnetic resonance spectroscopy. Brain Res. 2000;862(1–2):301–7. 29. Resmini E, Santos A, Gómez-Anson B, López-Mourelo O, Pires P, Vives-Gilabert Y, et al. Hippocampal dysfunction in cured Cushing’s syndrome patients, detected by 1H‐MR‐spectroscopy. Clin Endocrinol. 2013;79(5):700–7. 30. Crespo I, Santos A, Gómez-Ansón B, López-Mourelo O, Pires P, Vives-Gilabert Y, et al. Brain metabolite abnormalities in ventromedial prefrontal cortex are related to duration of hypercortisolism and anxiety in patients with Cushing’s syndrome. Endocrine. 2016;53(3):848–56. 31. Liu S, Wang Y, Xu K, Ping F, Li F, Wang R, et al. Voxel-based comparison of brain glucose metabolism between patients with Cushing’s disease and healthy subjects. Neuroimage Clin. 2018;17:354–8. 32. Cheng H, Gao L, Hou B, Feng F, Guo X, Wang Z, et al. Reversibility of cerebral blood flow in patients with Cushing’s disease after surgery treatment. Metabolism. 2020;104:154050. 33. Gao L, Liu L, Shi L, Luo Y, Wang Z, Guo X, et al. Dynamic changes of views on the brain changes of Cushing’s syndrome using different computer-assisted tool. Rev Endocr Metab Disord. 2020;21(1):185–200. 34. Piasecka M, Papakokkinou E, Valassi E, Santos A, Webb SM, de Vries F, et al. Psychiatric and neurocognitive consequences of endogenous hypercortisolism. J Intern Med. 2020;288(2):168–82. 35. Kelly DF. Transsphenoidal surgery for Cushing’s disease: a review of success rates, remission predictors, management of failed surgery, and Nelson’s Syndrome. Neurosurg Focus. 2007;23(3):E5. 36. Theodoropoulou M, Reincke M. Tumor-directed therapeutic targets in Cushing disease. J Clin Endocrinol Metab. 2019;104(3):925–33. 37. Maheu FS, Mazzone L, Merke DP, Keil MF, Stratakis CA, Pine DS, et al. Altered amygdala and hippocampus function in adolescents with hypercortisolemia: a functional magnetic resonance imaging study of Cushing syndrome. Dev Psychopathol. 2008;20(4):1177–89. 38. Langenecker SA, Weisenbach SL, Giordani B, Briceño EM, Guidotti Breting LM, Schallmo MP, et al. Impact of chronic hypercortisolemia on affective processing. Neuropharmacology. 2012;62(1):217–25. 39. Bas-Hoogendam JM, Andela CD, van der Werff SJA, Pannekoek JN, van Steenbergen H, Meijer OC, et al. Altered neural processing of emotional faces in remitted Cushing's disease. Psychoneuroendocrinology. 2015;59:134–46. 40. Van Der Werff SJ, Pannekoek JN, Andela CD, Meijer OC, Van Buchem MA, Rombouts SA, et al. Resting-state functional connectivity in patients with long-term remission of Cushing’s disease. Neuropsychopharmacology. 2015;40(8):1888–98. 41. Ragnarsson O, Stomby A, Dahlqvist P, Evang JA, Ryberg M, Olsson T, et al. Decreased prefrontal functional brain response during memory testing in women with Cushing’s syndrome in remission. Psychoneuroendocrinology. 2017;82:117–25. 42. Stomby A, Salami A, Dahlqvist P, Evang JA, Ryberg M, Bollerslev J, et al. Elevated resting-state connectivity in the medial temporal lobe and the prefrontal cortex among patients with Cushing’s syndrome in remission. Eur J Endocrinol. 2019;180(5):329–38. 43. Zung WW. A rating instrument for anxiety disorders. Psychosomatics. 1971;12(6):371–9. 44. Kim J-H, Lee J-M, Jo HJ, Kim SH, Lee JH, Kim ST, et al. Defining functional SMA and pre-SMA subregions in human MFC using resting state fMRI: functional connectivity-based parcellation method. NeuroImage. 2010;49(3):2375–86. 45. Power JD, Barnes KA, Snyder AZ, Schlaggar BL, Petersen SE. Spurious but systematic correlations in functional connectivity MRI networks arise from subject motion. NeuroImage. 2012;59(3):2142–54. 46. 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. 47. Du Y, Fan Y. Group information guided ICA for fMRI data analysis. NeuroImage. 2013;69:157–97. 48. Fan Y, Liu Y, Wu H, Hao Y, Liu H, Liu Z, et al. Discriminant analysis of functional connectivity patterns on Grassmann manifold. NeuroImage. 2011;56(4):2058–67. 49. Li P, Jing RX, Zhao RJ, Ding ZB, Shi L, Sun HQ, et al. Electroconvulsive therapy-induced brain functional connectivity predicts therapeutic efficacy in patients with schizophrenia: a multivariate pattern recognition study. NPJ Schizophr. 2017;3(1):21–9. 50. Forget H, Lacroix A, Bourdeau I, Cohen H. Long-term cognitive effects of glucocorticoid excess in Cushing’s syndrome. Psychoneuroendocrinology. 2016;65:26–33. 51. Mauri M, Sinforiani E, Bono G, Vignati F, Berselli M, Attanasio R, et al. Memory impairment in Cushing’s disease. Acta Neurol Scand. 1993;87(1):52–5. 52. Jiang H, He NY, Sun YH, Jian FF, Bian LG, Shen JK, et al. Altered spontaneous brain activity in Cushing’s disease: a resting-state functional MRI study. Clin Endocrinol. 2017;86(3):367–76. 53. Amaya JM, Viho EM, Sips HC, Lalai RA, Sahut-Barnola I, Dumontet T, et al. Gene expression changes in the brain of a Cushing’s syndrome mouse model. J Neuroendocrinol. 2022;34(4):e13125. 54. Bauduin SEEC, van der Pal Z, Pereira AM, Meijer OC, Giltay EJ, van der Wee NJA, et al. Cortical thickness abnormalities in long-term remitted Cushing’s disease. Transl Psychiatry. 2020;10(1):293. 55. Wang X, Zhou T, Wang P, Zhang L, Feng S, Meng X, et al. Dysregulation of resting-state functional connectivity in patients with Cushing’s disease. Neuroradiology. 2019;61(8):911–20. © 2023 The Author(s). Published by S. Karger AG, Basel Open Access License / Drug Dosage / Disclaimer This article is licensed under the Creative Commons Attribution-NonCommercial 4.0 International License (CC BY-NC). Usage and distribution for commercial purposes requires written permission. Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug. Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements. This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License . From https://karger.com/nen/article/doi/10.1159/000534789/869375/Reversibility-of-Impaired-Large-Scale-Functional
    1 point
  25. The following is a summary of “Diurnal Range and Intra-patient Variability of ACTH Is Restored With Remission in Cushing’s Disease,” published in the November 2023 issue of Endocrinology by Alvarez, et al. Distinguishing Cushing’s disease (CD) remission from other conditions using single adrenocorticotropic hormone (ACTH) measurements poses challenges. For a study, researchers sought to analyze changes in ACTH levels before and after transsphenoidal surgery (TSS) to identify trends confirming remission and establish ACTH cutoffs for targeted clinical trials. A retrospective analysis involved 253 CD patients undergoing TSS at a referral center from 2005 to 2019. Remission outcomes were assessed based on postoperative ACTH levels. Among 253 patients, 223 achieved remission post-TSS. The remission group exhibited higher ACTH variability at morning (AM) (P = .02) and evening (PM) (P < .001) time points compared to the nonremission group. Nonremission cases had a significantly narrower diurnal ACTH range (P < .0001). A ≥50% decrease in plasma ACTH from mean preoperative levels, especially in PM values, predicted remission. Absolute plasma ACTH concentration and the ratio of preoperative to postoperative values were associated with nonremission (adj P < .001 and .001, respectively). ACTH variability suppression was observed in CD, with remission linked to restored variability. A ≥50% decrease in plasma ACTH may predict CD remission post-TSS. The insights can guide clinicians in developing rational outcome measures for interventions targeting CD adenomas. Source: academic.oup.com/jcem/article-abstract/108/11/2812/7187942?redirectedFrom=fulltext
    1 point
  26. Dr. Theodore Friedman (the Wiz) will be giving a webinar on Optimal replacement for Hypopituitarism and Sheehan’s: Oxytocin, testosterone, growth hormone, stimulants and beyond Learn what most Endocrinologists don’t know about but will improve your quality of life Topics to be discussed include: • Oxytocin-the love hormone • Testosterone, not just for men • Stimulants to treat pituitary apathy • Growth hormone, not just for kids • Thyroid optimization • Cortisol, the right and wrong way to give • Learn about the common medicine you should never take if on growth hormone Wednesday • December 6th• 6 PM PST Via Zoom Click here to join the meeting or https://us02web.zoom.us/j/4209687343?pwd=amw4UzJLRDhBRXk1cS9ITU02V1pEQT09&omn=84521530646 OR +16699006833,,4209687343#,,,,*111116# Slides will be available before the webinar and recording after the meeting at slides or on Dr. Friedman’s YouTube channel OR Join on Facebook Live https://www.facebook.com/goodhormonehealth at 6 PM PST Meeting ID: 420 968 7343 Passcode: 111116 Your phone/computer will be muted on entry. There will be plenty of time for questions using the chat button. For more information, email us at mail@goodhormonehealth.com
    1 point
  27. Background: Cushing’s disease (CD) poses significant challenges in its treatment due to the lack of reliable biomarkers for predicting tumor localization or postoperative clinical outcomes. Sphingosine-1-phosphate (S1P) has been shown to increase cortisol biosynthesis and is regulated by adrenocorticotropic hormone (ACTH). Methods: We employed bilateral inferior petrosal sinus sampling (BIPSS), which is considered the gold standard for diagnosing pituitary sources of CD, to obtain blood samples and explore the clinical predictive value of the S1P concentration ratio in determining tumor laterality and postoperative remission. We evaluated 50 samples from 25 patients who underwent BIPSS to measure S1P levels in the inferior petrosal sinuses bilaterally. Results: Serum S1P levels in patients with CD were significantly higher on the adenoma side of the inferior petrosal sinus than on the nonadenoma side (397.7 ± 15.4 vs. 261.9 ± 14.88; P < 0.05). The accuracy of diagnosing tumor laterality with the interpetrosal S1P and ACTH ratios and the combination of the two was 64%, 56% and 73%, respectively. The receiver operating characteristic curve analysis revealed that the combination of interpetrosal S1P and ACTH ratios, as a predictor of tumor laterality, exhibited a sensitivity of 81.82% and a specificity of 75%, with an area under the curve value of 84.09%. Moreover, we observed that a high interpetrosal S1P ratio was associated with nonremission after surgery. Correlation analyses demonstrated that the interpetrosal S1P ratio was associated with preoperative follicle-stimulating hormone (FSH), luteinizing hormone (LH), and postoperative ACTH 8 am levels (P < 0.05). Conclusion: Our study demonstrated a significant association between the interpetrosal S1P ratio and tumor laterality, as well as postoperative remission in CD, suggesting that the interpetrosal S1P ratio could serve as a valuable biomarker in clinical practice. 1 Introduction Cushing’s disease (CD), also known as adrenocorticotropic hormone (ACTH)-secreting pituitary adenoma, arises from the pituitary corticotroph cells and induces endogenous hypercortisolism by stimulating the adrenal glands to produce excessive amount of cortisol (1). Patients with CD typically exhibit symptoms of hypercortisolism, such as hypertension, diabetes, purplish skin striae, mental disturbances, hyposexuality, hirsutism, menstrual disorders, acne, fatigue, obesity, and osteoporosis (1). The overall mortality of patients with CD is twice that of the general population, and if left untreated, hypercortisolism resulting from CD increases this rate to approximately four times the expected value (2–4). Transsphenoidal surgery continues to be the primary treatment for CD (5). However, previous studies reported variable remission rates, ranging from 45% to 95% (6–8). Long-term follow-up data have revealed recurrence in 3–66% of patients who had initially achieved complete remission (9, 10). The rate of surgical remission in CD can be influenced by various factors, including the size and location of the tumor, expertise of the neurosurgeon, and criteria used for assessing remission (11). Preoperative clinical variables, such as age, gender, disease duration, and severity of clinical signs and symptoms, cannot reliably identify patients at a higher risk of nonremission (12, 13). Therefore, predicting postsurgical remission in CD remains a challenging goal. Accumulating evidence has shown that sphingosine-1-phosphate (S1P), an intracellular pleiotropic bioactive sphingolipid metabolite synthesized by sphingosine kinase 1 (SPHK1), plays a pivotal role in diverse endocrine disorders (14–16). Overexpression of SPHK1 promotes the progression of multiple neuroendocrine tumors (17, 18). ACTH can rapidly activate sphingolipid metabolism, causing an increase in S1P secretion in the adrenal cortex (19). Furthermore, the activation of S1P signaling in H295R cells, a human adrenocortical tumor cell line, has been suggested to induce increased transcription of hormone-sensitive lipase and steroidogenic acute regulatory protein, ultimately elevating cortisol production (20). Recently, surgical removal of ACTH-secreting adenoma has been reported to cause a decline in sphingomyelin levels (21). However, whether they have a similar role in the pituitary gland remains to be investigated. Bilateral inferior petrosal sinus sampling (BIPSS) is a highly effective procedure for diagnosing pituitary sources of ACTH in CD (22, 23). Contemporaneous differences in ACTH concentration during venous sampling between the two sides of the adenoma can predict the location of the adenoma within the pituitary (on the side of the gland with a microadenoma) and may guide surgical treatment in cases with inconclusive magnetic resonance imaging findings. Previous studies demonstrated that an ACTH gradient of ≥1.4 between the inferior petrosal sinuses can indicate microadenoma lateralization in patients with CD (24–26). However, the correct lateralization only occurs in 57–68% of all cases (27–29). Therefore, we analyzed the clinical behavior of a well-characterized cohort of patients with CD who underwent BIPSS before surgery. We measured the difference in the concentration of S1P in bilateral petrosal sinus blood samples and explored the clinical predictive value of the S1P concentration ratio in determining tumor laterality and postoperative remission. 2 Materials and methods 2.1 Patients and study design This study was conducted at a tertiary center, involving a cohort of 25 patients diagnosed with CD who had undergone BIPSS and surgery, with a minimum follow-up duration of 2 years. Comprehensive chart reviews were conducted to collect data on demographics, clinical characteristics, pituitary imaging findings, tumor pathology, and biochemical tests. The criteria used for diagnosing CD encompassed the presence of characteristic signs and symptoms of hypercortisolism, along with biochemical evaluation of two urinary free cortisol measurements exceeding the normal range for the respective assay, serum cortisol level >1.8 μg/dL (50 nmol/L) after an overnight 1-mg dexamethasone suppression test, and two late-night salivary cortisol measurements exceeding the normal range for the respective assay (30). A diagnosis of Cushing’s syndrome was established if the patient had positive test results for at least two of the three aforementioned tests. Adrenal insufficiency was diagnosed if patients exhibited symptoms or signs of adrenal insufficiency or if serum cortisol levels were ≤3 μg/dL, even in the absence of clinical signs or symptoms. Remission was defined as normalization of the levels of 24-h urinary free cortisol, late-night salivary cortisol, and overnight 1-mg dexamethasone suppression test in patients without concurrent central adrenal insufficiency after surgery (31). 2.2 Patients and tissue/serum samples Surgical specimens of CD-affected tissues were collected from Xiangya Hospital, Central South University. Three normal pituitary tissues were obtained from cadaveric organ donors without any history of endocrine disease (Central South University). A total of 25 CD tissue samples were obtained for immunohistochemistry analysis. This study was conducted in compliance with the Helsinki Declaration and was ethically approved by the Xiangya Hospital Ethics Committee, Xiangya Hospital (Changsha, China). Tumor samples and corresponding clinical materials were obtained with written consent from all patients. 2.3 BIPSS After obtaining informed consent, BIPSS was performed using standard techniques described in previous studies (32, 33). Briefly, the patient’s head was immobilized to ensure midline positioning and prevent any potential bias towards asymmetric pituitary drainage by the petrosal sinuses. After placing peripheral catheters and cannulating both inferior petrosal sinuses, blood samples were collected at baseline and at 3, 5, 10, and 15 min following intravenous administration of DDAVP, which stimulates pituitary production of ACTH. Additional samples for experimental purposes were collected immediately following the 15-min sample collection to avoid interference with the patient’s diagnostic study. 2.4 Measurement of baseline plasma S1P concentration Blood samples were obtained from both petrosal sinuses and were centrifuged to remove cellular components. Samples that exhibited hemolysis or coagulation were excluded from the study. Plasma samples were stored at −80°C. The S1P levels in plasma were analyzed using a S1P competitive ELISA kit (Echelon Biosciences, Salt Lake City, UT) according to the manufacturer’s instructions (34). 2.5 Immunofluorescence staining The pituitary tissues were post-fixed and dehydrated with alcohol as follows: 70% for 24 h, 80% for 3 h, 90% for 4 h, 95% for 3 h, and finally in absolute alcohol for 2 h. Tissue slices with a 5-μm thickness were cut using a microtome (Thermo Fisher Scientific), blocked with 3% BSA, and then treated with primary antibodies to SPHK1 (CST, #3297) and ACTH (Proteintech, CL488-66358). Subsequently, the tissue slides were incubated with Alexa Fluor 488-conjugated anti-rabbit (Invitrogen, A21206, 1:200) or Alexa Fluor 555-conjugated anti-rabbit (Invitrogen, A21428, 1:200) secondary antibodies. Specimens were visualized and imaged using a fluorescence microscope. 2.6 Statistical analysis The Mann–Whitney U test was used to assess the clinical–molecular associations in adenoma samples, whereas the chi-square test was used to compare categorical data. The Kruskal–Wallis analysis and ANOVA were conducted for multiple comparisons. Statistical analyses were conducted using SPSS v20 and GraphPad Prism version 7. All results were presented in graphs and tables as median ± interquartile range. The distribution of each parameter was presented as the minimum–maximum range. Parametric or nonparametric statistical tests were applied, as appropriate, after testing for normality. The receiver operating characteristic curve was used to determine the cut-off value for predicting tumor laterality. Pearson correlation analyses was used to examine the correlations between variables. Proportions were expressed as percentages, and significance was defined as P < 0.05. 3 Results 3.1 Clinical characteristics of remission and nonremission in patients with CD This study included 25 patients with CD who underwent BIPSS before surgery (Figure 1). Among them, 12 patients had microadenomas, whereas the remaining 13 had inconclusive magnetic resonance imaging findings; clinicopathological data are summarized in Supplementary Table 1. Table 1 displays the demographics of patients who achieved remission (n = 16) and those who did not (n = 9). No significant differences were observed in terms of sex, age at diagnosis, or radiological variables between patients who achieved and those who did not achieve remission (P > 0.05). Patients who achieved remission exhibited a higher prevalence of emotional lability (P < 0.05). However, no significant differences were observed in other parameters (P > 0.05). Figure 1 Figure 1 Flowchart of the screening process employed to select eligible participants for the study. Table 1 Table 1 Baseline clinical features of patients with pituitary tumors secreting adrenocorticotropin. Several recent studies have established morning cortisol level measured on postoperative day 1 (POD1) as a predictive biomarker for long-term remission of CD (35, 36). For biochemical features, patients who did not achieve remission exhibited higher serum cortisol (19.16 ± 5.55 vs. 5.95 ± 1.42; P = 0.014) and median serum (8 am) ACTH (10.26 ± 8.24 vs. 5.15 ± 3.68; P = 0.042) levels on POD1. No significant differences were observed in the preoperative baseline 4 pm serum cortisol levels, preoperative baseline 0 am serum cortisol levels, preoperative 8 pm ACTH levels, 4 pm ACTH levels, and 0 am ACTH levels (P > 0.05) (Table 2). In addition preoperative FT3, FT4, TSH, GH, FSH, LH, and PRL levels were comparable in patients with and without remission. Table 2 Table 2 Baseline clinical and biochemical features of patients with pituitary tumors secreting adrenocorticotropin. 3.2 Overexpression of SPHK1 and higher concentrations of serum S1P on the tumor side in patients with CD Prior studies have demonstrated that ACTH acutely activates SPHK1 to increase S1P concentrations (19). Upregulation of SPHK1 is associated with poor prognosis in endocrine-related cancer (17, 18, 21). To investigate the role of SPHK1 in CD, we performed a heatmap analysis of key genes involved in phospholipid metabolism and signaling pathways in CD adenomas and surrounding normal tissues using the GEO dataset (GEO208107). This analysis revealed the activation of crucial genes involved in phospholipid metabolism and signaling pathways in ACTH-secreting pituitary adenomas (Supplementary Figure 1). Subsequently, we compared the association between pituitary SPHK1 expression and proopiomelanocortin, corticotropin-releasing hormone, corticotropin releasing hormone receptor 1, and corticotropin releasing hormone receptor 2 in pituitary tumor tissues and identified a positive correlation between SPHK1 and ACTH tumor-related genes in the TNM plot database (Supplementary Figure 2). To investigate the potential role of SPHK1 in CD, we compared the expression values of SPHK1 in the normal pituitary tissues and those obtained from patients with CD in the remission/nonremission groups. Immunofluorescence staining (Figures 2A, B; Supplementary Figure 3) revealed an increased number of double-positive cells for SPHK1 and ACTH in CD-affected pituitary tissues than those in the normal pituitary tissues. Furthermore, the proportion of double-positive cells for SPHK1 and ACTH was significantly higher in the nonremission CD adenomas tissues than that in the remission CD adenomas. Furthermore, we investigated the concentration of S1P in bilateral petrosal sinus blood samples and observed that the concentration was significantly higher on the adenoma side than that on the nonadenoma side (397.7 ± 15.4 vs. 261.9 ± 14.88; P < 0.05, Figure 2C). Thus, these findings suggested a close association between S1P concentration and the development of ACTH-secreting tumor. Figure 2 Figure 2 (A) Representative images of immunofluorescence double staining for SPHK1 (green) and ACTH (pink) in normal pituitary glands and ACTH-secreting pituitary adenomas from the remission and nonremission groups (Normal: n = 3, ACTH pituitary adenoma: remission vs. nonremission: n = 16 vs. 9); scale bars: 100-μm upper and 50-μm lower. (B) Quantitative analysis; white arrows indicate double-positive cells for ACTH and SPHK1. (C) The concentration of S1P in the plasma obtained from the inferior petrosal sinus of the adenoma side and nonadenoma side. ***P < 0.001. Bar represents mean ± SD. 3.3 Combination of interpetrosal S1P and ACTH ratios improved the diagnostic performance for adenoma laterality The pathology of patients with CD was classified based on adenomatous tissue with ACTH-positive immunostaining into adenoma or nonadenoma sides. To evaluate the correlation between the interpetrosal S1P ratio lateralization and tumor location, we compared the accuracy of predicting tumor laterality using the interpetrosal S1P ratio (>1) and interpetrosal ACTH ratio (>1.4) (the interpetrosal ACTH ratio >1.4 is acknowledged for its positive role in predicting tumor laterality), as well as their combination. Our results indicated that using the interpetrosal S1P or ACTH ratios alone yielded accuracies of 64% and 56% respectively. Notably, the combination of both demonstrated a significantly improved accuracy of 73% (Figure 3A). Figure 3 Figure 3 (A) Bar graph illustrating the accuracy of predicting tumor laterality. (B) Receiver operating characteristic (ROC) curve analysis of interpetrosal ACTH ratio to predict tumor location. (C) ROC curve analysis of the interpetrosal S1P ratio to predict tumor location. (D) ROC curve analysis of the combination of the interpetrosal S1P and ACTH ratios to predict tumor location. Thereafter, the receiver operating characteristic analysis was performed to determine the role of predicting tumor laterality. In particular, the interpetrosal ACTH ratio with an AUC of 75.32% (95% CI: 60.06–97.46%, P < 0.05) and the interpetrosal S1P ratio demonstrated a clinically significant diagnostic accuracy for lateralization, with an AUC of 79.17% (95% CI: 44.40–85.84%, P < 0.05). Furthermore, combining the interpetrosal S1P and ACTH ratios generated an receiver operating characteristic curve with an AUC of 84.09% (95% CI: 52.3–96.77%, P < 0.05) for predicting lateralization with tumor location (cutoff value: interpetrosal S1P ratio ≥1.06, interpetrosal ACTH ratio ≥2.8, 81.82% sensitivity, and 75% specificity) (Figures 3B–D). 3.4 Interpetrosal S1P ratio serves as a predictive factor for early remission in CD To investigate whether the interpetrosal S1P ratio is associated with early postoperative remission in CD, we compared the baseline interpetrosal S1P ratio between patients with CD in the remission and nonremission groups. Interestingly, we observed that the nonremission group exhibited higher interpetrosal S1P ratios than those of the remission group (median, 1.28 ± 0.25 vs. 1.10 ± 0.09, P = 0.012) (Figure 4). Figure 4 Figure 4 Left picture: Scatter plot of bilateral S1P concentrations in the remission and nonremission groups; the slope represents the interpetrosal S1P ratio, blue dots represent the remission group, and red dots represent the nonremission group. Right picture: The interpetrosal S1P ratio in the remission and nonremission groups. *P < 0.05. Bar represents mean ± SD. To investigate potential factors affecting the interpetrosal S1P ratio, we compared the correlation between interpetrosal S1P ratio and various clinical indicators. This analysis revealed that the interpetrosal S1P ratio positively correlated with preoperative FSH and LH levels, as well as with postoperative 8 am ACTH levels. No significant difference was observed between the interpetrosal S1P ratio and other indicators (Supplementary Figure 4). 4 Discussion The use of BIPSS involves collection of samples from each inferior petrosal sinus simultaneously, enabling a direct comparison of ACTH concentrations between the left and right petrosal sinuses. BIPSS is used for two purposes: 1) to assist in the differential diagnosis of Cushing’s syndrome; and 2) to determine which side of the pituitary gland contains an adenoma in patients with CD. The interpetrosal ACTH ratio is also useful in determining the location/lateralization of pituitary microadenomas (24, 30, 37), thereby providing guidance to the neurosurgeon during surgery. To our knowledge, this is the first study to demonstrate that serum S1P levels in patients with CD are significantly higher on the adenoma side of the inferior petrosal sinus than on the nonadenoma side. The interpetrosal S1P ratio exhibited a positive significance in predicting tumor laterality, and the predictive performance was improved when S1P was combined with the interpetrosal ACTH ratio. Notably, the interpetrosal S1P ratio exhibited a positive significance in predicting remission after surgery. Furthermore, the interpetrosal S1P ratio demonstrated a positive and significant correlation with preoperative FSH and LH levels, as well as 8 am ACTH levels on POD1. ACTH is recognized for its role in controlling the expression of genes involved in steroid production and cortisol synthesis in the human adrenal cortex through sphingolipid metabolism (19). Specifically, ACTH rapidly stimulates SPHK1 activity, leading to an increased in S1P levels, which in turn, increases the expression of multiple steroidogenic proteins (20). Our study demonstrated that higher S1P concentrations were present on the tumor side than on the nontumor side in patients with CD, indicating that the regulatory relationship between ACTH and S1P also exists in ACTH-secreting pituitary adenomas. Several pieces of evidence have supported the potential relationship between S1P and the occurrence of CD. Interestingly, SPHK1 and S1P are known to be integral to the regulation of epidermal growth factor receptor (EGFR) (38), which is highly expressed in human corticotropinomas, where it triggers proopiomelanocortin (the precursor of ACTH) transcription and ACTH synthesis (39). Blocking EGFR activity with an EGFR inhibitor can attenuate corticotroph tumor cell proliferation (40). Furthermore, SPHK1 and proopiomelanocortin share a common transcriptional coactivator, P300 (41, 42). Notably, S1P also directly binds to and inhibits histone deacetylase 2, thereby regulating histone acetylation and gene expression (43). Notably, histone deacetylase 2 expression is deficient in ACTH-pituitary adenomas in CD, contributing to glucocorticoid insensitivity (44), which is a hallmark of CD and a feature associated with nonremission. These studies further demonstrated an association between high S1P ratio and nonremission of CD. Our study, for the first time, established an association between SPHK1/S1P and ACTH adenoma. Nevertheless, further experimental verification is required to confirm the existence of common pathways linking SPHK1 and ACTH. Thus, these findings indicated that the S1P ratio can, to some extent, reflect the differences in ACTH levels and may serve as a surrogate marker for detecting ACTH-secreting pituitary adenomas. BIPSS is a highly effective procedure for diagnosing pituitary sources of ACTH in CD and remains the gold standard diagnostic method. However, some findings indicated certain limitations associated with the use of the inferior petrosal sinus sampling (IPSS) method in predicting tumor lateralization. The possible causes of error include asymmetrical or underdeveloped petrosal sinus anatomy and placement of the catheter (27). The present study revealed a notable increase in the interpetrosal ACTH ratio among patients with accurate predictions of tumor laterality than among those with inaccurate predictions, although the positive predictive value remained low. These findings suggested that other mechanisms may exist that contribute to false-positive results. The limitations on lateralization highlighted the need for further research to understand the underlying mechanisms contributing to the accuracy of IPSS in predicting tumor lateralization. Further investigation is required to understand these potential mechanisms and improve the accuracy of IPSS in predicting tumor lateralization. We observed that the interpetrosal S1P ratio was slightly more effective than the ACTH ratio in predicting tumor laterality. However, combining both methods significantly improved the diagnostic sensitivity and specificity. These results have important implications for clinical practice as accurate tumor lateralization is essential for the correct management and treatment of pituitary adenomas. Overall, these findings highlighted the importance of using multiple measures in predicting tumor lateralization and suggested that combining measures may be more effective than relying on any single measure alone. Future research should investigate additional measures to improve the accuracy of tumor lateralization and optimize the use of existing measures for making clinical decisions. The initial treatment recommendation for CD is surgery. However, long-term surveillance is necessary because of the high recurrence rate (12). Therefore, identifying patients who are at a greater recurrence risk would be helpful in establishing an effective surveillance strategy. Our study revealed that the expression of SPHK1 in pituitary tissue was higher in postoperative nonremission group than in postoperative remission group. Moreover, patients in the nonremission group exhibited significantly higher interpetrosal S1P ratios than those of patients in the remission group. SPHK1 catalyzes the direct phosphorylation synthesis of S1P, and the S1P ratio can thus reflect the expression level of SPHK1 in ACTH tumors. Since S1P can increase the expression of multiple steroidogenic proteins, including steroidogenic acute regulatory protein, 18-kDa translocator protein, low-density lipoprotein receptor, and scavenger receptor class B type I (20), the interpetrosal S1P ratios may be indicative of disease prognosis. This finding is consistent with previous findings indicating the overexpression of SPHK1 is associated with poor prognosis in various neuroendocrine tumors, as factors associated with tumor proliferation, S1P and SPHK1, may play a key role in the proliferation and survival of ACTH pituitary adenomas. The high proportions of SPHK1/ACTH double-positive cells are likely associated with greater phenotypic severity, and CD tumors with this phenotype may have a poor prognosis. These findings hold clinically significance for predicting early postoperative remission in patients with CD. As aforementioned, the interpetrosal S1P ratios have been suggested as a useful diagnostic tool for determining adenoma lateralization in CD, which can also serve as a prognostic indicator for postoperative remission. Pearson correlation analysis indicated that ACTH 8 am on POD1 and FSH/LH levels were significantly associated with the interpetrosal S1P ratio, suggesting that these pituitary dysfunctions may have a role in the early remission of CD. However, the sample size in this study was relatively small, and further studies with larger sample sizes are needed to confirm these findings. Additionally, other factors affecting surgical outcomes, such as the experience of the surgeon, extent of surgical resection, and use of adjuvant therapy, should be considered when predicting postoperative remission in patients with CD. This study has some limitations. First, the study was retrospective in design, which limited the control of confounding factors. Additionally, because of the limited sample size, we did not specifically investigate cases where the ACTH ratio failed to accurately identify the correct tumor location. Finally, we did not explore the functional evidence of a common pathway between SPHK1 and ACTH. Despite these limitations, the study contributes to our understanding of the potential utility of the interpetrosal S1P ratio as a biomarker for CD and provides a basis for future research in this area. In conclusion, our study demonstrated a significant association between the interpetrosal S1P ratio and tumor laterality, as well as in early remission in CD. These findings suggested that the interpetrosal S1P ratio could serve as a useful biomarker in clinical practice. Moreover, targeting genes and drugs related to SPHK1/S1P could provide novel therapeutic strategies for treating CD. Data availability statement The original contributions presented in the study are included in the article/Supplementary Material. Further inquiries can be directed to the corresponding author. Ethics statement The studies involving humans were approved by The Xiangya Hospital Ethics Committee, Xiangya Hospital (Changsha, China). The studies were conducted in accordance with the local legislation and institutional requirements. The participants provided their written informed consent to participate in this study. Author contributions HS: conceptualization, methodology, software, visualization, and investigation. CW and BH: software. YX: writing – review & editing. All authors contributed to the article and approved the submitted version. Funding The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article. Acknowledgments The authors gratefully acknowledge contributions from the GEO databases and TNMplot database (https://www.tnmplot.com/). 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.2023.1238573/full#supplementary-material References 1. Tritos NA, Miller K. Diagnosis and management of pituitary adenomas: A review. JAMA (2023) 329(16):1386–98. doi: 10.1001/jama.2023.5444 PubMed Abstract | CrossRef Full Text | Google Scholar 2. Ntali G, Hakami O, Wattegama M, Ahmed S, Karavitaki N. Mortality of patients with cushing's disease. Exp Clin Endocrinol Diabetes (2021) 129(3):203–7. doi: 10.1055/a-1197-6380 CrossRef Full Text | Google Scholar 3. Hakami O, Ahmed S, Karavitaki N. Epidemiology and mortality of Cushing's syndrome. Best Pract Res Clin Endocrinol Metab (2021) 35(1):101521. doi: 10.1016/j.beem.2021.101521 PubMed Abstract | CrossRef Full Text | Google Scholar 4. Clayton RN, Raskauskiene D, Reulen RC, Jones PW. Mortality and morbidity in Cushing's disease over 50 years in Stoke-on-Trent, UK: audit and meta-analysis of literature. J Clin Endocrinol Metab (2011) 96(3):632–42. doi: 10.1210/jc.2010-1942 PubMed Abstract | CrossRef Full Text | Google Scholar 5. Jones PS, Swearingen B. Pituitary surgery in Cushing’s disease: first line treatment and role of reoperation. Pituitary (2022) 25:713–717. doi: 10.1007/s11102-022-01254-8 PubMed Abstract | CrossRef Full Text | Google Scholar 6. Juszczak A, Ertorer ME, Grossman AB. The therapy of cushing’s disease in adults and children: an update. Hormone Metab Res (2012) 45:109–117. doi: 10.1055/s-0032-1330009 CrossRef Full Text | Google Scholar 7. Biller BM, Grossman AB, Stewart PM, Melmed S, Bertagna XY, Bertherat J, et al. Treatment of adrenocorticotropin-dependent Cushing's syndrome: a consensus statement. J Clin Endocrinol Metab (2008) 93(7):2454–62. doi: 10.1210/jc.2007-2734 PubMed Abstract | CrossRef Full Text | Google Scholar 8. Chandler WF, Barkan AL, Hollon TC, Sakharova AA, Sack JT, Brahma B, et al. Outcome of transsphenoidal surgery for cushing disease: A single-center experience over 32 years. Neurosurgery (2016) 78(2):216–23. doi: 10.1227/NEU.0000000000001011 PubMed Abstract | CrossRef Full Text | Google Scholar 9. Feng M, Liu Z, Liu X, Bao X, Yao Y, Deng K, et al. Diagnosis and outcomes of 341 patients with cushing's disease following transsphenoid surgery: A single-center experience. World Neurosurg (2018) 109:e75–80. doi: 10.1016/j.wneu.2017.09.105 PubMed Abstract | CrossRef Full Text | Google Scholar 10. Aranda GB, Ensenãat J, Mora M, Puig-Domingo M, Martínez de Osaba MJ, Casals G, et al. Long-term remission and recurrence rate in a cohort of Cushing’s disease: the need for long-term follow-up. Pituitary (2014) 18:142–9. doi: 10.1007/s11102-014-0567-8 CrossRef Full Text | Google Scholar 11. Nieman LK, Biller BM, Findling JW, Murad MH, Newell-Price JD, Savage MO, et al. Treatment of cushing's syndrome: an endocrine society clinical practice guideline. J Clin Endocrinol Metab (2015) 100(8):2807–31. doi: 10.1210/jc.2015-1818 PubMed Abstract | CrossRef Full Text | Google Scholar 12. Galiana PA, Montañana CF, Suárez PA, Vela JG, Escrivá CM, Lillo VR. Predictors of long-term remission after transsphenoidal surgery in Cushing's disease. Endocrinol y Nutricioín Oírgano La Sociedad Espanãola Endocrinol y Nutricioín (2013) 60:475–82. Google Scholar 13. Ayala AR, Manzano AJ. Detection of recurrent Cushing’s disease: proposal for standardized patient monitoring following transsphenoidal surgery. J Neuro-Oncol (2014) 119:235–242. doi: 10.1007/s11060-014-1508-0 CrossRef Full Text | Google Scholar 14. Liu M, Frej C, Langefeld CD, Divers J, Bowden DW, Carr JJ, et al. Plasma apoM and S1P levels are inversely associated with mortality in African Americans with type 2 diabetes mellitus. J Lipid Res (2019) 60:1425–1431. doi: 10.1194/jlr.P089409 PubMed Abstract | CrossRef Full Text | Google Scholar 15. Anderson AK, Lambert JM, Montefusco DJ, Tran BN, Roddy P, Holland WL, et al. Depletion of adipocyte sphingosine kinase 1 leads to cell hypertrophy, impaired lipolysis, and nonalcoholic fatty liver disease. J Lipid Res (2020) 61:1328–1340. doi: 10.1194/jlr.RA120000875 PubMed Abstract | CrossRef Full Text | Google Scholar 16. Smith C, Williams J, Hall C, Casas J, Caley M, O’Toole E, et al. Ichthyosis linked to sphingosine 1-phosphate lyase insufficiency is due to aberrant sphingolipid and calcium regulation. J Lipid Res (2023) 64(4):100351. doi: 10.1016/j.jlr.2023.100351 PubMed Abstract | CrossRef Full Text | Google Scholar 17. Maczis MA, Maceyka M, Waters MR, Newton J, Singh M, Rigsby MF, et al. Sphingosine kinase 1 activation by estrogen receptor α36 contributes to tamoxifen resistance in breast cancer. J Lipid Res (2018) 59:2297–2307. doi: 10.1194/jlr.M085191 PubMed Abstract | CrossRef Full Text | Google Scholar 18. Hii L-W, Chung FF, Mai CW, Yee ZY, Chan HH, Raja VJ, et al. Sphingosine kinase 1 regulates the survival of breast cancer stem cells and non-stem breast cancer cells by suppression of STAT1. Cells (2020) 9(4):886. doi: 10.3390/cells9040886 PubMed Abstract | CrossRef Full Text | Google Scholar 19. Ozbay T, Merrill AH, Sewer MB. ACTH regulates steroidogenic gene expression and cortisol biosynthesis in the human adrenal cortex via sphingolipid metabolism. Endocrine Res (2004) 30:787–794. doi: 10.1081/ERC-200044040 CrossRef Full Text | Google Scholar 20. Lucki NC, Li D, Sewer MB. Sphingosine-1-phosphate rapidly increases cortisol biosynthesis and the expression of genes involved in cholesterol uptake and transport in H295R adrenocortical cells. Mol Cell Endocrinol (2012) 348:165–75. doi: 10.1016/j.mce.2011.08.003 PubMed Abstract | CrossRef Full Text | Google Scholar 21. Lin K, Cheng W, Shen Q, Wang H, Wang R, Guo S, et al. Lipid profiling reveals lipidomic signatures of weight loss interventions. Nutrients (2023) 15(7):1784. doi: 10.3390/nu15071784 PubMed Abstract | CrossRef Full Text | Google Scholar 22. Detomas M, Ritzel K, Nasi-Kordhishti I, Schernthaner-Reiter MH, Losa M, Tröger V, et al. Bilateral inferior petrosal sinus sampling with human CRH stimulation in ACTH-dependent Cushing's syndrome: results from a retrospective multicenter study. Eur J Endocrinol (2023) 2023:lvad050. doi: 10.1093/ejendo/lvad050 CrossRef Full Text | Google Scholar 23. Zampetti B, Grossrubatscher EM, Dalino Ciaramella P, Boccardi E, Loli P. Bilateral inferior petrosal sinus sampling. Endocrine Connections (2016) 5:R12–R25. doi: 10.1530/EC-16-0029 PubMed Abstract | CrossRef Full Text | Google Scholar 24. Oldfield EH, Doppman JL, Nieman LK, Chrousos GP, Miller DL, Katz D, et al. Petrosal sinus sampling with and without corticotropin-releasing hormone for the differential diagnosis of Cushing's syndrome. New Engl J Med (1991) 325(13):897–905. doi: 10.1056/NEJM199109263251301 PubMed Abstract | CrossRef Full Text | Google Scholar 25. Oldfield EH, Chrousos GP, Schulte HM, Schaaf M, Mckeever PE, Krudy AG, et al. Preoperative lateralization of ACTH-secreting pituitary microadenomas by bilateral and simultaneous inferior petrosal venous sinus sampling. New Engl J Med (1985) 312(2):100–3. doi: 10.1056/NEJM198501103120207 PubMed Abstract | CrossRef Full Text | Google Scholar 26. Colao A, Faggiano A, Pivonello R, Pecori Giraldi F, Cavagnini F, Lombardi G. Inferior petrosal sinus sampling in the differential diagnosis of Cushing's syndrome: results of an Italian multicenter study. Eur J Endocrinol (2001) 144(5):499–507. doi: 10.1530/eje.0.1440499 PubMed Abstract | CrossRef Full Text | Google Scholar 27. Lefournier V, Martinie M, Vasdev A, Bessou P, Passagia JG, Labat-Moleur F, et al. Accuracy of bilateral inferior petrosal or cavernous sinuses sampling in predicting the lateralization of Cushing's disease pituitary microadenoma: influence of catheter position and anatomy of venous drainage. J Clin Endocrinol Metab (2003) 88(1):196–203. doi: 10.1210/jc.2002-020374 PubMed Abstract | CrossRef Full Text | Google Scholar 28. Liu C, Lo JC, Dowd CF, Wilson CB, Kunwar SM, Aron DC, et al. Cavernous and inferior petrosal sinus sampling in the evaluation of ACTH-dependent Cushing's syndrome. Clin Endocrinol (2004) 61(4):478–86. doi: 10.1111/j.1365-2265.2004.02115.x CrossRef Full Text | Google Scholar 29. Batista DL, Gennari M, Riar J, Chang R, Keil MF, Oldfield EH, et al. An assessment of petrosal sinus sampling for localization of pituitary microadenomas in children with Cushing disease. J Clin Endocrinol Metab (2006) 91(1):221–4. doi: 10.1210/jc.2005-1096 PubMed Abstract | CrossRef Full Text | Google Scholar 30. 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 31. Hinojosa-Amaya JoséM, Cuevas-Ramos D. “The definition of remission and recurrence of Cushing's disease.” Best practice & research. Clin Endocrinol Metab (2021) 35(1):101485. doi: 10.1016/j.beem.2021.101485 CrossRef Full Text | Google Scholar 32. Deipolyi AR, Karaosmanoğlu AD, Habito CM, Brannan SM, Wicky ST, Hirsch JA, et al. The role of bilateral inferior petrosal sinus sampling in the diagnostic evaluation of Cushing syndrome. Diagn Interventional Radiol (2012) 18(1):132–8. doi: 10.4261/1305-3825.DIR.4279-11.0 CrossRef Full Text | Google Scholar 33. Deipolyi AR, Bailin A, Hirsch JA, Walker TG, Oklu R. Bilateral inferior petrosal sinus sampling: experience in 327 patients. J neurointerv Surg (2016) 9:196–199. doi: 10.1136/neurintsurg-2015-012164 PubMed Abstract | CrossRef Full Text | Google Scholar 34. Chongsathidkiet P, Jackson C, Koyama S, Loebel F, Cui X, Farber SH, et al. Sequestration of T-cells in bone marrow in the setting of glioblastoma and other intracranial tumors. Nat Med (2018) 24:1459–1468. doi: 10.1038/s41591-018-0135-2 PubMed Abstract | CrossRef Full Text | Google Scholar 35. Catalino MP, Moore DT, Ironside N, Munoz AR, Coley J, Jonas R, et al. Post-operative serum cortisol and cushing disease recurrence in patients with corticotroph adenomas. J Clin Endocrinol Metab (2023) 2023:dgad347. doi: 10.1210/clinem/dgad347 CrossRef Full Text | Google Scholar 36. Wang F, Catalino MP, Bi WL, Dunn IF, Smith TR, Guo Y, et al. Post-operative day one morning cortisol value as a biomarker to predict long-term remission of cushing disease. J Clin Endocrinol Metab (2021) 106(1):e94-e102. doi: 10.1210/clinem/dgaa773 PubMed Abstract | CrossRef Full Text | Google Scholar 37. Newell-Price JD, Bertagna XY, Grossman AB, Nieman LK. Cushing's syndrome. Lancet (2005) 367(9522):1605–17. doi: 10.1383/medc.2005.33.11.11 CrossRef Full Text | Google Scholar 38. Tamashiro PM, Furuya H, Shimizu Y, Kawamori T. Sphingosine kinase 1 mediates head & neck squamous cell carcinoma invasion through sphingosine 1-phosphate receptor 1. Cancer Cell Int (2014) 14(1):76. doi: 10.1186/s12935-014-0076-x PubMed Abstract | CrossRef Full Text | Google Scholar 39. Perez-Rivas LG, Theodoropoulou M, Ferraù 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. doi: 10.1210/jc.2015-1453 PubMed Abstract | CrossRef Full Text | Google Scholar 40. Fukuoka H, Cooper O, Ben-Shlomo A, Mamelak A, Ren SG, Bruyette D, et al. EGFR as a therapeutic target for human, canine, and mouse ACTH-secreting pituitary adenomas. J Clin Invest (2011) 121(12):4712–21. doi: 10.1172/JCI60417 PubMed Abstract | CrossRef Full Text | Google Scholar 41. Postepska-Igielska A, Giwojna A, Gasri-Plotnitsky L, Schmitt N, Dold A, Ginsberg D, et al. LncRNA khps1 regulates expression of the proto-oncogene SPHK1 via triplex-mediated changes in chromatin structure. Mol Cell (2015) 60(4):626–36. doi: 10.1016/j.molcel.2015.10.001 PubMed Abstract | CrossRef Full Text | Google Scholar 42. Smith RG, Betancourt L, Sun Y. Molecular endocrinology and physiology of the aging central nervous system. Endocrine Rev (2005) 26(2):203–50. doi: 10.1210/er.2002-0017 CrossRef Full Text | Google Scholar 43. Hait NC, Allegood J, Maceyka M, Strub GM, Harikumar KB, Singh SK, et al. Regulation of histone acetylation in the nucleus by sphingosine-1-phosphate. Science (2009) 325:1254–7. doi: 10.1126/science.1176709 PubMed Abstract | CrossRef Full Text | Google Scholar 44. Bilodeau S, Vallette-Kasic S, Gauthier Y, Figarella-Branger D, Brue T, Berthelet F, et al. Role of Brg1 and HDAC2 in GR trans-repression of the pituitary POMC gene and misexpression in Cushing disease. Genes Dev (2006) 20:2871–86. doi: 10.1101/gad.1444606 PubMed Abstract | CrossRef Full Text | Google Scholar Keywords: ipss, sphingosine-1-phosphate, Cushing’s disease, remission, tumor laterality Citation: Sun H, Wu C, Hu B and Xiao Y (2023) Interpetrosal sphingosine-1-phosphate ratio predicting Cushing’s disease tumor laterality and remission after surgery. Front. Endocrinol. 14:1238573. doi: 10.3389/fendo.2023.1238573 Received: 12 June 2023; Accepted: 17 October 2023; Published: 31 October 2023. Edited by: Anton Luger, Medical University of Vienna, Austria Reviewed by: Guangwei Wang, Hunan University of Medicine, China Marie Helene Schernthaner-Reiter, Medical University of Vienna, Austria Copyright © 2023 Sun, Wu, Hu and Xiao. 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: Yuan Xiao, xiaoyuan2021@csu.edu.cn 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.2023.1238573/full
    1 point
  28. Abstract Gastrointestinal perforation is a well-addressed complication of exogenous hypercortisolism; however, patients with endogenous Cushing's syndrome (CS) do not usually experience this condition in clinical practice. The literature on this subject is limited and consists solely of clinical case reports/series with only 23 instances of gastrointestinal perforation occurring in individuals with endogenous Cushing's syndrome. This is mainly attributed to the rarity of Cushing's syndrome itself and the low chance of occurrence of such complications. We report a case of a recently diagnosed adrenocorticotropic hormone (ACTH)-dependent Cushing's syndrome in a 30-years-old female who presented initially with a three-month history of progressive weight gain, generalized weakness, acne, menstrual irregularity, and severe hypokalemia, and then developed a gastric ulcer perforation only one month after her ACTH-dependent Cushing's syndrome diagnosis and was managed through emergent surgery. Introduction A disorder of the endocrine system characterized by excessive cortisol production, known as Cushing's syndrome, rarely occurs. The main causes are pituitary tumors, ectopic adrenocorticotropic hormone (ACTH)-secreting tumors, or adrenal tumors that secrete cortisol independently [1]. Patients initially present with a wide range of symptoms, including weight gain, proximal myopathy, skin thinning, and abdominal striae [1]. Additionally, several metabolic disorders, such as diabetes mellitus, hypertension, and dyslipidemia, can occur, especially when the diagnosis is not established at an early stage [2]. There is a possibility of gastrointestinal complications among patients receiving exogenous glucocorticoids. However, there is limited information on gastrointestinal complications associated with endogenous hypercortisolemia [3,4]. Thus far, only 23 instances have been published addressing the co-occurrence of gastrointestinal perforation with endogenous Cushing's syndrome [5-17]. To the best of our knowledge, this is the first case reporting gastric perforation in an ACTH-dependent Cushing's syndrome, while the vast majority reported diverticular, sigmoid, or duodenal perforation with Cushing's syndrome [5-17]. Herein, we describe the medical history, physical examination, and investigatory findings of a 30-year-old female with a recent diagnosis of ACTH-dependent Cushing's syndrome that was complicated by gastric ulcer perforation, necessitating an urgent exploratory laparotomy. The primary motivator of this case report was the rarity of the described condition, the atypical location of the perforation in such patient group, and the relatively young age of the patient. Case Presentation History and examination A 30-year-old female with a history of mental retardation was admitted to our emergency department (ER) with progressive weakness and fatigue. Upon taking the history, she had been having menstrual irregularities, progressive weight gain, and generalized weakness, which was significant enough to limit her physical activity and hinder her movement for the past three months. Initial vital signs showed that the patient had a body temperature of 37°C, a pulse rate of 90 beats per minute, and a blood pressure of 130/80 mmHg. On physical examination, the patient had a moon face with supraclavicular fullness, dorsocervical fat pad, purple abdominal striae, facial signs of hirsutism, and acne all over the face, shoulders, chest, and back. Investigations In the initial laboratory examination, hypokalemia of 2.1 mEq/L, hyperglycemia of 12.1 mmol/L, and metabolic alkalosis were detected (Table 1). The cortisol level after 1 mg dexamethasone suppression test was 2204 nmol/L (normal range 140-690), ACTH 123 pg/mL (normal range 7.2-63.3), DHEA-S 27.85 umol/L (normal range 2.6-13.9), And 24-hour urine cortisol level was 1560 mg/day (normal range 30-350) (Table 1). No suppression was observed in cortisol level with 8 mg dexamethasone suppression test. Parameter Initial presentation Perforation presentation Refrence range Na+ 143 mEq/L 139 mmol/L 135-147 mEq/L Cl- 85 mEq/L 105 mmol/L 98-108 mEq/L K+ 2.1 mEq/L 2.8 mmol/L 3.5-5.0 mEq/L Mg2+ 0.79 mmol/L 0.77 mmol/L 0.85-1.110 mmol/L PO3- 0.88 mmol/L 1.23 mmol/L 0.97-1.46 mmol/L PH 7.54 7.36 7.35-7.45 PCO2 67.5 mmHg 42.7 mmHg 35-45 mmHg PO2 27.7 mmHg 62.2 mmHg 75-100 mmHg HCO3 49.8 mEq/L 23.6 mEq/L 22-26 mEq/L Random blood glucose 12.1 mmol/L 24.1 mmol/L <5.5 mmol/L Hemoglobin 13.5 g/dL 14.9 g/dL 13.7-16.8 g/dL White blood cells 9,720 /uL 11,100 /uL 3,300-8,600 /uL Lymphocyte 0.48% 0.33% - Neutrophil 8.55% 9.66% - Eosinophil 0.0% 0.0% - TSH 0.55 mIU/L Was not ordered 0.4-4.0 mIU/L Cortisol 2204 nmol/L 4842 nmol/L 140-690 nmol/L ACTH 123 pg/mL Was not ordered 7.2-63.3 pg/mL Table 1: Laboratory findings on initial presentation and on perforation day TSH - thyroid stimulating hormone; ACTH - adrenocorticotropic hormone A series of CT scans for the neck, chest, abdomen, and pelvis was performed and failed to localize any tumors acting as an ectopic source. A pituitary MRI was performed, and no adenoma was found. To complete the diagnostic workup, we decided to do an inferior petrosal sinus sampling (IPSS) and PET scan with Gallium 68; however, the patient's family refused and requested discharge and outpatient follow-ups. These results, together with the biochemical and clinical findings, supported the diagnostic hypothesis of ACTH-dependent Cushing's syndrome. Treatment/management When addressing the issue of hypokalemia that the patient presented with initially, it was found to be resistant and difficult to correct. The patient was put on spironolactone 50 mg BID, and potassium chloride 20 mEq q8h, and her potassium level barely reached 3.5 mmol/L after several days. In addition, her magnesium level was corrected with magnesium oxide 800 mg every six hours. Her blood glucose level was controlled with insulin glargine 6 units daily and Novorapid as per the sliding scale. The patient was discharged on spironolactone tablets 50 mg BID (oral), potassium chloride 20 mEq q8h, cholecalciferol, calcium carbonate, insulin glargine 6 units daily, and Novorapid 4 units TID before meals. Follow-up and outcomes Seven days after discharge, she presented to the ER complaining of a new onset of abdominal pain, constipation, and reduced urine output. Her Glasgow Coma Scale (GCS) was 15, her blood pressure measurement was 146/90 mmHg, her pulse rate was 66 beats per minute, her respiratory rate was 21 breaths per minute, and her temperature was 36.7°C. Upon physical examination, the patient had distended non-tender abdomen without any other significant findings. Blood work was done, including renal functions, and all parameters, including potassium, were within normal limits. A chest X-ray was also performed and revealed no evidence of pneumoperitoneum. The patient was clinically stable after managing her abdominal pain with acetaminophen injection and administering fleet enema for constipation. After instructions on when to come again to the ER were given, the patient was discharged home on lactulose and paracetamol, and a close outpatient follow-up appointment was scheduled. Five days after the ER visit, the patient presented again to the ER. She was still complaining of severe non-resolving abdominal pain, constipation, and reduced urine output. Upon physical examination in the ER, the patient was found to have developed a new onset of lower limb edema, abdominal rebound tenderness, and abdominal rigidity and guarding. She was hypotensive with a blood pressure of 91/46 mmHg, pulse rate of 80 beats per minute, respiratory rate of 16 breaths per minute, temperature of 38.2 °C, and SpO2 of 96%. The only significant laboratory finding was her potassium level dropping low to 2.8 mEq/L (Table 1). An X-ray of the chest was requested and showed a large pneumoperitoneum (Figure 1). Figure 1: Posteroanterior chest X-ray at the time of gastric perforation displaying severe air under the diaphragm with bilateral obstruction indicating massive pneumoperitoneum (red arrow) Abdominal CT was also urgently performed and confirmed the presence of gastric perforation likely related to an underlying perforated peptic ulcer with 0.8 cm defect at the distal greater curvature (Figures 2, 3). Figure 2: Coronal-section CT image of abdomen and pelvis at the time of gastric perforation showing features of gastric perforation likely related to the underlying perforated peptic ulcer with 0.8 cm defect at the distal greater curvature Figure 3: Horizontal-section CT image showing features of gastric perforation likely related to the underlying perforated peptic ulcer with 0.8 cm defect at the distal greater curvature The patient underwent an emergent gastric wedge resection for gastric perforation, and the pathology reported evidence of gastric ulcer with no evidence of malignancy. Furthermore, Helicobacter pylori test was performed on the sample, and it came back positive. The patient tolerated the surgery very well, and postoperative recovery was without any complications. Later, the patient was prescribed metyrapone 250 mg Q4h, which was then increased to 500 mg Q4h four days after surgery, and her cortisol level significantly dropped to 634nmol/L. During that time, a gastrin level test was also performed to exclude the presence of gastrinomas, and the level was 45 pg/ml (normal range 13-115). Discussion A small percentage of the population suffers from Cushing's syndrome, which is an endocrine disorder characterized by an endogenous overproduction of glucocorticoids, resulting in hypercortisolemia [1]. It is estimated to affect 0.7 to 2.4 people per million annually [1]. Hypercortisolemia alters psychologic, metabolic, and cardiovascular functions, resulting in increased mortality and morbidity rates, particularly if the diagnosis is delayed and long-term exposure to high cortisol levels occurs [2]. Women are more likely to suffer from this condition than men, and people in their 40s to 60s are most vulnerable to it [1]. Patients initially present with a wide range of symptoms, including weight gain, proximal myopathy, skin thinning, and abdominal striae [1]. Additionally, several metabolic disorders, such as diabetes mellitus, hypertension, and dyslipidemia, can occur [1]. Due to the rarity of this condition, there is often a significant delay in diagnosis and treatment, which could eventually lead to complications from prolonged hypercortisolism. From another standpoint, in a systematic review, the incidence of peptic ulcer perforation ranges from 3.8 to 14 per 100,000 individuals in the general population [18]. In under-developed countries, patients are typically young, tobacco-using males [19]. However, patients in industrialized countries are typically older with multiple co-morbidities and are on long-term non-steroidal anti-inflammatory drugs (NSAIDs) or steroid use [19]. Patients may present with an abrupt onset of abdominal discomfort, abdominal rigidity, and tachycardia in the early stages of a perforated peptic ulcer [19]. Later, abdominal distention, pyrexia, hypotension, fever, and vomiting can occur [19]. Furthermore, when the diagnosis is made early, a perforated ulcer often has a good prognosis. However, the risk of adverse events increases if there is a delay in the diagnosis [20]. Therefore, making an early detection through different imaging modalities is crucial [20]. A history of peptic ulcer disease, NSAIDs, physiological stress, smoking, corticosteroids, and Helicobacter pylori are some of the well-established risk factors for a perforated peptic ulcer [20]. The prevalence of Helicobacter pylori among Saudi patients is high; in one study, the overall prevalence was 46.5% in patients with dyspepsia using gastric biopsy [21]. Several studies have explored the relationship between Helicobacter pylori and gastrointestinal perforation, but the results have been mixed. Some studies have suggested a higher prevalence of Helicobacter pylori infection among individuals with gastrointestinal perforation compared to those without, indicating a potential association. However, other studies have found no significant difference in the prevalence of Helicobacter pylori infection between perforated and non-perforated gastrointestinal ulcer cases [22]. Furthermore, they suggested that the presence of other risk factors like the use of NSAIDs, smoking, and alcohol may interact with Helicobacter pylori infection and contribute to the development of complications such as gastrointestinal perforation [22]. However, in our case, the patient did not have any established risk factors for gastric perforation, such as NSAIDs, smoking, or alcohol. Therefore, considering the low incidence of gastrointestinal perforation and high prevalence of Helicobacter pylori, the conflicting data regarding the association between Helicobacter pylori and gastrointestinal perforation, and the lack of established risk factors for gastrointestinal perforation in our patient, we suggest that prolonged excess glucocorticoids from Cushing's syndrome may have contributed to the gastric perforation either independently or synergistically with Helicobacter pylori since hypercortisolism can lead to a weakened gastrointestinal wall integrity due to decreased collagen turnover and disruption of mucosal protection by prostacyclin [15]. In addition, because of hypercortisolism, perforation may not be contained or healed initially due to the immunosuppressive effects of hypercortisolism, whether endogenous or exogenous [15]. Additionally, high levels of cortisol may delay the diagnosis and treatment since it may mask the symptoms of the perforation [14]. Moreover, our patient was treated for severe hypokalemia with potassium supplementation for an extended period of time. Previous studies have linked potassium chloride supplementation to gastrointestinal ulceration and perforation, making this a possible additive cause of our patient's condition [23,24]. A limited number of studies have addressed gastrointestinal perforations associated with endogenous hypercortisolemia [5-17]. The correlation between Cushing's syndrome and gastrointestinal perforation is highlighted in our study and in the case reports that have been previously published (Table 2). Similar to our case, a female predominance was seen in gastrointestinal perforation among the reported cases of Cushing's syndrome [6,7,12,13,15,16]. Additionally, the average age at which gastrointestinal perforation occurred in patients with endogenous hypercortisolism ranged from 45 to 80, which is a noticeably higher age range than the case we are presenting here (aged 30) [6-10,12]. Furthermore, unlike our case, in which gastrointestinal perforation occurred four months after the onset of Cushing's symptoms, Intestinal perforation occurs approximately 9.8 months after Cushing's symptoms first appear [15]. Furthermore, in our patient, gastric perforation occurred while she was hypercortisolemic and not in a remission state. Hence, in association with Helicobacter pylori infection, severe hypercortisolemia could have been a secondary contributing factor to gastric perforation. The complications of gastric ulceration, specifically with endogenous Cushing's syndrome, have been addressed in two case reports [25,26]. It must be noted, however, that neither case is similar to ours. A case of gastric perforation was reported by Kubicka et al. in a patient who had a confirmed diagnosis of gastrinoma, and the patient was diagnosed with ectopic Cushing's syndrome seven months after gastric perforation [25]. Therefore, since ectopic Cushing's syndrome was diagnosed seven months after the perforation, it is more likely that the gastrinoma contributed to this complication. In contrast, our patient's serum gastrin level was within the normal range, ruling out gastrinoma. Further, Hoshino et al. reported a case of gastrointestinal bleeding in a 39-year-old man with a confirmed diagnosis of Cushing's disease secondary to pituitary adenoma [26]. He was found to have gastric ulceration and bleeding along with Helicobacter pylori infection and elevated cortisol levels [26]. In spite of the patient not developing a gastric perforation, it was suggested by the author that hypercortisolism might be a contributing factor for gastric ulcer complications by slowing down the ulcer healing process [26] Reference Year of publication Age, gender Highest cortisol level plasma cortisol (PC, nmol/L) / UFC (nmol/L) Cause of Cushing's syndrome Time from onset of Cushing's symptoms to perforation (months) Reported site of gastrointestinal perforation Current 2023 30, Female PC 4842 ACTH-dependant 4 Gastric perforation Ishinoda et al. [17] 2023 24, Male PC 1647 Cushing's disease 12 Sigmoid colon perforation Wijewickrama et al. [16] 2021 32, Female PC 1147 Pituitary microadenoma 1 Diverticular perforation Shahidi et al. [15] 2019 72, Female UFC 5296 Pancreatic neuroendocrine tumor 12 Diverticular perforation Shahidi et al. [15] 2019 61, Female PC 1925 Metastatic medullary carcinoma of thyroid 12 Sigmoid colon and diverticular perforation Shahidi et al. [15] 2019 68, Female UFC 410 Cushing's disease 12 Sigmoid colon perforation Shahidi et al. [15] 2019 71, Female UFC 1533 Cushing's disease 4 Diverticular perforation Shahidi et al. [15] 2019 54, Male UFC 374 Cushing's disease 3 Sigmoid colon perforation Shahidi et al. [15] 2019 52, Female UFC 885 Cushing's disease 16 Diverticular perforation Sater et al. [14] 2018 80, Female UFC 5601 Lung carcinoid 36 Diverticular perforation Sater et al. [14] 2018 60, Female UFC 72726 Metastatic islet cell carcinoma 36 Diverticular perforation Sater et al. [14] 2018 31, Male UFC 1297 Cushing's disease 20 Diverticular perforation Sater et al. [14] 2018 52, Female UFC 2371 Lung carcinoid 4 Diverticular perforation Sater et al. [14] 2018 67, Male UFC 3836 Ectopic ACTH 10 Diverticular perforation Sater et al. [14] 2018 51, Male UFC 13552 Metastatic thymic carcinoma 4 Diverticular perforation Kaya et al. [9] 2016 70, Male PC 1432 Small cell lung cancer 1 Diverticular perforation Dacruz et al. [12] 2016 60, Female UFC 4481 Metastatic parotid tumor 5 Sigmoid colon and diverticular perforation Matheny et al. [10] 2016 67, Male UFC 11119 Metastatic medullary carcinoma of thyroid 4 Diverticular perforation Flynn et al. [13] 2016 63, Female UFC 12465 Pheochromocytoma 1 Perforation at the splenic flexure Balestrieri et al. [11] 2016 75, Male PC 2272 Neuroendocrine tumor 1 Intestinal perforation Hara et al, [8] 2013 79, Male PC 1230 Cushing's disease 6 Diverticular perforation De Havenon et al. [7] 2011 71, Female PC 2593 Cushing's disease 9 Diverticular perforation Lutgers et al. [6] 2010 55, Female UFC 10152 Right pheochromocytoma 1 Sigmoid colon and diverticular perforation Drake et al. [5] 1998 35, Male PC 1442 Islet cell tumor 4 Duodenal perforation and rupture of pancreatic pseudocyst Table 2: Current case and previous reported 23 cases of patients with Cushing's syndrome and gastrointestinal perforation UFC - urinary free cortisol; PC - plasma cortisol; ACTH - adrenocorticotropic hormone Conclusions A high blood cortisol level can be associated with various clinical manifestations and diverse sets of complications. This case report sheds light on one of the less common complications of hypercortisolism in patients with Cushing's syndrome, which is gastrointestinal perforation. Our report further supports the published evidence that gastrointestinal perforation is a rare but potentially fatal complication among patients with Cushing's syndrome. Moreover, it highlights the possibility of developing gastric perforations in this patient group, even at younger ages than expected. This should elicit a high clinical suspicion and demand prompt investigation of Cushing's syndrome patients in a hypercortisolism state presenting with modest gastrointestinal symptoms. References Pivonello R, De Martino MC, De Leo M, Lombardi G, Colao A: Cushing's syndrome. Endocrinol Metab Clin North Am. 2008, 37:135-49. 10.1016/j.ecl.2007.10.010 Newell-Price J, Bertagna X, Grossman AB, Nieman LK: Cushing's syndrome. Lancet. 2006, 367:1605-17. 10.1016/S0140-6736(06)68699-6 Goethals L, Nieboer K, De Smet K, De Geeter E, Tabrizi NH, Van Eetvelde E, de Mey J: Cortisone associated diverticular perforation. JBR-BTR. 2011, 94:348-9. 10.5334/jbr-btr.705 Piekarek K, Israelsson LA: Perforated colonic diverticular disease: the importance of NSAIDs, opioids, corticosteroids, and calcium channel blockers. Int J Colorectal Dis. 2008, 23:1193-7. 10.1007/s00384-008-0555-4 Drake WM, Perry LA, Hinds CJ, Lowe DG, Reznek RH, Besser GM: Emergency and prolonged use of intravenous etomidate to control hypercortisolemia in a patient with Cushing's syndrome and peritonitis. J Clin Endocrinol Metab. 1998, 83:3542-4. 10.1210/jcem.83.10.5156 Lutgers HL, Vergragt J, Dong PV, de Vries J, Dullaart RP, van den Berg G, Ligtenberg JJ: Severe hypercortisolism: a medical emergency requiring urgent intervention. Crit Care Med. 2010, 38:1598-601. 10.1097/CCM.0b013e3181e47b7a de Havenon A, Ehrenkranz J: A perforated diverticulum in Cushing's disease. Int J Surg Case Rep. 2011, 2:215-7. 10.1016/j.ijscr.2011.06.009 Hara T, Akutsu H, Yamamoto T, Ishikawa E, Matsuda M, Matsumura A: Cushing's disease presenting with gastrointestinal perforation: a case report. Endocrinol Diabetes Metab Case Rep. 2013, 2013:130064. 10.1530/EDM-13-0064 Kaya T, Karacaer C, Açikgöz SB, Aydemir Y, Tamer A: Severe hypokalaemia, hypertension, and intestinal perforation in ectopic adrenocorticotropic hormone syndrome. J Clin Diagn Res. 2016, 10:OD09-11. 10.7860/JCDR/2016/17198.7127 Matheny LN, Wilson JR, Baum HB: Ectopic ACTH production leading to diagnosis of underlying medullary thyroid carcinoma. J Investig Med High Impact Case Rep. 2016, 4:2324709616643989. 10.1177/2324709616643989 Balestrieri A, Magnani E, Nuzzo F: Unusual Cushing's syndrome and hypercalcitoninaemia due to a small cell prostate carcinoma. Case Rep Endocrinol. 2016, 2016:6308058. 10.1155/2016/6308058 Dacruz T, Kalhan A, Rashid M, Obuobie K: An ectopic ACTH secreting metastatic parotid tumour. Case Rep Endocrinol. 2016, 2016:4852907. 10.1155/2016/4852907 Flynn E, Baqar S, Liu D, et al.: Bowel perforation complicating an ACTH-secreting phaeochromocytoma. Endocrinol Diabetes Metab Case Rep. 2016, 2016:10.1530/EDM-16-0061 Sater ZA, Jha S, McGlotten R, Hartley I, El Lakis M, Araque KA, Nieman LK: Diverticular perforation: A fatal complication to forestall in Cushing syndrome. J Clin Endocrinol Metab. 2018, 103:2811-4. 10.1210/jc.2018-00829 Shahidi M, Phillips RA, Chik CL: Intestinal perforation in ACTH-dependent Cushing's syndrome. Biomed Res Int. 2019, 2019:9721781. 10.1155/2019/9721781 Wijewickrama PS, Ratnasamy V, Somasundaram NP, Sumanatilleke M, Ambawatte SB: A challenging case of Cushing's disease complicated with multiple thrombotic phenomena following trans-sphenoidal surgery; a case report. BMC Endocr Disord. 2021, 21:29. 10.1186/s12902-021-00701-0 Ishinoda Y, Uto A, Meshino H, et al.: Bowel perforation associated with Cushing's disease: a case report with literature review. Endocr J. 2023, 70:933-9. 10.1507/endocrj.EJ23-0110 Lau JY, Sung J, Hill C, Henderson C, Howden CW, Metz DC: Systematic review of the epidemiology of complicated peptic ulcer disease: incidence, recurrence, risk factors and mortality. Digestion. 2011, 84:102-13. 10.1159/000323958 Chung KT, Shelat VG: Perforated peptic ulcer - an update. World J Gastrointest Surg. 2017, 9:1-12. 10.4240/wjgs.v9.i1.1 Weledji EP: An overview of gastroduodenal perforation. Front Surg. 2020, 7:573901. 10.3389/fsurg.2020.573901 Akeel M, Elmakki E, Shehata A, Elhafey A, Aboshouk T, Ageely H, Mahfouz MS: Prevalence and factors associated with H. pylori infection in Saudi patients with dyspepsia. Electron Physician. 2018, 10:7279-86. 10.19082/7279 Thirupathaiah K, Jayapal L, Amaranathan A, Vijayakumar C, Goneppanavar M, Nelamangala Ramakrishnaiah VP: The association between Helicobacter pylori and perforated gastroduodenal ulcer. Cureus. 2020, 12:e7406. 10.7759/cureus.7406 Farquharson-Roberts MA, Giddings AE, Nunn AJ: Perforation of small bowel due to slow release potassium chloride (slow-K). Br Med J. 1975, 3:206. 10.1136/bmj.3.5977.206 Payan H, Blaustein A: Potassium chloride and small bowel perforation. Gastroenterology. 1965, 48:877-8. 10.1016/S0016-5085(65)80073-7 Kubicka E, Zawadzka K, Syrycka J, Kałużny M, Pawluś A, Bolanowski M: A case of gastrinoma associated with ectopic Cushing syndrome. Pol Arch Intern Med. 2020, 130:328-9. 10.20452/pamw.15201 Hoshino C, Satoh N, Narita M, Kikuchi A, Inoue M: Another 'Cushing ulcer'. BMJ Case Rep. 2011, 2011:10.1136/bcr.02.2011.3888 From https://www.cureus.com/articles/196132-adrenocorticotropic-hormone-dependent-cushings-syndrome-complicated-with-gastric-ulcer-perforation-in-a-30-year-old-saudi-female-a-case-report-and-a-review-of-the-literature#!/
    1 point
  29. Abstract Summary This case report describes a rare presentation of ectopic Cushing’s syndrome (CS) due to ectopic corticotropin-releasing hormone (CRH) production from a medullary thyroid carcinoma (MTC). The patient, a 69-year-old man, presented with symptoms of muscle weakness, facial plethora, and easy bruising. An inferior petrosal sinus sampling test (IPSS) demonstrated pituitary adrenocorticotrophic hormone (ACTH) secretion, but a whole-body somatostatin receptor scintigraphy (68Ga-DOTATOC PET/CT) revealed enhanced uptake in the right thyroid lobe which, in addition to a grossly elevated serum calcitonin level, was indicative of an MTC. A 18F-DOPA PET/CT scan supported the diagnosis, and histology confirmed the presence of MTC with perinodal growth and regional lymph node metastasis. On immunohistochemical analysis, the tumor cell stained positively for calcitonin and CRH but negatively for ACTH. Distinctly elevated plasma CRH levels were documented. The patient therefore underwent thyroidectomy and bilateral adrenalectomy. This case shows that CS caused by ectopic CRH secretion may masquerade as CS due to a false positive IPSS test. It also highlights the importance of considering rare causes of CS when diagnostic test results are ambiguous. Learning points Medullary thyroid carcinoma may secrete CRH and cause ectopic CS. Ectopic CRH secretion entails a rare pitfall of inferior petrosal sinus sampling yielding a false positive test. Plasma CRH measurements can be useful in selected cases. Keywords: Adult; Male; White; Denmark; Pituitary; Pituitary; Thyroid; Error in diagnosis/pitfalls and caveats; September; 2023 Background The common denominator of Cushing’s syndrome (CS) is autonomous hypersecretion of cortisol (1) and it is subdivided into ACTH-dependent and ACTH-independent causes. The majority of CS cases are ACTH-dependent (80–85%) with a pituitary corticotroph tumor as the most prevalent cause (Cushing’s disease), and less frequently an ectopic ACTH-producing tumor (2). The gold standard method to ascertain the source of ACTH secretion in CS patients is inferior petrosal sinus sampling (IPSS) with measurement of plasma ACTH levels in response to systemic corticotropin-releasing hormone (CRH) stimulation (3). The IPSS has a very high sensitivity and specificity of 88–100% and 67–100%, respectively (4), but pitfalls do exist, including the rare ectopic CRH-producing tumor, which may yield a false positive test result (3). Here, we describe a very rare case masquerading as CS including a positive IPSS test. Case presentation A 69-year-old man presented at a local hospital with a 6-month history of progressive fatigue, muscle weakness and wasting, easy bruising, facial plethora, and fluid retention. His serum potassium level was 2.6 mmol/L (reference range: 3.5–4.2 mmol/L) without a history of diuretics use. His previous medical history included spinal stenosis, benign prostatic hyperplasia, and hypertension. An electromyography showed no sign of polyneuropathy and an echocardiography showed no signs of heart failure with an ejection fraction of 55%. MRI of the spine revealed multiple compression fractures, and the patient underwent spinal fusion and decompression surgery; during this admission he was diagnosed with type 2 diabetes (HbA1c: 55 mmol/mol). After spine surgery, the patient developed a pulmonary embolism and initiated treatment with rivaroxaban. Establishing the diagnosis of ACTH-dependent CS Six months after his spine surgery, the patient was referred to the regional department of endocrinology for osteoporosis management. Blood tests revealed a low serum testosterone level with non-elevated luteinizing hormone (LH) and follicle-stimulating hormone (FSH) levels (Table 1). An overnight 1 mg dexamethasone suppression test was positive with a morning cortisol level of 254 nmol/L and three consecutive 24-h urinary cortisol levels were markedly elevated with mean level of ≈600 nmol/24 h (reference range: 12–150 nmol/24 h). A single plasma ACTH was 37 ng/L (Table 1). Table 1 Baseline endocrine assessment. Parameters Patient’s values Reference range ACTH, ng/L 37 7–64 UFC, nmol/day 588 12–150 Urinary cortisol, nmol/L 600 171–536 OD, nmol/L 254 <50 Free testosterone, nmol/L 0.061 0.17–0.59 HbA1c, mmol/mol 55 <48 FSH, IU/L 7.4 1.2–15.8 LH, IU/L 2.2 1.7–8.6 ACTH, adrenocorticotropin; FSH, follicle-stimulating hormone; IU, international units; LH, luteinizing hormone; OD, plasma cortisol levels after a 1 mg overnight dexamethasone suppression test; UFC, urine free cortisol hormone. Differential diagnostic tests The patient was referred to a tertiary center for further examinations. Ketoconazole treatment was started to alleviate the consequences of hypercortisolism. A pituitary MRI revealed an intrasellar microtumor with a maximal diameter of 6 mm and an IPSS was ordered. A whole-body somatostatin receptor scintigraphy (68Ga-DOTATOC PET/CT) was also performed to evaluate the presence of a potential neuroendocrine tumor. This revealed multiple areas of enhanced uptake including the right thyroid lobe and cervical lymph nodes in the neck (with CT correlates), as well as in the duodenum (with no CT correlate). Concomitantly, a grossly elevated serum calcitonin level of 528 pmol/L (reference range <2.79 pmol/L) was measured. Subsequently, the IPSS revealed pituitary ACTH secretion with a central-to-peripheral ACTH ratio >3 (Table 2). The right petrosal sinus was not successfully catheterized; thus, lateralization could not be determined. To corroborate the diagnosis MTC, a 18F-DOPA PET/CT scan (FDOPA) was performed (5), which showed pathologically enhanced uptake in the right thyroid lobe and regional lymph nodes (Fig. 1). An ultrasound-guided core needle biopsy from the thyroid nodule was inconclusive; however, the patient underwent total thyroidectomy and regional lymph node resection, from which histology confirmed the diagnosis of disseminated MTC. Standard replacement with levothyroxine, calcium, and vitamin D was initiated. A blood sample was collected, and genomic DNA was extracted. The DNA analysis for RET germline mutation was negative. View Full Size Figure 1 18F-DOPA PET/CT scan with pathologically enhanced uptake in the right thyroid lobe (large blue arrow on the left side) and regional lymph nodes (small blue arrows). Citation: Endocrinology, Diabetes & Metabolism Case Reports 2023, 3; 10.1530/EDM-23-0057 Download Figure Download figure as PowerPoint slide Table 2 Results from the inferior petrosal sinus sampling.* Time (min) Left IPSS Peripheral L/P -5 42 36 1.2 -1 116 33 3.5 2 120 32 3.8 5 209 28 7.5 7 180 43 4.2 10 529 34 15.6 15 431 37 11.6 *Data represents ACTH levels in ng/L. IPSS Inferior petrosal sampling ACTH Adrenocorticotropin hormone CRH Corticotropin-releasing hormone, L/P Ratio of left (L) inferior petrosal sinus to peripheral venous ACTH concentrations. Pathology Total thyroidectomy and bilateral cervical lymph node dissection (level six and seven) were performed. Macroscopic evaluation of the right thyroid lobe revealed a 24 mm, irregular solid yellow tumor. Microscopically the tumor showed an infiltrating architecture with pseudofollicles and confluent solid areas. Calcification was prominent, but no amyloid deposition was seen. The tumor cells were pleomorphic with irregular nuclei and heterogenic chromatin structure. No mitotic activity or necrosis was observed. On immunohistochemical analysis, the tumor cells expressed thyroid transcription factor 1 and stained strongly for carcinoembryonic antigen and calcitonin; tumor cells were focally positive for cytokeratin 19. The tumor was completely negative for ACTH, thyroid peroxidase, and the Hector Battifora mesothelial-1 antigen. Further analysis revealed positive immunostaining for CRH (Fig. 2). The Ki-67 index was very low (0–1%), indicating a low cellular proliferation. Molecular testing for somatic RET mutation was not performed. View Full Size Figure 2 Histopathological findings and immunohistochemical studies of MTC. (A) Microscopic features of medullary thyroid carcinoma. (B) Polygonal tumor cells (hematoxylin and eosin, ×40). (C) Tumor cells stain for calcitonin (×20). (D) Immunohistochemical stain (×400) for CRH showing cells being positive (brown). (E) Pituitary tissue from healthy control staining positive for ACTH in comparison to (F) ACTH-negative cells MTC tissue from the patient (×20). Citation: Endocrinology, Diabetes & Metabolism Case Reports 2023, 3; 10.1530/EDM-23-0057 Download Figure Download figure as PowerPoint slide No malignancy was found in the left thyroid lobe and there was no evidence of C-cell hyperplasia. Regional lymph node metastasis was found in 13 out of 15 nodes with extranodal extension. Outcome and follow-up Follow-up Serum calcitonin levels declined after neck surgery but remained grossly elevated (118 pmol/L 3 weeks post surgery) and cortisol levels remained high. Ketoconazole treatment was poorly tolerated and not sufficiently effective. Plasma levels of CRH were measured by a competitive-ELISA kit (EKX-KIZI6R-96 Nordic BioSite), according to the instructions provided by the manufacturer. The intra- and interassay %CV (coefficient of variability) were below 8% and 10%, respectively, and the assay sensitivity was 1.4 pg/mL. The plasma CRH was distinctly elevated compared to in-house healthy controls both before and after thyroid surgery (Fig. 3). View Full Size Figure 3 Plasma CRH levels before and after total thyroidectomy compared to three healthy controls. Citation: Endocrinology, Diabetes & Metabolism Case Reports 2023, 3; 10.1530/EDM-23-0057 Download Figure Download figure as PowerPoint slide The patient subsequently underwent uneventful bilateral laparoscopic adrenalectomy followed by standard replacement therapy with hydrocortisone and fludrocortisone. The symptoms and signs of his CS gradually subsided. Pathology revealed bilateral cortical hyperplasia as expected. The patient continues follow-up at the Department of Oncology and the Department of Endocrinology and Internal Medicine. At 13-month follow-up, 68Ga-DOTATOC shows residual disease with pathologically enhanced uptake in two lymph nodes, whereas the previously described focal DOTATOC uptake in the duodenum was less pronounced (still no CT correlate). Serum calcitonin was 93 pmol/L at the 13-month follow-up. Discussion Diagnostic challenges remain in the distinction between pituitary and ectopic ACTH-dependent CS, and several diagnostic tools are used in combination, none of which is infallible, including IPSS (6). Our case and others illustrate that ectopic CRH secretion may yield a false positive IPSS test result (3). Measurement of circulating CRH levels is relevant if an ectopic CRH producing tumor is suspected, but the assay is not routinely available in clinical practice (Lynnette K Nieman M. Measurement of ACTH, CRH, and other hypothalamic and pituitary peptides https://www.uptodate.com/contents/measurement-of-acth-crh-and-other-hypothalamic-and-pituitary-peptides: UpToDate; 2019). In our case, the presence of elevated plasma CRH levels after thyroidectomy strengthened the indication for bilateral adrenalectomy. The most common neoplasm causing ectopic CS is small-cell lung cancer, whereas MTC accounts for 2–8% of ectopic cases (7). The development of CS in relation to MTC is generally associated with advanced disease and poor prognosis of an otherwise relatively indolent cancer (8), and the clinical progression of CS is usually rapid, why an early recognition and rapid control of hypercortisolemia and MTC is necessary to decrease morbidity and mortality (7, 9). Our case does have residual disease; however, he remains progression-free with stable and relatively low calcitonin levels within 1-year follow-up. Only a very limited number of cases of ectopic tumors with either combined ACTH and CRH secretion or isolated CRH secretion have been reported, with ectopic CRH secretion accounting for less than 1% of CS (9). An ACTH- or CRH-producing tumor can be difficult to localize and may include gastric ACTH/CRH-secreting neuroendocrine tumors (9). In our case, the 68Ga-DOTATOC identified a possible duodenal site, in addition to the MTC, but an upper gastrointestinal endoscopy revealed no pathological findings and there was no CT correlate. Thus, we concluded that the most likely and sole source of ectopic CRH was the MTC and its metastases. To our knowledge, no official guidelines for managing ectopic ACTH-dependent CS have been established. In a recent publication by Alba et al. (10), the authors demonstrated a clinical algorithm (The Mount Sinai Clinical Pathway, MSCP) and recommendation for the management of CS due to ectopic ACTH secretion. Essentially, our approach in this particular case followed these recommendations, including source location by CT and 68Ga-DOTATATE PET/CT imaging, acute management with ketoconazole, and finally, bilateral adrenalectomy as curative MTC surgery was not possible. In retrospect, performance of the IPSS could be questioned in view of the MTC diagnosis. In real time, however, a pituitary MRI performed early in the diagnostic process revealed a microadenoma, which prompted the IPSS. In parallel, a somatostatin receptor scintigraphy (68Ga-DOTATOC PET/CT) was also done, which raised the suspicion of an MTC. Conclusion We report a very rare case of an ectopic CS caused by a CRH-secreting MTC. Although IPSS has stood the test of time in the differential diagnosis of ACTH-dependent CS, this case illustrates a rare pitfall. Declaration of interest The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported. Funding This research did not receive any specific grant from any funding agency in the public, commercial, or not-for-profit sector. Patient consent Written informed consent for publication of their clinical details was obtained from the patient. Author contribution statement JOJ and MJO are the physicians responsible for the patient. LR performed the thyroidectomy and bilateral adrenalectomy. SHM and SLA assessed and reassessed the histopathology and the immunohistochemical analysis. MB measured plasma CRH. VM, JOJ, and MJO drafted the manuscript. All authors contributed to critical revision of the manuscript. References 1↑ Raff H, & Carroll T. Cushing's syndrome: from physiological principles to diagnosis and clinical care. Journal of Physiology 2015 593 493–506. (https://doi.org/10.1113/jphysiol.2014.282871) PubMed Search Google Scholar Export Citation 2↑ Hatipoglu BA. Cushing's syndrome. Journal of Surgical Oncology 2012 106 565–571. (https://doi.org/10.1002/jso.23197) PubMed Search Google Scholar Export Citation 3↑ Vassiliadi DA, Mourelatos P, Kratimenos T, & Tsagarakis S. Inferior petrosal sinus sampling in Cushing’s syndrome: usefulness and pitfalls. Endocrine 2021 73 530–539. (https://doi.org/10.1007/s12020-021-02764-4) PubMed Search Google Scholar Export Citation 4↑ Zampetti B, Grossrubatscher E, Dalino Ciaramella P, Boccardi E, & Loli P. Bilateral inferior petrosal sinus sampling. Endocrine Connections 2016 5 R12–R25. (https://doi.org/10.1530/EC-16-0029) PubMed Search Google Scholar Export Citation 5↑ Treglia G, Rufini V, Salvatori M, Giordano A, & Giovanella L. PET imaging in recurrent medullary thyroid carcinoma. International Journal of Molecular Imaging 2012 2012 324686. (https://doi.org/10.1155/2012/324686) PubMed Search Google Scholar Export Citation 6↑ Fasshauer M, Lincke T, Witzigmann H, Kluge R, Tannapfel A, Moche M, Buchfelder M, Petersenn S, Kratzsch J, Paschke R, et al.Ectopic Cushing' syndrome caused by a neuroendocrine carcinoma of the mesentery. BMC Cancer 2006 6 108. (https://doi.org/10.1186/1471-2407-6-108) PubMed Search Google Scholar Export Citation 7↑ Chrisoulidou A, Pazaitou-Panayiotou K, Georgiou E, Boudina M, Kontogeorgos G, Iakovou I, Efstratiou I, Patakiouta F, & Vainas I. Ectopic Cushing's syndrome due to CRH secreting liver metastasis in a patient with medullary thyroid carcinoma. Hormones 2008 7 259–262. (https://doi.org/10.1007/BF03401514) PubMed Search Google Scholar Export Citation 8↑ Corsello A, Ramunno V, Locantore P, Pacini G, Rossi ED, Torino F, Pontecorvi A, De Crea C, Paragliola RM, Raffaelli M, et al.Medullary thyroid cancer with ectopic Cushing's syndrome: a case report and systematic review of detailed cases from the literature. Thyroid 2022 32 1281–1298. (https://doi.org/10.1089/thy.2021.0696) PubMed Search Google Scholar Export Citation 9↑ Sharma ST, Nieman LK, & Feelders RA. Cushing's syndrome: epidemiology and developments in disease management. Clinical Epidemiology 2015 7 281–293. (https://doi.org/10.2147/CLEP.S44336) PubMed Search Google Scholar Export Citation 10↑ Alba EL, Japp EA, Fernandez-Ranvier G, Badani K, Wilck E, Ghesani M, Wolf A, Wolin EM, Corbett V, Steinmetz D, et al.The Mount Sinai clinical pathway for the diagnosis and management of hypercortisolism due to ectopic ACTH syndrome. Journal of the Endocrine Society 2022 6 bvac073. (https://doi.org/10.1210/jendso/bvac073) PubMed Search Google Scholar Export Citation From https://edm.bioscientifica.com/view/journals/edm/2023/3/EDM23-0057.xml
    1 point
  30. Ball-and-stick model of the cortisol (hydrocortisone) molecule. Credit: Public Domain A first-of-its kind hormone replacement therapy that more closely replicates the natural circadian and ultradian rhythms of our hormones has shown to improve symptoms in patients with adrenal conditions. Results from the University of Bristol-led clinical trial are published today in the Journal of Internal Medicine. Low levels of a key hormone called cortisol is typically a result of conditions such as Addison's and congenital adrenal hyperplasia. The hormone regulates a range of vital processes, from cognitive processes such as memory formation, metabolism and immune responses, through to blood pressure and blood sugar levels. When low, it can trigger symptoms of debilitating fatigue, nausea, muscle weakness, dangerously low blood pressure and depression. Although rare, these adrenal conditions require lifelong daily hydrocortisone replacement therapy. Although existing oral hormone replacement treatment can restore cortisol levels, it is still associated with an impaired quality of life for patients. Scientists believe this is because the current treatment does not mimic the body's normal physiological timing, missing cortisol's anticipatory rise and lacking its underlying ultradian and circadian rhythms. The new "pulsatility" therapy, the culmination of ten years' research by the Bristol team, is designed to deliver standard hydrocortisone replacement to patients via a pump which replicates more closely cortisol's natural rhythmic secretion pattern. The pulsatile subcutaneous pump has now revealed promising results in its first clinical trial. Twenty participants aged 18 to 64 years with adrenal insufficiency conditions were assessed during the double-blinded PULSES six-week trial and treated with usual dose hydrocortisone replacement therapy administered either via the pump or the standard three times daily oral treatment. While only psychological and metabolic symptoms were assessed during the trial, results revealed the pump therapy decreased fatigue by approximately 10%, improved mood and increased patient energy levels by 30% first thing in the morning—a key time frame when many patients struggle. Patient MRI scans also revealed alteration in the way that the brain processes emotional information. Dr. Georgina Russell, Honorary Lecturer at the University's Bristol Medical School, and the lead author, explained, "Patients on cortisol replacement therapy often have side effects which makes it difficult for them to lead normal lives. We hope this new therapy will offer greater hope for the thousands of people living with hormone insufficiency conditions." Stafford Lightman, a neuroendocrinology expert and Professor of Medicine at Bristol Medical School: Translational Health Sciences (THS), and the study's joint lead author, added, "Besides reduction in dosage, cortisol replacement has remained unchanged for many decades. It is widely recognized that current replacement therapy is unphysiological due to its lack of pre-awakening surge, ultradian rhythmicity, and post dose supraphysiological peaks. The new therapy clearly shows that the timing of cortisol delivery- in line with the body's own rhythmic pattern of cortisol secretion—is important for normal cognition and behavior. "Our findings support the administration of hormone therapy that mimics natural physiology, and is one of the first major advances in adrenal insufficiency treatment to date." Joe Miles, a participant on the PULSES trial, explained, "The Crono P pump has been life-changing. I noticed a very quick improvement compared to tablets when I was on the PULSES study. I went from feeling tired all the time to having sudden energy. "When the PULSES study ended and I had to return the pump, I simply couldn't cope with going back to how I used to be, so I made it my mission to write to as many doctors to have it prescribed privately. "I've now been on it for six years and have introduced a number of other people with Addison's disease to the pump, and all of them have said it's life changing. Some have gone from being seriously ill to feeling better than they have done for years." Dr. Russell said, "Approximately 1% of the UK population is taking steroids at any moment in time; these individuals can experience debilitating psychological side effects. This trial has shown that even at physiological levels, brain functioning is disrupted and that we need to explore not only the dose but the pattern of steroids delivery when considering any type of steroid treatment." More information: Ultradian hydrocortisone replacement alters neuronal processing, emotional ambiguity, affect and fatigue in adrenal insufficiency: The PULSES trial, Journal of Internal Medicine (2023). DOI: 10.1111/joim.13721 Journal information: Journal of Internal Medicine Provided by University of Bristol From https://medicalxpress.com/news/2023-10-first-of-its-kind-hormone-treatment-patient.html
    1 point
  31. I'm not sure I like this! STORY: Could artificial intelligence be used to make brain surgery safer? At this university in London, trainee surgeon Danyal Khan is taking part in a mock operation during which he's assisted by a real-time video feed, as is typical in brain surgery. But what's new here is that the footage is being analyzed by AI to help Khan better understand what he's seeing. The AI system, which is under development at University College London (UCL), highlights sensitive or critical structures in the brain. Neurosurgeon Hani Marcus believes it has the potential to make brain surgery safer and more effective: "So I'm very bullish that in the medium to long term, the A.I. will be helping lots of surgeons do lots of operations better than they otherwise can." Marcus says the AI system analyzed video of more than 200 pituitary gland tumor operations, and gained around 10 years-worth of experience in a fraction of the time. That knowledge means the AI can now not only help navigate to the correct area of the brain, but also know what should be happening at any stage of the procedure, making it a valuable training aid. "So, what we're really trying to do is apply AI or artificial intelligence to support surgeons doing brain tumor surgery at the base of the brain. And what this practically entails is us training the AI with hundreds of videos, telling it, if you like, what structures are what and then at some point over that period, the AI becomes really good itself at recognising things, and able to support other surgeons who're perhaps less experienced in advising them what to do next." Assistant Professor of Robotics and A.I. Sophia Bano explains how that might look in a real operation: "There can be scenarios where clinicians, unintentionally, are very close to a very critical structure such as the optic nerve. This can have, any damage or a slight more pressure on the optic nerve, can have long term complications on the patient side. So, this whole tool will alert the surgeon during the procedure if there is any risk of potential complication so they can recalibrate themselves during the procedure." Khan, who was also involved in developing the software that is now helping him learn, says the A.I. system has been a valuable tool in his training. It could also provide him with step-by-step guidance during a procedure, similar to having a senior surgeon standing over your shoulder. "I think as I progress as a surgeon, there might be stages where I wonder, you know, have I done enough of a particular part of the procedure and should I move on? And actually having that sort of assistant in the background as a reassurance to look at and say, 'well, yeah, actually, at this stage, out of the hundreds of videos of experts that this algorithm has watched, the experts would probably start moving on to the next phase'. It's a useful double check." According to UCL, the system could be ready to be used in operating theaters within two years. From https://news.yahoo.com/ai-could-help-brain-surgery-080156167.html
    1 point
  32. Abstract Background and Objectives Crooke cell adenomas (CCA) are a rare, aggressive subset of adrenocorticotrophin secreting pituitary corticotroph adenomas (sCTA) found in 5–10% of patients with Cushing’s disease. Multiple studies support worse outcomes in CCA but are limited by small sample size and single-institution databases. We compared outcomes in CCA and sCTA using a multicenter, international retrospective database of high-volume skull base centers. Methods Patients surgically treated for pituitary adenoma from January 2017 through December 2020 were included. Results 2826 patients from 12 international centers were compared (n=20 CCA and n=480 sCTA). No difference in baseline demographics, tumor characteristics or postoperative complications was seen. Microsurgical approaches (60% CCA vs. 62.3% sCTA) were most common. Gross total resection (GTR) was higher in CCA patients (100% vs. 83%, p=0.05). Among patients that had GTR according to intraoperative findings, fewer CCA patients had postoperative hormone normalization of pituitary function (50% vs. 77.8%, p<0.01) and remission of hypersecretion by 3-6 months (75% vs. 84.3%, p<0.01). This was present despite CCA having better local control rates (100% vs. 96%, p<0.01) and fewer patients with remnant on MRI (0% vs. 7.2%, p<0.01). A systematic literature review of 35 studies reporting on various treatment strategies reiterated the high rate of residual tumor, persistent hypercortisolism, and tumor-related mortality in CCA patients. Conclusion This modern, multicenter series of patients with CCA reflects their poor prognosis and reduced post-surgical hormonal normalization. Further work is necessary to better understand the pathophysiology of CCA to devise more targeted treatment approaches. References (0) Cited by (0) Previous presentations: none Previous publications: none Disclosures No relevant disclosures to report CREDIT statement Matthew Finlay: conceptualization, writing – review and editing Richard Drexler: conceptualization, writing – review and editing All: data curation, writing – review and editing Michael Karsy: conceptualization, data curation, methodology, writing – original draft, writing – review and editing, supervision Funding and Disclosures: none View full text From https://www.sciencedirect.com/science/article/abs/pii/S187887502301344X
    1 point
  33. Bridget Houser felt despairing. In the months before her 2018 wedding, Houser, who had never struggled with her weight, noticed that it inexplicably began to creep up. In response she doubled the length of her runs to eight miles, took back-to-back high intensity workout classes and often consumed only water, coffee and fruit during the day before a spartan, mostly vegetable, dinner. Yet no matter what Houser did, her weight stubbornly increased and her oval face grew round, a transformation that was glaringly obvious in comparison with her identical twin sister. Houser wondered whether the five pounds she gained despite her herculean effort was a corollary of other problems. For the previous two years she had battled a string of maladies: first daily headaches, then crippling anxiety, followed by insomnia, hair loss and acne, something she’d never endured as a teenager. “Stress was the universal explanation,” recalled Houser, a controller for a small business in Chicago. When doctors suggested that her upcoming marriage might be a cause of her problems, Houser considered, then rejected, the theory. It just didn’t jibe with her feelings. In early 2019, about six months after her wedding, Houser insisted that her doctors perform several tests. They ultimately revealed that her symptoms weren’t the result of stress or marital misgivings but of a serious illness that had been smoldering for years. After successful treatment followed by a long recovery Houser, now 34, feels far better than she did during those miserable years in her late 20s. “I wish I’d been nicer to myself and not blamed myself for what was going on,” she said. Getting through the wedding In 2016 Houser began experiencing daily pain in the back of her head, a common spot for tension headaches. When the headaches failed to improve with dietary changes or nonprescription pain relievers, she consulted her primary care doctor, followed by a neurologist who told her she had migraines. Houser, then 27, noticed that the headaches were worse when she wore contact lenses. “It was affecting my daily life and I talked myself into thinking the problem was my contacts,” she said. She decided Lasik surgery might help and in October 2017 underwent the procedure, which uses a laser to reshape the cornea, reducing or eliminating dependence on contacts or glasses. Her vision improved and the pain disappeared — briefly. A week after eye surgery, her headaches returned. “I wasn’t overly concerned,” Houser said. “I know a lot of people have headaches.” A few months later for no apparent reason Houser developed “really bad anxiety. It wasn’t just like I was anxious,” she recalled. “I couldn’t function. I’m Type A so I knew what anxiety is, but not to this degree.” One weekday morning in early 2018 she felt so overwhelmed that she took a sick day, then called her twin, Molly, and their mother and told them she needed help immediately. They managed to schedule a same-day appointment with a psychiatrist whom Houser began seeing regularly, along with a therapist. The psychiatrist zeroed in on her impending wedding and told Houser that the event can cause “huge anxiety.” She began taking an antidepressant along with Ativan, an anti-anxiety drug she used when things got really bad. She also ramped up her yoga practice, hoping it might calm her. Houser vividly remembers riding the escalator to her office one morning “and in my head I kept saying, ‘I’m in trouble, I’m in trouble,’” although she didn’t know what was wrong. Her changing appearance had become a source of great unhappiness. Although her weight remained in the normal range, Houser couldn’t figure out why she was gaining weight after drastically slashing her food intake and dramatically ramping up exercise. Her normally thick hair had thinned so noticeably that her hairdresser gently advised her to consult a doctor. Houser’s psychiatrist thought her hair loss might be caused by her antidepressant and switched medications. That didn’t seem to help. Houser was particularly bothered by her newly chubby face. “It was like a joke in my family,” she said, adding that she was teased about being overly sensitive. Even her wedding day was colored by unhappiness about her appearance and the intense amorphous anxiety that seemed omnipresent. “Rather than think about how excited I was,” Houser recalled, “it was ‘How can I get through this day?’” Normal thyroid After her wedding Houser felt worse. She developed severe insomnia, night sweats and acne. In February 2019 a nurse practitioner in her primary care practice ordered tests of her thyroid, which were normal. When Houser pressed for additional testing, she was referred to an endocrinologist. He told her she was stressed. Dissatisfied, she saw a second endocrinologist who agreed with the first. “She said ‘I don’t think there’s anything wrong with you’” metabolically, Houser recalled. The second endocrinologist’s nurse even revisited the marriage question in the presence of Houser’s husband, Doug, who had accompanied her to the appointment. “She said ‘I knew on my honeymoon I shouldn’t have gotten married,’” Houser remembered her saying. “‘Are you in a happy marriage?’ I couldn’t believe it.” Months earlier, the nurse practitioner who ordered the thyroid tests briefly mentioned measuring levels of cortisol, a hormone involved in the body’s response to stress and other functions. Elevated levels of cortisol can indicate Cushing’s syndrome, an uncommon hormonal disorder that occurs when the body produces too much of the hormone over a prolonged period. “She had thrown cortisol testing out there and I think it was always in the back of my mind,” Houser said. She asked the second endocrinologist to order cortisol tests. The doctor agreed, but not before telling Houser that she didn’t think she had Cushing’s because she lacked the classic symptoms: major weight gain, purple stretch marks and a fatty hump between the shoulders. Houser did have the “moon face” characteristic of Cushing’s that is also seen in people who take high doses of steroids for long periods to treat various illnesses — but Houser wasn’t taking steroids. Insomnia, headaches, acne and anxiety can be symptoms of Cushing’s. There are several forms of Cushing’s syndrome, which typically results from a tumor — usually benign but sometimes cancerous — in the pituitary or adrenal gland that pumps out excess cortisol. Sometimes tumors develop elsewhere in the body such as the lungs or pancreas. Cushing’s affects roughly five times as many women as men and typically occurs between the ages of 30 and 50. If left untreated, it can be fatal. A trio of tests measuring cortisol levels in Houser’s blood, urine and saliva were significantly elevated; the amount in her urine was eight times higher than normal. The formerly skeptical Chicago endocrinologist told Houser she had Cushing’s and referred her to James Findling, a Milwaukee endocrinologist who is internationally recognized for his treatment of the disease. “I was just so happy to have a diagnosis,” Houser recalled. Revealing photos Findling asked Houser to bring photographs taken several years earlier to her October 2018 appointment. It is a request he makes of patients as a way of spotting telltale physical manifestations. In Houser’s case, the facial change was particularly striking because she is an identical twin. Findling noted that delayed diagnosis is typical, because physical changes and other symptoms tend to occur gradually and insidiously. Houser, he added, “didn’t look like the typical Cushing’s patient. She wasn’t obese and she didn’t have diabetes or hypertension. It was more subtle than many cases.” The next step was determining the location of the tiny tumor. Tests found nothing in Houser’s pituitary or adrenal glands, and CT scans of her pelvis, chest and abdomen were clean. Findling ordered a dotatate PET scan, a highly sensitive CT scan that can find tumors that elude conventional imaging. The scan revealed a nodule in Houser’s left lung. Houser sought a second opinion from a thoracic surgeon in Chicago. While Findling and a thoracic surgeon at Milwaukee’s Froedtert Hospital strongly recommended that she undergo surgery to remove the tumor, the Chicago doctor disagreed. He said he didn’t think the lung nodule was causing Cushing’s and recommended that Houser continue therapy and anti-anxiety medication. “Do you know what it’s like to wake up from surgery and to not be better?” she remembers him asking her. After deliberating with her husband and conferring with her Milwaukee doctors, Houser opted for surgery performed Oct. 30, which removed part of her left lung. A pathologist determined that the nodule was a rare, slow-growing neuroendocrine lung cancer known as a bronchial carcinoid, which can cause Cushing’s. The Stage 2 cancer had spread to a nearby lymph node. “Fortunately I think we got it early,” Findling said. “She’s had a sustained remission and a cure of her Cushing’s.” “The cancer didn’t rock my world,” said Houser, who had previously had a melanoma skin cancer removed. (Doctors have told her they don’t think the cancers are related.) “It was about not having Cushing’s anymore, which was more important.” So why didn’t Houser’s doctors, among them endocrinologists, suspect Cushing’s? Findling, who estimates he has treated as many as 2,000 people with the disease in his 40-year career, said that while doctors are taught that Cushing’s is rare, it’s not. He cites a 2016 study, which that found that 26 of 353 endocrinology patients were found to have the disease. Textbook descriptions, which include the presence of purple stretch marks and a hump, are “almost a caricature,” Findling observed. “It’s pretty well recognized that Cushing’s is more subtle than that … and can cause neuropsychiatric and neurocognitive problems.” Houser’s normal weight and the fact that she didn’t have high blood pressure or diabetes may have misled doctors. “I think we’ve moved the needle a little bit, especially among endocrinologists,” he continued, adding that “the threshold for screening has got to change. Once you tell a primary care doctor that it’s a rare disorder, it goes in one ear and out the other. They think they’ll never see it.” “When you make this diagnosis it can have fabulous outcomes,” he added, citing Houser’s case. “That’s why I’m still doing this at my age.” Houser considers Findling to be her “literal lifesaver.” She spent the next year seeing him as she was slowly weaned off medications to normalize her hormone levels and recover her strength. She is monitored for Cushing’s annually, remains cancer-free and, other than residual fatigue, feels well. In October 2021 she gave birth to a daughter. Her son was born eight weeks ago. Houser regards the help provided by her family, particularly her husband whom she called “my biggest supporter,” as essential. That seems especially ironic because stress about their marriage had been blamed for symptoms that were actually caused by a cancer. “He was a huge help in calling doctors and making the necessary appointments when I didn’t have the energy to fight anymore.” His unwavering love, she said, was “a testament to our strong marriage.” From https://www.washingtonpost.com/wellness/2023/10/07/weight-anxiety-wedding-medical-mysteries/
    1 point
  34. Highlights The most common cause of ectopic ACTH syndrome is pulmonary carcinoid tumors and squamous cell lung cancer; however it is a relatively uncommon complication of pulmonary neoplasms. The most common cause of Cushing syndrome is iatrogenic corticosteroid use and it should be considered in all patients regardless of clinical background. Low urine cortisol levels may be associated with exogenous glucocorticoid exposure. Occult glucocorticoid exposure is rare but can be evaluated with liquid chromatography. Consumers should be aware of the potential risks of taking supplements, especially those advertised as joint pain relief products. Abstract Background Well differentiated bronchial neuroendocrine neoplasms often follow a clinically indolent course and rarely cause Ectopic ACTH syndrome. Iatrogenic corticosteroid use is the most common cause of Cushing syndrome and should be considered in all patients regardless of clinical background. Case report A 59 year old woman with an 11 year history of a 1.5 cm well differentiated bronchial carcinoid, presented with Cushingoid features. Laboratory results were not consistent with an ACTH dependent Cushing Syndrome and exogenous steroids were suspected. The patient received an FDA alert regarding a glucosamine supplement she had started 4 months prior for joint pain. Discussion Ectopic ACTH production is reported in less than 5% of patients with squamous cell lung cancer and 3% of patients with lung or pancreatic (non-MEN1) neuroendocrine tumors. Factitious corticoid exposure is rare and can be evaluated with synthetic corticosteroid serum testing. Conclusion Cushing syndrome due to supplements containing unreported corticosteroid doses should be considered in patients with typical Cushingoid features and contradictory hormonal testing. 1. Introduction Well differentiated bronchial neuroendocrine neoplasms often follow a clinically indolent course and can rarely exhibit Cushing syndrome due to ectopic production of adrenocorticotropic hormone (ACTH). However the most common cause of Cushing syndrome is iatrogenic corticosteroid use and should be considered in all patients regardless of clinical background (see Fig. 1, Fig. 2, Fig. 3, Fig. 4). Download : Download high-res image (243KB) Download : Download full-size image Fig. 1. DOTATATE PET/CT demonstrates a right upper lobe pulmonary nodule with intense uptake. Download : Download high-res image (201KB) Download : Download full-size image Fig. 2. DOTATATE PET/CT demonstrates intense uptake within a right upper lobe pulmonary nodule, consistent with biopsy-proven carcinoid tumor. There are no distant sites of abnormal uptake to suggest metastatic disease. Download : Download high-res image (399KB) Download : Download full-size image Fig. 3. Artri Ajo King Supplement (Source: FDA). The label claims that the product contains glucosamine, chondroitin, collagen, vitamin C, curcumin, nettle, omega 3, and methylsulfonylmethane. Download : Download high-res image (288KB) Download : Download full-size image Fig. 4. Artri King Supplement (Source: FDA). 2. Case report A 59–year old woman with an 11 year history of a 1.5 cm well-differentiated bronchial carcinoid, presented with 20 lb. weight gain, facial swelling, flushing, lower extremity edema and shortness of breath over 3 months. On exam, the patient was normotensive, centrally obese with mild hirsutism, facial fullness and ruddiness with evidence of a dorsocervical fat pad. Initially there was concern for hormonal activation of her known bronchial carcinoid. Testing resulted in a normal 24-hour urine 5-HIAA (6 mg/d, n < 15 mg/dL), elevated chromogranin A (201 ng/mL, n < 103 ng/mL), normal histamine (<1.5 ng/mL, n < 1.7 ng mL), low-normal 7 AM serum cortisol (5.1 μg/dL, n 3.6–19.3 μg/dL), normal 7 AM ACTH (17 pg/mL, n < 46 pg/mL) and a surprisingly low 24-hr urinary free cortisol (1.8 mcg/hr, n 4.0–50.0 mcg/hr). A late night saliva cortisol was 0.03 mcg/dL (n 3.4–16.8 mcg/dL). Testosterone, IGF-1, glucose and electrolytes were appropriate. An echocardiogram showed an ejection fraction of 60% with no evidence of carcinoid heart disease. A Dotatate PET-CT was obtained to evaluate for progression of the neuro-endocrine tumor and revealed a stable right upper lobe pulmonary nodule with no evidence of metastatic disease. Given low cortisol levels, ectopic Cushing syndrome was excluded and exogenous steroids were suspected, however the patient denied use of oral,inhaled, or injected steroids. A cosyntropin stimulation study yielded a pre-stimulation cortisol 6.2 μg/dL with an adequate post-stimulation cortisol 23.5 μg/dL. At this stage of evaluation, the patient received an FDA alert regarding a glucosamine supplement she had started 4 months prior for joint pain. The notification advised of hidden drug ingredients including dexamethasone, diclofenac, and methocarbamol contained within Artri King Glucosamine supplements not listed on the product label but verified by FDA lab analysis. The FDA had received several adverse event reports including liver toxicity and even death associated with such products. The patient's symptoms gradually improved after discontinuation of the supplement. 3. Discussion 3.1. Ectopic ACTH syndrome This patient's Cushingoid features were initially suspected to be secondary to the known bronchial neuroendocrine tumor. Ectopic ACTH production accounts for about 5–10% of all Cushing Syndrome cases [1]. The most common location of ectopic ACTH is the lungs with pulmonary carcinoid tumors being the most common cause, followed by squamous cell lung cancer [2]. Despite this patient's history of bronchial carcinoid tumor and positive chromogranin histopathological marker, her laboratory results were not consistent with an ACTH dependent Cushing Syndrome. In fact, Cushing syndrome is a relatively uncommon neuroendocrine neoplasm complication. The prevalence of ectopic ACTH production in patients with lung tumors is rare, at less than 5% in squamous cell lung cancer and about 3% in patients with lung or pancreatic (non-MEN1) neuroendocrine tumors1. Patients with ACTH dependent Cushing syndrome not suspected to originate from the pituitary, undergo further testing to evaluate for an ectopic ACTH secreting tumor. These tests include conventional imaging of the chest, abdomen and pelvis, as well as functional imaging such as octreotide scans, fluoride 18-fluorodeoxyglucose-positron emission tomography [18F-FDG PET], and gallium-68 DOTATATE positron emission tomography-computed tomography [Dotatate PET-CT] scan [3]. In our literature review, we found that there was insufficient evidence to determine the sensitivity and specificity of nuclear medicine imaging techniques [4,5]. In this case, the patient had no laboratory evidence for ACTH dependent Cushing Syndrome, but given the known bronchial carcinoid tumor, a repeat Dotatate PET-CT scan was obtained which demonstrated no indication of growth or spread of the known bronchial tumor. 3.2. Supplement induced Cushing Syndrome One of the most remarkable findings in this case was the patient's low urine cortisol level in the setting of her overt Cushingoid features. In our survey of the literature, we found that low urine cortisol levels were associated with exogenous glucocorticoid use [6,7]. The low urine cortisol levels may be reflective of intermittent glucocorticoid exposure. Indeed, this patient's Cushingoid features were determined to be secondary to prolonged use of Artri King supplement. Occult glucocorticoid use is difficult to diagnose even after performing a thorough medication reconciliation as patients may unknowingly consume unregulated doses of glucocorticoids in seemingly harmless supplements and medications. The incidence of supplement induced Cushing Syndrome is currently unknown as supplements are not regularly tested to detect hidden glucocorticoid doses. Additionally, the likelihood of developing supplement induced Cushing syndrome is dependent on dosage and duration of use. In our literature review we found nine published articles describing supplement induced Cushing Syndrome [[7], [8], [9], [10], [11], [12], [13], [14], [15]], one case report of tainted counterfeit medication causing Cushing Syndrome [16], and two cases of substances with probable glucocorticoid-like activity [17,18]. Of the nine published articles of supplement induced Cushing Syndrome, six were associated with supplements marketed as arthritic joint pain relief products including ArtriKing, Maajun, and AtriVid [[7], [8], [9], [10], [11], [12]]. These products later received government issued warnings in Mexico, Malaysia, and Colombia respectively [[19], [20], [21]]. To our knowledge there have been four published reports of ArtiKing supplement induced Cushing Syndrome [[7], [8], [9], [10]]. The first documented cases were reported in 2021 in Vera Cruz, Mexico; since then the Mexican medical community reported seeing a disproportionate increase in cases of iatrogenic Cushing Syndrome due to these supplements [7]. There have also been three American published articles describing a total of 4 cases of ArtriKing supplement induced Cushing syndrome [[8], [9], [10]]. In January 2022 the FDA issued a warning about Atri Ajo King containing diclofenac, which was not listed in the product label [22]. In April 2022 the FDA expanded its warning, advising consumers to avoid all Artri and Ortiga products after the FDA found these products contained dexamethasone and diclofenac [23]. In October 2022 the FDA issued warning letters to Amazon, Walmart, and Latin Foods market for distributing Artri and Ortiga products [24]. Many supplements are not regulated by the government and may contain hidden ingredients such as glucocorticoids. In these cases further evaluation of suspected products [25], medications [16], and patient serum [26] and urine [6] utilizing techniques such as liquid chromatography may be used to confirm occult glucocorticoid exposure. This case highlights the importance of educating patients to exercise caution when purchasing health products both online and abroad. Consumers should be aware of the potential risks of taking supplements, especially those advertised as joint pain relief products. 4. Conclusion Although the most common cause of ectopic ACTH syndrome is pulmonary carcinoid tumors and squamous cell lung cancer, it is a relatively uncommon complication of pulmonary neoplasms. Exogenous Cushing syndrome due to supplements containing unreported corticosteroid doses should be considered in patients with typical Cushingoid features and contradictory hormonal testing. Occult glucocorticoid exposure is rare but can be evaluated with liquid chromatography. This case report emphasizes the importance of teaching patients to be vigilant and appropriately research their health supplements. Patient consent Formal informed consent was obtained from the patient for publication of this case report. Declaration of competing interest The authors (Tomas Morales and Shanika Samarasinghe) of this case report declare that they have no financial conflicts of interest. Shanika Samrasinghe is an editorial member of the Journal of Clinical and Translational Endocrinology: Case Reports, and declares that she was not involved in the peer review and editorial decision making process for the publishing of this article. References [1] A.R. Hayes, A.B. Grossman The ectopic adrenocorticotropic hormone syndrome: rarely easy, always challenging Endocrinol Metab Clin N Am, 47 (2) (2018 Jun), pp. 409-425, 10.1016/j.ecl.2018.01.005 PMID: 29754641 View PDFView articleView in ScopusGoogle Scholar [2] A.M. Isidori, A. Lenzi Ectopic ACTH syndrome Arq Bras Endocrinol Metabol, 51 (8) (2007 Nov), pp. 1217-1225, 10.1590/s0004-27302007000800007 PMID: 18209859 View article View in ScopusGoogle Scholar [3] J. Young, M. Haissaguerre, O. Viera-Pinto, O. Chabre, E. Baudin, A. Tabarin Management of endocrine disease: cushing's syndrome due to ectopic ACTH secretion: an expert operational opinion Eur J Endocrinol, 182 (4) (2020 Apr), pp. R29-R58, 10.1530/EJE-19-0877 PMID: 31999619 View article View in ScopusGoogle Scholar [4] E. Varlamov, J.M. Hinojosa-Amaya, M. Stack, M. Fleseriu Diagnostic utility of Gallium-68-somatostatin receptor PET/CT in ectopic ACTH-secreting tumors: a systematic literature review and single-center clinical experience Pituitary, 22 (5) (2019 Oct), pp. 445-455, 10.1007/s11102-019-00972-w PMID: 31236798 View article View in ScopusGoogle Scholar [5] A.M. Isidori, E. Sbardella, M.C. Zatelli, M. Boschetti, G. Vitale, A. Colao, R. Pivonello, ABC Study Group Conventional and nuclear medicine imaging in ectopic cushing's syndrome: a systematic review J Clin Endocrinol Metab, 100 (9) (2015 Sep), pp. 3231-3244, 10.1210/JC.2015-1589 PMID: 26158607; PMCID: PMC4570166 View article View in ScopusGoogle Scholar [6] G. Cizza, L.K. Nieman, J.L. Doppman, M.D. Passaro, F.S. Czerwiec, G.P. Chrousos, G.B. Cutler Jr. Factitious cushing syndrome J Clin Endocrinol Metab, 81 (10) (1996 Oct), pp. 3573-3577, 10.1210/jcem.81.10.8855803 PMID: 8855803 View article View in ScopusGoogle Scholar [7] R. Patel, S. Sherf, N.B. Lai, R. Yu Exogenous cushing syndrome caused by a "herbal" supplement AACE Clin Case Rep, 8 (6) (2022 Aug 5), pp. 239-242, 10.1016/j.aace.2022.08.001 PMID: 36447831; PMCID: PMC9701910 View PDFView articleView in ScopusGoogle Scholar [8] C. Dunn, J. Amaya, P. Green A case of iatrogenic cushing's syndrome following use of an over-the-counter arthritis supplement 2023 Case Rep Endocrinol (2023 Mar 11), Article 4769258, 10.1155/2023/4769258 PMID: 36941974; PMCID: PMC10024620 View article View in ScopusGoogle Scholar [9] N. Mikhail, K. Kurator, E. Martey, A. Gaitonde, C. Cabrera, P. Balingit Iatrogenic cushing's syndrome caused by adulteration of a health product with dexamethasone Int J Endovascul Treatment Innovat Techn, 3 (1) (2022 Nov 23), pp. 6-9 Google Scholar [10] L. Del Carpio-Orantes, A.Q. Barrat-Hernández, A. Salas-González Iatrogenic Cushing syndrome due to fallacious herbal supplements. The case of ortiga ajo rey and Artri king Colegio de Medicina Interna de México, 37 (4) (2021), pp. 599-602 https://doi:10.24245/mim.v37i4.3912 Google Scholar [11] F. Wahab, R.A. Rahman, L.H. Yaacob, N.M. Noor, N. Draman A case report of steroid withdrawal syndrome Korean J Fam Med, 41 (5) (2020 Sep), pp. 359-362, 10.4082/kjfm.18.0181 Epub 2020 Sep 18. PMID: 32961047; PMCID: PMC7509117 View article View in ScopusGoogle Scholar [12] M. Zuluaga Quintero, A. Ramírez, A. Palacio, J.F. Botero, A. Clavijo Síndrome de Cushing exógeno e insuficiencia adrenal relacionada con consumo de producto natural Acta Méd Colomb, 42 (4) (2017), pp. 243-246, 10.36104/amc.2017.1006 View article Google Scholar [13] R. Patell, R. Dosi, S. Sheth, P. Jariwala Averting a crisis by 'add'ing up the clues 2014:bcr2014204685 BMJ Case Rep (2014 Jun 2), 10.1136/bcr-2014-204685 PMID: 24891489; PMCID: PMC4054156 View article Google Scholar [14] H. Hendarto Iatrogenic Cushing's syndrome caused by treatment with traditional herbal medicine, a case report 1st International Integrative Conference on Health, Life and Social Sciences (ICHLaS 2017) (2017 Dec), 10.2991/ichlas-17.2017.9 Atlantis Press View article Google Scholar [15] P.C. Oldenburg-Ligtenberg, M.M. van der Westerlaken A woman with Cushing's syndrome after use of an Indonesian herb: a case report Neth J Med, 65 (4) (2007 Apr), pp. 150-152 PMID: 17452765 View in ScopusGoogle Scholar [16] F. Azizi, A. Jahed, M. Hedayati, M. Lankarani, H.S. Bejestani, F. Esfahanian, N. Beyraghi, A. Noroozi, F. Kobarfard Outbreak of exogenous Cushing's syndrome due to unlicensed medications Clin Endocrinol, 69 (6) (2008 Dec), pp. 921-925, 10.1111/j.1365-2265.2008.03290.x Epub 2008 May 6. PMID: 18462262 View article View in ScopusGoogle Scholar [17] C. Martini, E. Zanchetta, M. Di Ruvo, A. Nalesso, M. Battocchio, E. Gentilin, E. Degli Uberti, R. Vettor, M.C. Zatelli Cushing in a leaf: endocrine disruption from a natural remedy J Clin Endocrinol Metab, 101 (8) (2016 Aug), pp. 3054-3060, 10.1210/jc.2016-1490 Epub 2016 May 24. PMID: 27218272 View article View in ScopusGoogle Scholar [18] A.J. Razenberg, J.W. Elte, A.P. Rietveld, H.C. van Zaanen, M.C. Cabezas A 'smart' type of Cushing's syndrome Eur J Endocrinol, 157 (6) (2007 Dec), pp. 779-781, 10.1530/EJE-07-0538 PMID: 18057386 View article View in ScopusGoogle Scholar [19] COFEPRIS (Federal Committee for Protection from Sanitary Risks) Public notification: COFEPRIS alerts about the illegal marketing of the product "ARTRI AJO KING", Which does not have a sanitary registration https://www.gob.mx/cofepris/articulos/cofepris-alerta-sobre-comercializacion-ilegal-del-producto-artri-ajo-king-el-cual-no-cuenta-con-registro-sanitario?idiom=es Google Scholar [20] Ministry of Health Malaysia Public notification: the truth about Maahun/Jamu http://www.myhealth.gov.my/en/the-truth-about-maajunjamu/ (2023) Google Scholar [21] INVIMA (National Food and Drug Surveillance Institute of Colombia) Health Alert: safety information about the product "ARTRIVID PLUS" promoted in different media of the country https://app.invima.gov.co/alertas/ckfinder/userfiles/files/ALERTAS%20SANITARIAS/medicamentos_pbiologicos/2015/Abril/ARTRIVID%20PLUS.pdf Google Scholar [22] FDA Public notification: Artri ajo king contains hidden drug ingredient https://www.fda.gov/drugs/medication-health-fraud/public-notification-artri-ajo-king-contains-hidden-drug-ingredient (2022) Google Scholar [23] FDA Public Notification: Artri King contains hidden drug ingredients https://www.fda.gov/drugs/medication-health-fraud/public-notification-artri-king-contains-hidden-drug-ingredients (2022) Google Scholar [24] FDA warns consumers not to purchase or use Artri and Ortiga products, which may contain hidden drug ingredients https://www.fda.gov/drugs/drug-safety-and-availability/fda-warns-consumers-not-purchase-or-use-artri-and-ortiga-products-which-may-contain-hidden-drug Google Scholar [25] P. Kempegowda, L. Quinn, L. Shepherd, S. Kauser, B. Johnson, A. Lawson, A. Bates Adrenal insufficiency from steroid-containing complementary therapy: importance of detailed history Endocrinol Diabetes Metab Case Rep, 2019 (1) (2019 Jul 26), pp. 1-4, 10.1530/EDM-19-0047 PMID: 31352697; PMCID: PMC6685090 View article Google Scholar [26] M.M. Pineyro, L. Redes, S. De Mattos, L. Sanchez, E. Brignardello, V. Bianchi, V. Ems, D. Centurión, M. Viola Factitious cushing's syndrome: a diagnosis to consider when evaluating hypercortisolism Front Endocrinol, 10 (2019 Mar 4), p. 129, 10.3389/fendo.2019.00129 PMID: 30886602; PMCID: PMC6409302 View article View in ScopusGoogle Scholar From https://www.sciencedirect.com/science/article/pii/S2214624523000199
    1 point
  35. Abstract Background As the population ages, the number of elderly patients with an indication for pituitary surgery is rising. Information on the outcome of patients aged over 75 is limited. This study reports a large series assessing the feasibility of surgical resection in this specific age range, focusing on surgical complications and postoperative results. Methods A retrospective cohort study of patients with pituitary adenomas and Rathke’s cleft cysts was conducted. All patients were aged 75 years or over and treated by a single expert neurosurgical team. A control population included 2379 younger adult patients operated by the same surgeons during the same period. Results Between 2008 and 2022, 155 patients underwent surgery. Indication was based on vision impairment in most patients (79%). Median follow-up was 13 months (range: 3–96). The first surgery was performed with an endoscopic transsellar approach, an extended endonasal transtuberculum approach and a microscopic transcranial approach in 96%, 3%, and 1% of patients, respectively. Single surgery was sufficient to obtain volume control in 97% of patients. From Kaplan-Meier estimates, 2-year and 5-year disease control with a single surgery were 97.3% and 86.2%, respectively. Resection higher than 80% was achieved in 77% of patients. No vision worsening occurred. In acromegaly and Cushing’s disease, endocrine remission was obtained in 90% of non-invasive adenomas. Surgical complications were noted in 5% of patients, with 30-day mortality, hematoma, cerebrospinal fluid leak, meningitis, and epistaxis occurring in 0.6%, 0.6%, 1.9%, 0.6%, and 1.3% respectively. New endocrine anterior deficits occurred in only 5%, while no persistent diabetes insipidus was noted. Compared with younger patients, the complication rate was not statistically different. Conclusions Surgery beyond the age of 75, mainly relying on an endoscopic endonasal transsellar approach, is effective and safe, provided that patients are managed in tertiary centers. This is a preview of subscription content, access via your institution. Abbreviations CSF: Cerebrospinal fluid ASA: American Society of Anesthesiologists Physical Status Classification System References Albano L, Losa M, Barzaghi LR, Niranjan A, Siddiqui Z, Flickinger JC, Lunsford LD, Mortini P (2021) Gamma Knife radiosurgery for pituitary tumors: a systematic review and meta-analysis. Cancers (Basel) 13(19):4998 Article PubMed Google Scholar Alexander TD, Chitguppi C, Collopy S et al (2022) Surgical outcomes of endoscopic transsphenoidal pituitary adenoma resection in elderly versus younger patients. J Neurol Surg B Skull Base 83(4):405–410 Article PubMed PubMed Central Google Scholar Baussart B, Declerck A, Gaillard S (2021) Mononostril endoscopic endonasal approach for pituitary surgery. Acta Neurochir (Wien) 163(3):655–659 Article PubMed Google Scholar Baussart B, Racy E, Gaillard S (2022) Double pedicled nasoseptal flap for skull base repair after endoscopic expanded endonasal approach. Acta Neurochir (Wien) 164(4):1111–1114 Article PubMed Google Scholar Baussart B, Venier A, Jouinot A, Reuter G, Gaillard S (2022) Closure strategy for endoscopic pituitary surgery: experience from 3015 patients. Front Oncol 12:1067312 Article PubMed Google Scholar Biamonte E, Betella N, Milani D, Lasio GB, Ariano S, Radice S, Lavezzi E, Mazziotti G, Lania A (2021) Impact of age on postsurgical outcomes of nonfunctioning pituitary adenomas. Endocrine 72(3):915–922 Article CAS PubMed Google Scholar Castinetti F, Nagai M, Dufour H, Kuhn J-M, Morange I, Jaquet P, Conte-Devolx B, Regis J, Brue T (2007) Gamma Knife radiosurgery is a successful adjunctive treatment in Cushing’s disease. Eur J Endocrinol 156(1):91–98 Article CAS PubMed Google Scholar Chalif EJ, Couldwell WT, Aghi MK (2022) Effect of facility volume on giant pituitary adenoma neurosurgical outcomes. J Neurosurg 14:1–10 Google Scholar Chen SH, Sprau A, Chieng L, Buttrick S, Alam ES, Ali SC, Madhavan K, Sargi ZB, Komotar R (2019) Transsphenoidal approach for pituitary adenomas in elderly patients. World Neurosurg 121:e670–e674 Article PubMed Google Scholar Ciric I, Ragin A, Baumgartner C, Pierce D (1997) Complications of transsphenoidal surgery: results of a national survey, review of the literature, and personal experience. Neurosurgery 40(2):225–236 (discussion 236-237) Article CAS PubMed Google Scholar Cossu G, Jouanneau E, Cavallo LM et al (2022) Surgical management of giant pituitary neuroendocrine tumors: meta-analysis and consensus statement on behalf of the EANS skull base section. Brain Spine 2:100878 Article PubMed PubMed Central Google Scholar Ding D, Mehta GU, Patibandla MR et al (2019) Stereotactic radiosurgery for acromegaly: an international multicenter retrospective cohort study. Neurosurg 84(3):717–725 Article Google Scholar Eichberg DG, Di L, Shah AH, Luther E, Richardson AM, Sarkiss CA, Ivan ME, Komotar RJ (2019) Brain tumor surgery is safe in octogenarians and nonagenarians: a single-surgeon 741 patient series. World Neurosurg 132:e185–e192 Article PubMed Google Scholar Engel JS, Tran J, Khalil N, Hladkowicz E, Lalu MM, Huang A, Wong CL, Hutton B, Dhesi JK, McIsaac DI (2023) A systematic review of perioperative clinical practice guidelines for care of older adults living with frailty. Br J Anaesth 130(3):262–271 Article PubMed Google Scholar Ezzat S, Asa SL, Couldwell WT, Barr CE, Dodge WE, Vance ML, McCutcheon IE (2004) The prevalence of pituitary adenomas: a systematic review. Cancer 101(3):613–619 Article PubMed Google Scholar Falk Erhag H, Guðnadóttir G, Alfredsson J, Cederholm T, Ekerstad N, Religa D, Nellgård B, Wilhelmson K (2023) The association between the clinical frailty scale and adverse health outcomes in older adults in acute clinical settings - a systematic review of the literature. Clin Interv Aging 18:249–261 Article PubMed PubMed Central Google Scholar Fleseriu M, Auchus R, Bancos I et al (2021) Consensus on diagnosis and management of Cushing’s disease: a guideline update. Lancet Diabetes Endocrinol 9(12):847–875 Article PubMed PubMed Central Google Scholar Frara S, Rodriguez-Carnero G, Formenti AM, Martinez-Olmos MA, Giustina A, Casanueva FF (2020) Pituitary tumors centers of excellence. Endocrinol Metab Clin North Am 49(3):553–564 Article PubMed Google Scholar Gaillard S (2014) The transition from microscopic to endoscopic transsphenoidal surgery in high-caseload neurosurgical centers: the experience of Foch Hospital. World Neurosurg 82(6 Suppl):S116-120 Article PubMed Google Scholar Giustina A, Chanson P, Bronstein MD et al (2010) A consensus on criteria for cure of acromegaly. J Clin Endocrinol Metab 95(7):3141–3148 Article CAS PubMed Google Scholar Gondim JA, Almeida JP, de Albuquerque LAF, Gomes E, Schops M, Mota JI (2015) Endoscopic endonasal transsphenoidal surgery in elderly patients with pituitary adenomas. J Neurosurg 123(1):31–38 Article PubMed Google Scholar Horvath B, Kloesel B, Todd MM, Cole DJ, Prielipp RC (2021) The evolution, current value, and future of the American Society of Anesthesiologists Physical Status Classification System. Anesthesiology 135(5):904–919 Article PubMed Google Scholar Kinoshita Y, Taguchi A, Tominaga A, Arita K, Yamasaki F (2021) Pseudocapsular resection in elderly patients with non-functioning pituitary adenoma. Clin Neurol Neurosurg 210:106997 Article PubMed Google Scholar Knosp E, Steiner E, Kitz K, Matula C (1993) Pituitary adenomas with invasion of the cavernous sinus space: a magnetic resonance imaging classification compared with surgical findings. Neurosurgery 33(4):610–617 (discussion 617-618) CAS PubMed Google Scholar Lee C-C, Sheehan JP (2016) Advances in Gamma Knife radiosurgery for pituitary tumors. Curr Opin Endocrinol Diabetes Obes 23(4):331–338 Article CAS PubMed Google Scholar Leiner T, Nemeth D, Hegyi P, Ocskay K, Virag M, Kiss S, Rottler M, Vajda M, Varadi A, Molnar Z (2022) Frailty and emergency surgery: results of a systematic review and meta-analysis. Front Med (Lausanne) 9:811524 Article PubMed Google Scholar Lin H-S, Watts JN, Peel NM, Hubbard RE (2016) Frailty and post-operative outcomes in older surgical patients: a systematic review. BMC Geriatr 16(1):157 Article PubMed PubMed Central Google Scholar Lu VM, Ravindran K, Perry A, Graffeo CS, Dawood HY, Van Gompel JJ, Mekary RA, Smith TR (2020) Recurrence of Rathke’s cleft cysts based on gross total resection of cyst wall: a meta-analysis. Neurosurg Rev 43(3):957–966 Article PubMed Google Scholar Maiuri F, Corvino S, Lorenzetti M, Franca RA, Esposito F, Caro Del Basso De M (2023) Intracranial meningiomas in patients aged ≥80 years: pathological features and surgical problems. World Neurosurg 173:e498–e508 Article PubMed Google Scholar Marcus HJ, Khan DZ, Borg A et al (2021) Pituitary society expert Delphi consensus: operative workflow in endoscopic transsphenoidal pituitary adenoma resection. Pituitary 24(6):839–853 Article PubMed PubMed Central Google Scholar Melmed S (ed) (2017) The pituitary, 4th edn. Elsevier/Academic Press, London, United Kingdom, San Diego Google Scholar Melmed S, Kaiser UB, Lopes MB et al (2022) Clinical biology of the pituitary adenoma. Endocr Rev 43(6):1003–1037 Article PubMed PubMed Central Google Scholar Memel Z, Chesney K, Pangal DJ, Bonney PA, Carmichael JD, Zada G (2019) Outcomes following transsphenoidal pituitary surgery in the elderly: a retrospective single-center review. Oper Neurosurg (Hagerstown) 16(3):302–309 Article PubMed Google Scholar Messerer M, De Battista JC, Raverot G, Kassis S, Dubourg J, Lapras V, Trouillas J, Perrin G, Jouanneau E (2011) Evidence of improved surgical outcome following endoscopy for nonfunctioning pituitary adenoma removal. Neurosurg Focus 30(4):E11 Article PubMed Google Scholar Micko A, Oberndorfer J, Weninger WJ, Vila G, Höftberger R, Wolfsberger S, Knosp E (2019) Challenging Knosp high-grade pituitary adenomas. J Neurosurg 132(6):1739–1746 Article PubMed Google Scholar Minniti G, Esposito V, Piccirilli M, Fratticci A, Santoro A, Jaffrain-Rea M-L (2005) Diagnosis and management of pituitary tumours in the elderly: a review based on personal experience and evidence of literature. Eur J Endocrinol 153(6):723–735 Article CAS PubMed Google Scholar Minniti G, Filippi AR, Osti MF, Ricardi U (2017) Radiation therapy for older patients with brain tumors. Radiat Oncol 12(1):101 Article PubMed PubMed Central Google Scholar Minniti G, Scaringi C, Poggi M, Jaffrain Rea ML, Trillò G, Esposito V, Bozzao A, Enrici MM, Toscano V, Enrici RM (2015) Fractionated stereotactic radiotherapy for large and invasive non-functioning pituitary adenomas: long-term clinical outcomes and volumetric MRI assessment of tumor response. Eur J Endocrinol 172(4):433–441 Article CAS PubMed Google Scholar Mortini P, Nocera G, Roncelli F, Losa M, Formenti AM, Giustina A (2020) The optimal numerosity of the referral population of pituitary tumors centers of excellence (PTCOE): a surgical perspective. Rev Endocr Metab Disord 21(4):527–536 Article PubMed Google Scholar Nidadavolu LS, Ehrlich AL, Sieber FE, Oh ES (2020) Preoperative evaluation of the frail patient. Anesth Analg 130(6):1493–1503 Article PubMed PubMed Central Google Scholar Ostrom QT, Cioffi G, Gittleman H, Patil N, Waite K, Kruchko C, Barnholtz-Sloan JS (2019) CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2012–2016. Neuro Oncol 21(Suppl 5):v1–v100 Article PubMed PubMed Central Google Scholar Pereira MP, Oh T, Joshi RS et al (2020) Clinical characteristics and outcomes in elderly patients undergoing transsphenoidal surgery for nonfunctioning pituitary adenoma. Neurosurg Focus 49(4):E19 Article PubMed Google Scholar Quah BL, Edwards-Bailey A, Gnanalingham K, Pathmanaban O, Vasilopoulos H, Roncaroli F, Kearney T, Balogun J, Karabatsou K (2022) Endoscopic transsphenoidal surgery for biochemically and clinically non-functioning adenohypophyseal tumours in the elderly: experience from a single UK centre. Endocrine 75(3):872–882 Article CAS PubMed Google Scholar Robenshtok E, Benbassat CA, Hirsch D, Tzvetov G, Cohen ZR, Iraqi HM, Gorshtein A, Toledano Y, Shimon I (2014) Clinical course and outcome of nonfunctioning pituitary adenomas in the elderly compared with younger age groups. Endocr Pract 20(2):159–164 Article PubMed Google Scholar Saleh A, Thirukumaran C, Mesfin A, Molinari RW (2017) Complications and readmission after lumbar spine surgery in elderly patients: an analysis of 2,320 patients. Spine J 17(8):1106–1112 Article PubMed Google Scholar Sheehan JM, Douds GL, Hill K, Farace E (2008) Transsphenoidal surgery for pituitary adenoma in elderly patients. Acta Neurochir (Wien) 150(6):571–574 (discussion 574) Article CAS PubMed Google Scholar Sheehan JP, Starke RM, Mathieu D et al (2013) Gamma Knife radiosurgery for the management of nonfunctioning pituitary adenomas: a multicenter study: clinical article. JNS 119(2):446–456 Article Google Scholar Sherlock M, Ayuk J, Tomlinson JW, Toogood AA, Aragon-Alonso A, Sheppard MC, Bates AS, Stewart PM (2010) Mortality in patients with pituitary disease. Endocr Rev 31(3):301–342 Article PubMed Google Scholar Spina A, Losa M, Mortini P (2019) Pituitary adenomas in elderly patients: clinical and surgical outcome analysis in a large series. Endocrine 65(3):637–645 Article CAS PubMed Google Scholar Tardivo V, Penner F, Garbossa D, Di Perna G, Pacca P, Salvati L, Altieri R, Grottoli S, Zenga F (2020) Surgical management of pituitary adenomas: does age matter? Pituitary 23(2):92–102 Article PubMed Google Scholar Thakur JD, Corlin A, Mallari RJ et al (2021) Pituitary adenomas in older adults (≥ 65 years): 90-day outcomes and readmissions: a 10-year endoscopic endonasal surgical experience. Pituitary 24(1):14–26 Article CAS PubMed Google Scholar Trouillas J, Jaffrain-Rea M-L, Vasiljevic A, Raverot G, Roncaroli F, Villa C (2020) How to classify the pituitary neuroendocrine tumors (PitNET)s in 2020. Cancers (Basel) 12(2):E514 Article Google Scholar Tuleasca C, Ducos Y, Leroy H-A, Chanson P, Knafo S, Levivier M, Aghakhani N, Parker F (2020) Transsphenoidal resection for pituitary adenoma in elderly versus younger patients: a systematic review and meta-analysis. Acta Neurochir (Wien) 162(6):1297–1308 Article PubMed Google Scholar Villa C, Vasiljevic A, Jaffrain-Rea ML et al (2019) A standardised diagnostic approach to pituitary neuroendocrine tumours (PitNETs): a European Pituitary Pathology Group (EPPG) proposal. Virchows Arch 475(6):687–692 Article CAS PubMed Google Scholar Wilson PJ, Omay SB, Kacker A, Anand VK, Schwartz TH (2018) Endonasal endoscopic pituitary surgery in the elderly. J Neurosurg 128(2):429–436 Article PubMed Google Scholar Yunoue S, Tokimura H, Tominaga A et al (2014) Transsphenoidal surgical treatment of pituitary adenomas in patients aged 80 years or older. Neurosurg Rev 37(2):269–276 (discussion 276-277) Article PubMed Google Scholar Zhan R, Ma Z, Wang D, Li X (2015) Pure endoscopic endonasal transsphenoidal approach for nonfunctioning pituitary adenomas in the elderly: surgical outcomes and complications in 158 patients. World Neurosurg 84(6):1572–1578 Article PubMed Google Scholar Zietlow KE, Wong S, Heflin MT, McDonald SR, Sickeler R, Devinney M, Blitz J, Lagoo-Deenadayalan S, Berger M (2022) Geriatric preoperative optimization: a review. Am J Med 135(1):39–48 Article PubMed Google Scholar Download references Author information Authors and Affiliations Department of Neurosurgery, La Pitié-Salpêtrière University Hospital, Assistance Publique-Hôpitaux de Paris, 47-83 Boulevard de L’Hôpital, 75013, Paris, France Marta Garvayo, Vincent Reina, Stephan Gaillard & Bertrand Baussart Department of Neurosurgery, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland Marta Garvayo & Mahmoud Messerer Université Paris Cité, CNRS, INSERM, Institut Cochin, 75014, Paris, France Chiara Villa, Anne Jouinot, Jérôme Bertherat, Guillaume Assié & Bertrand Baussart Department of Neuropathology, La Pitié-Salpêtière University Hospital, AP-HP, Sorbonne University, Paris, France Chiara Villa Department of Endocrinology, Assistance Publique-Hôpitaux de Paris, Hôpital Ambroise Paré, Boulogne Billancourt, France Mirella Hage & Marie-Laure Raffin-Sanson Université de Versailles Saint-Quentin-en-Yvelines UFR Des Sciences de La Santé Simone Veil, Montigny-Le-Bretonneux, France Mirella Hage & Marie-Laure Raffin-Sanson Department of Endocrinology and Reproductive Medicine, Centre de Référence Des Maladies Endocriniennes Rares de La Croissance Et du Développement, CRMERC, Endo-ERN, Pitié-Salpêtrière Hospital, AP-HP, Sorbonne University, Paris, France Carine Courtillot & Anne Bachelot Université Paris-Saclay, Inserm, Physiologie Et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d’Endocrinologie Et Des Maladies de La Reproduction, Centre de Référence des Maladies Rares de L’Hypophyse, Le Kremlin-Bicêtre, France Peter Kamenicky & Philippe Chanson Sorbonne University, Endocrine Unit, Reproductive Medicine, Centre de Référence Des Maladies Endocriniennes Rares de La Croissance Et du Développement (CRMERC), Endo-ERN (Id 739527), Saint-Antoine Hospital, AP-HP, Paris, France Camille Vatier & Sophie Christin-Maitre Inserm UMRS938, Saint-Antoine Research Center, Sorbonne University, 75012, Paris, France Camille Vatier INSERM UMR-833, Trousseau Hospital, Paris, France Sophie Christin-Maitre Department of Endocrinology, Center of Rare Adrenal Diseases, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France Jérôme Bertherat & Guillaume Assié Corresponding author Correspondence to Bertrand Baussart. Ethics declarations Conflict of interest The authors declare no competing interests. Additional information Publisher's Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Rights and permissions Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law. Reprints and Permissions From https://link.springer.com/article/10.1007/s00701-023-05809-x
    1 point
  36. Abstract Introduction Laparoscopic adrenalectomy is the standard treatment for adrenal tumors caused by Cushing's syndrome. However, few pregnant women have undergone adrenalectomy because of the risk of general anesthesia and surgery. Case presentation A 28-year-old woman presented with gradually worsening Cushing's signs at around 12 weeks of pregnancy. Magnetic resonance imaging displayed a 38-mm left adrenal tumor, which was the cause of the adrenal Cushing's syndrome. Metyrapone was started, which increased androgen levels. Since the management of Cushing's syndrome by medication alone is challenging, unilateral laparoscopic adrenalectomy by a retroperitoneal approach was performed at 23 weeks of the pregnancy. No perioperative complications were noted. Conclusion Adrenalectomy is considered safe in pregnant women with Cushing's syndrome. Laparoscopic adrenalectomy by retroperitoneal approach should be chosen and performed between 14 and 30 weeks of pregnancy to prevent mother and fetal complications. Abbreviations & Acronyms CS Cushing's syndrome MRI magnetic resonance imaging Keynote message We report a rare case of adrenalectomy performed via a retroperitoneal approach for Cushing's syndrome in a pregnant woman. Cushing's syndrome may affect the fetus, and surgery can be considered in addition to medical management. Adrenalectomy should be performed in the second trimester of pregnancy. Pneumoperitoneal pressure, position, and surgical approaches must receive careful attention. Introduction CS is characterized by excessive cortisol secretion and characteristic symptoms such as full moon-like facial features and central obesity. Premenopausal women with CS rarely become pregnant because excessive glucocorticoid secretion inhibits the synthesis of gonadotropins, leading to impaired ovarian and endometrial function, and causing amenorrhea or oligomenorrhea.1 Furthermore, even when women with CS become pregnant, the incidence of severe complications is high. CS can cause maternal hypertension, diabetes/glucose intolerance, osteopenia/osteoporosis, preeclampsia, pulmonary edema, heart failure, opportunistic infections, and even death. Additionally, CS can potentially cause stillbirth, prematurity, and intrauterine fetal growth restriction.1-6 Therefore, CS must be detected at an early stage in pregnancy; however, CS may go undetected because of the overlapping signs of preeclampsia and/or gestational diabetes. A cortisol-secreting adrenal tumor is the underlying cause of CS, and laparoscopic adrenalectomy is the standard treatment to it. Medical treatment of CS can include medications that inhibit 11β-hydroxylase, such as metyrapone and osilodrostat, but surgical treatment is considered if the disease is difficult to control with medical treatment. Nonobstetric surgery during pregnancy is performed in 1%–2% of pregnant women.7 Although general anesthesia is relatively safe during pregnancy, the indication for the surgery must be carefully considered because of potential risks such as neurodevelopmental delay, sudden death, etc. Herein, we present a case of a pregnant woman diagnosed with CS who underwent unilateral laparoscopic adrenalectomy by a retroperitoneal approach without any problems. Case presentation The patient was a 28-year-old primiparous woman. Since around 12 weeks of pregnancy, she has experienced facial and lower limb edema; gained 6-kg weight in 1 month; increased facial acne; and experienced subcutaneous bleeding on the forearms, red abdominal dermatitis, proximal muscle weakness, palpitations, insomnia, and decreased vision in eyes. Her symptoms gradually worsened from 14 weeks, and she was referred to our hospital to clarify the cause at 18 weeks of pregnancy. Adrenal CS was suspected on the basis of her Cushing's signs, cortisol 25 μg/dL, and adrenocorticotropic hormone <1.5 pg/mL. She had hypokalemia, hypogammaglobulinemia, and liver dysfunction, and her condition was rapidly worsening. Given her pregnant state, she was admitted for intensive testing for the case of CS from 19 weeks of pregnancy. MRI revealed a well-defined 38-mm left adrenal tumor, which was the cause of the adrenal CS (Fig. 1). She was started on metyrapone with 250 mg per day, which increased androgens (0.53–0.69 ng/mL in 1 week). We considered that the management of CS by medication alone would be challenging and performed adrenalectomy during her pregnancy. The dose of metyrapone was increased to 1000 mg per day eventually. Fig. 1 Open in figure viewerPowerPoint Magnetic resonance imaging on admission shows a left adrenal tumor with a long axis of 38 mm (arrowhead). Signal reduction was partially observed on opposed-phase images, leading to diagnosis of cortical adenoma. She was admitted to the hospital at 23 weeks and 2 days of gestation, and laparoscopic left adrenalectomy was performed via a retroperitoneal approach in the right lateral and jackknife position on the following day (Fig. S1). During the surgery, blood pressure was carefully controlled by an anesthesiologist and the patient's position and fetal heart rate were monitored by an obstetrician. The operation time, insufflation time, and general anesthesia time were 68, 59, and 123 min, respectively, and the blood loss volume was 75 mL, without any complications. Pathological findings revealed an adrenocortical adenoma. The specimen was positive for one of the nine Weiss criteria (Fig. 2). Fig. 2 Open in figure viewerPowerPoint (a) Intraoperative findings of the retroperitoneal approach. Arrowheads indicate the tumor. (b) Gross appearance of the resected adrenal tumor; a brownish-toned, substantial mass, 60 × 34 × 15 mm in size. (c, d) Hematoxylin–eosin staining showed that nodular lesion with a fibrous capsule, with foci of homogeneous cells with eosinophilic or pale, foamy sporangia and small round nuclei. Postoperatively, metyrapone was discontinued and both lower leg edema, facial acne, fatigue, and muscle weakness improved. Metyrapone was discontinued after surgery. Hydrocortisone, which had been administered at 150 mg/day during the perioperative period, was reduced every few weeks and was taken at 30 mg/day at delivery. She delivered by cesarean section at 38 weeks and 2 days of gestation, with good outcomes for the mother and her infant. Hydrocortisone was discontinued 15 weeks after delivery. We showed the changes in cortisol and ACTH from the first visit to postpartum (Fig. 3). Fig. 3 Open in figure viewerPowerPoint The transition of Cortisol and ACTH. Cortisol decreases rapidly after surgery and rises again before delivery. As cortisol improved, ACTH also increased. Discussion CS seldom occurs during pregnancy. Symptoms such as weight gain, skin striae, fatigue, and a round face can also occur in normal pregnancies. The dexamethasone suppression test can result in false positives because of ACTH produced by placenta in normal pregnancy. During pregnancy, there is a physiological state of high cortisol levels. The disappearance of diurnal rhythm is a useful indicator for diagnosis of CS in pregnancy because circadian rhythm is maintained in normal pregnancy. Useful diagnostic criteria include urine cortisol levels greater than three times the upper limit of normal, loss of diurnal cortisol rhythm, and presence of adrenal tumors on MRI. The pharmacologic treatment of endogenous cortisol is complex, and hormonal management is challenging. While the management of the cortisol levels is important, metyrapone is a risk factor for gestational hypertension and may inhibit fetal cortisol production by crossing the placenta.1-6, 8-12 In this case, because androgens were also elevated and drug management was expected to be challenging, the surgery was aggressively considered. Despite the reports of successful adrenalectomy is after 28 weeks of gestation,6, 13, 14 The surgery should be performed by an experienced team between 14 and 30 weeks of pregnancy, that is, after organogenesis phase and before the fetus grows too large.1, 13, 15 A few pregnant women with adrenal CS undergo adrenalectomy. However, the laparoscopic approach is safe, and maternal and fetal complications were higher in women who did not undergo surgery.16 Less postoperative pain, faster wound healing, and faster postoperative recovery are the main advantages of laparoscopic surgery.17 In pregnant women, pneumoperitoneal pressure should be kept <12 mmHg because increased intraabdominal pressure decreases placental blood flow and can cause fetal acidosis due to the absorption of carbon dioxide used for insufflation. Laparoscopic adrenalectomy can be safely performed through both transperitoneal and retroperitoneal approaches.18 However, in pregnant women, performing the surgery by the retroperitoneal approach in the lateral position is preferable to prevent putting pressure on the fetus during the surgery. The retroperitoneal approach is advantageous, as less pressure is placed on the uterus and adhesions are prevented. After taking the lateral position, the obstetrician is advised to check the position and confirm that the abdomen is not compressed and that the fetal heart rate is normal. Conclusions We present a case of a pregnant woman diagnosed with adrenal CS who underwent a unilateral laparoscopic adrenalectomy by a retroperitoneal approach without any problems. Adrenalectomy is a useful treatment when CS is difficult to control despite metyrapone and other medical support. Author contributions Nobuyoshi Takeuchi: Conceptualization; methodology; project administration; writing – original draft. Yusuke Imamura: Conceptualization; methodology; supervision; writing – review and editing. Kazuki Ishiwata: Data curation; supervision. Manato Kanesaka: Data curation; supervision. Yusuke Goto: Data curation; supervision. Tomokazu Sazuka: Data curation; supervision. Sawako Suzuki: Data curation; supervision. Hisashi Koide: Data curation; supervision. Shinichi Sakamoto: Data curation; supervision. Tomohiko Ichikawa: Data curation; supervision. Conflict of interest The authors declare no conflicts of interest. Approval of the research protocol by an Institutional Reviewer Board Not applicable. Informed consent Informed consent for the release of the case report and accompanying images has been obtained from the patient. Registry and the Registration No. of the study/trial Not applicable. From https://onlinelibrary.wiley.com/doi/10.1002/iju5.12637
    1 point
  37. Introduction to Endocrinology Endocrinology is a medical specialty that focuses on the diagnosis and treatment of diseases related to hormones. Endocrinologists are experts in managing and treating diseases related to the endocrine system, which includes the thyroid, pituitary, adrenal glands, and pancreas. Endocrinologists are trained to diagnose and treat conditions such as diabetes, thyroid disorders, pituitary disorders, and other conditions related to hormones. Endocrinologists also specialize in reproductive health and fertility issues, including PCOS. Endocrinology is a complex field that requires a deep understanding of the endocrine system and its role in regulating the body’s hormones. Endocrinologists must be able to interpret laboratory tests and understand the underlying causes of endocrine disorders. They must also be able to develop individualized treatment plans to address the specific needs of each patient. Diagnosing PCOS and Diabetes Endocrinologists are experts in diagnosing and managing PCOS and diabetes. PCOS is a hormonal disorder that affects the ovaries, and it is characterized by irregular menstrual cycles, excess facial and body hair, and infertility. To diagnose PCOS, an endocrinologist will perform a physical exam and order laboratory tests to measure hormone levels. The endocrinologist will also ask the patient about her symptoms and family history to determine if PCOS is the cause. Diabetes is a chronic condition that affects the body’s ability to process sugar. To diagnose diabetes, an endocrinologist will perform a physical exam and order laboratory tests to measure blood sugar levels. The endocrinologist may also order imaging tests to check for signs of diabetes-related complications. Treating PCOS and Diabetes Once the endocrinologist has diagnosed PCOS or diabetes, they will develop an individualized treatment plan to address the patient’s specific needs. For PCOS, the endocrinologist may recommend lifestyle changes such as weight loss, exercise, and dietary changes to help manage symptoms. The endocrinologist may also prescribe medications to regulate hormone levels and improve fertility. For diabetes, the endocrinologist may recommend lifestyle changes such as weight loss, exercise, and dietary changes to help manage blood sugar levels. The endocrinologist may also prescribe medications to help regulate blood sugar levels. In addition, the endocrinologist may recommend regular check-ups to monitor the patient’s progress and to adjust the treatment plan if needed. Conclusion Endocrinology plays an important role in managing PCOS and diabetes. Endocrinologists are experts in diagnosing and treating these conditions, and they are trained to develop individualized treatment plans that address the specific needs of each patient. By working with an endocrinologist, patients can get the help they need to manage their PCOS or diabetes and achieve their health goals. Endocrinology is a complex field that requires a deep understanding of the endocrine system and its role in regulating the body’s hormones. An endocrinologist can help patients with PCOS and diabetes manage their conditions and achieve their health goals. By working with an endocrinologist, patients can get the help they need to manage their PCOS or diabetes and achieve their health goals. From https://www.diabetesincontrol.com/the-role-of-endocrinology-in-managing-polycystic-ovary-syndrome-and-diabetes/
    1 point
  38. Key takeaways: Cushing’s syndrome symptoms moderately impact quality of life for adults with the condition. Weight gain, muscle fatigue and menstrual changes decline in severity from diagnosis to follow-up. Adults with endogenous Cushing’s syndrome reported that the condition moderately affects their quality of life and causes them to have symptoms about 16 days in a given month, according to findings published in Pituitary. “Our study aimed to evaluate the ongoing burden of Cushing’s syndrome in order to identify areas of unmet need,” Eliza B. Geer, MD, medical director of the Multidisciplinary Pituitary and Skull Base Tumor Center and associate attending of endocrinology and neurosurgery at Memorial Sloan Kettering Cancer Center, told Healio. “We found that patients with treated Cushing’s continue to experience ongoing symptoms more than half of the days in a given month, miss about 25 workdays per year and need twice the average number of outpatient visits per year, indicating a significant impact on daily function and work productivity. Some of these symptoms, like fatigue and pain, have not been well studied in Cushing’s patients, and need more attention.” Geer and colleagues administered a cross-sectional survey to 55 adults aged 21 years and older who had been diagnosed with Cushing’s syndrome at least 6 months before the survey and were receiving at least one pharmacologic therapy for their disease (85% women; mean age, 43.4 years). The survey was conducted online from June to August 2021. Five patient-reported outcome scales were included. The CushingQoL was used to analyze quality of life, a visual analog scale was included to assess pain, the Brief Fatigue Inventory was used to measure fatigue, the Sleep Disturbance v1.0 scale assessed perceptions of sleep and the PROMIS Short Form Anxiety v1.0-8a scale was used to measure fear, anxious misery, hyperarousal and somatic symptoms related to arousal. Participants self-reported the impact of Cushing’s syndrome on daily life and their physician’s level of awareness of Cushing’s syndrome. Some symptoms decline in severity over time Of the study group, 81% had pituitary or adrenal tumors, and 20% had ectopic adrenocorticotropic hormone-producing tumors; 80% of participants underwent surgery to treat their Cushing’s syndrome. The frequency of reported symptoms did not change from Cushing’s syndrome diagnosis to the time of the survey. The most frequently reported symptoms were weight gain, muscle fatigue and weakness and anxiety. Participants reported a decline in symptom severity for weight gain, muscle fatigue and weakness and menstrual changes from diagnosis to the survey. Though symptom severity declined, none of the three symptoms were entirely eliminated. Adults did not report declines in severity for other symptoms. Hirsutism and anxiety were reported by few participants, but were consistently scored high in severity among those who reported it. There were no changes in patient satisfaction with medications from their first appointment to the time of the survey. “It was surprising that anxiety and pain did not improve with treatment,” Geer said. “A quarter of patients at baseline reported anxiety and this percentage was exactly the same after treatment. Same for pain — nearly a quarter of patients reported pain despite treatment. While the presence of anxiety has been well-documented in Cushing’s patients, pain has not, and needs further study.” Nearly half of primary care providers unable to diagnose Cushing’s syndrome All participants reported having at least one challenge with being diagnosed with Cushing’s syndrome. Of the respondents, 49% said their primary care provider was unable to diagnose their Cushing’s syndrome and 33% initially received the wrong diagnosis. Physicians referred 49% of participants to a specialist, and 39% of adults said their doctor lacked knowledge or understanding of their condition. The study group had a moderate level of quality of life impairment as assessed through the CushingQoL scale. The mean pain score was 3.6 of a possible 10, indicating low levels of pain. Moderate to severe levels of fatigue were reported by 69% of participants. Self-reported sleep and anxiety scores were similar to what is observed in the general population. Participants said sexual activity, self-confidence and life satisfaction were most impacted by a Cushing’s syndrome diagnosis. Adults experienced symptoms a mean 16 days in a typical month and saw their outpatient physician an average of six times per year. Those who were employed said they miss 2 days of work per month, or about 25 days per year, due to Cushing’s syndrome. “Longitudinal assessment of clinically relevant patient-reported outcomes based on validated measures and coupled with biochemical and treatment data is needed in a large cohort of Cushing’s patients,” Geer said. “This will allow us to identify clinically meaningful changes in symptom burden within each patient, as well as predictors of outcomes — which patients improve on which symptoms, and which patients do not feel better despite biochemical normalization. We need to improve our ability to help our patients feel better, not just achieve normal cortisol levels.” For more information: Eliza B. Geer, MD, can be reached at geere@mskcc.org. From https://www.healio.com/news/endocrinology/20230830/adults-with-cushings-syndrome-report-high-burden-of-illness-despite-ongoing-treatment
    1 point
  39. Topics to be discussed include: New articles showing patients prefer desiccated thyroid Moving away from a TSH-centered approach New thyroid hormone preparations, including Adthyza •Does biotin affect thyroid tests? •Hypothyroidism diet? What is the difference between desiccated thyroid and synthetic thyroid hormones? Is rT3 important? Read more at https://cushings.invisionzone.com/topic/57621-webinar-dr-theodore-friedman-the-wiz-will-giving-a-webinar-on-updates-on-treating-hypothyroidism/
    1 point
  40. YOU’RE INVITED! A4M/Metabolic Medical Institute (MMI) Webinar on Updates on Treating Hypothyroidism Dr. Theodore Friedman (The Wiz) will giving a webinar on Updates on Treating Hypothyroidism Topics to be discussed include: New articles showing patients prefer desiccated thyroid Moving away from a TSH-centered approach New thyroid hormone preparations, including Adthyza •Does biotin affect thyroid tests? •Hypothyroidism diet? What is the difference between desiccated thyroid and synthetic thyroid hormones? Is rT3 important? Wednesday • August 2, 2023 • 4 PM PDT register at https://us02web.zoom.us/webinar/register/WN_kRyqZFlrSgKj54CuK7OQqQ Slides will be available on the day of the talk here. There will be plenty of time for questions using the chat button. For more information, email us at mail@goodhormonehealth.com
    1 point
  41. The most common procedure to remove pituitary tumors is transsphenoidal adenomectomy. It allows the removal of the tumor with minimal damage to the surrounding structures. The surgical team accesses the pituitary gland through the sphenoid sinus — a hollow space behind the nasal passages and below the pituitary gland. If performed in specialized centers and by an experienced pituitary surgeon, this type of surgery is reported to result in an overall cure rate, or full remission, of Cushing’s disease for 80% to 90% of patients. A higher success rate is seen with smaller tumors. However, reported remission rates vary considerably, mainly due to differences in the criteria used to define disease remission. In some cases, a second transsphenoidal adenomectomy is required to fully remove tumor tissue; in others, the initial surgical procedure is paired with a second form of treatment, such as radiation therapy or certain medications. Given the complexity of the procedure, the guidelines recommend patients undergo surgery in specialized Pituitary Tumor Centers of Excellence. Patients also are advised to have the surgery performed by an experienced pituitary neurosurgeon. Follow-up for all patients should be conducted by a multidisciplinary team, including a pituitary endocrinologist. Lifelong monitoring for disease recurrence is required.
    1 point
  42. Abstract Purpose. Few related factors of low bone mass in Cushing’s disease (CD) have been identified so far, and relevant sufficient powered studies in CD patients are rare. On account of the scarcity of data, we performed a well-powered study to identify related factors associated with low bone mass in young CD patients. Methods. This retrospective study included 153 CD patients (33 males and 120 females, under the age of 50 for men and premenopausal women). Bone mineral density (BMD) of the left hip and lumbar spine was measured by dual energy X-ray absorptiometry (DEXA). In this study, low bone mass was defined when the Z score was −2.0 or lower. Results. Among those CD patients, low bone mass occurred in 74 patients (48.37%). Compared to patients with normal BMD, those patients with low bone mass had a higher level of serum cortisol at midnight (22.31 (17.95-29.62) vs. 17.80 (13.75-22.77), ), testosterone in women (2.10 (1.33–2.89) vs. 1.54 (0.97–2.05), ), higher portion of male (32.43% vs. 11.54%, ) as well as hypertension (76.12% vs. 51.67%, ), and lower IGF-1 index (0.59 (0.43–0.76) vs. 0.79 (0.60–1.02), ). The Z score was positively associated with the IGF-1 index in both the lumbar spine (r = 0.35153, ) and the femoral neck (r = 0.24418, ). The Z score in the femoral neck was negatively associated with osteocalcin (r = −0.22744, ). Compared to the lowest tertile of the IGF-1 index (<0.5563), the patients with the highest tertile of the IGF-1 index (≥0.7993) had a lower prevalence of low bone mass (95% CI 0.02 (0.001–0.50), ), even after adjusting for confounders such as age, gender, duration, BMI, hypertension, serum cortisol at midnight, PTH, and osteocalcin. Conclusions. The higher IGF-1 index was independently associated with lower prevalence of low bone mass in young CD patients, and IGF-1 might play an important role in the pathogenesis of CD-caused low bone mass. 1. Introduction Cushing’s disease (CD), caused by an adrenocorticotropic hormone (ACTH)-secreting pituitary tumor, is a rare disease with approximately 1.2 to 2.4 new cases per million people each year [1]. Osteoporosis has been recognized as a serious consequence of endogenous hypercortisolism since the first description in 1932 [2]. The prevalence of osteoporosis is around 38–50%, and the rate of atraumatic compression fractures is 15.8% in CD patients [3]. After cortisol normalization and appropriate treatment, recovery of the bone impairment occurs slowly (6–9 years) and partially [4, 5]. Hypercortisolemia impairs bone quality through multiple mechanisms [6]. Growth hormone (GH) and insulin-like growth factor 1 (IGF-1) play a crucial role in bone growth and development [7]. IGF-1 is considered essential for the longitudinal growth of bone, skeletal maturity, and bone mass acquisition not only during growth but also in the maintenance of bone in adults [8]. Previous research studies revealed that low serum IGF-1 levels were associated with a 40% increased risk of fractures [9, 10], and serum IGF-1 levels could be clinically useful for evaluating the risk of spinal fractures [11]. In Marl Hotta’s research, extremely low or no response of plasma GH to recombinant human growth hormone (hGRH) injection was noted in CD patients. This result suggested that the diminished hGRH-induced GH secretion in patients with Cushing’s syndrome might be caused by the prolonged period of hypercortisolemia [12]. Other surveys indicated that glucocorticoids, suppressing GH–IGF-1 and the hypothalamic-pituitary-gonadal axes, lead to decreased number and dysfunction of osteoblast [13]. However, the exact mechanism is still unclear, and few risk factors for osteoporosis in CD have been identified so far. Until now, relevant and sufficiently powered studies in CD patients have been rare [14, 15]. Early recognition of the changes in bone mass in CD patients contributes to early diagnosis of bone mass loss and prompt treatment, which could help minimize the incidence of adverse events such as fractures. On account of the scarcity of data and pressing open questions concerning risk evaluation and management of osteoporosis, we performed a well-powered study to identify the related factors associated with low bone mass in young CD patients at the time of diagnosis. 2. Materials and Methods 2.1. Subjects This retrospective study enrolled 153 CD patients (33 males and 120 females) from the Department of Endocrinology and Metabolism of Huashan Hospital between January 2010 and February 2021. All subjects were evaluated by the same group of endocrinologists for detailed clinical evaluation. This study, which was in complete adherence to the Declaration of Helsinki, was approved by the Human Investigation Ethics Committee at Huashan Hospital, Fudan University (No. 2017M011). We collected data on demographic characteristics, laboratory tests, and bone mineral density. Inclusion criteria included the following: (1) willingness to participate in the study; (2) premenopausal women ≥18 years old, men ≥18 years old but younger than 50 years old, and young women (<50 years old) with menstrual abnormalities who were associated with CD after excluding menstrual abnormalities caused by other causes; (3) diagnosis of CD according to the updated diagnostic criteria [16]; and (4) pathological confirmation after transsphenoidal surgery (positive immunochemistry staining with ACTH). Exclusion criteria included Cushing’s syndrome other than pituitary origin. 2.2. Clinical and Biochemical Methods IGF-1 was measured using the Immulite 2000 enzyme-labeled chemiluminescent assay (Siemens Healthcare Diagnostic, Surrey, UK). Other endocrine hormones, including cortisol (F), 24-hour urinary free cortisol (24hUFC), adrenocorticotropic hormone (ACTH), prolactin (PRL), luteinizing hormone (LH), follicle stimulating hormone (FSH), estrogen (E2), progesterone (P), testosterone (T), thyroid stimulating hormone (TSH), and free thyroxine (FT4), were carried out by the chemiluminescence assay (Advia Centaur CP). Intra-assay and interassay coefficients of variation were less than 8 and 10%, respectively, for the estimation of all hormones. Bone metabolism markers included osteocalcin (OC), type I procollagen amino-terminal peptide (P1NP), parathyroid hormone (PTH), and 25-hydroxyvitamin D (25(OH)VD), measured in a Roche Cobas e411 analyzer using immunometric assays (Roche Diagnostics, Indianapolis, IN, USA). The IGF-1 index was defined as the ratio of the measured value to the respective upper limit of the reference range for age and sex. Body mass index (BMI) was calculated using the following formula: weight (kg)/height2 (m2). The bone mineral density (BMD) measuring instrument was Discovery type W dual energy X-ray absorptiometry from the American HOLOGIC company. Quality control tests were conducted every working day. Before examination, the date of birth, height, weight, and menopause date of the examiner were accurately recorded, and then BMD (g/cm2) of the left hip and lumbar spine were measured by DEXA. Z value was used for premenopausal women and men younger than 50 years old, and Z-value = (measured value − mean bone mineral density of peers)/standard deviation of BMD of peers [17, 18]. In this study, low bone mass was defined as a Z-value of −2.0 or lower. 2.3. Statistical Analysis The baseline characteristics were compared between CD patients with and without low bone mass by using the Student’s t-test for continuous variables and the χ2 test for category variables. Bone turnover markers, alanine aminotransferase (ALT), triglyceride (TG), IGF-1 index, thyroid stimulating hormone (TSH), free triiodothyronine (FT3), free thyroxine (FT4), testosterone (T), 24 hours of urine cortisol (24 h UFC), and serum cortisol at 8 a.m. (F8 am) and at midnight (F24 pm) were not in normal distribution, so variables mentioned above were Log10-transformed, which could be used as continuous variables during statistical analysis. Participants were categorized into three groups according to tertiles of the IGF-1 index: <0.5986, 0.5986–0.8380, and >0.8380. The linear trend across IGF-1 index tertiles was tested using linear regression analysis for continuous variables and the Cochran–Armitage test for categorical variables. We used a multivariate logistic regression model to identify related factors that are independently associated with the risk of low bone mass. Variables included in the multivariate logistic regression model were selected based on the Spearman rank correlation analysis and established traditional low bone mass risk factors as priors. The results were presented as odds ratios (OR) and the corresponding 95% confidence intervals (CI). Significance tests were two-tailed, with value <0.05 considered statistically significant for all analyses. Statistical analysis was performed using SAS version 9.3 (SAS Institute Inc, Cary, NC, USA). 3. Results 3.1. The Prevalence of Low Bone Mass in Young Cushing’s Disease Patients From the inpatient system of Huashan hospital, a total of 153 CD patients under the age of 50 for men and premenopausal women (some with menstrual abnormalities were associated with CD) were included, aged from 13 to 49 years, with an average age of 34.25 ± 8.39 years. There were 33 males (21.57%) and 120 females (78.43%). These CD patients included newly diagnosed CD, recurrences of CD, and CD without remission after treatment. There were no differences in the prevalence of different statuses of CD between the two groups (Table 1). Table 1 Clinical and biochemical preoperative characteristics of young Cushing’s disease patients according to status of bone mineral density at diagnosis. Among these CD patients, low bone mass occurred in 74 patients (48.37%), including 24 men and 50 women. The prevalence of low bone mass was 41.67% and 72.73% in female and male CD patients, respectively, and 42 (56.76%) patients suffered from low bone mass in the lumbar spine only, while 10 (13.51%) patients had low bone mass in the femoral neck only, and 22 (29.73%) patients had low bone mass in both parts. In female patients with low bone mass, 27 (54%) had low bone mass in the lumbar region only, 9 (18%) in the femoral neck only, and 14 (28%) had low bone mass in both parts. For male patients with low bone mass, 16 (66.67%) patients had low bone mass only in the lumbar region, and the rest (8, 33.33%) had low bone mass in both parts. Ten patients had a history of fragility fractures (6 ribs, 3 vertebrae, 1 femoral neck, and ribs), and all of them achieved low bone mass in BMD. 3.2. Baseline Characteristics of Cushing’s Disease Patients with and without Low Bone Mass These CD patients were divided into two groups with and without low bone mass (Table 1). Compared to patients without low bone mass, those low bone mass patients had a higher level of diastolic blood pressure (DBP) (97.07 ± 13.69 vs. 89.76 ± 13.43, ), serum creatinine (66.15 ± 24.33 vs. 55.90 ± 13.35, ), uric acid (0.36 ± 0.10 vs. 0.32 ± 0.10, ), cholesterol (5.57 ± 1.30 vs. 5.06 ± 1.47, ), testosterone in women (2.10 (1.33–2.89) vs. 1.54 (0.97–2.05), ), F24 pm (22.31 (17.95–29.62) vs. 17.80 (13.75–22.77), ), and higher portion of male (32.43% vs. 11.54%, ), as well as hypertension (76.12% vs. 51.67%, ). The low bone mass group had a lower IGF-1 index (0.59 (0.43–0.76) vs. 0.79 (0.60–1.02), ) and FT3 level (3.54 (3.16–4.04) vs. 3.98 (3.47–4.45), ) than those without low bone mass. CD patients without low bone mass were more likely to have serum IGF-1 above the upper limit of the normal reference range (ULN) with age-adjusted (18, 26.87% vs. 3, 4.84%, ). No differences of bone turnover makers were found between the two groups. 3.3. Association between Baseline Characteristics and BMD Spearman’s rank correlation analysis was used to explore the related factors of low bone mass in young CD patients (Table 2). The results indicated that the Z score in the lumbar spine was positively associated with age at diagnosis (r = 0.18801, ), IGF-1 index (r = 0.35153, ), FT3 level (r = 0.24117, ), estradiol in women (r = 0.2361, ), and occurrence of normal menstruation in females (r = 0.2267, ). Meanwhile, SBP (r = −0.21575, ), DBP (r = −0.32538, ), ALT (r = −0.17477, ), serum creatinine (r = −0.36072, ), cholesterol (r = −0.20205, ), testosterone in women (r = −0.2700, ), F8 am (r = −0.18998, ), and serum cortisol at midnight (r = −0.27273, ) were negatively associated with the Z-score in the lumbar spine. The results also illustrated that the Z-score in the femoral neck was positively associated with BMI (r = 0.33926, ), IGF-1 index (r = 0.24418, ), FT3 level (r = 0.20487, ), and occurrence of normal menstruation in females (r = 0.2393, ). Serum creatinine (r = −0.1932, ), osteocalcin (r = −0.22744, ), and testosterone in women (r = −0.2363, ) were negatively associated with the Z-score in the femoral neck. Table 2 Spearman rank correlation of BMD and various variables in Cushing’s disease patients. 3.4. IGF-1 Index and Low Bone Mass Participants were categorized into the following three groups according to tertiles of the preoperative IGF-1 index: <0.5986 (tertiles 1), 0.5986–0.8380 (tertiles 2), and >0.8380 (tertiles 3). With the IGF-1 index increasing, the level of PTH decreased (54.85 (38.35–66.2), 38.9 (26.6–66.9), 36 (25.5–47.05), and ), while other bone metabolism makers, including PINP, osteocalcin, and 25 (OH) VD, showed no differences among the three groups (Figures 1(a)–1(d)). With the increase in the IGF-1 index level, the Z-score of both vertebra lumbalis (tertiles 1: −2.4 (−3.3∼−1.5); tertiles 2: −1.9 (−2.3∼−1.0); tertiles 3: −1.15 (−1.9∼−0.4), ) and the neck of femur (tertiles 1: −1.7 (−2.3∼−0.95); tertiles 2: −1.2 (−1.9∼−0.5); tertiles 3: −1.0 (−1.5∼−0.5), ) increased gradually (Figures 2(a) and 2(b)). Meanwhile, prevalence of low bone mass decreased (68.29%, 53.33%, 23.81%, ) (Figure 3(a)) both in the vertebra lumbalis (63.41%, 48.89%, 16.67%, ) and the neck of femur (32.5%, 11.11%, 11.19%, ), with the increasing of the IGF-1 index level (Figures 3(b) and 3(c)). (d) (a) (b) (c) (d) (a) (b) (c) (d) (a) (b) (c) (d) Figure 1 Bone turnover makers in three groups according to tertiles of the preoperative IGF-1 index. Tertiles 1: <0.5986, tertiles 2: 0.5986–0.8380, and tertiles 3 >0.8380. a for PINP; b for osteocalcin; c for PTH; d for VD-OH25. (a) p for trend = 0.2601. (b) p for trend = 0.1310. (c) p for trend = 0.008. (d) p for trend = 0.7956. (b) (a) (b) (a) (b) (a) (b) Figure 2 Z-score of both the neck of femur and the vertebra lumbalis in three tertiles of the IGF-1 index. a for the neck of femur; b for the vertebra lumbalis. Tertiles 1: <0.5986, tertiles 2: 0.5986–0.8380, and tertiles 3 >0.8380. (a) p for trend = 0.0148. (b) p for trend < 0.0001. (c) (a) (b) (c) (a) (b) (c) (a) (b) (c) Figure 3 Prevalence of low bone mass according to tertiles of the preoperative IGF-1 index. With increment of the IGF-1 index level, prevalence of low bone mass decreased, both in the vertebra lumbalis and neck of femur. Tertiles 1: <0.5986, tertiles 2: 0.5986–0.8380, and tertiles 3 >0.8380. (a) p for trend = 0.0002. (b) p for trend = 0.0169. (c) p for trend < 0.0001. In the logistic regression analysis of the related factors of low bone mass, most of the potentially relevant factors were put into this model; only the IGF-1 index was still significantly negatively associated with the prevalence of low bone mass after adjusting for covariables. The results indicated that compared to the patients in the lowest tertile of the IGF-1 index (<0.5563), those with the highest tertile of the IGF-1 index (≥0.7993) had a lower prevalence of low bone mass (95% CI 0.16 (0.06–0.41), ). After adjusting for age, gender, and BMI, the patients in the highest tertile of the IGF-1 index still conferred a lower prevalence of low bone mass (95% CI 0.15 (0.06–0.42), ). The association between the IGF-1 index and low bone mass still existed (95% CI 0.02 (0.001–0.5), ) even after adjusting for age, gender, CD duration, BMI, hypertension, dyslipidemia, diabetes, ALT, Scr, FT3, F24 pm, PTH, and osteocalcin (Table 3). In comparison to the reference population, the participants in the middle tertile of the IGF-1 index (0.5563–0.7993) had no different risk of low bone mass. Table 3 Association between the preoperative IGF-1 index and the risk of low bone mass. 4. Discussion Our results revealed that low bone mass occurred in around half of young CD patients, affecting more males than females, and mostly in the lumbar spine. The CD patients in our study had a high prevalence (48.37%) of low bone mass at the baseline. This was in accordance with the findings of previous research, and the reported prevalence of osteoporosis due to excess endogenous cortisol ranges from 22% to 59% [19–25]. In this study, CD patients’ lumbar vertebrae were more severely affected than the neck of the femur. It is reported that lumbar vertebrae, containing more trabecular bone than femur neck, were more vulnerable to endogenous cortisol [26]. Our results also indicated that men were more prone to low bone mass than women in CD, which was in accordance with several other studies [23, 27, 28]; possibly, the deleterious effect of cortisol excess on BMD might overrule the protective effects of sex hormones, and men were more often hypogonadal compared with women in CD patients. In our study, patients with low bone mass had a significantly higher level of F24 pm. Both cortisol levels in the morning and at midnight, were negatively associated with the Z-score of BMD in the lumbar spine at diagnosis. But these results were not seen in the femoral neck at diagnosis. This further indicated that lumbar vertebrae were more vulnerable to endogenous cortisol. BMI was considered to be associated with bone mass [29]. In our study, higher BMI was associated with higher BMD at diagnosis in the femur neck but not in the lumbar vertebrae, consistent with other studies [30]. Interestingly, besides the above known related factors, we also found that a higher level of the IGF-1 index was strongly associated with a lower prevalence of low bone mass, both in the vertebra lumbalis and the neck of the femur, independently of age, gender, duration, BMI, hypertension, dyslipidemia, diabetes, level of ALT, creatinine, FT3, and F24 pm. The IGF-1 index was also positively associated with the BMD Z-score, both in the lumbar spine and the femoral neck. So far, there have been few studies concerning the association between IGF-1 and low bone mass in Cushing’s disease patients. As we know, GH [31, 32] and IGF-1 [33] have been demonstrated to increase both bone formation (e.g., collagen synthesis) and bone resorption. However, in CD patients, glucocorticoids resulted in decreased number and dysfunction of osteoblasts by inhibiting GH-IGF-1 axes [34, 35]. In vitro studies suggested that at high concentrations of glucocorticoids, a decreased release of GHRH had been reported [36–38]; therefore, GH-IGF-1 axes were inhibited. IGF-1 possessed anabolic mitogenic actions in osteoblasts while reducing the anabolic actions of TGF-β [39]. The decrease in IGF-1 might be a risk factor for low bone mass in CD patients. In vitro studies had also indicated that the suppressive effects of glucocorticoids on osteoblast function can be partially reversed by GH or IGF treatment [8]. In recent years, some studies have also shown that patients with untreated Cushing’s disease may have elevated IGF-1, and mildly elevated IGF-1 in Cushing’s disease does not imply pathological growth hormone excess. Higher IGF-1 levels could predict better outcomes in CD [40, 41]. Possible mechanisms were not clear, which might involve changes in IGF binding proteins (IGFBPs), interference in IGFBP fragments, IGF-1 synthesis or clearance, and/or the effects of hyperinsulinism induced by excess glucocorticoids. In our study, the results also showed that IGF-1 was an independent protective factor for low bone mass in CD patients. Our study was one of the few well-powered research studies on the association of IGF-1 levels with low bone mass in young CD patients. These represented important strengths of our study, especially given the rarity of CD. The main limitation of this study was its retrospective nature. This could not prove causality. A prospective study should be conducted to explore the causality between IGF-1 and osteoporosis in CD patients. In addition, this study lacked morphometric data for spinal fractures in all patients, which may underestimate the incidence of fractures and osteoporosis. However, our study indicated that a lower IGF-1 index level was significantly associated with low bone mass in young CD patients, which might provide a new aspect to understand the possible risk factors and mechanism of osteoporosis in CD patients. In conclusion, our study found that a higher IGF-1 index was independently and significantly associated with decreased prevalence of low bone mass in young CD patients, drawing attention to the role of IGF-1 in the pathogenesis of CD-caused low bone mass and may support the exploration of this pathway in therapeutic agent development in antiosteoporosis in CD. Data Availability The data used to support the findings of the study are available on request from the authors. Additional Points Through a retrospective study of a large sample of Cushing’s disease (CD) patients from a single center, we found that a higher IGF-1 index was independently associated with a lower prevalence of low bone mass in young CD patients and IGF-1 might play an important role in the pathogenesis of CD-caused low bone mass. Disclosure Wanwan Sun and Quanya Sun were the co-first authors. Conflicts of Interest The authors declare that they have no conflicts of interest. Authors’ Contributions Wanwan Sun analyzed the data and wrote the manuscript. Quanya Sun collected the data. Hongying Ye and Shuo Zhang conducted the study design and quality control. All authors read and approved the final manuscript. Wanwan Sun and Quanya Sun contributed equally to this work. Acknowledgments The present study was supported by grants from the initial funding of the Huashan Hospital (2021QD023). The study was also supported by grants from Multidisciplinary Diagnosis and Treatment (MDT) demonstration project in research hospitals (Shanghai Medical College, Fudan University, no: DGF501053-2/014). References H. Nishioka and S. Yamada, “Cushing's disease,” Journal of Clinical Medicine, vol. 8, no. 11, p. 1951, 2019. View at: Publisher Site | Google Scholar H. Cushing, “The basophil adenomas of the pituitary body and their clinical manifestations (pituitary basophilism),” Obesity Research, vol. 2, no. 5, pp. 486–508, 1994. View at: Publisher Site | Google Scholar R. A. Feelders, S. J. Pulgar, A. Kempel, and A. M. Pereira, “Management of endocrine disease: the burden of Cushing's disease: clinical and health-related quality of life aspects,” European Journal of Endocrinology, vol. 167, no. 3, pp. 311–326, 2012. View at: Publisher Site | Google Scholar R. Pivonello, M. De Leo, A. Cozzolino, and A. Colao, “The treatment of Cushing's disease,” Endocrine Reviews, vol. 36, no. 4, pp. 385–486, 2015. View at: Publisher Site | Google Scholar R. Pivonello, M. C. De Martino, M. De Leo, C. Simeoli, and A. Colao, “Cushing's disease: the burden of illness,” Endocrine, vol. 56, no. 1, pp. 10–18, 2017. View at: Publisher Site | Google Scholar R. S. Hardy, H. Zhou, M. J. Seibel, and M. S. Cooper, “Glucocorticoids and bone: consequences of endogenous and exogenous excess and replacement therapy,” Endocrine Reviews, vol. 39, no. 5, pp. 519–548, 2018. View at: Publisher Site | Google Scholar R. Bouillon and A. Prodonova, “Growth hormone deficiency and peak bone mass: laboratory for experimental medicine and Endocrinology, catholic university of leuven, gasthuisberg, leuven, Belgium,” Journal of Pediatric Endocrinology and Metabolism, vol. 13, no. s2, pp. 1327–1342, 2000. View at: Publisher Site | Google Scholar A. Giustina, G. Mazziotti, and E. Canalis, “Growth hormone, insulin-like growth factors, and the skeleton,” Endocrine Reviews, vol. 29, no. 5, pp. 535–559, 2008. View at: Publisher Site | Google Scholar T. Sugimoto, K. Nishiyama, F. Kuribayashi, and K. Chihara, “Serum levels of insulin-like growth factor (IGF) I, IGF-binding protein (IGFBP)-2, and IGFBP-3 in osteoporotic patients with and without spinal fractures,” Journal of Bone and Mineral Research, vol. 12, no. 8, pp. 1272–1279, 1997. View at: Publisher Site | Google Scholar P. Garnero, E. Sornay-Rendu, and P. D. Delmas, “Low serum IGF-1 and occurrence of osteoporotic fractures in postmenopausal women,” The Lancet, vol. 355, no. 9207, pp. 898-899, 2000. View at: Publisher Site | Google Scholar C. Ohlsson, D. Mellström, D. Carlzon et al., “Older men with low serum IGF-1 have an increased risk of incident fractures: the MrOS Sweden study,” Journal of Bone and Mineral Research, vol. 26, no. 4, pp. 865–872, 2011. View at: Publisher Site | Google Scholar M. Hotta, T. Shibasaki, A. Masuda et al., “Effect of human growth hormone-releasing hormone on GH secretion in Cushing's syndrome and non-endocrine disease patients treated with glucocorticoids,” Life Sciences, vol. 42, no. 9, pp. 979–984, 1988. View at: Publisher Site | Google Scholar L. T. Braun and M. Reincke, “The effect of biochemical remission on bone metabolism in Cushing's syndrome: a 2-year follow-up study,” Journal of Bone and Mineral Research, vol. 36, no. 11, pp. 2281-2282, 2021. View at: Publisher Site | Google Scholar I. Kanazawa, T. Yamaguchi, M. Yamamoto, M. Yamauchi, S. Yano, and T. Sugimoto, “Serum insulin-like growth factor-I level is associated with the presence of vertebral fractures in postmenopausal women with type 2 diabetes mellitus,” Osteoporosis International, vol. 18, no. 12, pp. 1675–1681, 2007. View at: Publisher Site | Google Scholar A. Scillitani, G. Mazziotti, C. Di Somma et al., “Treatment of skeletal impairment in patients with endogenous hypercortisolism: when and how?” Osteoporosis International, vol. 25, no. 2, pp. 441–446, 2014. View at: Publisher Site | Google Scholar M. Fleseriu, R. Auchus, I. Bancos et al., “Consensus on diagnosis and management of Cushing's disease: a guideline update,” Lancet Diabetes and Endocrinology, vol. 9, no. 12, pp. 847–875, 2021. View at: Publisher Site | Google Scholar J. M. Liu, D. L. Zhu, Y. M. Mu, and W. B. Xia, “Chinese Society of Osteoporosis and Bone Mineral Research, the Chinese Society of Endocrinology, Chinese Diabetes Society, Chinese Medical Association; Chinese Endocrinologist Association, Chinese Medical Doctor Association,” Management of fracture risk in patients with diabetes-Chinese Expert Consensus Journal of Diabetes, vol. 11, pp. 906–919, 2019. View at: Google Scholar P. M. Camacho, S. M. Petak, N. Binkley et al., “American association of clinical endocrinologists and American college of endocrinology clinical practice guidelines for the diagnosis and treatment of postmenopausal OSTEOPOROSIS-2020 update,” Endocrine Practice, vol. 26, no. 1, pp. 1–46, 2020. View at: Publisher Site | Google Scholar C. V. dos Santos, L. Vieira Neto, M. Madeira et al., “Bone density and microarchitecture in endogenous hypercortisolism,” Clinical Endocrinology, vol. 83, no. 4, pp. 468–474, 2015. View at: Publisher Site | Google Scholar N. Ohmori, K. Nomura, K. Ohmori, Y. Kato, T. Itoh, and K. Takano, “Osteoporosis is more prevalent in adrenal than in pituitary Cushing's syndrome,” Endocrine Journal, vol. 50, no. 1, pp. 1–7, 2003. View at: Publisher Site | Google Scholar M. E. Randazzo, E. Grossrubatscher, P. Dalino Ciaramella, A. Vanzulli, and P. Loli, “Spontaneous recovery of bone mass after cure of endogenous hypercortisolism,” Pituitary, vol. 15, no. 2, pp. 193–201, 2012. View at: Publisher Site | Google Scholar L. Tauchmanovà, R. Pivonello, C. Di Somma et al., “Bone demineralization and vertebral fractures in endogenous cortisol excess: role of disease etiology and gonadal status,” Journal of Clinical Endocrinology and Metabolism, vol. 91, no. 5, pp. 1779–1784, 2006. View at: Publisher Site | Google Scholar E. Valassi, A. Santos, M. Yaneva et al., “The European Registry on Cushing's syndrome: 2-year experience. Baseline demographic and clinical characteristics,” European Journal of Endocrinology, vol. 165, no. 3, pp. 383–392, 2011. View at: Publisher Site | Google Scholar L. Trementino, G. Appolloni, L. Ceccoli et al., “Bone complications in patients with Cushing's syndrome: looking for clinical, biochemical, and genetic determinants,” Osteoporosis International, vol. 25, no. 3, pp. 913–921, 2014. View at: Publisher Site | Google Scholar A. W. van der Eerden, M. den Heijer, W. J. Oyen, and A. R. Hermus, “Cushing's syndrome and bone mineral density: lowest Z scores in young patients,” The Netherlands Journal of Medicine, vol. 65, no. 4, pp. 137–141, 2007. View at: Google Scholar P. G. Lacativa and M. L. F. Farias, “Office practice of osteoporosis evaluation,” Arquivos Brasileiros de Endocrinologia and Metabologia, vol. 50, no. 4, pp. 674–684, 2006. View at: Publisher Site | Google Scholar L. H. A. Broersen, F. M. van Haalen, T. Kienitz et al., “Sex differences in presentation but not in outcome for ACTH-dependent Cushing's syndrome,” Frontiers in Endocrinology, vol. 10, p. 580, 2019. View at: Publisher Site | Google Scholar F. P. Giraldi, M. Moro, and F. Cavagnini, “Gender-related differences in the presentation and course of Cushing's disease,” Journal of Clinical Endocrinology and Metabolism, vol. 88, no. 4, pp. 1554–1558, 2003. View at: Publisher Site | Google Scholar S. Morin, J. F. Tsang, and W. D. Leslie, “Weight and body mass index predict bone mineral density and fractures in women aged 40 to 59 years,” Osteoporosis International, vol. 20, no. 3, pp. 363–370, 2009. View at: Publisher Site | Google Scholar M. Zilio, M. Barbot, F. Ceccato et al., “Diagnosis and complications of Cushing's disease: gender-related differences,” Clinical Endocrinology, vol. 80, no. 3, pp. 403–410, 2014. View at: Publisher Site | Google Scholar G. Amato, C. Carella, S. Fazio, G. La Montagna, A. Cittadini, and D. Sabatini, “Body composition, bone metabolism, and heart structure and function in growth hormone (GH)-deficient adults before and after GH replacement therapy at low doses,” Journal of Clinical Endocrinology and Metabolism, vol. 77, no. 6, pp. 1671–1676, 1993. View at: Publisher Site | Google Scholar S. A. Beshyah, E. Thomas, P. Kyd, P. Sharp, A. Fairney, and D. G. Johnston, “The effect of growth hormone replacement therapy in hypopituitary adults on calcium and bone metabolism,” Clinical Endocrinology, vol. 40, no. 3, pp. 383–391, 2010. View at: Publisher Site | Google Scholar P. R. Ebeling, J. D. Jones, W. M. O'Fallon, C. H. Janes, and B. L. Riggs, “Short-term effects of recombinant human insulin-like growth factor I on bone turnover in normal women,” Journal of Clinical Endocrinology and Metabolism, vol. 77, no. 5, pp. 1384–1387, 1993. View at: Publisher Site | Google Scholar G. Mazziotti and A. Giustina, “Glucocorticoids and the regulation of growth hormone secretion,” Nature Reviews Endocrinology, vol. 9, no. 5, pp. 265–276, 2013. View at: Publisher Site | Google Scholar N. A. Tritos, “Growth hormone deficiency in adults with Cushing's disease,” Best Practice and Research Clinical Endocrinology and Metabolism, vol. 35, no. 2, Article ID 101474, 2021. View at: Publisher Site | Google Scholar K. Nakagawa, T. Ishizuka, T. Obara, M. Matsubara, and K. Akikawa, “Dichotomic action of glucocorticoids on growth hormone secretion,” Acta Endocrinologica, vol. 116, no. 2, pp. 165–171, 1987. View at: Publisher Site | Google Scholar G. Fernández-Vázquez, L. Cacicedo, M. J. Lorenzo, R. Tolón, J. López, and F. Sánchez-Franco, “Corticosterone modulates growth hormone-releasing factor and somatostatin in fetal rat hypothalamic cultures,” Neuroendocrinology, vol. 61, no. 1, pp. 31–35, 1995. View at: Publisher Site | Google Scholar S. K. Fife, R. S. Brogan, A. Giustina, and W. B. Wehrenberg, “Immunocytochemical and molecular analysis of the effects of glucocorticoid treatment on the hypothalamic-somatotropic axis in the rat,” Neuroendocrinology, vol. 64, no. 2, pp. 131–138, 1996. View at: Publisher Site | Google Scholar T. L. McCarthy, M. Centrella, and E. Canalis, “Cortisol inhibits the synthesis of insulin-like growth factor-I in skeletal cells,” Endocrinology, vol. 126, no. 3, pp. 1569–1575, 1990. View at: Publisher Site | Google Scholar K. English, V. Chikani, G. Dimeski, and W. J. Inder, “Elevated insulin‐like growth factor‐1 in Cushing’s disease,” Clinical Endocrinology, vol. 91, no. 1, pp. 141–147, 2019. View at: Publisher Site | Google Scholar E. Gezer, B. Çetinarslan, A. Selek et al., “The association between insulin-like growth factor 1 levels within reference range and early postoperative remission rate in patients with Cushing’s disease,” Endocrine Research, vol. 46, no. 3, pp. 92–98, 2021. View at: Publisher Site | Google Scholar Copyright Copyright © 2023 Wanwan Sun et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. From https://www.hindawi.com/journals/ije/2023/3334982/
    1 point
  43. In this application note, Tecan presents a method for diagnosing Cushing's syndrome efficiently and accurately. The approach involves simultaneous the measurement of cortisol and dexamethasone levels using LC-MS/MS, which reduces false positives in dexamethasone suppression test (DSTs). The described LC-MS/MS method enables the tracking of multiple analytes, including cortisol, cortisone, and dexamethasone, in serum or plasma. Implementing this analytical approach offers clinical laboratories a straightforward means of performing DSTs, and the availability of a commercially available kit ensures reliable and reproducible results. Download this Article Download PDF >> From https://www.selectscience.net/application-articles/minimizing-the-number-of-false-positives-in-dexamethasone-suppression-testing-for-the-diagnosis-of-cushings-syndrome?artID=59632
    1 point
  44. In Italy it is estimated that there are about 3,000 patients suffering from Cushing’s syndrome, while in Europe the number rises to over 50,000. The Cushing’s syndrome, a disease caused by the excessive production of cortisol by the pituitary gland due to a benign tumor of the gland, has seen a breakthrough in its treatment. Thanks to a new drug called osilodrostat, approved in 2020 by the Food and Drug Administration and subsequently by Aifa in Italy, patients unfit for surgery can benefit from a treatment that offers the same effects as a scalpel. Furthermore, this drug reduced symptoms in 80% of cases. Cushing’s syndrome has been dubbed “full moon face disease” due to its most obvious visible effects, such as a rounding of the face caused by fat accumulation and visible weight gain also on the waist and back. Despite its symptomatic relevance, the disease has long been poorly understood by both healthcare professionals and the general public. To raise awareness of this syndrome, the #Thiscushing campaign has been launched, which aims to spread knowledge about the disease. The campaign recently stopped in Rome, during the Congress of the Italian Society of Endocrinology (SIE), where a photographic exhibition was organized which represents moments of daily life of people affected by Cushing’s syndrome and their difficulties. Despite the debilitating symptoms, Cushing’s syndrome is often underdiagnosed, resulting in delays in diagnosis of up to 5-7 years. The disease presents a wide range of symptoms, ranging from difficulty performing even simple daily activities such as tying your shoes or getting out of bed, to common manifestations such as high cholesterol, hypertension and hyperglycemia, which can be confused with symptoms of other less common pathologies. serious. It is for this reason that the EIS experts are appealing for the inclusion of Cushing’s syndrome in the list of rare pathologies recognized by the Ministry of Health, in order to facilitate timely diagnosis and faster access to the necessary treatments. From https://www.breakinglatest.news/health/cushings-syndrome-a-new-drug-allows-you-to-avoid-surgery/
    1 point
  45. Abstract Background The diagnosis of Cushing’s syndrome is challenging; however, through the clinical picture and the search for secondary causes of osteoporosis, it was possible to reach the diagnosis of the case reported. There was an independent, symptomatic ACTH hypercortisolism manifested by typical phenotypic changes, severe secondary osteoporosis and arterial hypertension in a young patient. Case presentation A 20-year-old Brazilian man with low back pain for 8 months. Radiographs showed fragility fractures in the thoracolumbar spine, and bone densitometry showed osteoporosis, especially when evaluating the Z Score (− 5.6 in the lumbar spine). On physical examination, there were wide violaceous streaks on the upper limbs and abdomen, plethora and fat increase in the temporal facial region, hump, ecchymosis on limbs, hypotrophy of arms and thighs, central obesity and kyphoscoliosis. His blood pressure was 150 × 90 mmHg. Cortisol after 1 mg of dexamethasone (24.1 µg/dL) and after Liddle 1 (28 µg/dL) were not suppressed, despite normal cortisoluria. Tomography showed bilateral adrenal nodules with more severe characteristics. Unfortunately, through the catheterization of adrenal veins, it was not possible to differentiate the nodules due to the achievement of cortisol levels that exceeded the upper limit of the dilution method. Among the hypotheses for the differential diagnosis of bilateral adrenal hyperplasia are primary bilateral macronodular adrenal hyperplasia, McCune–Albright syndrome and isolated bilateral primary pigmented nodular hyperplasia or associated with Carney’s complex. In this case, primary pigmented nodular hyperplasia or carcinoma became important etiological hypotheses when comparing the epidemiology in a young man and the clinical-laboratory-imaging findings of the differential diagnoses. After 6 months of drug inhibition of steroidogenesis, blood pressure control and anti-osteoporotic therapy, the levels and deleterious metabolic effects of hypercortisolism, which could also impair adrenalectomy in the short and long term, were reduced. Left adrenalectomy was chosen, given the possibility of malignancy in a young patient and to avoid unnecessary definitive surgical adrenal insufficiency if the adrenalectomy was bilateral. Anatomopathology of the left gland revealed expansion of the zona fasciculate with multiple nonencapsulated nodules. Conclusion The early identification of Cushing’s syndrome, with measures based on the assessment of risks and benefits, remains the best way to prevent its progression and reduce the morbidity of the condition. Despite the unavailability of genetic analysis for a precise etiological definition, it is possible to take efficient measures to avoid future damage. Peer Review reports Background Cushing’s syndrome may be exogenous or endogenous and, in this case, can be ACTH-dependent or independent. In the case reported, there was an independent, symptomatic ACTH hypercortisolism manifested by typical phenotypic changes, severe secondary osteoporosis and arterial hypertension in a young patient. Osteoporosis secondary to hypercortisolism occurs due to chronic reduction in bone formation, loss of osteocytes and increased reabsorption caused by intense binding of cortisol to glucocorticoid receptors present in bone cells [1]. In addition, excess cortisol impairs vitamin D metabolism and reduces endogenous parathyroid hormone secretion, intestinal calcium reabsorption, growth hormone release, and lean body mass [2]. Subclinical Cushing disease occurs in up to 11% of individuals diagnosed with early-onset osteoporosis and 0.5–1% of hypertension patients. [3] A cross-sectional study published in 2023 revealed a prevalence of 81.5% bone loss in 19 patients with Cushing’s syndrome [2]. The prevalence of osteopenia ranges from 60 to 80%, and the prevalence of osteoporosis ranges from 30 to 65% in patients with Cushing’s syndrome. Additionally, the incidence of fragility fractures ranges from 30 to 50% in these patients [4] and is considered the main cause of morbidity affecting the quality of life. The diagnosis is challenging, given the presence of confounding factors; however, through the clinical picture and the search for secondary causes of osteoporosis, it was possible to reach a syndromic diagnosis. Early identification of this syndrome, with measures based on the assessment of risks and benefits, remains the best way to prevent progression and reduce morbidity related to this disease [2]. Case presentation A 20-year-old Brazilian male patient reported low back pain that had evolved for 8 months, with no related trauma. He sought emergency care and performed spinal radiographs on this occasion (03/2019). Due to the several alterations observed in the images, he was referred to the Orthopedics Service of the Hospital of Federal University of Juiz de Fora, which prescribed orthopedic braces, indicated physical therapy and was referred again to the Osteometabolic Diseases outpatient clinic of the Endocrinology and Rheumatology Services of the Hospital of Federal University of Juiz de Fora on 10/2019. The radiographs showed a marked reduction in the density of bone structures, scoliotic deviation with convexity toward the left and reduction in the height of the lumbar vertebrae, with partial collapses of the vertebral bodies at the level of T12, L1, L2, L3 and L5, with recent collapses in T12 and L1, suggesting bone fragility fractures. The same can be seen in posterior magnetic resonance imaging (Fig. 1). Fig. 1 Radiography and Magnetic Resonance Imaging (MRI) of lumbosacral spine in profile Full size image Bone scintigraphy on 08/2019 did not reveal hyper flow or anomalous hyperemia in the topography of the thoracolumbar spine, and in the later images of the exam, there was a greater relative uptake of the tracer in the lumbar spine (vertebrae T10–T12, L2–L4), of nonspecific aspect, questioning the presence of osteoarticular processes or ankylosing spondylitis. It was also observed in the bone densitometry requested in October 2019, performed by dual-energy X-ray absorptiometry (DXA), low bone mineral density (BMD) in the lumbar spine, femoral neck and total femur, when comparing the results to evaluating the Z Score (Table 1). Table 1 Dual-energy X-ray absorptiometry (DXA) Full size table Thus, the diagnosis of osteoporosis was established, and treatment with vitamin D 7000 IU per week was started due to vitamin D3 insufficiency associated with the bisphosphonate alendronate 70 mg, also weekly. The patient had a past pathological history of fully treated syphilis (2018) and perianal condyloma with a surgical resection on 09/2017 and 02/2018. In the family history, it was reported that a maternal uncle died of systemic sclerosis. In the social context, the young person denied drinking alcohol and previous or current smoking. On physical examination, there were no lentiginous skin areas or blue nevi; however, wide violet streaks were observed on the upper limbs and abdomen, with plethora and increased fat in the temporal facial region and hump (Fig. 2a, b), limb ecchymosis, hypotrophy of the arms and thighs, central obesity and kyphoscoliosis. Systemic blood pressure (sitting) was 150 × 90 mmHg, BMI was 26.09 kg/m2, and waist circumference was 99 cm, with no reported reduction in height, maintained at 1.55 m. Fig. 2 Changes in the physical examination. a Violet streaks on the upper limbs, b Violet streaks on abdomen Full size image An investigation of secondary causes for osteoporosis was initiated, with the following laboratory test results (Table 2). Table 2 Laboratory tests Full size table Computed tomography of the abdomen with adrenal protocol performed on 08/13/2020 characterized isodense nodular formation in the body of the left adrenal and in the lateral portion of the right adrenal, measuring 1.5 cm and 0.6 cm, respectively. The lesions had attenuation of approximately 30 HU, showing enhancement by intravenous contrast, with an indeterminate washout pattern in the late phase after contrast (< 60%) (Fig. 3). Fig. 3 Computed tomography abdomen with adrenal protocol Full size image After contact with the interventional radiology of the Hospital of Federal University of Juiz de Fora, catheterization of adrenal veins was performed on 10/2020; however, it was not possible to perform adequate lesion characterization due to obtaining serum cortisol levels that extrapolated the dilutional upper limit of the method (Table 3). Table 3 Adrenal catheterization Full size table The calculation of the selectivity index was 6.63 (Reference Value (RV) > 3), confirming the good positioning of the catheter within the vessels during the procedure. The calculated lateralization index was 1.1296 (VR < 3), denoting bilateral hormone production. However, as aldosterone was not collected from a peripheral vein, it was not possible to obtain the contralateral rate and define whether there was contralateral suppression of aldosterone production [5]. Due to pending diagnoses for a better therapeutic decision and Cushing’s syndrome in clear evolution and causing organic damage, it was decided, after catheterization, to make changes in the patient’s drug prescription. Ketoconazole 400 mg per day was started, the dose of vitamin D was increased to 14,000 IU per week, and ramipril 5 mg per day was prescribed due to secondary hypertension. In addition, given the severity of osteoporosis, it was decided to replace previously prescribed alendronate with zoledronic acid. Magnetic resonance imaging of the upper abdomen was performed on 06/19/2021, which demonstrated lobulated nodular thickening in the left adrenal gland with areas of decreased signal intensity in the T1 out-phase sequence, denoting the presence of fat, and homogeneous enhancement using contrast, measuring approximately 1.7 × 1.5 × 1.3 cm, suggestive of an adenoma. There was also a small nodular thickening in the lateral arm of the right adrenal, measuring approximately 0.8 × 0.6 cm, which was difficult to characterize due to its small dimensions and nonspecific appearance. PPNAD or carcinoma became an important etiological hypothesis for the case described when comparing the epidemiology in a young man and the clinical-laboratory-imaging findings of the differential diagnoses. According to a dialog with the patient and family, the group of experts opted for unilateral glandular surgical resection on the left side (11/11/2021), where more significant changes were visualized, as there was a possibility of malignancy in a young patient and to avoid a definitive adrenal insufficiency condition because of bilateral adrenalectomy. This would first allow the analysis of the material and follow-up of the evolution of the condition with the permanence of the contralateral gland. In the macroscopic analysis of the adrenalectomy specimen, adrenal tissue weighing 20 g and measuring 9.3 × 5.5 × 2.0 cm was described, completely surrounded by adipose tissue. The gland has a multinodular surface and varies between 0.2 and 1.6 cm in thickness, showing a cortex of 0.1 cm in thickness and a medulla of 1.5 cm in thickness (Fig. 4). Fig. 4 Left adrenal Full size image The microscopic analysis described the expansion of the zona fasciculate, with the formation of multiple nonencapsulated nodules composed of polygonal cells with ample and eosinophilic cytoplasm and frequent depletion of intracytoplasmic lipid content. No areas of necrosis or mitotic activity were observed. The histopathological picture is suggestive of cortical pigmented micronodular hyperplasia of the adrenal gland. For the final etiological definition and an indication of contralateral adrenalectomy, which could be unnecessary and would avoid chronic corticosteroid therapy, or else, it would be necessary to protect the patient from future complications with the maintenance of the disease in the right adrenal gland, it would be essential to search for mutations in the PRKAR1A, PDE11A, PDE8B and PRKACA genes [15]; however, such genetic analysis is not yet widely available, and the impossibility of carrying it out at the local level did not allow a complete conclusion of the case. Discussion Through the clinical picture presented and the research of several secondary causes for osteoporosis, it was possible to arrive at the diagnosis of Cushing syndrome [6]. There was symptomatic independent ACTH hypercortisolism, manifested by typical phenotypic changes, severe secondary osteoporosis, and arterial hypertension in a young patient. The diagnosis of Cushing’s syndrome is always challenging, given the presence of confounding factors such as the following: Physiological states of hypercortisolism—pseudo Cushing (strenuous exercise, pregnancy, uncontrolled diabetes, sleep apnea, chronic pain, alcohol withdrawal, psychiatric disorders, stress, obesity, glucocorticoid resistance syndromes); Cyclic or mild—subclinical Cushing’s pictures; Frequent and, even unknown, short- and long-term use of corticosteroids under different presentations; Increase in the general population incidence of diabetes and obesity; Screening tests with singularities for collection and individualized for different patient profiles. It is important to note that the basal morning cortisol measurement is not the ideal test to assess hypercortisolism and is better applied to the assessment of adrenal insufficiency. However, the hypercortisolism of the case was unequivocal, and this test was also shown to be altered several times. As no test is 100% accurate, the current guidelines suggest the use of at least two first-line functional tests that focus on different aspects of the pathophysiology of the hypothalamic‒pituitary‒adrenal axis to confirm the hypercortisolism state: 24-hours cortisol, nocturnal salivary cortisol, morning serum cortisol after suppression with 1 mg of dexamethasone or after Liddle 1. Given that night-time salivary cortisol would require hospitalization, the other suggested tests were chosen, which are easier to perform in this context [7, 8]. Subsequently, tests were performed to determine the cause of hypercortisolism, such as serum ACTH levels and adrenal CT. The suppressed ACTH denoted the independence of its action. CT showed bilateral adrenal nodules with more severe features: solid lesion, attenuation > 10 UI on noncontrast images, and contrast washout speed < 60% in 10 minutes. In this case, it is essential to make a broad clinical decision and dialog with the patient to weigh and understand the risks and benefits of surgical treatment [9]. Among the main diagnostic hypotheses for the differential diagnosis of bilateral adrenal hyperplasia are primary bilateral macronodular adrenal hyperplasia, McCune–Albright syndrome (MAS) and bilateral primary pigmented nodular hyperplasia (PPNAD) isolated or associated with Carney’s complex. Another possibility would be bilateral adrenocorticotropic hormone (ACTH)-dependent macronodular hyperplasia secondary to long-term adrenal stimulation in patients with Cushing’s disease (ACTH-secreting pituitary tumor) or ectopic ACTH production, but the present case did not present with ACTH elevation. Primary macronodular adrenal hyperplasia (nodules > 1 cm) predominates in women aged 50–60 years and may also be detected in early childhood (before 5 years) in the context of McCune–Albright syndrome. Most cases are considered sporadic; however, there are now several reports of familial cases whose presentation suggests autosomal dominant transmission. Several pathogenic molecular causes were identified in the table, indicating that it is a heterogeneous disease [10]. The pathophysiology occurs through the expression of anomalous ectopic hormone receptors or amplified eutopic receptors in the adrenals. It usually manifests in an insidious and subclinical way, with cortisol secretion mediated through receptors for gastric inhibitory peptide (GIP), vasopressin (ADH), catecholamines, interleukin 1 (IL-1), leptin, luteinizing hormone (LH), serotonin or others. Nodular development is not always synchronous or multiple; thus, hypercortisolism only manifests when there is a considerable increase in the number of adrenocortical cells, with severe steroidogenesis observed by cortisoluria greater than 3 times the upper limit of normal. Patients with mild Cushing’s syndrome should undergo screening protocols to identify aberrant receptors, as this may alter the therapeutic strategy. If there is evidence of abnormal receptors, treatment with beta-blockers is suggested for patients with beta-adrenergic receptors or with gonadotropin-releasing hormone (GnRH) agonists (and sex steroid replacement) for patients with LH/hCG receptors. In patients in whom aberrant hormone receptors are not present or for whom no specific pharmacological blockade is available or effective, the definitive treatment is bilateral adrenalectomy, which is known to make the patient dependent on chronic corticosteroid therapy [11]. Studies have shown the effectiveness of unilateral surgery in the medium and long term, opting for the resection of the adrenal gland of greater volume and nodularity by CT, regardless of the values obtained by catheterization of adrenal veins, but with the possibility of persistence or recurrence in the contralateral gland. Another possibility would be total unilateral adrenalectomy associated with subtotal contralateral adrenalectomy [12]. In McCune–Albright syndrome (MAS), there are activating mutations in the G-protein GNAS1 gene, generating autonomic hyperfunction of several tissues, endocrine or not, and there may be, for example, a constant stimulus similar to ACTH on the adrenal gland. In this case, pituitary levels of ACTH are suppressed, and adrenal adenomas with Cushing’s syndrome appear. Hypercortisolism may occur as an isolated manifestation of the syndrome or be associated with the triad composed of polyostotic fibrous dysplasia, café au lait spots with irregular borders and gonadal hyperfunction with peripheral precocious puberty. The natural history of Cushing’s syndrome in McCune-Albright syndrome (MAS) is heterogeneous, with some children evolving with spontaneous resolution of hypercortisolism, while others have a more severe condition, eventually requiring bilateral adrenalectomy [13]. PPNAD predominates in females, in people younger than 30 years, multiple and small (< 6 mm) bilateral pigmented nodules (surrounded by atrophied cortex), which can reach 1.5 cm in adulthood, with family genetic inheritance (66%) or sporadic inheritance (33%), and as part of the Carney complex reported in 40% of cases. In 70% of cases, inactivating mutations are identified in the PKA regulatory 1-alpha subunit (PRKAR1A), a tumor suppressor gene [14]. Osteoporosis is often associated with this condition [15]. One test that can distinguish patients with PPNAD from other primary adrenocortical lesions is cortisoluria after sequential suppression with low- and high-dose dexamethasone. In contrast to most patients with primary adrenocortical disease, who demonstrate no change in urinary cortisol, 70% of PPNAD patients have a paradoxical increase in urinary cortisol excretion [16]. The treatment of choice for PPNAD is bilateral adrenalectomy due to the high recurrence rate for primary adrenal disease [17]. Carney complex is a multiple neoplastic syndrome with autosomal dominant transmission, characterized by freckle-like cutaneous hyperpigmentation (lentiginosis), endocrine tumors [(PPNAD), testicular and/or thyroid tumors and acromegaly] and nonendocrine tumors, including cutaneous, cardiac, mammary, and osteochondral myxomas, among others. In the above case, the transthoracic echocardiogram of the patient on 03/18/2021 showed cavities of normal dimensions, preserved systolic and diastolic functions, no valve changes and no lentiginous skin areas and blue nevi, making the diagnosis of the syndrome less likely. The definitive diagnosis of Carney requires two or more main manifestations. Several related clinical components may suggest the diagnosis but not define it. The diagnosis can also be made if a key criterion is present and a first-degree relative has Carney or an inactivating mutation of the gene encoding PRKAR1A [18]. The adenoma is usually small in size (< 3 cm), similar to the nodules in this case; however, it is usually unilateral, with an insidious and mild evolution, especially in adult women over 35 years of age, producing only 1 steroid class. Carcinomas are usually large (> 6 cm), and only 10% are bilateral. They should be suspected mainly when the tumor presents with hypercortisolism associated with hyperandrogenism. They have a bimodal age distribution, with peaks in childhood and adolescence, as well as at the end of life [3]. Conclusion Early identification of Cushing’s syndrome, with measures based on the assessment of risks and benefits, remains the best way to prevent progression and reduce morbidity [2]. After 6 months of drug inhibition of steroidogenesis, blood pressure control and anti-osteoporotic therapy, the objective was to minimize the levels and deleterious metabolic effects of hypercortisolism, which could also harm the surgical procedure in the short and long term through infections, dehiscence, nonimmediate bed mobilization and cardiovascular events. Unilateral adrenalectomy was chosen, given the possibility of malignancy in a young patient and to avoid definitive surgical adrenal insufficiency if the adrenalectomy was bilateral. Despite the unavailability of genetic analysis for a precise etiological definition, it is possible to take efficient measures to avoid unnecessary consequences or damage. Availability of data and materials All data generated or analysed during this study are included in this published article [and its Additional file 1]. The datasets generated and/or analysed during the current study are available in the link https://ufjfedubr-my.sharepoint.com/:v:/g/personal/barbara_reis_ufjf_edu_br/EVpIR005sPZGlQvMJhIwSaUB0Hig4KOjhkG4D4cMggUwHA?e=Dk8tng. Abbreviations ACTH: Adrenocorticotropic hormone PPNAD: Bilateral primary pigmented nodular hyperplasia DXA: Dual energy X-ray absorptiometry GIP: Gastric inhibitory peptide GnRH: Gonadotropin-releasing hormone IL-1: Interleukin 1 BMD: Low bone mineral density LH: Luteinizing hormone MAS: McCune–Albright syndrome PRKAR1A: PKA regulatory 1-alpha subunit ADH: Vasopressin References Pedro AO, Plapler PG, Szejnfeld VL. Manual brasileiro de osteoporose: orientações práticas para os profissionais de saúde. 1st ed. São Paulo: Editora Clannad; 2021. ISBN 978-65-89832-00-3. Naguib R, Elkemary EZ, Elsharkawi KM. The severity of bone loss: a comparison between Cushing’s disease and Cushing’s syndrome. J Endocrinol Metab. 2023;13(1):33–8. https://doi.org/10.14740/jem857. Article Google Scholar Vilar L, et al. Endocrinologia Clínica. 6th ed. Rio de Janeiro: Guanabara Koogan; 2016. Google Scholar Wang D, Dang CX, Hao YX, Yu X, Liu PF, Li JS. Relationship between osteoporosis and Cushing syndrome based on bioinformatics. Medicine (Baltimore). 2022;101(43): e31283. Article CAS PubMed Google Scholar Williams TA, Reincke M. Management of Endocrine Disease: diagnosis and management of primary aldosteronism: the Endocrine Society guideline 2016 revisited. Eur J Endocrinol. 2018;179(1):R19–29. https://doi.org/10.1530/EJE-17-0990. Article CAS PubMed Google Scholar Compston J, Cooper A, Cooper C, Gittoes N, Gregson C, Harvey N, National Osteoporosis Guideline Group (NOGG), et al. UK clinical guideline for the prevention and treatment of osteoporosis. Arch Osteoporos. 2017;12(1):43. https://doi.org/10.1007/s11657-017-0324-5. Article CAS PubMed PubMed Central Google Scholar Nieman LK. Diagnosis of Cushing’s syndrome in the modern era. Endocrinol Metab Clin N Am. 2018;47(2):259–73. https://doi.org/10.1016/j.ecl.2018.02.001. Article Google Scholar Herr K, Muglia VF, Koff WJ, Westphalen AC. Imaging of the adrenal gland lesions. Radiol Bras. 2014;47(4):228–39. https://doi.org/10.1590/0100-3984.2013.1762. Article PubMed PubMed Central Google Scholar Hsiao HP, Kirschner LS, Bourdeau I, Keil MF, Boikos SA, Verma S, et al. Clinical and genetic heterogeneity, overlap with other tumor syndromes, and atypical glucocorticoid hormone secretion in adrenocorticotropin-independent macronodular adrenal hyperplasia compared with other adrenocortical tumors. J Clin Endocrinol Metab. 2009;94(8):2930–7. https://doi.org/10.1210/jc.2009-0516. Article CAS PubMed PubMed Central Google Scholar Mircescu H, Jilwan J, N’Diaye N, et al. Are ectopic or abnormal membrane hormone receptors frequently present in adrenal Cushing’s syndrome? J Clin Endocrinol Metab. 2000;85(10):3531–6. https://doi.org/10.1210/jcem.85.10.6865. Article CAS PubMed Google Scholar Miller BS, Auchus RJ. Evaluation and treatment of patients with hypercortisolism: a review. JAMA Surg. 2020;155(12):1152–9. https://doi.org/10.1001/jamasurg.2020.3280. Article PubMed Google Scholar Haddad NG, Eugster EA. Peripheral precocious puberty including congenital adrenal hyperplasia: causes, consequences, management and outcomes. Best Pract Res Clin Endocrinol Metab. 2019;33(3):101273. https://doi.org/10.1016/j.beem.2019.04.007. Article PubMed Google Scholar Bonnet-Serrano F, Bertherat J. Genetics of tumors of the adrenal cortex. Endocr Relat Cancer. 2018;25(3):R131–52. https://doi.org/10.1530/ERC-17-0361. Article CAS PubMed Google Scholar Carney JA, Young WF Jr. Primary pigmented nodular adrenocortical disease and its associated conditions. Endocrinologist. 1992;2:6. Article Google Scholar Stratakis CA, Sarlis N, Kirschner LS, Carney JA, Doppman JL, Nieman LK, et al. Paradoxical response to dexamethasone in the diagnosis of primary pigmented nodular adrenocortical disease. Ann Intern Med. 1999;131(8):585–91. https://doi.org/10.7326/0003-4819-131-8-199910190-00006. Article CAS PubMed Google Scholar Powell AC, Stratakis CA, Patronas NJ, Steinberg SM, Batista D, Alexander HR, et al. Operative management of Cushing syndrome secondary to micronodular adrenal hyperplasia. Surgery. 2008;143(6):750–8. https://doi.org/10.1016/j.surg.2008.03.022. Article PubMed Google Scholar Almeida MQ, Stratakis CA. Carney complex and other conditions associated with micronodular adrenal hyperplasias. Best Pract Res Clin Endocrinol Metab. 2010;24(6):907–14. https://doi.org/10.1016/j.beem.2010.10.006. Article CAS PubMed PubMed Central Google Scholar Hannah-Shmouni F, Stratakis CA. A gene-based classification of primary adrenocortical hyperplasias. Horm Metab Res. 2020;52(3):133–41. https://doi.org/10.1055/a-1107-2972. Article CAS PubMed Google Scholar Download references Acknowledgements Not applicable. Funding Not applicable. Author information Authors and Affiliations Serviço de Endocrinologia, Hospital Universitário da Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil Bárbara Oliveira Reis, Christianne Toledo Sousa Leal, Danielle Guedes Andrade Ezequiel, Ana Carmen dos Santos Ribeiro Simões Juliano, Flávia Lopes de Macedo Veloso, Leila Marcia da Silva, Lize Vargas Ferreira, Mariana Ferreira & Gabriel Zeferino De Oliveira Souza Contributions All the authors contributed to the conception and design of the work and have approved the submitted version. All authors read and approved the final manuscript. Corresponding author Correspondence to Bárbara Oliveira Reis. Ethics declarations Ethics approval and consent to participate Not applicable. Consent for publication Written informed consent was obtained from the patient for publication of this case report and any accompanying images. A copy of the written consent is available for review by the Editor-in-Chief of this journal. 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. Supplementary Information Additional file 1. Surgical removal of adrenal gland. Rights and permissions Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data. Reprints and Permissions From https://jmedicalcasereports.biomedcentral.com/articles/10.1186/s13256-023-03968-0
    1 point
  46. Abstract The association between empty sella turcica (EST) syndrome and Cushing's disease has been rarely reported. It is plausible to hypothesize that EST syndrome in association with Cushing's disease can be attributed to intracranial hypertension. In this case report, we present a 47-year-old male patient who presented with weight loss, fatigue, easy bruising, acanthosis nigricans, and skin creases hyperpigmentation. Investigations revealed hypokalemia and confirmed the diagnosis of Cushing's disease. Magnetic resonance imaging (MRI) brain showed a partial EST syndrome and a new pituitary nodule as compared with previous brain imaging. Transsphenoidal surgery was pursued and was complicated by cerebrospinal fluid leakage. This case reflects the rare association of EST syndrome and Cushing's disease, suggesting the increased risk of postoperative complications in this setting and the diagnostic challenge that EST syndrome imposes. We review the literature for a possible mechanism of this association. Introduction Cushing's disease is commonly caused by an adrenocorticotropic hormone (ACTH)-producing pituitary adenoma, which can be very challenging to be seen on brain magnetic resonance imaging (MRI) [1]. Empty sella turcica (EST) syndrome is a radiological diagnosis of apparently empty turcica secondary to outpouching of the arachnoid mater into the turcica, which can be attributed to intracranial hypertension (ICHTN). This can make the visual diagnosis of pituitary adenoma even more challenging in clinical practice. ICHTN has been also associated with Cushing's disease and might explain this infrequent association between EST and Cushing's disease [1]. EST syndrome can be either partial or complete, primary or secondary and has been seen infrequently with Cushing's disease. In this setting, not only that it is likely to obscure an underlying pituitary lesion, but also it does contribute to the risk of postoperative complications [2]. Case Presentation A 47-year-old male presented to the emergency department (ED) with slowly progressive generalized limb muscle weakness affecting both distal and proximal muscles over a few weeks and gait instability for three days prior to presentation. He also reported unintentional 40 pounds weight loss over the previous four months. Past medical history was significant for type II diabetes mellitus, hypothyroidism, hypertension, and dyslipidemia. In the ED, vital signs included a blood pressure of 140/90 mmHg, a heart rate of 66 beats per minute, a respiratory rate of 16 cycles per minute, and SpO2 of 97% on room air. Body mass index has decreased to 22 kg/m2 from a baseline of 26 kg/m2 one month prior. On the physical exam, he exhibited cachexia, easy bruising, acanthosis nigricans, and hyperpigmentation of skin creases. All other systems were negative. Complete metabolic panel and complete blood count were obtained showing hyperglycemia of 311 mg/dl, see Table 1. Further lab evaluation showed elevated salivary cortisol at 2.96 microgram/dl (reference range 0.094-1.551 mcg/dl), elevated 24-hour urinary free cortisol at 156 mcg/24 hour (reference 10-100 mcg/24h), positive overnight dexamethasone suppression test with serum cortisol at 2.8 mcg/dl (reference more than 2 mcg/dl), negative anti-adrenal antibodies, normal aldosterone, and elevated dehydroepiandrostenedione at 401 mcg/dl (reference 32-240 mcg/dl), with lack of suppression of the ACTH level at 35.1 pg/ml (reference 10-60 pg/ml). This confirmed the diagnosis of Cushing's disease. Variable Finding Reference Random glucose 311 Less than 200 mg/dl Sodium 141 137-145 mmol/L Potassium 2.5 3.5-5.1 mmol/L Chloride 96 98-107 mmol/L Bicarbonate 32 22-30 mmol/L Blood urea nitrogen 32 9-20 mg/dl Creatinine 0.52 0.66-1.25 mg/dl Calcium 8.7 8.6-10.3 mg/dl Total protein 5.5 6.5-8.5 g/dl Albumin 3.3 3.5-5 g/dl Total bilirubin 0.6 0.2-1.3 mg/dl Alkaline phosphatase 115 38-126 U/L Aspartate transaminase 17 17-59 U/L Alanine transaminase 39 Less than 49 U/L White blood cell count 10x10^3 cells/mcl 4-10x1063 cells/mcl Hemoglobin 15.3 13.7-17.5 g/dl Platelet 281 150-400x10^3 cells/mcl Table 1: Lab Findings Computed tomography (CT) scan of the head was unremarkable. CT scan of the chest was also unremarkable. CT scan of abdomen and pelvis showed no adrenal mass. Ultrasound of the kidneys was unremarkable. Pituitary MRI brain protocol for adenoma showed a partial EST, shortening within neurohypophysis and a new 10 x 8 x 4 mm nodule along the floor of pituitary sella as compared to MRI four years ago (Figure 1). Figure 1: Magnetic Resonance Imaging (MRI) Brain MRI brain showing partially empty sella turcica syndrome ( black arrow) with a small nodule at the floor of the turcica (white arrow). The diagnosis of Cushing’s disease was confirmed, and the patient underwent trans-sphenoidal resection of pituitary adenoma. Histological examination showed positive CAM 5.2, positive chromogranin, and ACTH immunostains. The patient presented to the ED five days after discharge home. He stated that he noticed drainage from the nose that transitioned from bloody to clear fluid and has been increasing in quantity for two days with associated intermittent headaches since the surgery. He was afebrile with stable vital signs. No signs of infection were noted on basic labs. These were significant only for mild asymptomatic hyponatremia of 131 mmol/L and hypokalemia of 3.3 mmol/L. The patient was diagnosed with cerebrospinal fluid (CSF) leakage and had a lumbar drain trial. The trial was unsuccessful after several days, and the patient underwent a transnasal endoscopic repair of CSF rhinorrhea using nasoseptal flaps. At an outpatient follow-up one month and three months after the surgery, prior lab abnormalities including hypokalemia, hyponatremia, and hyperglycemia resolved. No further evidence of CSF leakage was appreciated, and he remained asymptomatic. Discussion EST syndrome is characterized by herniation of the subarachnoid space into the intrasellar space with compression of the pituitary gland into the posteroinferior wall [3]. This is likely to obscure the presence of underlying pituitary mass. The incidence of EST syndrome in the general population is estimated at 20%. The association between EST syndrome and Cushing's disease has been reported infrequently. A retrospective study of 68 patients with Cushing's disease found that 16% of these have EST syndrome [3]. Cushing's disease usually results from pituitary adenomas secreting ACTH, and even the smallest microadenomas can produce a systemic disease. These microadenomas can be very difficult to recognize on brain MRI [4]. This is complicated in EST syndrome and even further with the possibility of ectopic ACTH production. A retrospective study of 197 patients diagnosed with Cushing's disease concluded that EST syndrome is associated with higher prevalence of MRI-negative Cushing's disease. This was attributed to ICHTN and pituitary gland compression [1]. Although surgery is curative in 70-90% of cases, EST syndrome was found to have higher risk of postoperative complications among those with Cushing's disease including diabetes insipidus, hypopituitarism, and CSF leakage [3]. This is usually because in the case of MRI-negative Cushing's disease with total EST syndrome, empiric surgical exploration is sought after inferior petrosal sampling confirms the pituitary origin of excess ACTH, and postoperative remission indicates adequate tumor resection [2]. This entails a higher chance of uncertainty and injury to healthy pituitary tissue. EST syndrome can be either primarily due to defects in the sellar diaphragm or anatomical variant or secondary to ICHTN. EST syndrome has been reported in association with many conditions associated with elevated intracranial pressure including tumors, thrombosis, meningitis, hydrocephalus, and Arnold-Chiari malformation [5]. Reversal of EST syndrome has been reported in those with idiopathic ICHTN with therapy by acetazolamide, ventriculoperitoneal shunt, and lumbar puncture [6,7]. A study has shown correlation between CSF circulation impairment or blockage and EST syndrome [8]. The incidence of EST syndrome in association with symptomatic intracranial hypertension is variable and ranges from 2.5% for total EST syndrome to 94% for partial EST syndrome [9]. Impaired CSF circulation and dynamics have been reported in 77% of patients with EST syndrome [10]. In addition to intracranial hypertension, EST syndrome has also been described in association with obesity, meningioma, pediatric nevoid basal cell carcinoma, therapy for growth hormone deficiency and even in healthy individuals [9]. Lack of symptoms of intracranial hypertension in this patient does not rule it out as intracranial hypertension in EST syndrome represents a spectrum that ranges from asymptomatic, milder intracranial hypertension to symptomatic intracranial hypertension with headache, visual disturbance, and papilledema [10]. This explains the fact that only 8-14% of EST syndrome progress to symptomatic ICHTN, while symptomatic ICHTN has been associated with EST syndrome in 94% of cases. ICHTN has been seen in association with disturbance of the hypothalamic-pituitary-adrenal axis. This has been reported after surgical and medical treatment of Cushing's disease, withdrawal of long-term steroid therapy, initial presentation of Addison’s disease, or relative glucocorticoids deficiency [11]. Cortisol excess increases CSF production and reduces its absorption, hence increasing intracranial pressure [12]. Another possible mechanism is the expression of both mineralocorticoid responsive epithelial sodium channel receptors on the basolateral membrane of the CSF producing epithelial cells of the choroid plexus as well as the expression of 11-beta hydroxysteroid dehydrogenase type 1 enzyme, which is a bidirectional enzyme that mainly functions to convert the inactive cortisone to active cortisol. These mechanisms play a role in maintaining the balance between CSF production and absorption [13,14]. In this case, the patient presented some clinical findings that are rarely associated with Cushing's disease, combined with a radiological feature that masked the true diagnosis. Our patient presented with significant weight loss, rather than central obesity, which is normally associated with Cushing’s disease. Although possible, the increase in ACTH due to Cushing's disease is not sufficient to cause hyperpigmentation, which is a classical finding of Addison's disease, where the entire adrenal cortex is usually affected due to an autoimmune destruction; however, the zona glomerulosa of the adrenal cortex produces aldosterone and its deficiency would lead to hyperkalemia [15]. Our patient presented with both hyperpigmentation and hypokalemia. Conclusions EST syndrome is an uncommon radiological finding of apparently EST that has been reported in association with ICHTN. The latter has also been seen in association with Cushing's disease/syndrome. This is likely to result from glucocorticoid excess-induced change in CSF flow dynamics. EST has been infrequently described in association with Cushing's disease. This association has a clinical implication as it is likely to obscure the visualization of pituitary lesions responsible for Cushing's disease, contribute to diagnostic uncertainty, and increase the risk of healthy pituitary tissue injury and the risk of postoperative complications including CSF leakage. References Himes BT, Bhargav AG, Brown DA, Kaufmann TJ, Bancos I, Van Gompel JJ: Does pituitary compression/empty sella syndrome contribute to MRI-negative Cushing's disease? A single-institution experience. Neurosurg Focus. 2020, 48:E3. 10.3171/2020.3.FOCUS2084 Sun Y, Sun Q, Fan C, et al.: Diagnosis and therapy for Cushing's disease with negative dynamic MRI finding: a single-centre experience. Clin Endocrinol (Oxf). 2012, 76:868-76. 10.1111/j.1365-2265.2011.04279.x Manavela MP, Goodall CM, Katz SB, Moncet D, Bruno OD: The association of Cushing's disease and primary empty sella turcica. Pituitary. 2001, 4:145-51. 10.1023/a:1015310806063 Chatain GP, Patronas N, Smirniotopoulos JG, et al.: Potential utility of FLAIR in MRI-negative Cushing's disease. J Neurosurg. 2018, 129:620-8. 10.3171/2017.4.JNS17234 Friedman DI, Jacobson DM: Diagnostic criteria for idiopathic intracranial hypertension. Neurology. 2002, 59:1492-5. 10.1212/01.wnl.0000029570.69134.1b Triggiani V, Giagulli VA, Moschetta M, Guastamacchia E: An unusual case of reversible empty sella. Endocr Metab Immune Disord Drug Targets. 2016, 16:154-6. 10.2174/1871530315666151001141507 Wind JJ, Lonser RR, Nieman LK, DeVroom HL, Chang R, Oldfield EH: The lateralization accuracy of inferior petrosal sinus sampling in 501 patients with Cushing's disease. J Clin Endocrinol Metab. 2013, 98:2285-93. 10.1210/jc.2012-3943 Brismar K, Bergstrand G: CSF circulation in subjects with the empty sella syndrome. Neuroradiology. 1981, 21:167-75. 10.1007/BF00367338 Ranganathan S, Lee SH, Checkver A, Sklar E, Lam BL, Danton GH, Alperin N: Magnetic resonance imaging finding of empty sella in obesity related idiopathic intracranial hypertension is associated with enlarged sella turcica. Neuroradiology. 2013, 55:955-61. 10.1007/s00234-013-1207-0 Maira G, Anile C, Mangiola A: Primary empty sella syndrome in a series of 142 patients. J Neurosurg. 2005, 103:831-6. 10.3171/jns.2005.103.5.0831 Zada G, Tirosh A, Kaiser UB, Laws ER, Woodmansee WW: Cushing's disease and idiopathic intracranial hypertension: case report and review of underlying pathophysiological mechanisms. J Clin Endocrinol Metab. 2010, 95:4850-4. 10.1210/jc.2010-0896 Sinclair AJ, Ball AK, Burdon MA, Clarke CE, Stewart PM, Curnow SJ, Rauz S: Exploring the pathogenesis of IIH: an inflammatory perspective. J Neuroimmunol. 2008, 201:212-20. 10.1016/j.jneuroim.2008.06.029 Sinclair AJ, Onyimba CU, Khosla P, et al.: Corticosteroids, 11beta-hydroxysteroid dehydrogenase isozymes and the rabbit choroid plexus. J Neuroendocrinol. 2007, 19:614-20. 10.1111/j.1365-2826.2007.01569.x Amin MS, Wang HW, Reza E, Whitman SC, Tuana BS, Leenen FH: Distribution of epithelial sodium channels and mineralocorticoid receptors in cardiovascular regulatory centers in rat brain. Am J Physiol Regul Integr Comp Physiol. 2005, 289:R1787-97. 10.1152/ajpregu.00063.2005 Stratakis CA: Skin manifestations of Cushing's syndrome. Rev Endocr Metab Disord. 2016, 17:283-6. 10.1007/s11154-016-9399-3 From https://www.cureus.com/articles/161111-cushings-disease-associated-with-partially-empty-sella-turcica-syndrome-a-case-report#!/
    1 point
  47. Abstract Summary Cushing’s syndrome due to ectopic adrenocorticotropic hormone (ACTH) secretion (EAS) by a pheochromocytoma is a challenging condition. A woman with hypertension and an anamnestic report of a ‘non-secreting’ left adrenal mass developed uncontrolled blood pressure (BP), hyperglycaemia and severe hypokalaemia. ACTH-dependent severe hypercortisolism was ascertained in the absence of Cushingoid features, and a psycho-organic syndrome developed. Brain imaging revealed a splenial lesion of the corpus callosum and a pituitary microadenoma. The adrenal mass displayed high uptake on both 18F-FDG PET/CT and 68Ga-DOTATOC PET/CT; urinary metanephrine levels were greatly increased. The combination of antihypertensive drugs, high-dose potassium infusion, insulin and steroidogenesis inhibitor normalized BP, metabolic parameters and cortisol levels; laparoscopic left adrenalectomy under intravenous hydrocortisone infusion was performed. On combined histology and immunohistochemistry, an ACTH-secreting pheochromocytoma was diagnosed. The patient's clinical condition improved and remission of both hypercortisolism and catecholamine hypersecretion ensued. Brain magnetic resonance imaging showed a reduction of the splenial lesion. Off-therapy BP and metabolic parameters remained normal. The patient was discharged on cortisone replacement therapy for post-surgical hypocortisolism. EAS due to pheochromocytoma displays multifaceted clinical features and requires prompt diagnosis and multidisciplinary management in order to overcome the related severe clinical derangements. Learning points A small but significant number of cases of adrenocorticotropic hormone (ACTH)-dependent Cushing’s syndrome are caused by ectopic ACTH secretion by neuroendocrine tumours, which is usually associated with severe hypercortisolism causing severe clinical and metabolic derangements. Ectopic ACTH secretion by a pheochromocytoma is exceedingly rare but can be life-threatening, owing to the simultaneous excess of both cortisol and catecholamines. The combination of biochemical and hormonal testing and imaging procedures is mandatory for the diagnosis of ectopic ACTH secretion, and in the presence of an adrenal mass, the possibility of an ACTH-secreting pheochromocytoma should be taken into account. Immediate-acting steroidogenesis inhibitors are required for the treatment of hypercortisolism, and catecholamine excess should also be appropriately managed before surgical removal of the tumour. A multidisciplinary approach is required for the treatment of this challenging entity. Keywords: Adult; Female; White; Italy; Adrenal; Pituitary; Unique/unexpected symptoms or presentations of a disease; May; 2023 Background Cushing’s syndrome (CS) is a rare endocrine disease characterized by high levels of glucocorticoids; it increases morbidity and mortality due to cardiovascular and infectious diseases (1, 2, 3). To diagnose CS, adrenocorticotropic hormone (ACTH)-dependent disease must be distinguished from ACTH-independent disease, and pituitary ACTH production from ectopic production. About 20% of ACTH-dependent cases arise from ectopic ACTH secretion (EAS) (2, 3, 4). EAS is most often due to aberrant ACTH production by small-cell lung carcinoma or neuroendocrine tumours originating in the lungs or gastrointestinal tract; this, in turn, strongly increases cortisol production by the adrenal glands (3, 4, 5). Since the first-line treatment of EAS is the surgical removal of the ectopic ACTH-secreting tumour, its prompt and accurate localization is crucial. Rapid cortisol reduction by means of immediate-acting steroidogenesis inhibitors (4) is mandatory in order to treat the related endocrine, metabolic and electrolytic derangements. EAS by a pheochromocytoma is exceedingly rare and can be life-threatening. We describe the case of a woman with hypertension and a known ‘non-secreting’ left adrenal mass, who manifested uncontrolled blood pressure (BP), hyperglycaemia, hypokalaemia and psycho-organic syndrome associated with damage of the splenium of the corpus callosum. These findings were eventually seen to be related to an ACTH-secreting left pheochromocytoma, which was ascertained by hormonal evaluation and morphological and functional imaging assessment and confirmed by histopathology/immunostaining. Hormonal hypersecretion resolved after adrenalectomy and metabolic derangements normalized. Case presentation A 72-year-old woman with hypertension was taken to the emergency department because of increased BP (200/100 mm Hg). High BP (190/100 mmHg) was confirmed, whereas oxygen saturation (98%), heart rate (84 bpm) and lung and abdomen examination were normal. Electrocardiogram and chest x-ray were unremarkable. Captopril 50 mg orally, followed by intramuscular clonidine, normalized BP. The patient looked thin and reported significant weight loss (10 kg) over the previous 6 months; she was on antihypertensive therapy with bisoprolol 5 mg/day and irbesartan 150 mg/day, and ezetimibe 10 mg/day for dyslipidaemia. The patient’s records included a previous diagnosis in another hospital of normofunctioning multinodular goitre and a 2.5 cm-left solid inhomogeneous adrenal mass with well-defined margins, which was found on CT performed 6 years earlier during the work-up for hypertension. On the basis of hormonal data and absent uptake on 123I metaiodobenzylguanidine scintigraphy, the adrenal lesion had been deemed to be non-functioning and follow-up had been advised. Unfortunately, only initial cortisol (15.7 μg/dL) and 24-h urine-free cortisol (UFC) levels (32.5 μg/24 h) were retrievable; both proved normal. Investigations Blood chemistry showed neutrophilic leucocytosis, hyperglycaemia with increased glycated haemoglobin, severe hypokalaemia and metabolic alkalosis (Table 1). Potassium infusion (50 mEq in 500 mL saline/24 h) was rapidly started, together with a subcutaneous rapid-acting insulin analogue and prophylactic enoxaparin. The patient experienced mental confusion, hallucinations and restlessness; non-enhanced computed tomography (CT) of the brain revealed a hypodense area of the splenium of the corpus callosum, possibly due to metabolic damage (Fig. 1A). View Full Size Figure 1 Non-enhanced CT showing a hypodense area of the splenium of the corpus callosum (arrows), without mass effect (A, axial view). Contrast-enhanced MR image showing a hypointense pituitary lesion (arrow) which enhances more slowly than normal pituitary parenchyma, deemed suspicious for microadenoma (B, coronal view). FLAIR MR image showing hyperintense signal of the splenium of the corpus callosum (asterisk), which partially extended to the crux of the left fornix (arrow) (C, axial view). As the lesion showed no restricted diffusion on DWI (D, axial view), an ischaemic lesion was excluded. A progressive reduction in the extension of the hyperintense signal in the splenium of the corpus callosum (arrowheads) and in the crux of the left fornix (arrows) was observed on FLAIR MR images (2 months (E); 3 months (F); axial view). CT, computed tomography; DWI, diffusion-weighted imaging; FLAIR, fluid-attenuated inversion recovery; MR, magnetic resonance. Citation: Endocrinology, Diabetes & Metabolism Case Reports 2023, 2; 10.1530/EDM-22-0308 Download Figure Download figure as PowerPoint slide Table 1 Hormonal and biochemical evaluation of patient throughout hospitalization and follow-up. Normal range On hospital admission After surgery 10 days 2 months 3 months 6 months 9 months 12 months 16 months ACTH (pg/mL) 9–52 551 7 37 50 29.5 26 40.9 52 Morning cortisol† (µg/dL) 7–19.2 63.4 14 5.1 3.5 3.8 4.2 7.2 12.8 After 1 mg overnight dexamethasone  ACTH 583  Cortisol 60 DHEAS (µg/dL) 9.4–246 201 24-h urinalysis (µg/24 h)  Adrenaline 0–14.9 95.5  Noradrenaline 0–66 1133  Metanephrine 74–297 1927  Normetanephrine 105–354 1133 Chromogranin A 0–108 290 Renin (supine) (µU/mL) 2.4–29 3.9 14.6 Aldosterone (supine) (ng/dL) 3–15 3.4 12.5 LH (mIU/mL)* > 10 0.3 65.8 FSH (mIU/mL)* > 25 1.9 116 PRL (ng/mL) 3–24 13.7 FT4 (ng/dL) 0.9–1.7 1.1 1.2 FT3 (pg/mL) 1.8–4.6 1.1 2.7 TSH (µU/mL) 0.27–4.2 0.23 1.3 PTH (pg/mL) 15–65 166 Calcium (mg/dL) 8.2–10.2 8.2 Calcitonin (pg/mL) 0–10 1 Glycaemia (mg/dL) 60–110 212 69 73 83 Potassium (mEq/L) 3.5–5 2.4 3.3 3.9 4.2 3.7 5 4.4 3.9 Leucocytes (K/µL) 4.0–9.3 15.13 HbA1c (mmol/mol) 20–42 55 30 HCO3− (mEq/L) 22–26 41.8 *For menopausal age; †07:00–10:00 h. The patient was transferred to the internal medicine ward. Although potassium infusion was increased to 120 mEq/day, serum levels did not normalize; a mineralocorticoid receptor antagonist (potassium canreonate) was therefore introduced, but the effect was partial. In order to control BP, the irbersartan dose was increased (300 mg/day) and amlodipine (10 mg/day) was added. The combination of severe hypertension, newly occurring diabetes and resistant hypokalaemia prompted us to hypothesize a common endocrine aetiology. A thorough hormonal array showed very high ACTH and cortisol levels, whereas supine renin and aldosterone levels were in the low-normal range (Table 1). Since our patient proved repeatedly non-compliant with 24-h urine collection, UFC could not be measured. After an overnight 1 mg dexamethasone suppression test, cortisol levels remained unchanged, whereas ACTH levels slightly increased (Table 1). Notably, the patient showed no Cushingoid features. Gonadotropin levels were inappropriately low for the patient’s age; FT4 levels were normal, whereas FT3 and thyroid-stimulating hormone (TSH) levels were reduced and calcitonin levels were normal (Table 1). HbA1c levels were increased (Table 1). Finally, secondary hyperparathyroidism, associated with low-normal calcium levels and reduced vitamin D levels, was found (Table 1). Brain contrast-enhanced magnetic resonance (MR) imaging revealed a 5-mm median posterior pituitary microadenoma (Fig. 1B) and a hyperintense lesion of the splenium of the corpus callosum (Fig. 1C). Diffusion-weighted MR images of the lesion showed no restricted diffusion (Fig. 1D), thus excluding an ischaemic origin. Petrosal venous sampling for ACTH determination at baseline and after CRH stimulation was excluded, as it was deemed a high-risk procedure, given the patient's poor condition. Since the ACTH and cortisol levels were greatly increased and were associated with severe hypokalaemia, EAS was hypothesized; total-body contrast-enhanced CT revealed the left adrenal mass (3 cm), which showed regular margins and heterogeneous enhancement (Fig. 2A and B) and measured 25 Hounsfield units. There was no evidence of adrenal hyperplasia in the contralateral adrenal gland. The adrenal mass showed intense tracer uptake on both 18F-FDG PET/CT (Fig. 2C and D), suggestive of adrenal malignancy or functioning tumour, and 68Ga-DOTATOC PET/CT (Fig. 3), which is characteristic of a neuroendocrine lesion. No other sites of suspicious tracer uptake were detected. View Full Size Figure 2 Contrast-enhanced abdominal computed tomography showing a 3-cm left adrenal mass (arrow) with well-defined margins and inhomogeneus enhancement, deemed compatible with an adenoma (A, coronal view; B, axial view). The adrenal mass showed high uptake (SUV max: 7.3) on 18F-FDG PET/CT (C, coronal view; D, axial view). Citation: Endocrinology, Diabetes & Metabolism Case Reports 2023, 2; 10.1530/EDM-22-0308 Download Figure Download figure as PowerPoint slide View Full Size Figure 3 The left adrenal mass displaying very high uptake (SUV max: 40) on 68Ga-DOTATOC PET/CT (arrow, axial view). Citation: Endocrinology, Diabetes & Metabolism Case Reports 2023, 2; 10.1530/EDM-22-0308 Download Figure Download figure as PowerPoint slide Bisoprolol was withdrawn, and 24-h urinary catecholamine, metanephrine and normetanephrine levels proved significantly increased, as were chromogranin A levels (Table 1). In sum, an ACTH-secreting pheochromocytoma was suspected and the pituitary microadenoma was deemed a likely incidental finding. The patient’s mental state worsened, fluctuating from sopor to restlessness, which required parenteral neuroleptics and restraint. An electroencephalogram revealed a specific slowdown of cerebral electrical activity. Following rachicentesis, the cerebrospinal fluid showed pleocytosis (lympho-monocytosis), whereas a culture test and polymerase chain reaction for common neurotropic agents were negative. The neurologist hypothesized a psycho-organic syndrome secondary to severe metabolic derangement. Intravenous ampicillin, acyclovir and B vitamins were empirically started. The patient was transferred to the subintensive unit, where a nasogastric tube and central venous catheter were inserted, and enteral nutrition was started. Treatment Ketoconazole was started at a dosage of 200 mg twice daily; both cortisol and ACTH levels significantly decreased over a few days (Fig. 4), with a progressive decrease in glucose levels and normalization of potassium levels and BP on therapy. Subsequently, ketoconazole was titrated to 600 mg/day owing to a new increase in cortisol levels, which eventually normalized (Fig. 4). Of note, ACTH levels partially decreased on ketoconazole treatment (Fig. 4). View Full Size Figure 4 ACTH and cortisol levels throughout the patient’s hospitalization and follow-up. Citation: Endocrinology, Diabetes & Metabolism Case Reports 2023, 2; 10.1530/EDM-22-0308 Download Figure Download figure as PowerPoint slide Doxazosin 2 mg/day was added and the patient's systolic BP blood settled at around 100 mm Hg; after a few days, bisoprolol was restarted. Contrast-enhanced MR showed a partial reduction of the hyperintense splenial lesion (Fig. 1E). Despite the severe clinical condition and the high risks of adrenal surgery, the patient’s relatives strongly requested the procedure and laparoscopic left adrenalectomy was planned. Alpha-blocker and fluid infusion were continued, ketoconazole was withdrawn the day before surgery, and a 100 mg IV bolus of hydrocortisone was administered just before the operation, followed by 200 mg/day, at first in continuous infusion, then as a 100 mg bolus every 8 h. After the removal of the left adrenal mass, noradrenaline infusion was required, owing to the occurrence of severe hypotension. Outcome and follow-up Pathology revealed a 2.5 cm reddish-brown encapsulated tumour, which was compatible with pheochromocytoma (Fig. 5A and B); ACTH immunostaining was positive in about 30% of tumour cells (Fig. 5C). This confirmed the diagnostic hypothesis of an ACTH-secreting pheochromocytoma. The tumour was stained for Chromogranin A (Fig. 5D). There were no signs of adrenal cortex hyperplasia in the resected gland. Thorough germinal genetic testing, comprising the commonest pheochromocytoma/paraganglioma genes: CDKN1B, KIF1B, MEN1, RET, SDHA, SDHB, SDHC, SDHD, SDHAF2 and TMEM127, was negative. View Full Size Figure 5 Histological images of adrenal pheochromocytoma: the tumour is composed of well-defined nests of cells (‘zellballen’) (A; haematoxylin-eosin stain (HE), ×20) with pleomorphic nuclei with prominent nucleoli, basophilic or granular amphophilic cytoplasm (B; HE, ×40). The mitotic index was low: 1 mitosis per 30 high-power fields, and Ki-67 was 1%. On immunohistochemistry, cytoplasmatic ACTH staining was found in about 30% of tumour cells (C; ×20), whereas most tumour cells were stained for chromogranin A (D; ×20). Citation: Endocrinology, Diabetes & Metabolism Case Reports 2023, 2; 10.1530/EDM-22-0308 Download Figure Download figure as PowerPoint slide One week after surgery ACTH levels had dropped to a low-normal value: 7 pg/mL, and cortisol levels (before morning hydrocortisone bolus administration) were normal: 14 µg/dL (Fig. 4). The patient’s clinical status slowly improved and the nasogastric tube was removed; intravenous hydrocortisone was carefully tapered until withdrawal and high-dose oral cortisone acetate (62.5 mg/day) was started. This dose was initially required since BP remained low (systolic: 90 mm Hg); thereafter, cortisone was reduced to 37.5 mg/day. Plasma cortisol levels before morning cortisone administration were reduced (Fig. 4). A new MR of the brain showed a further partial reduction of the splenial lesion (Fig. 1F). The patient was discharged with normal off-therapy BP and metabolic parameters. During follow-up, she fully recovered, and BP and metabolic parameters remained normal. Gonadotropin levels became adequate for the patient’s age, and TSH and renin/aldosterone levels normalized (Table 1). Hypoadrenalism, however, persisted for more than 1 year; as the last hormonal evaluation, 16 months after surgery, showed normal baseline cortisol levels, the cortisone dose was tapered (12.5 mg/day) and further hormonal examination was scheduled (Table 1). ACTH and cortisol levels throughout the patient’s hospitalization and follow-up are shown in Fig. 4. Discussion The diagnosis of EAS is challenging and requires two steps: confirmation of increased ACTH and cortisol levels and anatomic distinction from pituitary sources of ACTH overproduction. Besides metabolic derangements (hyperglycaemia, hypertension), EAS-related severe hypercortisolism may cause profound hypokalaemia (3, 4, 5). In our patient, the combination of worsening hypertension, newly occurring diabetes and resistant hypokalaemia raised the suspicion of a common endocrine cause. ACTH-dependent severe hypercortisolism was ascertained, and subsequent brain MR revealed a pituitary microadenoma. The diagnosis of CS requires the combination of two abnormal test results: 24-h UFC, midnight salivary cortisol and/or abnormal 1 mg dexamethasone suppression testing (2, 6). ACTH evaluation (low/normal-high) is fundamental to tailoring the imaging technique. The very high cortisol levels found in our patient were unchanged after overnight dexamethasone testing, whereas UFC could not be assessed owing to the lack of compliance with urine collection. The accuracy of the UFC assays, however, may be impaired by cortisol precursors and metabolites. Salivary cortisol assessment was not performed since the specific assay is not available in our hospital. The combination of ACTH-dependent severe hypercortisolism and hypokalaemia prompted us to suspect EAS. The differential diagnosis between pituitary and ectopic ACTH-dependent CS involves high-dose (8 mg) dexamethasone suppression testing, which has relatively low diagnostic accuracy (6). Owing to the patient's very high cortisol levels and severe hypokalaemia, this testing was not performed, on account of the risks of administering corticosteroids in a patient already exposed to excessive levels (6). Furthermore, owing to the increase in ACTH levels observed after overnight dexamethasone testing, we postulated the possible occurrence of glucocorticoid-driven positive feedback on ACTH secretion, which has been described in EAS, including cases of pheochromocytoma (7). Finally, in the case of EAS suspected of being caused by pheochromocytoma, we do not recommend performing high-dose dexamethasone suppression testing, owing to the risk of triggering a catecholaminergic crisis (8). The dynamic tests commonly used to distinguish patients with EAS from those with Cushing's disease are the CRH stimulation test and the desmopressin stimulation test, either alone or in combination with CRH testing (6). Owing to the rapid worsening of our patient’s condition, dynamic testing was not done; however, the clinical picture and hormonal/biochemical data were suggestive of EAS. EAS is mainly (45–50%) due to neuroendocrine tumours, mostly of the lung (small-cell lung cancer and bronchial tumours), thymus or gastrointestinal tract; however, up to 20% of ACTH-secreting tumours remain occult (3, 4, 5). ACTH-secreting pheochromocytomas are responsible for about 5% of cases of EAS (3, 4, 9, 10). Indeed, this rate ranges widely, from 2.5% (11) to 15% (12), according to the different case series. Patients with EAS due to pheochromocytoma present with severe CS, overt diabetes mellitus, hypertension and hypokalaemia (3); symptoms of catecholamine excess may be unapparent (3), making the diagnosis more challenging. A recent review of 99 patients with ACTH- and/or CRH-secreting pheochromocytomas found that the vast majority displayed a Cushingoid phenotype (10); by contrast, another review of 24 patients reported that typical Cushingoid features were observed in only 30% of patients, whereas weight loss was a prevalent clinical finding (13). We hypothesized that the significant weight loss reported by our patient was largely due to the hypermetabolic state induced by catecholamines, which directly reduce visceral and subcutaneous fat, as recently reported (14). Our patient showed no classic stigmata of CS, owing to the rapid onset of severe hypercortisolism (10, 13), whereas she had worsening hypertension and newly occurring diabetes mellitus, which were related to both cortisol and catecholamine hypersecretion; hypokalaemia was deemed to be secondary to severe hypercortisolism. Indeed, greatly increased cortisol levels act on the mineralocorticoid receptors of the distal tubule after saturating 11β-hydroxysteroid dehydrogenase type 2, leading to hypokalaemia (4). Consequently, hypokalaemia is much more common (74–95% of patients) in EAS than in classic Cushing’s disease (10%) (3, 4, 10). This apparent mineralocorticoid excess suppresses renin and aldosterone secretion, as was ascertained in our patient. In this setting, the most effective way to manage hypokalaemia is to treat the hypercortisolism itself by administering immediate-acting steroidogenesis inhibitors, combined with potassium infusion and a mineralocorticoid receptor-antagonist (e.g. spironolactone) at an appropriate dosage (100–300 mg/day) (4). In ACTH-secreting pheochromocytoma, cortisol hypersecretion potentiates catecholamine-induced hypertension by stimulating the phenol-etholamine-N-methyl–transferase enzyme, which transforms noradrenaline to adrenaline (4). Indeed, in our patient, the significant ketoconazole-induced reduction in cortisol secretion led to satisfactory BP control on antihypertensive drugs. After the biochemical diagnosis of pheochromocytoma, a selective alpha-blocker was added, and after a few days, a beta-blocker was restarted in order to control reflex tachycardia (15). Our patient had greatly increased ACTH levels (>500 pg/mL) associated with very high cortisol levels (>60 µg/dL), which, together with the finding of hypokalaemia, prompted us to hypothesize EAS. With regard to these findings, ACTH levels are usually higher (>200 pg/mL) in patients with EAS than in those with CS due to a pituitary adenoma; however, considerable overlapping occurs (3, 11, 16). Most patients with ACTH-secreting pheochromocytomas in those series had ACTH levels >300 pg/mL, and a few had normal ACTH levels (9), thus complicating the diagnosis. In addition, patients with EAS usually have higher cortisol levels than those with ACTH-secreting adenomas (3, 11). In our patient, the left adrenal mass was deemed the culprit of EAS, and owing to very high urinary metanephrine levels, a pheochromocytoma was suspected. It can be assumed that the adrenal tumour, which was anamnestically reported as ‘non-secreting’, but on which only part of the initial hormonal data were available, was actually a pheochromocytoma at the time of the first diagnosis but displayed a silent clinical and hormonal behaviour. The mass subsequently showed significant uptake on both 18F-FDG PET/CT and 68Ga-DOTATOC PET/CT (4, 5). It is claimed that 68Ga-DOTATOC PET/CT provides a high grade (90%) of sensitivity and specificity in the diagnosis of tumours that cause EAS (4, 5); nevertheless, a recent systematic review reported much lower sensitivity (64%), which increased to 76% in histologically confirmed cases (17). In patients with EAS, immediate-acting steroidogenesis inhibitors are required in order to achieve prompt control of severe hypercortisolism (4). Ketoconazole is one of the drugs of choice since it inhibits adrenal steroidogenesis at several steps. In our patient, ketoconazole rapidly reduced cortisol levels to normal values, without causing hepatic toxicity (4). Moreover, ketoconazole proved effective at a moderate dosage (600 mg/day), which falls within the mean literature range (18, 19). However, dosages up to 1200–1600 mg/day are sometimes required in severe cases (usually EAS) (18, 19). Speculatively, our results might reflect an enhanced inhibitory action of ketoconazole at the adrenal level, which was able to override the strong ectopic ACTH stimulation. In addition, the finding that, following cortisol reduction, ACTH levels paradoxically decreased suggests an additive and direct effect of the drug. This effect has been observed in a few patients with EAS (20) and is supported by in vitro studies showing a direct anti-proliferative and pro-apoptotic effect of ketoconazole on ectopic ACTH secretion by tumours (21). Finally, the reduction in ACTH levels during treatment with steroidogenesis inhibitors prompts us to postulate the presence of glucocorticoid-driven positive feedback on ACTH secretion, as already described in neuroendocrine tumours (7, 20, 21). The coexistence of EAS and ACTH-producing pituitary adenoma is very rare but must be taken into account. In our case, we deemed the pituitary mass found on MR to be a non-secreting microadenoma. This hypothesis was strengthened by the finding that, following exeresis of the ACTH-secreting pheochromocytoma, ACTH normalized, hypercortisolism vanished and pituitary function recovered. These findings suggest that: (i) altered pituitary function at the baseline was secondary to the inhibitory effect of hypercortisolism; (ii) the excessive production of cortisol was driven by ACTH overproduction outside the pituitary gland, specifically within the adrenal gland tumour. In our patient, a few days after surgery, morning cortisol levels before hydrocortisone bolus administration were ‘normal’. Owing to both the half-life of hydrocortisone (8–12 h) and the supraphysiological dosage used, it is likely that a residual part of the drug, which cross-reacts in the cortisol assay, was still circulating at the time of blood collection, thus resulting in ‘normal’ cortisol values. Following the switch to oral cortisone, cortisol levels before therapy were low, thus confirming post-surgical hypocortisolism. Hypocortisolism remained throughout the first year after surgery, and glucocorticoid therapy was continued. Sixteen months after surgery, baseline cortisol levels returned to the normal range; cortisone therapy was therefore tapered and a further hormonal check was scheduled. Assessment of the cortisol response to ACTH stimulation testing would be helpful in order to check the resumption of the residual adrenal function. A peculiar aspect of our case was the occurrence of a psycho-organic syndrome together with the finding of a splenial lesion on brain imaging, which was deemed secondary to metabolic injury. Indeed, the increased cortisol levels present in patients with Cushing’s disease are detrimental to the white matter of the brain, including the corpus collosum, causing subsequent clinical derangements (22). Besides the direct effects of hypercortisolism, the splenial damage was also probably due to long-standing hypertension, worsened by newly occurring catecholamine hypersecretion and diabetes. Together with the normalization of cortisol and glycaemic levels, and of BP, a partial reduction in the splenial damage was observed on two subsequent MR examinations, and the patient's neurological condition slowly improved until she fully recovered. In our patient, thorough germinal genetic testing for the commonest pheochromocytoma/paraganglioma (PPGL) genes proved negative. Since approximately 40% of these tumours have germline mutations, genetic testing is recommended regardless of the patient’s age and family history. In the absence of syndromic, familial or metastatic presentation, the selection of genes for testing may be guided by the tumour location and biochemical phenotype. Alterations of the PPGL genes can be divided into two groups: 10 genes (RET, VHL, NF1, SDHD, SDHAF2, SDHC, SDHB, SDHA, TMEM127 and MAX) that have well-defined genotype–phenotype correlations, thus allowing to tailor imaging procedures and medical management, and a group of other emerging genes, which lack established genotype–phenotype associations; for patients in whom mutations of genes belonging to this second group are detected, and hence hereditary predisposition is established, only general medical surveillance and family screening can be planned (23, 24). In conclusion, our case highlights the importance of investigating patients with hypertension and metabolic derangements such as diabetes and hypokalaemia, since these findings may be a sign of newly occurring EAS, which, in rare cases, may be due to an ACTH-secreting pheochromocytoma. Since the additive effect of cortisol and catecholamine can cause dramatic clinical consequences, the possibility of an ACTH-secreting pheochromocytoma should be taken into account in the presence of an adrenal mass. EAS must be considered an endocrine emergency requiring urgent multi-specialist treatment. Surgery, whenever possible, is usually curative, and anatomic brain damage, as ascertained in our patient, may be at least partially reversible. Declaration of interest The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported. Funding This study did not receive any specific grant from any funding agency in the public, commercial or not-for-profit sector. The study was approved by the Local Ethics Committee (no: 732/2022). Patient consent The patient provided written informed consent. Author contribution statement All authors contributed equally to the conception, writing and editing of the manuscript. L Foppiani took care of the patient during hospitalization and in the outpatient department, performed the metabolic and endocrine work-up, conceived the study, analysed the data and wrote the manuscript. MG Poeta evaluated the patient during hospitalization with regard to neurological problems and planned the related work-up (brain imaging procedures and rachicentesis). M Rutigliani analysed the histological specimens and performed immunohistochemical studies. S Parodi performed CT and MR scans and analysed the related images. U Catrambone performed the left adrenalectomy. L Cavalleri performed general anaesthesia and assisted the patient during the surgical and post-surgical periods. G Antonucci revised the manuscript. P Del Monte helped in the endocrine work-up, in the evaluation of hormonal data and in the revision of the manuscript. A Piccardo performed 18F-FDG PET/CT and analysed the related images. Acknowledgement The work of Prof Silvia Morbelli in performing and analysing 68Ga-DOTATOC PET/CT is gratefully acknowledged. References 1↑ Pivonello R, Isidori AM, De Martino MC, Newell-Price J, Biller BMK, Colao A. Complications of Cushing's syndrome: state of the art. The Lancet Diabetes & Endocrinology 2016 4 611–629. (https://doi.org/10.1016/S2213-8587(1600086-3) Search Google Scholar Export Citation 2↑ Fleseriu M, Auchus R, Bancos I, Ben-Shlomo A, Bertherat J, Biermasz NR, Boguszewski CL, Bronstein MD, Buchfelder M, Carmichael JD, et al.Consensus on diagnosis and management of Cushing's disease: a guideline update. Lancet. Diabetes and Endocrinology 2021 9 847–875. (https://doi.org/10.1016/S2213-8587(2100235-7) PubMed Search Google Scholar Export Citation 3↑ Gabi JN, Milhem MM, Tovar YE, Karem ES, Gabi AY, & Khthir RA. Severe Cushing syndrome due to an ACTH-producing pheochromocytoma: a case presentation and review of the literature. Journal of the Endocrine Society 2018 2 621–630. (https://doi.org/10.1210/js.2018-00086) Search Google Scholar Export Citation 4↑ Young J, Haissaguerre M, Viera-Pinto O, Chabre O, Baudin E, & Tabarin A. Management of endocrine disease: Cushing's syndrome due to ectopic ACTH secretion: an expert operational opinion. European Journal of Endocrinology 2020 182 R29–R58. (https://doi.org/10.1530/EJE-19-0877) PubMed Search Google Scholar Export Citation 5↑ Hayes AR, & Grossman AB. The ectopic adrenocorticotropic hormone syndrome: rarely easy, always challenging. Endocrinology and Metabolism Clinics of North America 2018 47 409–425. (https://doi.org/10.1016/j.ecl.2018.01.005) PubMed Search Google Scholar Export Citation 6↑ Hayes AR, & Grossman AB. Distinguishing Cushing's disease from the ectopic ACTH syndrome: needles in a haystack or hiding in plain sight? Journal of Neuroendocrinology 2022 34 e13137. (https://doi.org/10.1111/jne.13137) Search Google Scholar Export Citation 7↑ Sakuma I, Higuchi S, Fujimoto M, Takiguchi T, Nakayama A, Tamura A, Kohno T, Komai E, Akina Shiga A, Nagano H, et al.Cushing syndrome due to ACTH-secreting pheochromocytoma, aggravated by glucocorticoid-driven positive-feedback loop. Journal of Clinical Endocrinology and Metabolism 2016 101 841–846. (https://doi.org/10.1210/jc.2015-2855) PubMed Search Google Scholar Export Citation 8↑ Barrett C, van Uum SH, & Lender JW. Risk of catecholaminergic crises following glucocorticoid administration in patients with an adrenal mass: a literature review. Clinical Endocrinology 2015 83 622–628. (https://doi.org/10.1111/cen.12813) Search Google Scholar Export Citation 9↑ Ballav C, Naziat A, Mihai R, Karavitaki N, Ansorge O, & Grossman AB. Mini-review: pheochromocytomas causing the ectopic ACTH syndrome. Endocrine 2012 42 69–73. (https://doi.org/10.1007/s12020-012-9646-7) PubMed Search Google Scholar Export Citation 10↑ Elliott PF, Berhane T, Ragnarsson O, & Falhammar H. Ectopic ACTH- and/or CRH-Producing pheochromocytomas. Journal of Clinical Endocrinology and Metabolism 2021 106 598–608. (https://doi.org/10.1210/clinem/dgaa488) Search Google Scholar Export Citation 11↑ Isidori AM, Kaltsas GA, Pozza C, Frajese V, Newell-Price J, Reznek RH, Jenkins PJ, Monson JP, Grossman AB, & Besser GM. The ectopic adrenocorticotropin syndrome: clinical features, diagnosis, management, and long-term follow-up. Journal of Clinical Endocrinology and Metabolism 2006 91 371–377.. (https://doi.org/10.1210/jc.2005-1542) PubMed Search Google Scholar Export Citation 12↑ Salgado LR, Fragoso MCB, Knoepfelmacher M, Machado MC, Domenice S, Pereira MA, & de Mendonça BB. Ectopic ACTH syndrome: our experience with 25 cases. European Journal of Endocrinology 2006 155 725–733. (https://doi.org/10.1530/eje.1.02278) PubMed Search Google Scholar Export Citation 13↑ Paleń-Tytko JE, Przybylik-Mazurek EM, Rzepka EJ, Pach DM, Sowa-Staszczak AS, Gilis-Januszewska A, & Hubalewska-Dydejczyk AB. Ectopic ACTH syndrome of different origin: diagnostic approach and clinical outcome. Experience of one clinical centre. PLoS One 2020 15 e0242679. (https://doi.org/10.1371/journal.pone.0242679) PubMed Search Google Scholar Export Citation 14↑ Krumeich LN, Cucchiara AJ, Nathanson KL, Kelz RR, Fishbein L, Fraker DL, Roses RE, Cohen DL, & Wachtel H. Correlation between plasma catecholamines, weight, and diabetes in pheochromocytoma and paraganglioma. Journal of Clinical Endocrinology and Metabolism 2021 106 e4028–e4038. (https://doi.org/10.1210/clinem/dgab401) PubMed Search Google Scholar Export Citation 15↑ Bihain F, Nomine-Criqui C, Guerci P, Gasman S, Klein M, & Brunaud L. Management of patients with treatment of pheochromocytoma: a critical appraisal. Cancers (Basel) 2022 14 3845. (https://doi.org/10.3390/cancers14163845) PubMed Search Google Scholar Export Citation 16↑ Ilias I, Torpy DJ, Pacak K, Mullen N, Wesley RA, & Nieman LK. Cushing’s syndrome due to ectopic corticotropin secretion: 20 years’ experience at the National Institute of Health. Journal of Clinical Endocrinology and Metabolism 2005 90 4955–4962. (https://doi.org/10.1210/jc.2004-2527) Search Google Scholar Export Citation 17↑ Varlamov E, Hinojosa-Amaya JM, Stack M, & Fleseriu M. Diagnostic utility of gallium-68-somatostatin receptor PET/CT in ectopic ACTH-secreting tumours: a systematic literature review and single-center clinical experience. Pituitary 2019 22 445–455. (https://doi.org/10.1007/s11102-019-00972-w) PubMed Search Google Scholar Export Citation 18↑ Varlamov EV, Han AJ, & Fleseriu M. Updates in adrenal steroidogenesis inhibitors for Cushing's syndrome. A practical guide. Best Practice and Research. Clinical Endocrinology and Metabolism 2021 35 101490. (https://doi.org/10.1016/j.beem.2021.101490) PubMed Search Google Scholar Export Citation 19↑ Marques JVO, & Boguszewski CL. Medical therapy in severe hypercortisolism. Best Practice and Research. Clinical Endocrinology and Metabolism 2021 35 101487. (https://doi.org/10.1016/j.beem.2021.101487) Search Google Scholar Export Citation 20↑ Sharma ST, & Nieman LK. Prolonged remission after long-term treatment with steroidogenesis inhibitors in Cushing's syndrome caused by ectopic ACTH secretion. European Journal of Endocrinology 2012 166 531–536. (https://doi.org/10.1530/EJE-11-0949) PubMed Search Google Scholar Export Citation 21↑ Martinez AD, Feelders RA, de Herder WW, Castaño JP, Moreno MAG, Dogan F, van Dungen R, van Koetsveld P, & Hofland LJ. Effects of ketoconazole on ACTH-producing and non-ACTH-producing neuroendocrine tumor cells. Hormones and Cancer 2019 107–119. (https://doi.org/10.1007/s12672-019-00361-6) Search Google Scholar Export Citation 22↑ Cui M, Zhou T, Feng S, Liu X, Wang F, Zhang Y, & Yu X. Altered microstructural pattern of white matter in Cushing’s disease identified by automated fiber quantification. NeuroImage. Clinical 2021 31 102770. (https://doi.org/10.1016/j.nicl.2021.102770) PubMed Search Google Scholar Export Citation 23↑ Lenders JWM, Duh QY, Eisenhofer G, Gimenez-Roqueplo AP, Grebe SKG, Hassan Murad MH, Naruse M, Pacak K, Young WF Jr & Endocrine Society. Pheochromocytoma and paraganglioma: an endocrine society clinical practice guideline. Journal of Clinical Endocrinology and Metabolism 2014 99 1915–1942. (https://doi.org/10.1210/jc.2014-1498) PubMed Search Google Scholar Export Citation 24↑ Sarkadi B, Saskoi E, Butz H, & Patocs A. Genetics of pheochromocytomas and paragangliomas determine the therapeutical approach. International Journal of Molecular Sciences 2022 23 1450. (https://doi.org/10.3390/ijms23031450) PubMed Search Google Scholar Export Citation From https://edm.bioscientifica.com/view/journals/edm/2023/2/EDM22-0308.xml
    1 point
×
×
  • Create New...