Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.140
Filter
1.
Dtsch Med Wochenschr ; 149(15): 871-878, 2024 Aug.
Article in German | MEDLINE | ID: mdl-39013407

ABSTRACT

Despite, or perhaps because of the rarity of neuroendocrine neoplasms, the diagnosis and treatment of these malignancies is of particular importance. Nuclear medicine can make an important contribution to this challenge. It offers the most sensitive and specific imaging of these tumor entities and can be helpful in treatment due to the radiotherapeutic drugs that have recently been approved. This theragnostic (fusion of therapeutic and diagnostic) concept is based on the frequent overexpression of somatostatin receptors on neuroendocrine tumor cells.Using diagnostic and therapeutic pharmaceuticals based on analogues from somatostatin, most applications from the nuclear medicine are successful, an additional therapeutic method is SIRT, also known as TARE, in which the hypervascularization of NEN-metastases is used as a therapeutic target.


Subject(s)
Neuroendocrine Tumors , Humans , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/therapy , Nuclear Medicine/methods , Radiopharmaceuticals/therapeutic use , Receptors, Somatostatin , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use
2.
Front Endocrinol (Lausanne) ; 15: 1385079, 2024.
Article in English | MEDLINE | ID: mdl-38948517

ABSTRACT

Background: 177Lu-oxodotreotide peptide receptor therapy (LuPRRT) is an efficient treatment for midgut neuroendocrine tumors (NETs) of variable radiological response. Several clinical, biological, and imaging parameters may be used to establish a relative disease prognosis but none is able to predict early efficacy or toxicities. We investigated expression levels for mRNA and miRNA involved in radiosensitivity and tumor progression searching for correlations related to patient outcome during LuPRRT therapy. Methods: Thirty-five patients received LuPRRT for G1/G2 midgut NETs between May 2019 and September 2021. Peripheral blood samples were collected prior to irradiation, before and 48 h after the second and the fourth LuPRRT, and at 6-month follow-up. Multiple regression analyses and Pearson correlations were performed to identify the miRNA/mRNA signature that will best predict response to LuPRRT. Results: Focusing on four mRNAs and three miRNAs, we identified a miRNA/mRNA signature enabling the early identification of responders to LuPRRT with significant reduced miRNA/mRNA expression after the first LuPRRT administration for patients with progressive disease at 1 year (p < 0.001). The relevance of this signature was reinforced by studying its evolution up to 6 months post-LuPRRT. Moreover, nadir absolute lymphocyte count within the first 2 months after the first LuPRRT administration was significantly related to low miRNA/mRNA expression level (p < 0.05) for patients with progressive disease. Conclusion: We present a pilot study exploring a miRNA/mRNA signature that correlates with early hematologic toxicity and therapeutic response 12 months following LuPRRT. This signature will be tested prospectively in a larger series of patients.


Subject(s)
Intestinal Neoplasms , MicroRNAs , Neuroendocrine Tumors , RNA, Messenger , Humans , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/blood , Neuroendocrine Tumors/therapy , Neuroendocrine Tumors/radiotherapy , Neuroendocrine Tumors/pathology , Male , Female , MicroRNAs/blood , MicroRNAs/genetics , Middle Aged , Intestinal Neoplasms/blood , Intestinal Neoplasms/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/drug therapy , RNA, Messenger/genetics , RNA, Messenger/blood , Aged , Follow-Up Studies , Adult , Prognosis , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Receptors, Peptide/genetics , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/administration & dosage , Lutetium , Radioisotopes
4.
Pituitary ; 27(3): 303-309, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38713317

ABSTRACT

INTRODUCTION: Skeletal fragility is a clinically relevant and not-reversible complication of acromegaly, involving around 30-40% of patients since the disease diagnosis. Few studies have investigated the effects on skeletal health of medical therapies for acromegaly. In this retrospective longitudinal monocentre study, we investigated the outcome of skeletal fragility in patients treated with Pasireotide Lar in combination with Pegvisomant (Pasi-Lar + Peg-V), also comparing those observed in patients treated with conventional therapies. RESULTS: We included 6 patients treated with Pasi-Lar + Peg-V, 5 patients treated with Peg-V in monotherapy (m-Peg-V), 16 patients treated with Peg-V plus first-generation somatostatin receptor ligands (fg-SRLs + Peg-V), 9 patients treated with Pasi-Lar. None of the patients treated with Pasi-Lar + Peg-V experienced worsening of spine and femoral bone mineral density (BMD) and incident vertebral fractures (i-VFs). Eight patients experienced i-VFs. The frequency of i-VFs was significantly lower in patients treated with the Pasi-Lar + Peg-V (0/8; 0%), as compared to those observed in m-Peg-V treated patients (4/8; 50%, p = 0.02). The frequency of i-VFs was slightly but not significantly higher in Pasi-Lar treated patients (1/8; 12.5% p = 0.6) and in fg-SRLs + Peg-V treated patients (3/8; 37.5% p = 0.364), concerning those treated with Pasi-Lar + Peg-V (0/8; 0%). I-VFs occurred more frequently in patients with higher GH levels at acromegaly diagnosis (p < 0.001), and in patients who experienced a BMD worsening (p = 0.005). CONCLUSION: Our preliminary data suggested that in conventional and multi-drug resistant acromegaly, the combination therapy Pasi-Lar + Peg-V may prevent the worsening of BMD and the occurrence of i-VFs. Prospective and translational studies should further validate these results and ascertain underlying physiopathology mechanisms.


Subject(s)
Acromegaly , Bone Density , Human Growth Hormone , Somatostatin , Humans , Acromegaly/drug therapy , Bone Density/drug effects , Middle Aged , Female , Male , Retrospective Studies , Adult , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Human Growth Hormone/analogs & derivatives , Pilot Projects , Aged , Longitudinal Studies
6.
Eur J Endocrinol ; 190(6): 421-433, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38701338

ABSTRACT

INTRODUCTION: Growth hormone (GH)-secreting pituitary tumors (GHomas) are the most common acromegaly cause. At diagnosis, most of them are macroadenomas, and up to 56% display cavernous sinus invasion. Biomarker assessment associated with tumor growth and invasion is important to optimize their management. OBJECTIVES: The study aims to identify clinical/hormonal/molecular biomarkers associated with tumor size and invasiveness in GHomas and to analyze the influence of pre-treatment with somatostatin analogs (SSAs) or dopamine agonists (DAs) in key molecular biomarker expression. METHODS: Clinical/analytical/radiological variables were evaluated in 192 patients from the REMAH study (ambispective multicenter post-surgery study of the Spanish Society of Endocrinology and Nutrition). The expression of somatostatin/ghrelin/dopamine system components and key pituitary/proliferation markers was evaluated in GHomas after the first surgery. Univariate/multivariate regression studies were performed to identify association between variables. RESULTS: Eighty percent of patients harbor macroadenomas (63.8% with extrasellar growth). Associations between larger and more invasive GHomas with younger age, visual abnormalities, higher IGF1 levels, extrasellar/suprasellar growth, and/or cavernous sinus invasion were found. Higher GH1 and lower PRL/POMC/CGA/AVPR1B/DRD2T/DRD2L expression levels (P < .05) were associated with tumor invasiveness. Least Absolute Shrinkage and Selection Operator's penalized regression identified combinations of clinical and molecular features with areas under the curve between 0.67 and 0.82. Pre-operative therapy with DA or SSAs did not alter the expression of any of the markers analyzed except for DRD1/AVPR1B (up-regulated with DA) and FSHB/CRHR1 (down-regulated with SSAs). CONCLUSIONS: A specific combination of clinical/analytical/molecular variables was found to be associated with tumor invasiveness and growth capacity in GHomas. Pre-treatment with first-line drugs for acromegaly did not significantly modify the expression of the most relevant biomarkers in our association model. These findings provide valuable insights for risk stratification and personalized management of GHomas.


Subject(s)
Acromegaly , Adenoma , Growth Hormone-Secreting Pituitary Adenoma , Neoplasm Invasiveness , Humans , Male , Female , Acromegaly/metabolism , Middle Aged , Adult , Growth Hormone-Secreting Pituitary Adenoma/pathology , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Adenoma/metabolism , Adenoma/pathology , Aged , Dopamine Agonists/therapeutic use , Biomarkers, Tumor/metabolism , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Human Growth Hormone/metabolism
7.
Endocr Relat Cancer ; 31(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38713182

ABSTRACT

The objective of the study was to evaluate the efficacy of second-line therapies in patients with acromegaly caused by a growth hormone (GH) and prolactin (PRL) co-secreting pituitary neuroendocrine tumor (GH&PRL-Pit-NET) compared to their efficacy in patients with acromegaly caused by a GH-secreting pituitary neuroendocrine tumor (GH-Pit-NET). This is a multicenter retrospective study of patients with acromegaly on treatment with pasireotide and/or pegvisomant. Patients were classified in two groups: GH&PRL-Pit-NETs when evidence of hyperprolactinemia and immunohistochemistry (IHC) for GH and PRL was positive or if PRL were >200 ng/dL regardless of the PRL-IHC and GH-Pit-NETs when the previously mentioned criteria were not met. A total of 28 cases with GH&PRL-Pit-NETs and 122 with GH-Pit-NETs met the inclusion criteria. GH&PRL-Pit-NETs presented at a younger age, caused hypopituitarism, and were invasive more frequently than GH-Pit-NETs. There were 124 patients treated with pegvisomant and 49 with pasireotide at any time. The efficacy of pegvisomant for IGF-1 normalization was of 81.5% and of pasireotide of 71.4%. No differences in IGF-1 control with pasireotide and with pegvisomant were observed between GH&PRL-Pit-NETs and GH-Pit-NETs. All GH&PRL-Pit-NET cases treated with pasireotide (n = 6) and 82.6% (n = 19/23) of the cases treated with pegvisomant normalized PRL levels. No differences in the rate of IGF-1 control between pegvisomant and pasireotide were detected in patients with GH&PRL-Pit-NETs (84.9% vs 66.7%, P = 0.178). We conclude that despite the more aggressive behavior of GH&PRL-Pit-NETs than GH-Pit-NETs, no differences in the rate of IGF-1 control with pegvisomant and pasireotide were observed between both groups, and both drugs have shown to be effective treatments to control IGF-1 and PRL hypersecretion in these tumors.


Subject(s)
Acromegaly , Human Growth Hormone , Neuroendocrine Tumors , Prolactin , Somatostatin , Humans , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Male , Female , Human Growth Hormone/analogs & derivatives , Human Growth Hormone/therapeutic use , Middle Aged , Adult , Prolactin/blood , Prolactin/metabolism , Retrospective Studies , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/metabolism , Acromegaly/drug therapy , Acromegaly/metabolism , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/metabolism , Aged , Young Adult
8.
Eur J Endocrinol ; 190(6): 458-466, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38771697

ABSTRACT

OBJECTIVE: The aim of this study is to compare the response to first-line medical treatment in treatment-naive acromegaly patients with pure growth hormone (GH)-secreting pituitary adenoma (GH-PA) and those with GH and prolactin cosecreting PA (GH&PRL-PA). DESIGN: This is a retrospective multicentric study of acromegaly patients followed from 2003 to 2023 in 33 tertiary Spanish hospitals with at least 6 months of first-line medical treatment. METHODS: Baseline characteristics, first-line medical treatment strategies, and outcomes were analyzed. We employed a multiple logistic regression full model to estimate the impact of some baseline characteristics on disease control after each treatment modality. RESULTS: Of the 144 patients included, 72.9% had a GH-PA, and 27.1% had a GH&PRL-PA. Patients with GH&PRL-PA were younger (43.9 ± 15.0 vs 51.9 ± 12.7 years, P < .01) and harboring more frequently macroadenomas (89.7% vs 72.1%, P = .03). First-generation somatostatin receptor ligand (fgSRL) as monotherapy was given to 106 (73.6%) and a combination treatment with fgSRL and cabergoline in the remaining 38 (26.4%). Patients with GH&PRL-PA received more frequently a combination therapy (56.4% vs 15.2%, P < .01). After 6 months of treatment, in the group of patients under fgSRL as monotherapy, those patients with GH&PRL-PA had worse control compared to GH-PAs (29.4% vs 55.1%, P = .04). However, these differences in the rate of disease control between both groups disappeared when both received combination treatment with fgSRL and cabergoline. CONCLUSION: In GH&PRL-PA, the biochemical control achieved with fgSRL as monotherapy is substantially worse than in patients harboring GH-PA, supporting the inclusion of cabergoline as first-line medical treatment in combination with fgSRLs in these subgroups of patients.


Subject(s)
Acromegaly , Cabergoline , Prolactin , Humans , Acromegaly/drug therapy , Acromegaly/blood , Female , Male , Middle Aged , Retrospective Studies , Adult , Cabergoline/therapeutic use , Treatment Outcome , Prolactin/blood , Growth Hormone-Secreting Pituitary Adenoma/drug therapy , Growth Hormone-Secreting Pituitary Adenoma/blood , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Human Growth Hormone , Adenoma/drug therapy , Adenoma/blood , Adenoma/metabolism , Adenoma/complications , Aged , Drug Therapy, Combination , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/blood , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/complications , Spain/epidemiology
9.
Expert Rev Gastroenterol Hepatol ; 18(4-5): 203-215, 2024.
Article in English | MEDLINE | ID: mdl-38725175

ABSTRACT

OBJECTIVE: To determine the effectiveness of the different pharmacological agents in preventing post-ERCP acute pancreatitis. METHODS: We included clinical trials of pharmacological interventions for prophylaxis of acute post-ERCP pancreatitis. The event evaluated was acute pancreatitis. We conducted a search strategy in MEDLINE (OVID), EMBASE, and Cochrane Central Register of Controlled Trials from inception to nowadays. We reported the information in terms of relative risks (RR) with a 95% confidence interval. We assessed the heterogeneity using the I2 test. RESULTS: We included 84 studies for analysis (30,463 patients). The mean age was 59.3 years (SD ± 7.01). Heterogeneity between studies was low (I2 = 34.4%) with no inconsistencies (p = 0.2567). Post ERCP pancreatitis was less in prophylaxis with NSAIDs (RR 0.65 95% CI [0.52 to 0.80]), aggressive hydration with Lactate Ringer (RR 0.32 95% CI [0.12-0.86]), NSAIDs + isosorbide dinitrate (RR 0.28 95% CI [0.11-0.71]) and somatostatin and analogues (RR 0.54 [0.43 to 0.68]) compared with placebo. CONCLUSIONS: NSAIDs, the Combination of NSAIDs + isosorbide dinitrate, somatostatin and analogues, and aggressive hydration with lactate ringer are pharmacological strategies that can prevent post-ERCP pancreatitis when compared to placebo. More clinical trials are required to determine the effectiveness of these drugs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Cholangiopancreatography, Endoscopic Retrograde , Pancreatitis , Aged , Humans , Middle Aged , Acute Disease , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Cholangiopancreatography, Endoscopic Retrograde/adverse effects , Fluid Therapy/methods , Network Meta-Analysis , Pancreatitis/prevention & control , Pancreatitis/etiology , Ringer's Lactate/therapeutic use , Ringer's Lactate/administration & dosage , Risk Factors , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Treatment Outcome
10.
Best Pract Res Clin Endocrinol Metab ; 38(4): 101893, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38575404

ABSTRACT

Although predictors of response to first-generation somatostatin receptor ligands (fg-SRLs), and to a lesser extent to pasireotide, have been studied in acromegaly for many years, their use is still not recommended in clinical guidelines. Is there insufficient evidence to use them? Numerous biomarkers including various clinical, functional, radiological and molecular markers have been identified. The first ones are applicable pre-surgery, while the molecular predictors are utilized for patients not cured after surgery. In this regard, factors predicting a good response to fg-SRLs are specifically: low basal GH, a low GH nadir in the acute octreotide test, T2 MRI hypointensity, a densely granulated pattern, high immunohistochemistry staining for somatostatin receptor 2 (SSTR2), and E-cadherin. However, there is still a lack of consensus regarding which of these biomarkers is more useful or how to integrate them into clinical practice. With classical statistical methods, it is complex to define reliable and generalizable cut-off values for a single biomarker. The potential solution to the limitations of traditional methods involves combining systems biology with artificial intelligence, which is currently providing answers to such long-standing questions that may eventually be finally included into the clinical guidelines and make personalized medicine a reality. The aim of this review is to describe the current knowledge of the main fg-SRLs and pasireotide response predictors, discuss their current usefulness, and point to future directions in the research of this field.


Subject(s)
Acromegaly , Receptors, Somatostatin , Somatostatin , Humans , Receptors, Somatostatin/metabolism , Receptors, Somatostatin/agonists , Acromegaly/drug therapy , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Ligands , Octreotide/therapeutic use , Human Growth Hormone/metabolism , Biomarkers/blood , Treatment Outcome
11.
Ann Surg ; 280(2): 179-187, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38662619

ABSTRACT

OBJECTIVE: Pharmacological prevention of postoperative pancreatic fistula (POPF) after pancreatectomy is open to debate. The present study compares clinically significant POPF rates in patients randomized between somatostatin versus octreotide as prophylactic treatment. METHODS: Multicentric randomized controlled open study in patient's candidate for pancreaticoduodenectomy (PD) or distal pancreatectomy (DP) comparing somatostatin continuous intravenous infusion for 7 days versus octreotid 100 µg, every 8 hours subcutaneous injection for 7 days, stratified by procedure (PD vs DP) and size of the main pancreatic duct (>4 mm) on grade B/C POPF rates at 90 days based on an intention-to-treat analysis. RESULTS: Of 763 eligible patients, 651 were randomized: 327 in the octreotide arm and 324 in the somatostatin arm, with comparable the stratification criteria - type of surgery and main pancreatic duct dilatation. Most patients had PD (n=480; 73.8%), on soft/normal pancreas (n=367; 63.2%) with a nondilated main pancreatic duct (n=472; 72.5%), most often for pancreatic adenocarcinoma (n=311; 47.8%). Almost all patients had abdominal drainage (n=621; 96.1%) and 121 (19.5%) left the hospital with the drain in place (median length of stay=16 days). A total of 153 patients (23.5%) developed a grade B/C POPF with no difference between both groups: 24.1%: somatostatin arm and 22.9%: octreotide arm (χ 2 test, P =0.73, ITT analysis). Absence of statistically significant difference persisted after adjustment for stratification variables and in per-protocol analysis. CONCLUSION: Continuous intravenous somatostatin is not statistically different from subcutaneous octreotide in the prevention of grade B/C POPF after pancreatectomy. FINDINGS: In the PREFIPS Randomized Clinical Trial including 651 patients, a total of 153 patients (23.5%) developed a grade B/C POPF with no significant difference between both groups: 24.1%: somatostatin arm and 22.9%: octreotide arm (χ 2 test, P =0.73, ITT analysis). Absence of statistically significant difference persisted after adjustment for stratification variables and in per-protocol analysis.


Subject(s)
Octreotide , Pancreatectomy , Pancreatic Fistula , Pancreaticoduodenectomy , Postoperative Complications , Somatostatin , Humans , Pancreatic Fistula/prevention & control , Pancreatic Fistula/etiology , Pancreatic Fistula/epidemiology , Octreotide/therapeutic use , Octreotide/administration & dosage , Male , Female , Somatostatin/administration & dosage , Somatostatin/therapeutic use , Pancreatectomy/adverse effects , Pancreaticoduodenectomy/adverse effects , Middle Aged , Postoperative Complications/prevention & control , Aged , Infusions, Intravenous , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/therapeutic use , Treatment Outcome , France/epidemiology , Adult , Injections, Subcutaneous
12.
Reprod Biol Endocrinol ; 22(1): 48, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38650041

ABSTRACT

CONTEXT: Acromegaly is a rare disease caused by excessive growth hormone (GH) secretion, mostly induced by pituitary adenomas. The care of pregnant women with acromegaly is challenging, in part due to existing clinical data being limited and not entirely consistent with regard to potential risks for mother and child. OBJECTIVE: To retrospectively examine data on pregnancy and maternal as well as neonatal outcomes in patients with acromegaly. DESIGN & METHODS: Retrospective data analysis from 47 pregnancies of 31 women treated in centers of the German Acromegaly Registry. RESULTS: 87.1% of the studied women underwent transsphenoidal surgery before pregnancy. In 51.1% a combination of dopamine agonists and somatostatin analogs were used before pregnancy. Three women did not receive any therapy for acromegaly. During pregnancy only 6.4% received either somatostatin analogs or dopamine agonists. In total, 70.2% of all documented pregnancies emerged spontaneously. Gestational diabetes was diagnosed in 10.6% and gravid hypertension in 6.4%. Overall, no preterm birth was detected. Indeed, 87% of acromegalic women experienced a delivery without complications. CONCLUSION: Pregnancies in women with acromegaly are possible and the course of pregnancy is in general safe for mother and child both with and without specific treatment for acromegaly. The prevalence of concomitant metabolic diseases such as gestational diabetes is comparable to the prevalence in healthy pregnant women. Nevertheless, larger studies with more data in pregnant patients with acromegaly are needed to provide safe and effective care for pregnant women with this condition.


Subject(s)
Acromegaly , Pregnancy Complications , Pregnancy Outcome , Registries , Humans , Female , Pregnancy , Acromegaly/epidemiology , Acromegaly/therapy , Retrospective Studies , Adult , Germany/epidemiology , Pregnancy Outcome/epidemiology , Pregnancy Complications/epidemiology , Diabetes, Gestational/epidemiology , Infant, Newborn , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use
13.
J Gastrointest Surg ; 28(3): 259-266, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38445918

ABSTRACT

BACKGROUND: Diversion colitis (DC) is a prevalent complication of colostomy characterized by intestinal inflammation. This study aimed to investigate the therapeutic potential of somatostatin (SST) in managing DC. METHODS: After establishing a rat DC model, SST was administered via Mini Osmotic Pumps 2001W at a pumping rate of 1.0 µL/h. Various techniques, including hematoxylin and eosin staining, periodic acid-Schiff staining, immunofluorescence staining, and electron microscopy were employed to assess the effects of SST. Intestinal barrier functions were evaluated using Evans blue, enzyme-linked immunosorbent assay, and MacConkey agar. RESULTS: After SST treatment, the significant weight loss and associated high mortality in the DC group were successfully mitigated. Upregulation of claudin-3 and claudin-4 restored mechanical barriers in colon epithelial tissue, whereas protection of goblet cells and stimulation of mucus secretion enhanced mucus barriers. SST effectively reduced leaky gut and alleviated systemic inflammation. CONCLUSION: This study provides initial evidence supporting the efficacy of SST in the treatment of DC. It offers insights into the role of SST in DC by elucidating its ability to restore damaged intestinal barriers.


Subject(s)
Colitis , Colostomy , Animals , Rats , Colostomy/adverse effects , Rivers , Colitis/drug therapy , Colitis/surgery , Somatostatin/therapeutic use , Inflammation
14.
Acta Gastroenterol Belg ; 87(1): 48-51, 2024.
Article in English | MEDLINE | ID: mdl-38431792

ABSTRACT

Cushing's syndrome (CS) secondary to adrenocorticotropic hormone (ACTH) producing tumours is a severe condition with a challenging diagnosis. Ectopic ACTH-secretion often involves neuroendocrine tumours (NET) in the respiratory tract. ACTH-secreting small intestine neuro-endocrine tumours (siNET) are extremely rare entities barely reported in literature. This review is illustrated by the case of a 75-year old woman with fulminant ectopic CS caused by a ACTH-secreting metastatic siNET. Severe hypokalemia, fluid retention and refractory hypertension were the presenting symptoms. Basal and dynamic laboratory studies were diagnostic for ACTH-dependent CS. Extensive imaging studies of the pituitary and thorax-abdomen areas were normal, while [68Ga]Ga-DOTATATE PET-CT revealed increased small intestine uptake in the left iliac fossa. The hypercortisolism was well controlled with somatostatin analogues, after which a debulking resection of the tumour was performed. Pathological investigation confirmed a well-differentiated NET with sporadic ACTH immunostaining and post-operative treatment with somatostatin analogues was continued with favourable disease control.


Subject(s)
Cushing Syndrome , Intestinal Neoplasms , Neuroendocrine Tumors , Female , Humans , Aged , Cushing Syndrome/diagnosis , Cushing Syndrome/etiology , Cushing Syndrome/pathology , Neuroendocrine Tumors/complications , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/pathology , Positron Emission Tomography Computed Tomography , Adrenocorticotropic Hormone , Intestinal Neoplasms/complications , Intestinal Neoplasms/diagnosis , Somatostatin/therapeutic use
15.
Int J Biol Sci ; 20(2): 801-817, 2024.
Article in English | MEDLINE | ID: mdl-38169563

ABSTRACT

Somatostatin analogues (SSTA) are first-line pharmacological treatment choice for acromegaly, which received satisfying tumor shrinkage and normalization of growth hormone. However, there are still patients unresponsive to SSTA, and the underline mechanism remains unknown. Besides, there is no evidence regarding the role of endoplasmic reticulum stress (ERS) and its transmission in SSTA resistance, which also require investigation. Primary growth hormone adenoma cells and cell lines were treated with SSTA; autophagy double-labeled LC3 (mRFP-GFP) adenovirus transfection, flow cytometry sorting, western blotting, calcium imaging as well as immunofluorescence staining were used to determine ERS and autophagy signal transmission; xenograft and syngeneic tumor in vivo model were exploited to confirm the ERS signal transmission mediated effect. Our results revealed that SSTA induces ERS in pituitary growth hormone (GH) adenoma cells. The ERS signals can be intercellularly transmitted, leading to less responsible to SSTA treatment. Moreover, SSTA stimulates inositol triphosphate (IP3) elevation, mediating ERS intercellular transfer. In addition, connexin 36 tunnels ERS transmission, and its blocker, Quinine, exhibits a synergistic effect with SSTA treating GH adenoma. Our study provided insight into ERS intercellular transmission mediated SSTA resistance, which could be translated into clinical usage to improve SSTA efficiency in GH adenoma treatment.


Subject(s)
Adenoma , Pituitary Neoplasms , Humans , Somatostatin/pharmacology , Somatostatin/therapeutic use , Growth Hormone/metabolism , Growth Hormone/therapeutic use , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Gap Junction delta-2 Protein , Adenoma/drug therapy , Endoplasmic Reticulum Stress
16.
J Endocrinol Invest ; 47(7): 1733-1741, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38244140

ABSTRACT

OBJECTIVE: Pasireotide LAR (PAS-LAR) was released in Italy in 2017 to treat acromegaly patients resistant to SRLs (Somatostatin Receptors Ligands). The long-term follow-up data of PAS-LAR therapy in Italy are limited. This study aimed to evaluate the efficacy and safety of PAS-LAR in acromegaly. DESIGN: Patients with acromegaly in PAS-LAR treatment were enrolled in three tertiary Italian endocrinological centers and evaluated by a retrospective observational real-life multicentre study. METHODS: Patients have been studied before (baseline) and 1, 6, 12, 24 and > 36 months after PAS-LAR start. Clinical, biochemical, and pituitary magnetic resonance data were collected, along with information on adverse events. Acromegaly disease activity was classified according to the IGF-1 index (normal value < 1.0). RESULTS: Fifty patients (female 23) were enrolled. PAS-LAR treatment (mean follow-up 24 ± 16 months) significantly decreased IGF-1 levels (IGF-1 index baseline vs last visit: 1.9 ± 0.6 vs 1.2 ± 0.6, p < 0.0001). At the last visit, 67% of patients had controlled disease, and 44% showed a decrease in tumor volume. Clinical and biochemical efficacy was observed as early as after 1-month of PAS-LAR treatment (IGF-1 index baseline vs 1-month: 1.9 ± 0.6 vs 1.4 ± 0.7, p < 0.0001). Also, 50% of patients referred headache improvement or disappearance. Fifteen patients discontinued PAS-LAR due to failure of treatment and poor glycaemic control. The prevalence of diabetes increased from 33% at the baseline to 54% at the last visit (p = 0.0072). CONCLUSION: In real-life settings, PAS-LAR significantly decreases symptoms, IGF-1 levels, and the size of adenoma in patients with acromegaly resistant to SRLs. Beneficial effects may occur early after the first injection.


Subject(s)
Acromegaly , Somatostatin , Humans , Female , Acromegaly/drug therapy , Male , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Somatostatin/administration & dosage , Middle Aged , Follow-Up Studies , Retrospective Studies , Adult , Treatment Outcome , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/metabolism , Italy/epidemiology , Growth Hormone-Secreting Pituitary Adenoma/drug therapy , Growth Hormone-Secreting Pituitary Adenoma/complications , Aged , Human Growth Hormone/blood
17.
Best Pract Res Clin Endocrinol Metab ; 38(4): 101875, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38290866

ABSTRACT

"First-generation" somatostatin receptor agonists (SSTRAs) octreotide and lanreotide are the most commonly used first-line pharmacological therapy for patients with acromegaly. A subset of patients respond only partially or not at all to the first-generation SSTRA, necessitating the use of additional pharmacological agents or other modes of therapy. Pasireotide is a "second-generation" SSTRA that has multi-receptor activity. Prospective studies have shown promise in the use of pasireotide in patients with poor response to first-generation SSTRA. Here we elucidate the molecular pathways of resistance to first-generation SSTRA, the mechanism of action, pre-clinical and clinical evidence of the use of pasireotide in patients having incomplete / lack of response to first-generation SSTRA. We also discuss the clinical, pathological, and radiological markers predicting response to pasireotide, and the difference in side-effect profiles of pasireotide, compared to first-generation SSTRA.


Subject(s)
Acromegaly , Somatostatin , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Humans , Acromegaly/drug therapy , Receptors, Somatostatin/agonists , Treatment Outcome
18.
J Nucl Med ; 65(3): 340-348, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38238038

ABSTRACT

Most well-differentiated neuroendocrine tumors (NETs) express high levels of somatostatin receptors, particularly subtypes 2 and 5. Somatostatin analogs (SSAs) bind to somatostatin receptors and are used for palliation of hormonal syndromes and control of tumor growth. The long-acting SSAs octreotide long-acting release and lanreotide are commonly used in the first-line metastatic setting because of their tolerable side effect profile. Radiolabeled SSAs are used both for imaging and for treatment of NETs. 177Lu-DOTATATE is a ß-emitting radiolabeled SSA that has been proven to significantly improve progression-free survival among patients with progressive midgut NETs and is approved for treatment of metastatic gastroenteropancreatic NETs. A key question in management of patients with gastroenteropancreatic and lung NETs is the sequencing of 177Lu-DOTATATE in relation to other systemic treatments (such as everolimus) or liver-directed therapies. This question is particularly complicated given the heterogeneity of NETs and the near absence of randomized trials comparing active treatment options. This state-of-the-art review examines the evidence supporting use of somatostatin-receptor-targeted treatments within the larger landscape of NET therapy and offers insights regarding optimal patient selection, assessment of benefit versus risk, and treatment sequencing.


Subject(s)
Carcinoma, Neuroendocrine , Neoplasms, Second Primary , Neuroendocrine Tumors , Humans , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/radiotherapy , Receptors, Somatostatin , Somatostatin/therapeutic use , Octreotide
19.
Oncologist ; 29(5): e643-e654, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38206830

ABSTRACT

BACKGROUND: In advanced neuroendocrine tumors (NET), antiproliferative treatment options beyond somatostatin analogs remain limited. Temozolomide (TMZ) has shown efficacy in NET alone or combined with other drugs. MATERIALS AND METHODS: SONNET (NCT02231762) was an open, multicenter, prospective, phase II study to evaluate lanreotide autogel 120 mg (LAN) plus TMZ in patients with progressive advanced/metastatic grade 1/2 gastroenteropancreatic (GEP) NET or of unknown primary. Patients could be enrolled at first-line or higher therapy line. The primary endpoint was disease control rate ([DCR], rate of stable disease [SD], partial [PR], and complete response [CR]) at 6 months of LAN and TMZ. Patients with nonfunctioning (NF) NET without progression at 6 months were randomized to 6-month LAN maintenance or watch and wait, patients with functioning (F)-NET with clinical benefit (PR, SD) continued on LAN. RESULTS: Fifty-seven patients were recruited. The majority of patients received the study drug at second or higher treatment line and had an NET G2. DCR at 6 months LAN and TMZ was 73.5%. After 6 months of further LAN maintenance, 54.5% of patients with F-NET and 71.4% with NF-NET had SD or PR vs 41.7% with NF-NET on observation only. LAN and TMZ were effective in all subgroups analyzed. At 12 months of follow-up, median progression-free survival was 11.1 months. Median serum chromogranin A decreased except in NF-NET on observation. O6-methylguanine DNA methyltransferase promoter methylation appeared to better reflect TMZ response than loss of gene expression. During combination therapy, the most frequent treatment-emergent adverse events grade 3/4 reported were nausea (14%), thrombocytopenia (12.3%), and neutropenia (8.8%). Four deaths were reported resulting from severe adverse events not considered related to study medication. CONCLUSIONS: LAN plus TMZ is a treatment option for patients with progressive GEP-NET with more aggressive biological profile showing a manageable safety profile.


Subject(s)
Intestinal Neoplasms , Neuroendocrine Tumors , Pancreatic Neoplasms , Peptides, Cyclic , Somatostatin , Somatostatin/analogs & derivatives , Temozolomide , Humans , Temozolomide/pharmacology , Temozolomide/therapeutic use , Temozolomide/administration & dosage , Somatostatin/therapeutic use , Somatostatin/pharmacology , Somatostatin/administration & dosage , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Male , Female , Middle Aged , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Aged , Peptides, Cyclic/therapeutic use , Peptides, Cyclic/pharmacology , Peptides, Cyclic/administration & dosage , Intestinal Neoplasms/drug therapy , Intestinal Neoplasms/pathology , Adult , Prospective Studies , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Aged, 80 and over
20.
Curr Probl Cancer ; 49: 101061, 2024 04.
Article in English | MEDLINE | ID: mdl-38281845

ABSTRACT

Pulmonary neuroendocrine tumors represent a spectrum of disease ranging from typical carcinoid tumors to small cell lung cancers. The incidence of low-grade pulmonary NETs has been increasing, leading to improved awareness and the need for more treatment options for this rare cancer. Somatostatin analogs continue to be the backbone of therapy and may be followed or accompanied by targeted therapy, chemotherapy, and immune therapy. The recent addition of peptide receptor radionuclide therapy (PRRT) to the treatment armamentarium of NETs has led to the development of targeted alpha therapy to overcome PRRT resistance and minimize off-target adverse effects. Herein, we aim to highlight current treatment options for patients with advanced low grade pulmonary NETs along with emerging therapies, sequencing of therapies, upcoming clinical trials, and the importance of a multidisciplinary team to improve patient outcomes.


Subject(s)
Lung Neoplasms , Neuroendocrine Tumors , Humans , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Lung Neoplasms/drug therapy , Neuroendocrine Tumors/therapy , Neuroendocrine Tumors/pathology , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Disease Management
SELECTION OF CITATIONS
SEARCH DETAIL