Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.762
Filter
1.
BMJ Open Ophthalmol ; 9(1)2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134323

ABSTRACT

AIMS: This study aimed to evaluate the effectiveness of somatostatin analogues (SA) for cystoid maculopathy (CM) in retinitis pigmentosa (RP) patients. MATERIALS AND METHODS: In this retrospective case series, clinical and imaging characteristics of 28 RP patients with CM, unresponsive to carbonic anhydrase inhibitors, were collected from medical charts. All patients received SA treatment as an alternative (octreotide long-acting release at 20 mg/month or 30 mg/month, or lanreotide at 90 mg/month or 120 mg/month). Outcome measures were mean reduction in foveal thickness (FT) and foveal volume (FV) and mean increase in best-corrected visual acuity at 3, 6 and 12 months of treatment initiation. Linear mixed models were used to calculate the effectiveness over time. RESULTS: 52 eyes of 28 RP patients were included; 39% were male. The median age at the start of treatment was 39 years (IQR 30-53). Median follow-up was 12 months (range 6-12). From baseline to 12 months, the mean FT decreased from 409±136 µm to 334±119 µm and the mean FV decreased from 0.31±0.10 mm3 to 0.25±0.04 mm3. Linear mixed model analyses showed a significant decrease in log FT and log FV at 3, 6 and 12 months after the start of treatment compared with baseline measurements (p<0.001, p<0.001, p<0.001). Mean best-corrected visual acuity did not increase significantly (0.46±0.35 logMAR to 0.45±0.38 logMAR after 12 months). DISCUSSION: SA may be an effective alternative treatment to reduce CM in RP patients.


Subject(s)
Retinitis Pigmentosa , Somatostatin , Tomography, Optical Coherence , Visual Acuity , Humans , Retinitis Pigmentosa/drug therapy , Male , Female , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Retrospective Studies , Middle Aged , Visual Acuity/drug effects , Adult , Peptides, Cyclic/therapeutic use , Octreotide/therapeutic use , Octreotide/administration & dosage , Treatment Outcome , Macular Edema/drug therapy , Macular Edema/etiology
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
3.
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
4.
Medicina (Kaunas) ; 60(7)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39064468

ABSTRACT

Background and Objectives: High-grade malignant neuroendocrine tumors (G3 NETs) and neuroendocrine carcinomas (NECs) are characterized by rapid proliferation, high metastatic capacity, and strong expression of somatostatin receptors (SSTRs). We aimed to analyze the presence of SSTRs in NET G3 and NEC, and to correlate their expression with the use of octreotide and pasireotide. Materials and Methods: For this purpose, we first performed a retrospective study of G3 NET and NEC patients, which included the determination of SSTR expression and response to octreotide treatment. Second, we selected the H69 small cell lung cancer cell line to determine the effect of octreotide and pasireotide. Results: Our results showed the traditional somatostatin analog (SSA) octreotide was ineffective in patients with NET G3 and NEC. On the other hand, RT-qPCR showed a high expression of SSTR2 and SSTR5 in H69 cells. Interestingly, while octreotide did not modify H69 cell proliferation, a strong inhibition of proliferation was detected with the use of pasireotide. Conclusions: In view of these results, a clinical trial in NET G3 and NEC patients using pasireotide is necessary to determine the usefulness of this drug in improving patient treatment.


Subject(s)
Neuroendocrine Tumors , Octreotide , Receptors, Somatostatin , Somatostatin , Humans , Octreotide/therapeutic use , Octreotide/pharmacology , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Somatostatin/pharmacology , Neuroendocrine Tumors/drug therapy , Receptors, Somatostatin/analysis , Retrospective Studies , Male , Middle Aged , Female , Aged , Cell Line, Tumor , Cell Proliferation/drug effects , Adult , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
5.
Endocr Relat Cancer ; 31(9)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39059428
7.
BMC Anesthesiol ; 24(1): 265, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085758

ABSTRACT

BACKGROUND: Carcinoid tumors are rare neuroendocrine malignancies presenting in an increasing number in our center. The incidence of carcinoid tumors is approximatively between 2.5 and 5 cases per 100,000 people of whom about 50% develop carcinoid syndrome. Once the carcinoid syndrome has developed, a carcinoid cardiomyopathy can occur. Carcinoid heart disease (CaHD) remains a serious and rare complication associated with a significant increase in morbidity and mortality. Although carcinoid tumors have been known and studied for several years, there are still scarce data on the anesthetic management and the peri operative period. CASE PRESENTATION: We describe a case of a Caucasian 44-year-old woman with an unusual presentation of left CaHD with an ileal neuroendocrine tumor and liver metastases. Our preoperative somatostatin administration protocol, limit the cardiac damage. The maintenance of stable hemodynamics, the use of balanced anesthetic technique, all along with a good understanding of the pathology, played a major role in the successful management of anesthesia. This case report allows us to introduce our decision algorithm for the management of this type of pathology in our tertiary hospital, Cliniques Universitaires Saint-Luc. CONCLUSION: Despite the paucity of data, anesthetic management of patients with carcinoid tumor can be safely performed with effective hemodynamic monitoring and a good understanding of the pathophysiology. Knowledge and application of a clear institutional algorithm for octreotide administration and multidisciplinary consultation at a referral center are essential for the management of these patients.


Subject(s)
Carcinoid Heart Disease , Ileal Neoplasms , Neuroendocrine Tumors , Humans , Female , Adult , Carcinoid Heart Disease/complications , Ileal Neoplasms/complications , Neuroendocrine Tumors/complications , Anesthesia/methods , Carcinoid Tumor/complications , Somatostatin/analogs & derivatives , Somatostatin/administration & dosage , Somatostatin/therapeutic use , Liver Neoplasms/secondary
8.
J Nucl Med ; 65(8): 1279-1285, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38991751

ABSTRACT

Myocardial somatostatin PET uptake is observed not only in most patients with acute myocarditis (AM) but also in some oncology patients referred for routine somatostatin PET. This raises concerns about the specificity of somatostatin PET for detecting myocarditis. The current study aims to identify factors associated with the detection of myocardial uptake on somatostatin PET scans recorded for oncology indications and differential PET criteria that characterize myocardial uptake in AM patients. Methods: We analyzed factors associated with the detection of myocardial [68Ga]Ga-DOTATOC uptake in 508 [68Ga]Ga-DOTATOC PET scans from 178 patients, performed for confirmed or suspected oncologic disease (Onc-PET) and PET criteria that could differentiate myocardial [68Ga]Ga-DOTATOC uptake in 31 patients with MRI-ascertained AM (AM-PET) from that in the Onc-PET group. Results: Significant myocardial uptake was detected in 137 (26.9%) Onc-PET scans and was independently associated with somatostatin analog treatment (exp(ß), 0.805; 95% CI, 0.728-0.890; P < 0.001) and age (exp(ß), 1.005; 95% CI, 1.001-1.009; P = 0.012). A comparable model was selected for predicting the myocardial-to-blood SUVmax ratio using somatostatin analog treatment (P < 0.001) and history of coronary artery disease (P = 0.022). Myocardial uptake was detected in 12.9% (25/193) of Onc-PET scans from patients treated with somatostatin analogs but in 43.4% (59/136) of untreated patients over the median age of 64 y. Myocardial uptake was apparent in all 31 AM-PET scans, with volume and intensity of uptake dramatically higher than in the 137 Onc-PET scans showing myocardial uptake. A myocardial-to-blood SUVmax ratio threshold of 2.20 provided a sensitivity of 87% (27/31) and a specificity of 88% (44/50) for differentiating myocardial uptake between the AM-PET group and an Onc-PET group restricted to patients with clinical characteristics comparable to those of patients in the AM-PET group (≤64 y of age, no coronary artery disease history, and no somatostatin agonists). A myocardial uptake volume threshold of 18 cm3 provided comparable diagnostic accuracy (sensitivity, 84% [26/31]; specificity, 94% [47/50]). Conclusion: Myocardial uptake was detected in 26.9% of somatostatin PET scans recorded for oncology indications. This rate was decreased by somatostatin analog treatments and increased in older individuals. However, somatostatin PET scans, analyzed with the quantitative criterion of uptake intensity or volume, are able to identify AM and to differentiate it from myocardial uptake of other origins.


Subject(s)
Myocarditis , Myocardium , Octreotide , Somatostatin , Adult , Aged , Female , Humans , Male , Middle Aged , Acute Disease , Biological Transport , Diagnosis, Differential , Heart/diagnostic imaging , Myocarditis/diagnostic imaging , Myocarditis/metabolism , Myocardium/metabolism , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Octreotide/analogs & derivatives , Octreotide/metabolism , Octreotide/pharmacokinetics , Organometallic Compounds/pharmacokinetics , Organometallic Compounds/metabolism , Positron-Emission Tomography , Radiopharmaceuticals/pharmacokinetics , Retrospective Studies , Somatostatin/analogs & derivatives , Somatostatin/metabolism
9.
Org Lett ; 26(26): 5447-5452, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38896796

ABSTRACT

Interest in electrocatalytic bioconjugation reactions has surged, particularly for modifying tryptophan and tyrosine residues in proteins. We used a cost-effective graphite felt electrode and low-current methodology to achieve selective bioconjugation of tryptophan with thiophenols, yielding up to 92%. This method exclusively labeled tryptophan residues and incorporated fluorinated tryptophan for NMR analysis. Eight polypeptides, including lanreotide and leuprorelin, were effectively coupled, demonstrating the method's versatility and potential for novel diagnostic and therapeutic agents.


Subject(s)
Peptides , Tryptophan , Tryptophan/chemistry , Peptides/chemistry , Electrochemical Techniques , Molecular Structure , Somatostatin/chemistry , Somatostatin/analogs & derivatives , Peptides, Cyclic/chemistry , Electrodes
11.
Expert Opin Drug Saf ; 23(8): 949-957, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38847075

ABSTRACT

INTRODUCTION: Neuroendocrine neoplasms (NENs) are a rare group of tumors originating from neuroendocrine cells in various organs. They include neuroendocrine tumors (NETs) and neuroendocrine carcinomas (NECs), which differ in biological behavior and prognosis. NETs are usually well-differentiated and slow-growing, while NECs are poorly differentiated and more aggressive. Management of NETs often involves somatostatin analogs like octreotide and lanreotide to control tumor growth and alleviate symptoms, especially in well-differentiated NETs. Lanreotide is used to control tumor growth, and both lanreotide and octreotide alleviate symptoms. Treatment approaches may vary depending on the specific type and grade of the neuroendocrine neoplasm. AREAS COVERED: This review provides an update on the safety of lanreotide autogel in treating patients with NETs, through a comprehensive review of clinical trials, post-marketing surveillance, real-world evidence, and its safety profile. Specific adverse events, side effects, and potential risks associated with lanreotide autogel are discussed, along with risk mitigation strategies and recommendations for patient monitoring. EXPERT OPINION: The findings highlight the overall safety of lanreotide autogel in managing NETs, focusing on its efficacy in controlling hormone secretion, tumor progression, and symptom management. New safety concerns and precautions are also addressed to help healthcare providers make informed decisions when prescribing lanreotide autogel.


Subject(s)
Antineoplastic Agents , Neuroendocrine Tumors , Peptides, Cyclic , Somatostatin , Humans , Somatostatin/analogs & derivatives , Somatostatin/administration & dosage , Somatostatin/adverse effects , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/adverse effects , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/pathology , Gels , Disease Progression
12.
Proc Natl Acad Sci U S A ; 121(26): e2321710121, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38885377

ABSTRACT

Somatostatin receptor 5 (SSTR5) is an important G protein-coupled receptor and drug target for neuroendocrine tumors and pituitary disorders. This study presents two high-resolution cryogenicelectron microscope structures of the SSTR5-Gi complexes bound to the cyclic neuropeptide agonists, cortistatin-17 (CST17) and octreotide, with resolutions of 2.7 Å and 2.9 Å, respectively. The structures reveal that binding of these peptides causes rearrangement of a "hydrophobic lock", consisting of residues from transmembrane helices TM3 and TM6. This rearrangement triggers outward movement of TM6, enabling Gαi protein engagement and receptor activation. In addition to hydrophobic interactions, CST17 forms conserved polar contacts similar to somatostatin-14 binding to SSTR2, while further structural and functional analysis shows that extracellular loops differently recognize CST17 and octreotide. These insights elucidate agonist selectivity and activation mechanisms of SSTR5, providing valuable guidance for structure-based drug development targeting this therapeutically relevant receptor.


Subject(s)
Octreotide , Receptors, Somatostatin , Receptors, Somatostatin/metabolism , Receptors, Somatostatin/agonists , Receptors, Somatostatin/chemistry , Humans , Octreotide/chemistry , Octreotide/pharmacology , Octreotide/metabolism , Neuropeptides/metabolism , Neuropeptides/chemistry , Cryoelectron Microscopy , Protein Binding , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Peptides, Cyclic/metabolism , Somatostatin/metabolism , Somatostatin/chemistry , Somatostatin/analogs & derivatives , Models, Molecular , HEK293 Cells
13.
J Neuroendocrinol ; 36(8): e13420, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38837825

ABSTRACT

[18F]AlF-NOTA-octreotide ([18F]AlF-OC) is a promising alternative for [68Ga]Ga-DOTA-somatostatin analogs (SSAs) in positron emission tomography (PET) imaging of the somatostatin receptor (SSTR). Our aim is to assess changes in TNM staging and differences in patient management between [18F]AlF-OC PET/CT and [68Ga]Ga-DOTA-SSA PET/CT in the work-up of neuroendocrine tumor (NET) patients. Patients who underwent both [18F]AlF-OC and [68Ga]Ga-DOTA-TATE or [68Ga]Ga-DOTA-NOC PET/CT in our multicenter study (Pauwels et al., J Nucl Med.2023;63:632-638) with a NET were included for analysis. TNM staging was determined and compared for both tracers. For each patient, the blinded [68Ga]Ga-DOTA-SSA or [18F]AlF-OC PET/CT images were presented in random order at a multidisciplinary team board. The images were presented together with clinical information and compared with previous SSTR and [18F]FDG PET/CT imaging. After a consensus decision for patient management was recorded, the board was presented with the PET/CT images from the other SSTR tracer and a decision was made for the second tracer. Differences in management were classified as major if it entailed an intermodality change and minor if it led to an intramodality change. Compared with [68Ga]Ga-DOTA-SSA, the use of [18F]AlF-OC led to a change in 16/75 patients: TNM staging changes in 10/75 patients (13.3%; downstaging in 3/10, upstaging in 7/10) and differences in clinical management were seen in 10/75 patients (13.3%), leading to a major difference in 7/10 cases and a minor change in 3/10 cases. All 10 cases with a difference in patient management between both PET tracers were caused by additional lesion detection by [18F]AlF-OC. The use of [18F]AlF-OC did not impact TNM staging or clinical management in the large majority of the patients (86.7%), further validating the potential for routine clinical use of [18F]AlF-OC PET/CT as an alternative for [68Ga]Ga-DOTA-SSA PET/CT. The trial is registered under ClinicalTrials.gov identifier NCT04552847 and EudraCT 2020-000549-15.


Subject(s)
Neuroendocrine Tumors , Positron Emission Tomography Computed Tomography , Humans , Positron Emission Tomography Computed Tomography/methods , Male , Middle Aged , Female , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/pathology , Aged , Prospective Studies , Adult , Octreotide/analogs & derivatives , Radiopharmaceuticals , Somatostatin/analogs & derivatives , Organometallic Compounds , Gallium Radioisotopes , Neoplasm Staging/methods
14.
Clin Endocrinol (Oxf) ; 101(3): 263-273, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38865284

ABSTRACT

OBJECTIVE: Acromegaly is associated with increased morbidity and mortality if left untreated. The therapeutic options include surgery, medical treatment, and radiotherapy. Several guidelines and recommendations on treatment algorithms and follow-up exist. However, not all recommendations are strictly evidence-based. To evaluate consensus on the treatment and follow-up of patients with acromegaly in the Nordic countries. METHODS: A Delphi process was used to map the landscape of acromegaly management in Denmark, Sweden, Norway, Finland, and Iceland. An expert panel developed 37 statements on the treatment and follow-up of patients with acromegaly. Dedicated endocrinologists (n = 47) from the Nordic countries were invited to rate their extent of agreement with the statements, using a Likert-type scale (1-7). Consensus was defined as ≥80% of panelists rating their agreement as ≥5 or ≤3 on the Likert-type scale. RESULTS: Consensus was reached in 41% (15/37) of the statements. Panelists agreed that pituitary surgery remains first line treatment. There was general agreement to recommend first-generation somatostatin analog (SSA) treatment after failed surgery and to consider repeat surgery. In addition, there was agreement to recommend combination therapy with first-generation SSA and pegvisomant as second- or third-line treatment. In more than 50% of the statements, consensus was not achieved. Considerable disagreement existed regarding pegvisomant monotherapy, and treatment with pasireotide and dopamine agonists. CONCLUSION: This consensus exploration study on the management of patients with acromegaly in the Nordic countries revealed a relatively large degree of disagreement among experts, which mirrors the complexity of the disease and the shortage of evidence-based data.


Subject(s)
Acromegaly , Delphi Technique , Somatostatin , Acromegaly/therapy , Humans , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Scandinavian and Nordic Countries/epidemiology , Consensus , Human Growth Hormone/therapeutic use , Human Growth Hormone/analogs & derivatives , Surveys and Questionnaires
15.
Endocr Relat Cancer ; 31(9)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38913539

ABSTRACT

Prospective data are lacking on early somatostatin analog (SSA) therapy in bronchopulmonary neuroendocrine tumors (BP-NETs; typical carcinoids and atypical carcinoids (TCs and ACs)). SPINET (EudraCT: 2015-004992-62; NCT02683941) was a phase III, double-blind study of lanreotide autogel/depot (LAN; 120 mg every 28 days) plus best supportive care (BSC) vs placebo plus BSC, with an optional open-label treatment phase (LAN plus BSC). Patients had metastatic/unresectable, somatostatin receptor (SSTR)-positive TCs or ACs. Recruitment was stopped early owing to slow accrual; eligible patients from the double-blind phase transitioned to open-label LAN. The adapted primary endpoint was progression-free survival (PFS) during either phase for patients receiving LAN. Seventy-seven patients were randomized (LAN, n = 51 (TCs, n = 29; ACs, n = 22); placebo, n = 26 (TCs, n = 16; ACs, n = 10)). Median (95% CI) PFS during double-blind and open-label phases in patients receiving LAN was 16.6 (11.3; 21.9) months overall (primary endpoint), 21.9 (12.8, not calculable (NC)) months in TCs, and 13.8 (5.4; 16.6) months in ACs. During double-blind treatment, median (95% CI) PFS was 16.6 (11.3; 21.9) months for LAN vs 13.6 (8.3; NC) months for placebo (not significant); corresponding values were 21.9 (13.8; NC) and 13.9 (13.4; NC) months, respectively, in TCs and 13.8 (5.4; 16.6) and 11.0 (2.8; 16.9) months, respectively, in ACs. Patients' quality of life did not deteriorate and LAN was well tolerated. Although recruitment stopped early and the predefined sample size was not met, SPINET is the largest prospective study to date of SSA therapy in SSTR-positive TCs and ACs and suggests clinical benefit in TCs.


Subject(s)
Neuroendocrine Tumors , Peptides, Cyclic , Somatostatin , Humans , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Somatostatin/administration & dosage , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/therapeutic use , Female , Male , Middle Aged , Double-Blind Method , Aged , Neuroendocrine Tumors/drug therapy , Adult , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/administration & dosage , Lung Neoplasms/drug therapy , Aged, 80 and over
16.
Neuroendocrinology ; 114(8): 775-785, 2024.
Article in English | MEDLINE | ID: mdl-38824926

ABSTRACT

INTRODUCTION: Aims of the study were to assess the differences in the diagnostic efficacy of 68Ga-somatostatin receptor analogs (68Ga-SSAs) and 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) for detecting bone metastases in neuroendocrine neoplasm (NEN) and to analyze the correlation between imaging features and clinical features of BMs. METHODS: We retrospectively analyzed the clinical and imaging data of 213 NEN patients who underwent 68Ga-SSA PET/CT and were finally diagnosed as BMs by pathology or follow-up. Of those, 103 patients underwent 18F-FDG PET/CT within 7 days after 68Ga-SSA PET/CT. RESULT: The BM detection rate of 68Ga-SSA PET/CT was higher than 18F-FDG PET/CT (86.4% vs. 66.0%, p = 0.02) in 103 patients with dual scanning. Meanwhile, the number of positive lesions in 68Ga-SSA PET/CT was significantly more than in 18F-FDG PET/CT (3.37 ± 1.95 vs. 2.23 ± 2.16, t = 4.137, p < 0.001). Most bone metastasis lesions presented as osteogenic change in CT (55.4%, 118/213). Concerning the primary tumor, the most frequent were of pancreatic origin (26.3%, 56/213), followed by rectal origin (22.5%, 48/213), thymic origin in 33 cases (15.5%), pulmonary origin in 29 cases (13.6%), paraganglioma in 20 cases (9.4%). The efficiency of 68Ga-SSA PET/CT to detect BMs was significantly correlated with the primary site (p = 0.02), with thymic carcinoid BMs being the most difficult to detect, and the positive rate was only 60.6% (20/33). However, 18F-FDG PET/CT positive rate was 76.92% (10/13) in thymic carcinoid BMs. In addition, the BMs of 7 patients in this study were detected by 68Ga-SSA PET earlier than CT for 4.57 months (range: 2-10 months). CONCLUSION: 68Ga-SSA PET/CT has higher sensitivity for detecting the BMs of NEN than 18F-FDG and detects the BM earlier than CT. Moreover, 18F-FDG PET/CT should be a complement for diagnosing the BMs of thymic carcinoids.


Subject(s)
Bone Neoplasms , Fluorodeoxyglucose F18 , Neuroendocrine Tumors , Positron Emission Tomography Computed Tomography , Humans , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/pathology , Male , Female , Middle Aged , Bone Neoplasms/secondary , Bone Neoplasms/diagnostic imaging , Retrospective Studies , Aged , Adult , Radiopharmaceuticals , Somatostatin/analogs & derivatives , Gallium Radioisotopes , Aged, 80 and over
17.
Int J Mol Sci ; 25(10)2024 May 19.
Article in English | MEDLINE | ID: mdl-38791582

ABSTRACT

A novel nanotechnology-based drug delivery system (DDS) targeted at pancreatic cancer cells was developed, characterized, and tested. The system consisted of liposomes as carriers, an anticancer drug (paclitaxel) as a chemotherapeutic agent, and a modified synthetic somatostatin analog, 5-pentacarbonyl-octreotide, a ligand for somatostatin receptor 2 (SSTR2), as a targeting moiety for pancreatic cancer. The cellular internalization, cytotoxicity, and antitumor activity of the DDS were tested in vitro using human pancreatic ductal adenocarcinoma (PDAC) cells with different expressions of the targeted SSTR2 receptors, and in vivo on immunodeficient mice bearing human PDAC xenografts. The targeted drug delivery system containing paclitaxel exhibited significantly enhanced cytotoxicity compared to non-targeted DDS, and this efficacy was directly related to the levels of SSTR2 expression. It was found that octreotide-targeted DDS proved exceptionally effective in suppressing the growth of PDAC tumors. This study underscores the potential of octreotide-targeted liposomal delivery systems to enhance the therapeutic outcomes for PDAC compared with non-targeted liposomal DDS and Paclitaxel-Cremophor® EL, suggesting a promising avenue for future cancer therapy innovations.


Subject(s)
Drug Delivery Systems , Liposomes , Octreotide , Paclitaxel , Pancreatic Neoplasms , Receptors, Somatostatin , Xenograft Model Antitumor Assays , Animals , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Receptors, Somatostatin/metabolism , Mice , Cell Line, Tumor , Paclitaxel/pharmacology , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Liposomes/chemistry , Drug Delivery Systems/methods , Octreotide/administration & dosage , Octreotide/pharmacology , Somatostatin/analogs & derivatives , Nanotechnology/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology
18.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL