Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 427
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1348990, 2024.
Article in English | MEDLINE | ID: mdl-38405148

ABSTRACT

Pasireotide is a somatostatin analogue for the treatment of acromegaly, a chronic condition caused by excess growth hormone. Despite the therapeutic benefits of pasireotide as a second-line treatment for inadequately controlled acromegaly, a major concern is its hyperglycemic side-effect. Here, we provide guidance on how to select appropriate patients with acromegaly for treatment with pasireotide. We summarize baseline characteristics of patients at high risk for pasireotide-associated hyperglycemia and recommend a monitoring strategy based on the risk profile. Self-monitoring of blood glucose levels (SMBG), measurements of fasting plasma glucose (FPG), postprandial plasma glucose (PPG) and regular HbA1c measurements are the foundation of our proposed monitoring approach. The pathophysiology of pasireotide-induced hyperglycemia involves decreased secretion of the incretin hormones GIP (glucose-dependent insulinotropic polypeptide) and GLP-1 (glucagon-like peptide-1). Our expert recommendations address the specific pathophysiology of pasireotide-induced hyperglycemia by recommending the incretin-based therapeutics dipeptidyl peptidase-4 inhibitors (DPP-4i) and glucagon-like peptide-1 receptor agonists (GLP-1 RA) in all appropriate patients as an alternative to first-line monotherapy with metformin. Furthermore, we emphasize the importance of adequate control of acromegaly, excellent diabetes education, nutrition and lifestyle guidance and advise to consult expert diabetologists in case of uncertainty in the management of patients with hyperglycemia under pasireotide.


Subject(s)
Acromegaly , Hyperglycemia , Somatostatin/analogs & derivatives , Humans , Acromegaly/drug therapy , Blood Glucose , Incretins , Somatostatin/adverse effects , Glucagon-Like Peptide 1
2.
Int J Clin Pharmacol Ther ; 62(1): 29-36, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37942928

ABSTRACT

OBJECTIVES: This study aimed to explore the risk factors of hypoglycemia in patients with gastrointestinal bleeding caused by somatostatin for injection and to establish a prediction model based on logistic regression combined with receiver operating characteristic (ROC) curve. MATERIALS AND METHODS: This retrospective study analyzed patients diagnosed with gastrointestinal bleeding and treated with somatostatin from January 2022 to May 2023 and collected hypoglycemic events. Univariate and multivariate logistic regression analysis were used to determine the independent influencing factors of somatostatin-induced hypoglycemia, and a prediction model was established. ROC analysis was used to evaluate the prediction model. RESULTS: A total of 331 patients were enrolled in this study, and 42 patients developed hypoglycemic events. Age and co-infection were found to be significant risk factors for hypoglycemia in patients with gastrointestinal bleeding induced by somatostatin. Binary logistic regression fitting established the hypoglycemia prediction model Logit (P) = -4.125+0.053Yage+1.366Yco-infection (co-infection: Xco-infection = 1, non-co-infection: Xno co-infection = 0), Hosmer-Lemeshow test results showed that the model had a good fit (χ2 = 10.552, df = 8, p = 0.228), and the AUC of the prediction model to predict the risk of hypoglycemia caused by somatostatin in patients with gastrointestinal bleeding was 0.744 (95% CI: 0.653 - 0.835, p < 0.001), the sensitivity was 57.14%, and the specificity was 93.77%. CONCLUSION: Among adult patients with gastrointestinal bleeding treated with somatostatin for injection, our study found that age and co-infection were significant risk factors for somatostatin-induced hypoglycemia in this patient population, and the fitted models had high predictive value in predicting the occurrence of hypoglycemia.


Subject(s)
Coinfection , Hypoglycemia , Adult , Humans , Retrospective Studies , Risk Factors , ROC Curve , Hypoglycemia/chemically induced , Hypoglycemia/epidemiology , Gastrointestinal Hemorrhage/chemically induced , Gastrointestinal Hemorrhage/diagnosis , Gastrointestinal Hemorrhage/epidemiology , Hypoglycemic Agents , Somatostatin/adverse effects
3.
J Hematol Oncol ; 16(1): 119, 2023 12 14.
Article in English | MEDLINE | ID: mdl-38098114

ABSTRACT

In retrospective studies, metformin use has been associated with better clinical outcomes in diabetic patients with advanced, well-differentiated neuroendocrine tumors (WDNETs). However, prospective evidence of metformin safety and activity is lacking. Here, we conducted the first-in-human phase Ib MetNET2 trial to investigate the safety and antitumor activity of metformin in combination with the somatostatin analog lanreotide autogel (ATG) in both diabetic and non-diabetic patients with advanced WDNETs of the gastrointestinal (GI) or thoracic tract. Enrolled patients received lanreotide ATG 120 mg plus oral metformin, up to a maximum dosage of 2550 mg/day. We enrolled 20 patients, of whom 18 (90%) and 2 (10%) had WDNETs of the GI and thoracic tract, respectively. Fourteen patients (70%) were non-diabetic. With a 5% incidence of SAEs, the study met its primary objective of demonstrating treatment safety. With a median follow-up of 39 months (95% CI 28-NE), median PFS was 24 months (95% CI 16-NE), with 12-month and 24-month PFS probability of 75% (95% CI 58-97) and 49% (95% CI 31-77), respectively. We found no statistically significant PFS differences between diabetic and non-diabetic patients. Among exploratory analyses, the presence of tumor genomic alterations in DNA damage pathways was associated with trend towards worse PFS, whereas a precocious reduction of HOMA-IR index and plasma cholesterol concentration showed a trend towards an association with better PFS. In conclusion, metformin plus lanreotide ATG is a safe and well tolerated combination treatment that is associated with promising antitumor activity in both non-diabetic and diabetic patients with WDNETs, and that warrants further investigation in larger clinical trials.


Subject(s)
Diabetes Mellitus , Metformin , Neuroendocrine Tumors , Pancreatic Neoplasms , Humans , Metformin/pharmacology , Metformin/therapeutic use , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/chemically induced , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Retrospective Studies , Prospective Studies , Somatostatin/adverse effects , Diabetes Mellitus/chemically induced , Diabetes Mellitus/drug therapy , Lung/pathology
4.
Eur J Drug Metab Pharmacokinet ; 48(6): 665-674, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37751056

ABSTRACT

BACKGROUND: In patients with kidney or hepatic diseases, an increment of circulating pasireotide is also expected. Therefore, this open-label, phase I study aimed to evaluate the pharmacokinetic profiles and safety of subcutaneous (SC) and long-acting release (LAR) intramuscular injections of pasireotide in male Taiwanese volunteers who are hyperendemic hepatitis B/C and chronic kidney disease (CKD). METHODS: A total of 45 male volunteers were randomized to receive one of nine treatment sequences, involving a single subcutaneous injection of 300, 600, or 900 µg pasireotide, a multiple SC injection of the same dosage of pasireotide [300, 600, or 900 µg, twice daily (b.i.d.) for 4 days and a single dose for 1 day], and a single dose of 20, 40, or 60 mg LAR pasireotide intramuscular injection. The pasireotide SC and LAR formulations were prepared and supplied to the study center by Novartis. Pharmacokinetic parameters were assessed from both formulations. All adverse events that occurred in participants throughout the study period, including abnormalities in fasting levels of glucose, insulin, and glucagon, as well as laboratory measurements and electrocardiograms, were recorded. RESULTS: Analysis of plasma concentration over time revealed a rapid absorption of pasireotide, with a maximal concentration at 0.5 h after SC injection(s) of pasireotide (300-900 µg). Following a single dose of pasireotide LAR (20-60 mg), a sustained release was observed following an initial increase on day 1, a plateau around day 20, and a decline over the next 7 weeks. CONCLUSIONS: Both pasireotide formulations showed dose-proportional pharmacokinetics and 300-900 µg of SC pasireotide and 20-60 mg LAR pasireotide treatment showed favorable safety profiles and was well-tolerated when administered in male Taiwanese volunteers who are hyperendemic hepatitis B/C and CKD.


Subject(s)
Hepatitis B , Renal Insufficiency, Chronic , Humans , Male , Somatostatin/adverse effects , Insulin , Renal Insufficiency, Chronic/drug therapy , Hepatitis B/chemically induced , Hepatitis B/drug therapy
5.
Adv Ther ; 40(2): 671-690, 2023 02.
Article in English | MEDLINE | ID: mdl-36502449

ABSTRACT

INTRODUCTION: Real-world data evaluating patients' injection experiences using the latest devices/formulations of the long-acting (LA) somatostatin analogs (SSAs) lanreotide Autogel/Depot (LAN; Somatuline®) and octreotide LA release (OCT; Sandostatin®) are limited. METHODS: PRESTO 2 was a 2020/2021 e-survey comparing injection experience of adults with neuroendocrine tumors (NETs) or acromegaly treated with LAN prefilled syringe versus OCT syringe for > 3 months in Canada, Ireland, the UK and the USA (planned sample size, 304). PRIMARY ENDPOINT: the proportion of patients with injection-site pain lasting > 2 days after their most recent injection, analyzed using a multivariate logistic regression model. Secondary endpoints included interference with daily life due to injection-site pain and technical injection problems in patients with current SSA use for ≥ 6 months. RESULTS: There were 304 respondents (acromegaly, n = 85; NETs, n = 219; LAN, n = 168; OCT, n = 136; 69.2% female; mean age, 59.6 years). Fewer patients had injection-site pain lasting > 2 days after the most recent injection with LAN (6.0%) than OCT (22.8%); the odds of pain lasting > 2 days were significantly lower for LAN than OCT, adjusted for disease subgroup and occurrence of injection-site reactions (odds ratio [95% confidence interval]: 0.13 [0.06-0.30]; p < 0.0001). Injection-site pain interfered with daily life "a little bit" or "quite a bit" in 37.2% and 3.8% (LAN) versus 52.5% and 7.5% (OCT) of patients, respectively. Among patients with ≥ 6 months' experience with current SSA (92.4% of patients), technical injection problems never occurred in 76.8% (LAN) and 42.9% (OCT) of patients. CONCLUSIONS: Compared with OCT, significantly fewer patients using LAN had injection-site pain lasting > 2 days after their most recent injection. Also, fewer LAN-treated patients experienced technical problems during injection. These findings demonstrate the importance of injection modality for overall LA SSA injection experience for patients with acromegaly or NETs.


Patients with neuroendocrine tumors or acromegaly often receive long-term monthly treatment with somatostatin analogs. These injectable drugs stop the body from making an excess of certain hormones. Understanding patients' experiences of these injections helps to provide better care. The PRESTO 2 online study surveyed 304 patients in Canada, Ireland, the UK and the USA with neuroendocrine tumors or acromegaly who were being treated with a somatostatin analog, either lanreotide Autogel/Depot (LAN) or octreotide long-acting release (OCT). The survey asked about injection experience, including injection-site pain lasting > 2 days and how it affected patients' lives, anxiety before injections and technical problems during injections (like syringe blockages). The survey showed fewer patients receiving LAN than OCT had injection-site pain that lasted > 2 days, and fewer said that the pain interfered with their daily lives. There were fewer technical injection problems with LAN than with OCT. However, more patients receiving LAN than OCT felt anxious before their injection. In some countries (including Canada, Ireland and the UK, but not the USA), the patient (or family member/friend) can inject LAN if they are on a stable dose, their doctor agrees, and they received training. A nurse/doctor must inject OCT. In PRESTO 2, about 40% of non-US patients who were eligible injected themselves (or were helped by a family member/friend). This may explain why more patients reported anxiety in the LAN group. PRESTO 2 provides important insights into patients' experiences of receiving somatostatin analogs and helps identify areas for improving patient care.


Subject(s)
Acromegaly , Injections , Neuroendocrine Tumors , Octreotide , Somatostatin , Female , Humans , Male , Middle Aged , Acromegaly/drug therapy , Neuroendocrine Tumors/drug therapy , Octreotide/adverse effects , Somatostatin/administration & dosage , Somatostatin/adverse effects , Somatostatin/analogs & derivatives , Surveys and Questionnaires , Delayed-Action Preparations/administration & dosage , Injections/adverse effects , Injections/instrumentation , Injections/methods
6.
HPB (Oxford) ; 24(11): 2029-2034, 2022 11.
Article in English | MEDLINE | ID: mdl-35953409

ABSTRACT

BACKGROUND: Clinically relevant postoperative pancreatic fistula (CR-POPF) is a significant contributor to morbidity after pancreatectomy. Somatostatin analogues have shown variable efficacy in the prevention of CR-POPF. Lanreotide is a somatostatin analogue ideally suited for perioperative use due to its long half-life and favorable side effect profile. METHODS: We conducted a phase II single-arm trial of a single dose of preoperative lanreotide (120 mg) in patients undergoing either pancreaticoduodenectomy (PD) or distal pancreatectomy (DP). The primary outcome was development of CR-POPF or intra-abdominal abscess. Secondary outcomes included biochemical leak and overall morbidity. RESULTS: A total of 98 patients completed the study. Sixty-two underwent PD (63.3%) and 36 underwent DP (36.7%). The primary outcome was observed in eight (8%) patients in the overall cohort, one from the DP group and seven from the PD group. Biochemical leak was detected in 12 (12.2%) patients in the overall cohort. Twenty-seven (27.5%) patients developed complications, of which 14 (14.2%) were major complications. Drug-related adverse events were limited to mild skin reactions in two (2%) patients. CONCLUSION: Patients who received preoperative lanreotide developed CR-POPF at rates significantly lower than historical controls or published literature. This provides strong justification for a randomized controlled trial.


Subject(s)
Pancreatic Fistula , Postoperative Complications , Somatostatin , Humans , Pancreatectomy/adverse effects , Pancreatic Fistula/etiology , Pancreatic Fistula/prevention & control , Pancreaticoduodenectomy/adverse effects , Postoperative Complications/etiology , Postoperative Complications/prevention & control , Retrospective Studies , Risk Factors , Somatostatin/adverse effects
7.
Rev Assoc Med Bras (1992) ; 68(4): 514-518, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35649076

ABSTRACT

OBJECTIVE: Long-acting depot formulations of somatostatin analogs, i.e., octreotide and lanreotide, are the first-line medical therapies for patients with acromegaly to whom surgery/radiotherapy cannot be performed or who have inadequate response. In this study, we aimed to evaluate the short-term local and systemic adverse reactions developed after the somatostatin analogs injections in the patients with acromegaly, in order to compare the side effects of somatostatin analogs injections. METHODS: Patients diagnosed with acromegaly who were referred to our endocrinology clinic for monthly somatostatin analogs injections were questionnaired. Wong-Baker Faces Pain Rating Scale was used to evaluate the injection-site pain at the time of injection. The existence of leg pain, nausea, diarrhea, and abdominal pain following the previous injection was also investigated during the next injection. RESULTS: A total of 49 patients were included in the study. The statistical difference could not be shown between the injection-site pain, anorexia, and leg pain frequencies of the groups, while the frequency of gastrointestinal disturbances, i.e., diarrhea and abdominal pain, was significantly lower in the octreotide group (p<0.001 and p=0.015, respectively). CONCLUSIONS: This is the first prospective study that compared the severity of the injection-site pain by using a scoring scale, following the long-acting somatostatin analogs injections. We have shown that there was no significant association of the injection-site pain severity with the somatostatin analogs regimen nor the dose differences within each somatostatin analogs treatment.


Subject(s)
Acromegaly , Abdominal Pain/chemically induced , Acromegaly/drug therapy , Diarrhea , Humans , Octreotide/adverse effects , Prospective Studies , Somatostatin/adverse effects
8.
Curr Treat Options Oncol ; 23(7): 1001-1013, 2022 07.
Article in English | MEDLINE | ID: mdl-35501552

ABSTRACT

OPINION STATEMENT: Neuroendocrine tumors (NET) represent a complex and heterogeneous group of malignancies arising from the diffuse endocrine cells and other cells derived from the neural crest. Advanced disease is observed at diagnosis in more than one-third of patients. Somatostatin analogs (SSA) are the cornerstone in advanced well-differentiated NET treatment. Unfortunately, most patients will eventually develop resistance to SSA treatment by different mechanisms that are not fully understood. In some cases of refractory carcinoid syndrome or progressive disease, the increase of SSA dose may help to reach out a symptomatic and/or tumor growth control. The clinical evidence behind above-label SSA administration is limited and should be individualized and discussed patient by patient. Some questions regarding high-dose SSA use are unsolved, such as the optimal dose to use, the frequency of administration, or the need of deepen molecular understanding that could help to adequately select patients for this approach.


Subject(s)
Malignant Carcinoid Syndrome , Neuroendocrine Tumors , Humans , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Somatostatin/adverse effects
9.
J Clin Endocrinol Metab ; 107(8): e3115-e3120, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35587448

ABSTRACT

CONTEXT: Congenital hyperinsulinism (HI) results in severe, persistent hypoglycemia and is associated with high risk of neurodevelopmental deficits. Sixty percent of HI cases are unresponsive to diazoxide, the only Food and Drug Administration-approved drug. Somatostatin analogs are used off-label as second-line treatment; the long-acting somatostatin analogue, lanreotide, has been used to treat HI over the past decade. Existing reports are limited to small case series. OBJECTIVE: To assess the effectiveness and safety of lanreotide in individuals with HI. DESIGN: Retrospective cohort study of individuals with HI treated with lanreotide between 2015 and 2020. SETTING: The Congenital Hyperinsulinism Center at The Children's Hospital of Philadelphia. PATIENTS: Fifty-four individuals with hyperinsulinism treated with lanreotide. MAIN OUTCOME MEASURES: Fasting duration with plasma glucose > 70 mg/dL; frequency of lanreotide-associated side effects. RESULTS: The median duration of lanreotide therapy was 28.7 (2.8-64.5) months. Thirty-four patients (63%) had HI due to inactivating mutations of the adenosine 5'-triphosphate (ATP) sensitive potassium channel (KATP-HI), and 39% had undergone a pancreatectomy. Of 52 patients receiving other HI therapies, 22 (42%) were able to discontinue other treatments and were managed on lanreotide alone. Fasting duration with plasma glucose > 70 mg/dL was significantly longer during therapy with lanreotide compared to prior to lanreotide initiation (8.6 ±â€…6.5 vs 5.1 ±â€…4.7 hours, P = 0.001). The most common side effects were subcutaneous nodules (26%) and gallstones (11%). CONCLUSIONS: Lanreotide is a well-tolerated treatment for patients with HI. It results in a longer duration of fasting and a simplification of treatment regimens.


Subject(s)
Congenital Hyperinsulinism , Hyperinsulinism , Blood Glucose , Child , Congenital Hyperinsulinism/genetics , Humans , Hyperinsulinism/complications , Peptides, Cyclic/adverse effects , Peptides, Cyclic/therapeutic use , Retrospective Studies , Somatostatin/adverse effects , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use
10.
Anesthesiology ; 136(5): 802-822, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35303056

ABSTRACT

BACKGROUND: Slick, a sodium-activated potassium channel, has been recently identified in somatosensory pathways, but its functional role is poorly understood. The authors of this study hypothesized that Slick is involved in processing sensations of pain and itch. METHODS: Immunostaining, in situ hybridization, Western blot, and real-time quantitative reverse transcription polymerase chain reaction were used to investigate the expression of Slick in dorsal root ganglia and the spinal cord. Mice lacking Slick globally (Slick-/-) or conditionally in neurons of the spinal dorsal horn (Lbx1-Slick-/-) were assessed in behavioral models. RESULTS: The authors found Slick to be enriched in nociceptive Aδ-fibers and in populations of interneurons in the spinal dorsal horn. Slick-/- mice, but not Lbx1-Slick-/- mice, showed enhanced responses to noxious heat in the hot plate and tail-immersion tests. Both Slick-/- and Lbx1-Slick-/- mice demonstrated prolonged paw licking after capsaicin injection (mean ± SD, 45.6 ± 30.1 s [95% CI, 19.8 to 71.4]; and 13.1 ± 16.1 s [95% CI, 1.8 to 28.0]; P = 0.006 [Slick-/- {n = 8} and wild-type {n = 7}, respectively]), which was paralleled by increased phosphorylation of the neuronal activity marker extracellular signal-regulated kinase in the spinal cord. In the spinal dorsal horn, Slick is colocalized with somatostatin receptor 2 (SSTR2), and intrathecal preadministration of the SSTR2 antagonist CYN-154806 prevented increased capsaicin-induced licking in Slick-/- and Lbx1-Slick-/- mice. Moreover, scratching after intrathecal delivery of the somatostatin analog octreotide was considerably reduced in Slick-/- and Lbx1-Slick-/- mice (Slick-/- [n = 8]: 6.1 ± 6.7 bouts [95% CI, 0.6 to 11.7]; wild-type [n =8]: 47.4 ± 51.1 bouts [95% CI, 4.8 to 90.2]; P = 0.039). CONCLUSIONS: Slick expressed in a subset of sensory neurons modulates heat-induced pain, while Slick expressed in spinal cord interneurons inhibits capsaicin-induced pain but facilitates somatostatin-induced itch.


Subject(s)
Capsaicin , Posterior Horn Cells , Animals , Capsaicin/adverse effects , Capsaicin/metabolism , Ganglia, Spinal/metabolism , Mice , Pain , Posterior Horn Cells/metabolism , Potassium Channels , Pruritus/chemically induced , Sensory Receptor Cells/metabolism , Sodium Channels , Somatostatin/adverse effects , Somatostatin/metabolism , Spinal Cord/metabolism
11.
J Nucl Med ; 63(9): 1326-1333, 2022 09.
Article in English | MEDLINE | ID: mdl-34992153

ABSTRACT

Peptide receptor radiotherapy with somatostatin analogs has been successfully used for years as a treatment for somatostatin-overexpressing tumors. Treatment of neuroendocrine tumors (NETs) with the ß-particle emitter 177Lu-DOTATATE is currently considered the standard of care for subjects with gastroenteropancreatic NETs. Despite the success of 177Lu-DOTATATE, there remains significant room for improvement in terms of both safety and efficacy. Targeted α-emitter therapy with isotopes such as 212Pb has the potential to improve both. Here, we present the preliminary results of the phase 1 first-in-humans dose-escalation trial evaluating 212Pb-DOTAMTATE (a bifunctional metal chelator [DOTAM] and the SSTR-targeting peptide [TATE]) in patients with somatostatin receptor-positive NETs. Methods: Twenty subjects with histologically confirmed NETs, prior positive somatostatin analog scans, and no prior history of 177Lu/90Y/111In peptide receptor radiotherapy, with different primary sites of the disease, were enrolled. Treatment began with single ascending doses of 212Pb-DOTAMTATE, with subsequent cohorts receiving an incremental 30% dose increase, which was continued until a tumor response or a dose-limiting toxicity was observed. This was followed by a multiple ascending dose regimen. The recommended phase 2 dose regimen consisted of 4 cycles of 2.50 MBq/kg (67.6 µCi/kg) of 212Pb-DOTAMTATE administered at 8-wk intervals, intravenously. Results: Ten subjects received the highest dose, 2.50 MBq/kg/cycle (67.6 µCi/kg/cycle). Treatment was well tolerated, with the most common treatment-emergent adverse events being nausea, fatigue, and alopecia. No serious treatment-emergent adverse events were related to the study drug, and no subjects required treatment delay or a dose reduction. An objective radiologic response of 80% was observed for the first 10 subjects treated at the recommended phase 2 dose. Conclusion: Targeted α-therapy with 212Pb-DOTAMTATE has been shown to be well tolerated. Preliminary efficacy results are highly promising. If these results are confirmed in a larger, multicenter clinical trial, 212Pb-DOTAMTATE would provide a substantial benefit over currently Food and Drug Administration-approved therapies for patients with metastatic or inoperable SSTR-expressing NETs regardless of the grade and location of the primary tumor.


Subject(s)
Neuroendocrine Tumors , Organometallic Compounds , Chelating Agents/adverse effects , Humans , Lead , Lead Radioisotopes , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/radiotherapy , Octreotide/adverse effects , Organometallic Compounds/adverse effects , Positron-Emission Tomography , Radionuclide Imaging , Radiopharmaceuticals/adverse effects , Receptors, Somatostatin , Somatostatin/adverse effects
12.
J Clin Endocrinol Metab ; 107(2): 297-308, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34618894

ABSTRACT

Currently, the first-generation somatostatin receptor ligands (fg-SRLs), octreotide LAR and lanreotide autogel, are the mainstays of acromegaly treatment and achieve biochemical control in approximately 40% of patients and tumor shrinkage in over 60% of patients. Pasireotide, a second-generation SRL, shows higher efficacy with respect to both biochemical control and tumor shrinkage but has a worse safety profile. In this review, we discuss the future perspectives of currently available SRLs, focusing on the use of biomarkers of response and precision medicine, new formulations of these SRLs and new drugs, which are under development. Precision medicine, which is based on biomarkers of response to treatment, will help guide the decision-making process by allowing physicians to choose the appropriate drug for each patient and improving response rates. New formulations of available SRLs, such as oral, subcutaneous depot, and nasal octreotide, may improve patients' adherence to treatment and quality of life since there will be more options available that better suit each patient. Finally, new drugs, such as paltusotine, somatropin, ONO-5788, and ONO-ST-468, may improve treatment adherence and present higher efficacy than currently available drugs.


Subject(s)
Acromegaly/drug therapy , Antineoplastic Agents, Hormonal/administration & dosage , Growth Hormone-Secreting Pituitary Adenoma/drug therapy , Receptors, Somatostatin/metabolism , Acromegaly/blood , Acromegaly/etiology , Antineoplastic Agents, Hormonal/adverse effects , Biomarkers, Tumor/blood , Growth Hormone-Secreting Pituitary Adenoma/blood , Growth Hormone-Secreting Pituitary Adenoma/complications , Growth Hormone-Secreting Pituitary Adenoma/genetics , Humans , Octreotide/administration & dosage , Octreotide/adverse effects , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/adverse effects , Precision Medicine/methods , Precision Medicine/trends , Quality of Life , Randomized Controlled Trials as Topic , Somatostatin/administration & dosage , Somatostatin/adverse effects , Somatostatin/analogs & derivatives , Treatment Outcome
13.
Pituitary ; 25(1): 180-190, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34498217

ABSTRACT

CONTEXT: Pasireotide-LAR, a second-generation somatostatin receptor ligand (SRL), is recommended for patients with acromegaly as second-line treatment. Its efficacy and safety were assessed in clinical trials; however, the real-world evidence is still scarce. OBJECTIVE: The aim of this study was to evaluate the impact of 1-year treatment with pasireotide-LAR on disease control and glucose metabolism in acromegaly patients resistant to first-generation SRLs. DESIGN: A single-center prospective study. METHODS: Twenty-eight patients with active acromegaly or acrogigantism on first-generation SRLs following ineffective pituitary surgery were switched to treatment with pasireotide-LAR 40 or 60 mg i.m. every 28 days. To assess the efficacy of the treatment GH and IGF-1 levels were measured every 3 months. Safety of treatment was carefully evaluated, especially its impact on glucose metabolism. RESULTS: Complete biochemical control (GH ≤ 1 ng/mL and IGF-1 ≤ 1 × ULN) was achieved in 26.9% of patients and partial + complete response (GH ≤ 2.5 ng/mL and IGF-1 ≤ 1.3 × ULN) in 50.0% of patients. Mean GH level decrease was the largest within first 6 months (P = 0.0001) and mean IGF-1 level decreased rapidly within the first 3 months (P < 0.0001) and they remained reduced during the study. Blood glucose and HbA1c levels increased significantly within 3 months (P = 0.0001) and stayed on stable level thereafter. Otherwise, the treatment was well tolerated and clinical improvement was noticed in majority of patients. CONCLUSIONS: This real-life study confirmed good effectiveness of pasireotide-LAR in patients resistant to first-generation SRLs. Pasireotide-LAR was overall safe and well tolerated, however significant glucose metabolism worsening was noted.


Subject(s)
Acromegaly , Somatostatin , Acromegaly/drug therapy , Human Growth Hormone , Humans , Insulin-Like Growth Factor I , Octreotide , Prospective Studies , Somatostatin/adverse effects , Somatostatin/analogs & derivatives , Treatment Outcome
14.
Clin Ther ; 43(12): 2136-2145.e2, 2021 12.
Article in English | MEDLINE | ID: mdl-34844770

ABSTRACT

PURPOSE: Inoperable malignant intestinal obstruction (IMIO) is a severe complication in patients with cancer, usually gastrointestinal or gynecologic in origin. For patients with IMIO, there is a need to relieve symptoms and limit nasogastric tube (NGT) use. Previous studies have suggested the efficacy of somatostatin analogues in relieving obstruction-related symptoms, such as nausea, vomiting, and pain. The purpose of this study was to assess the efficacy of lanreotide autogel 120 mg (LAN 120 mg) in the management of symptoms resulting from IMIO in patients with advanced cancer. METHODS: This single-arm, multicenter study enrolled 52 patients mostly with advanced gastrointestinal or ovarian malignant tumors (35 patients with NGT and 17 patients without NGT). Patients received 1 deep subcutaneous injection of LAN 120 mg. Evaluations were performed on days 7, 14, and 28. The primary end point was the percentage of responding patients before or at day 7. Response was defined as ≤2 vomiting episodes per day (for patients without NGT at baseline) or no vomiting recurrence (after NGT removal) during at least 3 consecutive days at any time point between treatment and day 7. Responders at day 28 were offered a second LAN 120 mg injection and followed up until day 56. FINDINGS: The proportion of responders in the intention-to-treat population was 24 of 52 (46.2%), which was significantly greater than the reference proportion of 30% (P = 0.0055). Patients without NGT had a higher response (88.2%) than patients with NGT (25.7%) and had a steady trend for clinical improvement that led to sustainable responses. Median time to response was 9 days for the overall population, 3 days for patients without NGT, and 14 days for patients with NGT (P < 0.0001). IMPLICATIONS: Our study is the first to use long-acting LAN 120 mg in patients with IMIO and suggests an effect in controlling clinical symptoms in patients with and without NGT at baseline. The safety profile of LAN 120 mg was similar to that reported in other indications. ClinicalTrials.gov identifier: NCT02275338.


Subject(s)
Intestinal Obstruction , Peptides, Cyclic , Somatostatin , Female , Humans , Intestinal Obstruction/drug therapy , Intestinal Obstruction/etiology , Peptides, Cyclic/adverse effects , Prospective Studies , Somatostatin/adverse effects
15.
Eur J Cancer ; 157: 403-414, 2021 11.
Article in English | MEDLINE | ID: mdl-34597974

ABSTRACT

INTRODUCTION: This prospective, single-arm, phase 2 study assessed the efficacy and safety of lanreotide autogel (LAN) administered at a reduced dosing interval in patients with progressive neuroendocrine tumours (NETs) after LAN standard regimen. METHODS: Patients had metastatic or locally advanced, grade 1 or 2 midgut NETs or pancreatic NETs (panNETs) and centrally assessed disease progression on LAN 120 mg every 28 days. They were treated with LAN 120 mg every 14 days for up to 96 weeks (midgut cohort) or 48 weeks (panNET cohort). The primary end-point was centrally assessed progression-free survival (PFS). PFS by Ki-67 categories was analysed post hoc. Secondary end-points included quality of life (QoL) and safety. RESULTS: Ninety-nine patients were enrolled (midgut, N = 51; panNET, N = 48). Median (95% CI) PFS was 8.3 (5.6-11.1) and 5.6 (5.5-8.3) months, respectively. In patients with Ki-67 ≤ 10%, median (95% CI) PFS was 8.6 (5.6-13.8) and 8.0 (5.6-8.3) months in the midgut and panNET cohorts, respectively. Patients' QoL did not deteriorate during the study. There were no treatment-related serious adverse events and only two withdrawals for treatment-related adverse events (both in the panNET cohort). CONCLUSIONS: In patients with progressive NETs following standard-regimen LAN, reducing the dosing interval to every 14 days provided encouraging PFS, particularly in patients with a Ki-67 ≤ 10% (post hoc); no safety concerns and no deterioration in QoL were observed. Increasing LAN dosing frequency could therefore be considered before escalation to less well-tolerated therapies.


Subject(s)
Antineoplastic Agents/administration & dosage , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Peptides, Cyclic/administration & dosage , Somatostatin/analogs & derivatives , Aged , Antineoplastic Agents/adverse effects , Disease Progression , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Gels , Humans , Male , Middle Aged , Neuroendocrine Tumors/mortality , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Peptides, Cyclic/adverse effects , Progression-Free Survival , Prospective Studies , Quality of Life , Somatostatin/administration & dosage , Somatostatin/adverse effects
16.
Front Endocrinol (Lausanne) ; 12: 745843, 2021.
Article in English | MEDLINE | ID: mdl-34690933

ABSTRACT

Objective: Efficacy of pharmacological treatments for acromegaly has been assessed in many clinical or real-world studies but no study was interested in economics evaluation of these treatments in France. Therefore, the objective of this study was to estimate the cost-utility of second-line pharmacological treatments in acromegaly patients. Methods: A Markov model was developed to follow a cohort of 1,000 patients for a lifetime horizon. First-generation somatostatin analogues (FGSA), pegvisomant, pasireotide and pegvisomant combined with FGSA (off label) were compared. Efficacy was defined as the normalization of insulin-like growth factor-1 (IGF-1) concentration and was obtained from pivotal trials and adjusted by a network meta-analysis. Costs data were obtained from French databases and literature. Utilities from the literature were used to estimate quality-adjusted life year (QALY). Results: The incremental cost-utility ratios (ICUR) of treatments compared to FGSA were estimated to be 562,463 € per QALY gained for pasireotide, 171,332 € per QALY gained for pegvisomant, and 186,242 € per QALY gained for pegvisomant + FGSA. Pasireotide seems to be the least cost-efficient treatment. Sensitivity analyses showed the robustness of the results. Conclusion: FGSA, pegvisomant and pegvisomant + FGSA were on the cost-effective frontier, therefore, depending on the willingness-to-pay for an additional QALY, they are the most cost-effective treatments. This medico-economic analysis highlighted the consistency of the efficiency results with the efficacy results assessed in the pivotal trials. However, most recent treatment guidelines recommend an individualized treatment strategy based on the patient and disease profile.


Subject(s)
Acromegaly/drug therapy , Drug Costs , Acromegaly/economics , Acromegaly/epidemiology , Aged , Aged, 80 and over , Cohort Studies , Cost-Benefit Analysis , Drug Costs/statistics & numerical data , Drug Therapy, Combination/adverse effects , Drug Therapy, Combination/economics , Female , France/epidemiology , Human Growth Hormone/administration & dosage , Human Growth Hormone/adverse effects , Human Growth Hormone/analogs & derivatives , Human Growth Hormone/economics , Humans , Male , Markov Chains , Middle Aged , Network Meta-Analysis , Octreotide/administration & dosage , Octreotide/adverse effects , Octreotide/economics , Quality-Adjusted Life Years , Somatostatin/administration & dosage , Somatostatin/adverse effects , Somatostatin/analogs & derivatives , Somatostatin/economics
17.
Expert Rev Clin Pharmacol ; 14(12): 1551-1560, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34664531

ABSTRACT

BACKGROUND: Patients with acromegaly require lifelong medication; a longer dosing interval would reduce treatment burden. This study investigated the pharmacokinetics, pharmacodynamics and safety profile of a new prolonged-release formulation (PRF) of lanreotide every 12 weeks. RESEARCH DESIGN AND METHODS: In this multicenter, open-label, dose-ascending study, cohorts of nine patients with acromegaly received single doses of lanreotide PRF according to a 3 + 3 + 3 scheme in order to determine the maximum tolerated dose (MTD). Following a 12-week treatment period, patients were followed up for a further 12 weeks. Serum lanreotide, insulin-like growth factor-1 and growth hormone concentrations were analyzed. Adverse events were monitored throughout the study. RESULTS: The MTD was not reached. Peak lanreotide serum concentration values were similar in all cohorts, whereas area under the curve values from time zero to 85 days increased but were not dose-proportional. The apparent elimination half-life of lanreotide PRF was approximately 54-63 days, in line with the expected prolonged-release characteristics. Growth hormone and insulin-like growth factor-1 levels were generally stable. CONCLUSIONS: The safety and tolerability profile was in-line with the known safety profile of lanreotide autogel. Lanreotide PRF was well tolerated and the pharmacokinetic profile suggests that a dosing interval of 12 weeks could be achievable. CLINICAL TRIAL REGISTRATION: www.clinicaltrials.gov identifier is NCT02396953; EudraCT 2014-002389-62.


Subject(s)
Acromegaly , Human Growth Hormone , Acromegaly/drug therapy , Human Growth Hormone/adverse effects , Humans , Peptides, Cyclic/adverse effects , Somatostatin/adverse effects , Somatostatin/analogs & derivatives
18.
Pituitary ; 24(6): 887-903, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34275099

ABSTRACT

PURPOSE: Pasireotide is an effective treatment for acromegaly and Cushing's disease, although treatment-emergent hyperglycemia can occur. The objective of this study was to assess incretin-based therapy versus insulin for managing pasireotide-associated hyperglycemia uncontrolled by metformin/other permitted oral antidiabetic drugs. METHODS: Multicenter, randomized, open-label, Phase IV study comprising a core phase (≤ 16-week pre-randomization period followed by 16-week randomized treatment period) and optional extension (ClinicalTrials.gov ID: NCT02060383). Adults with acromegaly (n = 190) or Cushing's disease (n = 59) received long-acting (starting 40 mg IM/28 days) or subcutaneous pasireotide (starting 600 µg bid), respectively. Patients with increased fasting plasma glucose (≥ 126 mg/dL on three consecutive days) during the 16-week pre-randomization period despite metformin/other oral antidiabetic drugs were randomized 1:1 to open-label incretin-based therapy (sitagliptin followed by liraglutide) or insulin for another 16 weeks. The primary objective was to evaluate the difference in mean change in HbA1c from randomization to end of core phase between incretin-based therapy and insulin treatment arms. RESULTS: Eighty-one (32.5%) patients were randomized to incretin-based therapy (n = 38 received sitagliptin, n = 28 subsequently switched to liraglutide; n = 12 received insulin as rescue therapy) or insulin (n = 43). Adjusted mean change in HbA1c between treatment arms was - 0.28% (95% CI - 0.63, 0.08) in favor of incretin-based therapy. The most common AE other than hyperglycemia was diarrhea (incretin-based therapy, 28.9%; insulin, 30.2%). Forty-six (18.5%) patients were managed on metformin (n = 43)/other OAD (n = 3), 103 (41.4%) patients did not require any oral antidiabetic drugs and 19 patients (7.6%) were receiving insulin at baseline and were not randomized. CONCLUSION: Many patients receiving pasireotide do not develop hyperglycemia requiring oral antidiabetic drugs. Metformin is an effective initial treatment, followed by incretin-based therapy if needed. ClinicalTrials.gov ID: NCT02060383.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Pituitary ACTH Hypersecretion , Adult , Blood Glucose , Humans , Hyperglycemia/chemically induced , Hypoglycemic Agents/adverse effects , Somatostatin/adverse effects , Somatostatin/analogs & derivatives
19.
Exp Clin Endocrinol Diabetes ; 129(7): 500-509, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34293802

ABSTRACT

BACKGROUND: The long-acting somatostatin analog lanreotide autogel is effective in the treatment of patients with neuroendocrine tumors. OBJECTIVE: To evaluate the long-term treatment response in patients with neuroendocrine tumors receiving lanreotide autogel in routine clinical practice. METHODS: Non-interventional, 24-month study in patients with neuroendocrine tumors treated with lanreotide autogel (NCT01840449). RESULTS: Patients (n=80) from 26 centers in Germany and Austria were enrolled. Neuroendocrine tumors were mainly grade 1/2, metastasized, intestinal, and associated with carcinoid syndrome; 88.9% had received previous neuroendocrine tumor treatment. Of those, 84.4% had previous surgery, 18.7% had received octreotide. The primary endpoint, defined by a <50% chromogranin A increase at month 12 compared with the lowest value between baseline and month 3 was achieved by 89.5% patients. Stable disease according to Response Evaluation Criteria in Solid Tumors 1.1 was observed in 76.9 and 75.0% patients at months 12 and 24 of lanreotide treatment, respectively. Mean change of chromogranin A levels from baseline to month 24 was -0.12 × upper limit of normal (95% CI, -0.22; -0.45). In a post hoc analysis, 38.5% of the subgroup of patients with carcinoid syndrome had daily diarrhea at baseline vs. 21.4% at month 24. At baseline, 27.8% of patients received lanreotide 120 mg every 4 weeks vs. 56.7% at month 24. Quality of life data were heterogeneous. No new safety issues arose and/or required further investigation. CONCLUSIONS: Our study reflects routine lanreotide autogel use in patients with advanced/metastatic neuroendocrine tumors. This analysis shows effectiveness with stabilization of disease-related symptoms and good tolerability of lanreotide autogel in clinical practice.


Subject(s)
Antineoplastic Agents/pharmacology , Malignant Carcinoid Syndrome/drug therapy , Neuroendocrine Tumors/drug therapy , Outcome Assessment, Health Care , Peptides, Cyclic/pharmacology , Somatostatin/analogs & derivatives , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Austria , Female , Germany , Humans , Male , Middle Aged , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/adverse effects , Quality of Life , Somatostatin/administration & dosage , Somatostatin/adverse effects , Somatostatin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...