Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.045
Filter
1.
BMC Pharmacol Toxicol ; 25(1): 44, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090674

ABSTRACT

BACKGROUND: Colorectal cancer (CRC), now the second most prevalent malignant tumor worldwide, is more prevalent in young adults. In recent decades, there has been progress in creating anti-colorectal cancer medications, including cytotoxic compounds. OBJECTIVES: Novel anticancer drugs are needed to surmount existing obstacles. A recent study investigated the effectiveness of novel formulations in preventing colorectal cancer. METHODS: During this study, we assessed a new kind of niosome called cyclo-Gly-L-DOPA (CG-Nio-CGLD) made from chitosan glutamate. We evaluated the anti-colorectal cancer properties of CG-Nio-CGLD utilizing CCK-8, invasion assay, MTT assay, flow cytometry, and cell cycle analysis. The transcription of genes associated with apoptosis was analyzed using quantitative real-time PCR. At the same time, the cytotoxicity of nanomaterials on both cancer and normal cell lines was assessed using MTT assays. Novel anticancer drugs are needed to surmount existing obstacles. A recent study investigated the effectiveness of newly developed formulations in preventing colorectal cancer. RESULTS: The Nio-CGLD and CG-Nio-CGLD were spherical mean diameters of 169.12 ± 1.87 and 179.26 ± 2.17 nm, respectively. Entrapment efficiency (EE%) measurements of the Nio-CGLD and CG-Nio-CGLD were 63.12 ± 0.51 and 76.43 ± 0.34%, respectively. In the CG-Nio-CGLD group, the percentages of early, late, necrotic, and viable CL40 cells were 341.93%, 23.27%, 9.32%, and 25.48%. The transcription of the genes PP53, cas3, and cas8 was noticeably higher in the treatment group compared to the control group (P > 0.001). Additionally, the treatment group had lower BCL2 and survivin gene expression levels than the control group (P < 0.01). Additionally, CG-Nio-CGLD formulations demonstrated a biocompatible nanoscale delivery mechanism and displayed little cytotoxicity toward the CCD 841 CoN reference cell line. CONCLUSION: These findings indicate that chitosan-based noisome encapsulation may enhance the effectiveness of CG-Nio-CGLD formulations in fighting cancer.


Subject(s)
Antineoplastic Agents , Chitosan , Colorectal Neoplasms , Liposomes , Humans , Chitosan/chemistry , Chitosan/administration & dosage , Colorectal Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Glutamic Acid , Peptides, Cyclic/chemistry , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacology , Apoptosis/drug effects , Survivin , Cell Survival/drug effects
2.
Int J Pharm ; 660: 124322, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38866082

ABSTRACT

Podocytes, cells of the glomerular filtration barrier, play a crucial role in kidney diseases and are gaining attention as potential targets for new therapies. Brain-Derived Neurotrophic Factor (BDNF) has shown promising results in repairing podocyte damage, but its efficacy via parenteral administration is limited by a short half-life. Low temperature sensitive liposomes (LTSL) are a promising tool for targeted BDNF delivery, preserving its activity after encapsulation. This study aimed to improve LTSL design for efficient BDNF encapsulation and targeted release to podocytes, while maintaining stability and biological activity, and exploiting the conjugation of targeting peptides. While cyclic RGD (cRGD) was used for targeting endothelial cells in vitro, a homing peptide (HITSLLS) was conjugated for more specific uptake by glomerular endothelial cells in vivo. BDNF-loaded LTSL successfully repaired cytoskeleton damage in podocytes and reduced albumin permeability in a glomerular co-culture model. cRGD conjugation enhanced endothelial cell targeting and uptake, highlighting an improved therapeutic effect when BDNF release was induced by thermoresponsive liposomal degradation. In vivo, targeted LTSL showed evidence of accumulation in the kidneys, and their BDNF delivery decreased proteinuria and ameliorated kidney histology. These findings highlight the potential of BDNF-LTSL formulations in restoring podocyte function and treating glomerular diseases.


Subject(s)
Brain-Derived Neurotrophic Factor , Drug Delivery Systems , Liposomes , Podocytes , Podocytes/drug effects , Podocytes/metabolism , Animals , Brain-Derived Neurotrophic Factor/administration & dosage , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Mice , Male , Cold Temperature , Coculture Techniques , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/chemistry , Kidney/drug effects , Kidney/metabolism , Mice, Inbred C57BL , Drug Liberation
3.
Anim Sci J ; 95(1): e13971, 2024.
Article in English | MEDLINE | ID: mdl-38899765

ABSTRACT

This study investigated the effects of cyclic antimicrobial lipopeptides (CLPs) from Bacillus subtilis on the growth performance, gut morphology, and cecal gene expression and microbiota in broilers; 120 1-day-old unsexed Arbor Acres chicks were randomly divided into four groups, with six replicates in each group and five broilers per cage. These groups were fed a basal diet (C), basal diet plus 10-mg enramycin/kg (E), and basal diet plus 51-mg CLPs/kg (L) or 102-mg CLPs/kg (H). The results indicated that CLP supplementation linearly increased the body weight compared with the C group at 35 days of age. Between 15 and 35 days and 1 and 35 days of age, CLP supplementation linearly increased the average daily gain compared with the C group. The duodenal villus height was significantly increased in the H group compared with the C and E groups. In the cecum, CLP supplementation linearly increased SOD and ZO-1 mRNA expression compared with the C group. ß diversity of microbiota indicated distinct clusters between the groups. CLP supplementation linearly increased the abundance of the genus Lactobacillus in the cecal digesta compared with the C group. These results demonstrate that B. subtilis-produced CLPs dose-dependently increase broilers' growth performance, improve their gut morphology, and modulate their gut microbiota.


Subject(s)
Bacillus subtilis , Cecum , Chickens , Diet , Dietary Supplements , Gastrointestinal Microbiome , Gene Expression , Lipopeptides , Animals , Chickens/growth & development , Chickens/microbiology , Cecum/microbiology , Gastrointestinal Microbiome/drug effects , Lipopeptides/pharmacology , Gene Expression/drug effects , Diet/veterinary , RNA, Messenger/metabolism , Animal Feed , Peptides, Cyclic/pharmacology , Peptides, Cyclic/administration & dosage , Lactobacillus , Intestines/anatomy & histology , Intestines/microbiology , Intestines/drug effects
4.
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
5.
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
6.
Biomater Sci ; 12(9): 2321-2330, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38488841

ABSTRACT

Nanotherapies, valued for their high efficacy and low toxicity, frequently serve as antitumor treatments, but do not readily penetrate deep into tumor tissues and cells. Here we developed an improved tumor-penetrating peptide (TPP)-based drug delivery system. Briefly, the established TPP iNGR was modified to generate a linear NGR peptide capable of transporting nanotherapeutic drugs into tumors through a CendR pathway-dependent, neuropilin-1 receptor-mediated process. Although TPPs have been reported to reach intended tumor targets, they often fail to penetrate cell membranes to deliver tumoricidal drugs to intracellular targets. We addressed this issue by harnessing cell penetrating peptide technology to develop a liposome-based multibarrier-penetrating delivery system (mbPDS) with improved synergistic drug penetration into deep tumor tissues and cells. The system incorporated doxorubicin-loaded liposomes coated with nona-arginine (R9) CPP and cyclic iNGR (CRNGRGPDC) molecules, yielding Lip-mbPDS. Lip-mbPDS tumor-targeting, tumor cell/tissue-penetrating and antitumor capabilities were assessed using CD13-positive human fibrosarcoma-derived cell (HT1080)-based in vitro and in vivo tumor models. Lip-mbPDS evaluation included three-dimensional layer-by-layer confocal laser scanning microscopy, cell internalization/toxicity assays, three-dimensional tumor spheroid-based penetration assays and antitumor efficacy assays conducted in an animal model. Lip-mbPDS provided enhanced synergistic drug penetration of multiple biointerfaces for potentially deep tumor therapeutic outcomes.


Subject(s)
Cell-Penetrating Peptides , Doxorubicin , Drug Delivery Systems , Liposomes , Humans , Animals , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Cell-Penetrating Peptides/chemistry , Cell Line, Tumor , Liposomes/chemistry , Mice , Drug Carriers/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Mice, Nude , Peptides, Cyclic/chemistry , Peptides, Cyclic/administration & dosage
7.
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
8.
Nat Chem Biol ; 20(5): 624-633, 2024 May.
Article in English | MEDLINE | ID: mdl-38155304

ABSTRACT

Cyclic peptides can bind challenging disease targets with high affinity and specificity, offering enormous opportunities for addressing unmet medical needs. However, as with biological drugs, most cyclic peptides cannot be applied orally because they are rapidly digested and/or display low absorption in the gastrointestinal tract, hampering their development as therapeutics. In this study, we developed a combinatorial synthesis and screening approach based on sequential cyclization and one-pot peptide acylation and screening, with the possibility of simultaneously interrogating activity and permeability. In a proof of concept, we synthesized a library of 8,448 cyclic peptides and screened them against the disease target thrombin. Our workflow allowed multiple iterative cycles of library synthesis and yielded cyclic peptides with nanomolar affinities, high stabilities and an oral bioavailability (%F) as high as 18% in rats. This method for generating orally available peptides is general and provides a promising push toward unlocking the full potential of peptides as therapeutics.


Subject(s)
Biological Availability , Peptides, Cyclic , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacology , Administration, Oral , Animals , Rats , Humans , Cyclization , Peptide Library , Thrombin/metabolism , Thrombin/chemistry , Male , Combinatorial Chemistry Techniques , Acylation
9.
Neuropathology ; 44(3): 247-251, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38099404

ABSTRACT

A dopamine agonist administered for prolactinoma treatment and pituitary stimulation tests are reported as risk factors for pituitary apoplexy. We report a case of an 82-year-old patient who suffered from pituitary apoplexy in an endocrinologically silent adenoma during lanreotide administration. The patient was diagnosed with a pancreatic neuroendocrine tumor with lymph node metastasis and treated with lanreotide for two years. An endoscopic endonasal transsphenoidal approach was used for tumor and hematoma removal. The specimen showed growth hormone and prolactin positivity and was diagnosed as pit1-lineage plurihormonal adenoma. The tumor also showed positivity for somatostatin receptor 2. Thus, lanreotide treatment is a risk factor for pituitary apoplexy even in silent adenoma.


Subject(s)
Adenoma , Neuroendocrine Tumors , Pancreatic Neoplasms , Peptides, Cyclic , Pituitary Apoplexy , Somatostatin , Humans , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Somatostatin/analogs & derivatives , Neuroendocrine Tumors/pathology , Aged, 80 and over , Pituitary Apoplexy/pathology , Adenoma/pathology , Adenoma/drug therapy , Peptides, Cyclic/administration & dosage , Male , Pituitary Neoplasms/pathology , Pituitary Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use
11.
J Psychopharmacol ; 36(2): 214-222, 2022 02.
Article in English | MEDLINE | ID: mdl-35102783

ABSTRACT

BACKGROUND: Cycloprolylglycine (CPG) is an endogenous dipeptide with a wide range of psychotropic activity and putative therapeutic potential for depression. A small but growing body of data suggests that antidepressant-like effect of CPG is associated with neuroplastic changes in the brain or 5-HT system modulation. However, the mechanisms of the dipeptide action remain elusive. AIMS: Here, we characterize the effects of chronic CPG administration on behavior and genes expression of antidepressants sensitive catalepsy (ASC) mice strain, characterized by depressive-like behavior. METHODS: ASC mice were injected with saline, fluoxetine (10 mg/kg/day), or CPG (1 and 2 mg/kg/day) during 2 weeks. Behavior was studied using the open field test, novel object test, elevated plus maze test, forced swim test, and tail suspension test (TST). The expressions of genes coding BDNF, CREB, 5-HT1A and 5-HT2A receptors, TPH2, and SERT in the brain were measured with quantitative real-time reverse transcription polymerase chain reaction (RT-PCR). RESULTS: Chronic intraperitoneal administration of 1 and 2 mg/kg of CPG revealed the significant antidepressant-like effect by decreasing immobility time in the TST. At the same time, CPG did not negatively affect locomotor activity, cognition, or anxiety. In the real-time quantitative polymerase chain reaction (PCR) assay, chronic CPG treatment (2 mg/kg for 14 days) increased Bdnf mRNA level in the frontal cortex. CONCLUSIONS: Our findings extend the evidence for the effectiveness of CPG to reduce depressive-like behaviors. The antidepressant-like effect of CPG is mediated, as least in part, by BDNF-dependent mechanism. The exact mechanism remains to be elucidated, and further studies are warranted.


Subject(s)
Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Depression/drug therapy , Peptides, Cyclic/pharmacology , Animals , Antidepressive Agents/administration & dosage , Behavior, Animal/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Fluoxetine/pharmacology , Frontal Lobe/drug effects , Gene Expression Regulation/drug effects , Male , Mice , Peptides, Cyclic/administration & dosage , RNA, Messenger/metabolism
12.
Article in English | MEDLINE | ID: mdl-34891048

ABSTRACT

Lanreotide is similar to a naturally occurring hormone, somatostatin; thus, it may be used to treat acromegaly or metastatic gastroenteropancreatic neuroendocrine tumours. Here, a bioanalytical method coupling ultra-performance liquid chromatography with tandem mass spectrometry to quantify lanreotide and an internal standard (IS) was developed and validated in dog plasma. The plasma samples were extracted using typical protein precipitation processes. The analyte and internal standard were separated on Phenomenex Kinetex® C18 with 0.1% formic acid and acetonitrile in the mobile phase at a flow rate of 0.4 mL/min. The fragmentation of precursor ions to product ions was optimized at m/z 548.8 â†’ 170.0 for lanreotide [M + 2H]2+ and 472.2 â†’ 436.2 for IS [M + H]+. The peak retention times of lanreotide and IS were 1.09 min and 1.22 min, respectively. The calibration curve samples in dog plasma ranged from 0.3 to 1000 ng/mL and showed good linearity, with a correlation coefficient of r2=0.9996. The lower limit of quantitation was 0.3 ng/mL. The intra- and inter-day precision (relative standard deviation) values for each quality control level were < 9.7 % and < 9.3 %, respectively; intra- and inter-day accuracy were < 109.3% and < 110.4%, respectively. Lanreotide in dog plasma was stable in various conditions. The maximum plasma concentration of lanreotide in male beagle dogs after subcutaneous injection of Somatuline® (lanreotide) Autogel 120 mg was 88.1 ng/mL. The half-life (T1/2) of lanreotide in beagle dogs was long, approximately 198.6 h; the area under the plasma-concentration curve from 0 to 840 h (day 35) was 6,995 ng⋅h/mL. This novel quantification method using UPLC-MS/MS was successfully applied to the pharmacokinetic analysis of lanreotide in dog plasma. The results will assist future studies of drug formulation and repurposing.


Subject(s)
Chromatography, Liquid/methods , Peptides, Cyclic/blood , Somatostatin/analogs & derivatives , Tandem Mass Spectrometry/methods , Animals , Dogs , Injections, Subcutaneous , Limit of Detection , Linear Models , Male , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacokinetics , Reproducibility of Results , Somatostatin/administration & dosage , Somatostatin/blood , Somatostatin/pharmacokinetics
13.
Nutr Neurosci ; 25(12): 2517-2527, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34565308

ABSTRACT

Cyclic glycine-proline (cGP) is a natural nutrient of breast milk and plays a role in regulating the function of insulin-like growth factor-1 (IGF-1). IGF-1 function is essential for post-natal brain development and adult cognitive function. We evaluated the effects of cGP on spatial memory and histological changes in the hippocampus of the adult rats following infancy administration. Infant rats were treated with either cGP or saline between post-natal days 8 and 22 via oral administration to lactating dams. The spatial memory was evaluated between post-natal days 70 and 75 using Morris water maze tests. The changes of capillaries, astrocytes, synaptophysin and glutamate receptor-1 were examined in the CA1 stratum radiatum of the hippocampus. Compared to saline-treated group, cGP-treated group showed higher path efficiency of entry and lower average heading errors to the platform zone. cGP-treated group also showed longer, larger and more astrocytic processes, more capillaries and higher glutamate receptor-1 expression. The rats made less average heading error to the platform zone have more capillaries, larger and longer astrocytic branches. Thus cGP treatment/supplementation during infancy moderately improved adulthood spatial memory. This long-lasting effect of cGP on memory could be mediated via promoting astrocytic plasticity, vascularization and glutamate trafficking. Therefore, cGP may have a role in regulating IGF-1 function during brain development.


Subject(s)
Brain , Insulin-Like Growth Factor I , Maternal Nutritional Physiological Phenomena , Peptides, Cyclic , Spatial Memory , Animals , Female , Rats , Astrocytes/metabolism , Hippocampus/metabolism , Insulin-Like Growth Factor I/metabolism , Lactation , Maze Learning , Receptors, Glutamate/metabolism , Peptides, Cyclic/administration & dosage , Brain/growth & development
14.
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
15.
Neuroendocrinology ; 112(1): 88-100, 2022.
Article in English | MEDLINE | ID: mdl-33508849

ABSTRACT

INTRODUCTION: Somatostatin analogs (SSA) prolong progression-free survival (PFS) in patients with well-differentiated gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs). However, the eligibility criteria in randomized clinical trials (RCTs) have been restricted, which contrasts with the vast heterogeneity found in NENs. METHODS: We identified patients with well-differentiated (Ki-67% ≤20%), metastatic GEP-NENs treated in first line with SSA monotherapy from the Spanish R-GETNE registry. The therapeutic effect was evaluated using a Bayesian Cox model. The objective was to compare survival-based outcomes from real-world clinical practice versus RCTs. RESULTS: The dataset contained 535 patients with a median age of 62 years (range: 26-89). The median Ki-67% was 4 (range: 0-20). The most common primary tumor sites were as follows: midgut, 46%; pancreas, 34%; unknown primary, 10%; and colorectal, 10%. Half of the patients received octreotide LAR (n = 266) and half, lanreotide autogel (n = 269). The median PFS was 28.0 months (95% CI: 22.1-32.0) for octreotide versus 30.1 months (95% CI: 23.1-38.0) for lanreotide. The overall hazard ratio for lanreotide versus octreotide was 0.90 (95% credible interval: 0.71-1.12). The probability of effect sizes >30% with lanreotide versus octreotide was 2 and 6% for midgut and foregut NENs, respectively. CONCLUSION: Our study evaluated the external validity of RCTs examining SSAs in the real world, as well as the main effect-modifying factors (progression status, symptoms, tumor site, specific metastases, and analytical data). Our results indicate that both octreotide LAR and lanreotide autogel had a similar effect on PFS. Consequently, both represent valid alternatives in patients with well-differentiated, metastatic GEP-NENs.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Intestinal Neoplasms/drug therapy , Neuroendocrine Tumors/drug therapy , Octreotide/pharmacology , Pancreatic Neoplasms/drug therapy , Peptides, Cyclic/pharmacology , Progression-Free Survival , Randomized Controlled Trials as Topic/standards , Registries , Somatostatin/analogs & derivatives , Somatostatin/analysis , Stomach Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Hormonal/administration & dosage , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Octreotide/administration & dosage , Peptides, Cyclic/administration & dosage , Prognosis , Reproducibility of Results , Somatostatin/administration & dosage , Somatostatin/pharmacology , Spain
16.
Medicina (Kaunas) ; 57(12)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34946232

ABSTRACT

Background and Objectives: Long-acting somatostatin analogues (SSA) (octreotide LAR and lanreotide Autogel) are recommended as first line treatment of locally advanced or metastatic well-differentiated neuroendocrine tumors (NETs) with a good expression of somatostatin receptor (SSTR). Both of these SSAs are usually administered via injections repeated every 4 weeks. The purpose of the study was to compare the route of SSA administration (injection performed by professional medical staff and self-administration of the drug) with progression-free survival. Materials and methods: 88 patients in 2019 and 96 patients in 2020 with locally advanced or metastatic well-differentiated NETs were included in the study. All patients had a good expression of SSTR type 2 and had been treated for at least 3 months with a stable dose of long-acting somatostatin analogue every 4 weeks. All of them had received training on drug self-injections from professional NET nurses at the beginning of the COVID-19 epidemic. Results: The rate of NET progression in the study group in 2020 was higher than in 2019 29.1% vs. 18.1% (28 vs. 16 cases), p = 0.081. Conclusions: The method of administration of long-acting SSA injection performed by professional medical staff vs. self-injection of the drug may significantly affect the risk of NET progression. The unequivocal confirmation of such a relationship requires further observation.


Subject(s)
Neuroendocrine Tumors , Octreotide/administration & dosage , Peptides, Cyclic/administration & dosage , Self Administration , Somatostatin/analogs & derivatives , Humans , Neuroendocrine Tumors/drug therapy , Somatostatin/administration & dosage , Treatment Outcome
17.
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
18.
Dalton Trans ; 50(44): 16156-16165, 2021 Nov 16.
Article in English | MEDLINE | ID: mdl-34704995

ABSTRACT

Radiotracers labelled with technetium-99m (99mTc) enable accessible diagnostic imaging of disease, provided that radiotracer preparation is simple. Whilst 99mTc radiopharmaceuticals for imaging perfusion are routinely prepared from kits, and regularly used in healthcare, there are no 99mTc-labelled receptor-targeted radiopharmaceuticals in widespread clinical use. This is in part due to the multistep radiosyntheses required for the latter. We demonstrate that the diphosphine, 2,3-bis(diphenylphosphino)maleic anhydride (BMA), is an excellent platform for preparation of kit-based, receptor-targeted 99mTc-labelled radiotracers: its conjugates are simple to prepare and can be easily labelled with 99mTc using one-step, kit-based protocols. Here, reaction of BMA with the αvß3-integrin receptor targeted cyclic peptide, Arg-Gly-Asp-DPhe-Lys (RGD), provided the first diphosphine-peptide conjugate, DP-RGD. DP-RGD was incorporated into a "kit", and addition of a saline solution containing 99mTcO4- to this kit, followed by heating, furnished the radiotracer [99mTcO2(DP-RGD)2]+ in consistently high radiochemical yields (>90%). The analogous [ReO2(DP-RGD)2]+ compound was prepared and characterised, revealing that both [99mTcO2(DP-RGD)2]+ and [ReO2(DP-RGD)2]+ consist of a mixture of cis and trans geometric isomers. Finally, [99mTcO2(DP-RGD)2]+ exhibited high metabolic stability, and selectively targeted αvß3-integrin receptors, enabling in vivo SPECT imaging of αvß3-integrin receptor expression in mice.


Subject(s)
Chelating Agents , Peptides, Cyclic , Phosphines , Radiopharmaceuticals , Technetium , Animals , Arthritis, Rheumatoid/diagnostic imaging , Arthritis, Rheumatoid/metabolism , Chelating Agents/administration & dosage , Chelating Agents/chemistry , Chelating Agents/pharmacokinetics , Female , Humans , Integrin alphaVbeta3/chemistry , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Phosphines/administration & dosage , Phosphines/chemistry , Phosphines/pharmacokinetics , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Technetium/administration & dosage , Technetium/chemistry , Technetium/pharmacokinetics , Tomography, Emission-Computed, Single-Photon
19.
Anticancer Res ; 41(9): 4277-4285, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34475047

ABSTRACT

BACKGROUND/AIM: To evaluate the antitumor effects of Plitidepsin against clear cell carcinoma (CCC) of the ovary. MATERIALS AND METHODS: The expression of eEF1A2 in ovarian cancer was assessed by immunohistochemistry. Using ovarian CCC cell lines, the antitumor effect of Plitidepsin was assessed both in vitro and in vivo. By over-expressing or knocking down the eEF1A2 expression, we investigated the role of eEF1A2 in the sensitivity of CCC cells to Plitidepsin. RESULTS: Immunoreactivity to eEF1A2 was observed in 76.2% of CCC, which was significantly higher than other histological subtypes of ovarian cancer. Plitidepsin exhibited significant antitumor activity toward chemonaive and chemoresistant CCC cells both in vitro and in vivo. Ectopic expression of eEF1A2 in CCC cells resulted in increased sensitivity to Plitidepsin. In contrast, eEF1A2 knockdown decreased sensitivity of CCC cells to plitidepsin. CONCLUSION: Plitidepsin, a novel anti-cancer agent that targets eEF1A2, may be a promising agent for treating ovarian CCC.


Subject(s)
Adenocarcinoma, Clear Cell/drug therapy , Depsipeptides/administration & dosage , Ovarian Neoplasms/drug therapy , Peptide Elongation Factor 1/metabolism , Peptides, Cyclic/administration & dosage , Up-Regulation/drug effects , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin , Depsipeptides/pharmacology , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Mice , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Peptide Elongation Factor 1/genetics , Peptides, Cyclic/pharmacology , Xenograft Model Antitumor Assays
20.
Mol Pharm ; 18(9): 3638-3648, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34424706

ABSTRACT

Targeting metastatic esophageal squamous cell carcinoma (ESCC) has been a challenge in clinical practice. Emerging evidence demonstrates that C-X-C chemokine receptor 4 (CXCR4) highly expresses in ESCC and plays a pivotal role in the process of tumor metastasis. We developed a copper-64 (t1/2 = 12.7 h, 19% beta+) labeling route of NOTA-CP01 derived from LY2510924, a cyclopeptide-based CXCR4 potent antagonist, in an attempt to noninvasively visualize CXCR4 expression in metastatic ESCC. Precursor NOTA-CP01 was designed by modifying the C-terminus of LY2510925 with bis-t-butyl NOTA via a butane-1,4-diamine linker. The radiolabeling process was finished within 15 min with high radiochemical yield (>95%), radiochemical purity (>99%), and specific activity (10.5-21 GBq/µmol) (non-decay-corrected). The in vitro solubility and stability tests revealed that [64Cu]NOTA-CP01 had a high water solubility (log P = -3.44 ± 0.12, n = 5) and high stability in saline and fetal bovine serum. [64Cu]NOTA-CP01 exhibited CXCR4-specific binding with a nanomolar affinity (IC50 = 1.61 ± 0.96 nM, Kd = 0.272 ± 0.14 nM) similar to that of the parental LY2510924. The in vitro cell uptake assay indicated that the [64Cu]NOTA-CP01-selective accumulation in EC109 cells was CXCR4-specific. Molecular docking of the CXCR4/NOTA-CP01 complex suggested that the Lys, Arg, and NOTA of this ligand have a strong polar interaction with the key residues of CXCR4, which explains the tight affinity of [64Cu]NOTA-CP01 for CXCR4. To test the target engagement in vivo, prolonged-time positron emission computed tomography (PET) imaging was performed at 0.5, 4, 6, 8, 12, 16, and 24 h postinjection of [64Cu]NOTA-CP01 to the EC109 tumor-bearing mice. The EC109 tumors were most visible with high contrast to the contralateral background at 6 h postinjection. The tracer revealed receptor-specific tumor accumulation, which was illustrated by effective blocking via coinjection with a blocking dose of LY2510924. Quantification analysis of the prolonged-time images showed that there was obvious radioactivity accumulation in the tumor (1.27 ± 0.19%ID/g) with the best tumor-to-blood ratio (4.79 ± 0.06) and tumor-to-muscle ratio (15.44 ± 2.94) at 6 h postinjection of the probe. The immunofluorescence and immunohistochemistry confirmed the positive expression of CXCR4 in the EC109 tumor and ESCC and metastatic lymph nodes of patients, respectively. We concluded that [64Cu]NOTA-CP01 possessed a very high target engagement for CXCR4-positive ESCC and could be a potential candidate in the clinical detection of metastatic ESCC.


Subject(s)
Esophageal Neoplasms/diagnostic imaging , Esophageal Squamous Cell Carcinoma/diagnostic imaging , Peptides, Cyclic/administration & dosage , Radiopharmaceuticals/administration & dosage , Receptors, CXCR4/antagonists & inhibitors , Animals , Cell Line, Tumor , Copper Radioisotopes , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/secondary , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Inhibitory Concentration 50 , Male , Mice , Molecular Docking Simulation , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Positron Emission Tomography Computed Tomography/methods , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , X-Ray Microtomography/methods , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL