Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Mol Biol Rep ; 51(1): 401, 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38457071

RESUMEN

BACKGROUND: Gastric ulcer (GU) is a common gastrointestinal tract illness. Aloe vera has anti-inflammatory, antioxidant, and healing characteristics. This research sought to explore the therapeutic impact of Aloe vera gel on ethanol-provoked GU in rats and to elucidate the underlying mechanisms involved. METHODS: An ethanol-induced GU rat model was constructed using forty male Wistar rats distributed at random into four groups: control, ulcer, pantoprazole, and Aloe vera. Gross evaluation of the stomach, ulcer index (UI), inhibition index, and gastric pH estimation were analyzed. Gastric malondialdehyde (MDA) and reduced glutathione (GSH) were determined using the spectrophotometric method, and serum gastrin level was measured by an enzyme-linked immunosorbent assay. Gastric nucleotide-binding domain, leucine-rich repeat, and pyrin domain PYD containing protein 3 (NLRP3) and gasdermin D (GSDMD) mRNA expression levels were estimated by quantitative real-time PCR. Finally, the histopathological examination of the glandular part of stomach tissue was done. RESULTS: The ulcer group revealed a significant increase in MDA, gastrin, NLRP3, and GSDMD and a decrease in gastric pH and GSH compared to the control group. Gross investigations of the ulcer group revealed a hemorrhagic lesion in the stomach and an increase in UI. Also, histopathological results for this group showed severe epithelial loss, haemorrhage, inflammatory cell infiltration, and blood vessel congestion. However, Aloe vera treatment improved the gross, biochemical, molecular, and histopathological alterations induced by ethanol when compared to the ulcer group. CONCLUSIONS: Aloe vera exerted antiulcer activities through modulation of oxidant/antioxidant status, anti-secretory properties, and mitigation of pyroptosis.


Asunto(s)
Preparaciones de Plantas , Úlcera Gástrica , Ratas , Masculino , Animales , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/tratamiento farmacológico , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Proteína con Dominio Pirina 3 de la Familia NLR , Etanol/efectos adversos , Úlcera/tratamiento farmacológico , Gastrinas/uso terapéutico , Piroptosis , Ratas Wistar , Extractos Vegetales/farmacología , Transducción de Señal
2.
Comput Math Methods Med ; 2022: 1221190, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36267315

RESUMEN

The present study was designed to observe the effect of quadruple therapy combined with probiotics on Helicobacter pylori-related peptic ulcer. The patients in the control group (n = 90) were given regular quadruple therapy including proton pump inhibitor ilaprazole enteric-coated tablet + two antibiotics amoxicillin dispersible tablet and metronidazole tablet + colloidal bismuth pectin capsule for 2 weeks. Patients in the study group (n = 90) were given abovementioned quadruple therapy combined with probiotics live combined Bifidobacterium, Lactobacillus, and Enterococcus Capsules, oral for 2 weeks. Then Hp clearance rate, recurrence rate, levels of gastrointestinal hormone makers, and advance reactions between two groups were compared. At the 2nd week after the treatment, the Helicobacter pylori clearance rate in the study group (87.79%) was significantly higher than the control group (78.89%), and the total recurrence rate in the study group (6.67%) was significantly lower than the control group (13.33%) (P < 0.05). Serum gastrin and motilin expression were lower, and somatostatin expressions was significantly higher than those in the control group (P < 0.05). There was no significant difference in the total incidence of adverse reactions between the two groups (P > 0.05). In summary, quadruple therapy combined with probiotics in the treatment of Helicobacter pylori-related peptic ulcer can improve the Helicobacter pylori clearance rate, reduce the Helicobacter pylori recurrence rate, and is beneficial to improving the level of gastrointestinal hormones, with certain safety.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Úlcera Péptica , Probióticos , Humanos , Infecciones por Helicobacter/tratamiento farmacológico , Bismuto/farmacología , Bismuto/uso terapéutico , Metronidazol/farmacología , Metronidazol/uso terapéutico , Inhibidores de la Bomba de Protones/farmacología , Gastrinas/farmacología , Gastrinas/uso terapéutico , Motilina/farmacología , Motilina/uso terapéutico , Comprimidos Recubiertos/farmacología , Comprimidos Recubiertos/uso terapéutico , Quimioterapia Combinada , Úlcera Péptica/tratamiento farmacológico , Úlcera Péptica/microbiología , Amoxicilina/uso terapéutico , Amoxicilina/farmacología , Antibacterianos/uso terapéutico , Probióticos/uso terapéutico , Pectinas/farmacología , Pectinas/uso terapéutico , Somatostatina/farmacología , Somatostatina/uso terapéutico
3.
Inflammopharmacology ; 30(5): 1659-1668, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35831736

RESUMEN

The purpose of this study was to investigate the anti-inflammatory effect of an aqueous extract of seed of broccoli (AESB) in Helicobacter pylori (HP)-infected patients without atrophic gastritis. This was a double-centre, randomized, double-blind, controlled study. A total of 110 HP-infected subjects were randomized to receive either AESB or placebo for 2 months. Inflammatory cytokine (IL-8, IFN-γ, TNF-α, CRP, IL-17A, IL-1ß, IL-18), pepsinogen I, II (PG I, PG II), and gastrin-17 (G-17) measurements and 13C-urea breath tests were performed at baseline and at 60 days. At 60 days, there was no significant difference in any of the inflammatory cytokines, pepsinogen or gastrin between the two groups. However, IL-8, IFN-γ, PG I, PG I/PG II ratio (PGR), and G-17 were reduced by 9.02 pg/mL, 5.08 pg/mL, 24.56 ng/mL, 1.75 and 0.3 pmol/L, respectively, in the AESB group compared with baseline (all P < 0.05). The HP eradication rates in the AESB group and placebo group were 11.11 and 3.70% at 60 days, respectively (P > 0.05). No treatment-related adverse events were reported. Thus, AESB may reduce the risk of gastric mucosal lesions and decrease the risk of gastric cancer by relieving inflammatory cytokines. The safety profile of AESB was satisfactory. This study is registered with the Chinese Clinical Trials Registry (Registration No. ChiCTR2100054249).


Asunto(s)
Brassica , Gastritis Atrófica , Infecciones por Helicobacter , Helicobacter pylori , Antiinflamatorios/uso terapéutico , Citocinas , Gastrinas/uso terapéutico , Gastritis Atrófica/tratamiento farmacológico , Gastritis Atrófica/patología , Infecciones por Helicobacter/tratamiento farmacológico , Humanos , Interleucina-17 , Interleucina-18 , Interleucina-8/uso terapéutico , Pepsinógeno A , Factor de Necrosis Tumoral alfa , Urea/uso terapéutico
4.
Br J Pharmacol ; 179(17): 4360-4377, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35484823

RESUMEN

BACKGROUND AND PURPOSE: Glucagon-like peptide-1 (GLP-1) and glucagon (GCG) receptor dual agonist have promising therapeutic effects in the treatment of obesity and diabetes. Moreover, GLP-1 and cholecystokinin 2 (CCK2 ) dual agonists have been shown to restore pancreas function and improve glycaemic control in preclinical studies. We describe, for the first time, the beneficial effects of GLP-1/glucagon receptor and GLP-1/CCK2 dual agonists, which can be integrated into one peptide, resulting in significant anti-diabetes and anti-obesity effectiveness. EXPERIMENTAL APPROACH: The in vitro potency of this novel peptide Xenopus (x) GLP-1/GCG/CCK2 tri-agonist (xGLP/GCG/gastrin) against GLP-1, GCG, CCK1 and CCK2 receptors was determined on cells expressing the corresponding receptors by cAMP accumulation or ERK1/2 phosphorylation assays. The in vivo anti-diabetes and anti-obesity effects of this tri-agonist xGLP/GCG/gastrin were studied in both db/db and diet induced obesity (DIO) mice. KEY RESULTS: xGLP/GCG/gastrin was a potent and selective GLP-1, GCG and CCK2 tri-agonist. In DIO mice, the metabolic benefits of xGLP-1/GCG/gastrin, such as reduction of body weight and hepatic lipid contents were significantly better than those of the peptide ZP3022 (GLP-1/CCK-2 dual agonist) and liraglutide. In a short-term study in db/db mice, xGLP/GCG/gastrin treatment had considerable effects, increasing islet numbers, islet areas and insulin content. In a long-term treatment study using db/db mice, xGLP-1/GCG/gastrin showed a significantly and sustained improvement in glucose tolerance and glucose control compared with that of liraglutide, ZP3022, cotadutide (GLP-1/GCG dual agonist) and xGLP/GCG-15. CONCLUSIONS AND IMPLICATIONS: These results demonstrate the therapeutic potential of xGLP-1/GCG/gastrin for the treatment of obesity and diabetes.


Asunto(s)
Diabetes Mellitus , Glucagón , Animales , Colecistoquinina , Diabetes Mellitus/tratamiento farmacológico , Gastrinas/agonistas , Gastrinas/uso terapéutico , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/agonistas , Hipoglucemiantes/farmacología , Liraglutida/farmacología , Liraglutida/uso terapéutico , Ratones , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Péptidos/farmacología , Receptores de Glucagón/agonistas , Receptores de Glucagón/metabolismo , Receptores de Glucagón/uso terapéutico
5.
Theranostics ; 10(24): 10861-10873, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33042258

RESUMEN

Rationale: A high tumor-to-healthy-tissue uptake ratio of radiolabeled ligands is an essential prerequisite for safe and effective peptide receptor radionuclide therapy (PRRT). In the present study, we searched for novel opportunities to increase tumor-specific uptake of the radiolabeled minigastrin analogue [177Lu]Lu-DOTA-(DGlu)6-Ala-Tyr-Gly-Trp-Nle-Asp-Phe-NH2 ([177Lu]Lu-PP-F11N), that targets the cholecystokinin B receptor (CCKBR) in human cancers. Methods: A kinase inhibitor library screen followed by proliferation and internalization assays were employed to identify compounds which can increase uptake of [177Lu]Lu-PP-F11N in CCKBR-transfected human epidermoid carcinoma A431 cells and natural CCKBR-expressing rat pancreatic acinar AR42J cells. Western blot (WB) analysis verified the inhibition of the signaling pathways and the CCKBR level, whereas the cell-based assay analyzed arrestin recruitment. Biodistribution and SPECT imaging of the A431/CCKBR xenograft mouse model as well as histological analysis of the dissected tumors were used for in vivo validation. Results: Our screen identified the inhibitors of mammalian target of rapamycin complex 1 (mTORC1), which increased cell uptake of [177Lu]Lu-PP-F11N. Pharmacological mTORC1 inhibition by RAD001 and metformin increased internalization of [177Lu]Lu-PP-F11N in A431/CCKBR and in AR42J cells. Analysis of protein lysates from RAD001-treated cells revealed increased levels of CCKBR (2.2-fold) and inhibition of S6 phosphorylation. PP-F11N induced recruitment of ß-arrestin1/2 and ERK1/2 phosphorylation. In A431/CCKBR-tumor bearing nude mice, 3 or 5 days of RAD001 pretreatment significantly enhanced tumor-specific uptake of [177Lu]Lu-PP-F11N (ratio [RAD001/Control] of 1.56 or 1.79, respectively), whereas metformin treatment did not show a significant difference. Quantification of SPECT/CT images confirmed higher uptake of [177Lu]Lu-PP-F11N in RAD001-treated tumors with ratios [RAD001/Control] of average and maximum concentration reaching 3.11 and 3.17, respectively. HE staining and IHC of RAD001-treated tumors showed a significant increase in necrosis (1.4% control vs.10.6% of necrotic area) and the reduction of proliferative (80% control vs. 61% of Ki67 positive cells) and mitotically active cells (1.08% control vs. 0.75% of mitotic figures). No significant difference in the tumor vascularization was observed after five-day RAD001 or metformin treatment. Conclusions: Our data demonstrates, that increased CCKBR protein level by RAD001 pretreatment has the potential to improve tumor uptake of [177Lu]Lu-PP-F11N and provides proof-of-concept for the development of molecular strategies aimed at enhancing the level of the targeted receptor, to increase the efficacy of PRRT and nuclear imaging.


Asunto(s)
Quimioradioterapia/métodos , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Neoplasias/terapia , Fragmentos de Péptidos/farmacología , Radiofármacos/farmacología , Animales , Línea Celular Tumoral , Everolimus/farmacología , Everolimus/uso terapéutico , Femenino , Gastrinas/genética , Gastrinas/farmacología , Gastrinas/uso terapéutico , Humanos , Lutecio , Ratones , Neoplasias/diagnóstico por imagen , Neoplasias/patología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/uso terapéutico , Radioisótopos , Radiofármacos/uso terapéutico , Ratas , Receptor de Colecistoquinina B/metabolismo , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Anticancer Agents Med Chem ; 20(4): 402-416, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31889492

RESUMEN

BACKGROUND: Cancer is the leading cause of death worldwide. Early detection can reduce the disadvantageous effects of diseases and the mortality in cancer. Nuclear medicine is a powerful tool that has the ability to diagnose malignancy without harming normal tissues. In recent years, radiolabeled peptides have been investigated as potent agents for cancer detection. Therefore, it is necessary to modify radiopeptides in order to achieve more effective agents. OBJECTIVE: This review describes modifications in the structure of radioconjugates with spacers who have improved the specificity and sensitivity of the peptides that are used in oncologic diagnosis and therapy. METHODS: To improve the biological activity, researchers have conjugated these peptide analogs to different spacers and bifunctional chelators. Many spacers of different kinds, such as hydrocarbon chain, amino acid sequence, and poly (ethyleneglycol) were introduced in order to modify the pharmacokinetic properties of these biomolecules. RESULTS: Different spacers have been applied to develop radiolabeled peptides as potential tracers in nuclear medicine. Spacers with different charge and hydrophilicity affect the characteristics of peptide conjugate. For example, the complex with uncharged and hydrophobic spacers leads to increased liver uptake, while the composition with positively charged spacers results in high kidney retention. Therefore, the pharmacokinetics of radio complexes correlates to the structure and total charge of the conjugates. CONCLUSION: Radio imaging technology has been successfully applied to detect a tumor in the earliest stage. For this purpose, the assessment of useful agents to diagnose the lesion is necessary. Developing peptide radiopharmaceuticals using spacers can improve in vitro and in vivo behavior of radiotracers leading to better noninvasive detection and monitoring of tumor growth.


Asunto(s)
Neoplasias/diagnóstico , Neoplasias/radioterapia , Péptidos/química , Péptidos/uso terapéutico , Radiofármacos/química , Radiofármacos/uso terapéutico , Animales , Bombesina/química , Bombesina/uso terapéutico , Gastrinas/química , Gastrinas/uso terapéutico , Humanos , Hormonas Estimuladoras de los Melanocitos/química , Hormonas Estimuladoras de los Melanocitos/uso terapéutico , Neurotensina/química , Neurotensina/uso terapéutico , Oligopéptidos/química , Oligopéptidos/uso terapéutico , Somatostatina/química , Somatostatina/uso terapéutico
7.
J Coll Physicians Surg Pak ; 30(12): 1269-1272, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33397051

RESUMEN

OBJECTIVE:  To explore the predictive value of serum gastrin (GAS), epidermal growth factor (EGF) levels in gastric ulcer complicated with acute upper gastrointestinal bleeding. STUDY DESIGN: A descriptive study. PLACE AND DURATION OF STUDY: Department of Emergency, Beijing Jiangong Hospital, China, from January 2019 to June 2020. METHODOLOGY: One hundred and twenty-five patients with gastric ulcer and acute upper gastrointestinal bleeding were selected as Group A. One hundred and twenty-five patients with gastric ulcer and no upper gastrointestinal bleeding were selected as Group B. Logistic regression analysis was used to analyse the risk factors of gastric ulcer complicated with acute upper gastrointestinal bleeding. The value of serum GAS, EGF in early diagnosis of gastric ulcer with upper gastrointestinal bleeding was evaluated by receiver operating characteristic (ROC) curve. RESULTS: Univariate analysis showed statistically significant differences between Group A and Group B in taking non-steroidal anti-inflammatory drugs (NSAIDs), helicobacter pylori (Hp) infection, serum GAS and EGF (all p <0.001). Logistic regression analysis showed that raised serum GAS and serum EGF were independent risk factors for gastric ulcer and upper gastrointestinal bleeding (both p <0.001). The ROC area of serum EGF to predict gastric ulcer and acute upper gastrointestinal bleeding was 0.810 (95% CI: 0.753-0.867, p <0.001), greater than ROC area of serum GAS. At serum EGF of ≤109.95 pg/mL, had the 84.8%, sensitivity to predict gastric ulcer and acute upper gastrointestinal bleeding with specificity of 68.8%. CONCLUSION: The predictive value of serum GAS and EGF is high for gastric ulcer complicated with acute upper gastrointestinal bleeding; the predictive value of serum EGF is greater than that of serum GAS. Key Words: Gastric ulcer, Acute upper gastrointestinal bleeding, Serum, Gastrin (GAS), Epidermal growth factor (EGF), Logistic regression, Receiver operating characteristic (ROC) curve.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Úlcera Gástrica , Antiinflamatorios no Esteroideos/efectos adversos , China , Factor de Crecimiento Epidérmico , Gastrinas/uso terapéutico , Hemorragia Gastrointestinal/diagnóstico , Hemorragia Gastrointestinal/etiología , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/diagnóstico , Humanos , Factores de Riesgo , Úlcera Gástrica/complicaciones , Úlcera Gástrica/diagnóstico
8.
Am J Physiol Gastrointest Liver Physiol ; 317(5): G682-G693, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31433212

RESUMEN

Growth of pancreatic cancer is stimulated by gastrin in both a paracrine and an autocrine fashion. Traditional therapies have not significantly improved survival, and recently pancreatic cancer has been deemed a "cold" tumor due to its poor response to immunotherapy. Strategies to improve survival of pancreatic cancer are desperately needed. In the current investigation, we studied the effects of an anti-gastrin cancer vaccine, polyclonal antibody stimulator (PAS; formerly called G17DT and Gastrimmune), used alone or in combination with a programmed cell death receptor (PD)-1 immune checkpoint antibody on pancreatic cancer growth, metastases, and the tumor microenvironment (TME). Immune-competent female C57BL/6 mice bearing syngeneic orthotopic murine pancreatic cancer treated with PAS had significantly smaller tumors and fewer metastases. Examination of the TME demonstrated decreased fibrosis with fewer M2 and more M1 tumor-associated macrophages. Expression of the E-cadherin gene was significantly increased and expression of the TGFßR2 gene was decreased compared with controls. Mice treated with PAS or the combination of PAS and PD-1 antibody exhibited significantly less tumor expression of phospho-paxillin, the focal adhesion protein ß-catenin, and matrix metalloproteinase-7. This study suggests that inhibition of the cancer-promoting effects of gastrin in pancreatic cancer can decrease metastases by altering the TME and decreasing pathways that activate the epithelial mesenchymal transition. The PAS vaccine appears to change the TME, making it more susceptible to therapy with an immune checkpoint antibody. This novel combination of two immunotherapies may improve survival of pancreatic cancer by decreasing both tumor growth and metastasis formation.NEW & NOTEWORTHY Survival from advanced pancreatic cancer is poor, in part due to dense fibrosis of the tumor microenvironment, increased number of M2-polarized macrophages that promote angiogenesis and invasion, and lack of "target-specific" therapy. Herein, we report that a tumor vaccine that selectively targets gastrin decreases pancreatic cancer growth and metastases. Furthermore, the gastrin vaccine polyclonal antibody stimulator alters the tumor microenvironment rendering it more responsive to immunotherapy with a programmed cell death receptor-1 immune checkpoint antibody.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Gastrinas/inmunología , Inmunoterapia/métodos , Neoplasias Pancreáticas/terapia , Animales , Cadherinas/genética , Cadherinas/metabolismo , Vacunas contra el Cáncer/uso terapéutico , Línea Celular , Línea Celular Tumoral , Femenino , Gastrinas/uso terapéutico , Macrófagos/metabolismo , Metaloproteinasa 7 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Neoplasias Pancreáticas/patología , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral , beta Catenina/genética , beta Catenina/metabolismo
9.
Pain ; 160(2): 345-357, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30281531

RESUMEN

Pain is associated with negative emotions such as anxiety, but the underlying neurocircuitry and modulators of the association of pain and anxiety remain unclear. The neuropeptide cholecystokinin (CCK) has both pronociceptive and anxiogenic properties, so we explored the role of CCK in anxiety and nociception in the central amygdala (CeA), a key area in control of emotions and descending pain pathways. Local infusion of CCK into the CeA of control rats increased anxiety, as measured in the light-dark box test, but had no effect on mechanical sensitivity. By contrast, intra-CeA CCK infusion 4 days after Complete Freund's Adjuvant (CFA) injection into the hindpaw resulted in analgesia, but also in loss of its anxiogenic capacity. Inflammatory conditions induced changes in the CeA CCK signaling system with an increase of CCK immunoreactivity and a decrease in CCK1, but not CCK2, receptor mRNA. In CFA rats, patch-clamp experiments revealed that CCK infusion increased CeA neuron excitability. It also partially blocked the discharge of wide dynamic range neurons in the dorsal spinal cord. These effects of CCK on CeA and spinal neurons in CFA rats were mimicked by the specific CCK2 receptor agonist, gastrin. This analgesic effect was likely mediated by identified CeA neurons projecting to the periaqueductal gray matter that express CCK receptors. Together, our data demonstrate that intra-CeA CCK infusion activated a descending CCK2 receptor-dependent pathway that inhibited spinal neuron discharge. Thus, persistent pain induces a functional switch to a newly identified analgesic capacity of CCK in the amygdala, indicating central emotion-related circuit controls pain transmission in spinal cord.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Colecistoquinina/metabolismo , Dolor/patología , Receptor de Colecistoquinina B/metabolismo , Transducción de Señal/fisiología , Amígdala del Cerebelo/patología , Animales , Adaptación a la Oscuridad/efectos de los fármacos , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Adyuvante de Freund/toxicidad , Gastrinas/uso terapéutico , Glutamato Descarboxilasa/metabolismo , Inflamación/inducido químicamente , Inflamación/complicaciones , Masculino , Neuronas/efectos de los fármacos , Neuronas/fisiología , Nocicepción/efectos de los fármacos , Dolor/etiología , Umbral del Dolor/efectos de los fármacos , Sustancia Gris Periacueductal/efectos de los fármacos , Sustancia Gris Periacueductal/fisiología , Ratas , Ratas Sprague-Dawley , Receptor de Colecistoquinina B/agonistas , Receptor de Colecistoquinina B/antagonistas & inhibidores , Receptor de Colecistoquinina B/genética , Transducción de Señal/efectos de los fármacos , Sincalida/uso terapéutico , Tetragastrina/análogos & derivados , Tetragastrina/uso terapéutico
10.
Nat Rev Endocrinol ; 15(2): 90-104, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30446744

RESUMEN

Obesity and its comorbidities, such as type 2 diabetes mellitus and cardiovascular disease, constitute growing challenges for public health and economies globally. The available treatment options for these metabolic disorders cannot reverse the disease in most individuals and have not substantially reduced disease prevalence, which underscores the unmet need for more efficacious interventions. Neurobiological resilience to energy homeostatic perturbations, combined with the heterogeneous pathophysiology of human metabolic disorders, has limited the sustainability and efficacy of current pharmacological options. Emerging insights into the molecular origins of eating behaviour, energy expenditure, dyslipidaemia and insulin resistance suggest that coordinated targeting of multiple signalling pathways is probably necessary for sizeable improvements to reverse the progression of these diseases. Accordingly, a broad set of combinatorial approaches targeting feeding circuits, energy expenditure and glucose metabolism in concert are currently being explored and developed. Notably, several classes of peptide-based multi-agonists and peptide-small molecule conjugates with superior preclinical efficacy have emerged and are currently undergoing clinical evaluation. Here, we summarize advances over the past decade in combination pharmacotherapy for the management of obesity and type 2 diabetes mellitus, exclusively focusing on large-molecule formats (notably enteroendocrine peptides and proteins) and discuss the associated therapeutic opportunities and challenges.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hormonas Gastrointestinales/uso terapéutico , Péptido 1 Similar al Glucagón/uso terapéutico , Terapia Molecular Dirigida/métodos , Obesidad/tratamiento farmacológico , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Animales , Índice de Masa Corporal , Diabetes Mellitus Tipo 2/diagnóstico , Esquema de Medicación , Quimioterapia Combinada , Femenino , Gastrinas/uso terapéutico , Humanos , Masculino , Enfermedades Metabólicas/diagnóstico , Enfermedades Metabólicas/tratamiento farmacológico , Metformina/uso terapéutico , Ratones , Obesidad/diagnóstico , Pronóstico , Medición de Riesgo , Resultado del Tratamiento
11.
Mol Biol Rep ; 45(6): 1759-1767, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30143975

RESUMEN

Treatment with radionuclide labeled regulatory peptides is a promising tool in the management of patients with inoperable receptor positive neuroendocrine tumors. Peptide receptor lutetium-177 radionuclide therapy currently has gained ample attention due to high specific accumulation of regulatory peptides at tumor cell surface and promising characteristics of ß- and γ-energy photons of lutetium-177 radionuclide. In this study gastrin peptides analogues were labeled with lutetium-177 by subsequent mixing of 177LuCl3 (~ 185 MBq), ammonium acetate buffer of 5 pH, gentistic acid, aqueous solution of gastrin peptide analogues (1 mg/mL) and heating the reaction mixture at 98 °C which resulted in high radiochemical yield (> 96%). Chromatographic analysis was carried out to analyze the radiochemical purity. The shelf life and serum stability results showed the labeled peptides are sufficiently stable up to 4-h. Glomerular filtration rate study results showed moderate filtration through kidneys. The GFR values of 177Lu-MGCL2 and 177Lu-MGCL4 was noted 48 mL/min and 45 mL/min, respectively. Biodistribution and scintigraphy study using rat and rabbit models showed minimal non-target accumulation, moderate uptake by liver and kidneys. The promising radiochemical yield, stability, GFR values and biodistribution results of 177Lu-MGCL2 & 4 indicate, the agents can be tested clinically for PRRT procedures.


Asunto(s)
Lutecio/uso terapéutico , Tumores Neuroendocrinos/terapia , Radioisótopos/uso terapéutico , Nanomedicina Teranóstica/métodos , Animales , Gastrinas/uso terapéutico , Masculino , Péptidos/uso terapéutico , Conejos , Radioquímica/métodos , Radiofármacos/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptor de Colecistoquinina B
12.
Mol Pharm ; 14(9): 3045-3058, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28728415

RESUMEN

Minigastrin (MG) analogues specifically target cholecystokinin-2 receptors (CCK2R) expressed in different tumors and enable targeted radiotherapy of advanced and disseminated disease when radiolabeled with a beta emitter such as 177Lu. Especially truncated MG analogues missing the penta-Glu sequence are associated with low kidney retention and seem therefore most promising for therapeutic use. Based on [d-Glu1,desGlu2-6]MG (MG11) we have designed the two cyclic MG analogues cyclo1,9[γ-d-Glu1,desGlu2-6,d-Lys9]MG (cyclo-MG1) and cyclo1,9[γ-d-Glu1,desGlu2-6,d-Lys9,Nle11]MG (cyclo-MG2). In the present work we have developed and preclinically evaluated a pharmaceutical kit formulation for the labeling with 177Lu of the two DOTA-conjugated cyclic MG analogues. The stability of the kits during storage as well as the stability of the radiolabeled peptides was investigated. A cell line stably transfected with human CCK2R and a control cell line without receptor expression were used for in vitro and in vivo studies with the radioligands prepared from kit formulations. In terms of stability 177Lu-DOTA-cyclo-MG2 showed advantages over 177Lu-DOTA-cyclo-MG1. Still, for both radioligands a high receptor-mediated cell uptake and favorable pharmacokinetic profile combining receptor-specific tumor uptake with low unspecific tissue uptake and low kidney retention were confirmed. Investigating the therapy efficacy and treatment toxicity in xenografted BALB/c nude mice a receptor-specific and comparable therapeutic effect could be demonstrated for both radioligands. A 1.7- to 2.6-fold increase in tumor volume doubling time was observed for receptor-positive tumors in treated versus untreated animals, which was 39-73% higher when compared to receptor-negative tumors. The treatment was connected with transient bone marrow toxicity and minor signs of kidney toxicity. All together the obtained results support further studies for the clinical translation of this new therapeutic approach.


Asunto(s)
Gastrinas/uso terapéutico , Receptor de Colecistoquinina B/metabolismo , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/radioterapia , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Receptores de Péptidos/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
Eur J Pharm Sci ; 85: 1-9, 2016 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-26826279

RESUMEN

INTRODUCTION: A variety of radiolabelled minigastrin analogues targeting the cholecystokinin 2 (CCK2) receptor were developed and compared in a concerted preclinical testing to select the most promising radiotracer for diagnosis and treatment of medullary thyroid carcinoma (MTC). DOTA-DGlu-DGlu-DGlu-DGlu-DGlu-DGlu-Ala-Tyr-Gly-Trp-Met-Asp-Phe-NH2 (CP04) after labelling with (111)In displayed excellent characteristics, such as high stability, receptor affinity, specific and persistent tumour uptake and low kidney retention in animal models. Therefore, it was selected for further clinical evaluation within the ERA-NET project GRAN-T-MTC. Here we report on the development of a pharmaceutical freeze-dried formulation of the precursor CP04 for a first multi-centre clinical trial with (111)In-CP04 in MTC patients. MATERIALS AND METHODS: The kit formulation was optimised by adjustment of buffer, additives and radiolabelling conditions. Three clinical grade batches of a final kit formulation with two different amounts of peptide (10 or 50 µg) were prepared and radiolabelled with (111)In. Quality control and stability assays of both the kits and the resulting radiolabelled compound were performed by HPLC analysis. RESULTS: Use of ascorbic acid buffer (pH4.5) allowed freeze-drying of the kit formulation with satisfactory pellet-formation. Addition of methionine and gentisic acid as well as careful selection of radiolabelling temperature was required to avoid extensive oxidation of the Met(11)-residue. Trace metal contamination, in particular Zn, was found to be a major challenge during the pharmaceutical filling process in particular for the 10 µg formulation. The final formulations contained 10 or 50 µg CP04, 25mg ascorbic acid, 0.5mg gentisic acid and 5mg L-methionine. The radiolabelling performed by incubation of 200-250 MBq (111)InCl3 at 90 °C for 15 min resulted in reproducible radiochemical purity (RCP) >94%. Kit-stability was proven for >6 months at +5 °C and at +25 °C. The radiolabelled product was stable for >4h at +25 °C. CONCLUSION: A kit formulation to prepare (111)In-CP04 for clinical application was developed, showing high stability of the kit as well as high RCP of the final product.


Asunto(s)
Gastrinas/química , Radioisótopos de Indio/química , Radioisótopos/química , Radiofármacos/química , Carcinoma Neuroendocrino/tratamiento farmacológico , Química Farmacéutica/métodos , Ensayos Clínicos como Asunto , Estabilidad de Medicamentos , Liofilización/métodos , Gastrinas/uso terapéutico , Humanos , Metionina/química , Estudios Multicéntricos como Asunto , Péptidos/química , Neoplasias de la Tiroides/tratamiento farmacológico
14.
Cancer Chemother Pharmacol ; 74(3): 479-86, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25030089

RESUMEN

BACKGROUND: The G17DT is a novel human immunogen that raises antibodies to the growth factor gastrin 17 (G17). The purpose of this study was to determine the safety and efficacy of G17DT in combination with irinotecan in patients refractory to irinotecan, and to correlate efficacy with anti-G17 immune response. METHODS: Patients received G17DT immunogen as a single intramuscular injection of 500 µg at weeks 1, 5, 9, and 26. Irinotecan was administered as an intravenous infusion of 125 mg/m(2) over 90 min starting at week 5. Each cycle of treatment consisted of irinotecan administered once weekly for 4 weeks, followed by a 2-week rest period. RESULTS: Of 161 patients who received G17DT, the best overall tumor response in the intent-to-treat population was complete response 0 (0 %), partial response 3 (3 %), stable disease 32 (32 %), and progressive disease 64 (65 %). Median survival was 217 days. About 94 (62 %) subjects evaluable for antibody titers were anti-G17 responders. Survival was significantly longer for anti-G17 responders compared with non-responders (9.0 vs. 5.6 months; P < 0.001). Toxicity was consistent with irinotecan (diarrhea, nausea, anemia, vomiting, fatigue, constipation, anorexia, and neutropenia) except for injection site reactions (pain 42 %, induration 13 %, edema 11 %, erythema 10 %, and three abscesses) attributed to G17DT in 52 % of the patients. CONCLUSION: Treatment with G17DT in combination with irinotecan results in an acceptable anti-G17 immune response, which correlated with promising survival activity in patients refractory to irinotecan-based chemotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Gastrinas/uso terapéutico , Anciano , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/efectos adversos , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/mortalidad , Diarrea/inducido químicamente , Esquema de Medicación , Femenino , Gastrinas/administración & dosificación , Gastrinas/efectos adversos , Humanos , Infusiones Intravenosas , Inyecciones Intramusculares , Irinotecán , Masculino , Persona de Mediana Edad , Náusea/inducido químicamente , Neutropenia/inducido químicamente , Resultado del Tratamiento , Vómitos/inducido químicamente
15.
Eur J Cancer Prev ; 23(4): 258-63, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24469263

RESUMEN

Gastrin has been identified as the principal effector of gastric secretion, but several studies have demonstrated its role as a biomarker of cancer risk and as a growth factor for colorectal, stomach, liver, and pancreatic cancer. Hypergastrinemia characterizes autoimmune gastritis, with body and fundic gland atrophy and increased risk for both gastric adenocarcinoma and neuroendocrine tumors. Gastric type I carcinoids develop in the context of autoimmune gastritis because of the stimulus exerted by gastrin on enterochromaffin-like cells and remain gastrin-sensitive for long durations because the removal of hypergastrinemia leads to tumor regression. The treatment of gastric carcinoid is still open to debate, but when the disease frequently relapses, or is multicentric or infiltrating, surgery is advocated or, in the alternative, a costly and long-lasting treatment with long-acting somatostatin analogues is prescribed. A technology allowing the preparation of an immunogen eliciting an immune system response with generation of antibodies against G17 has been developed. This vaccine has been tested in patients with colorectal, pancreatic or advanced gastric cancer. The vaccine has also been used in the treatment of gastric type I carcinoids, and the administration of G17DT in patients harboring these lesions leads to carcinoid regression. Antigastrin vaccination in the treatment of gastrointestinal cancer obviously needs validation, but this immunotherapy may well represent a simple, inexpensive, and active 'adjuvant' treatment.


Asunto(s)
Adenocarcinoma/metabolismo , Tumor Carcinoide/metabolismo , Neoplasias Colorrectales/metabolismo , Gastrinas/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Gástricas/metabolismo , Enfermedades Autoinmunes/metabolismo , Vacunas contra el Cáncer/uso terapéutico , Tumor Carcinoide/prevención & control , Tumor Carcinoide/terapia , Gastrinas/uso terapéutico , Gastritis/metabolismo , Humanos , Neoplasias Gástricas/prevención & control , Neoplasias Gástricas/terapia
16.
Pharm Res ; 30(11): 2843-54, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23793991

RESUMEN

PURPOSE: Our objective was to develop novel nanocarriers (protected graft copolymer, PGC) that improve the stability of heparin binding EGF (HBEGF) and gastrin and then to use PGC-formulated HBEGF (PGC-HBEGF) and Omeprazole (+/- PGC-gastrin) for normalizing fasting blood glucose (FBG) and improving islet function in diabetic mice. METHODS: HBEGF, PGC-HBEGF, Omeprazole, Omeprazole + PGC-HBEGF, Omeprazole + PGC-gastrin + PGC-HBEGF and epidermal growth factor (EGF) + gastrin were tested in multiple low dose streptozotocin diabetic mice. RESULTS: Omeprazole + PGC-HBEGF normalized FBG and is better than EGF + gastrin at improving islet function and decreasing insulitis. Groups treated with Omeprazole, Omeprazole + PGC-HBEGF, or EGF + gastrin have significantly improved islet function versus saline control. All animals that received PGC-HBEGF had significantly reduced islet insulitis versus saline control. Non-FBG was lower for Omeprazole + PGC-gastrin + PGC-HBEGF but Omeprazole + PGC-HBEGF alone showed better FBG and glucose tolerance. CONCLUSIONS: Omeprazole + PGC-HBEGF provides a sustained exposure to both EGFRA and gastrin, improves islet function, and decreases insulitis in multiple low dose streptozotocin diabetic mice. Although HBEGF or EGF elevates non-FBG, it facilitates a reduction of insulitis and, in the presence of Omeprazole, provides normalization of FBG at the end of treatment. The study demonstrates Omeprazole and PGC-HBEGF is a viable treatment for diabetes.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Portadores de Fármacos/química , Gastrinas/administración & dosificación , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Omeprazol/administración & dosificación , Animales , Glucemia/análisis , Diabetes Mellitus Experimental/patología , Gastrinas/farmacocinética , Gastrinas/uso terapéutico , Factor de Crecimiento Similar a EGF de Unión a Heparina , Péptidos y Proteínas de Señalización Intercelular/farmacocinética , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Masculino , Ratones , Nanoestructuras/química , Omeprazol/farmacocinética , Omeprazol/uso terapéutico , Páncreas/efectos de los fármacos , Páncreas/patología , Polímeros/química , Estreptozocina
18.
Immunotherapy ; 4(6): 587-99, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22788127

RESUMEN

Chronic atrophic gastritis, a precancerous change for gastric cancer, shows a loss of appropriate glands, Helicobacter pylori infection and autoimmune gastritis being the two main etiologic factors. While H. pylori eradication is the mandatory treatment for the former, no etiologic treatment is available for the latter, in which a Th1-type response, modulated by Tregs and Th17 cells, is involved. H. pylori-related atrophic gastritis is a risk factor for gastric adenocarcinoma, while autoimmune atrophic gastritis is also linked to a substantial risk of gastric type I carcinoid, related to the chronic stimulus exerted by hypergastrinemia on enterochromaffin-like cells. Several studies have been published on gastric cancer treatment through an active specific immunotherapy, aimed at improving the immunoregulatory response and increasing the circulating tumor-specific T cells. No study on immunotherapy of carcinoids is available but, in our experience, the administration of an antigastrin 17 vaccine induced carcinoid regression in two out of three patients treated.


Asunto(s)
Adenocarcinoma/terapia , Vacunas contra el Cáncer/uso terapéutico , Células Similares a las Enterocromafines/efectos de los fármacos , Gastrinas/uso terapéutico , Gastritis Atrófica/terapia , Inmunoterapia/métodos , Lesiones Precancerosas/terapia , Neoplasias Gástricas/terapia , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Vacunas contra el Cáncer/farmacología , Células Similares a las Enterocromafines/patología , Gastrinas/antagonistas & inhibidores , Gastrinas/inmunología , Gastrinas/farmacología , Gastritis Atrófica/inmunología , Gastritis Atrófica/patología , Humanos , Inmunoterapia/tendencias , Lesiones Precancerosas/inmunología , Lesiones Precancerosas/patología , Riesgo , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/patología
19.
Pancreas ; 41(3): 374-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22228104

RESUMEN

OBJECTIVES: This study aimed to investigate G17DT, an immunogen producing neutralizing antibodies against the tumor growth factors amidated and glycine-extended forms of gastrin-17, in the treatment of pancreatic cancer. METHODS: A randomized, double-blind, placebo-controlled, group-sequential multicenter trial of G17DT in patients with advanced pancreatic cancer unsuitable for or unwilling to take chemotherapy. Inclusion criteria were a Karnofsky performance score of 60 or higher and a life expectancy of more than 2 months. Patients received G17DT or placebo emulsion at weeks 0, 1, 3, 24, and 52. The primary end point was survival, and secondary end points were tolerability, Karnofsky performance. RESULTS: A total of 154 patients were recruited: 79 G17DT and 75 placebo. A final analysis of the intention-to-treat population, using a proportional hazards model, stratifying by disease stage and adjusting for interim analysis, gave a hazard ratio for mortality of 0.75 (95% confidence interval, 0.51-1.10, P = 0.138; G17DT/placebo). A conventional analysis without adjustment for disease stage or interim analysis, censoring for chemotherapy and excluding protocol violators, gave median survival periods of 151 (G17DT) and 82 days (placebo) (log-rank test, P = 0.03).Patients developing anti-G17DT responses (73.8%) survived longer than nonresponders or those on placebo (median survival, 176 vs 63 vs 83; log-rank test, P = 0.003). G17DT was well tolerated.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Gastrinas/inmunología , Gastrinas/uso terapéutico , Neoplasias Pancreáticas/terapia , Adulto , Anciano , Anciano de 80 o más Años , Vacunas contra el Cáncer/efectos adversos , Método Doble Ciego , Europa (Continente) , Femenino , Gastrinas/efectos adversos , Humanos , Estimación de Kaplan-Meier , Estado de Ejecución de Karnofsky , Esperanza de Vida , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Placebos , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Factores de Tiempo , Resultado del Tratamiento
20.
J Mol Med (Berl) ; 90(6): 707-18, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22228178

RESUMEN

Human anion exchanger 2 (AE2) is a plasma membrane protein that regulates intracellular pH and cell volume. AE2 contributes to transepithelial transport of chloride and bicarbonate in normal colon and other epithelial tissues. We now report that AE2 overexpression in colon cancer cells is correlated with expression of the nuclear proliferation marker, Ki67. Survival analysis of 24 patients with colon cancer in early stage or 33 patients with tubular adenocarcinoma demonstrated that expression of AE2 is correlated with poor prognosis. Cellular and molecular experiments indicated that AE2 expression promoted proliferation of colon cancer cells. In addition, we found that transcription factor EGR1 underlies AE2 upregulation and the AE2 sequester p16INK4a (P16) in the cytoplasm of colon cancer cells. Cytoplasmic P16 enhanced ERK phosphorylation and promoted proliferation of colon cancer cells. Gastrin inhibited proliferation of colon cancer cells by suppressing expression of EGR1 and AE2 and by blocking ERK phosphorylation. Taken together, our data describe a novel EGR1/AE2/P16/P-ERK signaling pathway in colon carcinogenesis, with implications for pathologic prognosis and for novel therapeutic approaches.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Antiportadores/metabolismo , Neoplasias del Colon/fisiopatología , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Gastrinas/farmacología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de Transporte de Anión/genética , Antineoplásicos/farmacología , Antiportadores/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Gastrinas/uso terapéutico , Humanos , Inmunohistoquímica , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Proteínas SLC4A
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...