Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 15: 1302015, 2024.
Article in English | MEDLINE | ID: mdl-38510652

ABSTRACT

Background: Elevated levels of somatostatin blunt glucagon counterregulation during hypoglycemia in type 1 diabetes (T1D) and this can be improved using somatostatin receptor 2 (SSTR2) antagonists. Hypoglycemia also occurs in late-stage type 2 diabetes (T2D), particularly when insulin therapy is initiated, but the utility of SSTR2 antagonists in ameliorating hypoglycemia in this disease state is unknown. We examined the efficacy of a single-dose of SSTR2 antagonists in a rodent model of T2D. Methods: High-fat fed (HFF), low dose streptozotocin (STZ, 35 mg/kg)-induced T2D and HFF only, nondiabetic (controls-no STZ) rats were treated with the SSTR2 antagonists ZT-01/PRL-2903 or vehicle (n = 9-11/group) 60 min before an insulin tolerance test (ITT; 2-12 U/kg insulin aspart) or an oral glucose tolerance test (OGTT; 2 g/kg glucose via oral gavage) on separate days. Results: This rodent model of T2D is characterized by higher baseline glucose and HbA1c levels relative to HFF controls. T2D rats also had lower c-peptide levels at baseline and a blunted glucagon counterregulatory response to hypoglycemia when subjected to the ITT. SSTR2 antagonists increased the glucagon response and reduced incidence of hypoglycemia, which was more pronounced with ZT-01 than PRL-2903. ZT-01 treatment in the T2D rats increased glucagon levels above the control response within 60 min of dosing, and values remained elevated during the ITT (glucagon Cmax: 156 ± 50 vs. 77 ± 46 pg/mL, p < 0.01). Hypoglycemia incidence was attenuated with ZT-01 vs. controls (63% vs. 100%) and average time to hypoglycemia onset was also delayed (103.1 ± 24.6 vs. 66.1 ± 23.6 min, p < 0.05). ZT-01 administration at the OGTT onset increased the glucagon response without exacerbating hyperglycemia (2877 ± 806 vs. 2982 ± 781), potentially due to the corresponding increase in c-peptide levels (6251 ± 5463 vs. 14008 ± 5495, p = 0.013). Conclusion: Treatment with SSTR2 antagonists increases glucagon responses in a rat model of T2D and results in less hypoglycemia exposure. Future studies are required to determine the best dosing periods for chronic SSTR2 antagonism treatment in T2D.

2.
Diabetes Obes Metab ; 25(6): 1547-1556, 2023 06.
Article in English | MEDLINE | ID: mdl-36734462

ABSTRACT

AIMS: To examine if glucagon counterregulatory defects exist in a rat model of prediabetes (pre-T2D) and to assess if a selective somatostatin receptor 2 antagonist (SSTR2a), ZT-01, enhances the glucagon response to insulin-induced hypoglycaemia. MATERIALS AND METHODS: Hyperglycaemia was induced in 8- to 9-week-old male, Sprague-Dawley rats via 7 weeks of high-fat diet followed by a single, low-dose intraperitoneal injection of streptozotocin (30 mg/kg). After 2 weeks of basal insulin therapy (0-4 U/d insulin glargine, administered subcutaneously [SC]) to facilitate partial glycaemic recovery and a pre-T2D phenotype, n = 17 pre-T2D and n = 10 normal chow-fed control rats underwent the first of two hypoglycaemic treatment-crossover experiments, separated by a 1-week washout period. On each experimental day, SSTR2a (3 mg/kg ZT-01, SC) or vehicle was administered 1 hour prior to insulin-induced hypoglycaemia (insulin aspart, 6 U/kg, SC). RESULTS: Glucagon counterregulation was marginally reduced with the induction of pre-T2D. Treatment with SSTR2a raised peak plasma glucagon levels and glucagon area under the curve before and after insulin overdose in both and pre-T2D rats. Blood glucose concentration was elevated by 30 minutes after SSTR2a treatment in pre-T2D rats, and hypoglycaemia onset (≤3.9 mmol/L) was delayed by 15 ± 12 minutes compared with vehicle (P < 0.001), despite similar glucose nadirs in the two treatment groups (1.4 ± 0.3 mmol/L). SSTR2a treatment had no effect on blood glucose levels in the control group or on the hypoglycaemia-induced decline in plasma C-peptide levels in either group. CONCLUSIONS: Treatment with an SSTR2a increases glucagon responsiveness and delays the onset of insulin-induced hypoglycaemia in this rat model of pre-T2D where only a modest deficiency in glucagon counterregulation exists.


Subject(s)
Diabetes Mellitus, Type 2 , Hypoglycemia , Prediabetic State , Male , Rats , Animals , Glucagon , Blood Glucose , Prediabetic State/chemically induced , Prediabetic State/drug therapy , Rats, Sprague-Dawley , Insulin Aspart , Diabetes Mellitus, Type 2/drug therapy
3.
Perit Dial Int ; 43(4): 324-333, 2023 07.
Article in English | MEDLINE | ID: mdl-36588412

ABSTRACT

BACKGROUND: Small hyperbranched polyglycerol (HPG) has been recently of interest for peritoneal dialysis, but its pharmacokinetics is barely understood. This study investigated the absorption, distribution and excretion of 1 and 3 kDa HPG. METHODS: Rats (naive, 5/6 nephrectomy (5/6 Nx) or bilateral nephrectomy (BNx)) received a single dose of 3H-labelled HPG-containing solutions intraperitoneally (IP) or intravenously (IV). Radioactivity in tissues, urine and faeces was counted using a scintillation counter. Pharmacokinetic parameters were calculated using WinNonlin software. RESULTS: During 8-h dwell with IP injected therapeutic dose of HPG-based hypertonic solutions, the plasma levels of 1 kDa HPG reached the peak at 2 h, followed by a decrease to the end, whereas 3 kDa HPG increased for the duration of the 8 h. At the experimental endpoint, the distribution of both sizes of HPG in major organs was minimal, whereas most of 1 kDa HPG was excreted via urine, and of 3 kDa remained in peritoneal cavity. The elimination of both 1 and 3 kDa HPG after either IP or IV administration was significantly delayed by 5/6 Nx or BNx as compared to naive controls. Further, 24-h faecal excretion of HPG (3 kDa) was <5% of injected dose that was not different between healthy and BNx rats. CONCLUSION: Data suggest size-dependent peritoneal absorption of osmotic HPG that are not specifically absorbed by any of the organs tested. The clearance of small HPG mainly depends on kidney excretion, implying the risk of HPG accumulation in patients with end-stage kidney disease who receive maintenance dialysis with HPG.


Subject(s)
Peritoneal Dialysis , Rats , Animals , Polymers , Peritoneal Cavity , Glycerol/pharmacokinetics
4.
Front Pharmacol ; 14: 1295639, 2023.
Article in English | MEDLINE | ID: mdl-38298268

ABSTRACT

Glucose homeostasis is primarily maintained by pancreatic hormones, insulin and glucagon, with an emerging role for a third islet hormone, somatostatin, in regulating insulin and glucagon responses. Under healthy conditions, somatostatin secreted from pancreatic islet δ-cells inhibits both insulin and glucagon release through somatostatin receptor- induced cAMP-mediated downregulation and paracrine inhibition of ß- and α-cells, respectively. Since glucagon is the body's most important anti-hypoglycemic hormone, and because glucagon counterregulation to hypoglycemia is lost in diabetes, the study of somatostatin biology has led to new investigational medications now in development that may help to restore glucagon counterregulation in type 1 diabetes. This review highlights the normal regulatory role of pancreatic somatostatin signaling in healthy islet function and how the inhibition of somatostatin receptor signaling in pancreatic α-cells may restore normal glucagon counterregulation in diabetes mellitus.

5.
Diabetes Obes Metab ; 24(5): 908-917, 2022 05.
Article in English | MEDLINE | ID: mdl-35060297

ABSTRACT

AIM: To evaluate the pharmacokinetics and efficacy of a novel somatostatin receptor 2 antagonist, ZT-01, to stimulate glucagon release in rats with type 1 diabetes (T1D). METHODS: The pharmacokinetics of ZT-01 and PRL-2903 were assessed following intraperitoneal or subcutaneous dosing at 10 mg/kg. We compared the efficacy of ZT-01 with PRL-2903 to prevent hypoglycaemia during an insulin bolus challenge and under hypoglycaemic clamp conditions. RESULTS: Within 1 hour after intraperitoneal administration, ZT-01 achieved more than 10-fold higher plasma Cmax compared with PRL-2903. Twenty-four hour exposure was 4.7× and 11.3× higher with ZT-01 by the intraperitoneal and subcutaneous routes, respectively. The median time to reach hypoglycaemia of more than 3.0 mmol/L was 60, 70, and 125 minutes following vehicle, PRL-2903, or ZT-01 administration, respectively. Furthermore, rats receiving ZT-01 had significantly higher glucose nadirs following insulin administration compared with PRL-2903- and vehicle-treated rats. During the hypoglycaemic clamp, ZT-01 increased peak glucagon responses by ~4-fold over PRL-2903. CONCLUSIONS: We conclude that ZT-01 may be effective in restoring glucagon responses and preventing the onset of hypoglycaemia in patients with T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Hypoglycemia , Receptors, Somatostatin , Animals , Blood Glucose , Diabetes Mellitus, Type 1/drug therapy , Glucagon , Humans , Hypoglycemia/chemically induced , Hypoglycemia/prevention & control , Insulin , Rats , Receptors, Somatostatin/antagonists & inhibitors
6.
Endocrinology ; 162(12)2021 12 01.
Article in English | MEDLINE | ID: mdl-34477204

ABSTRACT

Recent antecedent hypoglycemia is a known source of defective glucose counter-regulation in diabetes; the mechanisms perpetuating the cycle of progressive α-cell failure and recurrent hypoglycemia remain unknown. Somatostatin has been shown to suppress the glucagon response to acute hypoglycemia in rodent models of type 1 diabetes. We hypothesized that somatostatin receptor 2 antagonism (SSTR2a) would restore glucagon counterregulation and delay the onset of insulin-induced hypoglycemia in recurrently hypoglycemic, nondiabetic male rats. Healthy, male, Sprague-Dawley rats (n = 39) received bolus injections of insulin (10 U/kg, 8 U/kg, 5 U/kg) on 3 consecutive days to induce hypoglycemia. On day 4, animals were then treated with SSTR2a (10 mg/kg; n = 17) or vehicle (n = 12) 1 hour prior to the induction of hypoglycemia using insulin (5 U/kg). Plasma glucagon level during hypoglycemia was ~30% lower on day 3 (150 ± 75 pg/mL; P < .01), and 68% lower on day 4 in the vehicle group (70 ± 52 pg/mL; P < .001) compared with day 1 (219 ± 99 pg/mL). On day 4, SSTR2a prolonged euglycemia by 25 ± 5 minutes (P < .05) and restored the plasma glucagon response to hypoglycemia. Hepatic glycogen content of SSTR2a-treated rats was 35% lower than vehicle controls after hypoglycemia induction on day 4 (vehicle: 20 ± 7.0 vs SSTR2a: 13 ± 4.4 µmol/g; P < .01). SSTR2a treatment reverses the cumulative glucagon deficit resulting from 3 days of antecedent hypoglycemia in healthy rats. This reversal is associated with decreased hepatic glycogen content and delayed time to hypoglycemic onset. We conclude that recurrent hypoglycemia produces glucagon counterregulatory deficiency in healthy male rats, which can be improved by SSTR2a.


Subject(s)
Glucagon/metabolism , Hypoglycemia/metabolism , Peptides, Cyclic/pharmacology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Glucagon/drug effects , Glucose/metabolism , Hormone Antagonists/pharmacology , Hypoglycemia/pathology , Liver Glycogen/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, Somatostatin/antagonists & inhibitors , Recurrence
7.
BMC Nephrol ; 20(1): 181, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31113397

ABSTRACT

BACKGROUND: Glucose is a primary osmotic agent in peritoneal dialysis (PD) solutions, but its long-term use causes structural alteration of the peritoneal membrane (PM). Hyperbranched polyglycerol (HPG) is a promising alternative to glucose. This study was designed to compare the cellular responses of human peritoneal mesothelial cells (HPMCs) to these two different osmotic agents in a hypertonic solution using transcriptome analysis. METHODS: Cultured HPMCs were repeatedly exposed to HPG-based or Physioneal 40 (PYS, glucose 2.27%) hypertonic solutions. Transcriptome datasets were produced using Agilent SurePrint G3 Human GE 8 × 60 microarray. Cellular signaling pathways were examined by Ingenuity Pathway Analysis (IPA). Protein expression was examined by flow cytometry analysis and Western blotting. RESULTS: The HPG-containing solution was better tolerated compared with PYS, with less cell death and disruption of cell transcriptome. The levels of cell death in HPG- or PYS- exposed cells were positively correlated with the number of affected transcripts (HPG: 128 at day 3, 0 at day 7; PYS: 1799 at day 3, 212 at day 7). In addition to more affected "biosynthesis" and "cellular stress and death" pathways by PYS, both HPG and PYS commonly affected "sulfate biosynthesis", "unfolded protein response", "apoptosis signaling" and "NRF2-mediated oxidative stress response" pathways at day 3. PYS significantly up-regulated HLA-DMB and MMP12 in a time-dependent manner, and stimulated T cell adhesion to HPMCs. CONCLUSION: The lower cytotoxicity of hypertonic HPG solution is in agreement with its transient and minimal impact on the pathways for the "biosynthesis of cell constituents" and the "cellular stress and death". The significant up-regulation of HLA-DMB and MMP12 by PYS may be part of its initiation of immune response in the PM.


Subject(s)
Dialysis Solutions/administration & dosage , Gene Expression Profiling/methods , Peritoneal Cavity/cytology , Peritoneal Dialysis/trends , Signal Transduction/drug effects , Transcriptome/drug effects , Cell Line, Transformed , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Diuretics, Osmotic/administration & dosage , Humans , Jurkat Cells , Organic Chemicals/administration & dosage , Peritoneal Dialysis/methods , Polymethacrylic Acids/administration & dosage , Signal Transduction/genetics , Transcriptome/genetics
8.
J Thromb Haemost ; 17(10): 1645-1654, 2019 10.
Article in English | MEDLINE | ID: mdl-31145837

ABSTRACT

BACKGROUND: Endogenous fibrinolytic activation contributes to coagulopathy and mortality after trauma. Administering tranexamic acid (TXA), an antifibrinolytic agent, is one strategy to reduce bleeding; however, it must be given soon after injury to be effective and minimize adverse effects. Administering TXA topically to a wound site would decrease the time to treatment and could enable both local and systemic delivery if a suitable formulation existed to deliver the drug deep into wounds adequately. OBJECTIVES: To determine whether self-propelling particles could increase the efficacy of TXA. METHODS: Using previously developed self-propelling particles, which consist of calcium carbonate and generate CO2 gas, TXA was formulated to disperse in blood and wounds. The antifibrinolytic properties were assessed in vitro and in a murine tail bleeding assay. Self-propelled TXA was also tested in a swine model of junctional hemorrhage consisting of femoral arteriotomy without compression. RESULTS: Self-propelled TXA was more effective than non-propelled formulations in stabilizing clots from lysis in vitro and reducing blood loss in mice. It was well tolerated when administered subcutaneously in mice up to 300 to 1000 mg/kg. When it was incorporated in gauze, four of six pigs treated after a femoral arteriotomy and without compression survived, and systemic concentrations of TXA reached approximately 6 mg/L within the first hour. CONCLUSIONS: A formulation of TXA that disperses the drug in blood and wounds was effective in several models. It may have several advantages, including supporting local clot stabilization, reducing blood loss from wounds, and providing systemic delivery of TXA. This approach could both improve and simplify prehospital trauma care for penetrating injury.


Subject(s)
Antifibrinolytic Agents/administration & dosage , Calcium Carbonate/chemistry , Carbon Dioxide/chemistry , Drug Carriers , Fibrinolysis/drug effects , Hemorrhage/prevention & control , Tranexamic Acid/administration & dosage , Administration, Topical , Animals , Antifibrinolytic Agents/blood , Antifibrinolytic Agents/chemistry , Disease Models, Animal , Drug Compounding , Female , Hemorrhage/blood , Humans , Mice, Inbred C57BL , Sus scrofa , Time Factors , Tranexamic Acid/blood , Tranexamic Acid/chemistry
9.
Exp Mol Med ; 50(5): 1-11, 2018 05 30.
Article in English | MEDLINE | ID: mdl-29849046

ABSTRACT

Granzyme B (GzmB) is a serine protease that has long been thought to function exclusively in lymphocyte-mediated apoptosis. In recent years, this paradigm has been revisited due to the recognition that GzmB accumulates in the extracellular milieu in many autoimmune and chronic inflammatory disorders, and contributes to impaired tissue remodeling due to the cleavage of extracellular matrix proteins. Knockout studies suggest that GzmB-mediated cleavage of decorin (DCN) contributes to impaired collagen fibrillogenesis and remodeling. As DCN is anti-fibrotic and contributes to reduced hypertrophic scarring, GzmB-induced DCN cleavage could play a role in wound healing following burn injury. In the present study, a novel, gel-formulated, first-in-class small-molecule inhibitor of GzmB, VTI-1002, was assessed in a murine model of impaired, diabetic burn wound healing. VTI-1002 exhibited high specificity, potency, and target selectivity. Gel-formulated VTI-1002 was able to penetrate the stratum corneum and was retained in the skin with minimal systemic absorption. Daily topical administration of VTI-1002 gel for 30 days following thermal injury showed significantly accelerated wound closure, increased DCN protein levels, and collagen organization that was translated into significantly increased wound tensile strength compared to controls. Overall, VTI-1002 gel was well-tolerated in vivo and no adverse events were observed. Topical application of VTI-1002 represents a novel therapeutic approach for the treatment of cutaneous burn wounds.


Subject(s)
Burns/pathology , Granzymes/antagonists & inhibitors , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/pharmacology , Wound Healing/drug effects , Administration, Topical , Animals , Cicatrix/pathology , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Granzymes/metabolism , Male , Mice, Inbred C57BL
10.
BJU Int ; 122(5): 898-908, 2018 11.
Article in English | MEDLINE | ID: mdl-29862643

ABSTRACT

OBJECTIVE: To investigate the effect of three anticancer drugs (mitomycin c (MMC), doxorubicin or gemcitabine) on bladder wall morphology and the uptake of paclitaxel or docetaxel following coadministration. The primary objective of this study was to measure the uptake of MMC, doxorubicin or gemcitabine with or without exposure of the tissue to amine terminated cationic nanoparticles (CNPs) and to investigate any possible exfoliation effects of the three drugs on intact bladder tissue. The secondary objective was to investigate the uptake of taxane drugs (docetaxel, DTX) and paclitaxel, (PTX) from surfactant micelle formulations in the presence of MMC, doxorubicin or gemcitabine. MATERIALS AND METHODS: Sections of fresh pig bladder tissue were incubated in Franz diffusion cells with the urothelial side exposed to solutions of doxorubicin, MMC and gemcitabine containing radioactive drug for 90 min. Some tissue samples were simultaneously exposed to each of the three drugs in combination with the surfactant micelle formulations of PTX (Taxol) or DTX (Taxotere). Tissue sections were then cryostat sectioned for drug quantitation by liquid scintillation counting or fixed for scanning electron microscopy and haematoxylin and eosin staining. RESULTS: All three drugs caused exfoliation of the urothelial layer of bladder tissues. Drug uptake studies showed that all three drugs effectively penetrated the lamina propria through to the muscular layer over a 2-h incubation and these levels were unaffected by pre-treatment with CNPs. The uptake levels of the taxane drugs PTX and DTX were significantly enhanced following simultaneous treatment of bladders with MMC, doxorubicin or gemcitabine. CONCLUSION: The exfoliation effects of MMC, doxorubicin and gemcitabine allow for good tissue penetration of these drugs with no additional effect from CNP treatment of bladders. The observed exfoliation effect of these amine-containing drugs probably arises from a cationic interaction with the mucus and urothelium cell layer in a manner similar to that previously reported for CNPs. These studies suggest that the lack of long-term clinical efficacy of these drugs may not arise from poor intravesical drug penetration but may result from a rapid diffusion of the drugs into the deeper vascularised muscular region with rapid drug clearance. The enhanced uptake of PTX or DTX following co-administration with MMC, doxorubicin or gemcitabine probably arises from the removal of the urothelial barrier by exfoliation allowing for improved taxane partitioning into superficial layers. These effects may allow for dual drug intravesical strategies offering greatly improved taxane uptake and potential additive drug effects for improved efficacy.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Bridged-Ring Compounds/pharmacokinetics , Deoxycytidine/analogs & derivatives , Doxorubicin/pharmacokinetics , Mitomycin/pharmacokinetics , Taxoids/pharmacokinetics , Urinary Bladder , Animals , Cations , Deoxycytidine/pharmacokinetics , Male , Nanoparticles , Swine , Urinary Bladder/chemistry , Urinary Bladder/cytology , Urinary Bladder/metabolism , Urothelium/cytology , Urothelium/metabolism , Gemcitabine
11.
Basic Clin Pharmacol Toxicol ; 123(4): 494-503, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29753311

ABSTRACT

Metabolic syndrome (MetS) is commonly observed among peritoneal dialysis (PD) patients, and hyperbranched polyglycerol (HPG) is a promising glucose-sparing osmotic agent for PD. However, the biocompatibility of a HPG-based PD solution (HPG) in subjects with MetS has not been investigated. This study compared the local and systemic effects of a HPG solution with conventional physioneal (PYS) and icodextrin (ICO) PD solutions in rats with MetS. Obese type 2 diabetic ZSF1 rats received a daily intraperitoneal injection of PD solutions (10 mL) for 3 months. The peritoneal membrane (PM) function was determined by ultrafiltration (UF), and the systemic responses were determined by profiling blood metabolic substances, cytokines and oxidative status. Tissue damage was assessed by histology. At the end of the 3-month treatment with PD solutions, PM damage and UF loss in both the PYS and ICO groups were greater than those in the HPG group. Blood analyses showed that compared to the baseline control, the rats in the HPG group exhibited a significant decrease only in serum albumin and IL-6 and a minor glomerular injury, whereas in both the PYS and ICO groups, there were more significant decreases in serum albumin, antioxidant activity, IL-6, KC/GRO (CXCL1) and TNF-α (in ICO only) as well as a more substantial glomerular injury compared to the HPG group. Furthermore, PYS increased serum creatinine, serum glucose and urine production. In conclusion, compared to PYS or ICO solutions, the HPG solution had less adverse effects locally on the PM and systemically on distant organs (e.g. kidneys) and the plasma oxidative status in rats with MetS.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Dialysis Solutions/toxicity , Glycerol/toxicity , Icodextrin/toxicity , Kidney/drug effects , Obesity/metabolism , Peritoneal Dialysis/adverse effects , Peritoneum/drug effects , Polymers/toxicity , Animals , Biomarkers/blood , Cytokines/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Dialysis Solutions/administration & dosage , Disease Models, Animal , Glycerol/administration & dosage , Icodextrin/administration & dosage , Inflammation Mediators/blood , Injections, Intraperitoneal , Kidney/metabolism , Kidney/physiopathology , Male , Obesity/blood , Obesity/genetics , Obesity/physiopathology , Organic Chemicals/administration & dosage , Organic Chemicals/toxicity , Oxidative Stress/drug effects , Peritoneal Dialysis/methods , Peritoneum/metabolism , Peritoneum/physiopathology , Permeability , Polymers/administration & dosage , Rats, Zucker , Time Factors
13.
Shock ; 46(3 Suppl 1): 123-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27206277

ABSTRACT

Hemorrhage is the leading cause of preventable death in trauma, and hemorrhage from noncompressible junctional anatomic sites is particularly difficult to control. The current standard is QuikClot Combat Gauze packing, which requires 3 min of compression. We have created a novel dressing with calcium carbonate microparticles that can disperse and self-propel upstream against flowing blood. We loaded these microparticles with thrombin and tranexamic acid and tested their efficacy in a swine arterial bleeding model without wound compression. Anesthetized immature female swine received 5 mm femoral arteriotomies to induce severe junctional hemorrhage. Wounds were packed with kaolin-based QuikClot Combat Gauze (KG), propelled thrombin-microparticles with protonated tranexamic acid (PTG), or a non-propelling formulation of the same thrombin-microparticles with non-protonated tranexamic acid (NPTG). Wounds were not compressed after packing. Each animal then received one 15 mL/kg bolus of hydroxyethyl starch solution followed by Lactated Ringer as needed for hypotension (maximum: 100 mL/kg) for up to 3 h. Survival was improved with PTG (3-h survival: 8/8, 100%) compared with KG (3/8, 37.5%) and NPTG (2/8, 25%) (P <0.01). PTG animals maintained lower serum lactate and higher hemoglobin concentrations than NPTG (P <0.05) suggesting PTG decreased severity of subsequent hemorrhagic shock. However, total blood loss, Lactated Ringer infusion volumes, and mean arterial pressures of surviving animals were not different between groups (P >0.05). Thus, in this swine model of junctional arterial hemorrhage, gauze with self-propelled, prothrombotic microparticles improved survival and 2 indicators of hemorrhagic shock when applied without compression, suggesting this capability may enable better treatment of non-compressible junctional wounds.


Subject(s)
Bandages , Hemorrhage/drug therapy , Hemorrhage/therapy , Thrombin/administration & dosage , Thrombin/therapeutic use , Tranexamic Acid/administration & dosage , Tranexamic Acid/therapeutic use , Animals , Disease Models, Animal , Female , Hemostatics , Models, Statistical , Swine
14.
Diabetologia ; 59(8): 1724-31, 2016 08.
Article in English | MEDLINE | ID: mdl-27075449

ABSTRACT

AIMS/HYPOTHESIS: Regular exercise is at the cornerstone of care in type 1 diabetes. However, relative hyperinsulinaemia and a blunted glucagon response to exercise promote hypoglycaemia. Recently, a selective antagonist of somatostatin receptor 2, PRL-2903, was shown to improve glucagon counterregulation to hypoglycaemia in resting streptozotocin-induced diabetic rats. The aim of this study was to test the efficacy of PRL-2903 in enhancing glucagon counterregulation during repeated hyperinsulinaemic exercise. METHODS: Diabetic rats performed daily exercise for 1 week and were then exposed to saline (154 mmol/l NaCl) or PRL-2903, 10 mg/kg, before hyperinsulinaemic exercise on two separate occasions spaced 1 day apart. In the following week, animals crossed over to the alternate treatment for a third hyperinsulinaemic exercise protocol. RESULTS: Liver glycogen content was lower in diabetic rats compared with control rats, despite daily insulin therapy (p < 0.05). Glucagon levels failed to increase during exercise with saline but increased three-to-six fold with PRL-2903 (all p < 0.05). Glucose concentrations tended to be higher during exercise and early recovery with PRL-2903 on both days of treatment; this difference did not achieve statistical significance (p > 0.05). CONCLUSIONS/INTERPRETATION: PRL-2903 improves glucagon counterregulation during exercise. However, liver glycogen stores or other factors limit the prevention of exercise-induced hypoglycaemia in rats with streptozotocin-induced diabetes.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Glucagon/metabolism , Hypoglycemia/drug therapy , Hypoglycemia/etiology , Physical Conditioning, Animal/physiology , Receptors, Somatostatin/antagonists & inhibitors , Animals , Blood Glucose/drug effects , Diabetes Mellitus, Experimental/blood , Disease Models, Animal , Glucose/metabolism , Insulin/therapeutic use , Liver/drug effects , Liver/metabolism , Male , Peptides, Cyclic/therapeutic use , Rats , Rats, Sprague-Dawley
15.
Pharm Res ; 33(8): 1850-61, 2016 08.
Article in English | MEDLINE | ID: mdl-27091034

ABSTRACT

PURPOSE: Recently, efficacy studies in mice have shown that amine-terminated cationic (CNP) nanoparticulate carriers of DTX offer an improved formulation of the drug for intravesical delivery. It is hypothesized that this improved efficacy may arise from a carrier mediated bladder exfoliation process that removes the urothelial barrier allowing for increased drug uptake into bladder tissue. The objective of this study was to investigate exfoliation processes in fresh pig's bladders (ex vivo) exposed to three cationic polyglycerols with increasing degrees of amination (denoted 350, 580 and 780). The study also compared the tissue depth profile of DTX uptake into these tissues using these different carriers. MATERIALS AND METHODS: Aminated polyglycerols were synthesized and characterized in the laboratory with low (CNP-360), medium (CNP-580) and high (CNP-780) levels of amine content. CNP-based DTX solutions and commercial DTX solutions in polysorbate 80 (Taxotere®) were doped with (3)H-radiolabeled DTX and prepared by solvent evaporation from acetonitrile, followed by drying and reconstitution in pH 6.4 buffer. Sections of fresh pig's bladder tissue were clamped into Franz diffusion cells and the urothelial side was exposed to the DTX solutions for 2 h. Tissue sections were then frozen for sectioning by cryotome sectioning and subsequently processed for drug analysis by liquid scintillation counting. Alternatively tissue sections were fixed in 2% glutaraldehyde and 2% paraformaldehyde in 0.1 M sodium cacodylate buffer for the purposes of scanning electron microscopy (SEM). RESULTS: Exposure of the urothelial surface to the amine-terminated polyglycerol solutions resulted in the exfoliation of bladder tissues in a time- and concentration-dependent manner. Exfoliation was significantly more pronounced when using CNPs with a medium or high levels of amination whereas only minor levels of exfoliation were seen with low levels. Following incubation of tissues in Tween-based commercial formulations (Taxotere) of DTX (0.5 mg/mL) the drug was detectable at low levels (10-40 µg/g tissue) in all depths of tissue. Similar drug uptake was observed using the CNP-360 formulation. However drug uptake levels were increased to 60-100 µg/g tissue when samples were incubated with either the CNP-580 or CNP-780 formulations. CONCLUSION: The use of cationic polyglycerols with higher levels of amine termination allows for an enhanced uptake of DTX into bladder tissues as compared to commercial (Taxotere) formulations. These increased drug levels probably arise from exfoliation processes resulting in a temporary elimination of the urothelial permeability barrier and increased drug penetration into the tissue.


Subject(s)
Adhesives/metabolism , Nanoparticles/metabolism , Taxoids/metabolism , Urinary Bladder/metabolism , Adhesives/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Cations/administration & dosage , Cations/metabolism , Docetaxel , Drug Compounding , Nanoparticles/administration & dosage , Organ Culture Techniques , Permeability/drug effects , Swine , Taxoids/administration & dosage , Tissue Distribution/drug effects , Tissue Distribution/physiology , Urinary Bladder/drug effects
16.
Virus Res ; 210: 126-32, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26247418

ABSTRACT

Neuraminidase inhibitors (NAIs), including the most frequently prescribed oral therapeutic oseltamivir, play a critical role in the control of severe influenza virus (IFV) infections. However, recent reports of spread of an oseltamivir-resistant H1N1 pandemic strain in individuals who have never been exposed to oseltamivir highlight an urgent need for new antivirals against NAI-resistant IFVs. Difluorosialic acids (DFSAs) are a novel class of anti-IFV NAIs designed based on the mechanism of action of IFV NA, and distinguished by their covalent inhibition mode and their high structural similarity to the natural substrate, sialic acid. These characteristics should render the development of resistance a less rapid process. In this report, we evaluated the relative propensity of influenza A virus (IFV-A) NA to develop resistance against the DFSA class of inhibitor by passaging IFV-A strains in vitro in the presence of either oseltamivir or a representative DFSA (FeqGuDFSA). All the passage-selected lines gained mutations in hemagglutinin. Among the 12 oseltamivir-resistant passaged lines, five gained NA mutations and four of these were the well-defined H275Y mutation that causes oseltamivir resistance. In contrast, out of 15 DFSA-passaged lines, only 2 lines gained NA mutations. Further, NA inhibition assays indicated that these mutations did not change the sensitivity of NA to DFSA and thus the resistance to DFSA was not conferred by these NA mutations. These results strongly suggest that, compared to oseltamivir, IFV is less prone to development of resistance against DFSAs through NA mutations.


Subject(s)
Drug Resistance, Viral , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Mutation , Neuraminidase/antagonists & inhibitors , Oseltamivir/pharmacology , Sialic Acids/pharmacology , Animals , Dogs , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H3N2 Subtype/genetics , Madin Darby Canine Kidney Cells , Models, Molecular , Molecular Structure , Selection, Genetic , Serial Passage , Virus Cultivation
17.
Bioorg Med Chem Lett ; 25(12): 2505-9, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25980910

ABSTRACT

Difluorosialic acids (DFSAs) are potent inhibitors of viral neuraminidase that demonstrate activity against oseltamivir- and zanamivir-resistant strains of influenza. Unfortunately, low oral bioavailability precludes their development as second generation neuraminidase inhibitors for treating influenza as this leaves them unsuitable for use in an oral formulation. Herein is described the preparation of a series of DFSA prodrugs designed to increase oral bioavailability. These prodrugs were evaluated using a snapshot PK screen and stability tests, with successful candidates being further assessed with a full pharmacokinetic workup. These new prodrugs increased oral bioavailability by up to three times that seen for the parent DFSAs.


Subject(s)
Enzyme Inhibitors/chemistry , Neuraminidase/antagonists & inhibitors , Prodrugs/chemistry , Sialic Acids/chemistry , Viral Proteins/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Half-Life , Mice , Neuraminidase/metabolism , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Sialic Acids/chemical synthesis , Sialic Acids/pharmacokinetics , Viral Proteins/metabolism
18.
Science ; 340(6128): 71-5, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23429702

ABSTRACT

Influenza antiviral agents play important roles in modulating disease severity and in controlling pandemics while vaccines are prepared, but the development of resistance to agents like the commonly used neuraminidase inhibitor oseltamivir may limit their future utility. We report here on a new class of specific, mechanism-based anti-influenza drugs that function through the formation of a stabilized covalent intermediate in the influenza neuraminidase enzyme, and we confirm this mode of action with structural and mechanistic studies. These compounds function in cell-based assays and in animal models, with efficacies comparable to that of the neuraminidase inhibitor zanamivir and with broad-spectrum activity against drug-resistant strains in vitro. The similarity of their structure to that of the natural substrate and their mechanism-based design make these attractive antiviral candidates.


Subject(s)
Antiviral Agents/chemistry , Enzyme Inhibitors/chemistry , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae/drug effects , Sialic Acids/chemistry , Animals , Antiviral Agents/pharmacology , Crystallography, X-Ray , Dogs , Enzyme Inhibitors/pharmacology , Humans , Madin Darby Canine Kidney Cells , Neuraminidase/chemistry , Orthomyxoviridae/enzymology , Oseltamivir/chemistry , Oseltamivir/pharmacology , Protein Conformation , Sialic Acids/pharmacology , Structure-Activity Relationship , Zanamivir/chemistry , Zanamivir/pharmacology
19.
Biomaterials ; 33(2): 692-703, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22014457

ABSTRACT

Recently, we have reported that docetaxel (DTX) loaded, amine terminated hyperbranched polyglycerol (HPG-C(8/10)-MePEG-NH(2)) nanoparticles significantly increased drug uptake in mouse bladder tissues and was the most effective formulation to significantly inhibit tumor growth in an orthotopic model of bladder cancer. The objective of this study was to investigate the effects of HPG-C(8/10)-MePEG-NH(2) nanoparticles on bladder urothelial morphology and integrity, DTX uptake and permeability in bladder tissue and the extent of bladder urothelial recovery following exposure to, and then washout of, HPG-C(8/10)-MePEG-NH(2) nanoparticles. HPG-C(8/10)-MePEG-NH(2) nanoparticles significantly increased the uptake of DTX in both isolated pig bladder as well as in live mouse bladder tissues. Furthermore, HPG-C(8/10)-MePEG-NH(2) nanoparticles were demonstrated to increase the permeability of the urinary bladder wall by causing changes to the urothelial barrier function and morphology through opening of tight junctions and exfoliation of the superficial umbrella cells. These data suggest that exfoliation may be triggered by an apoptosis mechanism, which was followed by a rapid recovery of the urothelium within 24 h post-instillation of HPG-C(8/10)-MePEG-NH(2) nanoparticles. HPG-C(8/10)-MePEG-NH(2) nanoparticles cause significant but rapidly recoverable changes in the bladder urothelial morphology, which we believe may make them suitable for increasing drug permeability of bladder tissue and intravesical drug delivery.


Subject(s)
Glycerol/chemistry , Polymers/chemistry , Taxoids/pharmacokinetics , Urinary Bladder/drug effects , Urothelium/drug effects , Animals , Chemistry, Pharmaceutical , Disease Models, Animal , Docetaxel , Drug Delivery Systems , Female , Hydrophobic and Hydrophilic Interactions , Male , Mice , Mice, Nude , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Nanoparticles/chemistry , Swine , Urinary Bladder/pathology , Urinary Bladder Neoplasms/drug therapy , Urothelium/pathology
20.
Clin Cancer Res ; 17(9): 2788-98, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21357680

ABSTRACT

PURPOSE: The present work describes the development and in vitro and in vivo evaluation of a mucoadhesive nanoparticulate docetaxel (DTX) formulation for intravesical bladder cancer therapy. EXPERIMENTAL DESIGN: Mucoadhesive formulations based on hyperbranched polyglycerols (HPG), hydrophobically derivatized with C(8)/C(10) alkyl chains in the core and modified with methoxy-polyethylene glycol (MePEG) and amine groups in the shell (HPG-C(8/10)-MePEG-NH(2)) were synthesized and DTX was loaded into these by a solvent evaporation method. Both low-grade (RT4, MGHU3) and high-grade (UMUC3) human urothelial carcinoma cell lines were treated with various concentrations of DTX formulations in vitro. KU7 cells that stably express firefly luciferase (KU7-luc) were inoculated in female nude mice by intravesical instillation and quantified using bioluminescence imaging. Mice with established KU7-luc tumors were given a single intravesical instillation with PBS, Taxotere (DTX from Sanofi-aventis), and DTX-loaded HPG-C(8/10)-MePEG and/or HPG-C(8/10)-MePEG-NH(2). Drug uptake was conducted using LC/MS-MS (liquid chromatography/tandem mass spectrometry) and tumor microenvironment and uptake of rhodamine labeled HPGs was assessed. RESULTS: In vitro, all DTX formulations potently inhibited bladder cancer proliferation. However, in vivo, DTX-loaded HPG-C(8/10)-MePEG-NH(2) (mucoadhesive DTX) was the most effective formulation to inhibit tumor growth in an orthotopic model of bladder cancer. Furthermore, mucoadhesive DTX significantly increased drug uptake in mouse bladder tissues. In addition, rhodamine labeled HPG-C(8/10)-MePEG-NH(2) showed enhanced uptake of these nanoparticles in bladder tumor tissues. CONCLUSIONS: Our data show promising in vivo antitumor efficacy and provide preclinical proof of principle for the intravesical application of mucoadhesive nanoparticulate DTX formulation in the treatment of bladder cancer.


Subject(s)
Carcinoma/drug therapy , Mucous Membrane/metabolism , Taxoids/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Adhesiveness , Administration, Intravesical , Animals , Antineoplastic Agents/administration & dosage , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Docetaxel , Female , Humans , Mice , Mice, Nude , Mucous Membrane/drug effects , Mucous Membrane/pathology , Muscle, Smooth/pathology , Nanoparticles/administration & dosage , Neoplasm Invasiveness , Treatment Outcome , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...