Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
1.
J Clin Pharmacol ; 63(5): 593-603, 2023 05.
Article in English | MEDLINE | ID: mdl-36579617

ABSTRACT

This study aimed to develop a population pharmacokinetic (PK) model of ambrisentan in pediatric patients (8 to <18 years) with pulmonary arterial hypertension (PAH) and compare pediatric ambrisentan systemic exposure with previously reported adult data. Association of ambrisentan exposure with efficacy (6-minute walking distance) and safety (adverse events) were exploratory analyses. A population PK model was developed using pediatric PK data. Steady-state systemic exposure metrics were estimated for the pediatric population and compared with previously reported data in adult patients with PAH and healthy subjects. No covariates had a significant effect on PK parameters; therefore, the final covariate model was the same as the base model. The pediatric population PK model was a 2-compartment model including the effect of body weight (allometric scaling), first-order absorption and elimination, and absorption lag time. Steady-state ambrisentan exposure was similar between the pediatric and adult population when accounting for body weight differences. Geometric mean area under the concentration-time curve at steady state in pediatric patients receiving ambrisentan low dose was 3% lower than in the adult population (and similar in both populations receiving high dose). Geometric mean maximum plasma concentration at steady state in pediatric patients receiving low and high doses was 11% and 18% higher, respectively, than in the adult population. There was no apparent association in the pediatric or adult population between ambrisentan exposure and change in 6-minute walking distance or incidence of ambrisentan-related adverse events in pediatric patients. The similar ambrisentan exposure and exposure-response profiles observed in pediatric and adult populations with PAH suggests appropriateness of body-weight-based dosing in the pediatric population with PAH.


Subject(s)
Phenylpropionates , Pulmonary Arterial Hypertension , Pyridazines , Humans , Adult , Child , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/chemically induced , Antihypertensive Agents , Familial Primary Pulmonary Hypertension , Phenylpropionates/adverse effects , Phenylpropionates/pharmacokinetics , Pyridazines/adverse effects , Pyridazines/pharmacokinetics
2.
Open Forum Infect Dis ; 9(11): ofac550, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36420058

ABSTRACT

A 7-day course of glecaprevir/pibrentasvir started in the preoperative period prevented transmission of hepatitis C virus (HCV) from viremic donors to 10 HCV-negative recipients (2 heart, 1 lung, 6 kidney, 1 heart/kidney) with 100% sustained virological response at 12 weeks.

4.
J Pharm Pract ; 33(6): 912-914, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31248333

ABSTRACT

Solid organ transplant recipients have increased cancer risk due in part to chronic immunosuppression and opportunistic oncogenic viral infections. The management of drug interactions in transplant recipients being treated for cancer is important both to minimize the likelihood of drug-related toxicities and to optimize therapeutic outcomes. We present a case of a 41-year-old woman with a stable living-related kidney transplant maintained on an immunosuppressive regimen of cyclosporine, mycophenolate mofetil, and prednisone, who was subsequently diagnosed with a metastatic lobular breast carcinoma and papillary thyroid cancer and started palbociclib, a time-dependent CYP3A inhibitor. After initiation of palbociclib, cyclosporine trough and peak concentrations were increased by 159% and 81%, respectively, relative to the average cyclosporine concentrations pre-palbociclib. Using the Drug Interaction Probability Scale (DIPS), the interaction between palbociclib and cyclosporine was rated as "probable." Dose reductions of immunosuppressive agents that are CYP3A substrates are warranted if palbociclib is initiated, followed by close monitoring of blood concentrations. This report also highlights the challenges of coadministering a time-dependent inhibitor with a narrow therapeutic index drug that is metabolized by the same enzyme, particularly when the inhibitor is given in cycles with off-treatment periods.


Subject(s)
Kidney Transplantation , Adult , Cyclosporine/adverse effects , Female , Humans , Immunosuppressive Agents/adverse effects , Mycophenolic Acid , Piperazines/adverse effects , Pyridines/adverse effects
5.
Ann R Coll Surg Engl ; 101(2): e45-e47, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30354179

ABSTRACT

The incidence of biliary lithiasis after gastric surgery seems to be higher than in the general population. Endoscopic retrograde cholangiopancreatography (ERCP) allows several biliary and pancreatic pathologies to be dealt with; however, in patients with an altered anatomy of the upper and mid gastrointestinal tract, this endoscopic manoeuvre can be extremely challenging. We report a case of a 79-year-old woman with previous subtotal gastrectomy and Roux-en-Y reconstruction, admitted with a diagnosis of cholecystitis and choledocolithiasis. She was successfully treated with transjejunal laparoscopic-assisted ERCP and laparoscopic cholecystectomy, which appears to be a safe and useful procedure for choledocolithiasis treatment in patients with surgically altered anatomy.


Subject(s)
Cholangiopancreatography, Endoscopic Retrograde/methods , Cholecystectomy, Laparoscopic/methods , Cholecystitis/surgery , Choledocholithiasis/surgery , Gastrectomy , Jejunum/surgery , Postoperative Complications/surgery , Aged , Anastomosis, Roux-en-Y , Cholecystitis/etiology , Choledocholithiasis/etiology , Female , Humans
6.
Case Rep Transplant ; 2018: 2695178, 2018.
Article in English | MEDLINE | ID: mdl-29955411

ABSTRACT

Acute kidney injury is common among kidney transplant recipients. Postinfectious glomerulonephritis secondary to nephritogenic streptococci is one of the oldest known etiologies of acute kidney injury in native kidneys but rarely reported among kidney transplant recipients. This report is of a biopsy-proven case of acute kidney injury in a renal allograft recipient caused by de novo poststreptococcal glomerulonephritis.

7.
Pharmacol Res Perspect ; 4(5): e00251, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27713822

ABSTRACT

Safinamide is an orally administered α-aminoamide derivative with both dopaminergic and non-dopaminergic properties. Nonlinear mixed effects models for population pharmacokinetic (PK) and pharmacokinetic-pharmacodynamic (PKPD) analyses were developed using records from, respectively, 623 and 668 patients belonging to two Phase 3, randomized, placebo-controlled, double-blind efficacy studies. The aim was to estimate safinamide population PK parameters in patients with Parkinson's disease (PD) on stable levodopa therapy, and to develop a model of safinamide effect on the PD phase of normal functioning (ON-time). The final models were internally evaluated using visual predictive checks (VPCs), prediction corrected-VPC, and nonparametric bootstrap analysis. Safinamide profiles were adequately described by a linear one-compartmental model with first-order absorption and elimination. CL/F, Vd/F, and KA (95% confidence interval [CI]) were 4.96 (4.73-5.21) L/h, 166 (158-174) L, and 0.582 (0.335-0.829) h-1, respectively. CL/F and Vd/F increased with body weight, while age, gender, renal function, and exposure to levodopa did not influence safinamide PK. The observed ON-time values were adequately described by a linear model, with time in the study period as dependent variable, and rate of ON-time change and baseline plus offset effect as slope and intercept parameters. Safinamide treatment resulted in an increase in ON-time of 0.73 h (week 4), with further ON-time increase with the same slope as placebo. The increase was not influenced by age, levodopa, or safinamide exposure. The population models adequately describe the population PK of safinamide and safinamide effect on ON-time. No dose adjustments in elderly and mild to moderate renally impaired patients are requested.

8.
Case Rep Transplant ; 2014: 548243, 2014.
Article in English | MEDLINE | ID: mdl-24575309

ABSTRACT

Purpose. The case of a kidney transplant recipient who experienced a probable drug interaction between sirolimus and ranolazine is reported. Summary. The narrow therapeutic window of immunosuppressive therapy in transplant recipients requires close monitoring for potential drug-drug interactions. The patient, a 57-year-old Caucasian male kidney transplant recipient, was stable for years on sirolimus as his primary immunosuppressive agent and had a history of chronic angina, for which he was prescribed ranolazine. Upon addition and dose escalation of ranolazine, whole blood sirolimus levels more than tripled, rising to immeasurably high concentrations. After holding sirolimus on multiple occasions and reducing dosage more than 50%, blood levels returned to therapeutic range, while continuing ranolazine. Conclusion. Since ranolazine is a documented P-GP and CYP3A inhibitor, and sirolimus a known substrate for both pathways, it is proposed that ranolazine inhibition of P-GP and CYP3A4 contributed to the significant elevation in sirolimus exposure. No alternative causes for the rise in sirolimus exposure were found, and assessment with the Drug Interaction Probability Scale finds this interaction to be probable. Clinicians should be aware of the potential for this interaction to cause elevated sirolimus exposure and subsequent increase in clinical effect or toxicity, in this case overimmunosuppression.

9.
Target Oncol ; 9(3): 215-24, 2014 Sep.
Article in English | MEDLINE | ID: mdl-23832397

ABSTRACT

Aurora kinase inhibitors (AKIs) are a class of antimitotic, small-molecule anticancer agents. MSC1992371A is an AKI being evaluated for the treatment of patients with solid tumors. This phase I, open-label, dose-escalation study determined the maximum tolerated dose (MTD) of MSC1992371A in different dosing schedules in patients with locally advanced or metastatic solid tumors. MSC1992371A was administered on days 1 and 8 (schedule 1) or on days 1, 2, and 3 (schedule 2) of a 21-day cycle. The study was expanded with a third schedule (study drug on days 1-3 and 8-10). Adverse events were monitored throughout the study. Antitumor efficacy, drug pharmacokinetics, and pharmacodynamics were evaluated. Ninety-two patients were enrolled. MSC1992371A was dosed over eight levels in schedules 1 and 2, and the MTD was determined as 74 mg/m(2) per cycle for both schedules and as 60 mg/m(2) in schedule 3, albeit only in three patients due to discontinuation of the study. Overall, the most common grade 3 or 4 treatment-emergent adverse events were neutropenia, febrile neutropenia, thrombocytopenia, anemia, and fatigue. The most frequent dose-limiting toxicity over all schedules was neutropenia. MSC1992371A plasma concentrations tended to increase with increasing dose levels. Although no complete or partial responses were seen, stable disease ≥3 months was observed in 11 patients. Analysis for markers of target modulation and pharmacodynamics effects was unsuccessful. MSC1992371A was generally well tolerated in patients, with mainly transient hematologic toxicities apparent at an MTD of 60-74 mg/m(2)/21-day cycle, independent of dosing frequency.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Neoplasms/drug therapy , Norbornanes/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Administration, Oral , Adult , Aged , Aged, 80 and over , Cohort Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Food-Drug Interactions , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Norbornanes/adverse effects , Norbornanes/pharmacokinetics , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Treatment Outcome , Young Adult
10.
Clin Oncol (R Coll Radiol) ; 25(6): 336-42, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23615181

ABSTRACT

AIMS: This phase I dose-escalation study was designed to evaluate the combination of the mammalian target of rapamycin inhibitor ridaforolimus with the vascular endothelial growth factor inhibitor bevacizumab. MATERIALS AND METHODS: Seventeen adult patients with refractory advanced solid tumours received oral ridaforolimus (30 or 40 mg) once daily for 5 days per week (QDx5/wk) combined with intravenous bevacizumab (10 mg/kg every 2 weeks [Q2wk] or 15 mg/kg every 3 weeks [Q3wk]). Patients were evaluated for dose-limiting toxicities, safety and anti-tumour activity. RESULTS: A 40 mg dose of ridaforolimus with either bevacizumab dosing schedule was the recommended phase II dose. No dose-limiting toxicities were reported; the most common drug-related adverse events were mucosal inflammation and anorexia. Seven patients, with clinical features that included primary tumour of the abdominal origin (colorectal, pancreatic or gynaecological cancers) and previous abdominal radiotherapy, reported serious adverse events related to bowel perforations. There were no objective responses, but 65% of patients had a best response of stable disease. CONCLUSION: Oral ridaforolimus (40 mg QDx5/wk) is feasible to combine with standard doses of bevacizumab, although careful patient selection would be needed to mitigate the risk of bowel perforation-related adverse events. Combination therapy produced prolonged stable disease in several heavily pretreated patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Adult , Aged , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Bevacizumab , Cohort Studies , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Neoplasms/pathology , Sirolimus/administration & dosage , Sirolimus/adverse effects , Sirolimus/analogs & derivatives , Treatment Outcome , Young Adult
11.
Ann Oncol ; 24(4): 1104-11, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23211938

ABSTRACT

BACKGROUND: Ridaforolimus is an inhibitor of mTOR with evidence of antitumor activity in an I.V. formulation. This multicenter, open-label, 3 + 3 design nonrandomized, dose-escalation, phase I/IIa trial was conducted to determine the safety, pharmacokinetic (PK) and pharmacodynamic parameters, maximum tolerated dose, and antitumor activity of oral ridaforolimus. PATIENTS AND METHODS: Patients with metastatic or unresectable solid tumors refractory to therapy were eligible. Seven different continuous and intermittent dosing regimens were examined. RESULTS: One hundred and forty-seven patients were enrolled in this study among which 85 were patients with sarcoma. Stomatitis was the most common DLT observed. The dosing regimen, 40 mg QD × 5 days/week, provided the best combination of cumulative dose, dose density, and cumulative exposure, and was the recommended dosing regimen for subsequent clinical development. PK was nonlinear, with less than proportional increases in day-1 blood AUC0-∞ and Cmax, particularly with doses >40 mg. The terminal half-life estimate of ridaforolimus (QD × 5 40 mg) was 42.0 h, and the mean half-life ∼30-60 h. The clinical benefit rate, (complete response, partial response, or stable disease for ≥4 months was 24.5% for all patients and 27.1% for patients with sarcoma. CONCLUSION: Oral ridaforolimus had an acceptable safety profile and exhibited antitumor activity in patients with sarcoma and other malignancies. ClinicalTrials.gov Identifier NCT00112372.


Subject(s)
Neoplasms/drug therapy , Sarcoma/drug therapy , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/metabolism , Adult , Aged , Aged, 80 and over , Animals , Drug Administration Schedule , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Neoplasms/pathology , Sarcoma/pathology , Sirolimus/administration & dosage , Sirolimus/adverse effects , Sirolimus/antagonists & inhibitors , Sirolimus/pharmacokinetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , Treatment Outcome
12.
Ann Oncol ; 23(9): 2399-2408, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22357447

ABSTRACT

BACKGROUND: This phase I dose-escalation study investigated the maximum tolerated dose (MTD), safety, pharmacokinetics, pharmacodynamics (PDs), and preliminary antitumor activity of BGT226, a potent, oral dual phosphatidylinositol-3-kinase (PI3K)/mammalian target of rapamycin inhibitor. PATIENTS AND METHODS: Fifty-seven patients with advanced solid tumors received BGT226 2.5-125 mg/day three times weekly (TIW). Dose escalation was guided by an adaptive Bayesian logistic regression model with overdose control. Assessments included response per RECIST, [18F]-fluorodeoxyglucose uptake, and phosphorylated-S6 in skin and paired tumor samples. RESULTS: Three patients (125 mg cohort) had dose-limiting toxic effects (grade 3 nausea/vomiting, diarrhea). BGT226-related adverse events included nausea (68%), diarrhea (61%), vomiting (49%), and fatigue (19%). BGT226 demonstrated rapid absorption, variable systemic exposure, and a median half-life of 6-9 h. Seventeen patients (30%) had stable disease (SD) as best response. Nine patients had SD for ≥16 weeks. Thirty patients (53%) achieved stable metabolic disease as assessed by [18F]-fluorodeoxyglucose-positron emission tomography; however, no correlation between metabolic response and tumor shrinkage according to computed tomography was observed. PD changes suggested PI3K pathway inhibition but were inconsistent. CONCLUSIONS: The MTD of BGT226 was 125 mg/day TIW, and the clinically recommended dose was 100 mg/day TIW. Limited preliminary antitumor activity and inconsistent target inhibition were observed, potentially due to low systemic exposure.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Colonic Neoplasms/drug therapy , Imidazoles/therapeutic use , Prostatic Neoplasms/drug therapy , Quinolines/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/diagnostic imaging , Colonic Neoplasms/diagnostic imaging , Diarrhea/chemically induced , Female , Fluorodeoxyglucose F18 , Humans , Imidazoles/adverse effects , Imidazoles/pharmacokinetics , Male , Maximum Tolerated Dose , Middle Aged , Nausea/chemically induced , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms/diagnostic imaging , Quinolines/adverse effects , Quinolines/pharmacokinetics , Radionuclide Imaging , Radiopharmaceuticals , TOR Serine-Threonine Kinases/antagonists & inhibitors , Treatment Outcome , Young Adult
13.
Br J Cancer ; 105(7): 938-44, 2011 Sep 27.
Article in English | MEDLINE | ID: mdl-21878940

ABSTRACT

BACKGROUND: To evaluate the anticancer activity of erlotinib in patients with previously treated, advanced non-small cell lung cancer (NSCLC) whose dose is increased to that associated with a maximal level of tolerable skin toxicity (i.e., target rash (TR)); to characterise the pharmacokinetics (PK) and pharmacodynamics (PD) of higher doses of erlotinib. METHODS: Patients initially received erlotinib 150 mg per day. The dose was successively increased in each patient to that associated with a TR. Anticancer activity was evaluated. Plasma, skin, and hair were sampled for PK and PD studies. RESULTS: Erlotinib dose escalation to 200-475 mg per day was feasible in 38 (90%) of 42 patients. Twenty-four (57%) patients developed a TR, but 19 (79%) did so at 150 mg per day. Five (12%) patients, all of whom developed a TR, had a partial response. Median progression-free survival (PFS) was 2.3 months (95% CI: 1.61, 4.14); median PFS was 3.5 months and 1.9 months, respectively, for patients who did and did not experience a TR (hazard ratio, 0.51; P=0.051). Neither rash severity nor response correlated with erlotinib exposure. CONCLUSION: Intrapatient dose escalation of erlotinib does not appreciably increase the propensity to experience a maximal level of tolerable skin toxicity, or appear to increase the anticancer activity of erlotinib in NSCLC.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Squamous Cell/drug therapy , Exanthema/chemically induced , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Adenocarcinoma/pathology , Adult , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/pathology , Erlotinib Hydrochloride , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Prognosis , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/pharmacokinetics , Tissue Distribution
14.
Dev Biol ; 356(1): 19-27, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21600196

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) are found in the basement membrane and at the cell-surface where they modulate the binding and activity of a variety of growth factors and other molecules. Most of the functions of HSPGs are mediated by the variable sulfated glycosaminoglycan (GAG) chains attached to a core protein. Sulfation of the GAG chain is key as evidenced by the renal agenesis phenotype in mice deficient in the HS biosynthetic enzyme, heparan sulfate 2-O sulfotransferase (Hs2st; an enzyme which catalyzes the 2-O-sulfation of uronic acids in heparan sulfate). We have recently demonstrated that this phenotype is likely due to a defect in induction of the metanephric mesenchyme (MM), which along with the ureteric bud (UB), is responsible for the mutually inductive interactions in the developing kidney (Shah et al., 2010). Here, we sought to elucidate the role of variable HS sulfation in UB branching morphogenesis, particularly the role of 6-O sulfation. Endogenous HS was localized along the length of the UB suggesting a role in limiting growth factors and other molecules to specific regions of the UB. Treatment of cultures of whole embryonic kidney with variably desulfated heparin compounds indicated a requirement of 6O-sulfation in the growth and branching of the UB. In support of this notion, branching morphogenesis of the isolated UB was found to be more sensitive to the HS 6-O sulfation modification when compared to the 2-O sulfation modification. In addition, a variety of known UB branching morphogens (i.e., pleiotrophin, heregulin, FGF1 and GDNF) were found to have a higher affinity for 6-O sulfated heparin providing additional support for the notion that this HS modification is important for robust UB branching morphogenesis. Taken together with earlier studies, these findings suggest a general mechanism for spatio-temporal HS regulation of growth factor activity along the branching UB and in the developing MM and support the view that specific growth factor-HSPG interactions establish morphogen gradients and function as developmental switches during the stages of epithelial organogenesis (Shah et al., 2004).


Subject(s)
Growth Substances/physiology , Heparitin Sulfate/physiology , Kidney/embryology , Morphogenesis/physiology , Ureter/embryology , Animals , Carrier Proteins/pharmacology , Carrier Proteins/physiology , Cells, Cultured , Cytokines/pharmacology , Cytokines/physiology , Fibroblast Growth Factor 1/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Growth Substances/pharmacology , Heparitin Sulfate/pharmacology , Kidney/metabolism , Morphogenesis/drug effects , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Ureter/metabolism
16.
Dev Biol ; 347(2): 337-47, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20816800

ABSTRACT

Embryonic kidney development begins with the outgrowth of the ureteric bud (UB) from the Wolffian duct (WD) into the adjacent metanephric mesenchyme (MM). Both a GDNF-dependent and GDNF-independent (Maeshima et al., 2007) pathway have been identified. In vivo and in vitro, the GDNF-dependent pathway is inhibited by BMPs, one of the factors invoked to explain the limitation of UB formation in the unbudded regions of the WD surrounding the UB. However, the exact mechanism remains unknown. Here a previously described in vitro system that models UB budding from the WD was utilized to study this process. Because Protein kinase A (PKA) activation has been shown to prevent migration, morphogenesis and tubulogenesis of epithelial cells (Santos et al., 1993), its activity in budded and non-budded portions of the GDNF-induced WD was analyzed. The level of PKA activity was 15-fold higher in the unbudded portions of the WD compared to budded portions, suggesting that PKA activity plays a key role in controlling the site of UB emergence. Using well-characterized PKA agonists and antagonists, we demonstrated that at various levels of the PKA-signaling hierarchy, PKA regulates UB outgrowth from the WD by suppressing budding events. This process appeared to be PKA-2 isoform specific, and mediated by changes in the duct rather than the surrounding mesenchyme. In addition, it was not due to changes in either the sorting of junctional proteins, cell death, or cell proliferation. Furthermore, the suppressive effect of cAMP on budding did not appear to be mediated by spread to adjacent cells via gap junctions. Conversely, antagonism of PKA activity stimulated UB outgrowth from the WD and resulted in both an increase in the number of buds per unit length of WD as well as a larger surface area per bud. Using microarrays, analysis of gene expression in GDNF-treated WDs in which the PKA pathway had been activated revealed a nearly 14-fold decrease in Ret, a receptor for GDNF. A smaller decrease in GFRα1. a co-receptor for GDNF, was also observed. Using Ret-null WDs, we were able to demonstrate that PKA regulated GDNF-dependent budding but not GDNF-independent pathway for WD budding. We also found that BMP2 was higher in unbudded regions of the GDNF-stimulated WD. Treatment of isolated WDs with BMP2 suppressed budding and resulted in a 3-fold increase in PKA activity. The data suggests that the suppression of budding by BMPs and possibly other factors in non-budded zones of the WD may be regulated in part by increased PKA activity, probably partially through downregulation of Ret/GFRα1 coreceptor expression.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Kidney/embryology , Proto-Oncogene Proteins c-ret/metabolism , Ureter/embryology , Wolffian Ducts/embryology , Animals , Base Sequence , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/pharmacology , Cell Proliferation , DNA Primers/genetics , Embryonic Development/drug effects , Embryonic Development/genetics , Embryonic Development/physiology , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Male , Mesoderm/embryology , Mice , Mice, Knockout , Models, Biological , Pregnancy , Proto-Oncogene Proteins c-ret/deficiency , Proto-Oncogene Proteins c-ret/genetics , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Signal Transduction
18.
Dev Biol ; 339(2): 354-65, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20059993

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) are central modulators of developmental processes likely through their interaction with growth factors, such as GDNF, members of the FGF and TGFbeta superfamilies, EGF receptor ligands and HGF. Absence of the biosynthetic enzyme, heparan sulfate 2-O-sulfotransferase (Hs2st) leads to kidney agenesis. Using a novel combination of in vivo and in vitro approaches, we have reanalyzed the defect in morphogenesis of the Hs2st(-)(/)(-) kidney. Utilizing assays that separately model distinct stages of kidney branching morphogenesis, we found that the Hs2st(-/-) UB is able to undergo branching and induce mesenchymal-to-epithelial transformation when recombined with control MM, and the isolated Hs2st null UB is able to undergo branching morphogenesis in the presence of exogenous soluble pro-branching growth factors when embedded in an extracellular matrix, indicating that the UB is intrinsically competent. This is in contrast to the prevailing view that the defect underlying the renal agenesis phenotype is due to a primary role for 2-O sulfated HS in UB branching. Unexpectedly, the mutant MM was also fully capable of being induced in recombination experiments with wild-type tissue. Thus, both the mutant UB and mutant MM tissue appear competent in and of themselves, but the combination of mutant tissues fails in vivo and, as we show, in organ culture. We hypothesized a 2OS-dependent defect in the mutual inductive process, which could be on either the UB or MM side, since both progenitor tissues express Hs2st. In light of these observations, we specifically examined the role of the HS 2-O sulfation modification on the morphogenetic capacity of the UB and MM individually. We demonstrate that early UB branching morphogenesis is not primarily modulated by factors that depend on the HS 2-O sulfate modification; however, factors that contribute to MM induction are markedly sensitive to the 2-O sulfation modification. These data suggest that key defect in Hs2st null kidneys is the inability of MM to undergo induction either through a failure of mutual induction or a primary failure of MM morphogenesis. This results in normal UB formation but affects either T-shaped UB formation or iterative branching of the T-shaped UB (possibly two separate stages in collecting system development dependent upon HS). We discuss the possibility that a disruption in the interaction between HS and Wnts (e.g. Wnt 9b) may be an important aspect of the observed phenotype. This appears to be the first example of a defect in the MM preventing advancement of early UB branching past the first bifurcation stage, one of the limiting steps in early kidney development.


Subject(s)
Cell Differentiation , Kidney/metabolism , Mesoderm/cytology , Sulfotransferases/genetics , Ureter/cytology , Animals , Cells, Cultured , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Kidney/cytology , Kidney/embryology , Mesoderm/embryology , Mesoderm/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Morphogenesis , Sulfotransferases/metabolism , Ureter/embryology , Ureter/metabolism
19.
Ann Oncol ; 21(2): 376-381, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19633048

ABSTRACT

BACKGROUND: Lexatumumab (HGS-ETR2) is a fully human agonistic mAb to the tumor necrosis factor-related apoptosis-inducing ligand receptor 2 that activates the extrinsic apoptosis pathway and has potent preclinical antitumor activity. MATERIALS AND METHODS: This phase 1, dose escalation study assessed the safety, tolerability, pharmacokinetics (PKs) and immunogenicity of lexatumumab administered i.v. every 14 days in patients with advanced solid tumors. RESULTS: Thirty-one patients received lexatumumab over five dose levels (0.1-10 mg/kg). Most (26 of 31) received four or more cycles of treatment. One patient at 10 mg/kg experienced a possibly related dose-limiting toxicity of grade 3 hyperamylasemia. Nine patients achieved stable disease. One patient with chemotherapy-refractive Hodgkin's disease experienced a mixed response. Lexatumumab PKs were linear up to 10 mg/kg. At the 10 mg/kg dose, the mean (+/-standard deviation) t(1/2b) was 13.67 +/- 4.07 days, clearance was 4.95 +/- 1.93 ml/day/kg, V(1) was 45.55 ml/kg and V(ss) was 79.08 ml/kg, indicating that lexatumumab distributes outside the plasma compartment. No human antihuman antibodies were detected. CONCLUSIONS: Lexatumumab can be safely administered every 14 days at 10 mg/kg. The PK profile supports this schedule. Further evaluation of lexatumumab at this dose schedule is warranted, including combination trials with other agents.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Disease Progression , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Injections, Intravenous , Male , Middle Aged , Neoplasms/metabolism , Neoplasms/pathology , Treatment Outcome
20.
Development ; 136(24): 4213-24, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19934016

ABSTRACT

Ureteric bud (UB) emergence from the Wolffian duct (WD), the initiating step in metanephric kidney morphogenesis, is dependent on GDNF; however, GDNF by itself is generally insufficient to induce robust budding of the isolated WD in culture. Thus, additional factors, presumably peptides or polypeptide growth factors, might be involved. Microarray data from in vivo budding and non-budding conditions were analyzed using non-negative matrix factorization followed by gene ontology filtering and network analysis to identify sets of genes that are highly regulated during budding. These included the GDNF co-receptors GFRalpha1 and RET, as well as neuropeptide Y (NPY). By using ANOVA with pattern matching, NPY was also found to correlate most significantly to the budded condition with a high degree of connectedness to genes with developmental roles. Exogenous NPY [as well as its homolog, peptide YY (PYY)] augmented GDNF-dependent budding in the isolated WD culture; conversely, inhibition of NPY signaling or perturbation of NPY expression inhibited budding, confirming that NPY facilitates this process. NPY was also found to reverse the decreased budding, the downregulation of RET expression, the mislocalization of GFRalpha1, and the inhibition of AKT phosphorylation that resulted from the addition of BMP4 to the isolated WD cultures, suggesting that NPY acts through the budding pathway and is reciprocally regulated by GDNF and BMP4. Thus, the outgrowth of the UB from the WD might result from a combination of the upregulation of the GDNF receptors together with genes that support GDNF signaling in a feed-forward loop and/or counteraction of the inhibitory pathway regulated by BMP4.


Subject(s)
Neuropeptide Y/physiology , Wolffian Ducts/physiology , Animals , Bone Morphogenetic Protein 4/physiology , Cells, Cultured , Female , Gene Expression Regulation, Developmental , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Morphogenesis , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-ret/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction , Up-Regulation , Wolffian Ducts/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...