Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
J Pharmacol Exp Ther ; 382(3): 346-355, 2022 09.
Article in English | MEDLINE | ID: mdl-35840338

ABSTRACT

The benefit of once-weekly basal insulin is less frequent dosing, which has the potential to reduce the barrier to injection therapy and impact patient activation, adherence and compliance, quality of life, and outcomes. Basal Insulin Fc (BIF, LY3209590, or insulin efsitora alfa) is a once-weekly basal insulin in clinical testing for type 1 and type 2 diabetes mellitus. BIF is comprised of a novel single-chain variant of insulin fused to a human IgG2 fragment crystallizable region of an antibody domain using a peptide linker. The in vitro binding affinity of BIF for the human insulin receptor (IR) was two orders of magnitude weaker relative to human insulin. BIF stimulated IR phosphorylation in cells with reduced potency, yet full agonism, and exhibited a significantly faster dephosphorylation kinetic profile than human insulin or AspB10 insulin. BIF stimulated de novo lipogenesis in 3T3-L1 adipocytes and cell proliferation in SAOS-2 and H4IIE cells with ≥70-fold reduction in in vitro potency compared with human insulin. BIF possessed markedly reduced binding to hIGF-1R, making definitive measurements unattainable. In vivo pharmacology studies using streptozotocin-treated diabetic rats demonstrated a significant decrease in blood glucose compared with vehicle-treated animals 24 hours post-injection, persisting through 336 hours following subcutaneous administration. In streptozotocin-treated rats, BIF reached time at maximum concentration at 48 hours and possessed a clearance rate of ∼0.85 ml/h per kg, with a terminal half-life of ∼120 hours following subcutaneous administration. These results demonstrate BIF has an in vitro pharmacological profile similar to native insulin, with significantly reduced potency and an extended time-action profile in vivo that supports once-weekly dosing in humans. SIGNIFICANCE STATEMENT: BIF is a novel basal insulin Fc-fusion protein designed for once-weekly dosing. In this study, we demonstrate that BIF has an in vitro pharmacological profile similar to human insulin, but with weaker potency across assays for IR binding and activity. BIF has a PD and PK profile in STZ-treated rats supportive of weekly dosing in humans.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Humans , Insulin/metabolism , Quality of Life , Rats , Streptozocin
2.
J Pharmacol Exp Ther ; 358(2): 282-93, 2016 08.
Article in English | MEDLINE | ID: mdl-27231155

ABSTRACT

The autophagy pathway has been recognized as a mechanism of survival and therapy resistance in cancer, yet the extent of autophagy's function in metastatic progression is still unclear. Therefore, we used murine models of metastatic cancer to investigate the effect of autophagy modulation on metastasis development. Pharmacologic and genetic autophagy inhibition were able to impede cell proliferation in culture, but did not impact the development of experimentally induced 4T1 and B16-F10 metastases. Similarly, autophagy inhibition by adjuvant chloroquine (CQ) treatment did not delay metastasis in an orthotopic 4T1, tumor-resection model. However, neoadjuvant CQ treatment or genetic autophagy inhibition resulted in delayed metastasis development, whereas stimulation of autophagy by trehalose hastened development. Cisplatin was also administered either as a single agent or in combination with CQ. The combination of cisplatin and CQ was antagonistic. The effects of autophagy modulation on metastasis did not appear to be due to alterations in the intrinsic metastatic capability of the cells, as modulating autophagy had no impact on migration, invasion, or anchorage-independent growth in vitro. To explore the possibility of autophagy's influence on the metastatic microenvironment, bone marrow-derived cells (BMDCs), which mediate the establishment of the premetastatic niche, were measured in the lung and in circulation. Trehalose-treated mice had significantly more BMDCs than either vehicle- or CQ-treated mice. Autophagy inhibition may be most useful as a treatment to impede early metastatic development. However, modulating autophagy may also alter the efficacy of platinum-based therapies, requiring caution when considering combination therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chloroquine/pharmacology , Cisplatin/pharmacology , Drug Synergism , Female , Mice , Neoadjuvant Therapy , Neoplasm Metastasis , Neoplasm Staging , Time Factors , Trehalose/pharmacology
3.
J Pharmacol Exp Ther ; 357(3): 459-65, 2016 06.
Article in English | MEDLINE | ID: mdl-27026683

ABSTRACT

The aim of this research was to characterize the in vivo and in vitro properties of basal insulin peglispro (BIL), a new basal insulin, wherein insulin lispro was derivatized through the covalent and site-specific attachment of a 20-kDa polyethylene-glycol (PEG; specifically, methoxy-terminated) moiety to lysine B28. Addition of the PEG moiety increased the hydrodynamic size of the insulin lispro molecule. Studies show there is a prolonged duration of action and a reduction in clearance. Given the different physical properties of BIL, it was also important to assess the metabolic and mitogenic activity of the molecule. Streptozotocin (STZ)-treated diabetic rats were used to study the pharmacokinetic and pharmacodynamic characteristics of BIL. Binding affinity and functional characterization of BIL were compared with those of several therapeutic insulins, insulin AspB10, and insulin-like growth factor 1 (IGF-1). BIL exhibited a markedly longer time to maximum concentration after subcutaneous injection, a greater area under the concentration-time curve, and a longer duration of action in the STZ-treated diabetic rat than insulin lispro. BIL exhibited reduced binding affinity and functional potency as compared with insulin lispro and demonstrated greater selectivity for the human insulin receptor (hIR) as compared with the human insulin-like growth factor 1 receptor. Furthermore, BIL showed a more rapid rate of dephosphorylation following maximal hIR stimulation, and reduced mitogenic potential in an IGF-1 receptor-dominant cellular model. PEGylation of insulin lispro with a 20-kDa PEG moiety at lysine B28 alters the absorption, clearance, distribution, and activity profile receptor, but does not alter its selectivity and full agonist receptor properties.


Subject(s)
Insulin Lispro/chemistry , Insulin Lispro/pharmacology , Polyethylene Glycols/chemistry , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cell Differentiation/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Humans , Insulin Lispro/metabolism , Insulin Lispro/pharmacokinetics , Lipogenesis/drug effects , Male , Mice , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/metabolism , Receptor, Insulin/chemistry , Receptor, Insulin/metabolism , Substrate Specificity , Tyrosine/metabolism
4.
Br J Clin Pharmacol ; 81(5): 908-17, 2016 May.
Article in English | MEDLINE | ID: mdl-26648084

ABSTRACT

AIMS: Two phase 1 studies evaluated the pharmacokinetics (PK), safety and biological activity of tabalumab, a human monoclonal antibody against B-cell activating factor (BAFF), administered intravenously (i.v.) or subcutaneously (s.c.) in subjects with rheumatoid arthritis (RA) or systemic lupus erythematosus (SLE). METHODS: In study A, subjects with RA (n = 23) or SLE (n = 6) received a single i.v. dose of tabalumab (RA 0.01, 0.04, 0.125, 0.5, 2.0, and 8.0 mg kg(-1) and SLE 0.125 or 2.0 mg kg(-1) ) or placebo. In study B, subjects with RA received a single tabalumab dose i.v. (10 mg) (n = 12) or s.c. (20 mg) (n = 12). Serum tabalumab and CD20+ B cells were evaluated and safety was assessed throughout both studies. RESULTS: Tabalumab PK were non-linear across the 0.01 to 8.0 mg kg(-1) dose range. Clearance (CL) decreased from 2.9 to 0.1 l day(-1) and terminal half-life (t1/2 ) increased from about 1.6 to 25 days. Subjects with RA or SLE had similar PK. After s.c. dosing, tabalumab time to maximal concentration (tmax ) was 5.5 days. Absolute bioavailability (F) was approximately 62%. Following tabalumab dosing, CD20+ B cells transiently increased from baseline followed by a progressive decrease below baseline. CONCLUSION: A single tabalumab dose administered i.v. or s.c. was well tolerated and had non-linear CL over the dose range investigated in subjects with RA and SLE. The non-linearity likely reflects target-mediated CL due to binding to BAFF. Tabalumab showed biological activity based on changes in peripheral CD20+ lymphocyte numbers in both subjects with RA and SLE.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Arthritis, Rheumatoid/drug therapy , B-Cell Activating Factor/antagonists & inhibitors , Lupus Erythematosus, Systemic/drug therapy , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antigens, CD20/immunology , B-Lymphocyte Subsets/drug effects , B-Lymphocyte Subsets/immunology , Female , Humans , Injections, Intravenous , Injections, Subcutaneous , Male , Middle Aged
5.
Angew Chem Int Ed Engl ; 55(38): 11480-4, 2016 09 12.
Article in English | MEDLINE | ID: mdl-27510286

ABSTRACT

Hydrogen sulfide (H2 S) has emerged as a crucial biomolecule in physiology and cellular signaling. Key challenges associated with developing new chemical tools for understanding the biological roles of H2 S include developing platforms that enable reversible binding of this important biomolecule. The first synthetic small molecule receptor for the hydrosulfide anion, HS(-) , using only reversible, hydrogen-bonding interactions in a series of bis(ethynylaniline) derivatives, is reported. Binding constants of up to 90 300±8700 m(-1) were obtained in MeCN. The fundamental science of reversible sulfide binding, in this case featuring a key CH⋅⋅⋅S hydrogen bond, will expand the possibility for discovery of sulfide protein targets and molecular recognition agents.


Subject(s)
Hydrogen Sulfide/chemistry , Receptors, Artificial/chemistry , Anions/chemistry , Hydrogen Bonding , Hydrogen Sulfide/metabolism , Kinetics , Magnetic Resonance Spectroscopy , Molecular Conformation , Receptors, Artificial/metabolism , Spectrophotometry, Ultraviolet
6.
J Lipid Res ; 56(11): 2124-32, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26392590

ABSTRACT

Lilly PCSK9 antibody LY3015014 (LY) is a monoclonal antibody (mAb) that neutralizes proprotein convertase subtilisin-kexin type 9 (PCSK9). LY decreases LDL cholesterol in monkeys and, unlike other PCSK9 mAbs, does not cause an accumulation of intact PCSK9 in serum. Comparing the epitope of LY with other clinically tested PCSK9 mAbs, it was noted that the LY epitope excludes the furin cleavage site in PCSK9, whereas other mAbs span this site. In vitro exposure of PCSK9 to furin resulted in degradation of PCSK9 bound to LY, whereas cleavage was blocked by other mAbs. These other mAbs caused a significant accumulation of serum PCSK9 and displayed a shorter duration of LDL-cholesterol lowering than LY when administered to mice expressing the WT human PCSK9. In mice expressing a noncleavable variant of human PCSK9, LY behaved like a cleavage-blocking mAb, in that it caused significant PCSK9 accumulation, its duration of LDL lowering was reduced, and its clearance (CL) from serum was accelerated. Thus, LY neutralizes PCSK9 and allows its proteolytic degradation to proceed, which limits PCSK9 accumulation, reduces the CL rate of LY, and extends its duration of action. PCSK9 mAbs with this property are likely to achieve longer durability and require lower doses than mAbs that cause antigen to accumulate.


Subject(s)
Antibodies, Monoclonal/pharmacology , Anticholesteremic Agents/pharmacology , Proprotein Convertases/antagonists & inhibitors , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Anticholesteremic Agents/chemistry , Anticholesteremic Agents/pharmacokinetics , Cholesterol, LDL/blood , Drug Evaluation, Preclinical , Drug Stability , Furin/chemistry , Half-Life , Humans , Hypercholesterolemia/drug therapy , Macaca fascicularis , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Proprotein Convertase 9 , Proprotein Convertases/immunology , Protein Binding , Proteolysis , Serine Endopeptidases/immunology , Treatment Outcome
7.
J Am Chem Soc ; 137(47): 14959-67, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26539974

ABSTRACT

Aryl CH hydrogen bonds (HBs) are now commonly recognized as important factors in a number of fields, including molecular biology, stereoselective catalysis, and anion supramolecular chemistry. As the utility of CH HBs has grown, so to has the need to understand the structure-activity relationship for tuning both their strength and selectivity. Although there has been significant computational effort in this area, an experimental study of the substituent effects on CH HBs has not been previously undertaken. Herein we disclose a systematic study of a single CH HB by using traditional urea donors as directing groups in a supramolecular binding cavity. Experimentally determined association constants are examined by a combination of computational (electrostatic potential) and empirical (σm and σp) values for substituent effects. The dominance of electrostatic parameters, as observed in a computational DFT study, is consistent with current CH HB theory; however, a novel anion dependence of the substituent effects is revealed in solution.


Subject(s)
Carbon/chemistry , Hydrogen/chemistry , Anions , Crystallography, X-Ray , Hydrogen Bonding , Proton Magnetic Resonance Spectroscopy
8.
Vet Dermatol ; 26(4): 228-e50, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26178603

ABSTRACT

BACKGROUND: Placement of ciclosporin (Atopica(®); Novartis Animal Health, Greensboro, NC, USA) capsules in a freezer prior to administration may reduce the incidence of vomiting in dogs. However, its impact on ciclosporin stability and pharmacokinetics is unknown. HYPOTHESIS/OBJECTIVES: The purpose of this study was to evaluate the stability of Atopica(®) capsules and pharmacokinetics of ciclosporin in dogs after storage at -20°C in comparison with storage of capsules at 15-25°C. We hypothesized that there would be no difference in stability or pharmacokinetic parameters between freezer-stored and room-temperature Atopica(®) capsules. ANIMALS: Eight healthy research beagle dogs received one 5.0 mg/kg oral dose each of freezer-stored and room-temperature Atopica(®) capsules with a 1 week washout period between. METHODS: Ciclosporin concentrations of all available Atopica(®) capsule strengths were assessed for stability after -20°C storage at five time points over 30 days and at room temperature (15-25°C). A blinded, randomized cross-over study was also performed to compare blood concentrations of ciclosporin after capsule storage for 28 days at -20 versus 15-25°C. Blood samples were obtained over a 24 h period after administration. Capsule and whole-blood ciclosporin concentrations were assessed via high-performance liquid chromatography-tandem mass spectrometry. RESULTS: There was no significant difference in stability between freezer-stored and room-temperature Atopica(®) capsules at any time point. In the cross-over study, there were no significant differences in pharmacokinetic parameters assessed. CONCLUSIONS AND CLINICAL IMPORTANCE: Placing Atopica(®) capsules in a -20°C freezer for 28 days does not affect stability or absorption in the dog.


Subject(s)
Cyclosporine/pharmacokinetics , Drug Storage , Animals , Capsules , Cyclosporine/administration & dosage , Dogs , Drug Stability , Drug Storage/methods , Female , Freezing , Male , Temperature
9.
Vet Anaesth Analg ; 42(5): 472-83, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25327817

ABSTRACT

OBJECTIVE: To compare the pharmacokinetics and pharmacodynamics of propofol with or without 2% benzyl alcohol administered intravenously (IV) as a single induction dose in cats. STUDY DESIGN: Prospective experimental study. ANIMALS: Six healthy adult cats, three female intact, three male castrated, weighing 4.8 ± 1.8 kg. METHODS: Cats received 8 mg kg(-1) IV of propofol (P) or propofol with 2% benzyl alcohol (P28) using a randomized crossover design. Venous blood samples were collected at predetermined time points to 24 hours after drug administration to determine drug plasma concentrations. Physiologic and behavioral variables were also recorded. Propofol and benzyl alcohol concentrations were determined using high pressure liquid chromatography with fluorescence detection. Pharmacokinetic parameters were described using a 2-compartment model. Pharmacokinetic and pharmacodynamic parameters were analyzed using repeated measures anova (p < 0.05). RESULTS: Plasma concentrations of benzyl alcohol were below the lower limits of quantification (LLOQ) at all time points for two of the six cats (33%), and by 30 minutes for the remaining four cats. Propofol pharmacokinetics, with or without 2% benzyl alcohol, were characterized by rapid distribution, a long elimination phase, and a large volume of distribution. No differences were noted between treatments with the exception of clearance from the second compartment (CLD2), which was 23.6 and 38.8 mL kg(-1)  minute(-1) in the P and P28 treatments, respectively. Physiologic and behavioral variables were not different between treatments with the exception of heart rate at 4 hours post administration. CONCLUSIONS AND CLINICAL RELEVANCE: The addition of 2% benzyl alcohol as a preservative minimally altered the pharmacokinetics and pharmacodynamics of propofol 1% emulsion when administered as a single IV bolus in this group of cats. These data support the cautious use of propofol with 2% benzyl alcohol for induction of anesthesia in healthy cats.


Subject(s)
Anesthetics, Intravenous/pharmacology , Benzyl Alcohol/administration & dosage , Cats/physiology , Propofol/pharmacology , Anesthesia Recovery Period , Anesthesia, Intravenous/veterinary , Anesthetics, Intravenous/blood , Anesthetics, Intravenous/pharmacokinetics , Animals , Cats/metabolism , Cross-Over Studies , Drug Administration Schedule , Female , Male , Propofol/blood , Propofol/pharmacokinetics , Prospective Studies
10.
iScience ; 27(7): 109978, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39021796

ABSTRACT

High-grade serous ovarian cancers (HGSOCs) with homologous recombination deficiency (HRD) are initially responsive to poly (ADP-ribose) polymerase inhibitors (PARPi), but resistance ultimately emerges. HGSOC with CCNE1 amplification (CCNE1 amp) are associated with resistance to PARPi and platinum treatments. High replication stress in HRD and CCNE1 amp HGSOC leads to increased reliance on checkpoint kinase 1 (CHK1), a key regulator of cell cycle progression and the replication stress response. Here, we investigated the anti-tumor activity of the potent, highly selective, orally bioavailable CHK1 inhibitor (CHK1i), SRA737, in both acquired PARPi-resistant BRCA1/2 mutant and CCNE1 amp HGSOC models. We demonstrated that SRA737 increased replication stress and induced subsequent cell death in vitro. SRA737 monotherapy in vivo prolonged survival in CCNE1 amp models, suggesting a potential biomarker for CHK1i therapy. Combination SRA737 and PARPi therapy increased tumor regression in both PARPi-resistant and CCNE1 amp patient-derived xenograft models, warranting further study in these HGSOC subgroups.

11.
J Pharmacol Exp Ther ; 345(1): 125-38, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23318470

ABSTRACT

There are currently no registered drugs that slow the progression of neurodegenerative diseases, in part because translation from animal models to the clinic has been hampered by poor distribution to the brain. The present studies examined a selected series of para-phenyl-substituted diindolylmethane (C-DIM) compounds that display anti-inflammatory and neuroprotective efficacy in vitro. We postulated that the pharmacokinetic behavior of C-DIM compounds after oral administration would correlate with neuroprotective efficacy in vivo in a mouse model of Parkinson's disease. Pharmacokinetics and metabolism of 1,1-bis(3'-indolyl)-1-(p-methoxyphenyl)methane (C-DIM5), 1,1-bis(3'-indolyl)-1-(phenyl)methane, 1,1-bis(3'-indolyl)-1-(p-hydroxyphenyl)methane (C-DIM8), and 1,1-bis(3'-indolyl)-1-(p-chlorophenyl)methane (C-DIM12) were determined in plasma and brain of C57Bl/6 mice after oral and intravenous administration at 10 and 1 mg/Kg, respectively. Putative metabolites were measured in plasma, liver, and urine. C-DIM compounds given orally displayed the highest area under the curve, Cmax, and Tmax levels, and C-DIM12 exhibited the most favorable pharmacokinetics of the compounds tested. Oral bioavailability of each compound ranged from 6% (C-DIM8) to 42% (C-DIM12). After pharmacokinetic studies, the neuroprotective efficacy of C-DIM5, C-DIM8, and C-DIM12 (50 mg/Kg per oral) was examined in mice exposed to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and probenecid for 14 days, a model of progressive neurodegeneration with a strong neuroinflammatory component. C-DIM5 and C-DIM12 given orally once daily after one week of exposure to MPTP and probenecid prevented further loss of dopaminergic neurons in the substantia nigra pars compacta and striatal dopamine terminals, indicating that these compounds could be effective therapeutic agents to prevent neurodegeneration.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Indoles/pharmacokinetics , Indoles/therapeutic use , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/therapeutic use , Parkinsonian Disorders/drug therapy , Administration, Oral , Animals , Anti-Inflammatory Agents/chemistry , Chromatography, High Pressure Liquid , Corpus Striatum/drug effects , Corpus Striatum/pathology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Indoles/chemistry , Injections, Intravenous , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Molecular Structure , Neuroprotective Agents/chemistry , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Tandem Mass Spectrometry
12.
J Org Chem ; 78(13): 6550-7, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23735055

ABSTRACT

Hydrogen sulfide is an important biological signaling molecule and an important environmental target for detection. A major challenge in developing H2S detection methods is separating the often similar reactivity of thiols and other nucleophiles from H2S. To address this need, the nucleophilic aromatic substitution (SNAr) reaction of H2S with electron-poor aromatic electrophiles was developed as a strategy to separate H2S and thiol reactivity. Treatment of aqueous solutions of nitrobenzofurazan (7-nitro-1,2,3-benzoxadiazole, NBD) thioethers with H2S resulted in thiol extrusion and formation of nitrobenzofurazan thiol (λmax = 534 nm). This reactivity allows for unwanted thioether products to be converted to the desired nitrobenzofurazan thiol upon reaction with H2S. The scope of the reaction was investigated using a Hammett linear free energy relationship study, and the determined ρ = +0.34 is consistent with the proposed SN2Ar reaction mechanism. The efficacy of the developed probes was demonstrated in buffer and in serum with associated submicromolar detection limits as low as 190 nM (buffer) and 380 nM (serum). Furthermore, the sigmoidal response of nitrobenzofurazan electrophiles with H2S can be fit to accurately quantify H2S. The developed detection strategy offers a manifold for H2S detection that we foresee being applied in various future applications.


Subject(s)
Benzoxazoles/chemical synthesis , Hydrogen Sulfide/chemistry , Sulfhydryl Compounds/chemical synthesis , Benzoxazoles/chemistry , Colorimetry , Molecular Structure , Sulfhydryl Compounds/chemistry
13.
Br J Pharmacol ; 180(15): 1965-1980, 2023 08.
Article in English | MEDLINE | ID: mdl-36780899

ABSTRACT

BACKGROUND AND PURPOSE: Chronic heart failure, a progressive disease with limited treatment options currently available, especially in heart failure with preserved ejection fraction (HFpEF), represents an unmet medical need as well as an economic burden. The development of a novel therapeutic to slow or reverse disease progression would be highly impactful to patients and society. Relaxin-2 (relaxin) is a human hormone regulating cardiovascular, renal, and pulmonary adaptations during pregnancy. A short-acting recombinant relaxin, Serelaxin, demonstrated short-term heart failure symptom relief and biomarker improvement in acute heart failure trials. Here, we present the development of a long-acting relaxin analogue to be tested in the treatment of chronic heart failure. EXPERIMENTAL APPROACH: LY3540378 is a long-acting protein therapeutic composed of a human relaxin analogue and a serum albumin-binding VHH domain. KEY RESULTS: LY3540378 is a potent agonist of the relaxin family peptide receptor 1 (RXFP1) and maintains selectivity against RXFP2/3/4 comparable to native relaxin. The half-life of LY3540378 in preclinical species is extended through high affinity binding of the albumin-binding VHH domain to serum albumin. When tested in a single dose administration, LY3540378 elicited relaxin-mediated pharmacodynamic responses, such as reduced serum osmolality and increased renal blood flow in rats. In an isoproterenol-induced cardiac hypertrophy mouse model, treatment with LY3540378 significantly reduced cardiac hypertrophy and improved isovolumetric relaxation time. In a monkey cardiovascular safety study, there were no adverse observations from administration of LY3540378. CONCLUSION AND IMPLICATIONS: LY3540378 demonstrated to be a suitable clinical development candidate, and is progressing in clinical trials.


Subject(s)
Heart Diseases , Heart Failure , Relaxin , Animals , Female , Humans , Mice , Pregnancy , Rats , Cardiomegaly/drug therapy , Heart Diseases/drug therapy , Heart Failure/drug therapy , Relaxin/pharmacology , Relaxin/therapeutic use , Relaxin/metabolism , Stroke Volume
14.
J Vet Intern Med ; 34(2): 691-699, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31919893

ABSTRACT

BACKGROUND: Dogs infected with canine parvovirus (CPV) have compromised intestinal epithelial barrier integrity. Production of D-lactate by enteric bacteria may directly reflect disease severity or contribute to metabolic acid-base status in these dogs. HYPOTHESIS: Serum D-lactate concentration will be increased in CPV dogs compared to healthy controls and correlate with markers of disease severity and acid-base status. ANIMALS: Dogs with CPV undergoing treatment (n = 40) and healthy control dogs (n = 9). METHODS: Prospective observational study. Dogs with CPV had a baseline and daily CBC, venous blood gas with serum electrolyte concentrations, composite clinical severity score, and serum D-lactate concentration performed. A single serum D-lactate measurement was obtained from healthy control dogs. RESULTS: The CPV dogs had a higher D-lactate concentration (mean ± SD) of 469 ± 173 µM compared to controls, 306 ± 45 µM (P < .001). There was no difference in baseline D-lactate concentrations for CPV survivors (474 ± 28 µM), versus nonsurvivors (424 ± 116 µM; P = .70). D-lactate concentration decreased over the first 4 days of treatment (-9.6 µM/d; P = .46). Dogs hospitalized for <4 days had lower baseline D-lactate concentrations compared to those hospitalized ≥4 days (400 ± 178 µM versus 520 ± 152 µM; P = .03). No sustained correlation over time between serum D-lactate concentration and clinical severity score or recorded acid-base results. CONCLUSIONS AND CLINICAL IMPORTANCE: Serum D-lactate concentrations are higher in dogs with CPV compared to healthy controls but do not appear to be clinically relevant. No relationship identified between serum D-lactate concentrations and markers of CPV disease severity, acid-base status, or outcome.


Subject(s)
Dog Diseases/blood , Enteritis/veterinary , Lactic Acid/blood , Parvoviridae Infections/veterinary , Animals , Dogs , Enteritis/blood , Enteritis/virology , Parvoviridae Infections/blood , Parvoviridae Infections/pathology , Prospective Studies
15.
J Thorac Oncol ; 14(12): 2152-2163, 2019 12.
Article in English | MEDLINE | ID: mdl-31470128

ABSTRACT

INTRODUCTION: Despite the enthusiasm surrounding cancer immunotherapy, most SCLC patients show very modest response to immune checkpoint inhibitor monotherapy treatment. Therefore, there is growing interest in combining immune checkpoint blockade with chemotherapy and other treatments to enhance immune checkpoint blockade efficacy. Based on favorable clinical trial results, chemotherapy and immunotherapy combinations have been recently approved by the U.S. Food and Drug Administration for frontline treatment for SCLC. METHODS AND RESULTS: Here, we show that combined treatment of SRA737, an oral CHK1 inhibitor, and anti-programmed death ligand 1 (PD-L1) leads to an antitumor response in multiple cancer models, including SCLC. We further show that combining low, non-cytotoxic doses of gemcitabine with SRA737 + anti-PD-L1/anti-PD-1 significantly increased antitumorigenic CD8+ cytotoxic T cells, dendritic cells, and M1 macrophage populations in an SCLC model. This regimen also led to a significant decrease in immunosuppressive M2 macrophage and myeloid-derived suppressor cell populations, as well as an increase in the expression of the type I interferon beta 1 gene, IFNß, and chemokines, CCL5 and CXCL10. CONCLUSIONS: Given that anti-PD-L1/anti-PD-1 drugs have recently been approved as monotherapy and in combination with chemotherapy for the treatment of SCLC, and that the SRA737 + low dose gemcitabine regimen is currently in clinical trials for SCLC and other malignancies, our preclinical data provide a strong rational for combining this regimen with inhibitors of the PD-L1/PD-1 pathway.


Subject(s)
Combined Modality Therapy/methods , Deoxycytidine/analogs & derivatives , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Immunotherapy/methods , Lung Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Small Cell Lung Carcinoma/drug therapy , Tumor Microenvironment/immunology , Administration, Oral , Animals , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Female , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Lung Neoplasms/pathology , Mice , Small Cell Lung Carcinoma/pathology , Xenograft Model Antitumor Assays , Gemcitabine
16.
DNA Repair (Amst) ; 6(8): 1145-54, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17485251

ABSTRACT

O(6)-Methylguanine-DNA methyltransferase (MGMT) is a DNA repair protein that protects cells from the biological consequences of alkylating agents by removing alkyl groups from the O(6)-position of guanine. Cyclophosphamide and ifosfamide are oxazaphosphorines used clinically to treat a wide variety of cancers; however, the role of MGMT in recognizing DNA damage induced by these agents is unclear. In vitro evidence suggests that MGMT may protect against the urotoxic oxazaphosphorine metabolite, acrolein. Here, we demonstrate that Chinese hamster ovary cells transfected with MGMT are protected against cytotoxicity following treatment with chloroacetaldehyde (CAA), a neuro- and nephrotoxic metabolite of cyclophosphamide and ifosfamide. The mechanism by which MGMT recognizes damage induced by acrolein and CAA is unknown. CHO cells expressing a mutant form of MGMT (MGMT(R128A)), known to have >1000-fold less repair activity towards alkylated DNA while maintaining full active site transferase activity towards low molecular weight substrates, exhibited equivalent CAA- and acrolein-induced cytotoxicity to that of CHO cells transfected with plasmid control. These results imply that direct reaction of acrolein or CAA with the active site cysteine residue of MGMT, i.e. scavenging, is unlikely a mechanism to explain MGMT protection from CAA and acrolein-induced toxicity. In vivo, no difference was detected between Mgmt-/- and Mgmt+/+ mice in the lethal effects of cyclophosphamide. While MGMT may be important at the cellular level, mice deficient in MGMT are not significantly more susceptible to cyclophosphamide, acrolein or CAA. Thus, our data does not support targeting MGMT to improve oxazaphosphorine therapy.


Subject(s)
Cyclophosphamide/toxicity , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Acetaldehyde/analogs & derivatives , Acetaldehyde/toxicity , Acrolein/toxicity , Alkylating Agents/metabolism , Alkylating Agents/toxicity , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclophosphamide/metabolism , DNA Repair , Drug Resistance, Neoplasm , Ifosfamide/metabolism , Ifosfamide/toxicity , Mice , Mice, Knockout , Mutation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/enzymology , O(6)-Methylguanine-DNA Methyltransferase/deficiency , O(6)-Methylguanine-DNA Methyltransferase/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Transplantation, Heterologous
17.
Mutagenesis ; 23(5): 341-6, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18477655

ABSTRACT

O(6)-methylguanine DNA methyltransferase (MGMT) deficiency is associated with an increased susceptibility to alkylating agent toxicity. To understand the contribution of MGMT in protecting against cyclophosphamide (CP)-induced toxicity, mutagenesis and tumorigenesis, we compared the biological effects of this agent in transgenic Mgmt knockout and wild-type mice. In addition, neurofibromin (Nf1)+/- background was used to increase the likelihood of CP-induced tumorigenesis. Cohorts of Mgmt-proficient or -deficient mice (either Nf1+/+ or Nf1+/-) were given 6 weekly injections of a maximally tolerated dose of CP (250 mg/kg) or vehicle and followed for 15 months. CP-treated mice had more deaths than control mice but there was no difference in the long-term survival between Mgmt+/+ and Mgmt-/- mice (12 of 83 Mgmt+/+ mice died compared to 12 of 80 Mgmt-/- mice, disregarding Nf1 status). Lymphomas and adrenal tumours were the most frequent malignancies. Interestingly, CP-treated, Mgmt-deficient mice developed fewer tumours than controls. Ten of 71 (14%) Mgmt-proficient mice developed tumours after CP treatment compared to only 2 of 68 (3%) Mgmt-deficient mice (P = 0.02). Mgmt-/-, Nf1+/- mice developed fewer tumours (1 of 35, 3%) following CP compared to Mgmt+/+, Nf1+/- mice (7 of 37, 19%) (P = 0.03). Hypoxanthine-guanine phosphoribosyltransferase mutation assays showed no significant increases in mutant frequencies in Mgmt-/- (18.1 x 10(6)) compared to Mgmt+/+ mice (12.9 x 10(6)). These data indicate that MGMT deficiency does not protect against long-term toxicity or mutagenicity from CP and appears to attenuate the occurrence of CP-induced tumours in an Nf1+/- background.


Subject(s)
Alkylating Agents/toxicity , Cell Transformation, Neoplastic/chemically induced , Cyclophosphamide/toxicity , O(6)-Methylguanine-DNA Methyltransferase/physiology , Animals , Cell Transformation, Neoplastic/genetics , Hematopoiesis/drug effects , Hypoxanthine Phosphoribosyltransferase/genetics , Mice , Mice, Knockout , Mutagenesis , Mutation , Neurofibromin 1/genetics , O(6)-Methylguanine-DNA Methyltransferase/genetics
18.
J Feline Med Surg ; 20(8): 721-727, 2018 08.
Article in English | MEDLINE | ID: mdl-28905667

ABSTRACT

Objectives The objectives were to evaluate the pharmacokinetics (PK) of subcutaneous (SC) and intravenous (IV) dolasetron and the pharmacodynamics (PD) of SC dolasetron in healthy cats. Methods Five cats with unremarkable complete blood count, serum biochemistry and urinalyses were utilized. In the PK study, cats received 0.8 mg/kg SC and IV dolasetron in a crossover format. Serum samples were obtained via a jugular catheter at 0, 0.25, 0.5, 1, 2, 4, 8, 12, 24, 36 and 48 h after the administration of dolasetron. Dolasetron and the active metabolite hydrodolasetron were measured using liquid chromatography/tandem mass spectrometry. Non-compartmental PK analysis was performed. In the PD study, SC dolasetron (0.8 mg/kg and 1.0 mg/kg) and saline were administered 30 mins prior to administration of 0.44 mg/kg intramuscular xylazine in a randomized three-way crossover. Number of emetic events, lip licks, time to onset of emesis and visual nausea score were scored by a blinded observer. Results In the PK study, dolasetron was quickly metabolized to the active metabolite hydrodolasetron, limiting assessment of dolasetron PK parameters. Median (range) PK parameters for IV hydrodolasetron were as follows: maximum serum concentration (Cmax) 116 ng/ml (69-316 ng/ml), time to maximum concentration (Tmax) 0.5 h (0.3-0.5 h), half-life 3.3 h (2.9-7.2 h) and area under the curve until the last measurable concentration (AUClast) 323 h/ng/ml (138-454 h/ng/ml). Median (range) PK parameters for SC hydrodolasetron were as follows: Cmax 67.9 ng/ml (60.4-117 ng/ml), Tmax 0.5 h (0.5-1.0 h), half-life 3.8 h (2.9-5.3 h) and AUClast 437 h/ng/ml (221.5-621.8 h/ng/ml). There was no significant difference in exposure to hydrodolasetron between the routes of administration. With regard to PD, when dolasetron was administered prior to xylazine, there was no significant difference in the mean number of emetic events, lip licks, time to onset of emesis or visual nausea score when compared with saline. Conclusions and relevance Administration of 0.8 mg/kg dolasetron does not maintain serum concentrations of active metabolite for 24 h. Administration of dolasetron at 0.8 mg/kg and 1 mg/kg did not prevent xylazine-induced vomiting. Additional feline dose studies are needed to determine if a higher dose is efficacious.


Subject(s)
Cats/metabolism , Indoles/administration & dosage , Indoles/pharmacokinetics , Quinolizines/administration & dosage , Quinolizines/pharmacokinetics , Administration, Intravenous , Animals , Chromatography, Liquid , Cross-Over Studies , Double-Blind Method , Indoles/adverse effects , Indoles/blood , Infusions, Subcutaneous , Injections, Intramuscular , Quinolizines/adverse effects , Quinolizines/blood , Random Allocation , Tandem Mass Spectrometry , Xylazine/administration & dosage
19.
PLoS One ; 12(9): e0185089, 2017.
Article in English | MEDLINE | ID: mdl-28926611

ABSTRACT

Activation of the unfolded protein response (UPR) in eukaryotic cells represents an evolutionarily conserved response to physiological stress. Here, we report that the mTOR inhibitors rapamycin (sirolimus) and structurally related temsirolimus are capable of inducing UPR in sarcoma cells. However, this effect appears to be distinct from the classical role for these drugs as mTOR inhibitors. Instead, we detected these compounds to be associated with ribosomes isolated from treated cells. Specifically, temsirolimus treatment resulted in protection from chemical modification of several rRNA residues previously shown to bind rapamycin in prokaryotic cells. As an application for these findings, we demonstrate maximum tumor cell growth inhibition occurring only at doses which induce UPR and which have been shown to be safely achieved in human patients. These results are significant because they challenge the paradigm for the use of these drugs as anticancer agents and reveal a connection to UPR, a conserved biological response that has been implicated in tumor growth and response to therapy. As a result, eIF2 alpha phosphorylation and Xbp-1 splicing may serve as useful biomarkers of treatment response in future clinical trials using rapamycin and rapalogs.


Subject(s)
Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Unfolded Protein Response/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Eukaryotic Initiation Factor-2/metabolism , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA Splicing/drug effects , RNA, Messenger/metabolism , RNA, Ribosomal, 28S/metabolism , Sarcoma/metabolism , Sarcoma/pathology , Solvents/chemistry , Solvents/metabolism , TOR Serine-Threonine Kinases/metabolism , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
20.
Am J Vet Res ; 78(7): 862-866, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28650232

ABSTRACT

OBJECTIVE To characterize pharmacokinetics of cyclophosphamide and 4-hydoxycyclophosphamide (4-OHCP) in the plasma of healthy cats after oral, IV, and IP administration of cyclophosphamide. ANIMALS 6 healthy adult cats. PROCEDURES Cats were randomly assigned to receive cyclophosphamide (200 mg/m2) via each of 3 routes of administration (oral, IV, and IP); there was a 30-day washout period between successive treatments. Plasma samples were obtained at various time points for up to 8 hours after administration. Samples were treated with semicarbazide hydrochloride to trap the 4-OHCP in stable form, which allowed for cyclophosphamide and trapped 4-OHCP to be simultaneously measured by use of tandem mass spectrometry. Pharmacokinetic parameters were determined from drug concentration-versus-time data for both cyclophosphamide and 4-OHCP. RESULTS Cyclophosphamide was tolerated well regardless of route of administration. Pharmacokinetic parameters for 4-OHCP were similar after oral, IV, and IP administration. Area under the concentration-time curve for cyclophosphamide was lower after oral administration than after IV or IP administration. CONCLUSIONS AND CLINICAL RELEVANCE Cyclophosphamide can be administered interchangeably to cats as oral, IV, and IP formulations, which should provide benefits with regard to cost and ease of administration to certain feline patients.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Cats/metabolism , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/pharmacokinetics , Administration, Intravenous/veterinary , Administration, Oral , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Area Under Curve , Biological Availability , Cyclophosphamide/administration & dosage , Female , Half-Life , Injections, Intraperitoneal/veterinary , Male , Tandem Mass Spectrometry/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL