Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 387(3): 306-314, 2023 12.
Article in English | MEDLINE | ID: mdl-37699709

ABSTRACT

Blockade of the cluster of differentiation 40 (CD40)-CD40L interaction has potential for treating autoimmune diseases and preventing graft rejection. This first-in-human, randomized, double-blind, placebo-controlled study (NCT04497662) evaluated safety, pharmacokinetics, receptor occupancy, and pharmacodynamics of the humanized anti-CD40 monoclonal antibody KPL-404. Healthy volunteers were randomized to one of two single-ascending-dose groups: single intravenous KPL-404 dose 0.03, 0.3, 1, 3, or 10 mg/kg or single subcutaneous KPL-404 dose 1 or 5 mg/kg. There were no dose-limiting or dose-related safety findings. Nonlinear dose-dependent changes in various pharmacokinetic parameters were identified following the range of intravenous doses. At the 10 mg/kg intravenous dose level, the t1/2 was approximately 7 days, and full receptor occupancy was observed through Day 71, with complete suppression of T-cell-dependent antibody response (TDAR) to keyhole limpet hemocyanin (KLH) challenge on Day 1 and rechallenge on Day 29 through Day 57. With KPL-404 5 mg/kg subcutaneously, full receptor occupancy was observed through Day 43, with complete suppression of TDAR through at least Day 29. Antidrug antibodies to KPL-404 were suppressed for 57 days with 10 mg/kg intravenously and for 50 days with 5 mg/kg subcutaneously, further confirming prolonged target engagement and pharmacodynamics. These findings support continued investigation of KPL-404 intravenous and subcutaneous administration in a broad range of indications. SIGNIFICANCE STATEMENT: This first-in-human clinical trial of KPL-404, a fully humanized IgG4 monoclonal antibody, was designed with two independent (by route of administration) placebo-controlled single-ascending-dose-level groups, one with four intravenous single-dose cohorts and another with two subcutaneous single-dose cohorts. The pharmacokinetic profile, duration of full CD40 receptor occupancy, and magnitude and duration of memory immune response suppression observed confirm pharmacodynamic activity regardless of administration route. These data provide evidence that chronic KPL-404 dosing regimens (intravenous or subcutaneous) could be practical.


Subject(s)
Antibodies, Monoclonal , Antibody Formation , Humans , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Administration, Intravenous , T-Lymphocytes , Double-Blind Method
2.
Ann Rheum Dis ; 81(5): 653-661, 2022 05.
Article in English | MEDLINE | ID: mdl-35264321

ABSTRACT

OBJECTIVES: Granulocyte-macrophage colony-stimulating factor (GM-CSF) is implicated in pathogenesis of giant cell arteritis. We evaluated the efficacy of the GM-CSF receptor antagonist mavrilimumab in maintaining disease remission. METHODS: This phase 2, double-blind, placebo-controlled trial enrolled patients with biopsy-confirmed or imaging-confirmed giant cell arteritis in 50 centres (North America, Europe, Australia). Active disease within 6 weeks of baseline was required for inclusion. Patients in glucocorticoid-induced remission were randomly assigned (3:2 ratio) to mavrilimumab 150 mg or placebo injected subcutaneously every 2 weeks. Both groups received a 26-week prednisone taper. The primary outcome was time to adjudicated flare by week 26. A prespecified secondary efficacy outcome was sustained remission at week 26 by Kaplan-Meier estimation. Safety was also assessed. RESULTS: Of 42 mavrilimumab recipients, flare occurred in 19% (n=8). Of 28 placebo recipients, flare occurred in 46% (n=13). Median time to flare (primary outcome) was 25.1 weeks in the placebo group, but the median was not reached in the mavrilimumab group (HR 0.38; 95% CI 0.15 to 0.92; p=0.026). Sustained remission at week 26 was 83% for mavrilimumab and 50% for placebo recipients (p=0.0038). Adverse events occurred in 78.6% (n=33) of mavrilimumab and 89.3% (n=25) of placebo recipients. No deaths or vision loss occurred in either group. CONCLUSIONS: Mavrilimumab plus 26 weeks of prednisone was superior to placebo plus 26 weeks of prednisone for time to flare by week 26 and sustained remission in patients with giant cell arteritis. Longer treatment is needed to determine response durability and quantify the glucocorticoid-sparing potential of mavrilimumab. TRIAL REGISTRATION NUMBER: ClinicalTrials.gov number: NCT03827018, Europe (EUdraCT number: 2018-001003-36), and Australia (CT-2018-CTN-01 865-1).


Subject(s)
Giant Cell Arteritis , Antibodies, Monoclonal, Humanized/adverse effects , Double-Blind Method , Giant Cell Arteritis/drug therapy , Glucocorticoids/therapeutic use , Humans , Prednisone/adverse effects , Treatment Outcome
3.
J Med Chem ; 59(3): 854-66, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26789203

ABSTRACT

CRF mediates numerous stress-related endocrine, autonomic, metabolic, and behavioral responses. We present the synthesis and chemical and biological properties of astressin B analogues {cyclo(30-33)[D-Phe(12),Nle(21,38),C(α)MeLeu(27,40),Glu(30),Lys(33)]-acetyl-h/r-CRF(9-41)}. Out of 37 novel peptides, 17 (2, 4, 6-8, 10, 11, 16, 17, 27, 29, 30, 32-36) and 16 (3, 5, 9, 12-15, 18, 19, 22-26, 28, 31) had k(i) to CRF receptors in the high picomolar and low nanomole ranges, respectively. Peptides 1, 2, and 11 inhibited h/rCRF and urocortin 1-induced cAMP release from AtT20 and A7r5 cells. When Astressin C 2 was administered to adrenalectomized rats at 1.0 mg subcutaneously, it inhibited ACTH release for >7 d. Additional rat data based on the inhibitory effect of (2) on h/rCRF-induced stimulation of colonic secretory motor activity and urocortin 2-induced delayed gastric emptying also indicate a safe and long-lasting antagonistic effect. The overall properties of selected analogues may fulfill the criteria expected from clinical candidates.


Subject(s)
Corticotropin-Releasing Hormone/pharmacology , Peptide Fragments/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Animals , Corticotropin-Releasing Hormone/administration & dosage , Corticotropin-Releasing Hormone/chemistry , Cyclic AMP/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Molecular Structure , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Rats , Structure-Activity Relationship , Urocortins/antagonists & inhibitors
4.
PLoS One ; 9(2): e87704, 2014.
Article in English | MEDLINE | ID: mdl-24503632

ABSTRACT

The design, synthesis and pharmacology of novel long-acting exenatide analogs for the treatment of metabolic diseases are described. These molecules display enhanced pharmacokinetic profile and potent glucoregulatory and weight lowering actions compared to native exenatide. [Leu(14)]exenatide-ABD is an 88 residue peptide amide incorporating an Albumin Binding Domain (ABD) scaffold. [Leu(14)]exenatide-ABP is a 53 residue peptide incorporating a short Albumin Binding Peptide (ABP). [Leu(14)]exenatide-ABD and [Leu(14)]exenatide-ABP exhibited nanomolar functional GLP-1 receptor potency and were metabolically stable in vitro in human plasma and in a pancreatic digestive enzyme mixture. Both molecules displayed picomolar and nanomolar binding association with albumin across multiple species and circulating half lives of 16 and 11 hours, respectively, post a single IV dose in rats. Unlike exenatide, both molecules elicited robust glucose lowering when injected 1 day prior to an oral glucose tolerance test, indicative of their extended duration of action. [Leu(14)]exenatide-ABD was compared to exenatide in a Lep (ob/ob) mouse model of diabetes. Twice-weekly subcutaneously dosed [Leu(14)]exenatide-ABD displayed superior glucose lowering and weight loss in diabetic mice when compared to continuously infused exenatide at the same total weekly dose. A single oral administration of each molecule via an enteric coated capsule to cynomolgus monkeys showed superior pharmacokinetics for [Leu(14)]exenatide-ABD as compared to [Leu(14)]exenatide-ABP with detectable exposure longer than 14 days. These studies support the potential use of these novel long acting exenatide analogs with different routes of administration for the treatment of type 2 diabetes.


Subject(s)
Albumins/chemistry , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacokinetics , Peptides/chemistry , Peptides/pharmacokinetics , Protein Interaction Domains and Motifs , Venoms/chemistry , Venoms/pharmacokinetics , Administration, Oral , Albumins/metabolism , Animals , Binding Sites , Diabetes Mellitus, Experimental , Disease Models, Animal , Drug Stability , Exenatide , Glucagon-Like Peptide-1 Receptor , Glucose Tolerance Test , Humans , Hypoglycemic Agents/metabolism , Kinetics , Macaca fascicularis , Male , Mice , Peptides/metabolism , Protein Binding , Rats , Receptors, Glucagon/metabolism , Venoms/metabolism
5.
J Med Chem ; 51(9): 2676-81, 2008 May 08.
Article in English | MEDLINE | ID: mdl-18410083

ABSTRACT

H-DPhe (2)-c[Cys (3)-Phe (7)-DTrp (8)-Lys (9)-Thr (10)-Cys (14)]-Thr (15)-NH2 (1) (a somatostatin agonist, SRIF numbering) and H-Cpa (2)-c[DCys (3)-Tyr (7)-DTrp (8)-Lys (9)-Thr (10)-Cys (14)]-Nal (15)-NH2 (4) (a somatostatin antagonist) are based on the structure of octreotide that binds to three somatostatin receptor subtypes (sst 2/3/5) with significant binding affinity. Analogues of 1 and 4 were synthesized with norcysteine (Ncy), homocysteine (Hcy), or D-homocysteine (DHcy) at positions 3 and/or 14. Introducing Ncy at positions 3 and 14 constrained the backbone flexibility, resulting in loss of binding affinity at all sst s. The introduction of Hcy at positions 3 and 14 improved selectivity for sst 2 as a result of significant loss of binding affinity at the other sst s. Substitution by DHcy at position 3 in the antagonist scaffold (5), on the other hand, resulted in a significant loss of binding affinity at sst 2 and sst 3 as compared to the different affinities of the parent compound (4). The 3D NMR structures of the analogues in dimethylsulfoxide are consistent with the observed binding affinities.


Subject(s)
Octreotide/chemical synthesis , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors , Cell Line , Cysteine/chemistry , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Octreotide/chemistry , Octreotide/pharmacology , Radioligand Assay , Stereoisomerism , Structure-Activity Relationship
6.
J Med Chem ; 51(9): 2668-75, 2008 May 08.
Article in English | MEDLINE | ID: mdl-18410084

ABSTRACT

The synthesis, biological testing, and NMR studies of several analogues of H-c[Cys (3)-Phe (6)-Phe (7)-DTrp (8)-Lys (9)-Thr (10)-Phe (11)-Cys (14)]-OH (ODT-8, a pan-somatostatin analogue, 1) have been performed to assess the effect of changing the stereochemistry and the number of atoms in the disulfide bridge on binding affinity. Cysteine at positions 3 and/or 14 (somatostatin numbering) were/was substituted with d-cysteine, norcysteine, D-norcysteine, homocysteine, and/or D-homocysteine. The 3D structure analysis of selected partially selective, bioactive analogues (3, 18, 19, and 21) was carried out in dimethylsulfoxide. Interestingly and not unexpectedly, the 3D structures of these analogues comprised the pharmacophore for which the analogues had the highest binding affinities (i.e., sst 4 in all cases).


Subject(s)
Peptides, Cyclic/chemical synthesis , Receptors, Somatostatin/metabolism , Animals , Binding, Competitive , CHO Cells , Cricetinae , Cricetulus , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Radioligand Assay , Stereoisomerism , Structure-Activity Relationship
7.
J Med Chem ; 50(9): 2067-77, 2007 May 03.
Article in English | MEDLINE | ID: mdl-17402723

ABSTRACT

A series of acyline analogues incorporating l- and d-isomers of S-arylated/alkylated norcysteines [Ncy(R), where R is 2-naphthyl, methyl, and isopropyl] at positions 1, 4, 7, and 10 were synthesized. Some of these analogues were mono- and dioxidized to sulfoxides and sulfones. All of the analogues of acyline were screened for the antagonism of the GnRH-induced response in a reporter gene assay in HEK-293 cells expressing the human GnRH receptor. Nine of the analogues (9, 11, 15, 16, 17, 19, 20, 21, and 22) had antagonistic potency (IC50 < 2 nM) similar to that of acyline (IC50 = 0.52 nM) in this assay. Selected analogues (9, 11, 15, 16, 19, and 21) were tested in vitro for their antagonism at the rat GnRH-R in a reporter gene assay as well as in an in vivo intact male rat assay. Analogues 9 and 15 were the most potent in suppressing testosterone levels.


Subject(s)
Cysteine/analogs & derivatives , Cysteine/chemical synthesis , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Oligopeptides/chemical synthesis , Animals , Butyrates/chemical synthesis , Butyrates/pharmacology , Cell Line , Chlorocebus aethiops , Cysteine/pharmacology , Genes, Reporter , Gonadotropin-Releasing Hormone/genetics , Humans , Leucine/analogs & derivatives , Leucine/chemical synthesis , Leucine/pharmacology , Male , Naphthalenes/chemical synthesis , Naphthalenes/pharmacology , Oligopeptides/pharmacology , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Solubility , Stereoisomerism , Sulfones/chemical synthesis , Sulfones/pharmacology , Sulfoxides/chemical synthesis , Sulfoxides/pharmacology , Testosterone/antagonists & inhibitors , Testosterone/blood
8.
Tetrahedron Lett ; 48(29): 5107-5110, 2007 Jul 16.
Article in English | MEDLINE | ID: mdl-19562100

ABSTRACT

We report the synthesis of racemic Alloc-Ncy(Tmob)-OH, the resolution of its methyl ester, and demonstrate its application to form a norcystine bridge in octreotide-amide using the Fmoc-strategy on solid phase. N-Alloc and S-Tmob protections of norcysteine (Ncy) were found to be a preferred choice for Fmoc-strategy over three other protected norcysteines synthesized i.e. Fmoc-Ncy(tBu)-OH, Alloc-Ncy(tBu)-OH and Alloc-Ncy(Trt)-OH.

9.
J Med Chem ; 49(12): 3536-43, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16759096

ABSTRACT

Novel degarelix (Fe200486) analogues were screened for antagonism of GnRH-induced response (IC(50)) in a reporter gene assay. Inhibition of luteinizing hormone release over time was measured in the castrated male rat. N(omega)-Hydroxy- and N(omega)-methoxy-carbamoylation of Dab and Dap at position 3 (3-6), and N(omega)-hydroxy-,N(omega)-methoxy-carbamoylation and pegylation of 4Aph at positions 5 and 6 (7-10, 15-17, 22-25) were carried out. Modulation of hydrophobicity was achieved using different acylating groups at the N-terminus (11-14, 18-21, 26-28). Analogues 8, 15-17, 22, and 23 were equipotent to acyline (IC(50) = 0.69 nM) and degarelix (IC(50) = 0.58 nM) in vitro. Analogues 7, 17, and 23 were shorter acting than acyline, when 9, 11, 13, 15, 16, and 22 were longer acting. Only 9 and 14 were inactive at releasing histamine. No analogue exhibited a duration of action comparable to that of degarelix. Analogues with shorter and longer retention times on HPLC (a measure of hydrophilicity) than degarelix were identified.


Subject(s)
Butyric Acid/chemistry , Caprylates/chemistry , Oligopeptides/chemical synthesis , Polyethylene Glycols/chemistry , Urea/chemistry , Acylation , Animals , Cell Line , Chromatography, High Pressure Liquid , Female , Genes, Reporter , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropin-Releasing Hormone/blood , Histamine Release/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Male , Mast Cells/drug effects , Mast Cells/metabolism , Oligopeptides/chemistry , Oligopeptides/pharmacology , Orchiectomy , Ovulation/drug effects , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
10.
Org Lett ; 8(11): 2361-4, 2006 May 25.
Article in English | MEDLINE | ID: mdl-16706526

ABSTRACT

[reaction: see text] Norcysteine (Ncy) is an unnatural amino acid possessing an electronegative sulfur atom directly attached to the alpha-carbon atom. We describe the synthesis of Boc-D,L-Ncy(Mob)-OH, the resolution of its methyl ester, and the introduction of both D- and L-Ncy in GnRH analogues.


Subject(s)
Amino Acids/chemistry , Cysteine/analogs & derivatives , Cystine/analogs & derivatives , Cystine/chemistry , Gonadotropin-Releasing Hormone/chemistry , Peptides/chemical synthesis , Amino Acid Sequence , Cysteine/chemical synthesis , Cysteine/chemistry , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Humans , Luciferases/genetics , Luciferases/metabolism , Molecular Structure , Peptides/chemistry , Peptides/pharmacology , Receptors, LHRH/metabolism , Stereoisomerism , Structure-Activity Relationship
11.
J Med Chem ; 48(15): 4851-60, 2005 Jul 28.
Article in English | MEDLINE | ID: mdl-16033265

ABSTRACT

Degarelix (FE200486, Ac-d-2Nal(1)-d-4Cpa(2)-d-3Pal(3)-Ser(4)-4Aph(l-Hor)(5)-d-4Aph(Cbm)(6)-Leu(7)-ILys(8)-Pro(9)-d-Ala(10)-NH(2)) is a potent and very long acting antagonist of gonadotropin-releasing hormone (GnRH) after subcutaneous administration in mammals including humans. Analogues of degarelix were synthesized, characterized, and screened for the antagonism of GnRH-induced response in a reporter gene assay in HEK-293 cells expressing the human GnRH receptor. The duration of action was also determined in the castrated male rat assay to measure the extent (efficacy and duration of action) of inhibition of luteinizing hormone (LH) release. Structurally, this series of analogues has novel substitutions at positions 3, 7, and 8 and N(alpha)-methylation at positions 6, 7, and 8 in the structure of degarelix. These substitutions were designed to probe the spatial limitations of the receptor's cavity and to map the steric and ionic boundaries. Some functional groups were introduced that were hypothesized to influence the phamacokinetic properties of the analogues such as bioavailability, solubility, intra- or intermolecular hydrogen bond forming capacity, and ability to bind carrier proteins. Substitutions at positions 3 ([N(beta)-(2-pyridyl-methyl)d-Dap(3)]degarelix, IC(50) = 2.71 nM) (5), 7 ([Pra(7)]degarelix, IC(50) = 2.11 nM) (16), and 8 ([N(delta)-(IGly)Orn(8)]degarelix, IC(50) = 1.38 nM) (20) and N-methylation ([N(alpha)-methyl-Leu(7)]degarelix, IC(50) = 1.47 nM) (32) yielded analogues that were equipotent to degarelix (2) in vitro (IC(50) = 1.64 nM) but shorter acting in vivo. Out of the 33 novel analogues tested for the duration of action in this series, two analogues ([N(epsilon)-cyclohexyl-Lys(8)]degarelix, IC(50) = 1.50 nM) (23) and ([N(beta)-(IbetaAla)Dap(8)]degarelix, IC(50) = 1.98 nM) (26) had antagonist potencies and duration of action similar to that of azaline B {inhibited LH (>80%) release for >72 h after sc injection to castrated male rats at a standard dose of 50 mug/rat in 5% mannitol}. Under similar conditions analogues ([N(gamma)-(IGly)Dab(8)]degarelix, IC(50) = 1.56 nM) (21) and ([IOrn(8)]degarelix, IC(50) = 1.72 nM) (18) had a longer duration of action {inhibited LH (>96 h) release} than azaline B; however they were shorter acting than degarelix. Hydrophilicity of these analogues, a potential measure of their ability to be formulated for sustained release, was determined using RP-HPLC at neutral pH yielding analogues with shorter as well as longer retention times. No correlation was found between retention times and antagonist potency or duration of action.


Subject(s)
Gonadotropin-Releasing Hormone/antagonists & inhibitors , Oligopeptides/chemical synthesis , Animals , Cell Line , Chromatography, High Pressure Liquid , Genes, Reporter , Humans , Luciferases/genetics , Luteinizing Hormone/antagonists & inhibitors , Male , Oligopeptides/chemistry , Oligopeptides/pharmacology , Orchiectomy , Rats , Rats, Sprague-Dawley , Receptors, LHRH/antagonists & inhibitors , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 15(11): 2894-7, 2005 Jun 02.
Article in English | MEDLINE | ID: mdl-15911275

ABSTRACT

Several azaline B analogs (2-10) were synthesized and evaluated for their ability to antagonize GnRH in vitro and for duration of action in inhibiting luteinizing hormone secretion in a castrated male rat assay in vivo. Analogs, 8 (IC(50) = 1.85 nM), and 9 (IC(50) = 1.78 nM), are equipotent with azaline B (1, IC(50) = 1.36 nM) in vitro. Whereas 9 is short acting, 8 is as long acting as azaline B. Other analogs have IC(50) greater than 2.0 nM and are all short acting.


Subject(s)
Gonadotropin-Releasing Hormone/analogs & derivatives , Animals , Cell Line , Chromatography, High Pressure Liquid , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropin-Releasing Hormone/chemical synthesis , Gonadotropin-Releasing Hormone/chemistry , Gonadotropin-Releasing Hormone/pharmacology , Humans , Male , Rats
13.
Biopolymers ; 80(2-3): 386-91, 2005.
Article in English | MEDLINE | ID: mdl-15614806

ABSTRACT

Gonadotropin-releasing hormone (GnRH) antagonists with high potency and improved duration of action are needed for potential clinical applications. We synthesized four new antagonists (2-5) of GnRH homologues to Azaline B (1), with a common core sequence of [Aph(X)5, D-Aph(Cbm)6]Azaline B. In these analogs, (X) contains hydrophobic aromatic moieties (like homoveratoyl in 2, homovanillyl in 3, 2,5-dimethoxyphenylacetyl in 4, and 3,5-dimethoxyphenylacetyl in 5) designed to improve the duration of action over that of Azaline B. These analogs were tested in vitro for their ability to antagonize the GnRH receptor and in vivo for duration of action in a castrated male rat assay. Analogs 2, 4, and 5 were potent in vitro, but were found to be short acting in vivo. However, analog 3 [Aph(Hvn)5,D-Aph(Cbm)6]Azaline B is a potent human GnRH receptor antagonist in vitro (IC50 1.47 nM) and exhibits a longer duration of action than azaline B.


Subject(s)
Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Amino Acid Substitution , Animals , Cell Line , Gonadotropin-Releasing Hormone/chemical synthesis , Gonadotropin-Releasing Hormone/chemistry , Gonadotropin-Releasing Hormone/pharmacology , Humans , Male , Molecular Structure , Rats , Receptors, LHRH/drug effects , Recombinant Proteins/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...