Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(30): e2201067119, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35858423

ABSTRACT

The C-natriuretic peptide (CNP) analog vosoritide has recently been approved for treatment of achondroplasia in children. However, the regimen requires daily subcutaneous injections in pediatric patients over multiple years. The present work sought to develop a long-acting CNP that would provide efficacy equal to or greater than that of vosoritide but require less frequent injections. We used a technology for half-life extension, whereby a drug is attached to tetra-polyethylene glycol hydrogels (tetra-PEG) by ß-eliminative linkers that cleave at predetermined rates. These hydrogels-fabricated as uniform ∼60-µm microspheres-are injected subcutaneously, where they serve as a stationary depot to slowly release the drug into the systemic circulation. We prepared a highly active, stable CNP analog-[Gln6,14]CNP-38-composed of the 38 C-terminal amino acids of human CNP-53 containing Asn to Gln substitutions to preclude degradative deamidation. Two microsphere [Gln6,14]CNP-38 conjugates were prepared, with release rates designed to allow once-weekly and once-monthly administration. After subcutaneous injection of the conjugates in mice, [Gln6,14]CNP-38 was slowly released into the systemic circulation and showed biphasic elimination pharmacokinetics with terminal half-lives of ∼200 and ∼600 h. Both preparations increased growth of mice comparable to or exceeding that produced by daily vosoritide. Simulations of the pharmacokinetics in humans indicated that plasma [Gln6,14]CNP-38 levels should be maintained within a therapeutic window over weekly, biweekly, and likely, monthly dosing intervals. Compared with vosoritide, which requires ∼30 injections per month, microsphere [Gln6,14]CNP-38 conjugates-especially the biweekly and monthly dosing-could provide an alternative that would be well accepted by physicians, patients, and patient caregivers.


Subject(s)
Achondroplasia , Drug Development , Natriuretic Peptide, C-Type , Achondroplasia/drug therapy , Animals , Child , Delayed-Action Preparations , Humans , Hydrogels/chemistry , Injections, Subcutaneous , Mice , Microspheres , Natriuretic Peptide, C-Type/administration & dosage , Natriuretic Peptide, C-Type/analogs & derivatives , Natriuretic Peptide, C-Type/chemical synthesis , Natriuretic Peptide, C-Type/pharmacokinetics
2.
Bioconjug Chem ; 32(4): 794-800, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33822591

ABSTRACT

ß-Elimination of drugs tethered to macromolecular carbamates provides a platform for drug half-life extension. However, the macromolecular Michael acceptor products formed upon drug release can potentially react with biological amines and thiols and may raise concerns about safety. We desired to mitigate this possibility by developing linkers that have predictable rates of ß-elimination but suppressed rates of nucleophilic addition to their Michael acceptor products. We prepared Michael acceptor products of ß-eliminative linkers that contained a methyl group at the Cß carbon or a gem-dimethyl group at the Cγ carbon and studied the kinetics of their reactions with the most prevalent biological nucleophiles-amine and thiol groups. Aza-Michael reactions with glycine are slowed about 20-fold by methylation of the ß-carbon and 175-fold with a gem-dimethyl group at the γ-carbon. Likewise, addition of the glutathione thiol to γ-gem-dimethyl Michael acceptors was retarded 7-24-fold compared to parent unsubstituted linkers. It was estimated that in an in vivo environment of ∼0.5 mM macromolecular thiols or ∼20 mM macromolecular amines-as in plasma-the reaction half-life of a typical Michael acceptor with a γ-gem-dimethyl linker could exceed 3 years for thiols or 25 years for amines. We also prepared a large series of γ-gem-dimethyl ß-eliminative linkers and showed excellent structure-activity relationships of elimination rates with corresponding unsubstituted parent linkers. Finally, we compared the first-generation unsubstituted and new gem-dimethyl ß-eliminative linkers in a once-monthly drug delivery system of a 39 amino acid peptide. Both linkers provided the desired half-life extension of the peptide, but the Michael acceptor formed from the gem-dimethyl linker was much less reactive. We conclude that the γ-gem-dimethyl ß-eliminative linkers provide high flexibility and greatly reduce potential reactions of Michael acceptor products with biologically important nucleophiles.


Subject(s)
Pharmaceutical Preparations/chemistry , Carbamates/chemistry , Drug Delivery Systems , Drug Liberation , Half-Life , Kinetics , Structure-Activity Relationship
3.
Sci Rep ; 8(1): 10705, 2018 Jul 16.
Article in English | MEDLINE | ID: mdl-30013126

ABSTRACT

Human KLK8/neuropsin, a kallikrein-related serine peptidase, is mostly expressed in skin and the hippocampus regions of the brain, where it regulates memory formation by synaptic remodeling. Substrate profiles of recombinant KLK8 were analyzed with positional scanning using fluorogenic tetrapeptides and the proteomic PICS approach, which revealed the prime side specificity. Enzyme kinetics with optimized substrates showed stimulation by Ca2+ and inhibition by Zn2+, which are physiological regulators. Crystal structures of KLK8 with a ligand-free active site and with the inhibitor leupeptin explain the subsite specificity and display Ca2+ bound to the 75-loop. The variants D70K and H99A confirmed the antagonistic role of the cation binding sites. Molecular docking and dynamics calculations provided insights in substrate binding and the dual regulation of activity by Ca2+ and Zn2+, which are important in neuron and skin physiology. Both cations participate in the allosteric surface loop network present in related serine proteases. A comparison of the positional scanning data with substrates from brain suggests an adaptive recognition by KLK8, based on the tertiary structures of its targets. These combined findings provide a comprehensive picture of the molecular mechanisms underlying the enzyme activity of KLK8.


Subject(s)
Kallikreins/metabolism , Allosteric Regulation/drug effects , Allosteric Site/genetics , Calcium/metabolism , Cations, Divalent/metabolism , Crystallography, X-Ray , Kallikreins/antagonists & inhibitors , Kallikreins/genetics , Kallikreins/ultrastructure , Kinetics , Leupeptins/pharmacology , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Structure-Activity Relationship , Substrate Specificity/drug effects , Zinc/metabolism
4.
J Control Release ; 278: 74-79, 2018 05 28.
Article in English | MEDLINE | ID: mdl-29577950

ABSTRACT

We have developed a chemically-controlled drug delivery system in which a drug is covalently attached via a carbamate to hydrogel microspheres using a ß-eliminative linker; rate-determining proton removal from a CH bond adjacent to an electron withdrawing group results in a ß-elimination to cleave the carbamate and release the drug. After subcutaneous injection of the hydrogel-drug conjugate, the drug is slowly released into the systemic circulation and acquires an elimination t1/2,ß that matches the t1/2 of linker cleavage. A similar ß-eliminative linker with a slower cleavage rate is installed into crosslinks of the polymer to trigger gel degradation after drug release. We have now prepared ß-eliminative linkers that contain deuterium in place of the hydrogen whose removal initiates cleavage. In vitro model systems of drug release and degelation show large primary deuterium kinetic isotope effects of kH/kD ~ 2.5 to 3.5. Using a deuterated linker to attach the peptide octreotide to hydrogel-microspheres, the in vivo t1/2,ß of the drug was increased from ~1.5 to 4.5 weeks in the rat. Similarly, the in vivo time to biodegradation of hydrogels with deuterium-containing crosslinks could be extended ~2.5-fold compared to hydrogen-containing counterparts. Thus, the use of primary deuterium kinetic isotope effects in a single platform technology can control rates of ß-elimination reactions in drug release and polymer biodegradation rates.


Subject(s)
Deuterium/chemistry , Drug Delivery Systems , Octreotide/administration & dosage , Polymers/chemistry , Animals , Cross-Linking Reagents/chemistry , Drug Carriers/chemistry , Drug Liberation , Half-Life , Hydrogels , Octreotide/chemistry , Octreotide/pharmacokinetics , Rats
5.
Thromb Haemost ; 117(9): 1750-1760, 2017 08 30.
Article in English | MEDLINE | ID: mdl-28726978

ABSTRACT

Factor VII (FVII) activating protease (FSAP) is a circulating serine protease that is likely to be involved in a number of disease conditions such as stroke, atherosclerosis, liver fibrosis, thrombosis and cancer. To date, no systematic information is available about the substrate specificity of FSAP. Applying phage display and positional scanning substrate combinatorial library (PS-SCL) approaches we have characterised the specificity of FSAP towards small peptides. Results were evaluated in the context of known protein substrates as well as molecular modelling of the peptides in the active site of FSAP. The representative FSAP-cleaved sequence obtained from the phage display method was Val-Leu-Lys-Arg-Ser (P4-P1'). The sequence X-Lys/Arg-Nle-Lys/Arg (P4-P1) was derived from the PS-SCL method. These results show a predilection for cleavage at a cluster of basic amino acids on the nonprime side. Quenched fluorescent substrate (Ala-Lys-Nle-Arg-AMC) (amino methyl coumarin) and (Ala-Leu-Lys-Arg-AMC) had a higher selectivity for FSAP compared to other proteases from the hemostasis system. These substrates could be used to measure FSAP activity in a complex biological system such as plasma. In histone-treated plasma there was a specific activation of pro-FSAP as validated by the use of an FSAP inhibitory antibody, corn trypsin inhibitor to inhibit Factor XIIa and hirudin to inhibit thrombin, which may account for some of the haemostasis-related effects of histones. These results will aid the development of further selective FSAP activity probes as well as specific inhibitors that will help to increase the understanding of the functions of FSAP in vivo.


Subject(s)
Peptides/metabolism , Serine Endopeptidases/metabolism , Antithrombins/pharmacology , Catalytic Domain , Cell Surface Display Techniques , Enzyme Activation , Hirudins/pharmacology , Histones/metabolism , Humans , Kinetics , Molecular Docking Simulation , Peptide Library , Peptides/chemistry , Peptides/genetics , Plant Proteins/chemistry , Plant Proteins/pharmacology , Protein Binding , Protein Conformation , Serine Endopeptidases/blood , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Structure-Activity Relationship , Substrate Specificity , Thrombin/antagonists & inhibitors , Thrombin/metabolism
6.
ACS Chem Biol ; 12(8): 2107-2116, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28605180

ABSTRACT

We have developed a chemically controlled very long-acting delivery system to support once-monthly administration of a peptidic GLP-1R agonist. Initially, the prototypical GLP-1R agonist exenatide was covalently attached to hydrogel microspheres by a self-cleaving ß-eliminative linker; after subcutaneous injection in rats, the peptide was slowly released into the systemic circulation. However, the short serum exenatide half-life suggested its degradation in the subcutaneous depot. We found that exenatide undergoes deamidation at Asn28 with an in vitro and in vivo half-life of approximately 2 weeks. The [Gln28]exenatide variant and exenatide showed indistinguishable GLP-1R agonist activities as well as pharmacokinetic and pharmacodynamic effects in rodents; however, unlike exenatide, [Gln28]exenatide is stable for long periods. Two different hydrogel-[Gln28]exenatide conjugates were prepared using ß-eliminative linkers with different cleavage rates. After subcutaneous injection in rodents, the serum half-lives for the released [Gln28]exenatide from the two conjugates were about 2 weeks and one month. Two monthly injections of the latter in the Zucker diabetic fatty rat showed pharmacodynamic effects indistinguishable from two months of continuously infused exenatide. Pharmacokinetic simulations indicate that the delivery system should serve well as a once-monthly GLP-1R agonist for treatment of type 2 diabetes in humans.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Drug Delivery Systems , Glucagon-Like Peptide-1 Receptor/agonists , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Hypoglycemic Agents/administration & dosage , Microspheres , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/administration & dosage , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Molecular Structure , Time Factors
7.
Bioconjug Chem ; 27(10): 2534-2539, 2016 Oct 19.
Article in English | MEDLINE | ID: mdl-27657443

ABSTRACT

The utility of antigen-binding antibody fragments is often limited by their short half-lives. Half-life extension of such fragments is usually accomplished by attachment or binding to high-molecular-weight carriers that reduce the renal elimination rate. However, the higher hydrodynamic radius results in greater confinement in the vascular compartment and, thus, lower tissue distribution. We have developed a chemically controlled drug delivery system in which the drug is covalently attached to hydrogel microspheres by a self-cleaving ß-eliminative linker; upon subcutaneous injection, the t1/2,ß of the released drug acquires the t1/2 of linker cleavage. In the present work, we compared the pharmacokinetics of an anti-TNFα scFv, the same scFv attached to 40 kDa PEG by a stable linker, and the scFv attached to hydrogel microspheres by a self-cleaving linker. We also developed a general approach for the selective attachment of ß-eliminative linkers to the N-termini of proteins. In rats, the scFv had a t1/2,ß of 4 h and a high volume of distribution at steady state (Vd,SS), suggesting extensive tissue distribution. The PEG-scFv conjugate had an increased t1/2,ß of about 2 days but showed a reduced Vd,SS that was similar to the plasma volume. In contrast, the tissue-penetrable scFv released from the hydrogel system had a t1/2,ß of about 2 weeks. Thus, the cleavable microsphere-scFv conjugate releases its protein cargo with a prolonged half-life comparable to that of most full-length mAbs and in a form that has the high tissue distribution characteristic of smaller mAb fragments. Other antigen-binding antibody fragments should be amenable to the half-life extension approach described here.

8.
Bioconjug Chem ; 27(7): 1638-44, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27253622

ABSTRACT

We developed a long-acting drug-delivery system that supports subcutaneous administration of the peptidic somatostatin agonist octreotide-a blockbuster drug used to treat acromegaly and neuroendocrine tumors. The current once-a-month polymer-encapsulated octreotide, Sandostatin LAR, requires a painful intragluteal injection through a large needle by a health-care professional. To overcome such shortcomings, Tetra-PEG hydrogel microspheres were covalently attached to the α-amine of d-Phe(1) or the ε-amine of Lys(5) of octreotide by a self-cleaving ß-eliminative linker; upon subcutaneous injection in the rat using a small-bore needle, octreotide was slowly released. The released drug from the ε-octreotide conjugate showed a remarkably long serum half-life that exceeded two months. The α-octreotide conjugate had a half-life of ∼2 weeks, and showed an excellent correlation of in vitro and in vivo drug release. Pharmacokinetic models indicate these microspheres should support once-weekly to once-monthly self-administered subcutaneous dosing in humans. The hydrogel-octreotide conjugate shows the favorable pharmacokinetics of Sandostatin LAR without its drawbacks.


Subject(s)
Drug Carriers/chemistry , Hydrogels/chemistry , Octreotide/administration & dosage , Octreotide/chemistry , Animals , Delayed-Action Preparations , Injections, Subcutaneous , Microspheres , Polyethylene Glycols/chemistry , Rats
9.
Bioconjug Chem ; 27(5): 1210-5, 2016 05 18.
Article in English | MEDLINE | ID: mdl-26930186

ABSTRACT

We have developed a unique long-acting drug-delivery system for the GLP-1 agonist exenatide. The peptide was covalently attached to Tetra-PEG hydrogel microspheres by a cleavable ß-eliminative linker; upon s.c. injection, the exenatide is slowly released at a rate dictated by the linker. A second ß-eliminative linker with a slower cleavage rate was incorporated in polymer cross-links to trigger gel degradation after drug release. The uniform 40 µm microspheres were fabricated using a flow-focusing microfluidic device and in situ polymerization within droplets. The exenatide-laden microspheres were injected subcutaneously into the rat, and serum exenatide measured over a one-month period. Pharmacokinetic analysis showed a t1/2,ß of released exenatide of about 7 days which represents over a 300-fold half-life extension in the rat and exceeds the half-life of any currently approved long-acting GLP-1 agonist. Hydrogel-exenatide conjugates gave an excellent Level A in vitro-in vivo correlation of release rates of the peptide from the gel, and indicated that exenatide release was 3-fold faster in vivo than in vitro. Pharmacokinetic simulations indicate that the hydrogel-exenatide microspheres should support weekly or biweekly subcutaneous dosing in humans. The rare ability to modify in vivo pharmacokinetics by the chemical nature of the linker indicates that an even longer acting exenatide is feasible.


Subject(s)
Drug Carriers/chemistry , Hydrogels/chemistry , Peptides/administration & dosage , Peptides/chemistry , Venoms/administration & dosage , Venoms/chemistry , Animals , Drug Administration Schedule , Exenatide , Humans , Microspheres , Models, Molecular , Molecular Conformation , Peptides/pharmacokinetics , Polyethylene Glycols/chemistry , Rats , Venoms/pharmacokinetics
10.
Eur J Pharm Biopharm ; 93: 254-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25900863

ABSTRACT

We have previously developed a linker technology for half-life extension of peptides, proteins and small molecule drugs (1). The linkers undergo ß-elimination reactions with predictable cleavage rates to release the native drug. Here we utilize this technology for half-life extension of the 38 amino acid HIV-1 fusion inhibitor TRI-1144. Conjugation of TRI-1144 to 40 kDa PEG by an appropriate ß-eliminative linker and i.v. administration of the conjugate increased the in vivo half-life of the released peptide from 4 to 34 h in the rat, and the pharmacokinetic parameters were in excellent accord with a one-compartment model. From these data we simulated the pharmacokinetics of the PEG-TRI-1144 conjugate in humans, predicting a t1/2,ß of 70 h for the released peptide, and that a serum concentration of 25 nM could be maintained by weekly doses of 8 µmol of the conjugate. Using a non-circulating carrier (2) similar simulations indicated a t1/2,ß of 150 h for the peptide released from the conjugate and that dosing of only 1.8 µmol/week could maintain serum concentrations of TRI-1144 above 25 nM. Hence, releasable ß-eliminative linkers provide significant half-life extension to TRI-1144 and would be expected to do likewise for related peptides.


Subject(s)
HIV Envelope Protein gp41/pharmacokinetics , HIV Fusion Inhibitors/pharmacokinetics , Peptide Fragments/pharmacokinetics , Polyethylene Glycols/pharmacokinetics , Prodrugs/pharmacokinetics , Administration, Intravenous , Animals , Chemistry, Pharmaceutical , Computer Simulation , HIV Envelope Protein gp41/administration & dosage , HIV Envelope Protein gp41/blood , HIV Envelope Protein gp41/chemical synthesis , HIV Fusion Inhibitors/administration & dosage , HIV Fusion Inhibitors/blood , HIV Fusion Inhibitors/chemical synthesis , Half-Life , Hydrolysis , Male , Metabolic Clearance Rate , Models, Biological , Peptide Fragments/administration & dosage , Peptide Fragments/blood , Peptide Fragments/chemical synthesis , Peptides , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemical synthesis , Prodrugs/administration & dosage , Prodrugs/chemical synthesis , Rats, Sprague-Dawley , Technology, Pharmaceutical/methods
11.
Cancer Discov ; 4(11): 1310-25, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25122198

ABSTRACT

UNLABELLED: TMPRSS2 is an androgen-regulated cell-surface serine protease expressed predominantly in prostate epithelium. TMPRSS2 is expressed highly in localized high-grade prostate cancers and in the majority of human prostate cancer metastases. Through the generation of mouse models with a targeted deletion of Tmprss2, we demonstrate that the activity of this protease regulates cancer cell invasion and metastasis to distant organs. By screening combinatorial peptide libraries, we identified a spectrum of TMPRSS2 substrates that include pro-hepatocyte growth factor (HGF). HGF activated by TMPRSS2 promoted c-MET receptor tyrosine kinase signaling, and initiated a proinvasive epithelial-to-mesenchymal transition phenotype. Chemical library screens identified a potent bioavailable TMPRSS2 inhibitor that suppressed prostate cancer metastasis in vivo. Together, these findings provide a mechanistic link between androgen-regulated signaling programs and prostate cancer metastasis that operate via context-dependent interactions with extracellular constituents of the tumor microenvironment. SIGNIFICANCE: The vast majority of prostate cancer deaths are due to metastasis. Loss of TMPRSS2 activity dramatically attenuated the metastatic phenotype through mechanisms involving the HGF-c-MET axis. Therapeutic approaches directed toward inhibiting TMPRSS2 may reduce the incidence or progression of metastasis in patients with prostate cancer.


Subject(s)
Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Serine Endopeptidases/metabolism , Androgens/metabolism , Animals , Cell Line, Tumor , Hepatocyte Growth Factor/metabolism , Humans , Male , Mice, Knockout , Mice, SCID , Peptide Library , Proteolysis , Proto-Oncogene Proteins c-met/metabolism , Receptors, Androgen/metabolism , Tumor Microenvironment
12.
J Med Chem ; 57(6): 2303-14, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24494988

ABSTRACT

We have recently reported a chemical approach for half-life extension that utilizes ß-eliminative linkers to attach amine-containing drugs or prodrugs to macromolecules. The linkers release free drug or prodrug over periods ranging from a few hours to over 1 year. We adapted these linkers for use with phenol-containing drugs. Here, we prepared PEG conjugates of the irinotecan (CPT-11) active metabolite SN-38 via a phenyl ether that release the drug with predictable long half-lives. Pharmacokinetic studies in the rat indicate that, in contrast to other SN-38 prodrugs, the slowly released SN-38 shows a very low C(max), is kept above target concentrations for extended periods, and forms very little SN-38 glucuronide (the precursor of enterotoxic SN-38). The low SN-38 glucuronide is attributed to low hepatic uptake of SN-38. These macromolecular prodrugs have unique pharmacokinetic profiles that may translate to less intestinal toxicity and interpatient variability than the SN-38 prodrugs thus far studied.


Subject(s)
Camptothecin/analogs & derivatives , Glucuronides/chemical synthesis , Glucuronides/pharmacology , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Amines/chemistry , Animals , Camptothecin/chemical synthesis , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Chromatography, High Pressure Liquid , Delayed-Action Preparations , Female , Glucuronides/metabolism , Glucuronides/pharmacokinetics , Half-Life , Kinetics , Liver/metabolism , Macromolecular Substances , Phenols/chemistry , Prodrugs/pharmacokinetics , Rats , Rats, Sprague-Dawley
13.
Bioconjug Chem ; 24(12): 1990-7, 2013 Dec 18.
Article in English | MEDLINE | ID: mdl-24171387

ABSTRACT

We recently reported a chemical approach for half-life extension that utilizes sets of releasable linkers to attach drugs to macromolecules via a cleavable carbamate group (Santi et al., Proc. Nat. Acad. Sci. U.S.A. 2012, 109, 6211-6216). The linkers undergo a ß-elimination cleavage to release the free, native amine-containing drug. A limitation of the technology is the requirement for an amino group on the drug in order to form the carbamate bond, since most small molecules do not have an amine functional group. Here, we describe an approach to adapt these same ß-elimination carbamate linkers so they can be used to connect other acidic heteroatoms, in particular, phenolic hydroxyl groups. The approach utilizes a methylene adaptor to connect the drug to the carbamate nitrogen, and an electron-withdrawing group attached to carbamate nitrogen to stabilize the system against a pH-independent spontaneous cleavage. Carbamate cleavage is driven by ß-elimination to give a carboxylated aryl amino Mannich base which rapidly collapses to give the free drug, an aryl amine, and formaldehyde.


Subject(s)
Macromolecular Substances/chemistry , Nitrophenols/chemistry , Carbamates/chemistry , Drug Design , Kinetics , Polyethylene Glycols/chemistry , Solvents/chemistry , Structure-Activity Relationship
14.
J Biol Chem ; 288(15): 10588-98, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23447538

ABSTRACT

Human and mouse marapsins (Prss27) are serine proteases preferentially expressed by stratified squamous epithelia. However, mouse marapsin contains a transmembrane anchor absent from the human enzyme. To gain insights into physical forms, activities, inhibition, and roles in epithelial differentiation, we traced tail loss in human marapsin to a nonsense mutation in an ancestral ape, compared substrate preferences of mouse and human marapsins with those of the epithelial peptidase prostasin, designed a selective substrate and inhibitor, and generated Prss27-null mice. Phylogenetic analysis predicts that most marapsins are transmembrane proteins. However, nonsense mutations caused membrane anchor loss in three clades: human/bonobo/chimpanzee, guinea pig/degu/tuco-tuco/mole rat, and cattle/yak. Most marapsin-related proteases, including prostasins, are type I transmembrane proteins, but the closest relatives (prosemins) are not. Soluble mouse and human marapsins are tryptic with subsite preferences distinct from those of prostasin, lack general proteinase activity, and unlike prostasins resist antiproteases, including leupeptin, aprotinin, serpins, and α2-macroglobulin, suggesting the presence of non-canonical active sites. Prss27-null mice develop normally in barrier conditions and are fertile without overt epithelial defects, indicating that marapsin does not play critical, non-redundant roles in development, reproduction, or epithelial differentiation. In conclusion, marapsins are conserved, inhibitor-resistant, tryptic peptidases. Although marapsins are type I transmembrane proteins in their typical form, they mutated independently into anchorless forms in several mammalian clades, including one involving humans. Similar pathways appear to have been traversed by prosemins and tryptases, suggesting that mutational tail loss is an important means of evolving new functions of tryptic serine proteases from transmembrane ancestors.


Subject(s)
Evolution, Molecular , Membrane Proteins/genetics , Membrane Proteins/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Animals , CHO Cells , Cattle , Cricetinae , Cricetulus , Guinea Pigs , Humans , Membrane Proteins/antagonists & inhibitors , Mice , Mice, Mutant Strains , Mole Rats , Mutation , Pan paniscus , Pan troglodytes , Protease Inhibitors/pharmacology , Rats , Solubility , Species Specificity
15.
Proc Natl Acad Sci U S A ; 109(16): 6211-6, 2012 Apr 17.
Article in English | MEDLINE | ID: mdl-22474378

ABSTRACT

Conjugation to macromolecular carriers is a proven strategy for improving the pharmacokinetics of drugs, with many stable polyethylene glycol conjugates having reached the market. Stable conjugates suffer several limitations: loss of drug potency due to conjugation, confining the drug to the extracellular space, and the requirement for a circulating conjugate. Current research is directed toward overcoming such limitations through releasable conjugates in which the drug is covalently linked to the carrier through a cleavable linker. Satisfactory linkers that provide predictable cleavage rates tunable over a wide time range that are useful for both circulating and noncirculating conjugates are not yet available. We describe such conjugation linkers on the basis of a nonenzymatic ß-elimination reaction with preprogrammed, highly tunable cleavage rates. A set of modular linkers is described that bears a succinimidyl carbonate group for attachment to an amine-containing drug or prodrug, an azido group for conjugation to the carrier, and a tunable modulator that controls the rate of ß-eliminative cleavage. The linkers provide predictable, tunable release rates of ligands from macromolecular conjugates both in vitro and in vivo, with half-lives spanning from a range of hours to >1 y at physiological pH. A circulating PEG conjugate achieved a 56-fold half-life extension of the 39-aa peptide exenatide in rats, and a noncirculating s.c. hydrogel conjugate achieved a 150-fold extension. Using slow-cleaving linkers, the latter may provide a generic format for once-a-month dosage forms of potent drugs. The releasable linkers provide additional benefits that include lowering C(max) and pharmacokinetic coordination of drug combinations.


Subject(s)
Delayed-Action Preparations/pharmacokinetics , Macromolecular Substances/chemistry , Polyethylene Glycols/chemistry , Prodrugs/pharmacokinetics , Algorithms , Animals , Carbonates/chemistry , Chromatography, High Pressure Liquid , Delayed-Action Preparations/chemistry , Exenatide , Half-Life , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacokinetics , Kinetics , Male , Mice , Models, Biological , Models, Chemical , Molecular Structure , Peptides/chemistry , Peptides/pharmacokinetics , Prodrugs/chemistry , Rats, Sprague-Dawley , Succinimides/chemistry , Venoms/chemistry , Venoms/pharmacokinetics
16.
J Mol Biol ; 415(4): 699-715, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22154938

ABSTRACT

The type II transmembrane serine protease family consists of 18 closely related serine proteases that are implicated in multiple functions. To identify selective, inhibitory antibodies against one particular type II transmembrane serine protease, matriptase [MT-SP1 (membrane-type serine protease 1)], a phage display library was created with a natural repertoire of Fabs [fragment antigen binding (Fab)] from human naïve B cells. Fab A11 was identified with a 720 pM inhibition constant and high specificity for matriptase over other trypsin-fold serine proteases. A Trichoderma reesei system expressed A11 with a yield of ∼200 mg/L. The crystal structure of A11 in complex with matriptase has been determined and compared to the crystal structure of another antibody inhibitor (S4) in complex with matriptase. Previously discovered from a synthetic single-chain variable fragment library, S4 is also a highly selective and potent matriptase inhibitor. The crystal structures of the A11/matriptase and S4/matriptase complexes were solved to 2.1 Å and 1.5 Å, respectively. Although these antibodies, discovered from separate libraries, interact differently with the protease surface loops for their specificity, the structures reveal a similar novel mechanism of protease inhibition. Through the insertion of the H3 variable loop in a reverse orientation at the substrate-binding pocket, these antibodies bury a large surface area for potent inhibition and avoid proteolytic inactivation. This discovery highlights the critical role that the antibody scaffold plays in positioning loops to bind and inhibit protease function in a highly selective manner. Additionally, Fab A11 is a fully human antibody that specifically inhibits matriptase over other closely related proteases, suggesting that this approach could be useful for clinical applications.


Subject(s)
Antibodies/chemistry , Antibodies/pharmacology , Peptide Hydrolases/chemistry , Peptide Hydrolases/immunology , Protein Interaction Domains and Motifs/physiology , Amino Acid Sequence , Antibodies/isolation & purification , Antibodies/metabolism , Catalysis/drug effects , Catalytic Domain/genetics , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/isolation & purification , Immunoglobulin Fab Fragments/pharmacology , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Peptide Library , Protease Inhibitors/chemistry , Sequence Homology, Amino Acid , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Serine Endopeptidases/metabolism
17.
J Biol Chem ; 285(24): 18516-27, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20378535

ABSTRACT

Entamoeba histolytica cysteine proteinases (EhCPs) play a key role in disrupting the colonic epithelial barrier and the innate host immune response during invasion of E. histolytica, the protozoan cause of human amebiasis. EhCPs are encoded by 50 genes, of which ehcp4 (ehcp-a4) is the most up-regulated during invasion and colonization in a mouse cecal model of amebiasis. Up-regulation of ehcp4 in vivo correlated with our finding that co-culture of E. histolytica trophozoites with mucin-producing T84 cells increased ehcp4 expression up to 6-fold. We have expressed recombinant EhCP4, which was autocatalytically activated at acidic pH but had highest proteolytic activity at neutral pH. In contrast to the other amebic cysteine proteinases characterized so far, which have a preference for arginine in the P2 position, EhCP4 displayed a unique preference for valine and isoleucine at P2. This preference was confirmed by homology modeling, which revealed a shallow, hydrophobic S2 pocket. Endogenous EhCP4 localized to cytoplasmic vesicles, the nuclear region, and perinuclear endoplasmic reticulum (ER). Following co-culture with colonic cells, EhCP4 appeared in acidic vesicles and was released extracellularly. A specific vinyl sulfone inhibitor, WRR605, synthesized based on the substrate specificity of EhCP4, inhibited the recombinant enzyme in vitro and significantly reduced parasite burden and inflammation in the mouse cecal model. The unique expression pattern, localization, and biochemical properties of EhCP4 could be exploited as a potential target for drug design.


Subject(s)
Amebiasis/parasitology , Cysteine Proteases/chemistry , Cysteine Proteases/physiology , Entamoeba histolytica/metabolism , Animals , Cell Line, Tumor , Drug Design , Gene Expression Regulation, Enzymologic , Humans , Hydrogen-Ion Concentration , Kinetics , Mass Spectrometry/methods , Mice , Mice, Inbred C3H , Peptide Hydrolases/chemistry , Protease Inhibitors/pharmacology , Recombinant Proteins/chemistry , Thioredoxins/chemistry
18.
Cancer Res ; 70(4): 1505-12, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20145119

ABSTRACT

The cell surface protease membrane-type serine protease-1 (MT-SP1), also known as matriptase, is often upregulated in epithelial cancers. We hypothesized that dysregulation of MT-SP1 with regard to its cognate inhibitor hepatocyte growth factor activator inhibitor-1 (HAI-1), a situation that increases proteolytic activity, might be exploited for imaging purposes to differentiate malignant from normal tissue. In this study, we show that MT-SP1 is active on cancer cells and that its activity may be targeted in vivo for tumor detection. A proteolytic activity assay with several MT-SP1-positive human cancer cell lines showed that MT-SP1 antibodies that inhibit recombinant enzyme activity in vitro also bind and inhibit the full-length enzyme expressed on cells. In contrast, in the same assay, MT-SP1-negative cancer cell lines were inactive. Fluorescence microscopy confirmed the cell surface localization of labeled antibodies bound to MT-SP1-positive cells. To evaluate in vivo targeting capability, 0.7 to 2 nmoles of fluorescently labeled antibodies were administered to mice bearing tumors that were positive or negative for MT-SP1. Antibodies localized to MT-SP1-positive tumors (n = 3), permitting visualization of MT-SP1 activity, whereas MT-SP1-negative tumors (n = 2) were not visualized. Our findings define MT-SP1 activity as a useful biomarker to visualize epithelial cancers using a noninvasive antibody-based method.


Subject(s)
Diagnostic Imaging/methods , Neoplasms/diagnosis , Protein Processing, Post-Translational , Serine Endopeptidases/metabolism , Animals , Antibodies , Antibody Specificity , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Female , Fluorescent Antibody Technique/methods , HT29 Cells , Humans , Mice , Mice, Nude , Neoplasms/metabolism , Protein Processing, Post-Translational/drug effects , Serine Endopeptidases/immunology , Transplantation, Heterologous , Tumor Cells, Cultured
19.
PLoS Negl Trop Dis ; 3(6): e449, 2009 Jun 02.
Article in English | MEDLINE | ID: mdl-19488406

ABSTRACT

BACKGROUND: Blood flukes of the genus Schistosoma are platyhelminth parasites that infect 200 million people worldwide. Digestion of nutrients from the host bloodstream is essential for parasite development and reproduction. A network of proteolytic enzymes (proteases) facilitates hydrolysis of host hemoglobin and serum proteins. METHODOLOGY/PRINCIPAL FINDINGS: We identified a new cathepsin L termed SmCL3 using PCR strategies based on S. mansoni EST sequence data. An ortholog is present in Schistosoma japonicum. SmCL3 was heterologously expressed as an active enzyme in the yeast, Pichia pastoris. Recombinant SmCL3 has a broad pH activity range against peptidyl substrates and is inhibited by Clan CA protease inhibitors. Consistent with a function in degrading host proteins, SmCL3 hydrolyzes serum albumin and hemoglobin, is localized to the adult gastrodermis, and is expressed mainly in those life stages infecting the mammalian host. The predominant form of SmCL3 in the parasite exists as a zymogen, which is unusual for proteases. This zymogen includes an unusually long prodomain with alpha helical secondary structure motifs. The striking specificity of SmCL3 for amino acids with large aromatic side chains (Trp and Tyr) at the P2 substrate position, as determined with positional scanning-synthetic combinatorial library, is consistent with a molecular model that shows a large and deep S2 pocket. A sequence similarity network (SSN) view clusters SmCL3 and other cathepsins L in accordance with previous large-scale phylogenetic analyses that identify six super kingdoms. CONCLUSIONS/SIGNIFICANCE: SmCL3 is a gut-associated cathepsin L that may contribute to the network of proteases involved in degrading host blood proteins as nutrients. Furthermore, this enzyme exhibits some unusual sequence and biophysical features that may result in additional functions. The visualization of network inter-relationships among cathepsins L suggests that these enzymes are suitable 'marker sequences' for inclusion in future phylogenetic analyses.


Subject(s)
Cathepsin L/isolation & purification , Cathepsin L/metabolism , Schistosoma mansoni/enzymology , Amino Acid Sequence , Animals , Cathepsin L/genetics , Cricetinae , DNA, Protozoan/genetics , Enzyme Stability , Expressed Sequence Tags , Gastrointestinal Tract/chemistry , Gene Expression , Hemoglobins/metabolism , Humans , Hydrogen-Ion Concentration , Models, Molecular , Molecular Sequence Data , Phylogeny , Pichia/genetics , Polymerase Chain Reaction/methods , Protease Inhibitors/pharmacology , Schistosoma mansoni/genetics , Sequence Homology, Amino Acid , Serum Albumin/metabolism , Snails , Substrate Specificity
20.
Methods Mol Biol ; 539: 59-78, 2009.
Article in English | MEDLINE | ID: mdl-19377970

ABSTRACT

Determining the preferred substrate cleavage sequence of proteases is an important step toward understanding their roles in cancer development and progression. Knowledge of this sequence can aid in the design of new experimental tools for study as well as aid in the identification of endogenous protease substrates and signaling pathways. Various investigators have demonstrated a number of techniques to uncover these sequences, but most can be very time consuming. We have designed and successfully implemented a complete diverse ACC tetrapeptide positional scanning synthetic combinatorial library that allows for the rapid screening of proteases to determine their preferred residues at positions P1-P4. These sequences can be readily verified through kinetic measurements on single peptide substrates and utilized to further knowledge of the role of proteases in cancer.


Subject(s)
Coumarins/chemical synthesis , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Peptide Library , Peptides/chemistry , Peptides/metabolism , Amino Acid Sequence , Kinetics , Peptides/chemical synthesis , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...