Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(30): e2201067119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35858423

RESUMEN

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.


Asunto(s)
Acondroplasia , Desarrollo de Medicamentos , Péptido Natriurético Tipo-C , Acondroplasia/tratamiento farmacológico , Animales , Niño , Preparaciones de Acción Retardada , Humanos , Hidrogeles/química , Inyecciones Subcutáneas , Ratones , Microesferas , Péptido Natriurético Tipo-C/administración & dosificación , Péptido Natriurético Tipo-C/análogos & derivados , Péptido Natriurético Tipo-C/síntesis química , Péptido Natriurético Tipo-C/farmacocinética
2.
Bioconjug Chem ; 32(4): 794-800, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33822591

RESUMEN

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.


Asunto(s)
Preparaciones Farmacéuticas/química , Carbamatos/química , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Semivida , Cinética , Relación Estructura-Actividad
3.
Bioconjug Chem ; 27(7): 1638-44, 2016 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-27253622

RESUMEN

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.


Asunto(s)
Portadores de Fármacos/química , Hidrogeles/química , Octreótido/administración & dosificación , Octreótido/química , Animales , Preparaciones de Acción Retardada , Inyecciones Subcutáneas , Microesferas , Polietilenglicoles/química , Ratas
4.
Bioconjug Chem ; 27(5): 1210-5, 2016 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-26930186

RESUMEN

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.


Asunto(s)
Portadores de Fármacos/química , Hidrogeles/química , Péptidos/administración & dosificación , Péptidos/química , Ponzoñas/administración & dosificación , Ponzoñas/química , Animales , Esquema de Medicación , Exenatida , Humanos , Microesferas , Modelos Moleculares , Conformación Molecular , Péptidos/farmacocinética , Polietilenglicoles/química , Ratas , Ponzoñas/farmacocinética
5.
Bioconjug Chem ; 27(10): 2534-2539, 2016 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-27657443

RESUMEN

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.

6.
Proc Natl Acad Sci U S A ; 109(16): 6211-6, 2012 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-22474378

RESUMEN

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.


Asunto(s)
Preparaciones de Acción Retardada/farmacocinética , Sustancias Macromoleculares/química , Polietilenglicoles/química , Profármacos/farmacocinética , Algoritmos , Animales , Carbonatos/química , Cromatografía Líquida de Alta Presión , Preparaciones de Acción Retardada/química , Exenatida , Semivida , Hipoglucemiantes/química , Hipoglucemiantes/farmacocinética , Cinética , Masculino , Ratones , Modelos Biológicos , Modelos Químicos , Estructura Molecular , Péptidos/química , Péptidos/farmacocinética , Profármacos/química , Ratas Sprague-Dawley , Succinimidas/química , Ponzoñas/química , Ponzoñas/farmacocinética
7.
J Biol Chem ; 288(15): 10588-98, 2013 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-23447538

RESUMEN

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.


Asunto(s)
Evolución Molecular , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Animales , Células CHO , Bovinos , Cricetinae , Cricetulus , Cobayas , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Ratones Mutantes , Ratas Topo , Mutación , Pan paniscus , Pan troglodytes , Inhibidores de Proteasas/farmacología , Ratas , Solubilidad , Especificidad de la Especie
8.
Cancers (Basel) ; 16(11)2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38893160

RESUMEN

Malignant rhabdoid tumors (MRTs) are among the most aggressive and treatment-resistant malignancies affecting infants, originating in the kidney, brain, liver, and soft tissues. The 5-year event-free survival rate for these cancers is a mere 20%. In nearly all cases of MRT, the SMARCB1 gene (occasionally SMARCA4)-a pivotal component of the SWI/SNF chromatin remodeling complex-is homozygously deleted, although the precise etiology of these tumors remains unknown. While young patients with localized MRT generally show improved outcomes, especially those who are older and have early-stage disease, the overall prognosis remains poor despite optimal standard treatments. This highlights the urgent need for more effective treatment strategies. We investigated the antitumor activity of a PARP1 inhibitor (talazoparib, TLZ) combined with a DNA alkylating agent (temozolomide, TMZ) in MRT xenograft models. PARP1 is a widely targeted molecule in cancer treatment and, beyond its role in DNA repair, it participates in transcriptional regulation by recruiting chromatin remodeling complexes to modulate DNA accessibility for RNA polymerases. To widen the therapeutic window of the drug combination, we employed PEGylated TLZ (PEG~TLZ), which has been reported to reduce systemic toxicity through slow drug release. Remarkably, our findings indicate that five out of six MRT xenografts exhibited an objective response to PEG~TLZ+TMZ therapy. Significantly, the loss of SMARCB1 was found to confer a protective effect, correlating with higher expression levels of DNA damage and repair proteins in SMARCB1-deficient MRT cells. Additionally, we identified MGMT as a potential biomarker indicative of in vivo MRT response to PEG~TLZ+TMZ therapy. Moreover, our analysis revealed alterations in signaling pathways associated with the observed antitumor efficacy. This study presents a novel and efficacious therapeutic approach for MRT, along with a promising candidate biomarker for predicting tumor response.

9.
Sci Rep ; 14(1): 14175, 2024 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-38898077

RESUMEN

Central nervous system tumors have resisted effective chemotherapy because most therapeutics do not penetrate the blood-tumor-brain-barrier. Nanomedicines between ~ 10 and 100 nm accumulate in many solid tumors by the enhanced permeability and retention effect, but it is controversial whether the effect can be exploited for treatment of brain tumors. PLX038A is a long-acting prodrug of the topoisomerase 1 inhibitor SN-38. It is composed of a 15 nm 4-arm 40 kDa PEG tethered to four SN-38 moieties by linkers that slowly cleave to release the SN-38. The prodrug was remarkably effective at suppressing growth of intracranial breast cancer and glioblastoma (GBM), significantly increasing the life span of mice harboring them. We addressed the important issue of whether the prodrug releases SN-38 systemically and then penetrates the brain to exert anti-tumor effects, or whether it directly penetrates the blood-tumor-brain-barrier and releases the SN-38 cargo within the tumor. We argue that the amount of SN-38 formed systemically is insufficient to inhibit the tumors, and show by PET imaging that a close surrogate of the 40 kDa PEG carrier in PLX038A accumulates and is retained in the GBM. We conclude that the prodrug penetrates the blood-tumor-brain-barrier, accumulates in the tumor microenvironment and releases its SN-38 cargo from within. Based on our results, we pose the provocative question as to whether the 40 kDa nanomolecule PEG carrier might serve as a "Trojan horse" to carry other drugs past the blood-tumor-brain-barrier and release them into brain tumors.


Asunto(s)
Barrera Hematoencefálica , Neoplasias Encefálicas , Irinotecán , Profármacos , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Irinotecán/farmacocinética , Barrera Hematoencefálica/metabolismo , Ratones , Profármacos/farmacocinética , Profármacos/química , Profármacos/metabolismo , Humanos , Línea Celular Tumoral , Femenino , Ensayos Antitumor por Modelo de Xenoinjerto , Glioblastoma/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Camptotecina/análogos & derivados , Camptotecina/farmacocinética , Camptotecina/uso terapéutico
10.
Bioconjug Chem ; 24(12): 1990-7, 2013 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-24171387

RESUMEN

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.


Asunto(s)
Sustancias Macromoleculares/química , Nitrofenoles/química , Carbamatos/química , Diseño de Fármacos , Cinética , Polietilenglicoles/química , Solventes/química , Relación Estructura-Actividad
11.
J Biol Chem ; 285(24): 18516-27, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20378535

RESUMEN

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.


Asunto(s)
Amebiasis/parasitología , Proteasas de Cisteína/química , Proteasas de Cisteína/fisiología , Entamoeba histolytica/metabolismo , Animales , Línea Celular Tumoral , Diseño de Fármacos , Regulación Enzimológica de la Expresión Génica , Humanos , Concentración de Iones de Hidrógeno , Cinética , Espectrometría de Masas/métodos , Ratones , Ratones Endogámicos C3H , Péptido Hidrolasas/química , Inhibidores de Proteasas/farmacología , Proteínas Recombinantes/química , Tiorredoxinas/química
12.
Methods Mol Biol ; 539: 59-78, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19377970

RESUMEN

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.


Asunto(s)
Cumarinas/síntesis química , Péptido Hidrolasas/química , Péptido Hidrolasas/metabolismo , Biblioteca de Péptidos , Péptidos/química , Péptidos/metabolismo , Secuencia de Aminoácidos , Cinética , Péptidos/síntesis química , Especificidad por Sustrato
13.
J Control Release ; 278: 74-79, 2018 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-29577950

RESUMEN

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.


Asunto(s)
Deuterio/química , Sistemas de Liberación de Medicamentos , Octreótido/administración & dosificación , Polímeros/química , Animales , Reactivos de Enlaces Cruzados/química , Portadores de Fármacos/química , Liberación de Fármacos , Semivida , Hidrogeles , Octreótido/química , Octreótido/farmacocinética , Ratas
14.
Sci Rep ; 8(1): 10705, 2018 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-30013126

RESUMEN

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.


Asunto(s)
Calicreínas/metabolismo , Regulación Alostérica/efectos de los fármacos , Sitio Alostérico/genética , Calcio/metabolismo , Cationes Bivalentes/metabolismo , Cristalografía por Rayos X , Calicreínas/antagonistas & inhibidores , Calicreínas/genética , Calicreínas/ultraestructura , Cinética , Leupeptinas/farmacología , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Relación Estructura-Actividad , Especificidad por Sustrato/efectos de los fármacos , Zinc/metabolismo
15.
Int J Parasitol ; 37(7): 713-24, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17336985

RESUMEN

Ticks are ectoparasitic blood-feeders and important vectors for pathogens including arboviruses, rickettsiae, spirochetes and protozoa. As obligate blood-feeders, one possible strategy to retard disease transmission is disruption of the parasite's ability to digest host proteins. However, the constituent peptidases in the parasite gut and their potential interplay in the digestion of the blood meal are poorly understood. We have characterised a novel asparaginyl endopeptidase (legumain) from the hard tick Ixodes ricinus (termed IrAE), which we believe is the first such characterisation of a clan CD family C13 cysteine peptidase (protease) in arthropods. By RT-PCR of different tissues, IrAE mRNA was only expressed in the tick gut. Indirect immunofluorescence and EM localised IrAE in the digestive vesicles of gut cells and within the peritrophic matrix. IrAE was functionally expressed in Pichia pastoris and reacted with a specific peptidyl fluorogenic substrate, and acyloxymethyl ketone and aza-asparagine Michael acceptor inhibitors. IrAE activity was unstable at pH > or = 6.0 and was shown to have a strict specificity for asparagine at P1 using a positional scanning synthetic combinatorial library. The enzyme hydrolyzed protein substrates with a pH optimum of 4.5, consistent with the pH of gut cell digestive vesicles. Thus, IrAE cleaved the major protein of the blood meal, hemoglobin, to a predominant peptide of 4kDa. Also, IrAE trans-processed and activated the zymogen form of Schistosoma mansoni cathepsin B1 -- an enzyme contributing to hemoglobin digestion in the gut of that bloodfluke. The possible functions of IrAE in the gut digestive processes of I. ricinus are compared with those suggested for other hematophagous parasites.


Asunto(s)
Cisteína Endopeptidasas/genética , Sistema Digestivo/enzimología , Ixodes/enzimología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Catepsina B/metabolismo , Clonación Molecular , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/aislamiento & purificación , Cisteína Endopeptidasas/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Hemoglobinas/metabolismo , Ixodes/genética , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Filogenia , Pichia/genética , Pichia/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia
16.
Thromb Haemost ; 117(9): 1750-1760, 2017 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-28726978

RESUMEN

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.


Asunto(s)
Péptidos/metabolismo , Serina Endopeptidasas/metabolismo , Antitrombinas/farmacología , Dominio Catalítico , Técnicas de Visualización de Superficie Celular , Activación Enzimática , Hirudinas/farmacología , Histonas/metabolismo , Humanos , Cinética , Simulación del Acoplamiento Molecular , Biblioteca de Péptidos , Péptidos/química , Péptidos/genética , Proteínas de Plantas/química , Proteínas de Plantas/farmacología , Unión Proteica , Conformación Proteica , Serina Endopeptidasas/sangre , Serina Endopeptidasas/química , Serina Endopeptidasas/genética , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/farmacología , Relación Estructura-Actividad , Especificidad por Sustrato , Trombina/antagonistas & inhibidores , Trombina/metabolismo
17.
ACS Chem Biol ; 12(8): 2107-2116, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-28605180

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Receptor del Péptido 1 Similar al Glucagón/agonistas , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Hipoglucemiantes/administración & dosificación , Microesferas , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/administración & dosificación , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacocinética , Hipoglucemiantes/farmacocinética , Estructura Molecular , Factores de Tiempo
18.
Chem Biol ; 9(8): 881-9, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12204687

RESUMEN

The erythrocytic growth stage of Plasmodium falciparum involves hemoglobin proteolysis as the primary nutrient source with the concomitant release of free heme. The liberated heme is processed by the parasite into hemozoin, a polymeric porphyrin dimer. Histidine-rich protein binds heme and mediates the formation of hemozoin, which is inhibited by the antimalarial drug chloroquine. Interference with heme binding was determined using a microtiterplate assay. Combinatorial libraries were screened and tested against parasite growth, revealing a good correlation between heme binding interference and the inhibition of parasite growth. Several of these compounds retain their potency against a chloroquine-resistant strain of Plasmodium falciparum. The most potent compounds have IC(50) values less than or equal to 50 nM against chloroquine-resistant and chloroquine-sensitive parasites.


Asunto(s)
Antimaláricos/química , Hemo/metabolismo , Proteínas/metabolismo , Animales , Antimaláricos/farmacología , Técnicas Químicas Combinatorias , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Concentración 50 Inhibidora , Plasmodium falciparum/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteínas Protozoarias/metabolismo , Relación Estructura-Actividad
19.
Eur J Pharm Biopharm ; 93: 254-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25900863

RESUMEN

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.


Asunto(s)
Proteína gp41 de Envoltorio del VIH/farmacocinética , Inhibidores de Fusión de VIH/farmacocinética , Fragmentos de Péptidos/farmacocinética , Polietilenglicoles/farmacocinética , Profármacos/farmacocinética , Administración Intravenosa , Animales , Química Farmacéutica , Simulación por Computador , Proteína gp41 de Envoltorio del VIH/administración & dosificación , Proteína gp41 de Envoltorio del VIH/sangre , Proteína gp41 de Envoltorio del VIH/síntesis química , Inhibidores de Fusión de VIH/administración & dosificación , Inhibidores de Fusión de VIH/sangre , Inhibidores de Fusión de VIH/síntesis química , Semivida , Hidrólisis , Masculino , Tasa de Depuración Metabólica , Modelos Biológicos , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/síntesis química , Péptidos , Polietilenglicoles/administración & dosificación , Polietilenglicoles/síntesis química , Profármacos/administración & dosificación , Profármacos/síntesis química , Ratas Sprague-Dawley , Tecnología Farmacéutica/métodos
20.
J Med Chem ; 57(6): 2303-14, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24494988

RESUMEN

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.


Asunto(s)
Camptotecina/análogos & derivados , Glucurónidos/síntesis química , Glucurónidos/farmacología , Profármacos/síntesis química , Profármacos/farmacología , Aminas/química , Animales , Camptotecina/síntesis química , Camptotecina/farmacocinética , Camptotecina/farmacología , Cromatografía Líquida de Alta Presión , Preparaciones de Acción Retardada , Femenino , Glucurónidos/metabolismo , Glucurónidos/farmacocinética , Semivida , Cinética , Hígado/metabolismo , Sustancias Macromoleculares , Fenoles/química , Profármacos/farmacocinética , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA