Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Med Chem ; 67(11): 8585-8608, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38809766

RESUMO

The von Hippel-Lindau (VHL) protein plays a pivotal role in regulating the hypoxic stress response and has been extensively studied and utilized in the targeted protein degradation field, particularly in the context of bivalent degraders. In this study, we present a comprehensive peptidomimetic structure-activity relationship (SAR) approach, combined with cellular NanoBRET target engagement assays to enhance the existing VHL ligands. Through systematic modifications of the molecule, we identified the 1,2,3-triazole group as an optimal substitute of the left-hand side amide bond that yields 10-fold higher binding activity. Moreover, incorporating conformationally constrained alterations on the methylthiazole benzylamine moiety led to the development of highly potent VHL ligands with picomolar binding affinity and significantly improved oral bioavailability. We anticipate that our optimized VHL ligand, GNE7599, will serve as a valuable tool compound for investigating the VHL pathway and advancing the field of targeted protein degradation.


Assuntos
Disponibilidade Biológica , Peptidomiméticos , Proteína Supressora de Tumor Von Hippel-Lindau , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/química , Peptidomiméticos/química , Peptidomiméticos/farmacocinética , Peptidomiméticos/farmacologia , Humanos , Ligantes , Relação Estrutura-Atividade , Administração Oral , Animais
2.
Cancer Res ; 81(8): 2234-2245, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33622696

RESUMO

Targeted imaging and therapy approaches based on novel prostate-specific membrane antigen (PSMA) inhibitors have fundamentally changed the treatment regimen of prostate cancer. However, the exact mechanism of PSMA inhibitor internalization has not yet been studied, and the inhibitors' subcellular fate remains elusive. Here, we investigated the intracellular distribution of peptidomimetic PSMA inhibitors and of PSMA itself by stimulated emission depletion (STED) nanoscopy, applying a novel nonstandard live cell staining protocol. Imaging analysis confirmed PSMA cluster formation at the cell surface of prostate cancer cells and clathrin-dependent endocytosis of PSMA inhibitors. Following the endosomal pathway, PSMA inhibitors accumulated in prostate cancer cells at clinically relevant time points. In contrast with PSMA itself, PSMA inhibitors were found to eventually distribute homogeneously in the cytoplasm, a molecular condition that promises benefits for treatment as cytoplasmic and in particular perinuclear enrichment of the radionuclide carriers may better facilitate the radiation-mediated damage of cancerous cells. This study is the first to reveal the subcellular fate of PSMA/PSMA inhibitor complexes at the nanoscale and aims to inspire the development of new approaches in the field of prostate cancer research, diagnostics, and therapeutics. SIGNIFICANCE: This study uses STED fluorescence microscopy to reveal the subcellular fate of PSMA/PSMA inhibitor complexes near the molecular level, providing insights of great clinical interest and suggestive of advantageous targeted therapies. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2234/F1.large.jpg.


Assuntos
Citoplasma/metabolismo , Glutamato Carboxipeptidase II/antagonistas & inibidores , Terapia de Alvo Molecular/métodos , Neoplasias da Próstata/metabolismo , Animais , Antígenos de Superfície/metabolismo , Membrana Celular/metabolismo , Clatrina/metabolismo , Endocitose , Endossomos/metabolismo , Glutamato Carboxipeptidase II/metabolismo , Xenoenxertos , Humanos , Masculino , Camundongos , Microscopia de Fluorescência/métodos , Nanotecnologia/métodos , Peptidomiméticos/farmacocinética , Peptidomiméticos/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Coloração e Rotulagem
3.
Prep Biochem Biotechnol ; 51(6): 519-529, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33459157

RESUMO

Nowadays, proteins are frequently administered as therapeutic agents in human diseases. However, the main challenge regarding the clinical application of therapeutic proteins is short circulating plasma half-life that leads to more frequent injections for maintaining therapeutic plasma levels, increased therapy costs, immunogenic reactions, and low patient compliance. So, the development of novel strategies to enhance the pharmacokinetic profile of therapeutic proteins has attracted great attention in pharmaceuticals. So far, several techniques, each with their pros and cons, have been developed including chemical bonding to polymers, hyper glycosylation, Fc fusion, human serum albumin fusion, and recombinant PEG mimetics. These techniques mainly classify into three strategies; (i) the endosomal recycling of neonatal Fc receptor which is observed for immunoglobulins and albumin, (ii) decrease in receptor-mediated clearance, and (iii) increase in hydrodynamic radius through chemical and genetic modifications. Recently, novel PEG mimetic peptides like proline/alanine/serine repeat sequences are designed to overcome pitfalls associated with the previous technologies. Biodegradability, lack of or low immunogenicity, product homogeneity, and a simple production process, currently make these polypeptides as the preferred technology for plasma half-life extension of therapeutic proteins. In this review, challenges and pitfalls in the pharmacokinetic enhancement of therapeutic proteins using PEG-mimetic peptides will be discussed in detail.


Assuntos
Peptídeos , Peptidomiméticos , Proteínas Recombinantes de Fusão , Animais , Humanos , Peptídeos/química , Peptídeos/farmacocinética , Peptídeos/uso terapêutico , Peptidomiméticos/química , Peptidomiméticos/farmacocinética , Peptidomiméticos/uso terapêutico , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Polietilenoglicóis/uso terapêutico , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico
4.
Molecules ; 25(17)2020 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-32854201

RESUMO

High kidney uptake is a common feature of peptide-based radiopharmaceuticals, leading to reduced detection sensitivity for lesions adjacent to kidneys and lower maximum tolerated therapeutic dose. In this study, we evaluated if the Met-Val-Lys (MVK) linker could be used to lower kidney uptake of 68Ga-labeled DOTA-conjugated peptides and peptidomimetics. A model compound, [68Ga]Ga-DOTA-AmBz-MVK(Ac)-OH (AmBz: aminomethylbenzoyl), and its derivative, [68Ga]Ga-DOTA-AmBz-MVK(HTK01166)-OH, coupled with the PSMA (prostate-specific membrane antigen)-targeting motif of the previously reported HTK01166 were synthesized and evaluated to determine if they could be recognized and cleaved by the renal brush border enzymes. Additionally, positron emission tomography (PET) imaging, ex vivo biodistribution and in vivo stability studies were conducted in mice to evaluate their pharmacokinetics. [68Ga]Ga-DOTA-AmBz-MVK(Ac)-OH was effectively cleaved specifically by neutral endopeptidase (NEP) of renal brush border enzymes at the Met-Val amide bond, and the radio-metabolite [68Ga]Ga-DOTA-AmBz-Met-OH was rapidly excreted via the renal pathway with minimal kidney retention. [68Ga]Ga-DOTA-AmBz-MVK(HTK01166)-OH retained its PSMA-targeting capability and was also cleaved by NEP, although less effectively when compared to [68Ga]Ga-DOTA-AmBz-MVK(Ac)-OH. The kidney uptake of [68Ga]Ga-DOTA-AmBz-MVK(HTK01166)-OH was 30% less compared to that of [68Ga]Ga-HTK01166. Our data demonstrated that derivatives of [68Ga]Ga-DOTA-AmBz-MVK-OH can be cleaved specifically by NEP, and therefore, MVK can be a promising cleavable linker for use to reduce kidney uptake of radiolabeled DOTA-conjugated peptides and peptidomimetics.


Assuntos
Radioisótopos de Gálio , Rim , Peptídeos , Peptidomiméticos , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Animais , Linhagem Celular Tumoral , Radioisótopos de Gálio/química , Radioisótopos de Gálio/farmacocinética , Radioisótopos de Gálio/farmacologia , Humanos , Rim/diagnóstico por imagem , Rim/metabolismo , Camundongos , Neprilisina/química , Neprilisina/metabolismo , Peptídeos/síntese química , Peptídeos/química , Peptídeos/farmacocinética , Peptídeos/farmacologia , Peptidomiméticos/síntese química , Peptidomiméticos/química , Peptidomiméticos/farmacocinética , Peptidomiméticos/farmacologia , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Compostos Radiofarmacêuticos/farmacologia
5.
Bioorg Med Chem Lett ; 30(17): 127377, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32738988

RESUMO

The unprecedented pandemic of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is threatening global health. The virus emerged in late 2019 and can cause a severe disease associated with significant mortality. Several vaccine development and drug discovery campaigns are underway. The SARS-CoV-2 main protease is considered a promising drug target, as it is dissimilar to human proteases. Sequence and structure of the main protease are closely related to those from other betacoronaviruses, facilitating drug discovery attempts based on previous lead compounds. Covalently binding peptidomimetics and small molecules are investigated. Various compounds show antiviral activity in infected human cells.


Assuntos
Antivirais/farmacologia , Betacoronavirus/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Antivirais/farmacocinética , Betacoronavirus/enzimologia , Linhagem Celular , Proteases 3C de Coronavírus , Cisteína Endopeptidases/química , Inibidores de Cisteína Proteinase/farmacocinética , Descoberta de Drogas , Humanos , Peptidomiméticos/farmacocinética , Peptidomiméticos/farmacologia , SARS-CoV-2 , Especificidade por Substrato , Proteínas não Estruturais Virais/química
6.
Molecules ; 25(10)2020 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-32423178

RESUMO

The high affinity and specificity of peptides towards biological targets, in addition to their favorable pharmacological properties, has encouraged the development of many peptide-based pharmaceuticals, including peptide-based positron emission tomography (PET) radiopharmaceuticals. However, the poor in vivo stability of unmodified peptides against proteolysis is a major challenge that must be overcome, as it can result in an impractically short in vivo biological half-life and a subsequently poor bioavailability when used in imaging and therapeutic applications. Consequently, many biologically and pharmacologically interesting peptide-based drugs may never see application. A potential way to overcome this is using peptide analogues designed to mimic the pharmacophore of a native peptide while also containing unnatural modifications that act to maintain or improve the pharmacological properties. This review explores strategies that have been developed to increase the metabolic stability of peptide-based pharmaceuticals. It includes modifications of the C- and/or N-termini, introduction of d- or other unnatural amino acids, backbone modification, PEGylation and alkyl chain incorporation, cyclization and peptide bond substitution, and where those strategies have been, or could be, applied to PET peptide-based radiopharmaceuticals.


Assuntos
Peptídeos/síntese química , Peptidomiméticos/síntese química , Tomografia por Emissão de Pósitrons/métodos , Processamento de Proteína Pós-Traducional , Compostos Radiofarmacêuticos/síntese química , Acilação , Animais , Radioisótopos de Carbono/química , Radioisótopos de Carbono/farmacocinética , Ciclização , Radioisótopos de Flúor/química , Radioisótopos de Flúor/farmacocinética , Radioisótopos de Gálio/química , Radioisótopos de Gálio/farmacocinética , Meia-Vida , Humanos , Metilação , Peptídeos/farmacocinética , Peptidomiméticos/farmacocinética , Estabilidade Proteica , Compostos Radiofarmacêuticos/farmacocinética , Roedores
7.
J Med Chem ; 63(9): 4496-4505, 2020 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-32302130

RESUMO

The insertion of single 1,4-disubstituted 1,2,3-triazoles as metabolically stable bioisosteres of trans-amide bonds (triazole scan) was recently applied to the 177Lu-labeled tumor-targeting analog of minigastrin, [Nle15]MG11. The reported novel mono-triazolo-peptidomimetics of [Nle15]MG11 showed either improved resistance against enzymatic degradation or a significantly increased affinity toward the target receptor but never both. To enhance further the tumor-targeting properties of the minigastrin analogs, we studied conjugates with multiple amide-to-triazole substitutions for additive or synergistic effects. Promising candidates were identified by modification of two or three amide bonds, which yielded both improved stability and increased receptor affinity of the peptidomimetics in vitro. Biodistribution studies of radiolabeled multi-triazolo-peptidomimetics in mice bearing receptor-positive tumor xenografts revealed up to 4-fold increased tumor uptake in comparison to the all-amide reference compound [Nle15]MG11. In addition, we report here for the first time a linear peptidomimetic with three triazole insertions in its backbone and maintained biological activity.


Assuntos
Antineoplásicos/farmacologia , Gastrinas/farmacologia , Peptidomiméticos/farmacologia , Compostos Radiofarmacêuticos/farmacologia , Triazóis/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Desenho de Fármacos , Gastrinas/síntese química , Gastrinas/metabolismo , Gastrinas/farmacocinética , Humanos , Lutécio/química , Camundongos , Neoplasias/metabolismo , Peptidomiméticos/síntese química , Peptidomiméticos/metabolismo , Peptidomiméticos/farmacocinética , Ligação Proteica , Radioisótopos/química , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/metabolismo , Compostos Radiofarmacêuticos/farmacocinética , Receptor de Colecistocinina B/metabolismo , Triazóis/síntese química , Triazóis/metabolismo , Triazóis/farmacocinética
8.
J Med Chem ; 63(9): 4484-4495, 2020 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-32302139

RESUMO

MG11 is a truncated analog of minigastrin, a peptide with high affinity and specificity toward the cholecystokinin-2 receptor (CCK2R), which is overexpressed by different tumors. Thus, radiolabeled MG11 derivatives have great potential for use in cancer diagnosis and therapy. A drawback of MG11 is its fast degradation by proteases, leading to moderate tumor uptake in vivo. We introduced 1,4-disubstituted 1,2,3-triazoles as metabolically stable bioisosteres to replace labile amide bonds of the peptide. The "triazole scan" yielded peptidomimetics with improved resistance to enzymatic degradation and/or enhanced affinity toward the CCK2R. Remarkably, our lead compound achieved a 10-fold increase in receptor affinity, resulting in a 2.6-fold improved tumor uptake in vivo. Modeling of the ligand-CCK2R complex suggests that an additional cation-π interaction of the aromatic triazole moiety with the Arg356 residue of the receptor is accountable for these observations. We show for the first time that the amide-to-triazole substitution strategy offers new opportunities in drug development that go beyond the metabolic stabilization of bioactive peptides.


Assuntos
Antineoplásicos/farmacologia , Gastrinas/farmacologia , Peptidomiméticos/farmacologia , Compostos Radiofarmacêuticos/farmacologia , Triazóis/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Feminino , Gastrinas/síntese química , Gastrinas/metabolismo , Gastrinas/farmacocinética , Humanos , Lutécio/química , Camundongos , Neoplasias/metabolismo , Peptidomiméticos/síntese química , Peptidomiméticos/metabolismo , Peptidomiméticos/farmacocinética , Ligação Proteica , Conformação Proteica , Radioisótopos/química , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/metabolismo , Compostos Radiofarmacêuticos/farmacocinética , Receptor de Colecistocinina B/metabolismo , Triazóis/síntese química , Triazóis/metabolismo , Triazóis/farmacocinética
9.
Expert Opin Drug Discov ; 12(10): 1023-1030, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28699792

RESUMO

INTRODUCTION: The melanocortin system is a primordial and critical system for survival, involved in a wide variety of physiological functions. It includes melanocortin receptors (MCRs) and melanotropin ligands (MCLs). MCRs are important drug targets that can regulate several key physiological processes. Extensive efforts have been made to develop peptide and peptidomimetics targeting melanocortin receptors including MC1R, MC3R, MC4R and MC5R. Most research is focused on developing potent and selective melanotropins. However, developing bioavailable melanotropins remains challenging. Areas covered: Herein, the authors summarize promising strategies for developing bioavailable MCLs by using cyclized N-methylated melanotropins, and using cyclotide and tetrapeptide as templates. They discuss their unique advantages in oral availability and targeting MCRs in the central nervous system or in peripheral tissues. Finally, they discuss the observed differences in thepharmacology of MCRs between in vitro and in vivo tests. Expert opinion: N-methylated cyclized melanotropins have great potential to become bio- available drugs targeting MCRs in the brain, while MCR-grafted cyclotides tend to target MCRs in peripheral tissue. A better understanding of the biased signaling process is a new challenge and opportunity for the future discovery of bioavailable MCLs.


Assuntos
Desenho de Fármacos , Hormônios Estimuladores de Melanócitos/síntese química , Receptores de Melanocortina/metabolismo , Animais , Disponibilidade Biológica , Ciclotídeos/síntese química , Ciclotídeos/farmacocinética , Ciclotídeos/farmacologia , Humanos , Ligantes , Hormônios Estimuladores de Melanócitos/química , Hormônios Estimuladores de Melanócitos/farmacocinética , Peptídeos/síntese química , Peptídeos/farmacocinética , Peptídeos/farmacologia , Peptidomiméticos/síntese química , Peptidomiméticos/farmacocinética , Peptidomiméticos/farmacologia , Distribuição Tecidual
10.
Ann N Y Acad Sci ; 1397(1): 169-184, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28505395

RESUMO

The blood-brain barrier (BBB) formed by the microvascular endothelium limits cerebral drug delivery. The paraendothelial cleft is sealed by tight junctions (TJs) with a major contribution from claudin-5, which we selected as a target to modulate BBB permeability. For this purpose, drug-enhancer peptides were designed based on the first extracellular loop (ECL) of claudin-5 to allow transient BBB permeabilization. Peptidomimetics (C5C2 and derivatives, nanomolar affinity to claudin-5) size-selectively (≤40 kDa) and reversibly (12-48 h) increased the permeability of brain endothelial and claudin-5-transfected epithelial cell monolayers. Upon peptide uptake, the number of TJ strand particles diminished, claudin-5 was downregulated and redistributed from cell-cell contacts to the cytosol, and the cell shape was altered. Cellular permeability of doxorubicin (cytostatic drug, 580 Da) was enhanced after peptide administration. Mouse studies (3.5 µmol/kg i.v.) confirmed that, for both C5C2 and a d-amino acid derivative, brain uptake of Gd-diethylene-triamine penta-acetic acid (547 Da) was enhanced within 4 h of treatment. On the basis of our functional data, circular dichroism measurements, molecular modeling, and docking experiments, we suggest an association model between ß-sheets flanked by α-helices, formed by claudin-5 ECLs, and the peptides. In conclusion, we identified claudin-5 peptidomimetics that improve drug delivery through endothelial and epithelial barriers expressing claudin-5.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Claudina-5/farmacologia , Células Endoteliais/efeitos dos fármacos , Peptidomiméticos/farmacologia , Animais , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/farmacocinética , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/ultraestrutura , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular , Células Cultivadas , Dicroísmo Circular , Claudina-5/química , Claudina-5/farmacocinética , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Gadolínio DTPA/administração & dosagem , Gadolínio DTPA/farmacocinética , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Microscopia Eletrônica/métodos , Modelos Moleculares , Peptidomiméticos/química , Peptidomiméticos/farmacocinética , Permeabilidade/efeitos dos fármacos , Conformação Proteica , Ratos , Rodaminas/administração & dosagem , Rodaminas/farmacocinética , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo , Junções Íntimas/ultraestrutura , Imagem com Lapso de Tempo/métodos
11.
J Cell Biochem ; 118(4): 903-913, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27764887

RESUMO

Polymers of guanidinylated disulfide containing poly(amido amine)s (Gua-SS-PAAs), have shown high transfection efficiency and low cytotoxicity. Previously, we synthesized two Gua-SS-PAA polymers, using guanidino containing monomers (i.e., arginine and agmatine, denoted as ARG and AGM, respectively) and N,N'-cystaminebisacrylamide (CBA). In this study, these two polymers, AGM-CBA and ARG-CBA were complexed with plasmid DNA, and their uptake pathway was investigated. Complexes distribution in MCF-7 cells, and changes on cell endosomes/lysosomes and membrane after the cells were exposed to complexes were tested. In addition, how the transfection efficiency changed with the cell cycle status as well as endocytosis inhibitors were studied. The polymers of AGM-CBA and ARG-CBA can avoid endosomal/lysosomal trap, therefore, greatly delivering plasmid DNA (pDNA) to the cell nucleoli. It is the guanidine groups in the polymers that enhanced complexes' permeation through cell membrane with slight membrane damage, and targeting to the nucleoli. J. Cell. Biochem. 118: 903-913, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
DNA/administração & dosagem , Transfecção/métodos , Transporte Ativo do Núcleo Celular , Ciclo Celular , Nucléolo Celular/metabolismo , DNA/genética , Dissulfetos , Sistemas de Liberação de Medicamentos , Endocitose , Técnicas de Transferência de Genes , Guanidina , Humanos , Células MCF-7 , Peptidomiméticos/síntese química , Peptidomiméticos/química , Peptidomiméticos/farmacocinética , Plasmídeos/administração & dosagem , Plasmídeos/genética , Polímeros/síntese química , Polímeros/química , Polímeros/farmacocinética
12.
Eur J Med Chem ; 125: 914-924, 2017 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-27769032

RESUMO

Doxorubicin (DOX) belongs to the anthracycline class of drugs that are used in the treatment of various cancers. It has limited cystostatic effects in therapeutic doses, but higher doses can cause cardiotoxicity. In the current approach, we conjugated a peptidomimetic (Arg-aminonaphthylpropionic acid-Phe, compound 5) known to bind to HER2 protein to DOX via a glutaric anhydride linker. Antiproliferative assays suggest that the DOX-peptidomimetic conjugate has activity in the lower micromolar range. The conjugate exhibited higher toxicity in HER2-overexpressed cells than in MCF-7 and MCF-10A cells that do not overexpress HER2 protein. Cellular uptake studies using confocal microscope experiments showed that the conjugate binds to HER2-overexpressed cells and DOX is taken up into the cells in 4 h compared to conjugate in MCF-7 cells. Binding studies using surface plasmon resonance indicated that the conjugate binds to the HER2 extracellular domain with high affinity compared to compound 5 or DOX alone. The conjugate was stable in the presence of cells with a half-life of nearly 4 h and 1 h in human serum. DOX is released from the conjugate and internalized into the cells in 4 h, causing cellular toxicity. These results suggest that this conjugate can be used to target DOX to HER2-overexpressing cells and can improve the therapeutic index of DOX for HER2-positive cancer.


Assuntos
Antineoplásicos/farmacocinética , Doxorrubicina/farmacologia , Peptidomiméticos/farmacologia , Receptor ErbB-2/análise , Antineoplásicos/química , Antineoplásicos/farmacologia , Sítios de Ligação , Doxorrubicina/química , Doxorrubicina/farmacocinética , Feminino , Meia-Vida , Humanos , Células MCF-7 , Peptidomiméticos/química , Peptidomiméticos/farmacocinética , Ligação Proteica
13.
Sci Rep ; 6: 26908, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27245315

RESUMO

The neuropeptide kisspeptin and its receptor, KiSS1R, govern the reproductive timeline of mammals by triggering puberty onset and promoting ovulation by stimulating gonadotrophin-releasing hormone (GnRH) secretion. To overcome the drawback of kisspeptin short half-life we designed kisspeptin analogs combining original modifications, triazole peptidomimetic and albumin binding motif, to reduce proteolytic degradation and to slow down renal clearance, respectively. These analogs showed improved in vitro potency and dramatically enhanced pharmacodynamics. When injected intramuscularly into ewes (15 nmol/ewe) primed with a progestogen, the best analog (compound 6, C6) induced synchronized ovulations in both breeding and non-breeding seasons. Ovulations were fertile as demonstrated by the delivery of lambs at term. C6 was also fully active in both female and male mice but was completely inactive in KiSS1R KO mice. Electrophysiological recordings of GnRH neurons from brain slices of GnRH-GFP mice indicated that C6 exerted a direct excitatory action on GnRH neurons. Finally, in prepubertal female mice daily injections (0.3 nmol/mouse) for five days significantly advanced puberty. C6 ability to trigger ovulation and advance puberty demonstrates that kisspeptin analogs may find application in the management of livestock reproduction and opens new possibilities for the treatment of reproductive disorders in humans.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Kisspeptinas/genética , Ovulação/efeitos dos fármacos , Peptidomiméticos/farmacologia , Receptores de Kisspeptina-1/genética , Reprodução/efeitos dos fármacos , Maturidade Sexual/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Cruzamento/métodos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Hormônio Liberador de Gonadotropina/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Meia-Vida , Humanos , Kisspeptinas/metabolismo , Masculino , Camundongos , Camundongos Knockout , Ovulação/genética , Peptidomiméticos/síntese química , Peptidomiméticos/farmacocinética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Kisspeptina-1/deficiência , Reprodução/genética , Técnicas de Reprodução Assistida , Maturidade Sexual/genética , Ovinos
14.
ACS Appl Mater Interfaces ; 8(12): 7638-45, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26964858

RESUMO

RNA interference (RNAi) is a highly efficient approach for gene silencing. Regulation of gene expression at post-transcriptional level provides great potential for curing diseases caused by abnormal overexpression of disease-related genes. However, the application of RNAi in the clinic has been hindered by the lack of efficient and biocompatible delivery systems. Therefore, the development of a safe and tissue-targeted double-stranded interfering RNA (siRNA) carrier for clinical application is urgently needed. Here we report the discovery of a highly efficient liposomal siRNA delivery agent based on a novel peptidomimetic built from natural amino acids. Fine tuning of the composition of amino acids, the type of amide linkage in the peptidomimetic, as well as the formulation and the physicochemical parameters of the novel lipoplex resulted in a lipid nanoparticle (LNP) that efficiently encapsulates and carries siRNA to mouse liver. In vivo experiments showed that a single injection of unmodified siRNA complexed to one of the peptidomimetics at a clinically feasible dose induced significant RNAi in mouse liver, resulting in a 90% decrease in apolipoprotein B (ApoB) mRNA level, as well as a 60% decrease in serum ApoB protein level. Analysis of mouse serum by ELISA indicated that the novel peptidomimetic based lipoplex did not elevate the level of liver enzymes (ALT, AST) in the serum. Our novel peptidomimetic-based lipoplex demonstrated great potential for the development of a safe and efficient siRNA delivery agent for clinical applications.


Assuntos
Apolipoproteínas B/biossíntese , Inativação Gênica , Fígado/enzimologia , Peptidomiméticos , RNA Interferente Pequeno , Animais , Células Hep G2 , Humanos , Lipossomos , Camundongos , Peptidomiméticos/química , Peptidomiméticos/farmacocinética , Peptidomiméticos/farmacologia , RNA Interferente Pequeno/química , RNA Interferente Pequeno/farmacocinética , RNA Interferente Pequeno/farmacologia
15.
Eur J Nucl Med Mol Imaging ; 43(5): 953-963, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26497698

RESUMO

PURPOSE: Integrins are transmembrane receptors responsible for cell-cell adhesion and cell-extracellular matrix binding and play an important role in angiogenesis and tumour metastasis. For this reason, integrins are increasingly used as targets for molecular imaging. Up to now interest has mostly been focused on the integrin subtype αvß3. However, targeting of other subtypes such as the integrin α5ß1 is also of high interest due to its central role in colonization of metastatic cells, resistance of tumour cells to chemotherapy and ionizing radiation, and tumour aggressiveness. Recently, a highly active antagonist ligand (2,2'-(7-(1-carboxy-4-((6-((3-(4-(((S)-1-carboxy-2-(2-(3-guanidinobenzamido)acetamido)ethyl)carbamoyl)-3,5-dimethylphenoxy)propyl)amino)-6-oxohexyl)amino)-4-oxobutyl)-1,4,7-triazonane-1,4-diyl)diacetic acid, FR366) for the integrin subtype α5ß1 with high selectivity versus αvß3, has been developed and tested successfully in preliminary in vitro and in vivo experiments. Here, we present our results of an investigation of the use of (68)Ga-labelled α5ß1 ligand in PET imaging. METHODS: The free α5ß1 peptidomimetic ligand was functionalized with a spacer (6-aminohexanoic acid) and the bifunctional chelator 1-((1,3-dicarboxy)propyl)-4,7-(carboxymethyl)-1,4,7-triazacyclononane (NODAGA) to yield FR366 and labelled with (68)Ga. To confirm selective in vivo targeting of α5ß1, female BALB/c nude mice xenografted with α5ß1-expressing RKO cells in the right shoulder and α5ß1/αvß3-expressing M21 cells in the left shoulder were subjected to PET/CT scans and biodistribution experiments. Specificity of tracer uptake was proven by blocking studies. Metabolic stability of the injected tracer was measured in urine and in plasma. RESULTS: MicroPET/CT scans with radiolabelled FR366 showed a good tumour-to-normal tissue ratio with low uptake in the liver (0.32 ± 0.14 %ID/g) and good retention of (68)Ga-NODAGA-FR366 in the tumour (0.71 ± 0.20 %ID/g and 0.40 ± 0.12 %ID/g for RKO and M21 tumours, respectively, at 90 min after injection). Biodistribution experiments showed uptake in the α5ß1-expressing RKO tumour of 1.05 ± 0.23 %ID/g at 90 min after injection. Specificity of tracer uptake was demonstrated by injection of 5 mg/kg unlabelled ligand 10 min prior to tracer injection, resulting in a 67 % reduction in uptake in the RKO tumour. The tracer was found to be metabolically stable in urine and plasma 30 min after injection. CONCLUSION: Our results show that PET imaging of α5ß1 expression with the (68)Ga-labelled α5ß1-specific ligand is feasible with good image quality. Thus, FR366 is a promising new tool for investigating the role of α5ß1 in angiogenesis and the influence of this integrin subtype on cancer aggressiveness and metastatic potential.


Assuntos
Radioisótopos de Gálio/farmacocinética , Guanidinas/farmacocinética , Integrina alfa5beta1/metabolismo , Peptidomiméticos/farmacocinética , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/farmacocinética , Triazinas/farmacocinética , Animais , Linhagem Celular Tumoral , Feminino , Guanidinas/química , Guanidinas/farmacologia , Humanos , Integrina alfa5beta1/antagonistas & inibidores , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ligação Proteica , Distribuição Tecidual , Triazinas/química , Triazinas/farmacologia
16.
Bioconjug Chem ; 26(10): 2143-52, 2015 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-26347939

RESUMO

Neurotensin (NT) is a regulatory peptide with nanomolar affinity toward NT receptors, which are overexpressed by different clinically relevant tumors. Its binding sequence, NT(8-13), represents a promising vector for the development of peptidic radiotracers for tumor imaging and therapy. The main drawback of the peptide is its short biological half-life due to rapid proteolysis in vivo. Herein, we present an innovative strategy for the stabilization of peptides using nonhydrolizable 1,4-disubstituted, 1,2,3-triazoles as amide bond surrogates. A "triazole scan" of the peptide sequence yielded novel NT(8-13) analogues with enhanced stability, retained receptor affinity, and improved tumor targeting properties in vivo. The synthesis of libraries of triazole-based peptidomimetics was achieved efficiently on solid support by a combination of Fmoc-peptide chemistry, diazo transfer reactions, and the Cu(I)-catalyzed alkyne azide cycloaddition (CuAAC) employing methods that are fully compatible with standard solid phase peptide synthesis (SPPS) chemistry. Thus, the amide-to-triazole substitution strategy may represent a general methodology for the metabolic stabilization of biologically active peptides.


Assuntos
Antineoplásicos/farmacocinética , Neurotensina/química , Fragmentos de Peptídeos/química , Peptidomiméticos/química , Radioisótopos/química , Triazóis/química , Animais , Antineoplásicos/química , Técnicas de Química Sintética , Reação de Cicloadição , Feminino , Células HT29 , Meia-Vida , Humanos , Marcação por Isótopo/métodos , Lutécio/química , Camundongos Nus , Terapia de Alvo Molecular/métodos , Peptidomiméticos/farmacocinética , Receptores de Neurotensina/metabolismo , Relação Estrutura-Atividade , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Artigo em Russo | MEDLINE | ID: mdl-26356399

RESUMO

Authors present an overview of theirs author's works on the design of low-molecular mimetic of the nerve growth factor and studies of mechanisms of action and pharmacological properties of the compound. The original working hypothesis, underlying the design of the compound, posited that different neurotrophin hairpin loops could activate different signaling cascades by interaction with the receptor and so be responsible for different effects. The mimetic bis(N-succinyl-L-glutamyl-L-lysine)hexametylendiamide (GK-2), that was designed on the basis of NGF loop 4 ß-turn sequence, activated TrkA and PI3K/Akt, but not MAPK/Erk. GK-2 showed neuroprotective activity in concentrations up to 10-9М against H(2)O(2) or glutamate or MPTP-induced neurotoxicity in РС12, НТ22 cells and primary rat hippocampal neurons. At that, GK-2 has no differentiating activity. In in vivo experiments, GK-2 exhibited significant anti-ischemic, anti-parkinsonic effect, reversed impaired cognitive functions in models of Alzheimer's disease in doses 0.01 - 5 mg/kg intraperitoneally and 5-10 mg/kg orally, but does not induce side effects accompanying the full-length neurotrophin treatment, which are hyperalgesia and weight loss. It was shown that GK-2 was a low-toxicity compound (LD50=700 mg/kg, intraperitoneally, mice) and capable of crossing the blood-brain barrier. The agent GK-2 is promising for development as a neuroprotective agent and is currently in preclinical studies.


Assuntos
Dipeptídeos/química , Dipeptídeos/farmacologia , Fator de Crescimento Neural/química , Peptidomiméticos/química , Peptidomiméticos/farmacologia , Animais , Barreira Hematoencefálica/metabolismo , Dipeptídeos/farmacocinética , Desenho de Fármacos , Flavonoides/farmacologia , Proteínas de Choque Térmico HSP70/biossíntese , Heme Oxigenase-1/biossíntese , Humanos , Camundongos , Células PC12 , Peptidomiméticos/farmacocinética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/agonistas , Ratos , Receptor trkA/agonistas
18.
J Med Chem ; 58(18): 7475-84, 2015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26309061

RESUMO

Novel backbone-modified radiolabeled analogs based on the tumor targeting peptide bombesin were synthesized and fully evaluated in vitro and in vivo. We have recently introduced the use of 1,4-disubstituted 1,2,3-triazoles as metabolically stable trans-amide bond surrogates in radiolabeled peptides in order to improve their tumor targeting. As an extension of our approach, we now report several backbone-modified analogs of the studied bombesin peptide bearing multiple triazole substitutions. We investigated the effect of the modifications on several biological parameters including the internalization of the radiopeptidomimetics into tumor cells, their affinity toward the gastrin releasing peptide receptor (GRPr), metabolic stability in blood plasma, and biodistribution in mice bearing GRPr-expressing xenografts. The backbone-modified radiotracers exhibited a significantly increased resistance to proteolytic degradation. In addition, some of the radiopeptidomimetics retained a nanomolar affinity toward GRPr, resulting in an up to 2-fold increased tumor uptake in vivo in comparison to a (all amide bond) reference compound.


Assuntos
Amidas/química , Bombesina/análogos & derivados , Lutécio , Fragmentos de Peptídeos/química , Peptidomiméticos/química , Compostos Radiofarmacêuticos/química , Triazóis/química , Amidas/farmacocinética , Amidas/farmacologia , Animais , Bombesina/química , Bombesina/farmacocinética , Bombesina/farmacologia , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Camundongos Nus , Transplante de Neoplasias , Fragmentos de Peptídeos/farmacocinética , Fragmentos de Peptídeos/farmacologia , Peptidomiméticos/farmacocinética , Peptidomiméticos/farmacologia , Proteólise , Compostos Radiofarmacêuticos/farmacocinética , Compostos Radiofarmacêuticos/farmacologia , Receptores da Bombesina/metabolismo , Relação Estrutura-Atividade , Distribuição Tecidual , Triazóis/farmacocinética , Triazóis/farmacologia
19.
Nucl Med Biol ; 42(10): 780-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26169882

RESUMO

INTRODUCTION: In this study, a structurally modified phosphoramidate scaffold, with improved prostate-specific membrane antigen (PSMA) avidity, stability and in vivo characteristics, as a PET imaging agent for prostate cancer (PCa), was prepared and evaluated. METHODS: p-Fluorobenzoyl-aminohexanoate and 2-(3-hydroxypropyl)glycine were introduced into the PSMA-targeting scaffold yielding phosphoramidate 5. X-ray crystallography was performed on the PSMA/5 complex. [(18)F]5 was synthesized, and cell uptake and internalization studies were conducted in PSMA(+) LNCaP and CWR22Rv1 cells and PSMA(-) PC-3 cells. In vivo PET imaging and biodistribution studies were performed at 1 and 4 h post injection in mice bearing CWR22Rv1 tumor, with or without blocking agent. RESULTS: The crystallographic data showed interaction of the p-fluorobenzoyl group with an arene-binding cleft on the PSMA surface. In vitro studies revealed elevated uptake of [(18)F]5 in PSMA(+) cells (2.2% in CWR22Rv1 and 12.1% in LNCaP) compared to PSMA(-) cells (0.08%) at 4 h. In vivo tumor uptake of 2.33% ID/g and tumor-to-blood ratio of 265:1 was observed at 4 h. CONCLUSIONS: We have successfully synthesized, radiolabeled and evaluated a new PSMA-targeted PET agent. The crystal structure of the PSMA/5 complex highlighted the interactions within the arene-binding cleft contributing to the overall complex stability. The high target uptake and rapid non-target clearance exhibited by [(18)F]5 in PSMA(+) xenografts substantiates its potential use for PET imaging of PCa. ADVANCES IN KNOWLEDGE: The only FDA-approved imaging agent for PCa, Prostascint®, targets PSMA but suffers from inherent shortcomings. The data acquired in this manuscript confirmed that our new generation of [(18)F]-labeled PSMA inhibitor exhibited promising in vivo performance as a PET imaging agent for PCa and is well-positioned for subsequent clinical trials. Implications for Patient Care Our preliminary data demonstrate that this tracer possesses the required imaging characteristics to be sensitive and specific for PCa imaging in patients at all stages of the disease.


Assuntos
Amidas/química , Radioisótopos de Flúor , Glutamato Carboxipeptidase II/antagonistas & inibidores , Peptidomiméticos/química , Peptidomiméticos/farmacologia , Ácidos Fosfóricos/química , Tomografia por Emissão de Pósitrons/métodos , Neoplasias da Próstata/diagnóstico por imagem , Animais , Antígenos de Superfície/química , Transporte Biológico , Linhagem Celular Tumoral , Estabilidade de Medicamentos , Glutamato Carboxipeptidase II/química , Humanos , Concentração Inibidora 50 , Marcação por Isótopo , Masculino , Camundongos , Modelos Moleculares , Peptidomiméticos/metabolismo , Peptidomiméticos/farmacocinética , Neoplasias da Próstata/patologia , Inibidores de Proteases/química , Inibidores de Proteases/metabolismo , Inibidores de Proteases/farmacocinética , Inibidores de Proteases/farmacologia , Conformação Proteica , Distribuição Tecidual
20.
J Med Chem ; 56(22): 9328-41, 2013 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-24144329

RESUMO

Peptide hybrids (phybrids) comprising covalently linked peptide hormones can leverage independent biological pathways for additive or synergistic metabolic benefits. PEGylation of biologics offers enhanced stability, duration, and reduced immunogenicity. These two modalities can be combined to produce long-acting therapeutics with dual pharmacology and enhanced efficacy. Compound 10 is composed of an exenatide (AC2993) analogue, AC3174, and an amylinomimetic, davalintide (AC2307), with an intervening 40 kD PEG moiety. It displayed dose-dependent and prolonged efficacy for glucose control and body weight reduction in rodents with superior in vitro and in vivo activities compared to those of a side-chain PEGylated phybrid 6. In diet-induced obese (DIO) rats, the weight-loss efficacy of 10 was similar to that of a combination of PEG-parents 3 and 4. A single dose of 10 elicited sustained body weight reduction in DIO rats for at least 21 days. Compound 10's terminal half-life of ~27 h should translate favorably to weekly dosing in humans.


Assuntos
Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Doenças Metabólicas/tratamento farmacológico , Hormônios Peptídicos/metabolismo , Peptídeos/farmacologia , Peptidomiméticos/farmacologia , Polietilenoglicóis/química , Peçonhas/farmacologia , Animais , Desenho de Fármacos , Exenatida , Feminino , Humanos , Masculino , Camundongos , Modelos Moleculares , Obesidade/tratamento farmacológico , Peptídeos/química , Peptídeos/farmacocinética , Peptídeos/uso terapêutico , Peptidomiméticos/química , Peptidomiméticos/farmacocinética , Peptidomiméticos/uso terapêutico , Estrutura Secundária de Proteína , Ratos , Fatores de Tempo , Peçonhas/química , Peçonhas/farmacocinética , Peçonhas/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA