Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Tissue Eng Regen Med ; 20(6): 965-979, 2023 10.
Article in English | MEDLINE | ID: mdl-37589886

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) is an incurable disease that negatively influences the quality of life of patients. Current and emerging therapies target proinflammatory cytokines and/or receptors to downregulate proinflammatory responses, but insufficient remission requires other therapeutic agents. Herein, we report that the synthetic anti-inflammatory peptide 15 (SAP15) is capable of cell penetration and anti-inflammatory activity in human macrophages. METHODS: SAP15 was labeled with fluorescence and administered to human leukemia monocytic cells (THP-1) cells for cell penetration analysis. Using biolayer interferometry analysis, the binding affinity of SAP15 with histone deacetylase 5 (HDAC5) was measured. SAP15-treated THP-1 cells were analyzed by protein phosphorylation assay, flow cytometry, and enzyme-linked immunosorbent assay (ELISA). In addition, in vivo analysis of the therapeutic effect on IBD was observed in a dextran sulfate sodium (DSS)-induced model. Samples from SAP15-treated mice were analyzed at both the macroscopic and microscopic levels using ELISA, myeloperoxidase (MPO) assays, and histological evaluations. RESULTS: SAP15 was internalized within the cytosol and nucleus of THP-1 cells and bound to the HDAC5 protein. SAP15-treated macrophages were assessed for protein phosphorylation and showed inhibited phosphorylation of HDAC5 and other immune-related proteins, which led to increased M2-like macrophage markers and decreased M1-like macrophage markers and tumor necrosis factor-α and interleukin-6 cytokine levels. The SAP15 treatment on IBD model showed significant recovery of colon length. Further histological analysis of colon demonstrated the therapeutic effect of SAP15 on mucosal layer. Moreover, proinflammatory cytokine levels and MPO activity from the plasma show that SAP15 is effective in reduced proinflammatory responses. CONCLUSION: These findings suggest that SAP15 is a novel peptide with a novel cell-penetrating peptide with anti-inflammatory property that can be used as a therapeutic agent for IBD and other inflammatory diseases.


Subject(s)
Cell-Penetrating Peptides , Inflammatory Bowel Diseases , Humans , Animals , Mice , Cell-Penetrating Peptides/adverse effects , Quality of Life , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/pathology , Cytokines/metabolism , Anti-Inflammatory Agents/pharmacology , Histone Deacetylases/adverse effects
2.
Int J Cancer ; 149(6): 1313-1321, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34019700

ABSTRACT

CIGB-552 is a synthetic peptide that interacts with COMMD1 and upregulates its protein levels. The objectives of this phase I study were safety, pharmacokinetic profile, evaluation of the lymphocytes CD4+ and CD8+ and preliminary activity in patients with advanced tumors. A 3 + 3 dose-escalation design with seven dose levels was implemented. Patients were included until a grade 3 related adverse event occurred and the maximum tolerated dose was reached. The patients received subcutaneous administration of CIGB-552 three times per week for 2 weeks. Single-dose plasma pharmacokinetics was characterized at two dose levels, and tumor responses were classified by RECIST 1.1. Twenty-four patients received CIGB-552. Dose-limiting toxicity was associated with a transient grade 3 pruritic maculopapular rash at a dose of 7.0 mg. The maximum tolerated dose was defined as 4.7 mg. Ten patients were assessable for immunological status. Seven patients had significant changes in the ratio CD4/CD8 in response to CIGB-552 treatment; three patients did not modify the immunological status. Stable disease was observed in five patients, including two metastatic soft sarcomas. We conclude that CIGB-552 at dose 4.7 mg was well tolerated with no significant adverse events and appeared to provide some clinical benefits.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell-Penetrating Peptides/administration & dosage , NF-kappa B/drug effects , Neoplasms/drug therapy , Adaptor Proteins, Signal Transducing/metabolism , Adult , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Injections, Subcutaneous , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Neoplasms/metabolism , Neoplasms/pathology , Research Design , Treatment Outcome
3.
Expert Opin Investig Drugs ; 30(2): 167-176, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33393390

ABSTRACT

INTRODUCTION: Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder caused by mutations in the dystrophin (DMD) gene. Most patients die from respiratory failure or cardiomyopathy. There are significant unmet needs for treatments for DMD as the standard of care is principally limited to symptom relief through treatments including steroids. AREAS COVERED: This review summarizes safety and efficacy in promising areas of DMD therapeutics - small molecules, stop codon readthrough, gene replacement, and exon skipping - under clinical examination from 2015-2020 as demonstrated in the NIH Clinical Trials and PubMed search engines. EXPERT OPINION: Currently, steroids persist as the most accessible medicine for DMD. Stop-codon readthrough, gene replacement, and exon-skipping therapies all aim to restore dystrophin expression. Of these strategies, gene replacement therapy has recently gained momentum while exon-skipping retains great traction. The  FDA approval of three exon-skipping antisense oligonucleotides illustrate this regulatory momentum, though the effectiveness and sequence design of eteplirsen remain controversial. Cell-penetrating peptides promise to more efficaciously treat DMD-related cardiomyopathy.The recent success of antisense therapies, however, poses major regulatory challenges. To fully realize the benefits of exon-skipping, including cocktail oligonucleotide-mediated multiple exon-skipping and oligonucleotide drugs for very rare mutations, regulatory challenges need to be addressed in coordination with scientific advances.


Subject(s)
Cell-Penetrating Peptides/therapeutic use , Dystrophin/genetics , Genetic Therapy , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides/therapeutic use , Animals , Cell-Penetrating Peptides/adverse effects , Codon, Terminator , Drug Development , Drugs, Investigational/adverse effects , Drugs, Investigational/therapeutic use , Exons , Gene Expression Regulation , Genetic Predisposition to Disease , Genetic Therapy/adverse effects , Humans , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/genetics , Mutation , Oligonucleotides/adverse effects , Steroids/adverse effects , Steroids/therapeutic use , Treatment Outcome
4.
Arterioscler Thromb Vasc Biol ; 40(12): 2990-3003, 2020 12.
Article in English | MEDLINE | ID: mdl-33028101

ABSTRACT

OBJECTIVE: Arterial thrombosis leading to ischemic injury worsens the prognosis of many patients with cardiovascular disease. PZ-128 is a first-in-class pepducin that reversibly inhibits PAR1 (protease-activated receptor 1) on platelets and other vascular cells by targeting the intracellular surface of the receptor. The TRIP-PCI (Thrombin Receptor Inhibitory Pepducin in Percutaneous Coronary Intervention) trial was conducted to assess the safety and efficacy of PZ-128 in patients undergoing cardiac catheterization with intent to perform percutaneous coronary intervention. Approach and Results: In this randomized, double-blind, placebo-controlled, phase 2 trial, 100 patients were randomly assigned (2:1) to receive PZ-128 (0.3 or 0.5 mg/kg), or placebo in a 2-hour infusion initiated just before the start of cardiac catheterization, on top of standard oral antiplatelet therapy. Rates of the primary end point of bleeding were not different between the combined PZ-128 doses (1.6%, 1/62) and placebo group (0%, 0/35). The secondary end points of major adverse coronary events at 30 and 90 days did not significantly differ but were numerically lower in the PZ-128 groups (0% and 2% in the PZ-128 groups, 6% and 6% with placebo, p=0.13, p=0.29, respectively). In the subgroup of patients with elevated baseline cardiac troponin I, the exploratory end point of 30-day major adverse coronary events + myocardial injury showed 83% events in the placebo group versus 31% events in the combined PZ-128 drug groups, an adjusted relative risk of 0.14 (95% CI, 0.02-0.75); P=0.02. CONCLUSIONS: In this first-in-patient experience, PZ-128 added to standard antiplatelet therapy appeared to be safe, well tolerated, and potentially reduced periprocedural myonecrosis, thus providing the basis for further clinical trials. Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT02561000.


Subject(s)
Acute Coronary Syndrome/therapy , Blood Platelets/drug effects , Cardiac Catheterization , Cell-Penetrating Peptides/administration & dosage , Coronary Artery Disease/therapy , Lipopeptides/administration & dosage , Myocardium/pathology , Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/administration & dosage , Receptor, PAR-1/agonists , Thrombosis/prevention & control , Acute Coronary Syndrome/diagnostic imaging , Aged , Blood Platelets/metabolism , Cardiac Catheterization/adverse effects , Cardiac Catheterization/instrumentation , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/pharmacokinetics , Coronary Artery Disease/diagnostic imaging , Double-Blind Method , Female , Humans , Infusions, Intravenous , Lipopeptides/adverse effects , Lipopeptides/pharmacokinetics , Male , Middle Aged , Necrosis , Percutaneous Coronary Intervention/adverse effects , Percutaneous Coronary Intervention/instrumentation , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Proof of Concept Study , Prospective Studies , Receptor, PAR-1/metabolism , Recurrence , Stents , Thrombosis/blood , Thrombosis/etiology , Time Factors , Treatment Outcome , United States
5.
Pharmacol Res Perspect ; 7(6): e00547, 2019 12.
Article in English | MEDLINE | ID: mdl-31832205

ABSTRACT

Activation of MrgX2, an orphan G protein-coupled receptor expressed on mast cells, leads to degranulation and histamine release. Human MrgX2 binds promiscuously to structurally diverse peptides and small molecules that tend to have basic properties (basic secretagogues), resulting in acute histamine-like adverse drug reactions of injected therapeutic agents. We set out to identify MrgX2 orthologues from other mammalian species used in nonclinical stages of drug development. Previously, the only known orthologue of human MrgX2 was from mouse, encoded by Mrgprb2. MrgX2 genes of rat, dog (beagle), minipig, pig, and Rhesus and cynomolgus monkey were identified by bioinformatic approaches and verified by their ability to mediate calcium mobilization in transfected cells in response to the classical MrgX2 agonist, compound 48/80. The peptide GSK3212448 is an inhibitor of the PRC2 epigenetic regulator that caused profound anaphylactoid reactions upon intravenous infusion to rat. We showed GSK3212448 to be a potent MrgX2 agonist particularly at rat MrgX2. We screened sets of drug-like molecules and peptides to confirm the highly promiscuous nature of MrgX2. Approximately 20% of drug-like molecules activated MrgX2 (pEC50 ranging from 4.5 to 6), with the principle determinant being basicity. All peptides tested of net charge +3 or greater exhibited agonist activity, including the cell penetrating peptides polyarginine (acetyl-Arg9-amide) and TAT (49-60), a fragment of HIV-1 TAT protein. Finally, we showed that the glycopeptide antibiotic vancomycin, which is associated with clinical pseudo-allergic reactions known as red man syndrome, is an agonist of MrgX2.


Subject(s)
Anaphylaxis/chemically induced , Mast Cells/drug effects , Nerve Tissue Proteins/agonists , Peptide Fragments/adverse effects , Receptors, G-Protein-Coupled/agonists , Receptors, Neuropeptide/agonists , Vancomycin/adverse effects , Anaphylaxis/immunology , Animals , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cell Line, Tumor , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/adverse effects , Disease Models, Animal , Drug Evaluation, Preclinical/adverse effects , HEK293 Cells , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Humans , Mast Cells/immunology , Mast Cells/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Peptide Fragments/administration & dosage , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/immunology , Receptors, Neuropeptide/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Syndrome , Vancomycin/administration & dosage , p-Methoxy-N-methylphenethylamine/pharmacology
6.
Molecules ; 24(5)2019 Mar 07.
Article in English | MEDLINE | ID: mdl-30866424

ABSTRACT

Cell-penetrating-peptides (CPPs) are small amino-acid sequences characterized by their ability to cross cellular membranes. They can transport various bioactive cargos inside cells including nucleic acids, large proteins, and other chemical compounds. Since 1988, natural and synthetic CPPs have been developed for applications ranging from fundamental to applied biology (cell imaging, gene editing, therapeutics delivery). In recent years, a great number of studies reported the potential of CPPs as carriers for the treatment of various diseases. Apart from a good efficacy due to a rapid and potent delivery, a crucial advantage of CPP-based therapies is the peptides low toxicity compared to most drug carriers. On the other hand, they are quite unstable and lack specificity. Higher specificity can be obtained using a cell-specific CPP to transport the therapeutic agent or using a non-specific CPP to transport a cargo with a targeted activity. CPP-cargo complexes can also be conjugated to another moiety that brings cell- or tissue-specificity. Studies based on all these approaches are showing promising results. Here, we focus on recent advances in the potential usage of CPPs in the context of cancer therapy, with a particular interest in CPP-mediated delivery of anti-tumoral proteins.


Subject(s)
Cell-Penetrating Peptides/chemistry , Drug Carriers/chemistry , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell-Penetrating Peptides/adverse effects , Drug Carriers/adverse effects , Humans , Organ Specificity
7.
Sci Rep ; 9(1): 3312, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30824773

ABSTRACT

Activated proximal tubular epithelial cells (PTECs) play a crucial role in progressive tubulo-interstitial fibrosis in native and transplanted kidneys. Targeting PTECs by non-viral delivery vectors might be useful to influence the expression of important genes and/or proteins in order to slow down renal function loss. However, no clinical therapies that specifically target PTECs are available at present. We earlier showed that a cationic cell penetrating peptide isolated from South American rattlesnake venom, named crotamine, recognizes cell surface heparan sulfate proteoglycans and accumulates in cells. In healthy mice, crotamine accumulates mainly in kidneys after intraperitoneal (ip) injection. Herein we demonstrate for the first time, the overall safety of acute or long-term treatment with daily ip administrated crotamine for kidneys functions. Accumulation of ip injected crotamine in the kidney brush border zone of PTECs, and its presence inside these cells were observed. In addition, significant lower in vitro crotamine binding, uptake and reporter gene transport and expression could be observed in syndecan-1 deficient HK-2 PTECs compared to wild-type cells, indicating that the absence of syndecan-1 impairs crotamine uptake into PTECs. Taken together, our present data show the safety of in vivo long-term treatment with crotamine, and its preferential uptake into PTECs, which are especially rich in HSPGs such as syndecan-1. In addition to the demonstrated in vitro gene delivery mediated by crotamine in HK-2 cells, the potential applicability of crotamine as prototypic non-viral (gene) delivery nanocarrier to modulate PTEC gene and/or protein expression was confirmed.


Subject(s)
Cell-Penetrating Peptides , Crotalid Venoms , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Animals , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/pharmacokinetics , Cell-Penetrating Peptides/pharmacology , Crotalid Venoms/adverse effects , Crotalid Venoms/pharmacokinetics , Crotalid Venoms/pharmacology , Epithelial Cells/cytology , Kidney Tubules, Proximal/cytology , Male , Mice
8.
São Paulo; s.n; s.n; 2019. 126 p. ilus, graf, tab.
Thesis in Portuguese | LILACS | ID: biblio-1007440

ABSTRACT

O desenvolvimento de resistência antimicrobiana e a consequente seleção de microrganismos multirresistentes consolidam-se como grandes ameaças à saúde global. Neste contexto, a busca por novas drogas antimicrobianas/microbicidas é fundamental e compostos como os peptídeos antimicrobianos (AMPs) tornaram-se alvos atraentes. Os AMPs são compostos químicos de massa molar média e grande diversidade estrutural, produzidos por todos os seres vivos e com capacidade de inibir o crescimento de e/ou matar microrganismos. O AMP Cheferina I (Chef I) foi isolado das raízes de Capsella bursa-pastoris e é resultado da proteólise de uma proteína da família das proteínas ricas em glicina, que em plantas estão relacionadas às funções de defesa e cicatrização. O nosso grupo de pesquisa foi pioneiro no desenvolvimento e estudo de análogos truncados amidados deste AMP atípico rico em glicina (67,9%) e histidina (28,6%), que se mostraram ativos frente às diferentes cepas de Candida e a S. cerevisiae pela internalização/ação celular acompanhada de manutenção da integridade da membrana plasmática; o análogo amidado (Chef Ia) e o análogo marcado com 5(6)-carboxifluoresceína/FAM (FAM-Chef Ia) tiveram as suas atividades antifúngicas potencializadas por íons Zn2+. Este trabalho deu continuidade ao estudo do efeito dos íons metálicos divalentes Zn2+, Cu2+, Ca2+ e Mg2+ nas atividades anticandida/fungistática e candidacida/fungicida a diferentes pHs e forças iônicas, estruturas e localizações intracelulares destes análogos. Os resultados na ausência de íons em pH 5,1 revelaram maior atividade do análogo fluorescente em relação à do não fluorescente. Neste mesmo pH, as atividades anticandida e candidacida de Chef Ia foram influenciadas negativamente pelos íons Ca2+ e Mg2+ (2-4 vezes) enquanto que, na presença de íons Zn2+ as atividades anticandida de ambos os análogos foram aumentadas (Chef Ia: 8-64 vezes; FAM-Chef Ia: 4-32 vezes). Os íons Cu2+ aumentaram a atividade anticandida de Chef Ia (2-4 vezes), mas não a do análogo fluorescente, mas as atividades candidacidas de ambos foram melhoradas (Chef Ia: 2-8 vezes; FAM-Chef Ia: 2 vezes). Em pH 5,1, os íons Zn2+ mantiveram a atividade anticandida de Chef Ia em alta força iônica, mas só FAM-Chef Ia exibiu atividade candidacida. Em pH 7,4 ambos análogos foram inativos em baixa e alta forças iônicas na ausência e presença de Zn2+ ou Cu2+. As maiores porcentagens de folhas-ß-antiparalelas e dobras foram observadas no espectro de DC de Chef Ia em pH 7,4, sendo que aqueles registrados em pH 5,1 e 7,4 em presença de íons Zn2 e Cu2+ indicaram a formação de quelatos estruturalmente distintos. Ambos os peptídeos são bioquelantes em potencial, sendo as proporções peptídeo: íon obtidas as seguintes: FAM-Chef Ia = 1:2 para Cu2+, 1:10 para Zn2+; Chef Ia = 1:1 para Cu2+. A análise da internalização celular de FAM-Chef Ia permitiu a suposição de dois mecanismos de internalização (translocação direta e endocitose), sendo que nas células vivas a presença de Zn2+ afetou negativamente a translocação direta (p 0,0343) e potencializou a endocitose (p 0,0002)


The development of antimicrobial resistance and the consequent selection of multiresistant microorganisms have become major threats to global health. In this context, the search for new antimicrobial/microbicidal drugs is crucial and the antimicrobial peptides (AMPs) have been seen as attractive targets. AMPs are chemical compounds of medium molecular mass and high structural diversity produced by all living beings, capable of inhibiting the growth of microorganisms and killing them. The AMP Shepherin I (Shep I) was isolated from the roots of Capsella bursa-pastoris, being a bioactive peptide encrypted in a glycine-rich protein from a family that in plants are strictly related to defense and healing functions. Our research group has pioneered the development and study of amidated truncated analogues of this atypical glycine- (67.9%) and histidine-rich (28.6%) AMP, which has shown activity against different strains of Candida and S. cerevisiae through cellular internalization with maintenance of the plasma membrane integrity. The amide analogue (Chef Ia) and its fluorescent analog labeled with 5 (6) - carboxyfluorescein / FAM (FAM-Chef Ia) had their antifungal activities potentiated by Zn2+ ions, so the present work continued examining the effect of the divalent metallic ions Zn2+, Cu2+, Ca2+ and Mg2+ on the anticandidal/fungistatic and candidacidal/fungicide activities at different pHs and ionic forces, structures and intracellular locations of these analogues. The results in the absence of those ions at pH 5.1 revealed that the fluorescently labelled analog was more potent than the nonfluorescent. At the same pH, Shep Ia anticandidal and candidacidal activities were negatively influenced by Ca2+ and Mg2+ ions (2-4 fold), whereas in the presence of Zn2+ ions the anticandidal activities of both analogues were increased (Shep Ia: 8-64 fold, FAM- Shep Ia: 4-32 fold). Cu2+ ions increased Shep Ia anticandidal activity (2-4 fold) but not that of FAM-Shep Ia, nevertheless, the candidacidal activities of both analogues were increased (Shep Ia: 2-8 fold, FAM-Shep Ia: 2 fold). Also at pH 5.1, the Zn2+ ions helped retaining the anticandidal activity of Shep Ia at high ionic strength, although only FAM-Shep Ia exhibited candidacidal activity. At pH 7.4 both analogues were inactive at low and high ionic strengths in the absence or presence of Zn2+ or Cu2+. The highest percentages of antiparallel ß-sheet and turns were observed in Shep Ia CD spectrum at pH 7.4, while those recorded at pH 5.1 and 7.4 in the presence of Zn2+ or Cu2+ ions indicated the formation of structurally different chelates. Both peptides are potential biochelates, with the following peptide:ion ratios: FAM-Shep Ia = 1: 2 for Cu2+, 1:10 for Zn2+; Shep Ia = 1: 1 for Cu2+. The analysis of the cellular internalization of FAM-Chef Ia allowed the assumption of two mechanisms of internalization (direct translocation and endocytosis) and in the living cells the presence of Zn2+ negatively affected the direct translocation (p 0.0343) and potentiated endocytosis (p 0.0002)


Subject(s)
Cell-Penetrating Peptides/adverse effects , Anti-Infective Agents/analysis , Plant Roots/adverse effects , Capsella/anatomy & histology
9.
Biomed Pharmacother ; 108: 1090-1096, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30372809

ABSTRACT

INTRODUCTION: The biological membranes as natural permeable barriers are required for survival and function of living cells. However, these natural barriers could be a major obstacle for the efficient intracellular delivery of therapeutic agents. MATERIALS AND METHODS: In recent two decades, the use of peptides as novel carriers for intracellular cargo delivery has been received more attention by introducing the cell penetrating peptides (CPPs). CPPs, protein transduction domains, are an attractive class of short peptide sequences which can translocate across the cell membrane. CONCLUSION: Owing to the ability of CPPs to transport across cellular membrane, they can employ as an appropriate carrier for various cargos include nucleic acid, proteins, SiRNA, therapeutic agents, nanoparticles and so on. In this review, we describe the classifications of CPPs, their uptake mechanisms as well as biomedical applications of the CPPs.


Subject(s)
Biomedical Technology , Cell-Penetrating Peptides/pharmacology , Amino Acid Sequence , Animals , Cell Death/drug effects , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/chemistry , Endocytosis , Humans
10.
Drug Deliv ; 25(1): 757-765, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29528244

ABSTRACT

Tumor-targeted delivery is considered a crucial component of current anticancer drug development and is the best approach to increase the efficacy and reduce the toxicity. Nanomedicine, particularly ligand-based nanoparticles have shown a great potential for active targeting of tumor. Cell penetrating peptide is one of the promising ligands in a targeted cancer therapy. In this study, the gambogic acid-loaded nanostructured lipid carrier (GA-NLC) was modified with two kinds of cell penetrating peptides (cRGD and RGERPPR). The GA-NLC was prepared by emulsification and solvent evaporation method and coupled with cRGD, RGERPPR, and combination cRGD and RGERPPR to form GA-NLC-cRGD, GA-NLC-RGE, and GA-NLC-cRGD/RGE, respectively. The formulations were characterized by their particle size and morphology, zeta potential, encapsulation efficiency, and differential scanning calorimetry. In vitro cytotoxicity and cellular uptake study of the formulations were performed against breast cancer cell (MDA-MB-231). Furthermore, in vivo biodistribution and antitumor activity of the formulations were determined by in vivo imaging and in tumor-bearing nude mice, respectively. The result of in vitro cytotoxicity study showed that GA-NLC-RGE exhibited a significantly higher cytotoxicity on MDA-MB-231 as compared with GA-NLC and GA-Sol. Similarly, RGE-Cou-6-NLC showed remarkably higher uptake by the cells than other NLCs over the incubation period. The in vivo imaging study has demonstrated that among the formulations, the RGE-decorated DiR-NLC were more accumulated in the tumor site. The in vivo antitumor activity revealed that RGE-GA-NLC inhibits the tumor growth more efficiently than other formulations. In conclusion, RGERPPR has a potential as an effective carrier in targeting drug delivery of anticancer agents.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Breast Neoplasms/drug therapy , Cell-Penetrating Peptides/chemistry , Drug Carriers/administration & dosage , Nanostructures/chemistry , Xanthones/administration & dosage , Absorption, Physiological , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell-Penetrating Peptides/adverse effects , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Drug Carriers/therapeutic use , Drug Compounding , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Microscopy, Electron, Transmission , Nanostructures/adverse effects , Nanostructures/ultrastructure , Particle Size , Random Allocation , Surface Properties , Tissue Distribution , Tumor Burden/drug effects , Xanthones/pharmacokinetics , Xanthones/pharmacology , Xanthones/therapeutic use , Xenograft Model Antitumor Assays
11.
Tissue Barriers ; 4(2): e1156805, 2016.
Article in English | MEDLINE | ID: mdl-27358754

ABSTRACT

Numerous approaches have been explored to date in the pursuit of delivering peptides or proteins via the oral route. One such example is chemical modification, whereby the native structure of a peptide or protein is tailored to provide a more efficient uptake across the epithelial barrier of the gastrointestinal tract via incorporation of a chemical motif or moiety. In this regard, a diverse array of concepts have been reported, ranging from the exploitation of endogenous transport mechanisms to incorporation of physicochemical modifications in the molecule, which promote more favorable interactions with the absorptive membrane at the cell surface. This review provides an overview of the modification technologies described in the literature and offers insights into some pragmatic considerations pertaining to their translation into clinically viable concepts.


Subject(s)
Administration, Oral , Cell-Penetrating Peptides/pharmacokinetics , Animals , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/chemistry , Humans , Intestinal Absorption
12.
Tissue Barriers ; 4(2): e1178369, 2016.
Article in English | MEDLINE | ID: mdl-27358757

ABSTRACT

Non-injectable delivery of peptide and protein drugs is hampered by their labile nature, hydrophilicity, and large molecular size; thus limiting their permeation across mucosae, which represent major biochemical and physical barriers to drugs administered via e.g. the oral, nasal, and pulmonary routes. However, in recent years cell-penetrating peptides (CPP) have emerged as promising tools to enhance mucosal delivery of co-administered or conjugated peptide and protein cargo and more advanced CPP-cargo formulations are emerging. CPPs act as transepithelial delivery vectors, but the mechanism(s) by which CPPs mediate cargo translocation across an epithelium is so far poorly understood; both due to the fact that multiple factors influence the resulting uptake and trafficking mechanisms as well as to the complicated nature of sensitive studies of this. In addition to a proper mechanistic understanding, documentation of CPP-mediated delivery in higher animal species than rodent as well as extensive toxicological studies are necessary for CPP-containing non-injectable DDSs to reach the clinic.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Drug Delivery Systems/methods , Epithelium/metabolism , Animals , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/pharmacokinetics , Humans , Intestinal Absorption
13.
Theranostics ; 6(2): 177-91, 2016.
Article in English | MEDLINE | ID: mdl-26877777

ABSTRACT

Cell penetrating peptides (CPPs) were widely used for drug delivery to tumor. However, the nonselective in vivo penetration greatly limited the application of CPPs-mediated drug delivery systems. And the treatment of malignant tumors is usually followed by poor prognosis and relapse due to the existence of extravascular core regions of tumor. Thus it is important to endue selective targeting and stronger intratumoral diffusion abilities to CPPs. In this study, an RGD reverse sequence dGR was conjugated to a CPP octa-arginine to form a CendR (R/KXXR/K) motif contained tandem peptide R8-dGR (RRRRRRRRdGR) which could bind to both integrin αvß3 and neuropilin-1 receptors. The dual receptor recognizing peptide R8-dGR displayed increased cellular uptake and efficient penetration ability into glioma spheroids in vitro. The following in vivo studies indicated the active targeting and intratumoral diffusion capabilities of R8-dGR modified liposomes. When paclitaxel was loaded in the liposomes, PTX-R8-dGR-Lip induced the strongest anti-proliferation effect on both tumor cells and cancer stem cells, and inhibited the formation of vasculogenic mimicry channels in vitro. Finally, the R8-dGR liposomal drug delivery system prolonged the medium survival time of intracranial C6 bearing mice by 2.1-fold compared to the untreated group, and achieved an exhaustive anti-glioma therapy including anti-tumor cells, anti-vasculogenic mimicry and anti-brain cancer stem cells. To sum up, all the results demonstrated that R8-dGR was an ideal dual receptor recognizing CPP with selective glioma targeting and efficient intratumoral diffusion, which could be further used to equip drug delivery system for effective glioma therapy.


Subject(s)
Brain Neoplasms/drug therapy , Cell-Penetrating Peptides/pharmacokinetics , Glioma/drug therapy , Integrin alphaVbeta3/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/chemistry , HeLa Cells , Humans , Liposomes/adverse effects , Liposomes/pharmacokinetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Oligopeptides/adverse effects , Oligopeptides/physiology , Protein Binding
14.
Arterioscler Thromb Vasc Biol ; 36(1): 189-97, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26681756

ABSTRACT

OBJECTIVE: Pepducins are membrane-tethered, cell-penetrating lipopeptides that target the cytoplasmic surface of their cognate receptor. Here, we report the first human use of a protease-activated receptor-1-based pepducin, which is intended as an antiplatelet agent to prevent ischemic complications of percutaneous coronary interventions. APPROACH AND RESULTS: PZ-128 was administered by 1 to 2 hours continuous intravenous infusion (0.01-2 mg/kg) to 31 subjects with coronary artery disease or multiple coronary artery disease risk factors. Safety, antiplatelet efficacy, and pharmacokinetics were assessed at baseline and 0.5, 1, 2, 6, 24 hours, and 7 to 10 days postdosing. The inhibitory effects of PZ-128 on platelet aggregation stimulated by the protease-activated receptor-1 agonist SFLLRN (8 µmol/L) at 30 minutes to 6 hours were dose dependent with 20% to 40% inhibition at 0.3 mg/kg, 40% to 60% at 0.5 mg/kg, and ≥ 80% to 100% at 1 to 2 mg/kg. The subgroup receiving aspirin in the 0.5 and 1-mg/kg dose cohorts had 65% to 100% inhibition of final aggregation to SFLLRN at 30 minutes to 2 hours and 95% to 100% inhibition by 6 hours. The inhibitory effects of 0.5 mg/kg PZ-128 were reversible with 50% recovery of aggregation to SFLLRN by 24 hours. There were no significant effects of PZ-128 on aggregation induced by AYPGKF, ADP, or collagen, indicating that the observed effects were specific to protease-activated receptor-1. The plasma half-life was 1.3 to 1.8 hours, and PZ-128 was nondetectable in urine. There were no effects on bleeding, coagulation, clinical chemistry, or ECG parameters. CONCLUSIONS: PZ-128 is a promising antiplatelet agent that provides rapid, specific, dose dependent, and reversible inhibition of platelet protease-activated receptor-1 through a novel intracellular mechanism. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT01806077.


Subject(s)
Blood Platelets/drug effects , Cell-Penetrating Peptides/administration & dosage , Coronary Artery Disease/therapy , Lipopeptides/administration & dosage , Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation/drug effects , Receptor, PAR-1/antagonists & inhibitors , Adult , Aged , Blood Platelets/metabolism , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/pharmacokinetics , Coronary Artery Disease/blood , Coronary Artery Disease/diagnosis , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Infusions, Intravenous , Lipopeptides/adverse effects , Lipopeptides/pharmacokinetics , Male , Middle Aged , Percutaneous Coronary Intervention/adverse effects , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Function Tests , Receptor, PAR-1/metabolism , Treatment Outcome
16.
Mol Pharm ; 12(5): 1584-91, 2015 May 04.
Article in English | MEDLINE | ID: mdl-25828697

ABSTRACT

A drug delivery system designed specifically for oligonucleotide therapeutics can ameliorate the problems associated with the in vivo delivery of these molecules. The internalization of free oligonucleotides is challenging, and cytotoxicity is the main obstacle for current transfection vehicles. To develop nontoxic delivery vehicles for efficient transfection of oligonucleotides, we designed a self-assembling peptide amphiphile (PA) nanosphere delivery system decorated with cell penetrating peptides (CPPs) containing multiple arginine residues (R4 and R8), and a cell surface binding peptide (KRSR), and report the efficiency of this system in delivering G-3129, a Bcl-2 antisense oligonucleotide (AON). PA/AON (peptide amphiphile/antisense oligonucleotide) complexes were characterized with regards to their size and secondary structure, and their cellular internalization efficiencies were evaluated. The effect of the number of arginine residues on the cellular internalization was investigated by both flow cytometry and confocal imaging, and the results revealed that uptake efficiency improved as the number of arginines in the sequence increased. The combined effect of cell penetration and surface binding property on the cellular internalization and its uptake mechanism was also evaluated by mixing R8-PA and KRSR-PA. R8 and R8/KRSR decorated PAs were found to drastically increase the internalization of AONs compared to nonbioactive PA control. Overall, the KRSR-decorated self-assembled PA nanospheres were demonstrated to be noncytotoxic delivery vectors with high transfection rates and may serve as a promising delivery system for AONs.


Subject(s)
Cell-Penetrating Peptides/chemistry , Nanospheres/chemistry , Oligonucleotides, Antisense/chemistry , Cell Survival/drug effects , Cell-Penetrating Peptides/adverse effects , Circular Dichroism , Flow Cytometry , Humans , MCF-7 Cells , Microscopy, Confocal , Microscopy, Electron, Transmission
17.
Eur J Pharm Biopharm ; 91: 91-102, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25660910

ABSTRACT

The aim of the present study was to develop a novel strategy to deliver intracellularly the peptide GSE24.2 for the treatment of Dyskeratosis congenita (DC) and other defective telomerase disorders. For this purpose, biodegradable polymeric nanoparticles using poly(lactic-co-glycolic acid) (PLGA NPs) or poly(lactic-co-glycolic acid)-poly ethylene glycol (PLGA-PEG NPs) attached to either polycations or cell-penetrating peptides (CPPs) were prepared in order to increase their cellular uptake. The particles exhibited an adequate size and zeta potential, with good peptide loading and a biphasic pattern obtained in the in vitro release assay, showing an initial burst release and a later sustained release. GSE24.2 structural integrity after encapsulation was assessed using SDS-PAGE, revealing an unaltered peptide after the NPs elaboration. According to the cytotoxicity results, cell viability was not affected by uncoated polymeric NPs, but the incorporation of surface modifiers slightly decreased the viability of cells. The intracellular uptake exhibited a remarkable improvement of the internalization, when the NPs were conjugated to the CPPs. Finally, the bioactivity, addressed by measuring DNA damage rescue and telomerase reactivation, showed that some formulations had the lowest cytotoxicity and highest biological activity. These results proved that GSE24.2-loaded NPs could be delivered to cells, and therefore, become an effective approach for the treatment of DC and other defective telomerase syndromes.


Subject(s)
Biocompatible Materials/chemistry , Cell Cycle Proteins/chemistry , Drug Delivery Systems , Enzyme Reactivators/chemistry , Nanoparticles/chemistry , Nuclear Proteins/chemistry , Peptide Fragments/chemistry , Animals , Biocompatible Materials/adverse effects , Biological Transport , Cell Cycle Proteins/administration & dosage , Cell Cycle Proteins/adverse effects , Cell Cycle Proteins/genetics , Cell Line , Cell Survival/drug effects , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/chemistry , Cells, Cultured , Chemical Phenomena , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/adverse effects , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/therapeutic use , Drug Compounding , Drug Delivery Systems/adverse effects , Drug Liberation , Drug Stability , Dyskeratosis Congenita/drug therapy , Enzyme Reactivators/administration & dosage , Enzyme Reactivators/adverse effects , Enzyme Reactivators/therapeutic use , Humans , Lactic Acid/adverse effects , Lactic Acid/chemistry , Mice , Nanoparticles/adverse effects , Nuclear Proteins/administration & dosage , Nuclear Proteins/adverse effects , Nuclear Proteins/genetics , Peptide Fragments/administration & dosage , Peptide Fragments/adverse effects , Peptide Fragments/genetics , Polyamines/adverse effects , Polyamines/chemistry , Polyelectrolytes , Polyethylene Glycols/adverse effects , Polyethylene Glycols/chemistry , Polyglactin 910/adverse effects , Polyglactin 910/chemistry , Polyglycolic Acid/adverse effects , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Protein Stability , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/chemistry , Recombinant Proteins/therapeutic use
18.
Drug Dev Ind Pharm ; 41(4): 617-22, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24564798

ABSTRACT

The generation 4-poly-amidoamine-dendrimers (PAMAM G4 dendrimer, P) was conjugated to Tat peptide (Tat, T), a cell-penetrating peptide, in search of an efficient anti-tumor drug delivery vehicle for cancer therapy. In this study, we synthesized BODIPY-labeled Tat-Conjugated PAMAM dendrimers (BPTs) as a novel nanosized anticancer drug carriers and systemically investigated their biodistribution and the tumor accumulation in Sarcoma 180-bearing mice. In addition, the uptake and the cytotoxicity to S180 cells of BPTs thereof were evaluated. The unmodified dendrimer (BP) showed a soon clearance from the blood stream and nonspecific accumulation in tumor. In contrast, the Tat-modified dendrimer, BPT(64) with appropriate particle size showed a better retention in blood and could be accumulated effectively in tumor tissue via the enhanced permeability and retention (EPR) effect. Moreover, BPTs with a high Tat modification rate was accumulated more effectively in tumor tissue. In vitro experiments, these BPTs displayed low cytotoxicity on S180 cells and high uptake to S180 cells. These findings indicate that the nanoparticulate system on the basis of Tat-conjugated PAMAM dendrimers is safer and effective in the concentration range (below 20 µg/ml) to be used as a carrier of anti-tumor drugs for tumor targeting by intravenous administration.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell-Penetrating Peptides/chemistry , Dendrimers/chemistry , Drug Delivery Systems , Nanostructures/chemistry , Sarcoma 180/drug therapy , tat Gene Products, Human Immunodeficiency Virus/chemistry , Absorption, Physiological , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biological Availability , Cell Line, Tumor , Cell Survival/drug effects , Cell-Penetrating Peptides/adverse effects , Chemical Phenomena , Dendrimers/adverse effects , Fluorescent Dyes/chemistry , Injections, Intravenous , Male , Mice, Inbred BALB C , Nanostructures/adverse effects , Particle Size , Peptide Fragments/adverse effects , Peptide Fragments/chemistry , Sarcoma 180/blood , Sarcoma 180/metabolism , Tissue Distribution , tat Gene Products, Human Immunodeficiency Virus/adverse effects
19.
J Pept Sci ; 20(10): 760-84, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25112216

ABSTRACT

The penetration of polar or badly soluble compounds through a cell membrane into live cells requires mechanical support or chemical helpers. Cell-penetrating peptides (CPPs) are very promising chemical helpers. Because of their low cytotoxicity and final degradation to amino acids, they are particularly favored in in vivo studies and for clinical applications. Clearly, the future of CPP research is bright; however, the required optimization studies for each drug require considerable individualized attention. Thus, CPPs are not the philosopher's stone. As of today, a large number of such transporter peptides with very different sequences have been identified. These have different uptake mechanisms and can transport different cargos. Intracellular concentrations of cargos can reach a low micromole range and are able to influence intracellular reactions. Internalized ribonucleic acids such as small interfering RNA (siRNA) and mimics of RNA such as peptide nucleic acids, morpholino nucleic acids, and triesters of oligonucleotides can influence transcription and translation. Despite the highly efficient internalization of antibodies, enzymes, and other protein factors, as well as siRNA and RNA mimics, the uptake and stabile insertion of DNA into the genome of the host cells remain substantially challenging. This review describes a wide array of differing CPPs, cargos, cell lines, and tissues. The application of CPPs is compared with electroporation, magnetofection, lipofection, viral vectors, dendrimers, and nanoparticles, including commercially available products. The limitations of CPPs include low cell and tissue selectivity of the first generation and the necessity for formation of fusion proteins, conjugates, or noncovalent complexes to different cargos and of cargo release from intracellular vesicles. Furthermore, the noncovalent complexes require a strong molar excess of CPPs, and extensive experimentation is required to determine the most optimal CPP for any given cargo and cell type. Yet to predict which CPP is optimal for any given target remains a complex question. More recently, there have been promising developments: the enhancement of cell specificity using activatable CPPs, specific transport into cell organelles by insertion of corresponding localization sequences, and the transport of drugs through blood-brain barriers, through the conjunctiva of eyes, skin, and into nerve cells. Proteins, siRNA, and mimics of oligonucleotides can be efficiently transported into cells and have been tested for treatment of certain diseases. The recent state of the art in CPP research is discussed together with the overall scope, limitations, and some recommendations for future research directions.


Subject(s)
Cell-Penetrating Peptides/therapeutic use , Drug Delivery Systems/methods , Genetic Therapy/methods , Models, Biological , Animals , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/pharmacology , Drug Delivery Systems/adverse effects , Endocytosis , Genetic Therapy/adverse effects , Humans , Organ Specificity , Protein Transport , Translational Research, Biomedical/trends
20.
Mol Pharm ; 11(5): 1471-8, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24708261

ABSTRACT

New therapeutic strategies are required to develop candidate drugs and ensure efficient delivery of these drugs to the brain and the central nervous system (CNS). Small interfering RNA (siRNA)-based therapies have been investigated as potential novel approaches for the treatment of brain disorders. Previously, we showed that Tat, a cell-penetrating peptide derived from HIV-Tat, and the modified block copolymers (MPEG-PCL-Tat) can form stable complexes with siRNA or can be loaded with an anticancer drug and efficiently deliver the drugs to the brain tissue via intranasal delivery. In this study, to develop a novel, efficient, and safe therapeutic strategy for managing brain disorders, we used MPEG-PCL-Tat micelles with a nose-to-brain delivery system to investigate its therapeutic effects on a rat model of malignant glioma using siRNA with a Raf-1 (siRaf-1)/camptothecin (CPT) codelivery system. MPEG-PCL-Tat and CPT-loaded MPEG-PCL-Tat can form a stable complex with siRNA with a particle size from 60 to 200 nm and a positive charge at N/P ratios up to 5. Additionally, MPEG-PCL-Tat/siRaf-1 and CPT-loaded MPEG-PCL-Tat/siRaf-1 have fostered cell death in rat glioma cells after the high cellular uptake of siRaf-1/drug by the MPEG-PCL-Tat carrier. Furthermore, compared to the unloaded MPEG-PCL-Tat/siRaf-1 complex, a CPT-loaded MPEG-PCL-Tat/siRaf-1 complex achieved the high therapeutic effect because of the additive effects of CPT and siRaf-1. These results indicate that drug/siRNA codelivery using MPEG-PCL-Tat nanomicelles with nose-to-brain delivery is an excellent therapeutic approach for brain and CNS diseases.


Subject(s)
Brain/metabolism , Camptothecin/therapeutic use , Cell-Penetrating Peptides/chemistry , Micelles , RNA, Small Interfering/physiology , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Brain/drug effects , Camptothecin/administration & dosage , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell-Penetrating Peptides/adverse effects , Disease Models, Animal , Glioma , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Male , Proto-Oncogene Proteins c-raf , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL