Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Liposome Res ; : 1-15, 2024 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-38459750

RESUMO

Recently, metformin (Met) has shown to have antineoplastic properties in cancer treatment by improving hypoxic tumor conditions, and causing reduction in the synthesis of biomolecules, which are vital for cancer growth. However, as an orally administered drug, Met has low bioavailability and rapid renal clearance. Thus, the goal of this study was to vectorize Met inside liposomes in the context of triple negative breast cancer (TNBC), which currently lacks treatment options when compared to other types of breast cancer. Vectorization of Met inside liposomes was done using Bangham method by implementing double design of experiment methodology to increase Met drug loading (minimum-run resolution V characterization design and Box-Behnken design), as it is generally extremely low for hydrophilic molecules. Optimization of Met-loaded liposome synthesis was successfully achieved with drug loading of 190 mg/g (19% w/w). The optimal Met-liposomes were 170 nm in diameter with low PdI (< 0.1) and negative surface charge (-20 mV), exhibiting sustained Met release at pH 7.4. The liposomal Met delivery system was stable over several months, and successfully reduced TNBC cell proliferation due to the encapsulated drug. This study is one the first reports addressing liposome formulation through thin-film hydration using two design of experiment methods aiming to increase drug loading of Met.

2.
Biomacromolecules ; 20(8): 3076-3086, 2019 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-31305991

RESUMO

The use of small interfering RNA (siRNA) to regulate oncogenes appears as a promising strategy in the context of cancer therapy, especially if they are vectorized by a smart delivery system. In this study, we investigated the cellular trafficking of a siRNA nanovector (called CS-MSN) functionalized with the cell-penetrating peptide gH625 in a triple-negative breast cancer model. With complementary techniques, we showed that siRNA nanovectors were internalized by both clathrin- and caveolae-mediated endocytosis. The presence of gH625 at the surface of the siRNA nanovector did not modify the entry pathway of CS-MSN, but it increased the amount of siRNA found inside the cells. Results suggested an escape of siRNA from endosomes, which is enhanced by the presence of the peptide gH625, whereas nanoparticles continued their trafficking into lysosomes. The efficiency of CS-MSN to inhibit the GFP in MDA-MB-231 cells was 1.7-fold higher than that of the nanovectors without gH625.


Assuntos
Peptídeos Penetradores de Células/administração & dosagem , Endocitose , Endossomos/metabolismo , Proteínas de Fluorescência Verde/antagonistas & inibidores , Nanopartículas/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Neoplasias de Mama Triplo Negativas/metabolismo , Movimento Celular , Feminino , Inativação Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lisossomos/metabolismo , Nanopartículas/química , RNA Interferente Pequeno/genética , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Células Tumorais Cultivadas
3.
Bioconjug Chem ; 29(11): 3516-3521, 2018 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-30352511

RESUMO

Antibody-drug conjugates (ADC) are spearheading vectorized chemotherapy against cancer, with 4 FDA-approved ADCs and 79 in clinical trials. However, most ADCs are produced using a stochastic bioconjugation method, target hematological cancers, and are derived from a full immunoglobulin-G (IgG). These factors limit their efficacy, especially against solid tumors which remain difficult to treat. Here we report the site-specific conjugation of a single auristatin derivative onto an engineered anti-HER2 single chain fragment variable (scFv) of the trastuzumab antibody, generating new scFv-drug conjugates (SDC). Two cysteines were judiciously incorporated at the beginning of the scFv hexahistidine tag, in order to allow controlled bioconjugation of a heterobifunctional linker including a second generation maleimide (SGM), either cleavable (for monomethyl auristatin E) or noncleavable (for monomethyl auristatin F). Our data indicated that both SDCs conserved their affinity to HER2 in comparison to the native scFv, and were efficiently able to kill in vitro HER2-positive SK-BR-3 cells at subnanomolar concentrations (EC50 of 0.68 nM and 0.32 nM). No effect was observed on HER2-negative MCF-7 cells. Ours results showed efficient targeting of site-specific SDCs against HER2-positive breast cancer cells. This work represents a first important step in the design of more effective small conjugates, paving the way for future in vivo translation to evaluate their full potential.


Assuntos
Aminobenzoatos/química , Neoplasias da Mama/tratamento farmacológico , Imunoconjugados/química , Imunoconjugados/farmacologia , Fatores Imunológicos/química , Fatores Imunológicos/farmacologia , Maleimidas/química , Oligopeptídeos/química , Receptor ErbB-2/efeitos dos fármacos , Anticorpos de Cadeia Única/química , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/imunologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Imunoconjugados/uso terapêutico , Fatores Imunológicos/uso terapêutico , Engenharia de Proteínas , Trastuzumab/química , Trastuzumab/imunologia
4.
Bioconjug Chem ; 29(5): 1553-1559, 2018 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-29553717

RESUMO

Biocompatible multifunctional nanomedicines (NMs) are known to be an attractive platform for targeted anticancer theranosis. However, these nanomedicines are of interest only if they efficiently target diseased cells and accumulate in tumors. Here we report the synthesis of a new generation of immunotargeted nanomedicines composed of a superparamagnetic iron oxide nanoparticle (SPION) core, polyethylene glycol coating and the anti-HER2 single chain fragment variable (scFv) of Trastuzumab antibody. We developed two novel bioengineered scFv carrying two cysteines located (i) at the end (4D5.1-cys2) or (ii) at the beginning (4D5.2-cys2) of its hexahistidine tag. The scFv bioconjugation was controlled via heterobifunctional linkers including a second generation maleimide (SGM). Our data indicated that the insertion of cysteines at the beginning of the hexahistidine tag was allowed to obtain nearly 2-fold conjugation efficiency (13 scFv/NP) compared to NMs using classical maleimide. As a result, the NMs-4D5.2 built using the optimal 4D5-cys2 and linkers equipped with SGM showed the enhanced recognition of HER2 in an ELISA format and on the surface of SK-BR-3 breast cancer cells in vitro. Their stability in serum was also significantly improved compared to the NMs-4D5. Our results showed the fundamental importance of the controlled ligand conjugation in the perspective of rational design of NMs with tailored physicochemical and biological properties.


Assuntos
Antineoplásicos Imunológicos/química , Imunoconjugados/química , Nanopartículas de Magnetita/química , Maleimidas/química , Anticorpos de Cadeia Única/química , Trastuzumab/química , Anticorpos Imobilizados/química , Anticorpos Imobilizados/farmacologia , Antineoplásicos Imunológicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Feminino , Humanos , Imunoconjugados/farmacologia , Maleimidas/farmacologia , Modelos Moleculares , Anticorpos de Cadeia Única/farmacologia , Trastuzumab/farmacologia
5.
Org Biomol Chem ; 16(11): 1882-1889, 2018 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-29473076

RESUMO

Herein we describe the synthesis and evaluation of four novel HER2-targeting, cathepsin B-sensitive antibody-drug conjugates bearing a monomethylauristatin E (MMAE) cytotoxic payload, constructed via the conjugation of cleavable linkers to trastuzumab using a site-specific bioconjugation methodology. These linkers vary by both cleavable trigger motif and hydrophilicity, containing one of two cathepsin B sensitive dipeptides (Val-Cit and Val-Ala), and engendered with either hydrophilic or hydrophobic character via application of a PEG12 spacer. Through evaluation of physical properties, in vitro cytotoxicity, and receptor affinity of the resulting antibody-drug conjugates (ADCs), we have demonstrated that while both dipeptide triggers are effective, the increased hydrophobicity of the Val-Ala pair limits its utility within this type of linker. In addition, while PEGylation augments linker hydrophilicity, this change does not translate to more favourable ADC hydrophilicity or potency. While all described structures demonstrated excellent and similar in vitro cytotoxicity, the ADC with the ValCitPABMMAE linker shows the most promising combination of in vitro potency, structural homogeneity, and hydrophilicity, warranting further evaluation into its therapeutic potential.


Assuntos
Antineoplásicos Imunológicos/química , Catepsina B/metabolismo , Imunoconjugados/química , Oligopeptídeos/química , Receptor ErbB-2/metabolismo , Trastuzumab/química , Antineoplásicos Imunológicos/síntese química , Antineoplásicos Imunológicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Química Sintética/métodos , Sistemas de Liberação de Medicamentos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Imunoconjugados/farmacologia , Neoplasias/tratamento farmacológico , Oligopeptídeos/síntese química , Oligopeptídeos/farmacologia , Trastuzumab/farmacologia
6.
J Nanobiotechnology ; 16(1): 18, 2018 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-29466990

RESUMO

BACKGROUND: Recent advances in nanomedicine have shown the great interest of active targeting associated to nanoparticles. Single chain variable fragments (scFv) of disease-specific antibodies are very promising targeting entities because they are small, not immunogenic and able to bind their specific antigens. The present paper is devoted to biological properties in vitro and in vivo of fluorescent and pegylated iron oxide nanoparticles (SPIONs-Cy-PEG-scFv) functionalized with scFv targeting Human Epithelial growth Receptor 2 (HER2). RESULTS: Thanks to a site-selective scFv conjugation, the resultant nanoprobes demonstrated high affinity and specific binding to HER2 breast cancer cells. The cellular uptake of SPIONs-Cy-PEG-scFv was threefold higher than that for untargeted PEGylated iron oxide nanoparticles (SPIONs-Cy-PEG) and is correlated to the expression of HER2 on cells. In vivo, the decrease of MR signals in HER2+ xenograft tumor is about 30% at 24 h after the injection. CONCLUSIONS: These results all indicate that SPIONs-Cy-PEG-scFv are relevant tumor-targeting magnetic resonance imaging agents, suitable for diagnosis of HER2 overexpressing breast tumor.


Assuntos
Neoplasias da Mama/diagnóstico por imagem , Compostos Férricos/química , Corantes Fluorescentes/química , Nanopartículas/química , Polietilenoglicóis/química , Receptor ErbB-2/análise , Anticorpos de Cadeia Única/química , Animais , Linhagem Celular Tumoral , Meios de Contraste/química , Feminino , Humanos , Imageamento por Ressonância Magnética/métodos , Camundongos Nus
7.
Biochim Biophys Acta Gen Subj ; 1861(6): 1578-1586, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27919801

RESUMO

BACKGROUND: This work is focused on mechanisms of uptake in cancer cells of rationally designed, covalently assembled nanoparticles, made of superparamagnetic iron oxide nanoparticles (SPIONs), fluorophores (doxorubicin or Nile Blue), polyethylene glycol (PEG) and folic acid (FA), referred hereinafter as SFP-FA. METHODS: SFP-FA were characterized by DLS, zetametry and fluorescence spectroscopy. The SFP-FA uptake in cancer cells was monitored using fluorescence-based methods like fluorescence-assisted cell sorting, CLSM with single-photon and two-photon excitation. The SFP-FA endocytosis was also analyzed with electron microscopy approaches: TEM, HAADF-STEM and EELS. RESULTS: The SFP-FA have zeta potential below -6mW and stable hydrodynamic diameter close to 100nm in aqueous suspensions of pH range from 5 to 8. They contain ca. 109 PEG-FA, 480 PEG-OCH3 and 22-27 fluorophore molecules per SPION. The fluorophores protected under the PEG shell allows a reliable detection of intracellular NPs. SFP-FA readily enter into all the cancer cell lines studied and accumulate in lysosomes, mostly via clathrin-dependent endocytosis, whatever the FR status on the cells. CONCLUSIONS: The present study highlights the advantages of rational design of nanosystems as well as the possible involvement of direct molecular interactions of PEG and FA with cellular membranes, not limited to FA-FR recognition, in the mechanisms of their endocytosis. GENERAL SIGNIFICANCE: Composition, magnetic and optical properties of the SFP-FA as well their ability to enter cancer cells are promising for their applications in cancer theranosis. Combination of complementary analytical approaches is relevant to understand the nanoparticles behavior in suspension and in contact with cells.


Assuntos
Antibióticos Antineoplásicos/metabolismo , Neoplasias da Mama/metabolismo , Clatrina/metabolismo , Doxorrubicina/metabolismo , Portadores de Fármacos , Endocitose , Ácido Fólico/metabolismo , Magnetismo/métodos , Nanopartículas de Magnetita , Nanomedicina/métodos , Polietilenoglicóis/química , Neoplasias do Colo do Útero/metabolismo , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Cavéolas/metabolismo , Vesículas Revestidas por Clatrina/metabolismo , Doxorrubicina/química , Doxorrubicina/farmacologia , Endossomos/metabolismo , Feminino , Ácido Fólico/química , Células HeLa , Humanos , Lisossomos/metabolismo , Células MCF-7 , Nanopartículas de Magnetita/química , Microscopia Confocal , Microscopia Eletrônica de Transmissão e Varredura , Microscopia de Fluorescência por Excitação Multifotônica , Espectroscopia de Perda de Energia de Elétrons , Neoplasias do Colo do Útero/tratamento farmacológico
8.
Anal Chem ; 87(22): 11233-41, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26488315

RESUMO

Novel magneto-plasmonic nanoprobes were designed for multimodal diagnosis of cancer by combination of magnetic resonance imaging (MRI), surface-enhanced resonance Raman scattering (SERRS), and fluorescence emission in the very near infrared (VNIR). A controlled electrostatic assembly of silver nanoparticles (AgNPs), superparamagnetic iron oxide nanoparticles (SPIONs), VNIR dye Nile Blue (NB), and biopolymer chitosan (Chi) was used to formulate the AgIONs-Chi nanoprobes. The formulation protocol did not involve organic solvents and was rapid and efficient as confirmed by magnetic sorting. The SERRS response of the nanoprobes was very intense and constant for days. It decreased linearly upon 1000-fold dilution and was still recognizable at 0.1 nM NB concentration. After 30 days of storage, the SERRS loss was less than 30% and the hydrodynamic size of the AgIONs-Chi in PBS remained below 200 nm. The gradual decrease of the ratio SERRS/fluorescence allowed one to monitor the release of the fluorescent molecule upon long-term nanoprobe dissociation. The AgIONs-Chi exhibited 2-fold higher MRI contrast than that of commercially available SPION suspensions. Finally, the nanoprobes were actively uptaken by HeLa cancer cells and ensured trimodal MRI-SERRS-fluorescence detection of 10 µL cell inclusions in cm-sized agarose gels used here as phantom models of microtumors. The above results show that the magneto-plasmonic AgIONs-Chi are promising substrates for SERRS analysis in solution and for multimodal imaging of cancer cells.


Assuntos
Separação Celular/métodos , Fluorescência , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita/química , Neoplasias/patologia , Análise de Célula Única , Células HeLa , Humanos , Campos Magnéticos , Tamanho da Partícula , Análise Espectral Raman , Propriedades de Superfície , Células Tumorais Cultivadas
9.
J Control Release ; 366: 567-584, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38215985

RESUMO

Trastuzumab emtansine (Kadcyla®) was the first antibody-drug conjugate (ADC) approved by the Food and Drug Administration in 2013 against a solid tumor, and the first ADC to treat human epidermal growth factor receptor 2 positive (HER2+) breast cancer. However, this second generation ADC is burden by several limitations included heterogeneity, limited activity against heterogeneous tumor (regarding antigen expression) and suboptimal tumor penetration. To address this, different development strategies are oriented towards homogeneous conjugation, new drugs, optimized linkers and/or smaller antibody formats. To reach better developed next generation ADCs, a key parameter to consider is the management of the hydrophobicity associated with the linker-drug, increasing with and limiting the drug-to-antibody ratio (DAR) of the ADC. Here, an innovative branched pegylated linker was developed, to control the hydrophobicity of the monomethyl auristatin E (MMAE) and its cathepsin B-sensitive trigger. This branched pegylated linker-MMAE was then used for the efficient generation of internalizing homogeneous ADC of DAR 8 and minibody-drug conjugate of DAR 4, targeting HER2. Both immunoconjugates were then evaluated in vitro and in vivo on breast cancer models. Interestingly, this study highlighted that the minibody-MMAE conjugate of DAR 4 was the best immunoconjugate regarding in vitro cellular internalization and cytotoxicity, gamma imaging, ex vivo biodistribution profile in mice and efficient reduction of tumor size in vivo. These results are very promising and encourage us to explore further fragment-drug conjugate development.


Assuntos
Aminobenzoatos , Neoplasias da Mama , Imunoconjugados , Oligopeptídeos , Estados Unidos , Camundongos , Humanos , Animais , Feminino , Neoplasias da Mama/tratamento farmacológico , Preparações Farmacêuticas , Distribuição Tecidual , Linhagem Celular Tumoral , Imunoconjugados/uso terapêutico , Ado-Trastuzumab Emtansina , Interações Hidrofóbicas e Hidrofílicas , Polietilenoglicóis
10.
Int J Pharm ; 632: 122335, 2023 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-36283640

RESUMO

This article was withdrawn from International Journal of Pharmaceutics in order to be published in International Journal of Pharmaceutics: X. The Publisher apologizes for any inconvenience this may cause.

11.
Int J Pharm X ; 4: 100126, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36147518

RESUMO

Chemoresistance and hence the consequent treatment failure is considerably challenging in clinical cancer therapeutics. The understanding of the genetic variations in chemoresistance acquisition encouraged the use of gene modulatory approaches to restore anti-cancer drug efficacy. Many smart nanoparticles are designed and optimized to mediate combinational therapy between nucleic acid and anti-cancer drugs. This review aims to define a rational design of such co-loaded nanocarriers with the aim of chemoresistance reversal at various cellular levels to improve the therapeutic outcome of anticancer treatment. Going through the principles of therapeutics loading, physicochemical characteristics tuning, and different nanocarrier modifications, also looking at combination effectiveness on chemosensitivity restoration. Up to now, these emerging nanocarriers are in development status but are expected to introduce outstanding outcomes.

12.
Pharmaceuticals (Basel) ; 15(12)2022 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-36558904

RESUMO

In this work, lipid-based nanoparticles (LBNP) were designed to combine tyrosine kinase inhibitor (TKI) Lapatinib (LAPA) with siRNA directed against apoptosis inhibitor protein Survivin (siSurvivin) in an injectable form. This nanosystem is based on lipid nanocapsules (LNCs) coated with a cationic polymeric shell composed of chitosan grafted through a transacylation reaction. The hydrophobic LAPA is solubilized in the inner oily core, while hydrophilic siRNA is associated electrostatically onto the nanocarrier's surface. The co-loaded LBNP showed a narrow size distribution (polydispersity index (PDI) < 0.3), a size of 130 nm, and a slightly positive zeta potential (+21 mV). LAPA and siRNA were loaded in LBNP at a high rate of >90% (10.6 mM) and 100% (4.6 µM), respectively. The siRNA-LAPA_LBNP was readily uptaken by the human epidermal growth factor receptor 2 overexpressed (HER2+) breast cancer cell line SK-BR-3. Moreover, the cytotoxicity studies confirmed that the blank chitosan decorated LBNP is not toxic to the cells with the tested concentrations, which correspond to LAPA concentrations from 1 to 10 µM, at different incubation times up to 96 h. Furthermore, siCtrl.-LAPA_LBNP had a more cytotoxic effect than Lapatinib salt, while siSurvivin-LAPA_LBNP had a significant synergistic cytotoxic effect compared to siCtrl.-LAPA_LBNP. All these findings suggested that the developed modified LBNP could potentiate anti-Survivin siRNA and LAPA anti-cancer activity.

13.
Pharmaceutics ; 14(8)2022 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-35893800

RESUMO

Self-assembled peptides possess remarkable potential as targeted drug delivery systems and key applications dwell anti-cancer therapy. Peptides can self-assemble into nanostructures of diverse sizes and shapes in response to changing environmental conditions (pH, temperature, ionic strength). Herein, we investigated the development of self-assembled peptide-based nanofibers (NFs) with the inclusion of a cell-penetrating peptide (namely gH625) and a matrix metalloproteinase-9 (MMP-9) responsive sequence, which proved to enhance respectively the penetration and tumor-triggered cleavage to release Doxorubicin in Triple Negative Breast Cancer cells where MMP-9 levels are elevated. The NFs formulation has been optimized via critical micelle concentration measurements, fluorescence, and circular dichroism. The final nanovectors were characterized for morphology (TEM), size (hydrodynamic diameter), and surface charge (zeta potential). The Doxo loading and release kinetics were studied in situ, by optical microspectroscopy (fluorescence and surface-enhanced Raman scattering-SERS). Confocal spectral imaging of the Doxo fluorescence was used to study the TNBC models in vitro, in cells with various MMP-9 levels, the drug delivery to cells as well as the resulting cytotoxicity profiles. The results confirm that these NFs are a promising platform to develop novel nanovectors of Doxo, namely in the framework of TNBC treatment.

14.
Pharmaceutics ; 14(11)2022 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-36432729

RESUMO

According to Globocan 2020, breast cancer is considered one of the most common cancers affecting women and is one of the leading causes of death in over 100 countries. The available classical treatment options do not always give satisfactory outcomes, and some patients develop resistance to these treatments. This study aims to investigate the combination of nanovectorized siRNA directed against anti-apoptotic protein Survivin (siSurvivin) by targeted stealth magnetic siRNA nanovectors (TS-MSN), designed in our lab, with Doxorubicin (DOX), as an option for HER2+ breast cancer treatment. The hypothesis is that the pretreatment of the HER2+ breast cancer cell line SK-BR-3 with siSurvivin will induce apoptosis in the cancer cells and enhance the therapeutic efficacy of DOX, allowing a dose reduction of DOX and hence a reduction of potential side effects. TS-MSN are based on superparamagnetic iron oxide nanoparticles (SPIONs) covalently coupled with a fluorophore sulfocyanine-5 and polyethylene glycol 5000 (PEG5000) and functionalized with single-chain variable fragments (scFv) of an antibody targeting the HER2 membrane receptor. These covalently functionalized SPIONs are then complexed via electrostatic interactions with therapeutic siRNA and the cationic polymers, chitosan, and poly-L-arginine. TS-MSNsiSurvivin had an average size of 144 ± 30 nm, a PDI of 0.3, and a slightly positive zeta potential value of 10.56 ± 05.70 mV. The agarose gel electrophoresis assay confirmed that the siRNA is well-complexed into TS-MSN without leakage, as no free siRNA was detected. Moreover, siRNA in TS-MSN was protected from RNAse A degradation for up to 6 h at 37 °C. Formulations of TS-MSN with siSurvivin demonstrated in vitro gene knockdown up to 89% in the HER2+ breast cancer cell line SK-BR-3. Furthermore, qRT-PCR confirmed a significant Survivin mRNA relative expression inhibition (about 50%) compared to control siRNA or untreated cells. A combination protocol was evaluated between TS-MSN and Doxorubicin (DOX) for the first time. Therefore, SK-BR-3 cells were pretreated with TS-MSN formulated with siSurvivin at 50 nM for 24 h alone, before a DOX treatment at a concentration of 0.5 µM (corresponding to the IC50) was added for 48 h. The MTT cytotoxicity tests, performed after 72 h of treatment, revealed that the combination had a significant synergistic cytotoxic effect on SK-BR-3 cells compared to monotherapies or untreated cells. We confirmed that pretreatment of cells with siSurvivin potentializes the cytotoxic effect of DOX as an alternative approach for treating HER2+ breast cancer. In conclusion, a combination of anti-Survivin siRNA and DOX would be a good alternative in HER2+ breast cancer therapy.

15.
Int J Pharm X ; 4: 100139, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36420371

RESUMO

Apoptosis is an important process that directly affects the response of cancer cells to anticancer drugs. Among different factors involved in this process, the BcL-xL protein plays a critical role in inhibiting apoptosis induced by chemotherapy agents. Henceforth, its downregulation may have a synergistic activity that lowers the necessary dose of anticancer agents. In this study, anti-Bcl-xL siRNA were formulated within an EGFR-targeted nanomedicine with scFv ligands (NM-scFv) and its activity was tested in the non-small cell lung cancer (NSCLC) cell line H460. The obtained NMs-scFv anti-Bcl-xL were suitable for intravenous injection with sizes around 100 nm, a high monodispersity level and good siRNA complexation capacity. The nanocomplex's functionalization with anti-EGFR scFv ligands was shown to allow an active gene delivery into H460 cells and led to approximately 63% of gene silencing at both mRNA and protein levels. The NM-scFv anti-Bcl-xL improved the apoptotic activity of cisplatin and reduced the cisplatin IC50 value in H460 cells by a factor of around three from 0.68 ± 0.12 µM to 2.21 ± 0.18 µM (p < 0.01), respectively, in comparison to that of NM-scFv formulated with control siRNA (p > 0.05).

16.
Pharmaceutics ; 14(12)2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36559172

RESUMO

Following our previous study on the development of EGFR-targeted nanomedicine (NM-scFv) for the active delivery of siRNA in EGFR-positive cancers, this study focuses on the development and the quality control of a radiolabeling method to track it in in vivo conditions with nuclear imaging. Our NM-scFv is based on the electrostatic complexation of targeted nanovector (NV-scFv), siRNA and two cationic polymers. NV-scFv comprises an inorganic core, a fluorescent dye, a polymer layer and anti-EGFR ligands. To track NM-scFv in vivo with nuclear imaging, the DTPA chemistry was used to radiolabel NM-scFv with 111In. DTPA was thiolated and introduced onto NV-scFv via the maleimide chemistry. To obtain suitable radiolabeling efficiency, different DTPA/NV-scFv ratios were tested, including 0.03, 0.3 and 0.6. At the optimized ratio (where the DTPA/NV-scFv ratio was 0.3), a high radiolabeling yield was achieved (98%) and neither DTPA-derivatization nor indium-radiolabeling showed any impact on NM-scFv's physicochemical characteristics (DH ~100 nm, PDi < 0.24). The selected NM-scFv-DTPA demonstrated good siRNA protection capacity and comparable in vitro transfection efficiency into EGFR-overexpressing cells in comparison to that of non-derivatized NM-scFv (around 67%). Eventually, it was able to track both qualitatively and quantitatively NM-scFv in in vivo environments with nuclear imaging. Both the radiolabeling and the NM-scFv showed a high in vivo stability level. Altogether, a radiolabeling method using DTPA chemistry was developed with success in this study to track our NM-scFv in in vivo conditions without any impact on its active targeting and physicochemical properties, highlighting the potential of our NM-scFv for future theranostic applications in EGFR-overexpressing cancers.

17.
Pharmaceutics ; 14(8)2022 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-35893780

RESUMO

Antibody-drug conjugates (ADCs) derived from a full immunoglobulin-G (IgG) are associated with suboptimal solid-tumor penetration and Fc-mediated toxicities. Antibody fragment-drug conjugates (FDCs) could be an alternative. Nevertheless, innovative solutions are needed to implant cysteines as conjugation sites in the single-chain fragment variable (scFv) format, which is the backbone from which many other antibody formats are built. In addition, the bioconjugation site has the utmost importance to optimize the safety and efficacy of bioconjugates. Our previous intra-tag cysteine (ITC) strategy consisted of introducing a bioconjugation motif at the C-terminal position of the 4D5.2 scFv, but this motif was subjected to proteolysis when the scFv was produced in CHO cells. Considering these data, using three intra-domain cysteine (IDC) strategies, several parameters were studied to assess the impact of different locations of a site-specific bioconjugation motif in the variable domains of an anti-HER2 scFv. In comparison to the ITC strategy, our new IDC strategy allowed us to identify new fragment-drug conjugates (FDCs) devoid of proteolysis and exhibiting enhanced stability profiles, better affinity, and better ability to kill selectively HER2-positive SK-BR-3 cells in vitro at picomolar concentrations. Thus, this work represents an important optimization step in the design of more complex and effective conjugates.

18.
Eur J Med Chem ; 229: 114063, 2022 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-34974337

RESUMO

Antibody-drug conjugates (ADCs) are targeted therapies, mainly used in oncology, consisting in a three-component molecular architecture combining a highly potent drug conjugated via a linker onto a monoclonal antibody (mAb), designed for the selective delivery of the drug to the tumor site. The linker is a key component, defining the ADC stability and mechanism of action, and particularly the drug release strategy. In this study, we have developed and synthesized a cleavable linker, which possesses an Asn-Pro-Val (NPV) sequence sensitive to the human neutrophil elastase (HNE), overexpressed in the tumor microenvironment. This linker permitted the site-specific conjugation of the cell-permeable drug, monomethyl auristatin E (MMAE), onto trastuzumab, using a disulfide re-bridging technology. The resulting ADC was then evaluated in vitro. This conjugate demonstrated retained antigen (Ag) binding affinity and exhibited high subnanomolar potency against Ag-positive tumor cells after internalization, suggesting an intracellular mechanism of linker cleavage. While no internalization and cytotoxic activity of this ADC was observed on Ag-negative cells in classical conditions, the supplementation of exogenous HNE permitted to restore a nanomolar activity on these cells, suggesting an extracellular mechanism of drug release in these conditions. This in vitro proof of concept tends to prove that the NPV sequence could allow a dual intra- and extracellular mechanism of drug release. This work represents a first step in the design of original ADCs with a new dual intra- and extracellular drug delivery system and opens the way to further experimentations to evaluate their full potential in vivo.


Assuntos
Antineoplásicos/química , Imunoconjugados/química , Elastase de Leucócito/metabolismo , Oligopeptídeos/química , Trastuzumab/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Dipeptídeos/química , Dissulfetos/química , Composição de Medicamentos , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Humanos , Imunoconjugados/farmacologia , Ligação Proteica , Conformação Proteica , Trastuzumab/farmacologia
19.
Nanoscale ; 14(15): 5884-5898, 2022 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-35373226

RESUMO

The impact of nanoparticle surface chemistry on cell interactions and especially cell uptake has become evident over the last few years in nanomedicine. Since PEG polymers have proved to be ideal tools for attaining stealthiness and favor escape from the in vivo mononuclear phagocytotic system, the accurate control of their geometry is of primary importance and can be achieved through reversible addition-fragmentation transfer (RAFT) polymerization. In this study, we demonstrate that the residual groups of the chain transfer agents (CTAs) introduced in the main chain exert a significant impact on the cellular internalization of functionalized nanoparticles. High-resolution magic angle spinning 1H NMR spectroscopy and fluorescence spectroscopy permitted by the magneto-fluorescence properties of nanoassemblies (NAs) revealed the compaction of the PEG comb-like shell incorporating CTAs with a long alkyl chain, without changing the overall surface potential. As a consequence of the capability of alkyl units to self-assemble at the NA surface while hardly contributing more than 0.5% to the total polyelectrolyte weight, denser PEGylated NAs showed notably less internalization in all cells of the tumor microenvironment (tumor cells, macrophages and healthy cells). Interestingly, such differentiated uptake is also observed between pro-inflammatory M1-like and immunosuppressive M2-like macrophages, with the latter more efficiently phagocytizing NAs coated with a less compact PEGylated shell. In contrast, the NA diffusion inside multicellular spheroids, used to mimic solid tumors, appeared to be independent of the NA coating. These results provide a novel effort-saving approach where the sole variation of the chemical nature of CTAs in RAFT PEGylated polymers strikingly modulate the cell uptake of nanoparticles upon the organization of their surface coating and open the pathway toward selectively addressing macrophage populations for cancer immunotherapy.


Assuntos
Nanopartículas , Polímeros , Corantes , Nanopartículas/química , Polietilenoglicóis/química , Polimerização , Polímeros/química , Polímeros/farmacologia , Microambiente Tumoral
20.
Int J Pharm ; 605: 120795, 2021 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-34119579

RESUMO

Recently, active targeting using nanocarriers with biological ligands has emerged as a novel strategy for improving the delivery of therapeutic and/or imaging agents to tumor cells. The presence of active targeting moieties on the surface of nanomedicines has been shown to play an important role in enhancing their accumulation in tumoral cells and tissues versus healthy ones. This property not only helps to increase the therapeutic index but also to minimize possible side effects of the designed nanocarriers. Since the overexpression of epidermal growth factor receptors (EGFR) is a common occurrence linked to the progression of a broad variety of cancers, the potential application of anti-EGFR immunotherapy and EGFR-targeting ligands in active targeting nanomedicines is getting increasing attention. Henceforth, the EGFR-targeted nanomedicines were extensively studied in vitro and in vivo but exhibited both satisfactory and disappointing results, depending on used protocols. This review is designed to give an overview of a variety of EGFR-targeting ligands available for nanomedicines, how to conjugate them onto the surface of nanoparticles, and the main analytical methods to confirm this successful conjugation.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias , Antineoplásicos/uso terapêutico , Portadores de Fármacos/uso terapêutico , Sistemas de Liberação de Medicamentos , Família de Proteínas EGF/uso terapêutico , Humanos , Ligantes , Nanomedicina , Neoplasias/tratamento farmacológico , Controle de Qualidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA