Refinar búsqueda
Resultados 1-10 de 22
Modulating inflammation through the neutralization of Interleukin-6 and tumor necrosis factor-α by biofunctionalized nanoparticles Texto completo
2021 | 1000
Lima, Ana Cláudia Fernandes | Amorim, Diana Alexandra Silva | Laranjeira, Inês | Almeida, Armando | Reis, R. L. | Ferreira, Helena Susana Costa Machado | Pinto-Ribeiro, Filipa | Neves, N. M.
Biological agents that neutralize the activity of pro-inflammatory cytokines are revolutionizing the treatment of inflammatory conditions. However, the antibodies (Abs) short half-life and off-target distribution critically limit their efficacy and safety. Therefore, this work proposes the immobilization of anti-TNF-α and anti-IL-6 Abs at the surface of polymeric nanoparticles (NPs) in order to extend and increase the Abs therapeutic efficacy, owing to the protection from degradation that the NPs provide, and to avoid off-target side effects through local administration. In an in vitro model of inflammation, biofunctionalized NPs were able to reduce the harmful effects on human chondrocytes provided by inflammatory macrophages, being demonstrated the additive effects of the dual neutralization. Significantly, biofunctionalized NPs ameliorated inflammation more efficiently than soluble Abs in an in vivo experimental model of inflammation, exhibiting a safe profile, a prolonged action, and a stronger efficacy. Hence, as this strategy is able to increase the therapeutic efficacy of the currently available treatments, it is a promising potential therapeutic option for inflammatory conditions. | Authors acknowledge the financial support from FCT/MCTES (Portuguese Foundation for Science and Technology/Ministry of Science, Technology and Higher Education) and the FSE/POCH (European Social Fund through the Operational Program of Human Capital), for the PhD scholarship PD/BD/11384/2015 of A. C. Lima (PD/59/2013), FCT for the projects PTDC/CTM-BIO/4388/2014 – SPARTAN and PTDC/BTMSAL/28882/2017 – Cells4_IDs, and the Northern Portugal Regional Operational Programme (NORTE 2020), under the Portugal 2020 Partnership Agreement, through the European Regional Development Fund (FEDER) for the projects NORTE-01-0145-FEDER-000023 – FROnTHERA and NORTE-01-0145-FEDER-000021. Authors also acknowledge REMIX Project, funded by the European Union’s Horizon 2020 Research and Innovation Programme under the Maria Sklodowska Curie Grant (Agreement No. 778078).
Mostrar más [+] Menos [-]The potential of tumor-specific Th2 cells for cancer immunotherapy by adoptive transfer Texto completo
1000 | 2013
Fauskanger, Marte Hotvedt | Alexandre Corthay, Bjarne Bogen
Immunotherapy based on adoptive cell transfer (ACT) of tumor-specific T cells, either naturally occurring or gene engineered, represents a promising strategy to cure cancer. To date the majority of ACT trials have focused on cytotoxic CD8+ T cells, which cave the ability to recognize and kill cancer cells directly. The potential of CD4+ T helper cells has in the recent years been increasingly appreciated. There are several subsets of CD4+ T cells, including Th1, Th2, Th17, and regulatory T cells, which differ greatly in terms of cytokine secretion and effector functions. It is not known which subset can provide the greatest efficacy for ACT. Here, we have investigated the efficacy of in vitro expanded, tumor-specific Th2 cells for the treatment of mice with MHC class II negative myeloma. The transferred Th2 cells were confirmed to have a true Th2 phenotype, producing the Th2-prototype cytokines IL-4, IL- and IL-13. The Th2 cells could very efficiently provide tumor cell eradication after intravenous transfer into immunodeficient mice challenged with subcutaneous myeloma inoculation. The Th2-accosiated cytokines IL-4 and IL-13 are known to induce alternative activation of macrophages into an M2 phenotype. M2 macrophages produce the enzyme arginase-1 and the potential of this enzyme being an effector molecule, was investigated. Blocking experiments revealed arginase-1 was required for efficient cancer eradication. These results show that there might be a great potential for Th2 cells in ACT, and that the Th2 cells possibly cause cancer eradication through the differentiation of macrophages into an arginase-1-producing M2 phenotype.
Mostrar más [+] Menos [-]Combination therapy with CD147-targeted nanoparticles carrying phenformin decreases lung cancer growth Texto completo
2023 | 1000
Pereira-Nunes, Andreia | Ferreira, Helena Susana Costa Machado | Abreu, Sara | Guedes, Marta | Neves, N. M. | Baltazar, Fátima | Granja, Sara
Lung cancer is one of the most fatal cancers worldwide. Resistance to conventional therapies remains a hindrance to patient treatment. Therefore, the development of more effective anti-cancer therapeutic strategies is imperative. Solid tumors exhibit a hyperglycolytic phenotype, leading to enhanced lactate production; and, consequently, its extrusion to the tumor microenvironment. Previous data reveals that inhibition of CD147, the chaperone of lactate transporters (MCTs), decreases lactate export in lung cancer cells and sensitizes them to phenformin, leading to a drastic decrease in cell growth. In this study, the development of anti-CD147 targeted liposomes (LUVs) carrying phenformin is envisioned, and their efficacy is evaluated to eliminate lung cancer cells. Herein, the therapeutic effect of free phenformin and anti-CD147 antibody, as well as the efficacy of anti-CD147 LUVs carrying phenformin on A549, H292, and PC-9 cell growth, metabolism, and invasion, are evaluated. Data reveals that phenformin decreases 2D and 3D-cancer cell growth and that the anti-CD147 antibody reduces cell invasion. Importantly, anti-CD147 LUVs carrying phenformin are internalized by cancer cells and impaired lung cancer cell growth in vitro and in vivo. Overall, these results provide evidence for the effectiveness of anti-CD147 LUVs carrying phenformin in compromising lung cancer cell aggressiveness. | This work was supported by the Fundação para a Ciência e a Tecnologia (FCT) - project UIDB/50026/2020 and UIDP/50026/2020 and by the project NORTE-01-0145-FEDER-000055,supported by Norte Portugal Regional Operational Programme NORTE 2020), under the PORTUGAL 2020Partnership Agreement, through the European Regional Development Fund (ERDF).”. SG and AP-N received fellowships from FCT (SFRH/BPD/117858/2016 and SFRH/BD/148476/2019, respectively).
Mostrar más [+] Menos [-]Microfluidic mixing system for precise PLGA-PEG nanoparticles size control Texto completo
1000 | 2022
Gimondi, Sara | Guimarães, Carlos F. | Vieira, Sara Filipa Fontoura | Gonçalves, Virgínia M. F. | Tiritan, M. E. | Reis, R. L. | Ferreira, Helena Susana Costa Machado | Neves, N. M.
In this study, a microfluidic device was employed to produce polymeric nanoparticles (NPs) with well-controlled sizes. The influence of several parameters in the synthesis process, namely, polymer concentration, flow rate and flow rate ratio between the aqueous and organic solutions was investigated. To evaluate the NPs size effect, three diameters were selected (30, 50 and 70 nm). Their cytocompatibility was demonstrated on endothelial cells and macrophages. Additionally, their efficacy to act as drug carriers was assessed in an in vitro inflammatory scenario. NPs loaded and released diclofenac (DCF) in a size-dependent profile (smaller sizes presented lower DCF content and higher release rate). Moreover, 30 nm NPs were the most effective in reducing prostaglandin E2 concentration. Therefore, this study demonstrates that microfluidics can generate stable NPs with controlled sizes, high monodispersity and enhanced batch-to-batch reproducibility. Indeed, NPs size is a crucial parameter for drug encapsulation, release and overall biological efficacy. | The authors would like to thank funding that allowed to carry out this work namely, the Fundacao para a Ciencia e a Tecnologia (FCT) for the S. Gimondi (PD/BD/143140/2019), C. F. Guimaraes (PD/BD135253/2017) and S. F. Vieira (PD/BD/135246/2017) fellowships. This work was also supported by FROnTHERA (NORTE-01-0145-FEDER-000023), Cells4_IDs (PTDC/BTM-SAL/28882/2017) and the NORTE 2020 Structured Project, co-funded by Norte2020 (NORTE-01-0145-FEDER-000021). We also thank Dr. Isabel Leonor for the precious support for the SEM microscopy and AFM images.
Mostrar más [+] Menos [-]The role of organ-on-a-chip technology in advancing personalized medicine Texto completo
2023 | 1000
Caballero, David | Reis, R. L. | Kundu, Subhas C
Organ-on-a-chip technology has emerged as a powerful tool to model human physiology and disease in vitro. This technology integrates microfluidic systems and cell microenvironments to create dynamic microscale devices that mimic the structure and functional unit of human organs and tissues. Organ-on-a-chip models have shown great promise in drug discovery, toxicology testing, and disease modeling. As a result, they have the potential to revolutionize personalized medicine by enabling the testing of drugs and therapies on patient-specific cells or tissues, thus improving the efficacy of treatments. This chapter provides an overview of recent advances in organ-on-a-chip technology and its applications in personalized medicine, focusing on its potential impact on cancer research. We will discuss how these microfluidic-based models can contribute to advancing individualized treatments and enabling the stratification of patients. | This work was funded by the Portuguese Foundation for Science and Technology (FCT) under the program CEEC Individual 2017 (CEECIND/00352/2017) and the RECOVER project (2022.02260.PTDC).
Mostrar más [+] Menos [-]Fucoidan/chitosan nanoparticles functionalized with anti-ErbB-2 target breast cancer cells and impair tumor growth in vivo Texto completo
2021 | 1000
Oliveira, Catarina | Gonçalves, Céline S. | Martins, Eduarda P. | Neves, N. M. | Reis, R. L. | Costa, Bruno M. | Silva, Tiago H. | Martins, Albino
The work herein presented reports the development of fucoidan/chitosan nanoparticles (NPs) loaded with gemcitabine and functionalized with ErbB-2 antibody at their surface (NPs + Gem + Ab). The maximum immobilization of ErbB-2 on NPs' surface was set at 10 mu g mL(-1) and resulted in NPs with a size around 160 nm, a polydispersity index of 0.18, and a zeta potential of 21 mV. ErbB-2 is overexpressed in some subtypes of breast cancers, and the targeting capability of the NPs + Gem + Ab system was confirmed by an increased cellular uptake of SKBR3 cells (ErbB-2 positive) when compared to MDA-MB-231 (ErbB-2 negative). To validate the targeting efficacy of NPs + Gem + Ab, a co-culture system with human endothelial and SKBR3 cells was established. Cytotoxic effects over endothelial cells were similar for all the tested conditions (between 25 and 30%). However, the NPs + Gem + Ab system presented increased toxicity over breast cancer cells, above 80% after 24 h, when compared to free Gem and NPs + Gem (around 15% and 20%, respectively). In vivo studies demonstrated that the developed targeting system significantly reduced tumor growth and the appearance of lung metastasis compared to untreated controls. In summary, the efficacy of the NPs + Gem + Ab system to target cancer cells was established and validated both in vitro and in vivo, being a compelling alternative strategy to current chemotherapeutic approaches. | This work was developed under the scope of the Structured projects for R&D&I NORTE-01-0145-FEDER-000013/21/23 supported by the Northern Portugal Regional Operational Programme Norte 2020, under the Portugal 2020 Partnership Agreement. The authors would like also to thank Norte 2020 for financing the PhD scholarship of C.O. "Norte-085369-000037" and the Portuguese Foundation for Science and Technology for the Investigator grant of A.M. (IF/00376/2014). The authors would also like to acknowledge Teresa Oliveira for her insights regarding the histological analysis performed for the in vivo studies.
Mostrar más [+] Menos [-]Class I and Class II Lanthipeptides Produced by Bacillus spp. Texto completo
1000 | 2015
Barbosa, Joana | Caetano, Tânia | Mendo, Sónia
The increasing number of multidrug-resistant pathogens, along with the small number of new antimicrobials under development, leads to an increased need for novel alternatives. Class I and class II lanthipeptides (also known as lantibiotics) have been considered promising alternatives to classical antibiotics. In addition to their relevant medical applications, they are used as probiotics, prophylactics, preservatives, and additives in cosmetics and personal-care products. The genus Bacillus is a prolific source of bioactive compounds including ribosomally and nonribosomally synthesized antibacterial peptides. Accordingly, there is significant interest in the biotechnological potential of members of the genus Bacillus as producers of antimicrobial lanthipeptides. The present review focuses on aspects of the biosynthesis, gene cluster organization, structure, antibacterial spectrum, and bioengineering approaches of lanthipeptides produced by Bacillus strains. Their efficacy and potency against some clinically relevant strains, including MRSA and VRE, are also discussed. Although no lanthipeptides are currently in clinical use, the information herein highlights the potential of these compounds.
Mostrar más [+] Menos [-]Light-triggered release of photocaged therapeutics: where are we now? Texto completo
1000 | 2019
Silva, Joana Maria Marques | Silva, Eduardo | Reis, R. L.
The current available therapeutics face several challenges such as the development of ideal drug delivery systems towards the goal of personalized treatments for patients benefit. The application of light as an exogenous activation mechanism has shown promising outcomes, owning to the spatiotemporal confinement of the treatment in the vicinity of the diseased tissue, which offers many intriguing possibilities. Engineering therapeutics with light responsive moieties have been explored to enhance the bioavailability, and drug efficacy either in vitro or in vivo. The tailor-made character turns the so-called photocaged compounds highly desirable to reduce the side effects of drugs and, therefore, have received wide research attention. Herein, we seek to highlight the potential of photocaged compounds to obtain a clear understanding of the mechanisms behind its use in therapeutic delivery. A deep overview on the progress achieved in the design, fabrication as well as current and possible future applications in therapeutics of photocaged compounds is provided, so that novel formulations for biomedical field can be designed. | The authors acknowledge the financial support of Portuguese Foundation for Science and Technology (FCT) through the post-doctoral grant number SFRH/BPD/116779/2016 and the project PTDC/ CTM-BIO/4706/2014.
Mostrar más [+] Menos [-]Fibronectin bound to a fibrous substrate has chondrogenic induction properties Texto completo
1000 | 2020
Casanova, Marta Alexandra Rodrigues | Reis, R. L. | Martins, Albino | Neves, N. M.
Articular cartilage is an avascular tissue characterized by a dense and specific extracellular matrix (ECM). Fibronectin (FN) is a key constituent of the pericellular ECM, assembled into a fibrillar matrix through a cell-mediated process, being implicated in chondrogenic events. In this study, we evaluate the chondrogenic potential of FN bound to the surface of an electrospun nanofibrous mesh (NFM). For that, an anti-FN antibody was immobilized at the surface of NFMs, rendering them capable of selectively binding endogenous FN (eFN) from blood plasma. The chondrogenic potential of bound eFN was further assessed by culturing human bone marrow-derived mesenchymal stem cells (hBM-MSCs) for 28 days, in a basal growth medium. The biological results indicate that NFMs functionalized with eFN were able to successfully induce the chondrogenesis of hBM-MSCs, as demonstrated by the high expression of SOX9, Aggrecan, and Collagen type II. Therefore, biofunctionalized nanofibrous substrates comprising eFN significantly enhance the efficacy of a cartilage tissue-engineering strategy. | The authors would like to acknowledge the Portuguese Foundation for Science and Technology (FCT) for the Ph.D. grant of M.R.C. (PD/BD/113797/2015) financed by the FCT Doctoral Program on Advanced Therapies for Health (PATH) (FSE/POCH/PD/169/2013), the IF grant of A.M. (IF/00376/2014), and the project SPARTAN (PTDC/CTMBIO/4388/2014)
Mostrar más [+] Menos [-]Lyotropic liquid crystalline 2D and 3D mesophases: Advanced materials for multifunctional anticancer nanosystems Texto completo
2023 | 1000
Araújo-Silva, Henrique | Teixeira, Patricia V | Gomes, Andreia C | Lúcio, M. | Lopes, Carla M
Cancer remains a leading cause of mortality. Despite significant breakthroughs in conventional therapies, treatment is still far from ideal due to high toxicity in normal tissues and therapeutic inefficiency caused by short drug lifetime in the body and resistance mechanisms. Current research moves towards the development of multifunctional nanosystems for delivery of chemotherapeutic drugs, bioactives and/or radionuclides that can be combined with other therapeutic modalities, like gene therapy, or imaging to use in therapeutic screening and diagnosis. The preparation and characterization of Lyotropic Liquid Crystalline (LLC) mesophases self-assembled as 2D and 3D structures are addressed, with an emphasis on the unique properties of these nanoassemblies. A comprehensive review of LLC nanoassemblies is also presented, highlighting the most recent advances and their outstanding advantages as drug delivery systems, including tailoring strategies that can be used to overcome cancer challenges. Therapeutic agents loaded in LLC nanoassemblies offer qualitative and quantitative enhancements that are superior to conventional chemotherapy, particularly in terms of preferential accumulation at tumor sites and promoting enhanced cancer cell uptake, lowering tumor volume and weight, improving survival rates, and increasing the cytotoxicity of their loaded therapeutic agents. In terms of quantitative anticancer efficacy, loaded LLC nanoassemblies reduced the IC50 values from 1.4-fold against lung cancer cells to 125-fold against ovarian cancer cells. | This work was supported by the Portuguese Foundation for Science and Technology (FCT) in the framework of the Strategic Funding UIDB/04650/2020 and UIDB/04050/2020 (CF-UM-UP and CBMA); UIDP/04378/2020 and UIDB/04378/2020 of the Research Unit on Applied Molecular Biosciences—UCIBIO and the project LA/P/0140/2020 of the Associate Laboratory Institute for Health and Bioeconomy—i4HB.
Mostrar más [+] Menos [-]