Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
3.
Ann Surg Oncol ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38869763

ABSTRACT

BACKGROUND: The highly metastatic nature of pancreatic ductal adenocarcinoma (PDAC) and the difficulty to achieve favorable patient outcomes emphasize the need for novel therapeutic solutions. For preclinical evaluations, genetically engineered mouse models are often used to mimic human PDAC but frequently fail to replicate synchronous development and metastatic spread. This study aimed to develop a transplantation model to achieve synchronous and homogenous PDAC growth with controlled metastatic patterns in the liver. METHODS: To generate an orthotopic PDAC model, the DT6606 cell line was injected into the pancreas head of C57BL/6 mice, and their survival was monitored over time. To generate a heterotopic transplantation model, growing doses of three PDAC cell lines (DT6606, DT6606lm, and K8484) were injected into the portal vein of mice. Magnetic resonance imaging (MRI) was used to monitor metastatic progression, and histologic analysis was performed. RESULTS: Orthotopically injected mice succumbed to the tumor within an 11-week period (average survival time, 78.2 ± 4.45 days). Post-mortem examinations failed to identify liver metastasis. In the intraportal model, 2 × 105 DT6606 cells resulted in an absence of liver metastases by day 21, whereas 5 × 104 DT6606lm cells and 7 × 104 K8484 cells resulted in steady metastatic growth. Higher doses caused significant metastatic liver involvement. The use of K8484 cells ensured the growth of tumors closely resembling the histopathologic characteristics of human PDAC. CONCLUSIONS: This report details the authors' efforts to establish an "optimal" murine model for inducing metastatic PDAC, which is critical for advancing our understanding of the disease and developing more effective treatments.

4.
Int J Biol Sci ; 19(1): 156-166, 2023.
Article in English | MEDLINE | ID: mdl-36594095

ABSTRACT

Rationale: The αvß6- and αvß8-integrins, two cell-adhesion receptors upregulated in many tumors and involved in the activation of the latency associated peptide (LAP)/TGFß complex, represent potential targets for tumor imaging and therapy. We investigated the tumor-homing properties of a chromogranin A-derived peptide containing an RGDL motif followed by a chemically stapled alpha-helix (called "5a"), which selectively recognizes the LAP/TGFß complex-binding site of αvß6 and αvß8. Methods: Peptide 5a was labeled with IRDye 800CW (a near-infrared fluorescent dye) or with 18F-NOTA (a label for positron emission tomography (PET)); the integrin-binding properties of free peptide and conjugates were then investigated using purified αvß6/αvß8 integrins and various αvß6/αvß8 single - or double-positive cancer cells; tumor-homing, biodistribution and imaging properties of the conjugates were investigated in subcutaneous and orthotopic αvß6-positive carcinomas of the pancreas, and in mice bearing subcutaneous αvß8-positive prostate tumors. Results: In vitro studies showed that 5a can bind both integrins with high affinity and inhibits cell-mediated TGFß activation. The 5a-IRDye and 5a-NOTA conjugates could bind purified αvß6/αvß8 integrins with no loss of affinity compared to free peptide, and selectively recognized various αvß6/αvß8 single- or double-positive cancer cells, including cells from pancreatic carcinoma, melanoma, oral mucosa, bladder and prostate cancer. In vivo static and dynamic optical near-infrared and PET/CT imaging and biodistribution studies, performed in mice with subcutaneous and orthotopic αvß6-positive carcinomas of the pancreas, showed high target-specific uptake of fluorescence- and radio-labeled peptide by tumors and low non-specific uptake in other organs and tissues, except for excretory organs. Significant target-specific uptake of fluorescence-labeled peptide was also observed in mice bearing αvß8-positive prostate tumors. Conclusions: The results indicate that 5a can home to αvß6- and/or αvß8-positive tumors, suggesting that this peptide can be exploited as a ligand for delivering imaging or anticancer agents to αvß6/αvß8 single- or double-positive tumors, or as a tumor-homing inhibitor of these TGFß activators.


Subject(s)
Carcinoma , Pancreatic Neoplasms , Prostatic Neoplasms , Male , Animals , Mice , Humans , Chromogranin A/metabolism , Positron Emission Tomography Computed Tomography , Tissue Distribution , Peptides/chemistry , Integrins/metabolism , Transforming Growth Factor beta/metabolism
5.
Int J Mol Sci ; 23(18)2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36142575

ABSTRACT

The aim of this study is to provide a comprehensive characterization of stemness in pancreatic ductal adenocarcinoma (PDAC) cell lines. Seventeen cell lines were evaluated for the expression of cancer stem cell (CSC) markers. The two putative pancreatic CSC phenotypes were expressed heterogeneously ranging from 0 to 99.35% (median 3.46) for ESA+CD24+CD44+ and 0 to 1.94% (median 0.13) for CXCR4+CD133+. Cell lines were classified according to ESA+CD24+CD44+ expression as: Low-Stemness (LS; <5%, n = 9, median 0.31%); Medium-Stemness (MS; 6−20%, n = 4, median 12.4%); and High-Stemness (HS; >20%, n = 4, median 95.8%) cell lines. Higher degree of stemness was associated with in vivo tumorigenicity but not with in vitro growth kinetics, clonogenicity, and chemo-resistance. A wide characterization (chemokine receptors, factors involved in pancreatic organogenesis, markers of epithelial−mesenchymal transition, and secretome) revealed that the degree of stemness was associated with KRT19 and NKX2.2 mRNA expression, with CD49a and CA19.9/Tie2 protein expression, and with the secretion of VEGF, IL-7, IL-12p70, IL-6, CCL3, IL-10, and CXCL9. The expression of stem cell markers was also evaluated on primary tumor cells from 55 PDAC patients who underwent pancreatectomy with radical intent, revealing that CXCR4+/CD133+ and CD24+ cells, but not ESA+CD24+CD44+, are independent predictors of mortality.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CD24 Antigen/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line , Cell Line, Tumor , Humans , Hyaluronan Receptors/metabolism , Integrin alpha1 , Interleukin-10/metabolism , Interleukin-6/metabolism , Interleukin-7/metabolism , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/pathology , RNA, Messenger/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Vascular Endothelial Growth Factor A/metabolism , Pancreatic Neoplasms
6.
Cancers (Basel) ; 13(17)2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34503252

ABSTRACT

The stroma is a relevant player in driving and supporting the progression of pancreatic ductal adenocarcinoma (PDAC), and a large body of evidence highlights its role in hindering the efficacy of current therapies. In fact, the dense extracellular matrix (ECM) characterizing this tumor acts as a natural physical barrier, impairing drug penetration. Consequently, all of the approaches combining stroma-targeting and anticancer therapy constitute an appealing option for improving drug penetration. Several strategies have been adopted in order to target the PDAC stroma, such as the depletion of ECM components and the targeting of cancer-associated fibroblasts (CAFs), which are responsible for the increased matrix deposition in cancer. Additionally, the leaky and collapsing blood vessels characterizing the tumor might be normalized, thus restoring blood perfusion and allowing drug penetration. Even though many stroma-targeting strategies have reported disappointing results in clinical trials, the ECM offers a wide range of potential therapeutic targets that are now being investigated. The dense ECM might be bypassed by implementing nanoparticle-based systems or by using mesenchymal stem cells as drug carriers. The present review aims to provide an overview of the principal mechanisms involved in the ECM remodeling and of new promising therapeutic strategies for PDAC.

7.
Nanotechnology ; 32(32)2021 May 17.
Article in English | MEDLINE | ID: mdl-33892482

ABSTRACT

Conventional chemotherapy used against cancer is mostly limited due to their non-targeted nature, affecting normal tissue and causing undesirable toxic effects to the affected tissue. With the aim of improving these treatments both therapeutically and in terms of their safety, numerous studies are currently being carried out using nanoparticles (NPs) as a vector combining tumor targeting and carrying therapeutic tools. In this context, it appears that nucleolin, a molecule over-expressed on the surface of tumor cells, is an interesting therapeutic target. Several ligands, antagonists of nucleolin of various origins, such as AS1411, the F3 peptide and the multivalent pseudopeptide N6L have been developed and studied as therapeutic tools against cancer. Over the last ten years or so, numerous studies have been published demonstrating that these antagonists can be used as tumor targeting agents with NPs from various origins. Focusing on nucleolin ligands, the aim of this article is to review the literature recently published or under experimentation in our research team to evaluate the efficacy and future development of these tools as anti-tumor agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Aptamers, Nucleotide/therapeutic use , Neoplasms/drug therapy , Oligodeoxyribonucleotides/therapeutic use , Peptide Fragments/therapeutic use , Peptides/therapeutic use , Phosphoproteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Antineoplastic Agents/chemistry , Aptamers, Nucleotide/chemistry , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Gene Expression , HMGB2 Protein/genetics , HMGB2 Protein/metabolism , Humans , Ligands , Molecular Targeted Therapy/methods , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oligodeoxyribonucleotides/chemistry , Peptide Fragments/chemistry , Peptides/chemistry , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Theranostic Nanomedicine/methods , Nucleolin
8.
Int J Pharm ; 590: 119930, 2020 Nov 30.
Article in English | MEDLINE | ID: mdl-33010394

ABSTRACT

Large-pore mesoporous silica (LPMS) microspheres with tunable pore size have received intensive interest in the field of drug delivery due to their high storage capacity and fast delivery rate of drugs. In this work, a facile salt-assisted spray-drying method has been developed to fabricate LPMS microspheres using continuous spray-drying of simple inorganic salts as pore templates and colloidal SiO2 nanoparticles as building blocks, followed by washing with water to remove the templates. More importantly, the porosity of the LPMS microspheres can be finely tuned by adjusting the furnace temperature and relative concentration of the salt to SiO2, which could lead to optimal pharmaceutical outcomes. Then, the biological roles of these LPMS microspheres were evaluated in antibacterial and cancer therapy. In this regard, rhodamine b as a probe was initially loaded inside the LPMS microspheres. The obtained particles not only showed high entrapment efficiency (up to 30%) and a pH-responsive drug release but also presented pore-size-controlled drug release performance. Then, in vitro antibacterial activities of multiple antibiotics, namely nalidixic acid, chloramphenicol, and ciprofloxacin, loaded in the LPMS particles were investigated against two pathogenic bacteria, Escherichia coli (Gram-negative) and Staphylococcus aureus (Gram-positive). The results indicated bacterial inhibition up to 70% and 20% in less than 2 h for Escherichia coli and Staphylococcus aureus, respectively. This inhibition of bacterial growth was accompanied by no bacterial regrowth within 30 h. Finally, the versatility of LPMS microspheres as drug carriers in pancreatic cancer treatment was explored. In this regard, a pro-apoptotic NCL antagonist agent (N6L) as an antitumor agent was successfully loaded onto LPMS microspheres. Interestingly, the resulting particles showed pore-size-dependent anticancer activity with inhibition of cancer cell growth up to 60%.


Subject(s)
Pancreatic Neoplasms , Silicon Dioxide , Anti-Bacterial Agents/pharmacology , Drug Compounding , Humans , Microspheres , Pancreatic Neoplasms/drug therapy , Particle Size , Porosity
9.
J Control Release ; 320: 112-124, 2020 04 10.
Article in English | MEDLINE | ID: mdl-31962094

ABSTRACT

Inducible T-cell costimulator (ICOS) upon binding to its ligand (ICOSL) mediates adaptive immunity and antitumor response. Thus, antitumor therapies targeting the ICOS/ICOSL pathway hold great promise for cancer treatment. In this regard, ICOSL triggering by a soluble recombinant form of ICOS (ICOS-Fc) hampered adhesiveness and migration of dendritic, endothelial, and tumor cells in vitro. Furthermore, in vivo treatment with ICOS-Fc previously showed the capability to inhibit lung metastatization of ICOSL+ B16-F10 melanoma cells when injected intravenously in mice, but it failed to block the growth of established subcutaneous B16-F10 murine tumors. Thus, we asked whether passive targeting of solid tumors with ICOS-Fc-loaded biocompatible and biodegradable nanoparticles (NPs) could instead prove effectiveness in reducing tumor growth. Here, ICOS-Fc was loaded in two types of polymer nanoparticles, i.e. cross-linked ß-cyclodextrin nanosponges (CDNS) and poly(lactic-co-glycolic acid) (PLGA) NPs and in vitro characterized. In vivo experiments showed that treatment of C57BL6/J mice with ICOS-Fc loaded into the two nanoformulations inhibits the growth of established subcutaneous B16-F10 tumors. This anticancer activity appears to involve both anti-angiogenic and immunoregulatory effects, as shown by decreased tumor vascularization and downmodulation of IL-10 and Foxp3, two markers of regulatory T cells (Tregs). Overall, the substantial in vivo anticancer activity of ICOS-Fc-loaded CDNS and PLGA NPs against different components of the tumor microenvironment makes these nanoformulations attractive candidates for future combination cancer therapy.


Subject(s)
Melanoma, Experimental , Nanoparticles , Animals , Immunity, Cellular , Immunotherapy , Inducible T-Cell Co-Stimulator Ligand , Inducible T-Cell Co-Stimulator Protein , Melanoma, Experimental/drug therapy , Mice , Tumor Microenvironment
10.
Cancers (Basel) ; 12(1)2020 Jan 09.
Article in English | MEDLINE | ID: mdl-31936526

ABSTRACT

Doxorubicin (DOX) is an anthracycline widely used in cancer therapy and in particular in breast cancer treatment. The treatment with DOX appears successful, but it is limited by a severe cardiotoxicity. This work evaluated the in vitro and in vivo anticancer effect of a new formulation of ß-cyclodextrin nanosponges containing DOX (BNS-DOX). The BNS-DOX effectiveness was evaluated in human and mouse breast cancer cell lines in vitro in terms of effect on cell growth, cell cycle distribution, and apoptosis induction; and in vivo in BALB-neuT mice developing spontaneous breast cancer in terms of biodistribution, cancer growth inhibition, and heart toxicity. BNS-DOX significantly inhibited cancer cell proliferation, through the induction of apoptosis, with higher efficiency than free DOX. The breast cancer growth in BALB-neuT mice was inhibited by 60% by a BNS-DOX dose five times lower than the DOX therapeutic dose, with substantial reduction of tumor neoangiogenesis and lymphangiogenesis. Biodistribution after BNS-DOX treatment revealed a high accumulation of DOX in the tumor site and a low accumulation in the hearts of mice. Results indicated that use of BNS may be an efficient strategy to deliver DOX in the treatment of breast cancer, since it improves the anti-cancer effectiveness and reduces cardiotoxicity.

11.
Front Pharmacol ; 10: 776, 2019.
Article in English | MEDLINE | ID: mdl-31354491

ABSTRACT

This study investigated the effects of free paclitaxel (PTX) and PTX-loaded in pyromellitic nanosponges (PTX-PNS) in reducing in vitro and in vivo melanoma cell growth and invasivity, and in inhibiting angiogenesis. To test the response of cells to the two PTX formulations, the cell viability was evaluated by MTT assay in seven continuous cell lines, in primary melanoma cells, both in 2D and 3D cultures, and in human umbilical vein endothelial cells (HUVECs) after exposure to different concentrations of PTX or PTX-PNS. Cell motility was assessed by a scratch assay or Boyden chamber assay, evaluating cell migration in presence or absence of diverse concentrations of PTX or PTX-PNS. The effect of PTX and PTX-PNS on angiogenesis was evaluated as endothelial tube formation assay, a test able to estimate the formation of three-dimensional vessels in vitro. To assess the anticancer effect of PTX and PTX-PNS in in vivo experiments, the two drug formulations were tested in a melanoma mouse model obtained by B16-BL6 cell implantation in C57/BL6 mice. Results obtained were as follows: 1) MTT analysis revealed that cell proliferation was more affected by PTX-PNS than by PTX in all tested cell lines, in both 2D and 3D cultures; 2) the analysis of the cell migration showed that PTX-PNS acted at very lower concentrations than PTX; 3) tube formation assay showed that PTX-PNS were more effective in inhibiting tube formation than free PTX; and 4) in vivo experiments demonstrated that tumor weights, volumes, and growth were significantly reduced by PTX-PNS treatment with respect to PTX; the angiogenesis and the cell proliferation, detected in the tumor samples with CD31 and Ki-67 antibodies, respectively, indicated that, in the PTX-PNS-treated tumors, the tube formation was inhibited, and a low amount of proliferating cells was present. Taken together, our data demonstrated that our new PTX nanoformulation can respond to some important issues related to PTX treatment, lowering the anti-tumor effective doses and increasing the effectiveness in inhibiting melanoma growth in vivo.

12.
Nanomaterials (Basel) ; 9(2)2019 Feb 08.
Article in English | MEDLINE | ID: mdl-30744025

ABSTRACT

Background: Solid lipid nanoparticles (SLNs) are attractive drug delivery systems for lipophilic molecules like curcumin (CURC) with low chemical stability. Methods: A simple, innovative, and cold-operating method, named "cold dilution of microemulsion" is developed by the authors to produce SLNs. An oil-in-water microemulsion (µE), whose disperse phase consisted of a solution of trilaurin in a partially water-miscible solvent, was prepared after mutually saturating solvent and water. Trilaurin SLNs precipitated following solvent removal upon water dilution of the µE. After SLN characterization (mean size, Zeta potential, CURC entrapment efficiency, and over time stability), they were tested for in vitro cytotoxicity studies on pancreatic adenocarcinoma cell lines and for in vivo preliminary biodistribution studies in Wistar healthy rats. Results: CURC loaded SLNs (SLN-CURC) had mean diameters around 200 nm, were negatively charged, stable over time, and able to entrap CURC up to almost 90%, consequently improving its stability. SLN-CURC inhibited in vitro pancreatic carcinoma cell growth in concentration-dependent manner. Their in vivo intravenous administration suggested a possible long circulation. Conclusions: These results, according to a concomitant study on chitosan-coated SLNs, confirm the possibility to apply the developed SLN-based delivery systems as a means to entrap CURC, to improve both its water dispersibility and chemical stability, facilitating its application in therapy.

13.
Phytomedicine ; 56: 156-164, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30668336

ABSTRACT

BACKGROUND: Ailanthone (Aila) is a natural active compound isolated from the Ailanthus altissima, which has been shown to possess an "in vitro" growth-inhibitory effect against several cancer cell lines. Advanced bladder cancer is a common disease characterized by a frequent onset of resistance to cisplatin-based therapy. The cisplatin (CDDP) resistance is accompanied by an increase in Nrf2 protein expression which contributes to conferring resistance. Recently, we demonstrated a cross-talk between Nrf2 and YAP. YAP has also been demonstrated to play an important role in chemoresistance of bladder cancer. PURPOSE: We analyzed the antitumor effect of Aila in sensitive and CDDP-resistant bladder cancer cells and the molecular mechanisms involved in Aila activity. STUDY DESIGN: Sensitive and CDDP-resistant 253J B-V and 253J bladder cancer cells, intrinsically CDDP-resistant T24 bladder cancer cells and HK-2 human renal cortex cells were used. Cells were treated with diverse concentrations of Aila and proliferation, cell cycle, apoptosis and gene expressions were determined. METHODS: Aila toxicity and proliferation were determined by MTT and colony forming methods, respectively. Cell cycle was determined by cytofluorimetric analysis through PI staining method. Apoptosis was detected using Annexin V and PI double staining followed by quantitative flow cytometry. Expressions of Nrf2, Yap, c-Myc, and house-keeping genes were determined by western blot with specific antibodies. Cell migration was detected by wound healing and Boyden chamber analysis. RESULTS: Aila inhibited the growth of sensitive and CDDP-resistant bladder cancer cells with the same effectiveness. On the contrary, the growth of HK-2 cells was only slightly reduced by Aila. Cell cycle analysis revealed an accumulation of Aila-treated bladder cancer cells in the G0/G1 phase. Interestingly, Aila strongly reduced Nrf2 expression in these cell lines. Moreover, Aila significantly reduced YAP, and c-Myc protein expression. The random and the oriented migration of bladder cancer cells were strongly inhibited by Aila treatment, in particular in CDDP-resistant cells. CONCLUSION: Aila inhibited proliferation and invasiveness of bladder cancer cells. Its high effectiveness in CDDP resistant cells could be related to the inhibition of Nrf2, YAP, and c-Myc expressions. Aila could represent a new tool to treating CDDP-resistant bladder cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Quassins/pharmacology , Urinary Bladder Neoplasms/drug therapy , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Drug Resistance, Neoplasm/physiology , Humans , NF-E2-Related Factor 2/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , YAP-Signaling Proteins
14.
Oncotarget ; 9(88): 35813-35829, 2018 Nov 09.
Article in English | MEDLINE | ID: mdl-30533197

ABSTRACT

Strigolactones (SLs) are carotenoid-derived plant hormones that exhibit anti-cancer activities. We previously demonstrated that two SL analogues, MEB55 and ST362, inhibit the growth and survival of various cancer cell lines. However, these compounds have low aqueous solubility and stability at physiological pH. Here, we generated SL-loaded glutathione/pH-responsive nanosponges (GSH/pH-NS) to selectively deliver SLs to prostate cancer cells and enhance their therapeutic efficacy. The SLs were readily incorporated into the GSH/pH-NS. The drug loading efficiency was 13.9% for MEB55 and 15.4% for ST362, and the encapsulation efficiency was 88.7% and 96.5%, respectively. Kinetic analysis revealed that release of MEB55 and ST362 from the GSH/pH-NS was accelerated at acidic pH and in the presence of a high GSH concentration. Evaluation of the effects of MEB55- and ST362-loaded GSH/pH-NS on the growth of DU145 (high GSH) and PC-3 (low GSH) prostate cancer cells revealed that the GSH/pH-NS inhibited the proliferation of DU145 cells to a greater extent than free MEB55 or ST362 over a range of concentrations. These findings indicate GSH/pH-NS are efficient tools for controlled delivery of SLs to prostate cancer cells and may enhance the therapeutic efficacy of these compounds.

15.
J Nanosci Nanotechnol ; 18(1): 556-563, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29768881

ABSTRACT

Floxuridine is a very effective drug with high potency in the treatment of various tumors but its utility is limited by its low efficiency of cellular uptake. In order to improve the floxuridine efficiency of cellular uptake, lipophilic prodrug of floxuridine (3',5'-distearoyl-5-fluoro-2'-deoxyuridine) was synthetized and loaded into behenic acid nanoparticles produced by fatty acid coacervation technique. Generally, spherical shaped SLN with mean diameters below 300 nm were obtained. Distearoyl-floxuridine was loaded in SLN with high entrapment efficiency (from 70.8 to 82.8%). In Vitro cytotoxicity studies on different human cancer cell lines (M14, HT-29 and MDA-MB231) were performed in order to test the ability of distearoyl-floxuridine-SLN to inhibit the cancer cell growth. In MTT test distearoyl floxuridine SLN showed a greater efficacy than floxuridine on all cancer cell lines revealing an efficiency about 100 times higher. Also clonogenic assay showed a higher cytotoxicity of distearoyl-floxuridine-SLN compared to floxuridine but the difference between the formulations was only about 10 times. In conclusion, SLN proved to be a promising vehicle to increase the floxuridine efficacy in cancer therapy.


Subject(s)
Floxuridine , Nanoparticles , Neoplasms , Prodrugs , Cell Line , Cell Line, Tumor , Drug Carriers/therapeutic use , Floxuridine/pharmacology , Floxuridine/therapeutic use , Humans , Lipids , Neoplasms/drug therapy , Particle Size , Prodrugs/pharmacology
16.
Int J Mol Sci ; 19(2)2018 Jan 24.
Article in English | MEDLINE | ID: mdl-29364157

ABSTRACT

AIM: To develop an innovative delivery system for temozolomide (TMZ) in solid lipid nanoparticles (SLN), which has been preliminarily investigated for the treatment of melanoma. MATERIALS AND METHODS: SLN-TMZ was obtained through fatty acid coacervation. Its pharmacological effects were assessed and compared with free TMZ in in vitro and in vivo models of melanoma and glioblastoma. RESULTS: Compared to the standard free TMZ, SLN-TMZ exerted larger effects, when cell proliferation of melanoma cells, and neoangiogeneis were evaluated. SLN-TMZ also inhibited growth and vascularization of B16-F10 melanoma in C57/BL6 mice, without apparent toxic effects. CONCLUSION: SLN could be a promising strategy for the delivery of TMZ, allowing an increased stability of the drug and thereby its employment in the treatment of aggressive malignacies.


Subject(s)
Dacarbazine/analogs & derivatives , Melanoma/pathology , Nanoparticles , Animals , Biomarkers , Cell Line, Tumor , Dacarbazine/administration & dosage , Dacarbazine/chemistry , Disease Models, Animal , Drug Stability , Female , Humans , Immunohistochemistry , Melanoma/drug therapy , Melanoma/metabolism , Melanoma, Experimental , Mice , Molecular Structure , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Neoplastic Stem Cells , Temozolomide
17.
World J Gastroenterol ; 23(23): 4200-4210, 2017 Jun 21.
Article in English | MEDLINE | ID: mdl-28694660

ABSTRACT

AIM: To improve anti-inflammatory activity while reducing drug doses, we developed a nanoformulation carrying dexamethasone and butyrate. METHODS: Dexamethasone cholesteryl butyrate-solid lipid nanoparticles (DxCb-SLN) were obtained with the warm microemulsion method. The anti-inflammatory activity of this novel nanoformulation has been investigated in vitro (cell adhesion to human vascular endothelial cells and pro-inflammatory cytokine release by lipopolysaccharide-induced polymorphonuclear cells) and in vivo (disease activity index and cytokine plasma concentrations in a dextran sulfate sodium-induced mouse colitis) models. Each drug was also administered separately to compare its effects with those induced by their co-administration in SLN at the same concentrations. RESULTS: DxCb-SLN at the lowest concentration tested (Dx 2.5 nmol/L and Cb 0.1 µmol/L) were able to exert a more than additive effect compared to the sum of the individual effects of each drug, inducing a significant in vitro inhibition of cell adhesion and a significant decrease of pro-inflammatory cytokine (IL-1ß and TNF-α) in both in vitro and in vivo models. Notably, only the DxCb nanoformulation administration was able to achieve a significant cytokine decrease compared to the cytokine plasma concentration of the untreated mice with dextran sulfate sodium-induced colitis. Specifically, DxCb-SLN induced a IL-1ß plasma concentration of 61.77% ± 3.19%, whereas Dx or Cb used separately induced a concentration of 90.0% ± 2.8% and 91.40% ± 7.5%, respectively; DxCb-SLN induced a TNF-α plasma concentration of 30.8% ± 8.9%, whereas Dx or Cb used separately induced ones of 99.5% ± 4.9% and 71.1% ± 10.9%, respectively. CONCLUSION: Our results indicate that the co-administration of dexamethasone and butyrate by nanoparticles may be beneficial for inflammatory bowel disease treatment.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Drug Delivery Systems , Inflammatory Bowel Diseases/drug therapy , Nanoparticles/chemistry , Animals , Butyrates/administration & dosage , Cell Adhesion , Colitis/chemically induced , Colitis/metabolism , Cytokines/metabolism , Dexamethasone/administration & dosage , Human Umbilical Vein Endothelial Cells , Humans , Inflammation , Leukocytes, Mononuclear/cytology , Lipopolysaccharides , Male , Mice , Mice, Inbred BALB C , Neutrophils/cytology
18.
Drug Deliv ; 24(1): 670-680, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28368209

ABSTRACT

Anaplastic carcinoma of the thyroid (ATC) is a lethal human malignant cancer with median survival of 6 months. To date, no treatment has substantially changed its course, which makes urgent need for the development of novel drugs or novel formulations for drug delivery. Nanomedicine has enormous potential to improve the accuracy of cancer therapy by enhancing availability and stability, decreasing effective doses and reducing side effects of drugs. Camptothecin (CPT) is an inhibitor of DNA topoisomerase-I with several anticancer properties but has poor solubility and a high degradation rate. Previously, we reported that CPT encapsulated in ß-cyclodextrin-nanosponges (CN-CPT) increased solubility, was protected from degradation and inhibited the growth of prostate tumor cells both in vitro and in vivo. The aim of this study was to extend that work by assessing the CN-CPT effectiveness on ATC both in vitro and in vivo. Results showed that CN-CPT significantly inhibited viability, clonogenic capacity and cell-cycle progression of ATC cell lines showing a faster and enhanced effect compared to free CPT. Moreover, CN-CPT inhibited tumor cell adhesion to vascular endothelial cells, migration, secretion of pro-angiogenic factors (IL-8 and VEGF-α), expression of ß-PIX, belonging to the Rho family activators, and phosphorylation of the Erk1/2 MAPK. Finally, CN-CPT significantly inhibited the growth, the metastatization and the vascularization of orthotopic ATC xenografts in SCID/beige mice without apparent toxic effects in vivo. This work extends the previous insight showing that ß-cyclodextrin-nanosponges are a promising tool for the treatment of ATC.


Subject(s)
Thyroid Carcinoma, Anaplastic , Animals , Camptothecin , Cell Line, Tumor , Heterografts , Humans , Male , Mice , Mice, SCID
19.
Sci Rep ; 7: 43718, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28276434

ABSTRACT

Osteopontin is a pleiotropic cytokine that is involved in several diseases including multiple sclerosis. Secreted osteopontin is cleaved by few known proteases, modulating its pro-inflammatory activities. Here we show by in vitro experiments that secreted osteopontin can be processed by extracellular proteasomes, thereby producing fragments with novel chemotactic activity. Furthermore, osteopontin reduces the release of proteasomes in the extracellular space. The latter phenomenon seems to occur in vivo in multiple sclerosis, where it reflects the remission/relapse alternation. The extracellular proteasome-mediated inflammatory pathway may represent a general mechanism to control inflammation in inflammatory diseases.


Subject(s)
Multiple Sclerosis/metabolism , Osteopontin/metabolism , Proteasome Endopeptidase Complex/metabolism , Amino Acid Sequence , Chemotaxis/immunology , Endothelial Cells/metabolism , Extracellular Space/metabolism , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Humans , Lymphocytes/immunology , Lymphocytes/metabolism , Models, Molecular , Multiple Sclerosis/immunology , Osteopontin/chemistry , Osteopontin/immunology , Proteasome Endopeptidase Complex/immunology , Protein Conformation , Structure-Activity Relationship
20.
Gels ; 3(2)2017 Jun 08.
Article in English | MEDLINE | ID: mdl-30920519

ABSTRACT

Glucocorticoids are widely prescribed in treatment of rheumatoid arthritis, asthma, systemic lupus erythematosus, lymphoid neoplasia, skin and eye inflammations. However, well-documented adverse effects offset their therapeutic advantages. In this work, novel nano-hydrogels for the sustained delivery of dexamethasone were designed to increase both bioavailability and duration of the administered drug and reducing the therapeutic dose. Hydrogels are soft materials consisting of water-swollen cross-linked polymers to which the insertion of cyclodextrin (CD) moieties adds hydrophobic drug-complexing sites. Polyamidoamines (PAAs) are biocompatible and biodegradable polymers apt to create CD moieties in hydrogels. In this work, ß or γ-CD/PAA nanogels have been developed. In vitro studies showed that a pretreatment for 24⁻48 h with dexamethasone-loaded, ß-CD/PAA nanogel (nanodexa) inhibits adhesion of Jurkat cells to human umbilical vein endothelial cells (HUVEC) in conditions mimicking inflammation. This inhibitory effect was faster and higher than that displayed by free dexamethasone. Moreover, nanodexa inhibited COX-2 expression induced by PMA+A23187 in Jurkat cells after 24⁻48 h incubation in the 10-8⁻10-5 M concentration range, while dexamethasone was effective only at 10-5 M after 48 h treatment. Hence, the novel nanogel-dexamethasone formulation combines faster action with lower doses, suggesting the potential for being more manageable than the free drug, reducing its adverse side effects.

SELECTION OF CITATIONS
SEARCH DETAIL
...