lactoferrin has been researched along with Glioblastoma* in 13 studies
1 review(s) available for lactoferrin and Glioblastoma
Article | Year |
---|---|
Natural substances to potentiate canonical glioblastoma chemotherapy.
Topics: Anthraquinones; Biological Products; Brain Neoplasms; Catechols; Cell Cycle; Dose-Response Relationship, Drug; Drug Synergism; Glioblastoma; Lactoferrin; Tea Tree Oil; Temozolomide | 2021 |
12 other study(ies) available for lactoferrin and Glioblastoma
Article | Year |
---|---|
Gastrointestinally absorbable lactoferrin-heparin conjugate with anti-angiogenic activity for treatment of brain tumor.
Glioblastoma multiforme (GBM) is a central nervous system disease with poor prognosis. Curative treatments for GBM involve chemotherapy, radiotherapy, and surgical pathways. Recently, antiangiogenic therapy through medications has been tried to slow tumor growth, but the drugs can induce side effects. To overcome these limitations, we developed a new orally absorbable form of heparin that can attenuate angiogenic activity by binding to growth factors around the tumor tissue. We conjugated lactoferrin (Lf) to heparin because Lf can be orally absorbed, and it interacts with the lactoferrin receptor (Lf-R) expressed on the intestine, blood-brain barrier (BBB), and glioma tumor masses. We successfully conjugated Lf and heparin by amide bond formation, as evidenced by advanced physicochemical properties such as pharmacokinetics and stability in acidic condition. This new material inhibited angiogenesis in vitro without toxicity. In addition, Lf-heparin administered orally to GBM orthotopic mice was absorbed in the small intestine and delivered specifically to the brain tumor by receptor transcytosis (Lf-R). Lf-heparin further attenuated angiogenesis progression in GBM orthotopic mice. Based on these results, Lf-heparin shows potential as a new oral medication for treatment of glioblastoma. Topics: Animals; Blood-Brain Barrier; Brain Neoplasms; Glioblastoma; Heparin; Lactoferrin; Mice | 2023 |
The MIF promoter SNP rs755622 is associated with immune activation in glioblastoma.
Intratumoral heterogeneity is a defining hallmark of glioblastoma, driving drug resistance and ultimately recurrence. Many somatic drivers of microenvironmental change have been shown to affect this heterogeneity and, ultimately, the treatment response. However, little is known about how germline mutations affect the tumoral microenvironment. Here, we find that the single-nucleotide polymorphism (SNP) rs755622 in the promoter of the cytokine macrophage migration inhibitory factor (MIF) is associated with increased leukocyte infiltration in glioblastoma. Furthermore, we identified an association between rs755622 and lactotransferrin expression, which could also be used as a biomarker for immune-infiltrated tumors. These findings demonstrate that a germline SNP in the promoter region of MIF may affect the immune microenvironment and further reveal a link between lactotransferrin and immune activation. Topics: Glioblastoma; Humans; Intramolecular Oxidoreductases; Lactoferrin; Macrophage Migration-Inhibitory Factors; Polymorphism, Single Nucleotide; Promoter Regions, Genetic; Tumor Microenvironment | 2023 |
Enhanced activity of AZD5582 and SM-164 in rabies virus glycoprotein-lactoferrin-liposomes to downregulate inhibitors of apoptosis proteins in glioblastoma.
Upregulated proliferation of neoplastic cells from suppressing apoptotic signals associated with the inhibitors of apoptosis proteins (IAP) makes difficult the achievement of therapeutic efficiency against glioblastoma multiforme. Studies in the last few years have witnessed a paradigm focusing on targeting IAP using its antagonists, such as Smac mimetics, to restrain tumor malignancy. A Smac mimetic compound needs to penetrate the blood-brain barrier (BBB), and must be internalized into cerebral tumor for improved chemotherapy. Rabies virus glycoprotein (RVG) and lactoferrin (Lf)-grafted liposomes were developed in this study to carry two IAP antagonists, AZD5582 and SM-164, across the BBB and to induce apoptosis in U87 MG and human brain cancer stem cells (HBCSCs). Liposomes modified with RVG slightly reduced BBB tightness and enhanced capability of AZD5582 and SM-164 for traversing the barrier because of their brain-targeting ability. Immunofluorescence and western-blot results revealed that AZD5582- and SM-164-encapsulated liposomes facilitated mutual curative intensity, effectively triggered apoptosis of U87 MG and HBCSCs, reduced the expression of cellular IAP 1 (cIAP1) and X-linked IAP (XIAP), and enhanced the expression of caspase-3. Hence, RGV-Lf-liposomes carrying AZD5582 and SM-164 can be promising formulations to activate apoptosis of U87 MG and HBCSCs, and this functionalized drug delivery system targeting cIAP and XIAP is a potential strategy to cure glioblastoma in clinical cancer management. Topics: Alkynes; Antineoplastic Agents; Apoptosis; Brain Neoplasms; Bridged Bicyclo Compounds, Heterocyclic; Cell Line, Tumor; Glioblastoma; Humans; Lactoferrin; Liposomes; Oligopeptides; Rabies virus; Triazoles | 2022 |
Facile synthesis of lactoferrin conjugated ultra small large pore silica nanoparticles for the treatment of glioblastoma.
The blood brain barrier (BBB) and blood tumour barrier (BTB) remain a major roadblock for delivering therapies to treat brain cancer. Amongst brain cancers, glioblastoma (GBM) is notoriously difficult to treat due to the challenge of delivering chemotherapeutic drugs across the BBB and into the tumour microenvironment. Consequently, GBM has high rates of tumour recurrence. Currently, limited numbers of chemotherapies are available that can cross the BBB to treat GBM. Nanomedicine is an attractive solution for treating GBM as it can augment drug penetration across the BBB and into the heterogeneous tumour site. However, very few nanomedicines exist that can easily overcome both the BBB and BTB owing to difficulty in synthesizing nanoparticles that meet the small size and surface functionality restrictions. In this study, we have developed for the first-time, a room temperature protocol to synthesise ultra-small size with large pore silica nanoparticles (USLP, size ∼30 nm, pore size >7 nm) with the ability to load high concentrations of chemotherapeutic drugs and conjugate a targeting moiety to their surface. The nanoparticles were conjugated with lactoferrin (>80 kDa), whose receptors are overexpressed by both the BBB and GBM, to achieve additional active targeting. Lactoferrin conjugated USLP (USLP-Lf) were loaded with doxorubicin - a chemotherapy agent that is known to be highly effective against GBM Topics: Blood-Brain Barrier; Brain Neoplasms; Cell Line, Tumor; Drug Delivery Systems; Glioblastoma; Humans; Lactoferrin; Nanoparticles; Silicon Dioxide; Tumor Microenvironment | 2021 |
Native and iron-saturated bovine lactoferrin differently hinder migration in a model of human glioblastoma by reverting epithelial-to-mesenchymal transition-like process and inhibiting interleukin-6/STAT3 axis.
Glioblastoma, the most lethal form of brain cancer, is characterized by fast growth, migration and invasion of the surrounding parenchyma, with epithelial-to-mesenchymal transition (EMT)-like process being mostly responsible for tumour spreading and dissemination. A number of actors, including cadherins, vimentin, transcriptional factors such as SNAIL, play critical roles in the EMT process. The interleukin (IL)-6/STAT3 axis has been related to enhanced glioblastoma's migration and invasion abilities as well. Here, we present data on the differential effects of native and iron-saturated bovine lactoferrin (bLf), an iron-chelating glycoprotein of the innate immune response, in inhibiting migration in a human glioblastoma cell line. Through a wound healing assay, we found that bLf was able to partially or completely hinder cell migration, depending on its iron saturation rate. At a molecular level, bLf down-regulated both SNAIL and vimentin expression, while inducing a notable increase in cadherins' levels and inhibiting IL-6/STAT3 axis. Again, these effects positively correlated to bLf iron-saturation state, with the Holo-form resulting more efficient than the native one. Overall, our data suggest that bLf could represent a novel and efficient adjuvant treatment for glioblastoma's standard therapeutic approaches. Topics: Cadherins; Cell Line, Tumor; Cell Movement; Down-Regulation; Epithelial-Mesenchymal Transition; Gene Expression Regulation, Neoplastic; Glioblastoma; Humans; Interleukin-6; Iron; Lactoferrin; Snail Family Transcription Factors; STAT3 Transcription Factor; Up-Regulation; Vimentin | 2020 |
Hyaluronic acid tethered pH-responsive alloy-drug nanoconjugates for multimodal therapy of glioblastoma: An intranasal route approach.
In the present investigation, FePt alloy nanoparticles were synthesized with controlled size and elemental composition followed by surface modification using (3-Aminopropyl) triethoxysilane (APTES). Lenalidomide was covalently bound to FePt-NH Topics: Administration, Intranasal; Alloys; Animals; Blood-Brain Barrier; Cell Line, Tumor; Cell Survival; Dogs; Drug Liberation; Endocytosis; Glioblastoma; Humans; Hyaluronic Acid; Hydrogen-Ion Concentration; Hyperthermia, Induced; Iron; Lactoferrin; Lenalidomide; Male; Mucins; Nanoconjugates; Oleic Acid; Photoelectron Spectroscopy; Phototherapy; Platinum; Rats, Wistar; Reactive Oxygen Species; Spectroscopy, Fourier Transform Infrared; Swine | 2019 |
Aurora kinase B siRNA-loaded lactoferrin nanoparticles potentiate the efficacy of temozolomide in treating glioblastoma.
To investigate the efficacy of lactoferrin nanoparticles (LfNPs) in delivering siRNA across the blood-brain barrier to treat glioblastoma multiforme (GBM) and with an additional objective of potentiation of conventional temozolomide (TMZ) chemotherapy.. Aurora kinase B (AKB) siRNA-loaded nanoparticles (AKB-LfNPs) were prepared with milk protein, lactoferrin, by water in oil emulsion method. AKB-LfNPs were tested in cell lines and in GBM orthotopic mouse model with and without TMZ treatment.. AKB silencing, cytotoxicity and cell cycle arrest by these LfNPs were shown to be effective on GL261 cells. Tumor growth was significantly lower in AKB-LfNPs alone and in combination with TMZ treated mice and increased the survival by 2.5-times.. Treatment of AKB-LfNPs to GBM mice improves life expectancy and has potential to combine with conventional chemotherapy. Topics: Animals; Apoptosis; Aurora Kinase B; Blood-Brain Barrier; Cell Line, Tumor; Cell Proliferation; Cell Survival; Dacarbazine; Glioblastoma; Humans; Lactoferrin; Mice; RNA, Small Interfering; Temozolomide; Xenograft Model Antitumor Assays | 2018 |
Brain targeted delivery of carmustine using solid lipid nanoparticles modified with tamoxifen and lactoferrin for antitumor proliferation.
Solid lipid nanoparticles (SLNs) conjugated with tamoxifen (TX) and lactoferrin (Lf) were applied to carry anticancer carmustine (BCNU) across the blood-brain barrier (BBB) for enhanced antiproliferation against glioblastoma multiforme (GBM). BCNU-loaded SLNs with modified TX and Lf (TX-Lf-BCNU-SLNs) were used to penetrate a monolayer of human brain-microvascular endothelial cells (HBMECs) and human astrocytes and to target malignant U87MG cells. The surface TX and Lf on TX-Lf-BCNU-SLNs improved the characteristics of sustained release for BCNU. When compared with BCNU-loaded SLNs, TX-Lf-BCNU-SLNs increased the BBB permeability coefficient for BCNU about ten times. In addition, TX-BCNU-SLNs considerably promoted the fluorescent intensity of intracellular acetomethoxy derivative of calcein (calcein-AM) in HBMECs via endocytosis. However, the conjugated Lf could only slightly increase the fluorescence of calcein-AM. Moreover, the order of formulation in the inhibition to U87MG cells was TX-Lf-BCNU-SLNs>TX-BCNU-SLNs>Lf-BCNU-SLNs>BCNU-SLNs. TX-Lf-BCNU-SLNs can be effective in infiltrating the BBB and delivering BCNU to GBM for future chemotherapy application. Topics: Antineoplastic Agents; Astrocytes; Blood-Brain Barrier; Brain; Carmustine; Cell Line, Tumor; Delayed-Action Preparations; Drug Delivery Systems; Endothelial Cells; Fluoresceins; Glioblastoma; Humans; Lactoferrin; Lipids; Nanoparticles; Permeability; Tamoxifen | 2016 |
Exogenous cathepsin G upregulates cell surface MHC class I molecules on immune and glioblastoma cells.
Major histocompatibility complex (MHC) class I molecules present antigenic peptides to cytotoxic T cells. During an adaptive immune response, MHC molecules are regulated by several mechanisms including lipopolysaccharide (LPS) and interferon gamma (IFN-g). However, it is unclear whether the serine protease cathepsin G (CatG), which is generally secreted by neutrophils at the site of inflammation, might regulate MHC I molecules. We identified CatG, and to a higher extend CatG and lactoferrin (LF), as an exogenous regulator of cell surface MHC I expression of immune cells and glioblastoma stem cells. In addition, levels of MHC I molecules are reduced on dendritic cells from CatG deficient mice compared to their wild type counterparts. Furthermore, cell surface CatG on immune cells, including T cells, B cells, and NK cells triggers MHC I on THP-1 monocytes suggesting a novel mechanism for CatG to facilitate intercellular communication between infiltrating cells and the respective target cell. Subsequently, our findings highlight the pivotal role of CatG as a checkpoint protease which might force target cells to display their intracellular MHC I:antigen repertoire. Topics: Animals; Cathepsin G; Cell Line; Cell Line, Tumor; Cell Membrane; Disease Models, Animal; Female; Gene Expression Regulation, Neoplastic; Glioblastoma; Histocompatibility Antigens Class I; Humans; Immune System; Lactoferrin; Male; Mice; Mice, Knockout; Monocytes; Neoplastic Stem Cells; Proteolysis | 2016 |
Targeting delivery of etoposide to inhibit the growth of human glioblastoma multiforme using lactoferrin- and folic acid-grafted poly(lactide-co-glycolide) nanoparticles.
Lactoferrin (Lf) and folic acid (FA) were crosslinked on poly(lactide-co-glycolide) (PLGA) nanoparticles (NPs) for transporting etoposide across the blood-brain barrier (BBB) and treating human brain malignant glioblastoma. Lf- and FA-grafted PLGA NPs (Lf/FA/PLGA NPs) were employed to permeate the monolayer of human brain-microvascular endothelial cells (HBMECs) regulated by human astrocytes and to inhibit the multiplication of U87MG cells. Lf/FA/PLGA NPs showed a satisfactory entrapment efficiency of etoposide and characteristics of sustained drug release. When compared with PLGA NPs, the permeability coefficient for etoposide across the BBB using Lf/FA/PLGA NPs increased about twofold. The antiproliferative efficacy against the growth of U87MG cells was in the following order: Lf/FA/PLGA NPs>FA/PLGA NPs>PLGA NPs>free etoposide solution. In addition, the targeting ability of Lf/FA/PLGA NPs was evidenced by immunostaining of Lf receptor on HBMECs and folate receptor on U87MG cells during endocytosis. Lf/FA/PLGA NPs with loaded etoposide can be a promising anticancer pharmacotherapy to enhance the delivery of etoposide to malignant brain tumors for preclinical trials. Topics: Antineoplastic Agents, Phytogenic; Astrocytes; Blood-Brain Barrier; Brain Neoplasms; Cell Line, Tumor; Cell Survival; Cells, Cultured; Drug Delivery Systems; Endothelial Cells; Etoposide; Folate Receptors, GPI-Anchored; Folic Acid; Glioblastoma; Humans; Lactic Acid; Lactoferrin; Microscopy, Electron, Scanning; Microscopy, Electron, Transmission; Nanoparticles; Polyglycolic Acid; Polylactic Acid-Polyglycolic Acid Copolymer; Receptors, Cell Surface; Topoisomerase II Inhibitors | 2015 |
In vitro and in vivo effect of human lactoferrin on glioblastoma growth.
Human lactoferrin (HLF) is a natural protein with antitumor activity. The aim of this study was to investigate the effects of HLF alone and in combination with temozolomide (TMZ), a conventional chemotherapeutic, on human glioblastoma (GBM) cells.. The authors cultured fresh human primary cell lines NMD and FN and the continuous cell line U87MG to evaluate proliferation in the presence of HLF alone at different doses (1, 10, and 100 mg/ml, and 1 mg/ml) and in combination with TMZ. In in vivo experiments they assessed tumor size reduction in CD1 nude mice carrying an orthotopic GBM xenograft and orally treated with HLF.. Lactoferrin causes growth inhibition in the NMD and FN primary cell lines and in the U87MG continuous cell line. This inhibition seemed to be modulated by the downregulation of cyclin D1 and D4. Western blot and fluorescence-activated cell sorting analysis showed inhibition of the cell cycle in G0/G1 and G2 phases. When administered in nude mice, HLF (60 mg/kg/day) decreased tumor size about 30%, as shown in both histological analyses and high-field brain MRI. Administration of HLF with TMZ enhanced the effect of chemotherapy both in vitro and in vivo.. This study demonstrated that HLF can inhibit GBM cell growth, suggesting that this nontoxic substance may have a role in potentiating the effect of current TMZ treatment of GBM. Topics: Animals; Antineoplastic Agents, Alkylating; Cell Proliferation; Dacarbazine; Drug Therapy, Combination; Glioblastoma; Humans; Lactoferrin; Male; Mice; Mice, Nude; Temozolomide; Tumor Cells, Cultured | 2015 |
Immunohistochemical detection of lactoferrin in human astrocytomas and multiforme glioblastomas.
The presence of lactoferrin in astrocytomas, anaplastic astrocytomas and multiforme glioblastomas was determined by immunohistochemistry; the staining intensity and the percentage of neoplastic stained cells were graded and statistical analysis was performed by non-parametric methods. A moderate to strong diffuse immunoreactivity for lactoferrin was shown in glial elements of astrocytomas, while the positivity was progressively reduced in anaplastic astrocytomas and in multiforme glioblastomas, some of which were unstained; a highly significant difference was found between scores relative to astrocytomas and glioblastomas. We suggest that the lactoferrin may be produced by neoplastic astrocytes which permits a greater availability of iron for metabolic cellular processes. Alternatively, the cytoplasmic localization of lactoferrin in neoplastic astrocytes may be the consequence of defective or functionally impaired lactoferrin receptors at the cellular surface. Topics: Astrocytoma; Brain Neoplasms; Female; Glioblastoma; Humans; Immunohistochemistry; Iron; Lactoferrin; Male; Middle Aged; Staining and Labeling | 1999 |