Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 179
1.
Leukemia ; 36(1): 138-154, 2022 01.
Article En | MEDLINE | ID: mdl-34290359

Immune profiling in patients with monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM), and multiple myeloma (MM) provides the framework for developing novel immunotherapeutic strategies. Here, we demonstrate decreased CD4+ Th cells, increased Treg and G-type MDSC, and upregulation of immune checkpoints on effector/regulatory and CD138+ cells in MM patients, compared MGUS/SMM patients or healthy individuals. Among the checkpoints profiled, LAG3 was most highly expressed on proliferating CD4+ Th and CD8+ Tc cells in MM patients BMMC and PBMC. Treatment with antibody targeting LAG3 significantly enhanced T cells proliferation and activities against MM. XBP1/CD138/CS1-specific CTL generated in vitro displayed anti-MM activity, which was further enhanced following anti-LAG3 treatment, within the antigen-specific memory T cells. Treg and G-type MDSC weakly express LAG3 and were minimally impacted by anti-LAG3. CD138+ MM cells express GAL-3, a ligand for LAG3, and anti-GAL-3 treatment increased MM-specific responses, as observed for anti-LAG3. Finally, we demonstrate checkpoint inhibitor treatment evokes non-targeted checkpoints as a cause of resistance and propose combination therapeutic strategies to overcome this resistance. These studies identify and validate blockade of LAG3/GAL-3, alone or in combination with immune strategies including XBP1/CD138/CS1 multipeptide vaccination, to enhance anti-tumor responses and improve patient outcome in MM.


Antigens, CD/chemistry , Blood Proteins/antagonists & inhibitors , Galectins/antagonists & inhibitors , Immunosuppression Therapy/methods , Leukocytes, Mononuclear/immunology , Monoclonal Gammopathy of Undetermined Significance/immunology , Multiple Myeloma/immunology , T-Lymphocytes, Cytotoxic/immunology , Apoptosis , Case-Control Studies , Cell Proliferation , Follow-Up Studies , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Lymphocyte Activation , Monoclonal Gammopathy of Undetermined Significance/metabolism , Monoclonal Gammopathy of Undetermined Significance/pathology , Monoclonal Gammopathy of Undetermined Significance/therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Prognosis , Tumor Cells, Cultured , Lymphocyte Activation Gene 3 Protein
2.
Molecules ; 26(21)2021 Nov 05.
Article En | MEDLINE | ID: mdl-34771095

Previous reports have shown that consumption of wine has several health benefits; however, there are different types of wine. In the present study, red wines were investigated for their compositions of active ingredients. The interaction of each component in terms of its binding mode with different serum proteins was unraveled, and the components were implicated as drug candidates in clinical settings. Overall, the study indicates that red wines have a composition of flavonoids, non-flavonoids, and phenolic acids that can interact with the key regions of proteins to enhance their biological activity. Among them, rutin, resveratrol, and tannic acid have shown good binding affinity and possess beneficial properties that can enhance their role in clinical applications.


Biomarkers/blood , Blood Proteins/antagonists & inhibitors , Flavonoids/pharmacology , Wine/analysis , Alcoholic Beverages , Antioxidants/analysis , Binding Sites , Blood Proteins/chemistry , Blood Proteins/metabolism , Flavonoids/chemistry , Flavonoids/pharmacokinetics , Fluorometry/methods , Humans , Ligands , Models, Molecular , Molecular Conformation , Phenols , Protein Binding , Structure-Activity Relationship , Wine/adverse effects
3.
Glycobiology ; 31(10): 1390-1400, 2021 11 18.
Article En | MEDLINE | ID: mdl-34228782

Galectin-3 (Gal-3), a ß-galactoside-binding lectin, has been implicated in a plethora of pathological disorders including fibrosis, inflammation, cancer and metabolic diseases. TD139-a thio-digalactoside inhibitor developed by Galecto Biotech as a potential therapeutic for idiopathic pulmonary fibrosis-is the most advanced small-molecule Gal-3 inhibitor in clinical studies. It binds to human Gal-3 with high affinity but has lower affinity towards mouse and rat homologs, which is also manifested in the differential inhibition of Gal-3 function. Using biophysical methods and high-resolution X-ray co-crystal structures of TD139 and Gal-3 proteins, we demonstrate that a single amino acid change corresponding to A146 in human Gal-3 is sufficient for the observed reduction in the binding affinity of TD139 in rodents. Site-directed mutagenesis of A146V (in human Gal-3) and V160A (in mouse Gal-3) was sufficient to interchange the affinities, mainly by affecting the off rates of the inhibitor binding. In addition, molecular dynamics simulations of both wild-type and mutant structures revealed the sustained favorable noncovalent interactions between the fluorophenyl ring and the active site A146 (human Gal-3 and mouse V160A) that corroborate the finding from biophysical studies. Current findings have ramifications in the context of optimization of drug candidates against Gal-3.


Blood Proteins , Galectins , Thiogalactosides , Humans , Binding Sites/drug effects , Blood Proteins/antagonists & inhibitors , Blood Proteins/metabolism , Galectins/antagonists & inhibitors , Galectins/metabolism , Thiogalactosides/metabolism , Thiogalactosides/pharmacology
4.
Mol Med Rep ; 24(4)2021 10.
Article En | MEDLINE | ID: mdl-34328195

Oral squamous cell carcinoma (OSCC) is a cancer associated with high mortality (accounting for 3.1/100,000 deaths per year in Brazil in 2013) and a high frequency of amplification in the expression of the epidermal growth factor receptor (EGFR). Treatment with the EGFR inhibitor cetuximab leads to drug resistance in patients with OSCC due to unknown mechanisms. Galectin­3 (Gal­3) is a ß­galactoside binding lectin that regulates multiple signaling pathways in cells. The present study aimed to investigate the effect of Gal­3 in cetuximab­resistant (cet­R) OSCC. The OSCC HSC3 cell line was selected to establish a mouse xenograft model, which was treated with cetuximab to induce resistance. Subsequently, a Gal­3 inhibitor was used to treat cet­R tumors, and the tumor volume was monitored. The expression of Gal­3, phosphorylated (p)­ERK1/2 and p­Akt was assessed using immunohistochemistry. The combined effect of cetuximab and the Gal­3 inhibitor on HSC3 tumor xenografts was also investigated. HSC3 cells were cultured in vitro to investigate the regulatory effects of Gal­3 on ERK1/2 and Akt via western blotting. In addition, the effects of the Gal­3 inhibitor on the proliferation, colony formation, invasion and apoptosis of HSC3 cells were investigated by performing Cell Counting Kit­8, colony formation, Transwell and apoptosis assays, respectively. In cet­R OSCC tumors, increased expression of Gal­3, p­ERK1/2 and p­Akt was observed. Further research demonstrated that Gal­3 regulated the expression of both ERK1/2 and Akt in HSC3 cells by promoting phosphorylation. Moreover, the Gal­3 inhibitor decreased the proliferation and invasion, but increased the apoptosis of cet­R HSC3 cells. In addition, the Gal­3 inhibitor suppressed the growth of cet­R tumors. Collectively, the results indicated that the Gal­3 inhibitor and cetuximab displayed a synergistic inhibitory effect on OSCC tumors. In summary, the present study demonstrated that Gal­3 may serve an important role in cet­R OSCC. The combination of cetuximab and the Gal­3 inhibitor may display a synergistic antitumor effect, thereby inhibiting the development of cetuximab resistance in OSCC.


Blood Proteins/antagonists & inhibitors , Carcinoma, Squamous Cell/drug therapy , Drug Resistance, Neoplasm , Galectins/antagonists & inhibitors , Mouth Neoplasms/drug therapy , Animals , Antineoplastic Agents, Immunological/pharmacology , Apoptosis/drug effects , Blood Proteins/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cetuximab/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Galectins/genetics , Gene Knockdown Techniques , Humans , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
5.
Eur J Med Chem ; 220: 113500, 2021 Aug 05.
Article En | MEDLINE | ID: mdl-33962190

Galectin-3 plays a crucial role in cancerogenesis; its targeting is a prospective pathway in cancer diagnostics and therapy. Multivalent presentation of glycans was shown to strongly increase the affinity of glycoconjugates to galectin-3. Further strengthening of interaction with galectin-3 may be accomplished using artificial glycomimetics with apt aryl substitutions. We established a new, as yet undescribed chemoenzymatic method to produce selective C-3-substituted N,N'-diacetyllactosamine glycomimetics and coupled them to human serum albumin. From a library of enzymes, only ß-N-acetylhexosaminidase from Talaromyces flavus was able to efficiently synthesize the C-3-propargylated disaccharide. Various aryl residues were attached to the functionalized N,N'-diacetyllactosamine via click chemistry to assess the impact of the aromatic substitution. In ELISA-type assays with galectin-3, free glycomimetics exhibited up to 43-fold stronger inhibitory potency to Gal-3 than the lactose standard. Coupling to human serum albumin afforded multivalent neo-glycoproteins with up to 4209-fold increased inhibitory potency per glycan compared to the monovalent lactose standard. Surface plasmon resonance brought further information on the kinetics of galectin-3 inhibition. The potential of prepared neo-glycoproteins to target galectin-3 was demonstrated on colorectal adenocarcinoma DLD-1 cells. We investigated the uptake of neo-glycoproteins into cells and observed limited non-specific transport into the cytoplasm. Therefore, neo-glycoproteins primarily act as efficient scavengers of exogenous galectin-3 of cancer cells, inhibiting its interaction with the cell surface, and protecting T-lymphocytes against galectin-3-induced apoptosis. The present neo-glycoproteins combine the advantage of a straightforward synthesis, selectivity, non-toxicity, and high efficiency for targeting exogenous galectin-3, with possible application in the immunomodulatory treatment of galectin-3-overexpressing cancers.


Biomimetic Materials/pharmacology , Blood Proteins/antagonists & inhibitors , Galectins/antagonists & inhibitors , Glycoproteins/metabolism , Biomimetic Materials/chemical synthesis , Biomimetic Materials/chemistry , Blood Proteins/genetics , Blood Proteins/metabolism , Dose-Response Relationship, Drug , Galectins/genetics , Galectins/metabolism , Glycoproteins/chemistry , Humans , Kinetics , Molecular Structure , Structure-Activity Relationship
6.
J Immunother Cancer ; 9(4)2021 04.
Article En | MEDLINE | ID: mdl-33837055

BACKGROUND: PD-1/PD-L1 engagement and overexpression of galectin-3 (Gal-3) are critical mechanisms of tumor-induced immune suppression that contribute to immunotherapy resistance. We hypothesized that Gal-3 blockade with belapectin (GR-MD-02) plus anti-PD-1 (pembrolizumab) would enhance tumor response in patients with metastatic melanoma (MM) and head and neck squamous cell carcinoma (HNSCC). METHODS: We performed a phase I dose escalation study of belapectin+pembrolizumab in patients with advanced MM or HNSCC (NCT02575404). Belapectin was administered at 2, 4, or 8 mg/kg IV 60 min before pembrolizumab (200 mg IV every 3 weeks for five cycles). Responding patients continued pembrolizumab monotherapy for up to 17 cycles. Main eligibility requirements were a functional Eastern Cooperative Oncology Group status of 0-2, measurable or assessable disease, and no active autoimmune disease. Prior T-cell checkpoint antibody therapy was permitted. RESULTS: Objective response was observed in 50% of MM (7/14) and and 33% of HNSCC (2/6) patients. Belapectin+pembrolizumab was associated with fewer immune-mediated adverse events than anticipated with pembrolizumab monotherapy. There were no dose-limiting toxicities for belapectin within the dose range investigated. Significantly increased effector memory T-cell activation and reduced monocytic myeloid-derived suppressor cells (M-MDSCs) were observed in responders compared with non-responders. Increased baseline expression of Gal-3+ tumor cells and PD-1+CD8+ T cells in the periphery correlated with response as did higher serum trough levels of pembrolizumab. CONCLUSIONS: Belapectin+pembrolizumab therapy has activity in MM and HNSCC. Increased Gal-3 expression, expansion of effector memory T cells, and decreased M-MDSCs correlated with clinical response. Further investigation is planned.


Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood Proteins/antagonists & inhibitors , Galectins/antagonists & inhibitors , Head and Neck Neoplasms/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Multiple Myeloma/drug therapy , Pectins/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Squamous Cell Carcinoma of Head and Neck/drug therapy , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Blood Proteins/immunology , Female , Galectins/immunology , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/immunology , Humans , Immune Checkpoint Inhibitors/adverse effects , Male , Memory T Cells/drug effects , Memory T Cells/immunology , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/immunology , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Pectins/adverse effects , Programmed Cell Death 1 Receptor/immunology , Squamous Cell Carcinoma of Head and Neck/diagnosis , Squamous Cell Carcinoma of Head and Neck/immunology , Time Factors , Treatment Outcome
7.
Expert Opin Ther Pat ; 31(8): 709-721, 2021 Aug.
Article En | MEDLINE | ID: mdl-33749494

INTRODUCTION: Galectins are ubiquitous in nature. They have established themselves as a protein family of high therapeutic potential and play a role in a wide variety of diseases like cancer, fibrosis, and Alzheimer's. Within the galectin family, galectin- 1 and galectin- 3 have been widely studied and their roles and functions have now been well established. AREAS COVERED: In this review, we discuss the important advancements in the development of galectin-1 & 3 inhibitors. All patents filed detailing the divergent strategies to inhibit galectin-1 & 3 from 2016 to present have been covered and discussed. EXPERT OPINION: Over the past couple of decades, distinct galectin inhibitors have been synthesized, reported and studied. Among all, the mono and disaccharide-based antagonists have been found to be considerably successful. However, the cumbersome synthetic route followed to develop this class of inhibitors, in addition to complexity involved in making selective modifications within these molecules has posed a significant challenge. Recently, there have been numerous reports on heterocyclic-based galectin inhibitors. If these are established as potent galectin inhibitors, their ease of synthesis and tunability could overcome the potential drawbacks of carbohydrate-based inhibitors and could thus be exploited to develop efficient and highly specific galectin inhibitors.


Blood Proteins/antagonists & inhibitors , Galectin 1/antagonists & inhibitors , Galectins/antagonists & inhibitors , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Blood Proteins/metabolism , Drug Development , Fibrosis/drug therapy , Fibrosis/pathology , Galectin 1/metabolism , Galectins/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Patents as Topic
8.
Cancer Res ; 81(8): 2207-2219, 2021 04 15.
Article En | MEDLINE | ID: mdl-33608316

Chemotherapy-induced peripheral neuropathy (CIPN) is a severe dose-limiting side effect of taxanes such as paclitaxel and docetaxel. Despite the high medical needs, insufficient understanding of the complex mechanism underlying CIPN pathogenesis precludes any endorsed causal therapy to prevent or relieve CIPN. In this study, we report that elevation of plasma galectin-3 level is a pathologic change common to both patients with taxane-treated breast cancer with CIPN and a mouse model of taxane-related CIPN. Following multiple intraperitoneal injections of paclitaxel in mice, galectin-3 levels were elevated in Schwann cells within the sciatic nerve but not in other peripheral organs or cells expressing galectin-3. Consistent with this, paclitaxel treatment of primary cultures of rat Schwann cells induced upregulation and secretion of galectin-3. In vitro migration assays revealed that recombinant galectin-3 induced a chemotactic response of the murine macrophage cell line RAW 264.7. In addition, perineural administration of galectin-3 to the sciatic nerve of naive mice mimicked paclitaxel-induced macrophage infiltration and mechanical hypersensitivity. By contrast, chemical depletion of macrophages by clodronate liposomes suppressed paclitaxel-induced mechanical hypersensitivity despite the higher level of plasma galectin-3. Deficiency (Galectin-3 -/- mice) or pharmacologic inhibition of galectin-3 inhibited paclitaxel-induced macrophage infiltration and mechanical hypersensitivity. In conclusion, we propose that Schwann cell-derived galectin-3 plays a pronociceptive role via macrophage infiltration in the pathogenesis of taxane-induced peripheral neuropathy. Therapies targeting this phenomenon, which is common to patients with CIPN and mouse models, represent a novel approach to suppress taxane-related CIPN. SIGNIFICANCE: These findings demonstrate that the elevation of plasma galectin-3 is a CIPN-related pathologic change common to humans and mice, and that targeting galectin-3 is a therapeutic option to delay CIPN progression.


Galectins/blood , Macrophages/physiology , Pain Perception/physiology , Peripheral Nervous System Diseases/physiopathology , Schwann Cells/metabolism , Sciatic Nerve/metabolism , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Blood Proteins/antagonists & inhibitors , Blood Proteins/pharmacology , Blood Proteins/physiology , Cell Movement , Chemotaxis , Clodronic Acid/pharmacology , Disease Models, Animal , Docetaxel/adverse effects , Female , Galectins/antagonists & inhibitors , Galectins/pharmacology , Galectins/physiology , Humans , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Paclitaxel/adverse effects , Peripheral Nervous System Diseases/blood , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/prevention & control , Prospective Studies , Rats , Schwann Cells/drug effects , Sciatic Nerve/cytology , Sciatic Nerve/drug effects , Up-Regulation
9.
Expert Rev Clin Pharmacol ; 14(4): 457-464, 2021 Apr.
Article En | MEDLINE | ID: mdl-33612037

INTRODUCTION: Galectin-3 (Gal-3) is a ß-galactoside binding protein associated with many disease pathologies, including chronic inflammation and fibrogenesis. It has been implicated in the disease severity of NASH, although its precise role is unknown. Inhibition of Gal-3 has shown to improve and prevent fibrosis progression and has now reached phase III clinical trial in NASH patients. AREAS COVERED: This discusses the role of Gal-3 in NASH. It brings together the current findings of Gal-3 in NASH and hepatic fibrosis by analyzing recent data from animal model studies and clinical trials. EXPERT OPINION: Gal-3 inhibitors, in particular, Belapectin (GR-MD-02), have shown promising results for NASH with advanced fibrosis. In a phase 2 trial, Belapectin did not meet the primary endpoint. However, a sub-analysis of Belapectin among a separate group of patients without esophageal varices showed 2 mg/kg of GR-MD-02 reduced HVPG and the development of new varices. A subsequent study is under way, aiming to replicate the positive findings in phase 2 and demonstrate greater efficacy. If Belapectin is shown to be effective, it will be coupled with other drugs that target steatohepatitis to maximize efficacy and disease reversal.


Blood Proteins/antagonists & inhibitors , Galectins/antagonists & inhibitors , Liver Cirrhosis/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Disease Models, Animal , Disease Progression , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/physiopathology , Non-alcoholic Fatty Liver Disease/physiopathology , Pectins/administration & dosage , Pectins/pharmacology , Severity of Illness Index
10.
Bioorg Med Chem Lett ; 37: 127835, 2021 04 01.
Article En | MEDLINE | ID: mdl-33556574

Fluorescent biosensors are indispensable tools for molecular imaging, detection, and drug screening. Conventionally, fluorescent biosensors were constructed by incorporating fluorophores into ligands. Here, to develop ligand-independent biosensors, we demonstrated biosensor selection from a fluorophore-modified peptide phage library. In this library, the peptides were designed to form α-helical structures, and one cysteine, the probe modification site, was located at the center of four randomized residues on the same face of the helix. By conjugation with 4-nitrobenzoxadiazole (NBD), we constructed an NBD-modified phage library. We conducted selection against galectin-3 (Gal-3), a galactose-specific lectin associated with various biological events such as tumor metastasis and insulin resistance. After biopanning, we obtained NBD-modified peptides that selectively bind to Gal-3 from the library. The fluorescence intensity of the hit biosensors increased with the concentration of Gal-3, and this fluorescent response was visually observed.


Biosensing Techniques , Blood Proteins/antagonists & inhibitors , Fluorescent Dyes/pharmacology , Galectins/antagonists & inhibitors , Nitro Compounds/pharmacology , Oxadiazoles/pharmacology , Peptides/pharmacology , Blood Proteins/metabolism , Dose-Response Relationship, Drug , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Galectins/metabolism , Humans , Molecular Structure , Nitro Compounds/chemistry , Oxadiazoles/chemistry , Peptides/chemical synthesis , Peptides/chemistry , Structure-Activity Relationship
11.
Int J Biol Macromol ; 171: 550-559, 2021 Feb 28.
Article En | MEDLINE | ID: mdl-33444654

Larch arabinogalactan (AG), extracted from Larix gmelinii sawdust, was depolymerized by H2O2 oxidation and purified by gel column to yield a novel degraded fraction (AGD2). The structural analysis indicated AGD2 had lower arabinose content and molecular weight compared with AG, in which the ratio of galactose and arabinose was changed from 7:3 to 16:1, the molecular weight was decreased from 50.2 kDa to 3.7 kDa, and the chain conformation spread from highly branched structure to flexible strand. It was one kind of ß-D-(1 â†’ 3)-galactan with fewer ß-D-(1 â†’ 6)-Galp side branches at O-6 position. Further, the results of the Gal-3 binding and immunomodulatory assay suggested that the unbinding force of AGD2 onto Gal-3 was as twice as AG to be 76 ± 11 pN at the loading rate of 0.15 µm/s. It could better promote the secretion of pro-inflammatory cytokines (TNF-α, IL-6 and IL-1ß) than AG in a dose-dependent manner.


Galactans/isolation & purification , Larix/chemistry , Wood/chemistry , Animals , Arabinose/analysis , Blood Proteins/antagonists & inhibitors , Carbohydrate Conformation , Dose-Response Relationship, Drug , Galactans/chemistry , Galactans/pharmacology , Galactose/analysis , Galectins/antagonists & inhibitors , Immunologic Factors/chemistry , Immunologic Factors/isolation & purification , Immunologic Factors/pharmacology , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Methylation , Mice , Microscopy, Atomic Force , Nuclear Magnetic Resonance, Biomolecular , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/biosynthesis
12.
Curr Drug Discov Technol ; 18(3): 451-456, 2021.
Article En | MEDLINE | ID: mdl-31969105

AIMS: To utilize in silico-based approach for investigating the ability of PEGylated rapamycin as a competitive inhibitor to Galectin-3 for curing various diseases or that may provide an attractive strategy for treatment of a wide variety of tumors. BACKGROUND: Galectin-3 (Gal-3) signaling protein is a unique member of lectin family present at the cell surface, intracellularly in both the nucleus and cytoplasm and extracellularly in the general circulation. Circulating Gal-3 is present in both normal and cancer cells. High levels of circulating Gal-3 have been proven to be associated with inflammation and fibrosis in several acute and chronic conditions, which include neurological degeneration, inflammatory and immune responses, autoimmune diseases, diabetes, heart failure, atherosclerosis, response to infection, wound healing, liver, lung, and kidney disease. Gal-3 is known to regulate many biological activities including cell adhesion, angiogenesis, growth, apoptosis, migration, and metastasis. Rapamycin has been examined alone or in combination with other drugs for treatment of various cancers in clinical studies. Although it has shown promising therapeutic effects, its clinical development was interrupted by poor aqueous solubility and limited preferential distribution. To overcome these limitations, RA has been chemically modified to hydrophilic analogues, such as everolimus (EV). However, all these approaches can only partially increase the solubility, but has little effect on the blood distribution and pharmacokinetics. Therefore, it is necessary to explore other RA conjugates to improve aqueous solubility and tissue distribution profile. Recently we reported that RP-MPEG inhibits the growth of various cancer cell lines by acting on mammalian target of RP (mTOR) receptor site and it can be used for gastric cancer. OBJECTIVE: To construct various molecular weight RP-MPEG by replacing MPEG chain in 40-O-(2- hydroxyethyl) position of the EV and analyze their binding affinity to Gal-3. METHODS: The chemical structures of various molecular weight RP-MPEG were built using ChemSketch software. The molecular docking study was performed to find the best probable structure of RP-MPEG for competitive inhibition of the CRD, based on the interaction score. For that purpose, the 3D structures of RP and EV were obtained from NCBI PubChem compound database, where the structural protein-co-crystallized ligand complex of Gal-3 (TD2, as a native ligand) was retrieved from RCSB Protein Data Bank. All structures of the selected compounds, served as molecules for molecular modeling, were optimized through MOE.2014 software before docking. Hydrogen atoms and partial charges were added to the protein. Protein minimization was performed in gas solvation with the side chains, keeping it rigid and the ligand flexible. The selected site was isolated and minimized, followed by protonating the protein. The 3D ligands were minimized using MMFF94x with cutoffs of 10 to 12 Å. The hydrogens and charges were fixed, and the RMS gradient was set to 0.001 kcal/mol. The docking results were analyzed to identify and assess the binding affinity of these compounds to CRD using drug discovery software. RESULTS: Our results indicated that RP-MPEG with MW 1178.51 g/mol has a logP value of 3.79 and has possessed the strongest binding affinity toward CRD of Gal-3 with a docking score of -6.87. Compared with TD2, there were additional close contacts for RP-MPEG (MW 1178.51 g/mol), coming from three hydrogen bonds with Asp148, Arg162, and Arg144 which suggest that this ligand is a strong competitive inhibitor among the other molecules for Gal-3. CONCLUSION: RP-MPEG with the MW 1178.51 g/mol could be a promising blocker for various biological action of Gal-3 includes profibrotic activity, modulation of immune responses and inflammatory responses to cancer that contributes to neoplastic transformation, angiogenesis and metastasis. Other: The 95% confidence intervals (CIs) of the binding affinity (according to their mean and standard errors) were estimated with 2.5 and 97.5 percentile as the lower and upper bounds.


Antibiotics, Antineoplastic/pharmacology , Blood Proteins/antagonists & inhibitors , Galectins/antagonists & inhibitors , Neoplasms/drug therapy , Sirolimus/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/therapeutic use , Blood Proteins/metabolism , Galectins/metabolism , Humans , Molecular Docking Simulation , Molecular Weight , Polyethylene Glycols/chemistry , Protein Binding , Sirolimus/chemistry , Sirolimus/therapeutic use
13.
ChemMedChem ; 16(4): 713-723, 2021 02 17.
Article En | MEDLINE | ID: mdl-33156953

Calix[4]arene PTX008 is an angiostatic agent that inhibits tumor growth in mice by binding to galectin-1, a ß-galactoside-binding lectin. To assess the affinity profile of PTX008 for galectins, we used 15 N,1 H HSQC NMR spectroscopy to show that PTX008 also binds to galectin-3 (Gal-3), albeit more weakly. We identified the contact site for PTX008 on the F-face of the Gal-3 carbohydrate recognition domain. STD NMR revealed that the hydrophobic phenyl ring crown of the calixarene is the binding epitope. With this information, we performed molecular modeling of the complex to assist in improving the rather low affinity of PTX008 for Gal-3. By removing the N-dimethyl alkyl chain amide groups, we produced PTX013 whose reduced alkyl chain length and polar character led to an approximately eightfold stronger binding than PTX008. PTX013 also binds Gal-1 more strongly than PTX008, whereas neither interacts strongly, if at all, with Gal-7. In addition, PTX013, like PTX008, is an allosteric inhibitor of galectin binding to the canonical ligand lactose. This study broadens the scope for galectin targeting by calixarene-based compounds and opens the perspective for selective galectin blocking.


Blood Proteins/antagonists & inhibitors , Calixarenes/pharmacology , Galectins/antagonists & inhibitors , Phenols/pharmacology , Polysaccharides/pharmacology , Binding Sites , Blood Proteins/metabolism , Calixarenes/chemistry , Dose-Response Relationship, Drug , Galectins/metabolism , Humans , Magnetic Resonance Spectroscopy , Molecular Structure , Phenols/chemistry , Polysaccharides/chemistry , Structure-Activity Relationship
14.
Biomed Pharmacother ; 133: 111066, 2021 Jan.
Article En | MEDLINE | ID: mdl-33378967

Galectin-3 (gal-3), a member of the galectin family, is a glycoprotein with high affinity for ß-galactoside. Gal-3 is a cytoplasmically synthesized protein that can shuttle between the cytoplasm and nucleus and can even be transported to the membrane and secreted into the extracellular environment. Cardio/cerebrovascular diseases generally refer to ischemic or hemorrhagic diseases occurring in the heart, brain and systemic tissues, which are characterized by high morbidity, high disability rates and high mortality rates. To date, considerable research has demonstrated that gal-3 expression is aberrantly increased and plays important roles in cardio/cerebrovascular diseases, such as acute ischemic stroke (AIS), myocardial fibrosis, acute coronary syndrome (ACS), and heart failure (HF). Hence, understanding the biological roles of gal-3 in these diseases may be essential for cardio/cerebrovascular disease treatment and diagnosis to improve patient quality of life. In this review, we summarize current research on the roles of gal-3 in human cardiovascular diseases and potential inhibitors of gal-3, which may provide new strategies for disease therapies.


Blood Proteins/metabolism , Cardiovascular Diseases/metabolism , Cerebrovascular Disorders/metabolism , Galectins/metabolism , Signal Transduction , Animals , Blood Proteins/antagonists & inhibitors , Blood Proteins/genetics , Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/genetics , Cerebrovascular Disorders/drug therapy , Cerebrovascular Disorders/genetics , Galectins/antagonists & inhibitors , Galectins/genetics , Humans , Molecular Targeted Therapy , Signal Transduction/drug effects , Up-Regulation
15.
Int J Biochem Cell Biol ; 130: 105881, 2021 01.
Article En | MEDLINE | ID: mdl-33181315

Galectin-3 is a beta-galactoside-binding mammalian lectin and part of the 15 member galectin family that are evolutionarily highly conserved. It is the only chimeric protein with a C-terminal carbohydrate recognition domain (CRD) linked to a proline, glycine, and tyrosine rich additional N-terminal domain. Galectin-3 binds several cell surface glycoproteins via its CRD domain as well as undergoing oligomerization, via binding at the N-terminal or the CRD, resulting in the formation of a galectin-3 lattice on the cell surface. The galectin-3 lattice has been regarded as being a crucial mechanism whereby extracellular galectin-3 modulates cellular signalling by prolonging retention time or retarding lateral movement of cell surface receptors in the plasma membrane. As such galectin-3 can regulate various cellular functions such as diffusion, compartmentalization and endocytosis of plasma membrane glycoproteins and glycolipids and the functionality of membrane receptors. In multiple models of organ fibrosis, it has been demonstrated that galectin-3 is potently pro-fibrotic and modulates the activity of fibroblasts and macrophages in chronically inflamed organs. Increased galectin-3 expression also activates myofibroblasts resulting in scar formation and may therefore impact common fibrotic pathways leading to fibrosis in multiple organs. Over the last decade there has been a marked increase in the scientific literature investigating galectin-3 in a range of fibrotic diseases as well as the clinical development of new galectin-3 inhibitors. In this review we will examine the role of galectin-3 in fibrosis, the therapeutic strategies for inhibiting galectin-3 in fibrotic disease and the clinical landscape to date.


Endocytosis , Fibrosis/drug therapy , Galectins/antagonists & inhibitors , Inflammation/prevention & control , Animals , Blood Proteins/antagonists & inhibitors , Fibrosis/metabolism , Fibrosis/pathology , Galectins/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology
16.
Int J Mol Sci ; 21(23)2020 Dec 03.
Article En | MEDLINE | ID: mdl-33287402

Galectin-3 (Gal-3) is a ß-galactoside-binding protein belonging to the lectin family with pleiotropic regulatory activities and several physiological cellular functions, such as cellular growth, proliferation, apoptosis, differentiation, cellular adhesion, and tissue repair. Inflammation, tissue fibrosis and angiogenesis are the main processes in which Gal-3 is involved. It is implicated in the pathogenesis of several diseases, including organ fibrosis, chronic inflammation, cancer, atherosclerosis and other cardiovascular diseases (CVDs). This review aims to explore the connections of Gal-3 with cardiovascular diseases since they represent a major cause of morbidity and mortality. We herein discuss the evidence on the pro-inflammatory role of Gal-3 in the atherogenic process as well as the association with plaque features linked to lesion stability. We report the biological role and molecular mechanisms of Gal-3 in other CVDs, highlighting its involvement in the development of cardiac fibrosis and impaired myocardium remodelling, resulting in heart failure and atrial fibrillation. The role of Gal-3 as a prognostic marker of heart failure is described together with possible diagnostic applications to other CVDs. Finally, we report the tentative use of Gal-3 inhibition as a therapeutic approach to prevent cardiac inflammation and fibrosis.


Blood Proteins/genetics , Blood Proteins/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Galectins/genetics , Galectins/metabolism , Animals , Biomarkers , Blood Proteins/antagonists & inhibitors , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology , Disease Models, Animal , Disease Susceptibility , Fibrosis , Galectins/antagonists & inhibitors , Heart Failure , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation/complications , Inflammation/etiology , Inflammation/metabolism , Molecular Targeted Therapy
17.
Front Immunol ; 11: 550366, 2020.
Article En | MEDLINE | ID: mdl-33072090

Non-steroidal anti-inflammatory drugs (NSAIDs) induce ulcers in the gastrointestinal tract, including the stomach and small intestine. NSAID-induced gastric ulcers can be prevented by taking acid-neutralizing/inhibitory drugs and cytoprotective agents. In contrast, there are no medicines to control NSAID-induced small intestinal ulcers, which are accompanied by a mucosal invasion of bacteria and subsequent activation of immune cells. Galectin-3 (Gal3), an endogenous lectin, has anti-microbial and pro-inflammatory functions. In the small intestine, since Gal3 is highly expressed in epithelial cells constitutively and macrophages inducibly, the Gal3 level can affect microbiota composition and macrophage activation. We hypothesized that the modulation of Gal3 expression could be beneficial in NSAID-induced intestinal ulcers. Using Gal3 knockout (Gal3KO) mice, we determined whether Gal3 could be a therapeutic target in NSAID-induced intestinal ulcers. Following the administration of indomethacin, an NSAID, we found that small intestinal ulcers were less severe in Gal3KO mice than in wild-type (WT) mice. We also found that the composition of intestinal microbiota was different between WT and Gal3KO mice and that bactericidal antibiotic polymyxin B treatment significantly suppressed NSAID-induced ulcers. Furthermore, clodronate, a macrophage modulator, attenuated NSAID-induced ulcers. Therefore, Gal3 could be an exacerbating factor in NSAID-induced intestinal ulcers by affecting the intestinal microbiota population and macrophage activity. Inhibition of Gal3 may be a therapeutic strategy in NSAID-induced intestinal ulcers. Clinical Trial Registration: www.ClinicalTrials.gov, identifier NCT03832946.


Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Blood Proteins/metabolism , Galectins/metabolism , Intestinal Diseases/etiology , Intestinal Diseases/metabolism , Ulcer/etiology , Ulcer/metabolism , Animals , Biomarkers , Blood Proteins/antagonists & inhibitors , Disease Management , Disease Models, Animal , Disease Susceptibility , Galectins/antagonists & inhibitors , Immunophenotyping , Intestinal Diseases/drug therapy , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Molecular Targeted Therapy , Ulcer/drug therapy
18.
Int J Mol Sci ; 21(19)2020 Sep 26.
Article En | MEDLINE | ID: mdl-32993166

In this study, we aimed to develop a multifunctional drug/gene delivery system for the treatment of glioblastoma multiforme by combining the ligand-mediated active targeting and the pH-triggered drug release features of graphene oxide (GO). Toward this end, we load irinotecan (CPT-11) to cetuximab (CET)-conjugated GO (GO-CET/CPT11) for pH-responsive drug release after endocytosis by epidermal growth factor receptor (EGFR) over-expressed U87 human glioblastoma cells. The ultimate injectable drug/gene delivery system was designed by co-entrapping stomatin-like protein 2 (SLP2) short hairpin RNA (shRNA) and GO-CET/CPT11 in thermosensitive chitosan-g-poly(N-isopropylacrylamide) (CPN) polymer solution, which offers a hydrogel depot for localized, sustained delivery of the therapeutics after the in situ formation of CPN@GO-CET/CPT11@shRNA hydrogel. An optimal drug formulation was achieved by considering both the loading efficiency and loading content of CPT-11 on GO-CET. A sustained and controlled release behavior was found for CPT-11 and shRNA from CPN hydrogel. Confocal microscopy analysis confirmed the intracellular trafficking for the targeted delivery of CPT-11 through interactions of CET with EGFR on the U87 cell surface. The efficient transfection of U87 using SLP2 shRNA was achieved using CPN as a delivery milieu, possibly by the formation of shRNA/CPN polyplex after hydrogel degradation. In vitro cell culture experiments confirmed cell apoptosis induced by CPT-11 released from acid organelles in the cytoplasm by flow cytometry, as well as reduced SLP2 protein expression and inhibited cell migration due to gene silencing. Finally, in vivo therapeutic efficacy was demonstrated using the xenograft of U87 tumor-bearing nude mice through non-invasive intratumoral delivery of CPN@GO-CET/CPT11@shRNA by injection. Overall, we have demonstrated the novelty of this thermosensitive hydrogel to be an excellent depot for the co-delivery of anticancer drugs and siRNA. The in situ forming hydrogel will not only provide extended drug release but also combine the advantages offered by the chitosan-based copolymer structure for siRNA delivery to broaden treatment modalities in cancer therapy.


Blood Proteins , Chitosan , Drug Delivery Systems , Gene Transfer Techniques , Glioblastoma , Graphite , Irinotecan , Membrane Proteins , Neoplasm Proteins , RNA, Small Interfering , Blood Proteins/antagonists & inhibitors , Blood Proteins/genetics , Blood Proteins/metabolism , Cell Line, Tumor , Chitosan/chemistry , Chitosan/pharmacology , ErbB Receptors/agonists , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/therapy , Graphite/chemistry , Graphite/pharmacology , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Irinotecan/chemistry , Irinotecan/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology
19.
Carbohydr Polym ; 245: 116526, 2020 Oct 01.
Article En | MEDLINE | ID: mdl-32718630

Rhamnogalacturonan I (RG-I) pectin are regarded as strong galectin-3 (Gal-3) antagonist because of galactan sidechains. The present study focused on discussing the effects of more structural regions in pectin on the anti-Gal-3 activity. The water-soluble pectin (WSP) recovered from citrus canning processing water was categorized as RG-I pectin. The controlled enzymatic hydrolysis was employed to sequentially remove the α-1,5-arabinan, homogalaturonan and ß-1,4-galactan in WSP. The Gal-3-binding affinity KD (kd/ka) of WSP and debranched pectins were calculated to be 0.32 µM, 0.48 µM, 0.56 µM and 1.93 µM. Moreover, based on the more sensitive cell line (MCF-7) model, the IC30 value of WSP was lower than these of modified pectins, indicating decreased anti-Gal-3 activity. Our results suggested that the total amount of neutral sugar sidechains, the length of arabinan and cooperation between HG and RG-I played important roles in the anti-Gal-3 activity of WSP, not the Gal/Ara ratio or RG-I/HG ratio. These results provided a new insight into structure-activity relationship of citrus segment membrane RG-I as a galectin-3 antagonist and a new functional food.


Blood Proteins/antagonists & inhibitors , Cell Membrane/chemistry , Citrus/chemistry , Galactans/pharmacology , Galectins/antagonists & inhibitors , Pectins/chemistry , Pectins/pharmacology , Blood Proteins/metabolism , Cell Wall/chemistry , Fruit/chemistry , Galectins/metabolism , Humans , Hydrolysis , MCF-7 Cells , Pectins/metabolism , Plant Cells , Polysaccharides/chemistry , Protein Binding , Solubility , Structure-Activity Relationship , Water/chemistry
20.
Org Biomol Chem ; 18(20): 3903-3907, 2020 05 27.
Article En | MEDLINE | ID: mdl-32400847

Selective galectin inhibitors are valuable research tools and could also be used as drug candidates. In that context, TD139, a thiodigalactoside galectin-3 inhibitor, is currently being evaluated clinically for the treatment of idiopathic pulmonary fibrosis. Herein, we describe a new strategy for the preparation of TD139. Starting from inexpensive levoglucosan, we used a rarely employed reaction cascade: Payne rearrangement/azidation process leading to 3-azido-galactopyranose. The latter intermediate was efficiently converted into TD139 in a few simple and practical steps.


Blood Proteins/antagonists & inhibitors , Galectins/antagonists & inhibitors , Thiogalactosides/pharmacology , Triazoles/pharmacology , Blood Proteins/metabolism , Carbohydrate Conformation , Crystallography, X-Ray , Galectins/metabolism , Humans , Models, Molecular , Thiogalactosides/chemical synthesis , Thiogalactosides/chemistry , Triazoles/chemical synthesis , Triazoles/chemistry
...