Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 668
Filter
1.
Bioconjug Chem ; 35(5): 674-681, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38695582

ABSTRACT

Aptamers are widely used molecular recognition tools in targeted therapy, but their ability to effectively penetrate deep into solid tumors remains a significant challenge, leading to suboptimal treatment efficacy. Here, we developed a polyfluoroalkyl (PFA) decoration strategy to enhance aptamer recognition, cell internalization, and solid tumor penetration. Our results indicate that PFA with around 11 fluorine atoms significantly improves aptamer internalization both in vitro and in vivo settings. However, we also observed that the use of PFA tags containing 19 and 23 fluorine atoms on aptamers resulted in nonspecific cell anchoring in control cell lines, affecting the specificity of aptamers. Overall, we found that using a chemical modification strategy could enhance the deep tumor penetration ability of aptamers and validate their effectiveness in vivo. This approach has significant practical applications in targeted drug delivery for cancer treatment.


Subject(s)
Aptamers, Nucleotide , Receptor Protein-Tyrosine Kinases , Aptamers, Nucleotide/chemistry , Humans , Animals , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Cell Line, Tumor , Mice , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/metabolism , Drug Delivery Systems/methods
2.
Hepatol Commun ; 8(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38668724

ABSTRACT

BACKGROUND: Primary sclerosing cholangitis is a progressive inflammatory liver disease characterized by biliary and liver fibrosis. Vascular adhesion protein-1 (VAP-1) is important in the inflammatory process driving liver fibrosis. We evaluated the safety and efficacy of VAP-1 blockade with a monoclonal antibody (timolumab, BTT1023) in patients with primary sclerosing cholangitis. METHODS: BUTEO was a prospective, single-arm, open-label, multicenter, phase II trial, conducted in 6 centers in the United Kingdom. Patients with primary sclerosing cholangitis aged 18-75 years had an alkaline phosphatase value of >1.5 times the upper limit of normal. The dose-confirmatory stage aimed to confirm the safety of timolumab through the incidence of dose-limiting toxicity and sufficient trough levels of circulating antibody to block VAP-1 function. The primary outcome of the dose-expansion portion of the trial was patient's response to timolumab at day 99, as measured by a reduction in serum alkaline phosphatase by 25% or more from baseline to day 99. RESULTS: Twenty-three patients were recruited: 7 into the initial dose-confirmatory stage and a further 16 into an expansion stage. Timolumab (8 mg/kg) was confirmed to be safe for the duration of administration with sufficient circulating levels. Only 2 of the 18 evaluable patients (11.1%) achieved a reduction in alkaline phosphatase levels of 25% or more, and both the proportion of circulating inflammatory cell populations and biomarkers of fibrosis remained unchanged from baseline. CONCLUSIONS: The BUTEO trial confirmed 8 mg/kg timolumab had no short-term safety signals and resulted in sufficient circulating levels of VAP-1 blocking timolumab. However, the trial was stopped after an interim assessment due to a lack of efficacy as determined by no significant change in serum liver tests.


Subject(s)
Amine Oxidase (Copper-Containing) , Cell Adhesion Molecules , Cholangitis, Sclerosing , Humans , Male , Female , Middle Aged , Adult , Cholangitis, Sclerosing/drug therapy , Cholangitis, Sclerosing/blood , Amine Oxidase (Copper-Containing)/blood , Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Cell Adhesion Molecules/blood , Cell Adhesion Molecules/antagonists & inhibitors , Prospective Studies , Aged , Treatment Outcome , Young Adult , Alkaline Phosphatase/blood , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/adverse effects , Adolescent
3.
Small ; 20(19): e2307045, 2024 May.
Article in English | MEDLINE | ID: mdl-38100142

ABSTRACT

Since WHO has declared the COVID-19 outbreak a global pandemic, nearly seven million deaths have been reported. This efficient spread of Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is facilitated by the ability of the spike glycoprotein to bind multiple cell membrane receptors. Although ACE2 is identified as the main receptor for SARS-CoV-2, other receptors could play a role in viral entry. Among others, C-type lectins such as DC-SIGN are identified as efficient trans-receptor for SARS-CoV-2 infection, so the use of glycomimetics to inhibit the infection through the DC-SIGN blockade is an encouraging approach. In this regard, multivalent nanostructures based on glycosylated [60]fullerenes linked to a central porphyrin scaffold have been designed and tested against DC-SIGN-mediated SARS-CoV-2 infection. First results show an outstanding inhibition of the trans-infection up to 90%. In addition, a deeper understanding of nanostructure-receptor binding is achieved through microscopy techniques, high-resolution NMR experiments, Quartz Crystal Microbalance experiments, and molecular dynamic simulations.


Subject(s)
Cell Adhesion Molecules , Fullerenes , Lectins, C-Type , Porphyrins , Receptors, Cell Surface , SARS-CoV-2 , SARS-CoV-2/drug effects , Lectins, C-Type/metabolism , Lectins, C-Type/antagonists & inhibitors , Humans , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Fullerenes/chemistry , Fullerenes/pharmacology , Porphyrins/chemistry , Porphyrins/pharmacology , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , COVID-19/virology , Protein Binding , Molecular Dynamics Simulation , COVID-19 Drug Treatment
4.
Curr Treat Options Oncol ; 23(4): 578-593, 2022 04.
Article in English | MEDLINE | ID: mdl-35312963

ABSTRACT

OPINION STATEMENT: Nectin-4 is a tumor-associated antigen that is highly expressed on various cancer cells, and it has been further proposed to have roles in tumor development and propagation ranging from cellular proliferation to motility and invasion. Nectin-4 blockade reduces tumor proliferation and induces apoptosis in several malignancies. Nectin-4 has been used as a potential target in antibody-drug conjugate (ADC) development. Enfortumab vedotin, an ADC against Nectin-4, has demonstrated efficacy against solid tumor malignancies. Enfortumab vedotin has received US Food and Drug Administration approval for treating urothelial cancer. Furthermore, the efficacy of ADCs against Nectin-4 against solid tumors other than urothelial cancer has been demonstrated in preclinical studies, and clinical trials examining the effects of enfortumab vedotin are ongoing. Recently, Nectin-4 was reported to be highly expressed in several skin cancers, including malignant melanoma, cutaneous squamous cell carcinoma, and extramammary Paget's disease, and involved in tumor progression and survival in retrospective studies. Nectin-4-targeted therapies and ADCs against Nectin-4 could therefore be novel therapeutic options for skin cancers. This review highlights current knowledge on Nectin-4 in malignant tumors, the efficacy of enfortumab vedotin in clinical trials, and the prospects of Nectin-4-targeted agents against skin cancers.


Subject(s)
Antibodies, Monoclonal , Carcinoma, Squamous Cell , Cell Adhesion Molecules , Immunoconjugates , Molecular Targeted Therapy , Skin Neoplasms , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Cell Adhesion Molecules/antagonists & inhibitors , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Retrospective Studies , Skin Neoplasms/drug therapy , Urinary Bladder Neoplasms/drug therapy , Urologic Neoplasms/drug therapy
5.
Int J Mol Sci ; 23(3)2022 Jan 22.
Article in English | MEDLINE | ID: mdl-35163164

ABSTRACT

The involvement of periostin (POSTN) in non-small-cell lung cancer (NSCLC) migration, invasion, and its underlying mechanisms has not been well established. The present study aims to determine epithelial POSTN expression in NSCLC and to assess associations with clinicopathological factors and prognosis as well as to explore the effects of POSTN knockdown on tumor microenvironment and the migration and invasion of lung cancer cells. Immunohistochemistry was used to evaluate epithelial POSTN expression in NSCLC. POSTN mRNA expression in the dissected lung cancer cells was confirmed by laser capture microdissection and real-time PCR. A549 cells were used for transfecting shRNA-POSTN lentiviral particles. Wound healing and Transwell invasion assays were used to assess the migratory and invasive abilities of A549 cells transfected with POSTN-specific short hairpin (sh)RNA. The results demonstrated significantly higher cytoplasmic POSTN expression in the whole NSCLC group compared to non-malignant lung tissue (NMLT). POSTN expression in cancer cells may be considered to be an independent prognostic factor for survival in NSCLC. POSTN knockdown significantly inhibited A549 cell migration and invasion capabilities in vitro. The activity and the expression level of matrix metalloproteinase-2 (MMP-2) were significantly decreased in A549.shRNA compared to control cells. In summary, POSTN may regulate lung cancer cell invasiveness by modulating the expression of MMP-2 and may represent a potential target for novel therapeutic intervention for NSCLC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion Molecules/metabolism , Cell Movement , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Silencing , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Matrix Metalloproteinase 2/genetics , Neoplasm Invasiveness , Prognosis , Survival Rate , Tumor Cells, Cultured , Tumor Microenvironment
6.
Cell Mol Life Sci ; 79(2): 88, 2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35067832

ABSTRACT

Junctional adhesion molecule (JAM)-A is a cell adhesion receptor localized at epithelial cell-cell contacts with enrichment at the tight junctions. Its role during cell-cell contact formation and epithelial barrier formation has intensively been studied. In contrast, its role during collective cell migration is largely unexplored. Here, we show that JAM-A regulates collective cell migration of polarized epithelial cells. Depletion of JAM-A in MDCK cells enhances the motility of singly migrating cells but reduces cell motility of cells embedded in a collective by impairing the dynamics of cryptic lamellipodia formation. This activity of JAM-A is observed in cells grown on laminin and collagen-I but not on fibronectin or vitronectin. Accordingly, we find that JAM-A exists in a complex with the laminin- and collagen-I-binding α3ß1 integrin. We also find that JAM-A interacts with tetraspanins CD151 and CD9, which both interact with α3ß1 integrin and regulate α3ß1 integrin activity in different contexts. Mapping experiments indicate that JAM-A associates with α3ß1 integrin and tetraspanins CD151 and CD9 through its extracellular domain. Similar to depletion of JAM-A, depletion of either α3ß1 integrin or tetraspanins CD151 and CD9 in MDCK cells slows down collective cell migration. Our findings suggest that JAM-A exists with α3ß1 integrin and tetraspanins CD151 and CD9 in a functional complex to regulate collective cell migration of polarized epithelial cells.


Subject(s)
Cell Adhesion Molecules/metabolism , Integrin alpha3beta1/metabolism , Tetraspanin 24/metabolism , Tetraspanin 29/metabolism , Animals , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Line , Cell Movement/drug effects , Dogs , Doxorubicin/pharmacology , Humans , Junctional Adhesion Molecule A/antagonists & inhibitors , Junctional Adhesion Molecule A/genetics , Madin Darby Canine Kidney Cells , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism
7.
Chem Biol Interact ; 353: 109811, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-35016848

ABSTRACT

Pancreatic cancer will be the second leading cause of cancer-related mortality worldwide due to its high rate of metastasis. Cathepsins (CATs) are effectors of invasive growth in various cancers. Currently, targeting CATs represents an attractive strategy for the treatment of highly metastatic cancers with high CATs activity, such as pancreatic cancer. To develop a stronger antimetastatic agent, ASPER-29, a novel inhibitor of CATs designed by using the asperphenamate derivative BBP as a lead compound, was synthesized, and its therapeutic potential in pancreatic cancer metastasis was investigated in this study. Molecular docking and enzyme inhibition assays proved that ASPER-29 can inhibit the activity of CAT-L and CAT-S by binding with these enzymes in classical action modes. Furthermore, ASPER-29 significantly inhibited the activity of CAT-L and CAT-S but had no effect on their expression in PANC-1 and BxPC-3 cells. The in vitro antimetastatic activities of ASPER-29 were examined by wound healing and Transwell chamber assays. We found that ASPER-29 inhibited the migration and invasion of PANC-1 and BxPC-3 cells in a concentration-dependent manner. Moreover, the in vivo antimetastatic effects of ASPER-29 were confirmed in a mouse xenotransplantation model. H&E staining and immunohistochemistry assays of Ki67 and CEACAM6 proved that ASPER-29 treatment significantly blocked the metastasis of pancreatic cancer cells to lung and liver tissues. Additionally, the activity of both CAT-L and CAT-S was markedly inhibited in the lung and liver tissues of ASPER-29-administered mice compared with the mice in the model group, suggesting that the metastasis-blocking effect of ASPER-29 should be mediated via inhibition of the activity of CAT-L and CAT-S in pancreatic cancer cells. Together, our results demonstrated that ASPER-29, as a novel inhibitor of CAT-L and CAT-S, possessed the evident ability to block the metastasis of pancreatic cancer cells.


Subject(s)
Cathepsin L/antagonists & inhibitors , Cathepsins/antagonists & inhibitors , Cell Movement/drug effects , Protease Inhibitors/pharmacology , Animals , Antigens, CD/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Binding Sites , Cathepsin L/metabolism , Cathepsins/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Humans , Liver/metabolism , Liver/pathology , Lung/metabolism , Lung/pathology , Mice , Mice, Nude , Molecular Docking Simulation , Neoplasm Metastasis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Protease Inhibitors/therapeutic use , Transplantation, Heterologous
8.
Pharmacol Rep ; 74(1): 40-46, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35001320

ABSTRACT

Vascular adhesion protein-1 (VAP-1) is a bifunctional protein that has the ability to catalyze the deamination of primary amines and is involved in the production of hydrogen peroxide, aldehydes, and advanced glycation end products (AGEs). VAP-1 is usually stored in intracellular vesicles of endothelial cells, smooth muscles, and adipocytes. It is responsible for leukocyte transmigration and adhesion. Overexpression of VAP-1 exacerbates oxidative stress and modulates a variety of inflammatory mediators linked with diabetic complications. Numerous studies have suggested the association of increased insulin levels with serum VAP-1 (sVAP-1). Preclinical research evidence suggests the increased activity of sVAP-1 in type 1 and 2 diabetes. Scientific reports on VAP-1 inhibitors have shown a reduction in severity in diabetic animal models. VAP-1 is a potential target of a therapeutically effective line of treatment for diabetes and diabetic complications such as nephropathy and retinopathy. The primary focus of this review is the role of VAP-1 in diabetes and its associated microvascular complications.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Cell Adhesion Molecules/metabolism , Diabetes Mellitus , Diabetic Angiopathies , Hypoglycemic Agents/pharmacology , Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Animals , Cell Adhesion Molecules/antagonists & inhibitors , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Diabetic Angiopathies/drug therapy , Diabetic Angiopathies/metabolism , Humans
9.
Biomolecules ; 11(11)2021 10 27.
Article in English | MEDLINE | ID: mdl-34827585

ABSTRACT

Glycosylation is an important post-translational modification that affects a wide variety of physiological functions. DC-SIGN (Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin) is a protein expressed in antigen-presenting cells that recognizes a variety of glycan epitopes. Until now, the binding of DC-SIGN to SARS-CoV-2 Spike glycoprotein has been reported in various articles and is regarded to be a factor in systemic infection and cytokine storm. The mechanism of DC-SIGN recognition offers an alternative method for discovering new medication for COVID-19 treatment. Here, we discovered three potential pockets that hold different glycan epitopes by performing molecular dynamics simulations of previously reported oligosaccharides. The "EPN" motif, "NDD" motif, and Glu354 form the most critical pocket, which is known as the Core site. We proposed that the type of glycan epitopes, rather than the precise amino acid sequence, determines the recognition. Furthermore, we deduced that oligosaccharides could occupy an additional site, which adds to their higher affinity than monosaccharides. Based on our findings and previously described glycoforms on the SARS-CoV-2 Spike, we predicted the potential glycan epitopes for DC-SIGN. It suggested that glycan epitopes could be recognized at multiple sites, not just Asn234, Asn149 and Asn343. Subsequently, we found that Saikosaponin A and Liquiritin, two plant glycosides, were promising DC-SIGN antagonists in silico.


Subject(s)
COVID-19/immunology , Cell Adhesion Molecules/antagonists & inhibitors , Epitopes/chemistry , Glycosides/chemistry , Lectins, C-Type/antagonists & inhibitors , Polysaccharides/chemistry , Receptors, Cell Surface/antagonists & inhibitors , Amino Acid Motifs , Binding Sites , COVID-19/metabolism , Computer Simulation , Cytokines/metabolism , Flavanones/chemistry , Glucosides/chemistry , Humans , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Monosaccharides/chemistry , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/chemistry , Saponins/chemistry , Spike Glycoprotein, Coronavirus/chemistry
10.
J Neuroinflammation ; 18(1): 233, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34654450

ABSTRACT

BACKGROUND: Changes in dopaminergic neural function can be induced by an acute inflammatory state that, by altering the integrity of the neurovasculature, induces neuronal stress, cell death and causes functional deficits. Effectively blocking these effects of inflammation could, therefore, reduce both neuronal and functional decline. To test this hypothesis, we inhibited vascular adhesion protein 1 (VAP-1), a membrane-bound protein expressed on the endothelial cell surface, that mediates leukocyte extravasation and induces oxidative stress. METHOD: We induced dopaminergic neuronal loss by infusing lipopolysaccharide (LPS) directly into the substantia nigra (SN) in rats and administered the VAP-1 inhibitor, PXS-4681A, daily. RESULTS: LPS produced: an acute inflammatory response, the loss of dopaminergic neurons in the SN, reduced the dopaminergic projection to SN target regions, particularly the dorsolateral striatum (DLS), and a deficit in habit learning, a key function of the DLS. In an attempt to protect SN neurons from this inflammatory response we found that VAP-1 inhibition not only reduced neutrophil infiltration in the SN and striatum, but also reduced the associated striatal microglia and astrocyte response. We found VAP-1 inhibition protected dopamine neurons in the SN, their projections to the striatum and promoted the functional recovery of habit learning. Thus, we reversed the loss of habitual actions, a function usually dependent on dopamine release in DLS and sensitive to striatal dysfunction. CONCLUSIONS: We establish, therefore, that VAP-1 inhibition has an anti-inflammatory profile that may be beneficial in the treatment of dopamine neuron dysfunction caused by an acute inflammatory state in the brain.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Adhesion Molecules/metabolism , Corpus Striatum/metabolism , Dopaminergic Neurons/metabolism , Habits , Learning/physiology , Allyl Compounds/pharmacology , Allyl Compounds/therapeutic use , Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cell Adhesion Molecules/antagonists & inhibitors , Corpus Striatum/drug effects , Dopaminergic Neurons/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Learning/drug effects , Male , Rats , Rats, Wistar , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
11.
Int J Oncol ; 59(5)2021 11.
Article in English | MEDLINE | ID: mdl-34664682

ABSTRACT

The Nectin cell adhesion molecule (Nectin) family members are Ca2+­independent immunoglobulin­like cellular adhesion molecules (including Nectins 1­4), involved in cell adhesion via homophilic/heterophilic interplay. In addition, the Nectin family plays a significant role in enhancing cellular viability and movement ability. In contrast to enrichment of Nectins 1­3 in normal tissues, Nectin­4 is particularly overexpressed in a number of tumor types, including breast, lung, urothelial, colorectal, pancreatic and ovarian cancer. Moreover, the upregulation of Nectin­4 is an independent biomarker for overall survival in numerous cancer types. A large number of studies have revealed that high expression of Nectin­4 is closely related to tumor occurrence and development in various cancer types, but the manner in which Nectin­4 protein contributes to the onset and development of these malignancies is yet unknown. The present review summarizes the molecular mechanisms and functions of Nectin­4 protein in the biological processes and current advances with regard to its expression and regulation in various cancer types.


Subject(s)
Cell Adhesion Molecules/physiology , Neoplasms/etiology , Antibodies, Monoclonal/pharmacology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/chemistry , Clinical Trials as Topic , Epithelial-Mesenchymal Transition , Humans , Neoplasms/therapy , Neovascularization, Pathologic/etiology , Oncolytic Virotherapy , Signal Transduction/physiology
12.
J Med Chem ; 64(19): 14332-14343, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34524803

ABSTRACT

In addition to a variety of viral-glycoprotein receptors (e.g., heparan sulfate, Niemann-Pick C1, etc.), dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN), from the C-type lectin receptor family, plays one of the most important pathogenic functions for a wide range of viruses (e.g., Ebola, human cytomegalovirus (HCMV), HIV-1, severe acute respiratory syndrome coronavirus 2, etc.) that invade host cells before replication; thus, its inhibition represents a relevant extracellular antiviral therapy. We report two novel p-tBu-calixarene glycoclusters 1 and 2, bearing tetrahydroxamic acid groups, which exhibit micromolar inhibition of soluble DC-SIGN binding and provide nanomolar IC50 inhibition of both DC-SIGN-dependent Jurkat cis-cell infection by viral particle pseudotyped with Ebola virus glycoprotein and the HCMV-gB-recombinant glycoprotein interaction with monocyte-derived dendritic cells expressing DC-SIGN. A unique cooperative involvement of sugar, linker, and calixarene core is likely behind the strong avidity of DC-SIGN for these low-valent systems. We claim herein new promising candidates for the rational development of a large spectrum of antiviral therapeutics.


Subject(s)
Calixarenes/chemistry , Cell Adhesion Molecules/antagonists & inhibitors , Glycoconjugates/metabolism , Glycoproteins/antagonists & inhibitors , Hydroxamic Acids/chemistry , Lectins, C-Type/antagonists & inhibitors , Phenols/chemistry , Receptors, Cell Surface/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Cell Adhesion Molecules/metabolism , Cell Line , Cytomegalovirus/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Ebolavirus/physiology , Glycoconjugates/chemistry , Glycoconjugates/pharmacology , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Jurkat Cells , Lectins, C-Type/metabolism , Models, Biological , Protein Binding , Receptors, Cell Surface/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Viral Proteins/genetics , Viral Proteins/metabolism
13.
Mol Pharm ; 18(9): 3464-3474, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34448393

ABSTRACT

Optimal cytoreduction for ovarian cancer is often challenging because of aggressive tumor biology and advanced stage. It is a critical issue since the extent of residual disease after surgery is the key predictor of ovarian cancer patient survival. For a limited number of cancers, fluorescence-guided surgery has emerged as an effective aid for tumor delineation and effective cytoreduction. The intravenously administered fluorescent agent, most commonly indocyanine green (ICG), accumulates preferentially in tumors, which are visualized under a fluorescent light source to aid surgery. Insufficient tumor specificity has limited the broad application of these agents in surgical oncology including for ovarian cancer. In this study, we developed a novel tumor-selective fluorescent agent by chemically linking ICG to mouse monoclonal antibody 10D7 that specifically recognizes an ovarian cancer-enriched cell surface receptor, CUB-domain-containing protein 1 (CDCP1). 10D7ICG has high affinity for purified recombinant CDCP1 and CDCP1 that is located on the surface of ovarian cancer cells in vitro and in vivo. Our results show that intravenously administered 10D7ICG accumulates preferentially in ovarian cancer, permitting visualization of xenograft tumors in mice. The data suggest CDCP1 as a rational target for tumor-specific fluorescence-guided surgery for ovarian cancer.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Cell Adhesion Molecules/antagonists & inhibitors , Fluorescent Dyes/administration & dosage , Optical Imaging/methods , Ovarian Neoplasms/diagnosis , Animals , Antibodies, Monoclonal/chemistry , Antigens, Neoplasm , Cell Line, Tumor , Female , Fluorescent Dyes/chemistry , Humans , Indocyanine Green/administration & dosage , Indocyanine Green/chemistry , Injections, Intravenous , Mice , Ovarian Neoplasms/pathology , Xenograft Model Antitumor Assays
14.
Drug Dev Res ; 82(8): 1096-1110, 2021 12.
Article in English | MEDLINE | ID: mdl-34462935

ABSTRACT

Trophoblast cell-surface antigen 2 (Trop 2) is a transmembrane glycoprotein that is highly expressed in various cancer types with relatively low or no baseline expression in most normal tissues. Its overexpression is associated with tumor growth and poor prognosis; Trop 2 is, therefore, an ideal therapeutic target for epithelial cancers. Several Trop 2 targeted therapeutics have recently been developed for the treatment of cancers, such as anti-Trop 2 antibodies and antibody-drug conjugates (ADCs), as well as Trop 2-specific cell therapy. In particular, the safety and clinical benefit of Trop 2-based ADCs have been demonstrated in clinical trials across multiple tumor types, including those with limited treatment options, such as triple-negative breast cancer, platinum-resistant urothelial cancer, and heavily pretreated non-small cell lung cancer. In this review, we elaborate on recent advances in Trop 2 targeted modalities and provide an overview of novel insights for future developments in this field.


Subject(s)
Cell Adhesion Molecules/antagonists & inhibitors , Neoplasms/drug therapy , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Antigens, Neoplasm/physiology , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/physiology , Humans , Immunoconjugates/therapeutic use , Immunotherapy, Adoptive
15.
ChemMedChem ; 16(15): 2345-2353, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34061468

ABSTRACT

The C-type lectin receptor DC-SIGN mediates interactions with envelope glycoproteins of many viruses such as SARS-CoV-2, ebola, and HIV and contributes to virus internalization and dissemination. In the context of the recent SARS-CoV-2 pandemic, involvement of DC-SIGN has been linked to severe cases of COVID-19. Inhibition of the interaction between DC-SIGN and viral glycoproteins has the potential to generate broad spectrum antiviral agents. Here, we demonstrate that mannose-functionalized poly-l-lysine glycoconjugates efficiently inhibit the attachment of viral glycoproteins to DC-SIGN-presenting cells with picomolar affinity. Treatment of these cells leads to prolonged receptor internalization and inhibition of virus binding for up to 6 h. Furthermore, the polymers are fully bio-compatible and readily cleared by target cells. The thermodynamic analysis of the multivalent interactions reveals enhanced enthalpy-driven affinities and promising perspectives for the future development of multivalent therapeutics.


Subject(s)
Antiviral Agents/pharmacology , Cell Adhesion Molecules/antagonists & inhibitors , Glycoconjugates/pharmacology , Lectins, C-Type/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Virus Attachment/drug effects , Antiviral Agents/chemical synthesis , Antiviral Agents/metabolism , Cell Adhesion Molecules/metabolism , Glycoconjugates/chemical synthesis , Glycoconjugates/metabolism , Humans , Lectins, C-Type/metabolism , Mannose/analogs & derivatives , Mannose/metabolism , Mannose/pharmacology , Microbial Sensitivity Tests , Polylysine/analogs & derivatives , Polylysine/metabolism , Polylysine/pharmacology , Protein Binding/drug effects , Receptors, Cell Surface/metabolism , SARS-CoV-2/drug effects , THP-1 Cells , Thermodynamics , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/metabolism
16.
Clin Cancer Res ; 27(16): 4511-4520, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34083232

ABSTRACT

PURPOSE: We investigated safety, tolerability, pharmacokinetics, and antitumor activity of the protein tyrosine kinase 7 (PTK7)-targeted, auristatin-based antibody-drug conjugate (ADC) PF-06647020/cofetuzumab pelidotin (NCT02222922). PATIENTS AND METHODS: Patients received PF-06647020 intravenously every 3 weeks at 0.2-3.7 mg/kg or every 2 weeks at 2.1-3.2 mg/kg, in sequential dose escalation, following a modified toxicity probability interval method. In dose expansion, pretreated patients with advanced, platinum-resistant ovarian cancer, non-small cell lung cancer (NSCLC), or triple-negative breast cancer (TNBC) received PF-06647020 2.8 mg/kg every 3 weeks. RESULTS: The most common, treatment-related adverse events for PF-06647020 administered every 3 weeks were nausea, alopecia, fatigue, headache, neutropenia, and vomiting (45%-25%); 25% of patients had grade ≥ 3 neutropenia. Two patients experienced dose-limiting toxicities (grade 3 headache and fatigue) at the highest every 3 weeks dose evaluated. The recommended phase II dose was 2.8 mg/kg every 3 weeks. The overall safety profile observed with PF-06647020 administered every 2 weeks was similar to that of the every 3 weeks regimen. Systemic exposure for the ADC and total antibody generally increased in a dose-proportional manner. Antitumor activity was observed in treated patients with overall objective response rates of 27% in ovarian cancer (n = 63), 19% in NSCLC (n = 31), and 21% in TNBC (n = 29). Responders tended to have moderate or high PTK7 tumor expression by IHC. CONCLUSIONS: This PTK7-targeted ADC demonstrated therapeutic activity in previously treated patients with ovarian cancer, NSCLC, and TNBC at a dose range of 2.1-3.2 mg/kg, supporting further clinical evaluation to refine dose, schedule, and predictive tissue biomarker testing in patients with advanced malignancies.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma, Ovarian Epithelial , Immunoconjugates , Lung Neoplasms , Ovarian Neoplasms , Triple Negative Breast Neoplasms , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/pathology , Cell Adhesion Molecules/antagonists & inhibitors , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Neoplasm Staging , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology
17.
Oncol Rep ; 46(1)2021 07.
Article in English | MEDLINE | ID: mdl-34013368

ABSTRACT

Trophoblast cell surface antigen 2 (TROP2), reported to be overexpressed in several types of cancer, is involved in cell proliferation, invasion, metastasis, and poor prognosis of many types of cancer. Previously, a highly sensitive anti­TROP2 monoclonal antibody (clone TrMab­6; mouse IgG2b, κ) was developed using a Cell­Based Immunization and Screening (CBIS) method. TrMab­6 was useful for investigations using flow cytometry, western blot, and immunohistochemistry. The aim of the present study was to investigate whether TrMab­6 possesses in vitro antibody­dependent cellular cytotoxicity (ADCC) or complement­dependent cytotoxicity (CDC) activities or in vivo antitumor activities using mouse xenograft models of TROP2­overexpressed CHO­K1 (CHO/TROP2) and breast cancer cell lines, including MCF7, MDA­MB­231, and MDA­MB­468. In vitro experiments revealed that TrMab­6 strongly induced ADCC and CDC activities against CHO/TROP2 and the three breast cancer cell lines, whereas it did not show those activities against parental CHO­K1 and MCF7/TROP2­knockout cells. Furthermore, in vivo experiments on CHO/TROP2 and MCF7 xenografts revealed that TrMab­6 significantly reduced tumor growth, whereas it did not show antitumor activities against parental CHO­K1 and MCF7/TROP2­knockout xenografts. The findings suggest that TrMab­6 is a promising treatment option for TROP2­expressing breast cancers.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibody-Dependent Cell Cytotoxicity/drug effects , Breast Neoplasms/drug therapy , Cell Adhesion Molecules/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CHO Cells , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cricetulus , Female , Gene Knockout Techniques , Humans , MCF-7 Cells , Mice , Treatment Outcome , Xenograft Model Antitumor Assays
18.
Front Immunol ; 12: 656452, 2021.
Article in English | MEDLINE | ID: mdl-34017333

ABSTRACT

Inflammatory bowel diseases (IBDs), including Crohn's disease (CD) and ulcerative colitis (UC) are multifactorial diseases with still unknown aetiology and an increasing prevalence and incidence worldwide. Despite plentiful therapeutic options for IBDs, the lack or loss of response in certain patients demands the development of further treatments to tackle this unmet medical need. In recent years, the success of the anti-α4ß7 antibody vedolizumab highlighted the potential of targeting the homing of immune cells, which is now an important pillar of IBD therapy. Due to its complexity, leukocyte trafficking and the involved molecules offer a largely untapped resource for a plethora of potential therapeutic interventions. In this review, we aim to summarise current and future directions of specifically interfering with immune cell trafficking. We will comment on concepts of homing, retention and recirculation and particularly focus on the role of tissue-derived chemokines. Moreover, we will give an overview of the mode of action of drugs currently in use or still in the pipeline, highlighting their mechanisms and potential to reduce disease burden.


Subject(s)
Cell Movement/immunology , Inflammatory Bowel Diseases/immunology , Animals , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Chemokines/antagonists & inhibitors , Chemokines/metabolism , Disease Susceptibility/immunology , Drug Development , Humans , Inflammatory Bowel Diseases/metabolism , Integrins/antagonists & inhibitors , Integrins/metabolism , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/metabolism , Molecular Targeted Therapy , Sphingosine-1-Phosphate Receptors/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
19.
Immunol Lett ; 235: 32-40, 2021 07.
Article in English | MEDLINE | ID: mdl-34000305

ABSTRACT

The junctional adhesion molecule-A (JAM-A) is an adhesion molecule present in the surface of several cell types, such as endothelial cells and leukocytes as well as Dendritic Cells (DC). Given the potential relevance of JAM-A in diverse pathological conditions such as inflammatory diseases and cancer, we investigated the role of JAM-A in CD4+ T cell priming. We demonstrate that JAM-A is present in the immunological synapse formed between T cells and DC during priming. Furthermore, an antagonistic anti-JAM-A mAb could disrupt the interaction between CD4+ T cell and DC. Antagonism of JAM-A also attenuated T cell activation and proliferation with a decrease in T-bet expression and increased IL-6 and IL-17 secretion. These findings demonstrate a functional role for JAM-A in interactions between CD4+ T cells and DCs during T cell priming as a positive regulator of Th1 differentiation.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Differentiation/immunology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Receptors, Cell Surface/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Autoimmunity , Biomarkers , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Adhesion/immunology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Communication , Coculture Techniques , Cytokines/biosynthesis , Disease Susceptibility , Humans , Immunological Synapses/metabolism , Immunophenotyping , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Th1 Cells/metabolism
20.
Front Immunol ; 12: 625808, 2021.
Article in English | MEDLINE | ID: mdl-33841409

ABSTRACT

B7 family members and their receptors play key roles in regulating T cell responses, and constitute very attractive targets for developing immunotherapeutic drugs. V-Set and Immunoglobulin domain containing 3 (VSIG3), a ligand for the novel B7 family immune checkpoint V-domain immunoglobulin suppressor of T cell activation (VISTA), can significantly inhibit T cell functions. Inhibitors targeting the VISTA/VSIG3 pathway are of great significance in tumor immunology. Here, we show the crystal structure of the extracellular domain (ECD) of the human VSIG3 protein at 2.64 angstrom resolution, and we produce recombinant human VSIG-3 ECD in both CHO cells and E. coli. Furthermore, we demonstrated the interaction of VISTA and VSIG3 by coimmunoprecipitation (Co-IP). Based on protein-protein docking for VISTA and VSIG3, we report a small molecule inhibitor of VSIG3 K284-3046 and evaluate its biological activities in vitro. This study was the first to reveal the crystal structure of VSIG3, and provides the structural basis for designing antibodies or compounds for the unique VSIG3/VISTA coinhibitory pathway in the treatment of cancers, autoimmune diseases and may be beneficial of designing vaccines.


Subject(s)
B7 Antigens/metabolism , Cell Adhesion Molecules/metabolism , Immunoglobulins/metabolism , Animals , B7 Antigens/chemistry , B7 Antigens/genetics , CHO Cells , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Proliferation/drug effects , Cricetulus , Crystallography, X-Ray , Cytokines/metabolism , Drug Design , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Immunoglobulins/chemistry , Immunoglobulins/genetics , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Ligands , Lymphocyte Activation/drug effects , Molecular Docking Simulation , Protein Binding , Protein Interaction Domains and Motifs , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...