Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
MAbs ; 16(1): 2362432, 2024.
Article in English | MEDLINE | ID: mdl-38849989

ABSTRACT

In contrast to natural antibodies that rely mainly on the heavy chain to establish contacts with their cognate antigen, we have developed a bispecific antibody format in which the light chain (LC) drives antigen binding and specificity. To better understand epitope-paratope interactions in this context, we determined the X-ray crystallographic structures of an antigen binding fragment (Fab) in complex with human CD47 and another Fab in complex with human PD-L1. These Fabs contain a κ-LC and a λ-LC, respectively, which are paired with an identical heavy chain (HC). The structural analysis of these complexes revealed the dominant contribution of the LCs to antigen binding, but also that the common HC provides some contacts in both CD47 and PD-L1 Fab complexes. The anti-CD47 Fab was affinity optimized by diversifying complementary-determining regions of the LC followed by phage display selections. Using homology modeling, the contributions of the amino acid modification to the affinity increase were analyzed. Our results demonstrate that, despite a less prominent role in natural antibodies, the LC can mediate high affinity binding to different antigens and neutralize their biological function. Importantly, Fabs containing a common variable heavy (VH) domain enable the generation of bispecific antibodies retaining a truly native structure, maximizing their therapeutic potential.


Subject(s)
Antibodies, Bispecific , B7-H1 Antigen , CD47 Antigen , Immunoglobulin Fab Fragments , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/immunology , Humans , CD47 Antigen/immunology , CD47 Antigen/chemistry , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , B7-H1 Antigen/immunology , B7-H1 Antigen/chemistry , B7-H1 Antigen/antagonists & inhibitors , Crystallography, X-Ray , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/immunology , Models, Molecular
2.
MAbs ; 12(1): 1739408, 2020.
Article in English | MEDLINE | ID: mdl-32191151

ABSTRACT

Mesothelin (MSLN) is a cell surface glycoprotein overexpressed in several solid malignancies, including gastric, lung, mesothelioma, pancreatic and ovarian cancers. While several MSLN-targeting therapeutic approaches are in development, only limited efficacy has been achieved in patients. A potential shortcoming of several described antibody-based approaches is that they target the membrane distal region of MSLN and, additionally, are known to be handicapped by the high levels of circulating soluble MSLN in patients. We show here, using monoclonal antibodies (mAbs) targeting different MSLN-spanning epitopes, that the membrane-proximal region resulted in more efficient killing of MSLN-positive tumor cells in antibody-dependent cell-mediated cytotoxicity (ADCC) assays. Surprisingly, no augmented killing was observed in antibody-dependent cellular phagocytosis (ADCP) by mAbs targeting this membrane-proximal region. To further increase the ADCP potential, we, therefore, generated bispecific antibodies (bsAbs) coupling a high-affinity MSLN binding arm to a blocking CD47 arm. Here, targeting the membrane-proximal domain of MSLN demonstrated enhanced ADCP activity compared to membrane-distal domains when the bsAbs were used in in vitro phagocytosis killing assays. Importantly, the superior anti-tumor activity was also translated in xenograft tumor models. Furthermore, we show that the bsAb approach targeting the membrane-proximal epitope of MSLN optimized ADCC activity by augmenting FcγR-IIIA activation and enhanced ADCP via a more efficient blockade of the CD47/SIRPα axis.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antineoplastic Agents/immunology , CD47 Antigen/immunology , GPI-Linked Proteins/immunology , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/pharmacology , Epitopes/immunology , Humans , Immunotherapy/methods , Mesothelin , Mice , Phagocytosis/immunology , Xenograft Model Antitumor Assays
3.
Sci Signal ; 11(561)2018 12 18.
Article in English | MEDLINE | ID: mdl-30563864

ABSTRACT

Irritable bowel syndrome (IBS) is a common gastrointestinal disorder that is characterized by chronic abdominal pain concurrent with altered bowel habit. Polyunsaturated fatty acid (PUFA) metabolites are increased in abundance in IBS and are implicated in the alteration of sensation to mechanical stimuli, which is defined as visceral hypersensitivity. We sought to quantify PUFA metabolites in patients with IBS and evaluate their role in pain. Quantification of PUFA metabolites by mass spectrometry in colonic biopsies showed an increased abundance of 5-oxoeicosatetraenoic acid (5-oxoETE) only in biopsies taken from patients with IBS with predominant constipation (IBS-C). Local administration of 5-oxoETE to mice induced somatic and visceral hypersensitivity to mechanical stimuli without causing tissue inflammation. We found that 5-oxoETE directly acted on both human and mouse sensory neurons as shown by lumbar splanchnic nerve recordings and Ca2+ imaging of dorsal root ganglion (DRG) neurons. We showed that 5-oxoETE selectively stimulated nonpeptidergic, isolectin B4 (IB4)-positive DRG neurons through a phospholipase C (PLC)- and pertussis toxin-dependent mechanism, suggesting that the effect was mediated by a G protein-coupled receptor (GPCR). The MAS-related GPCR D (Mrgprd) was found in mouse colonic DRG afferents and was identified as being implicated in the noxious effects of 5-oxoETE. Together, these data suggest that 5-oxoETE, a potential biomarker of IBS-C, induces somatic and visceral hyperalgesia without inflammation in an Mrgprd-dependent manner. Thus, 5-oxoETE may play a pivotal role in the abdominal pain associated with IBS-C.


Subject(s)
Arachidonic Acids/metabolism , Irritable Bowel Syndrome/pathology , Nociception , Receptors, G-Protein-Coupled/physiology , Sensory Receptor Cells/pathology , Animals , Calcium/metabolism , Case-Control Studies , Colon/drug effects , Colon/metabolism , Colon/pathology , Constipation/chemically induced , Constipation/physiopathology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Humans , Irritable Bowel Syndrome/etiology , Irritable Bowel Syndrome/metabolism , Male , Mice , Mice, Inbred C57BL , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Signal Transduction
4.
Pain ; 159(7): 1257-1267, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29554016

ABSTRACT

Proteases and protease-activated receptors (PARs) are major mediators involved in irritable bowel syndrome (IBS). Our objectives were to decipher the expression and functionality (calcium signaling) of PARs in human dorsal root ganglia (DRG) neurons and to define mechanisms involved in human sensory neuron signaling by IBS patient mediators. Human thoracic DRG were obtained from the national disease resource interchange. Expression of PAR1, PAR2, and PAR4 was assessed by immunohistochemistry and quantitative reverse transcription PCR (RT-qPCR) in whole DRG or in primary cultures of isolated neurons. Calcium signaling in response to PAR agonist peptides (PAR-AP), their inactive peptides (PAR-IP), thrombin (10 U/mL), supernatants from colonic biopsies of patients with IBS, or healthy controls, with or without PAR1 or PAR4 antagonist were studied in cultured human DRG neurons. PAR1, PAR2, and PAR4 were all expressed in human DRG, respectively, in 20%, 40%, and 40% of the sensory neurons. PAR1-AP increased intracellular calcium concentration in a dose-dependent manner. This increase was inhibited by PAR1 antagonism. By contrast, PAR2-AP, PAR4-AP, and PAR-IP did not cause calcium mobilization. PAR1-AP-induced calcium flux was significantly reduced by preincubation with PAR4-AP, but not with PAR2-AP. Thrombin increased calcium flux, which was inhibited by a PAR1 antagonist and increased by a PAR4 antagonist. Supernatants from colonic biopsies of patients with IBS induced calcium flux in human sensory neurons compared with healthy controls, and this induction was reversed by a PAR1 antagonist. Taken together, our results highlight that PAR1 antagonism should be investigated as a new therapeutic target for IBS symptoms.


Subject(s)
Ganglia, Spinal/metabolism , Irritable Bowel Syndrome/metabolism , Receptor, PAR-1/metabolism , Sensory Receptor Cells/metabolism , Signal Transduction/physiology , Thorax/innervation , Calcium Signaling , Colon/metabolism , Humans , Visceral Pain/metabolism
5.
Mol Metab ; 10: 100-108, 2018 04.
Article in English | MEDLINE | ID: mdl-29428595

ABSTRACT

OBJECTIVE: Decreasing duodenal contraction is now considered as a major focus for the treatment of type 2 diabetes. Therefore, identifying bioactive molecules able to target the enteric nervous system, which controls the motility of intestinal smooth muscle cells, represents a new therapeutic avenue. For this reason, we chose to study the impact of oral galanin on this system in diabetic mice. METHODS: Enteric neurotransmission, duodenal contraction, glucose absorption, modification of gut-brain axis, and glucose metabolism (glucose tolerance, insulinemia, glucose entry in tissue, hepatic glucose metabolism) were assessed. RESULTS: We show that galanin, a neuropeptide expressed in the small intestine, decreases duodenal contraction by stimulating nitric oxide release from enteric neurons. This is associated with modification of hypothalamic nitric oxide release that favors glucose uptake in metabolic tissues such as skeletal muscle, liver, and adipose tissue. Oral chronic gavage with galanin in diabetic mice increases insulin sensitivity, which is associated with an improvement of several metabolic parameters such as glucose tolerance, fasting blood glucose, and insulin. CONCLUSION: Here, we demonstrate that oral galanin administration improves glucose homeostasis via the enteric nervous system and could be considered a therapeutic potential for the treatment of T2D.


Subject(s)
Blood Glucose/metabolism , Enteric Nervous System/drug effects , Galanin/pharmacology , Hypoglycemic Agents/pharmacology , Neurons/drug effects , Administration, Oral , Animals , Enteric Nervous System/metabolism , Galanin/administration & dosage , Hypoglycemic Agents/administration & dosage , Hypothalamus/metabolism , Insulin/blood , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism
6.
Sci Rep ; 6: 31849, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27549402

ABSTRACT

Apelin is a bioactive peptide involved in the control of energy metabolism. In the hypothalamus, chronic exposure to high levels of apelin is associated with an increase in hepatic glucose production, and then contributes to the onset of type 2 diabetes. However, the molecular mechanisms behind deleterious effects of chronic apelin in the brain and consequences on energy expenditure and thermogenesis are currently unknown. We aimed to evaluate the effects of chronic intracerebroventricular (icv) infusion of apelin in normal mice on hypothalamic inflammatory gene expression, energy expenditure, thermogenesis and brown adipose tissue functions. We have shown that chronic icv infusion of apelin increases the expression of pro-inflammatory factors in the hypothalamus associated with an increase in plasma interleukin-1 beta. In parallel, mice infused with icv apelin exhibit a significant lower energy expenditure coupled to a decrease in PGC1alpha, PRDM16 and UCP1 expression in brown adipose tissue which could explain the alteration of thermogenesis in these mice. These data provide compelling evidence that central apelin contributes to the development of type 2 diabetes by altering energy expenditure, thermogenesis and fat browning.


Subject(s)
Adipose Tissue, Brown/drug effects , Apelin/pharmacology , Energy Metabolism/drug effects , Thermogenesis/drug effects , Adipose Tissue, Brown/metabolism , Animals , Apelin/administration & dosage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Expression/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Infusions, Intraventricular , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
7.
Gastroenterology ; 149(2): 433-44.e7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25911511

ABSTRACT

BACKGROUND & AIMS: In mice, activation of the transient receptor potential cation channels (TRP) TRPV1, TRPV4, and TRPA1 causes visceral hypersensitivity. These receptors and their agonists might be involved in development of irritable bowel syndrome (IBS). We investigated whether polyunsaturated fatty acid (PUFA) metabolites, which activate TRPs, are present in colon tissues from patients with IBS and act as endogenous agonists to induce hypersensitivity. METHODS: We analyzed colon biopsy samples from 40 patients with IBS (IBS biopsies) and 11 healthy individuals undergoing colorectal cancer screening (controls), collected during colonoscopy at the University of Bologna, Italy. Levels of the PUFA metabolites that activate TRPV1 (12-hydroperoxyeicosatetraenoic acid, 15-hydroxyeicosatetraenoic acid, 5-hydroxyeicosatetraenoic acid, and leukotriene B4), TRPV4 (5,6-epoxyeicosatrienoic acid [EET] and 8,9-EET), and TRPA1 (PGA1, 8-iso-prostaglandin A2, and 15-deoxy-Δ-prostaglandin J2) were measured in biopsies and their supernatants using liquid chromatography and tandem mass spectrometry; we also measured levels of the PUFA metabolites prostaglandin E2 (PGE2) and resolvins. C57Bl6 mice were given intrathecal injections of small interfering RNAs to reduce levels of TRPV4, or control small interfering RNAs, along with colonic injections of biopsy supernatants; visceral hypersensitivity was measured based on response to colorectal distension. Mouse sensory neurons were cultured and incubated with biopsy supernatants and lipids extracted from biopsies or colons of mice. Immunohistochemistry was used to detect TRPV4 in human dorsal root ganglia samples (from the National Disease Research Interchange). RESULTS: Levels of the TRPV4 agonist 5,6-EET, but not levels of TRPV1 or TRPA1 agonists, were increased in IBS biopsies compared with controls; increases correlated with pain and bloating scores. Supernatants from IBS biopsies, but not from controls, induced visceral hypersensitivity in mice. Small interfering RNA knockdown of TRPV4 in mouse primary afferent neurons inhibited the hypersensitivity caused by supernatants from IBS biopsies. Levels of 5,6-EET and 15-HETE were increased in colons of mice with, but not without, visceral hypersensitivity. PUFA metabolites extracted from IBS biopsies or colons of mice with visceral hypersensitivity activated mouse sensory neurons in vitro, by activating TRPV4. Mouse sensory neurons exposed to supernatants from IBS biopsies produced 5,6-EET via a mechanism that involved the proteinase-activated receptor-2 and cytochrome epoxygenase. In human dorsal root ganglia, TPV4 was expressed by 35% of neurons. CONCLUSIONS: Colon tissues from patients with IBS have increased levels of specific PUFA metabolites. These stimulate sensory neurons from mice and generate visceral hypersensitivity via activation of TRPV4.


Subject(s)
Calcium Channels/metabolism , Colon/metabolism , Fatty Acids, Unsaturated/metabolism , Irritable Bowel Syndrome/metabolism , Nerve Tissue Proteins/metabolism , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Adult , Aged , Animals , Biopsy , Chromatography, Liquid , Colon/cytology , Colon/innervation , Dinoprostone/metabolism , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Humans , Italy , Male , Mice , Mice, Inbred C57BL , Middle Aged , RNA, Small Interfering/genetics , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Signal Transduction , TRPA1 Cation Channel , Tandem Mass Spectrometry , Young Adult
8.
Eur J Pharm Biopharm ; 81(2): 379-85, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22386911

ABSTRACT

Topical delivery of 5-aminosalicylic acid (5-ASA) to the colonic mucosa is important in order to achieve effective drug concentration in the site of inflammation and to minimize its systemic availability. 5-ASA loaded pellets were prepared by an extrusion/spheronization method. Mucoadhesive biopolymer chitosan was incorporated into the pellets, and drug delivery to the colon was controlled by the pH-sensitive polymer Eudragit® FS. Dissolution profiles of coated pellets revealed no drug release at pH 1.2 within 2h and release as intended in the simulated distal ileum and colon. In vivo, chitosan-core drug loaded pellets (AMCh) showed 2.5-fold higher drug metabolite concentration than after chitosan free pellets (AM) administration in the inflamed colonic tissue. Additionally, AMCh demonstrated decreased in AUC in colitis group (1507 ± 400 ng h/ml) compared with AM (1907 ± 122 ng h/ml). In terms of therapeutic efficiency, administration of pellets markedly decreased the colon/body weight ratio (colitis: 0.0355 ± 0.0028; AM 0.0092 ± 0.0033; AMCh 0.0086 ± 0.0022) and myeloperoxidase activity (colitis: 3212 ± 294 U/g tissue; AM 796 ± 211 U/g; AMCh 552 ± 319 U/g). Bioadhesive chitosan pellets showed additional beneficial properties for colonic 5-ASA delivery in the treatment of inflammatory bowel disease by increasing the drug concentration locally.


Subject(s)
Chitosan/administration & dosage , Chitosan/chemistry , Colitis/metabolism , Mesalamine/administration & dosage , Mesalamine/chemistry , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Biopolymers/administration & dosage , Biopolymers/chemistry , Biopolymers/pharmacokinetics , Chitosan/pharmacokinetics , Colitis/drug therapy , Colon/drug effects , Colon/metabolism , Drug Delivery Systems/methods , Drug Implants/administration & dosage , Drug Implants/chemistry , Drug Implants/pharmacokinetics , Hydrogen-Ion Concentration , Inflammation/drug therapy , Inflammation/metabolism , Male , Mesalamine/pharmacokinetics , Particle Size , Peroxidase/metabolism , Rats , Rats, Wistar , Solubility
9.
Int J Pharm ; 422(1-2): 151-9, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22079717

ABSTRACT

Preparation of coated pellets intended for rutin colon delivery, their evaluation in vitro and in vivo in experimental colitis in rats was the purpose of this study. Pellets were obtained using extrusion/spheronization and coated with three types of coatings (caffeic acid/hypromellose/alginic acid; sodium alginate/hypromellose/zinc acetate; sodium alginate/chitosan). Dissolution using buffers of pH values, ß-glucosidase and times corresponding to gastrointestinal tract (GIT) was provided. Pellets coated with alginate/chitosan showed low rutin dissolution (12-14%) in upper GIT conditions and fast release (87-89%) under colon conditions; that is a good presumption of intended rutin release. After colitis induction and development, the rats were treated with pellets and rutin solution administered orally, solution also rectally. Colon/body weight ratio, myeloperoxidase activity and histological evaluation were performed. Rutin was able to promote colonic healing at the dose of 10mg/kg: colon/body weight ratio decreased and myeloperoxidase activity was significantly suppressed. Pellets coated with alginate/chitosan applied orally and rutin solution administered rectally showed the best efficacy. The combination of rutin as natural product, mucoadhesive chitosan degraded in the colon and sodium alginate as the main coating substance in the form of pellets create a promising preparation for therapy of this severe illness.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chitosan/chemistry , Colitis/drug therapy , Colon/drug effects , Gastrointestinal Agents/pharmacology , Rutin/pharmacology , Administration, Oral , Alginates/chemistry , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Buffers , Caffeic Acids/chemistry , Chemistry, Pharmaceutical , Colitis/chemically induced , Colitis/pathology , Colon/pathology , Disease Models, Animal , Drug Compounding , Drug Implants , Drug Stability , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/chemistry , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Hydrogen-Ion Concentration , Hypromellose Derivatives , Male , Methylcellulose/analogs & derivatives , Methylcellulose/chemistry , Rats , Rats, Wistar , Rutin/administration & dosage , Rutin/chemistry , Solubility , Technology, Pharmaceutical/methods , Time Factors , Trinitrobenzenesulfonic Acid , Zinc Acetate/chemistry
10.
Drug Dev Ind Pharm ; 37(9): 1100-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21417610

ABSTRACT

BACKGROUND: In recent years, many achievements have been realized in the therapy of inflammatory bowel disease (IBD) although its etiology remains unknown. Thus IBD treatment is symptomatic and targets general inflammatory mechanisms. Oral formulations containing 5-aminosalicylic acid (5-ASA) have become the standard therapy for mild-to-moderate IBD. OBJECTIVE: This article is a review of recently published research dealing with new 5-ASA dosage forms. Thus promising candidates for IBD treatment evaluated in vitro are reported; systems tested in vivo in trinitrobenzene sulfonic acid (TNBS)-induced colitis in rats are mentioned; and 5-ASA formulations used in clinical studies are presented. Moreover, all oral dosage forms containing 5-ASA or its prodrugs are reviewed; their characteristics and utilization in IBD treatment are discussed. CONCLUSION: In several clinical studies, it has been shown that multiparticulates such as pellets offer more advantages as compared with single unit forms, that is, coated tablets. Prolonged presence close to the site of the action, improved drug bioavailability, and easier administration of large drug doses belong to the benefits of pellets.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Inflammatory Bowel Diseases/drug therapy , Mesalamine/administration & dosage , Animals , Clinical Trials as Topic , Drug Compounding/methods , Drug Delivery Systems/methods , Humans , Inflammatory Bowel Diseases/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...