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 ; 13(1): 1938796, 2021.
Article in English | MEDLINE | ID: mdl-34241561

ABSTRACT

New challenges and other topics in non-clinical safety testing of biotherapeutics were presented and discussed at the nineth European BioSafe Annual General Membership meeting in November 2019. The session topics were selected by European BioSafe organization committee members based on recent company achievements, agency interactions and new data obtained in the non-clinical safety testing of biotherapeutics, for which data sharing would be of interest and considered as valuable information. The presented session topics ranged from strategies of in vitro testing, immunogenicity prediction, bioimaging, and developmental and reproductive toxicology (DART) assessments to first-in-human (FIH) dose prediction and bioanalytical challenges, reflecting the entire space of different areas of expertise and different molecular modalities. During the 9th meeting of the European BioSafe members, the following topics were presented and discussed in 6 main sessions (with 3 or 4 presentations per session) and in three small group breakout sessions: 1) DART assessment with biotherapeutics: what did we learn and where to go?; 2) Non-animal testing strategies; 3) Seeing is believing: new frontiers in imaging; 4) Predicting immunogenicity during early drug development: hope or despair?; 5) Challenges in FIH dose projections; and 6) Non-canonical biologics formats: challenges in bioanalytics, PKPD and biotransformation for complex biologics formats. Small group breakout sessions were organized for team discussion about 3 specific topics: 1) Testing of cellular immune function in vitro and in vivo; 2) MABEL approach (toxicology and pharmacokinetic perspective); and 3) mRNA treatments. This workshop report presents the sessions and discussions at the meeting.


Subject(s)
Drug Evaluation, Preclinical/methods , Humans
2.
Clin Cancer Res ; 27(14): 4036-4053, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33771854

ABSTRACT

PURPOSE: CD40 agonists hold great promise for cancer immunotherapy (CIT) as they enhance dendritic cell (DC) activation and concomitant tumor-specific T-cell priming. However, the broad expression of CD40 accounts for sink and side effects, hampering the efficacy of anti-CD40 antibodies. We hypothesized that these limitations can be overcome by selectively targeting CD40 agonism to the tumor. Therefore, we developed a bispecific FAP-CD40 antibody, which induces CD40 stimulation solely in presence of fibroblast activation protein α (FAP), a protease specifically expressed in the tumor stroma. EXPERIMENTAL DESIGN: FAP-CD40's in vitro activity and FAP specificity were validated by antigen-presenting cell (APC) activation and T-cell priming assays. In addition, FAP-CD40 was tested in subcutaneous MC38-FAP and KPC-4662-huCEA murine tumor models. RESULTS: FAP-CD40 triggered a potent, strictly FAP-dependent CD40 stimulation in vitro. In vivo, FAP-CD40 strongly enhanced T-cell inflammation and growth inhibition of KPC-4662-huCEA tumors. Unlike nontargeted CD40 agonists, FAP-CD40 mediated complete regression of MC38-FAP tumors, entailing long-term protection. A high dose of FAP-CD40 was indispensable for these effects. While nontargeted CD40 agonists induced substantial side effects, highly dosed FAP-CD40 was well tolerated. FAP-CD40 preferentially accumulated in the tumor, inducing predominantly intratumoral immune activation, whereas nontargeted CD40 agonists displayed strong systemic but limited intratumoral effects. CONCLUSIONS: FAP-CD40 abrogates the systemic toxicity associated with nontargeted CD40 agonists. This enables administration of high doses, essential for overcoming CD40 sink effects and inducing antitumor immunity. Consequently, FAP-targeted CD40 agonism represents a promising strategy to exploit the full potential of CD40 signaling for CIT.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , CD40 Antigens/agonists , Endopeptidases/drug effects , Immunotherapy/methods , Membrane Proteins/drug effects , Neoplasms/drug therapy , Animals , Mice , Tumor Cells, Cultured
3.
J Immunol ; 194(8): 3646-55, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25769923

ABSTRACT

Mouse experimental autoimmune glomerulonephritis, a model of human antiglomerular basement membrane disease, depends on both Ab and T cell responses to the Goodpasture Ag noncollagenous domain 1 of the α3-chain of type IV collagen (α3IV-NC1). The aim of our study was to further characterize the T cell-mediated immune response. Repeated immunization with mouse α3IV-NC1 caused fatal glomerulonephritis in DBA/1 mice. Although two immunizations were sufficient to generate high α3IV-NC1-specific IgG titers, Ab and complement deposition along the glomerular basement membranes, and a nephrotic syndrome, two additional immunizations were needed to induce a necrotizing/crescentic glomerulonephritis. Ten days after the first immunization, α3IV-NC1-specific CD4(+) cells producing TNF-α, IFN-γ, or IL-17A were detected in the spleen. With the emergence of necrotizing/crescentic glomerulonephritis, ∼0.15% of renal CD4(+) cells were specific for α3IV-NC1. Using peptides spanning the whole α3IV-NC1 domain, three immunodominant T cell epitopes were identified. Immunization with these peptides did not lead to clinical signs of experimental autoimmune glomerulonephritis or necrotizing/crescentic glomerulonephritis. However, mice immunized with one of the peptides (STVKAGDLEKIISRC) developed circulating Abs against mouse α3IV-NC1 first detected at 8 wk, and 50% of the mice showed mild proteinuria at 18-24 wk due to membranous glomerulopathy. Taken together, our results suggest that autoreactive T cells are able to induce the formation of pathologic autoantibodies. The quality and quantity of α3IV-NC1-specific Ab and T cell responses are critical for the phenotype of the glomerulonephritis.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Collagen Type IV/metabolism , Glomerulonephritis, Membranous/immunology , Immunization , Peptides/immunology , Animals , Autoantibodies/immunology , Autoantigens/metabolism , Autoantigens/toxicity , CD4-Positive T-Lymphocytes/pathology , Collagen Type IV/toxicity , Cytokines/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/toxicity , Glomerulonephritis, Membranous/chemically induced , Glomerulonephritis, Membranous/pathology , Humans , Mice , Peptides/toxicity , Proteinuria/chemically induced , Proteinuria/immunology , Proteinuria/pathology , Spleen/immunology , Spleen/pathology
4.
Mol Oncol ; 8(2): 401-16, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24423492

ABSTRACT

An epithelial-mesenchymal transition (EMT) is a critical process during embryonic development and the progression of epithelial tumors to metastatic cancers. Gene expression profiling has uncovered the transcription factor LIM homeobox gene 2 (Lhx2) with up-regulated expression during TGFß-induced EMT in normal and cancerous breast epithelial cells. Loss and gain of function experiments in transgenic mouse models of breast cancer and of insulinoma in vivo and in breast cancer cells in vitro indicate that Lhx2 plays a critical role in primary tumor growth and metastasis. Notably, the transgenic expression of Lhx2 during breast carcinogenesis promotes vessel maturation, primary tumor growth, tumor cell intravasation and metastasis by directly inducing the expression of platelet-derived growth factor (PDGF)-B in tumor cells and by indirectly increasing the expression of PDGF receptor-ß (PDGFRß) on tumor cells and pericytes. Pharmacological inhibition of PDGF-B/PDGFRß signaling reduces vessel functionality and tumor growth and Lhx2-induced cell migration and cell invasion. The data indicate a dual role of Lhx2 during EMT and tumor progression: by inducing the expression of PDGF-B, Lhx2 provokes an autocrine PDGF-B/PDGFRß loop required for cell migration, invasion and metastatic dissemination and paracrine PDGF-B/PDGFRß signaling to support blood vessel functionality and, thus, primary tumor growth.


Subject(s)
Autocrine Communication , Breast Neoplasms/metabolism , LIM-Homeodomain Proteins/metabolism , Mammary Neoplasms, Experimental/metabolism , Paracrine Communication , Proto-Oncogene Proteins c-sis/biosynthesis , Signal Transduction , Transcription Factors/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Humans , LIM-Homeodomain Proteins/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Proto-Oncogene Proteins c-sis/genetics , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/genetics , Transcription Factors/genetics
5.
Cancer Res ; 74(5): 1566-75, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24413534

ABSTRACT

An epithelial-mesenchymal transition (EMT) underlies malignant tumor progression and metastatic spread by enabling cancer cells to depart from the primary tumor, invade surrounding tissue, and disseminate to distant organs. EMT also enriches for cancer stem cells (CSC) and increases the capacity of cancer cells to initiate and propagate tumors upon transplantation into immune-deficient mice, a major hallmark of CSCs. However, the molecular mechanisms promoting the tumorigenicity of cancer cells undergoing an EMT and of CSCs have remained widely elusive. We here report that EMT confers efficient tumorigenicity to murine breast cancer cells by the upregulated expression of the proangiogenic factor VEGF-A and by increased tumor angiogenesis. On the basis of these data, we propose a novel interpretation of the features of CSCs with EMT-induced, VEGF-A-mediated angiogenesis as the connecting mechanism between cancer cell stemness and tumor initiation.


Subject(s)
Carcinogenesis/pathology , Epithelial-Mesenchymal Transition/genetics , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Neoplastic Stem Cells/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism
6.
J Am Soc Nephrol ; 22(4): 649-63, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21372207

ABSTRACT

Mesangial cells in diabetic mice and human kidneys with diabetic nephropathy exhibit increased type VIII collagen, a nonfibrillar protein that exists as a heterodimer composed of α1(VIII) and α2(VIII), encoded by Col8a1 and Col8a2, respectively. Because TGF-ß1 promotes the development of diabetic glomerulosclerosis, we studied whether type VIII collagen modulates the effects of TGF-ß1 in mesangial cells. We obtained primary cultures of mesangial cells from wild-type, doubly heterozygous (Col8a1(+/-)/Col8a2(+/-)), and double-knockout (Col8a1(-/-)/Col8a2(-/-)) mice. TGF-ß1 bound normally to double-knockout mesangial cells. In wild-type mesangial cells, TGF-ß1 inhibited proliferation, but in double-knockout cells, it stimulated proliferation, promoted cell cycle progression, and reduced apoptosis; we could reverse this effect by reconstituting α1(VIII). Furthermore, in wild-type cells, TGF-ß1 mainly stimulated the Smad pathways, whereas in double-knockout cells, it activated the MAPK and PI3K/Akt pathways and induced expression of fibroblast growth factor 21 (FGF21). Inhibiting FGF21 expression by either interfering with activation of the MAPK and PI3K/Akt pathways or by FGF21 siRNA attenuated the TGF-ß1-induced proliferation of double-knockout mesangial cells. In vivo, diabetic double-knockout mice had significantly higher expression of renal FGF21 mRNA and protein compared with diabetic wild-type mice. Immunohistochemistry revealed strong expression of FGF21 in both glomerular (mesangial) and tubular cells of diabetic mice. Taken together, these data suggest that type VIII collagen significantly modulates the effect of TGF-ß1 on mesangial cells and may therefore play a role in the pathogenesis of diabetic nephropathy.


Subject(s)
Cell Proliferation/drug effects , Collagen Type VIII/metabolism , Mesangial Cells/metabolism , Mesangial Cells/pathology , Transforming Growth Factor beta1/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Cycle/drug effects , Cell Cycle/physiology , Cells, Cultured , Collagen Type VIII/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Disease Models, Animal , Fibroblast Growth Factors/metabolism , Mesangial Cells/drug effects , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Streptozocin , Transforming Growth Factor beta1/physiology
7.
Diabetes ; 58(7): 1672-81, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19401424

ABSTRACT

OBJECTIVE: Key features of diabetic nephropathy include the accumulation of extracellular matrix proteins. In recent studies, increased expression of type VIII collagen in the glomeruli and tubulointerstitium of diabetic kidneys has been noted. The objectives of this study were to assess whether type VIII collagen affects the development of diabetic nephropathy and to determine type VIII collagen-dependent pathways in diabetic nephropathy in the mouse model of streptozotocin (STZ)-induced diabetes. RESEARCH DESIGN AND METHODS: Diabetes was induced by STZ injections in collagen VIII-deficient or wild-type mice. Functional and histological analyses were performed 40 days after induction of diabetes. Type VIII collagen expression was assessed by Northern blots, immunohistochemistry, and real-time PCR. Proliferation of primary mesangial cells was measured by thymidine incorporation and direct cell counting. Expression of phosphorylated extracellular signal-regulated kinase (ERK1/2) and p27(Kip1) was assessed by Western blots. Finally, Col8a1 was stably overexpressed in mesangial cells. RESULTS: Diabetic wild-type mice showed a strong renal induction of type VIII collagen. Diabetic Col8a1(-)/Col8a2(-) animals revealed reduced mesangial expansion and cellularity and extracellular matrix expansion compared with the wild type. These were associated with less albuminuria. High-glucose medium as well as various cytokines induced Col8a1 in cultured mesangial cells. Col8a1(-)/Col8a2(-) mesangial cells revealed decreased proliferation, less phosphorylation of Erk1/2, and increased p27(Kip1) expression. Overexpression of Col8a1 in mesangial cells induced proliferation. CONCLUSIONS: Lack of type VIII collagen confers renoprotection in diabetic nephropathy. One possible mechanism is that type VIII collagen permits and/or fosters mesangial cell proliferation in early diabetic nephropathy.


Subject(s)
Collagen Type VIII/deficiency , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/prevention & control , Glomerular Mesangium/pathology , Animals , Cell Division , Collagen Type VIII/genetics , DNA Primers , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Male , Mice , Mice, Knockout , Polymerase Chain Reaction , RNA/genetics
8.
J Biol Chem ; 281(13): 8645-55, 2006 Mar 31.
Article in English | MEDLINE | ID: mdl-16407229

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1), a transcriptional complex composed of an oxygen-sensitive alpha- and a beta-subunit, plays a pivotal role in cellular adaptation to low oxygen availability. Under normoxia, the alpha-subunit of HIF-1 is hydroxylated by a family of prolyl hydroxylases (PHDs) and consequently targeted for proteasomal degradation. Three different PHDs have been identified, but the difference among their in vivo roles remain unclear. PHD3 is strikingly expressed by hypoxia, displays high substrate specificity, and has been identified in other signaling pathways. PHD3 may therefore hydroxylate divergent substrates and/or connect divergent cellular responses with HIF. We identified a novel WD-repeat protein, recently designated Morg1 (MAPK organizer 1), by screening a cDNA library with yeast two-hybrid assays. The interaction between PHD3 and Morg1 was confirmed in vitro and in vivo. We found seven WD-repeat domains by cloning the full-length cDNA of Morg1. By confocal microscopy both proteins co-localize within the cytoplasm and the nucleus and display a similar tissue expression pattern in Northern blots. Binding occurs at a conserved region predicted to the top surface of one propeller blade. Finally, HIF-mediated reporter gene activity is decreased by Morg1 and reduced to basal levels when Morg1 is co-expressed with PHD3. Suppression of Morg1 or PHD3 by stealth RNA leads to a marked increase of HIF-1 activity. These results indicate that Morg1 specifically interacts with PHD3 most likely by acting as a molecular scaffold. This interaction may provide a molecular framework between HIF regulation and other signaling pathways.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Hypoxia-Inducible Factor 1/metabolism , Procollagen-Proline Dioxygenase/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Amino Acid Sequence , Animals , Base Pairing , Base Sequence , Binding Sites , Blotting, Northern , Carrier Proteins/biosynthesis , Cell Hypoxia , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA, Complementary , Enzyme-Linked Immunosorbent Assay , Genes, Reporter , Hypoxia-Inducible Factor 1/genetics , Luciferases/metabolism , Microscopy, Confocal , Molecular Sequence Data , Molecular Weight , PC12 Cells , Precipitin Tests , Procollagen-Proline Dioxygenase/genetics , Protein Binding , Protein Structure, Tertiary , Rats , Transcription, Genetic , Trypsin/pharmacology , Two-Hybrid System Techniques
9.
Arterioscler Thromb Vasc Biol ; 26(1): 56-61, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16269661

ABSTRACT

OBJECTIVE: Type VIII collagen is upregulated after vascular injury and in atherosclerosis. However, the role of type VIII collagen endogenously expressed by smooth muscle cells (SMCs) and in the context of the vascular matrix microenvironment, which is rich in type I collagen, is not known. To address this, we have compared aortic SMCs from wild-type (WT) mice to SMCs from type VIII collagen-deficient (KO) mice when plated on type I collagen. METHODS AND RESULTS: Type VIII collagen was upregulated after wounding of WT SMCs. KO SMCs exhibited greater adhesion to type I collagen than WT SMCs (optical density [OD595]=0.458+/-0.044 versus 0.193+/-0.071). By contrast, the WT SMCs spread more (389+/-75% versus 108+/-14% increase in cell area), migrated further (total distance 80.6+/-6.2 microm versus 64.2+/-4.4 microm), and exhibited increased [3H]-thymidine uptake (160,000+/-22,300 versus 63,100+/-12,100 counts per minute) when compared with KO SMCs. Gelatin zymograms showed that WT SMCs expressed latent matrix metalloproteinase 2, whereas KO SMCs did not. Addition of exogenous type VIII collagen returned levels of KO SMC adhesion (OD595=0.316+/-0.038), migration (79.5+/-5.8 microm), and latent matrix metalloproteinase 2 expression to levels comparable to WT SMCs. CONCLUSIONS: This study suggests that SMCs can modify the matrix microenvironment by producing type VIII collagen, using it to overlay type I collagen, and generating a substrate favorable for migration.


Subject(s)
Cell Movement/physiology , Collagen Type VIII/genetics , Collagen Type VIII/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Alleles , Animals , Aorta/cytology , Cell Division/physiology , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Extracellular Matrix/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Microscopy, Video , Up-Regulation/physiology
10.
FASEB J ; 19(10): 1232-44, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16051690

ABSTRACT

Collagen VIII is localized in subendothelial and subepithelial extracellular matrices. It is a major component of Descemet's membrane, a thick basement membrane under the corneal endothelium, where it forms a hexagonal lattice structure; a similar structure, albeit less extensive, may be formed in other basement membranes. We have examined the function of collagen VIII in mice by targeted inactivation of the genes encoding the two polypeptide subunits, Col8a1 and Col8a2. Analysis of these mice reveals no major structural defects in most organs, but demonstrates that type VIII collagen is required for normal anterior eye development, particularly the formation of a corneal stroma with the appropriate number of fibroblastic cell layers and Descemet's membrane of appropriate thickness. Complete lack of type VIII collagen leads to dysgenesis of the anterior segment of the eye: a globoid, keratoglobus-like protrusion of the anterior chamber with a thin corneal stroma. Descemet's membrane is markedly thinned. The corneal endothelial cells are enlarged and reduced in number, and show a decreased ability to proliferate in response to different growth factors in vitro. An important function of collagen VIII may therefore be to generate a peri- or subcellular matrix environment that permits or stimulates cell proliferation.


Subject(s)
Anterior Eye Segment/abnormalities , Collagen Type VIII/physiology , Animals , Anterior Eye Segment/embryology , Aorta/metabolism , Cell Proliferation , Collagen Type VIII/genetics , Cornea/ultrastructure , Endothelium, Corneal/pathology , Eye Abnormalities/etiology , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...