Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Exp Clin Cancer Res ; 38(1): 332, 2019 Jul 30.
Article in English | MEDLINE | ID: mdl-31362764

ABSTRACT

BACKGROUND: Immunotherapeutic approaches designed to augment T and B cell mediated killing of tumor cells has met with clinical success in recent years suggesting tremendous potential for treatment in a broad spectrum of tumor types. After complex recognition of target cells by T and B cells, delivery of the serine protease granzyme B (GrB) to tumor cells comprises the cytotoxic insult resulting in a well-characterized, multimodal apoptotic cascade. METHODS: We designed a recombinant fusion construct, GrB-Fc-4D5, composed of a humanized anti-HER2 scFv fused to active GrB for recognition of tumor cells and internal delivery of GrB, simulating T and B cell therapy. We assessed the construct's antigen-binding specificity and GrB enzymatic activity, as well as in vitro cytotoxicity and internalization into target and control cells. We also assessed pharmacokinetic and toxicology parameters in vivo. RESULTS: GrB-Fc-4D5 was highly cytotoxic to Her2 positive cells such as SKBR3, MCF7 and MDA-MB-231 with IC50 values of 56, 99 and 27 nM, respectively, and against a panel of HER2+ cell lines regardless of endogenous expression levels of the PI-9 inhibitor. Contemporaneous studies with Kadcyla demonstrated similar levels of in vitro activity against virtually all cells tested. GrB-Fc-4D5 internalized rapidly into target SKOV3 cells within 1 h of exposure rapidly delivering GrB to the cytoplasmic compartment. In keeping with its relatively high molecular weight (160 kDa), the construct demonstrated a terminal-phase serum half-life in mice of 39.2 h. Toxicity studies conducted on BALB/c mice demonstrated no statistically significant changes in SGPT, SGOT or serum LDH. Histopathologic analysis of tissues from treated mice demonstrated no drug-related changes in any tissues examined. CONCLUSION: GrB-Fc-4D5 shows excellent, specific cytotoxicity and demonstrates no significant toxicity in normal, antigen-negative murine models. This construct constitutes a novel approach against HER2-expressing tumors and is an excellent candidate for further development.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Drug Development , Molecular Targeted Therapy , Receptor, ErbB-2/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/isolation & purification , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Drug Delivery Systems , Gene Expression , Genetic Vectors/genetics , Granzymes/administration & dosage , Granzymes/genetics , Humans , Mice , Molecular Targeted Therapy/methods , Protein Binding/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Single-Chain Antibodies/genetics , Xenograft Model Antitumor Assays
2.
Cancer Immunol Immunother ; 67(8): 1251-1260, 2018 08.
Article in English | MEDLINE | ID: mdl-29869168

ABSTRACT

The recruitment of T-cells by bispecific antibodies secreted from adoptively transferred, gene-modified autologous cells has shown satisfactory results in preclinical cancer models. Even so, the approach's translation into the clinic will require incremental improvements to its efficacy and reduction of its toxicity. Here, we characterized a tandem T-cell recruiting bispecific antibody intended to benefit gene-based immunotherapy approaches, which we call the light T-cell engager (LiTE), consisting of an EGFR-specific single-domain VHH antibody fused to a CD3-specific scFv. We generated two LiTEs with the anti-EGFR VHH and the anti-CD3 scFv arranged in both possible orders. Both constructs were well expressed in mammalian cells as highly homogenous monomers in solution with molecular weights of 43 and 41 kDa, respectively. In situ secreted LiTEs bound the cognate antigens of both parental antibodies and triggered the specific cytolysis of EGFR-expressing cancer cells without inducing T-cell activation and cytotoxicity spontaneously or against EGFR-negative cells. Light T-cell engagers are, therefore, suitable for future applications in gene-based immunotherapy approaches.


Subject(s)
Antibodies, Bispecific/therapeutic use , CD3 Complex/immunology , ErbB Receptors/immunology , Immunotherapy , Neoplasms/therapy , Single-Chain Antibodies/therapeutic use , T-Lymphocytes/immunology , Cell Proliferation/drug effects , ErbB Receptors/metabolism , Humans , Lymphocyte Activation , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Tumor Cells, Cultured
3.
Oncoimmunology ; 7(1): e1377874, 2017.
Article in English | MEDLINE | ID: mdl-29296540

ABSTRACT

The redirection of T cell activity using bispecific antibodies is one of the most promising cancer immunotherapy approaches currently in development, but it is limited by cytokine storm-related toxicities, as well as the pharmacokinetics and tumor-penetrating capabilities of current bispecific antibody formats. Here, we have engineered the ATTACK (Asymmetric Tandem Trimerbody for T cell Activation and Cancer Killing), a novel T cell-recruiting bispecific antibody which combines three EGFR-binding single-domain antibodies (VHH; clone EgA1) with a single CD3-binding single-chain variable fragment (scFv; clone OKT3) in an intermediate molecular weight package. The two specificities are oriented in opposite directions in order to simultaneously engage cancer cells and T cell effectors, and thereby promote immunological synapse formation. EgA1 ATTACK was expressed as a homogenous, non-aggregating, soluble protein by mammalian cells and demonstrated an enhanced binding to EGFR, but not CD3, when compared to the previously characterized tandem bispecific antibody which has one EgA1 VHH and one OKT3 scFv per molecule. EgA1 ATTACK induced synapse formation and early signaling pathways downstream of TCR engagement at lower concentrations than the tandem VHH-scFv bispecific antibody. Furthermore, it demonstrated extremely potent, dose-dependent cytotoxicity when retargeting human T cells towards EGFR-expressing cells, with an efficacy over 15-fold higher than that of the tandem VHH-scFv bispecific antibody. These results suggest that the ATTACK is an ideal format for the development of the next-generation of T cell-redirecting bispecific antibodies.

4.
Sci Rep ; 6: 28643, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27345490

ABSTRACT

Here, we describe a new strategy that allows the rapid and efficient engineering of mono and multispecific trivalent antibodies. By fusing single-domain antibodies from camelid heavy-chain-only immunoglobulins (VHHs) to the N-terminus of a human collagen XVIII trimerization domain (TIE(XVIII)) we produced monospecific trimerbodies that were efficiently secreted as soluble functional proteins by mammalian cells. The purified VHH-TIE(XVIII) trimerbodies were trimeric in solution and exhibited excellent antigen binding capacity. Furthermore, by connecting with two additional glycine-serine-based linkers three VHH-TIE(XVIII) modules on a single polypeptide chain, we present an approach for the rational design of multispecific tandem trimerbodies with defined stoichiometry and controlled orientation. Using this technology we report here the construction and characterization of a tandem VHH-based trimerbody capable of simultaneously binding to three different antigens: carcinoembryonic antigen (CEA), epidermal growth factor receptor (EGFR) and green fluorescence protein (GFP). Multispecific tandem VHH-based trimerbodies were well expressed in mammalian cells, had good biophysical properties and were capable of simultaneously binding their targeted antigens. Importantly, these antibodies were very effective in inhibiting the proliferation of human epidermoid carcinoma A431 cells. Multispecific VHH-based trimerbodies are therefore ideal candidates for future applications in various therapeutic areas.


Subject(s)
Antibodies, Bispecific , Protein Engineering , Recombinant Fusion Proteins , Single-Chain Antibodies , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Camelids, New World , Humans , Mice , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics
5.
AMB Express ; 5(1): 137, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26239030

ABSTRACT

Recombinant antibodies are used with great success in many different diagnostic and therapeutic applications. A variety of protein expression systems are available, but nowadays almost all therapeutic antibodies are produced in mammalian cell lines due to their complex structure and glycosylation requirements. However, production of clinical-grade antibodies in mammalian cells is very expensive and time-consuming. On the other hand, Escherichia coli (E. coli) is known to be the simplest, fastest and most cost-effective recombinant expression system, which usually achieves higher protein yields than mammalian cells. Indeed, it is one of the most popular host in the industry for the expression of recombinant proteins. In this work, a trivalent single-chain fragment variable (scFv)-based N-terminal trimerbody, specific for native laminin-111, was expressed in human embryonic kidney 293 cells and in E. coli. Mammalian and bacterially produced anti-laminin trimerbody molecules display comparable functional and structural properties, although importantly the yield of trimerbody expressed in E. coli was considerably higher than in human cells. These results demonstrated that E. coli is a versatile and efficient expression system for multivalent trimerbody-based molecules that is suitable for their industrial production.

6.
Microb Cell Fact ; 13: 116, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25112455

ABSTRACT

BACKGROUND: Recombinant antibodies are highly successful in many different pathological conditions and currently enjoy overwhelming recognition of their potential. There are a wide variety of protein expression systems available, but almost all therapeutic antibodies are produced in mammalian cell lines, which mimic human glycosylation. The production of clinical-grade antibodies in mammalian cells is, however, extremely expensive. Compared to mammalian systems, protein production in yeast strains such as Pichia pastoris, is simpler, faster and usually results in higher yields. RESULTS: In this work, a trivalent single-chain fragment variable (scFv)-based N-terminal trimerbody, specific for the human carcinoembryonic antigen (CEA), was expressed in human embryonic kidney 293 cells and in Pichia pastoris. Mammalian- and yeast-produced anti-CEA trimerbody molecules display similar functional and structural properties, yet, the yield of trimerbody expressed in P. pastoris is about 20-fold higher than in human cells. CONCLUSIONS: P. pastoris is an efficient expression system for multivalent trimerbody molecules, suitable for their commercial production.


Subject(s)
Biotechnology/methods , Pichia/metabolism , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/chemistry , Carcinoembryonic Antigen/immunology , Enzyme-Linked Immunosorbent Assay , Genetic Vectors , HEK293 Cells , Humans , Immobilized Proteins/metabolism , Protein Stability , Protein Structure, Tertiary , Serum/metabolism , Single-Chain Antibodies/isolation & purification
7.
Oncoimmunology ; 3: e28810, 2014.
Article in English | MEDLINE | ID: mdl-25057445

ABSTRACT

Gene therapy to achieve in vivo secretion of recombinant anti-CD3 x anti-tumor bispecific antibodies in cancer patients is being explored as a strategy to counterbalance rapid renal elimination, thereby sustaining levels of bispecific antibodies in the therapeutic range. Here, we performed a comparative analysis between single- and two-chain configurations for anti-CD3 x anti-CEA (carcinoembryonic antigen) bispecific antibodies secreted by genetically-modified human cells. We demonstrate that tandem single-chain variable fragment (scFv) antibodies and two-chain diabodies are expressed as soluble secreted proteins with similar yields. However, we found significant differences in their biological functionality (i.e., antigen binding) and in their ability to induce non-specific T cell activation. Whereas single-chain tandem scFvs induced human T cell activation and proliferation in an antigen-independent manner, secreted two-chain diabodies exerted almost no proliferative stimulus when human T cells were cultured alone or in co-cultures with CEA negative cells. Thus, our data suggest that two-chain diabodies are preferable to single-chain tandem scFvs for immunotherapeutic strategies comprising in vivo secretion of bispecific antibodies aiming to recruit and activate anticancer specific lymphocytic effector T cells.

8.
J Biol Chem ; 289(4): 2457-68, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24307174

ABSTRACT

Pericytes and mesenchymal stem cells (MSCs) are ontogenically related, and in fact, no significant phenotypic differences could be observed by flow cytometry. Transcriptome analysis of human pericytes and MSCs revealed that 43 genes were up-regulated more than 10-fold in pericytes compared with MSCs. Identification of Toll-like receptor 4 (TLR4) as one of the most abundant RNA species in pericytes with respect to MSCs and confirmation of TLR4 expression on the cell surface led us to obtain a comprehensive overview of the expression program of lipopolysaccharide (LPS)-stimulated pericytes. Transcriptional profiling of LPS-treated cells revealed that 22 genes were up-regulated more than 5-fold. Of them, 10 genes encoded chemokines and cytokines (CXCL10, CCL20, IL8, CXCL1, IL6, CCL2, IL1B, CXCL2, IL1A, and CXCL6), and three genes encoded adhesion molecules (ICAM1, VCAM1, and SELE). LPS induced nuclear translocation of the transcription factor NF-κB in stimulated pericytes. Moreover, inhibition of NF-κB activation by SC-514 blocked LPS-induced up-regulation of a subset of chemokine genes, confirming the key role of NF-κB in LPS signaling in pericytes. At the protein level, we assessed the secretion of the proinflammatory cytokines and chemokines IL-6, IL-8, CXCL1, CXCL2, CXCL3, and CCL2 not only after LPS treatment but also in HMGB1-stimulated pericytes. Up-regulation of the adhesion molecules ICAM-1 and VCAM-1 resulted in an increased adhesion of peripheral blood leukocytes to an LPS-treated pericyte monolayer. The role of pericytes in the inflammatory context has been scarcely addressed; according to these results, pericytes should be considered as active players in the inflammatory cascade with potential physiopathological implications.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Cytokines/biosynthesis , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Pericytes/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/agonists , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/immunology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Adhesion Molecules/immunology , Cell Nucleus/immunology , Cell Nucleus/metabolism , Cells, Cultured , Coculture Techniques , Cytokines/immunology , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Leukocytes/cytology , Leukocytes/immunology , Leukocytes/metabolism , NF-kappa B/immunology , Pericytes/cytology , Pericytes/immunology , Signal Transduction/immunology , Thiophenes/pharmacology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Up-Regulation/drug effects , Up-Regulation/immunology
9.
Int J Cancer ; 134(12): 2984-90, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24242829

ABSTRACT

Snail1 is a transcriptional factor that plays an important role in epithelial-mesenchymal transition and in the acquisition of invasive properties by epithelial cells. In colon tumors, Snail1 expression in the stroma correlates with lower specific survival of cancer patients. However, the role(s) of Snail1 expression in stroma and its association with patients' survival have not been determined. We used human primary carcinoma-associated fibroblasts (CAFs) or normal fibroblasts (NFs) and fibroblast cell lines to analyze the effects of Snail1 expression on the protumorigenic capabilities in colon cancer cells. Snail1 expression was higher in CAFs than in NFs and, as well as α-SMA, a classic marker of activated CAFs. Moreover, in tumor samples from 50 colon cancer patients, SNAI1 expression was associated with expression of other CAF markers, such as α-SMA and fibroblast activation protein. Interestingly, coculture of CAFs with colon cells induced a significant increase in epithelial cell migration and proliferation, which was associated with endogenous SNAI1 expression levels. Ectopic manipulation of Snail1 in fibroblasts demonstrated that Snail1 expression controlled migration as well as proliferation of cocultured colon cancer cells in a paracrine manner. Furthermore, expression of Snail1 in fibroblasts was required for the coadjuvant effect of these cells on colon cancer cell growth and invasion when coxenografted in nude mice. Finally, cytokine profile changes, particularly MCP-3 expression, in fibroblasts are put forward as mediators of Snail1-derived effects on colon tumor cell migration. In summary, these studies demonstrate that Snail1 is necessary for the protumorigenic effects of fibroblasts on colon cancer cells.


Subject(s)
Carcinogenesis , Colonic Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Cycle , Cell Movement , Cell Proliferation , Coculture Techniques , Colonic Neoplasms/genetics , Cytokines/metabolism , Endopeptidases , Female , Fibroblasts/pathology , Gelatinases/genetics , Gelatinases/metabolism , Gene Expression , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Messenger/biosynthesis , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Snail Family Transcription Factors , Tumor Cells, Cultured
10.
PLoS One ; 8(8): e72957, 2013.
Article in English | MEDLINE | ID: mdl-23951338

ABSTRACT

Metastatic breast cancer is the leading cause of death by malignancy in women worldwide. Tumor metastasis is a multistep process encompassing local invasion of cancer cells at primary tumor site, intravasation into the blood vessel, survival in systemic circulation, and extravasation across the endothelium to metastasize at a secondary site. However, only a small percentage of circulating cancer cells initiate metastatic colonies. This fact, together with the inaccessibility and structural complexity of target tissues has hampered the study of the later steps in cancer metastasis. In addition, most data are derived from in vivo models where critical steps such as intravasation/extravasation of human cancer cells are mediated by murine endothelial cells. Here, we developed a new mouse model to study the molecular and cellular mechanisms underlying late steps of the metastatic cascade. We have shown that a network of functional human blood vessels can be formed by co-implantation of human endothelial cells and mesenchymal cells, embedded within a reconstituted basement membrane-like matrix and inoculated subcutaneously into immunodeficient mice. The ability of circulating cancer cells to colonize these human vascularized organoids was next assessed in an orthotopic model of human breast cancer by bioluminescent imaging, molecular techniques and immunohistological analysis. We demonstrate that disseminated human breast cancer cells efficiently colonize organoids containing a functional microvessel network composed of human endothelial cells, connected to the mouse circulatory system. Human breast cancer cells could be clearly detected at different stages of the metastatic process: initial arrest in the human microvasculature, extravasation, and growth into avascular micrometastases. This new mouse model may help us to map the extravasation process with unprecedented detail, opening the way for the identification of relevant targets for therapeutic intervention.


Subject(s)
Breast Neoplasms/pathology , Neoplasm Metastasis/pathology , Neoplastic Cells, Circulating/pathology , Organoids/blood supply , Organoids/pathology , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Nude
11.
Proc Natl Acad Sci U S A ; 110(34): 13791-6, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23918357

ABSTRACT

Antibody cancer therapies rely on systemically accessible targets and suitable antibodies that exert a functional activity or deliver a payload to the tumor site. Here, we present proof-of-principle of in vivo selection of human antibodies in tumor-bearing mice that identified a tumor-specific antibody able to deliver a payload and unveils the target antigen. By using an ex vivo enrichment process against freshly disaggregated tumors to purge the repertoire, in combination with in vivo biopanning at optimized phage circulation time, we have identified a human domain antibody capable of mediating selective localization of phage to human prostate cancer xenografts. Affinity chromatography followed by mass spectrometry analysis showed that the antibody recognizes the proteasome activator complex PA28. The specificity of soluble antibody was confirmed by demonstrating its binding to the active human PA28αß complex. Whereas systemically administered control phage was confined in the lumen of blood vessels of both normal tissues and tumors, the selected phage spread from tumor vessels into the perivascular tumor parenchyma. In these areas, the selected phage partially colocalized with PA28 complex. Furthermore, we found that the expression of the α subunit of PA28 [proteasome activator complex subunit 1 (PSME1)] is elevated in primary and metastatic human prostate cancer and used anti-PSME1 antibodies to show that PSME1 is an accessible marker in mouse xenograft tumors. These results support the use of PA28 as a tumor marker and a potential target for therapeutic intervention in prostate cancer.


Subject(s)
Antibodies, Neoplasm/immunology , Biomarkers, Tumor/immunology , Immunotherapy/methods , Muscle Proteins/metabolism , Prostatic Neoplasms/immunology , Proteasome Endopeptidase Complex/metabolism , Animals , Antibodies, Neoplasm/metabolism , Antibody Specificity , Blotting, Western , Cell Surface Display Techniques , Chromatography, Affinity , Chromatography, Liquid , Drug Delivery Systems/methods , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Immunoprecipitation , Male , Mice , Mice, Nude , Prostatic Neoplasms/therapy , Statistics, Nonparametric , Tandem Mass Spectrometry
12.
MAbs ; 5(1): 70-9, 2013.
Article in English | MEDLINE | ID: mdl-23221741

ABSTRACT

Here, we describe a new class of multivalent and multispecific antibody-based reagents for therapy. The molecules, termed "trimerbodies," use a modified version of the N-terminal trimerization region of human collagen XVIII noncollagenous 1 domain flanked by two flexible linkers as trimerizing scaffold. By fusing single-chain variable fragments (scFv) with the same or different specificity to both N- and C-terminus of the trimerizing scaffold domain, we produced monospecific or bispecific hexavalent molecules that were efficiently secreted as soluble proteins by transfected mammalian cells. A bispecific anti-laminin x anti-CD3 N-/C-trimerbody was found to be trimeric in solution, very efficient at recognizing purified plastic-immobilized laminin and CD3 expressed at the surface of T cells, and remarkably stable in human serum. The bispecificity was further demonstrated in T cell activation studies. In the presence of laminin-rich substrate, the bispecific anti-laminin x anti-CD3 N-/C-trimerbody stimulated a high percentage of human T cells to express surface activation markers. These results suggest that the trimerbody platform offers promising opportunities for the development of the next-generation therapeutic antibodies, i.e., multivalent and bispecific molecules with a format optimized for the desired pharmacokinetics and adapted to the pathological context.


Subject(s)
Antibodies, Bispecific , Collagen Type XVIII , Recombinant Fusion Proteins , Single-Chain Antibodies , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibodies, Bispecific/metabolism , Antibody Affinity , Antibody Specificity , CD3 Complex/immunology , CD3 Complex/metabolism , Collagen Type XVIII/chemistry , Collagen Type XVIII/genetics , Collagen Type XVIII/immunology , Collagen Type XVIII/metabolism , HEK293 Cells , Humans , Jurkat Cells , Laminin/immunology , Laminin/metabolism , Lymphocyte Activation , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/metabolism , T-Lymphocytes/immunology , Transfection
13.
PLoS One ; 7(6): e39097, 2012.
Article in English | MEDLINE | ID: mdl-22723936

ABSTRACT

Laminins are large heterotrimeric cross-shaped extracellular matrix glycoproteins with terminal globular domains and a coiled-coil region through which the three chains are assembled and covalently linked. Laminins are key components of basement membranes, and they serve as attachment sites for cell adhesion, migration and proliferation. In this work, we produced a recombinant fragment comprising the entire laminin coiled-coil of the α1-, ß1-, and γ1-chains that assemble into a stable heterotrimeric coiled-coil structure independently of the rest of the molecule. This domain was biologically active and not only failed to serve as a substrate for cell attachment, spreading and focal adhesion formation but also inhibited cell adhesion to laminin when added to cells in a soluble form at the time of seeding. Furthermore, gene array expression profiling in cells cultured in the presence of the laminin coiled-coil domain revealed up-regulation of genes involved in cell motility and invasion. These findings were confirmed by real-time quantitative PCR and zymography assays. In conclusion, this study shows for the first time that the laminin coiled-coil domain displays anti-adhesive functions and has potential implications for cell migration during matrix remodeling.


Subject(s)
Cell Movement/genetics , Laminin/genetics , Laminin/metabolism , Protein Interaction Domains and Motifs/physiology , Protein Multimerization , Animals , Cell Adhesion/genetics , Cell Line, Tumor , Enzyme Activation , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Laminin/chemistry , Matrix Metalloproteinase 2/metabolism , Mice , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Reproducibility of Results
14.
MAbs ; 4(2): 226-32, 2012.
Article in English | MEDLINE | ID: mdl-22453098

ABSTRACT

We recently described the in vitro and in vivo properties of an engineered homotrimeric antibody made by fusing the N-terminal trimerization region of collagen XVIII NC1 domain to the C-terminus of a scFv fragment [trimerbody (scFv-NC1) 3; 110 kDa]. Here, we demonstrated the utility of the N-terminal trimerization region of collagen XV NC1 domain in the engineering of trivalent antibodies. We constructed several scFv-based trimerbodies containing the human type XV trimerization domain and demonstrated that all the purified trimerbodies were trimeric in solution and exhibited excellent antigen binding capacity. Importantly, type XV trimerbodies demonstrated substantially greater thermal and serum stability and resistance to protease digestion than type XVIII trimerbodies. In summary, the small size, high expression level, solubility and stability of the trimerization domain of type XV collagen make it the ideal choice for engineering homotrimeric antibodies for cancer detection and therapy.


Subject(s)
Antibodies, Neoplasm , Collagen , Recombinant Fusion Proteins , Single-Chain Antibodies , Animals , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Collagen/biosynthesis , Collagen/genetics , Collagen/immunology , HEK293 Cells , Humans , Mice , Neoplasms/immunology , Neoplasms/pathology , Protein Stability , Protein Structure, Tertiary , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...