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










Publication year range
1.
Front Med (Lausanne) ; 9: 1058455, 2022.
Article in English | MEDLINE | ID: mdl-36507540

ABSTRACT

Triple-negative breast cancer (TNBC) is characterized by aggressiveness and high rates of metastasis. The identification of relevant biomarkers is crucial to improve outcomes for TNBC patients. Membrane type 1-matrix metalloproteinase (MT1-MMP) could be a good candidate because its expression has been reported to correlate with tumor malignancy, progression and metastasis. Moreover, single-domain variable regions (VHHs or Nanobodies) derived from camelid heavy-chain-only antibodies have demonstrated improvements in tissue penetration and blood clearance, important characteristics for cancer imaging. Here, we have developed a nanobody-based PET imaging strategy for TNBC detection that targets MT1-MMP. A llama-derived library was screened against the catalytic domain of MT1-MMP and a panel of specific nanobodies were identified. After a deep characterization, two nanobodies were selected to be labeled with gallium-68 (68Ga). ImmunoPET imaging with both ([68Ga]Ga-NOTA-3TPA14 and [68Ga]Ga-NOTA-3CMP75) in a TNBC mouse model showed precise tumor-targeting capacity in vivo with high signal-to-background ratios. (68Ga)Ga-NOTA-3CMP75 exhibited higher tumor uptake compared to (68Ga)Ga-NOTA-3TPA14. Furthermore, imaging data correlated perfectly with the immunohistochemistry staining results. In conclusion, we found a promising candidate for nanobody-based PET imaging to be further investigated as a diagnostic tool in TNBC.

2.
Elife ; 92020 09 08.
Article in English | MEDLINE | ID: mdl-32897857

ABSTRACT

Endothelial integrity is vital for homeostasis and adjusted to tissue demands. Although fluid uptake by lymphatic capillaries is a critical attribute of the lymphatic vasculature, the barrier function of collecting lymphatic vessels is also important by ensuring efficient fluid drainage as well as lymph node delivery of antigens and immune cells. Here, we identified the transmembrane ligand EphrinB2 and its receptor EphB4 as critical homeostatic regulators of collecting lymphatic vessel integrity. Conditional gene deletion in mice revealed that EphrinB2/EphB4 signalling is dispensable for blood endothelial barrier function, but required for stabilization of lymphatic endothelial cell (LEC) junctions in different organs of juvenile and adult mice. Studies in primary human LECs further showed that basal EphrinB2/EphB4 signalling controls junctional localisation of the tight junction protein CLDN5 and junction stability via Rac1/Rho-mediated regulation of cytoskeletal contractility. EphrinB2/EphB4 signalling therefore provides a potential therapeutic target to selectively modulate lymphatic vessel permeability and function.


Lymph vessels are thin walled tubes that, similar to blood vessels, carry white blood cells, fluids and waste. Unlike veins and arteries, however, lymph vessels do not carry red blood cells and their main function is to remove excess fluid from tissues. The cells that line vessels in the body are called endothelial cells, and they are tightly linked together by proteins to control what goes into and comes out of the vessels. The chemical, physical and mechanical signals that control the junctions between endothelial cells are often the same in different vessel types, but their effects can vary. The endothelial cells of both blood and lymph vessels have two interacting proteins on their membrane known as EphrinB2 and its receptor, EphB4. When these two proteins interact, the EphB4 receptor becomes activated, which leads to changes in the junctions that link endothelial cells together. Frye et al. examined the role of EphrinB2 and EphB4 in the lymphatic system of mice. When either EphrinB2 or EphB4 are genetically removed in newborn or adult mice, lymph vessels become disrupted, but no significant effect is observed on blood vessels. The reason for the different responses in blood and lymph vessels is unknown. The results further showed that lymphatic endothelial cells need EphB4 and EphrinB2 to be constantly interacting to maintain the integrity of the lymph vessels. Further examination of human endothelial cells grown in the laboratory revealed that this constant signalling controls the internal protein scaffold that determines a cell's shape and integrity. Changes in the internal scaffold affect the organization of the junctions that link neighboring lymphatic endothelial cells together. The loss of signalling between EphrinB2 and EphB4 in lymph vessels reflects the increase in vessel leakage seen in response to bacterial infections and in some genetic conditions such as lymphoedema. Finding ways to control the signalling between these two proteins could help treat these conditions by developing drugs that improve endothelial cell integrity in lymph vessels.


Subject(s)
Endothelial Cells/metabolism , Ephrin-B2/genetics , Homeostasis , Intercellular Junctions/metabolism , Lymphatic Vessels/physiology , Receptor, EphB4/genetics , Signal Transduction , Animals , Claudin-5/genetics , Ephrin-B2/metabolism , Gene Deletion , Mice , Receptor, EphB4/metabolism
3.
Clin Cancer Res ; 25(11): 3352-3365, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30944125

ABSTRACT

PURPOSE: A driving factor in pancreatic ductal adenocarcinoma (PDAC) treatment resistance is the tumor microenvironment, which is highly immunosuppressive. One potent immunologic adjuvant is radiotherapy. Radiation, however, has also been shown to induce immunosuppressive factors, which can contribute to tumor progression and formation of fibrotic tumor stroma. To capitalize on the immunogenic effects of radiation and obtain a durable tumor response, radiation must be rationally combined with targeted therapies to mitigate the influx of immunosuppressive cells and fibrosis. One such target is ephrinB2, which is overexpressed in PDAC and correlates negatively with prognosis.Experimental Design: On the basis of previous studies of ephrinB2 ligand-EphB4 receptor signaling, we hypothesized that inhibition of ephrinB2-EphB4 combined with radiation can regulate the microenvironment response postradiation, leading to increased tumor control in PDAC. This hypothesis was explored using both cell lines and in vivo human and mouse tumor models. RESULTS: Our data show this treatment regimen significantly reduces regulatory T-cell, macrophage, and neutrophil infiltration and stromal fibrosis, enhances effector T-cell activation, and decreases tumor growth. Furthermore, our data show that depletion of regulatory T cells in combination with radiation reduces tumor growth and fibrosis. CONCLUSIONS: These are the first findings to suggest that in PDAC, ephrinB2-EphB4 interaction has a profibrotic, protumorigenic role, presenting a novel and promising therapeutic target.


Subject(s)
Ephrin-B2/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Receptor, EphB4/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers , Cell Line, Tumor , Disease Models, Animal , Ephrin-B2/antagonists & inhibitors , Ephrin-B2/genetics , Female , Flow Cytometry , Gene Expression , Gene Knockdown Techniques , Humans , Immunohistochemistry , Mice , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/methods , Neutrophils/immunology , Neutrophils/metabolism , Pancreatic Neoplasms/therapy , Radiotherapy/adverse effects , Radiotherapy/methods , Receptor, EphB4/antagonists & inhibitors , Receptor, EphB4/genetics , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Xenograft Model Antitumor Assays
4.
Clin Cancer Res ; 24(15): 3550-3559, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29588308

ABSTRACT

Purpose: Despite the wide use of antiangiogenic drugs in the clinical setting, predictive biomarkers of response to these drugs are still unknown.Experimental Design: We applied whole-exome sequencing of matched germline and basal plasma cell-free DNA samples (WES-cfDNA) on a RAS/BRAF/PIK3CA wild-type metastatic colorectal cancer patient with primary resistance to standard treatment regimens, including inhibitors to the VEGF:VEGFR2 pathway. We performed extensive functional experiments, including ectopic expression of VEGFR2 mutants in different cell lines, kinase and drug sensitivity assays, and cell- and patient-derived xenografts.Results: WES-cfDNA yielded a 77% concordance rate with tumor exome sequencing and enabled the identification of the KDR/VEGFR2 L840F clonal, somatic mutation as the cause of therapy refractoriness in our patient. In addition, we found that 1% to 3% of samples from cancer sequencing projects harbor KDR somatic mutations located in protein residues frequently mutated in other cancer-relevant kinases, such as EGFR, ABL1, and ALK. Our in vitro and in vivo functional assays confirmed that L840F causes strong resistance to antiangiogenic drugs, whereas the KDR hot-spot mutant R1032Q confers sensitivity to strong VEGFR2 inhibitors. Moreover, we showed that the D717V, G800D, G800R, L840F, G843D, S925F, R1022Q, R1032Q, and S1100F VEGFR2 mutants promote tumor growth in mice.Conclusions: Our study supports WES-cfDNA as a powerful platform for portraying the somatic mutation landscape of cancer and discovery of new resistance mechanisms to cancer therapies. Importantly, we discovered that VEGFR2 is somatically mutated across tumor types and that VEGFR2 mutants can be oncogenic and control sensitivity/resistance to antiangiogenic drugs. Clin Cancer Res; 24(15); 3550-9. ©2018 AACR.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Colorectal Neoplasms/genetics , Neovascularization, Pathologic/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Anaplastic Lymphoma Kinase/genetics , Angiogenesis Inhibitors/adverse effects , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Colorectal Neoplasms/blood , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , ErbB Receptors/genetics , Exome/genetics , Female , Heterografts , Humans , Mice , Mutation , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Protein Conformation/drug effects , Proto-Oncogene Proteins c-abl/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/blood , Vascular Endothelial Growth Factor Receptor-2/chemistry , Exome Sequencing
5.
Nature ; 546(7660): 676-680, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28658220

ABSTRACT

Cutaneous melanoma is a type of cancer with an inherent potential for lymph node colonization, which is generally preceded by neolymphangiogenesis. However, sentinel lymph node removal does not necessarily extend the overall survival of patients with melanoma. Moreover, lymphatic vessels collapse and become dysfunctional as melanomas progress. Therefore, it is unclear whether (and how) lymphangiogenesis contributes to visceral metastasis. Soluble and vesicle-associated proteins secreted by tumours and/or their stroma have been proposed to condition pre-metastatic sites in patients with melanoma. Still, the identities and prognostic value of lymphangiogenic mediators remain unclear. Moreover, our understanding of lymphangiogenesis (in melanomas and other tumour types) is limited by the paucity of mouse models for live imaging of distal pre-metastatic niches. Injectable lymphatic tracers have been developed, but their limited diffusion precludes whole-body imaging at visceral sites. Vascular endothelial growth factor receptor 3 (VEGFR3) is an attractive 'lymphoreporter' because its expression is strongly downregulated in normal adult lymphatic endothelial cells, but is activated in pathological situations such as inflammation and cancer. Here, we exploit this inducibility of VEGFR3 to engineer mouse melanoma models for whole-body imaging of metastasis generated by human cells, clinical biopsies or endogenously deregulated oncogenic pathways. This strategy revealed early induction of distal pre-metastatic niches uncoupled from lymphangiogenesis at primary lesions. Analyses of the melanoma secretome and validation in clinical specimens showed that the heparin-binding factor midkine is a systemic inducer of neo-lymphangiogenesis that defines patient prognosis. This role of midkine was linked to a paracrine activation of the mTOR pathway in lymphatic endothelial cells. These data support the use of VEGFR3 reporter mice as a 'MetAlert' discovery platform for drivers and inhibitors of metastasis.


Subject(s)
Cytokines/metabolism , Lymphatic Vessels/metabolism , Neoplasm Metastasis/diagnostic imaging , Neoplasm Metastasis/pathology , Whole Body Imaging/methods , Animals , Disease Models, Animal , Disease Progression , Endothelial Cells/metabolism , Female , Genes, Reporter , Humans , Lymphangiogenesis , Lymphatic Vessels/pathology , Male , Melanoma/diagnostic imaging , Melanoma/metabolism , Melanoma/pathology , Mice , Midkine , Paracrine Communication , Prognosis , Recurrence , Reproducibility of Results , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor Receptor-3/analysis , Vascular Endothelial Growth Factor Receptor-3/metabolism , Xenograft Model Antitumor Assays
6.
J Clin Invest ; 126(12): 4554-4568, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27820703

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) reside in the bone marrow. Stress signals from cancer and other conditions promote HSPC mobilization into circulation and subsequent homing to tissue microenvironments. HSPC infiltration into tissue microenvironments can influence disease progression; notably, in cancer, HSPCs encourage tumor growth. Here we have uncovered a mutually exclusive distribution of EPHB4 receptors in bone marrow sinusoids and ephrin B2 ligands in hematopoietic cells. We determined that signaling interactions between EPHB4 and ephrin B2 control HSPC mobilization from the bone marrow. In mice, blockade of the EPHB4/ephrin B2 signaling pathway reduced mobilization of HSPCs and other myeloid cells to the circulation. EPHB4/ephrin B2 blockade also reduced HSPC infiltration into tumors as well as tumor progression in murine models of melanoma and mammary cancer. These results identify EPHB4/ephrin B2 signaling as critical to HSPC mobilization from bone marrow and provide a potential strategy for reducing cancer progression by targeting the bone marrow.


Subject(s)
Bone Marrow/metabolism , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Receptor, EphB4/metabolism , Signal Transduction/physiology , Stem Cell Niche/physiology , Animals , Cell Line , Ephrin-B2/genetics , Ephrin-B2/metabolism , Mice , Receptor, EphB4/genetics
7.
Sci Rep ; 6: 30293, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27456546

ABSTRACT

WW domains are small domains present in many human proteins with a wide array of functions and acting through the recognition of proline-rich sequences. The WW domain belonging to polyglutamine tract-binding protein 1 (PQBP1) is of particular interest due to its direct involvement in several X chromosome-linked intellectual disabilities, including Golabi-Ito-Hall (GIH) syndrome, where a single point mutation (Y65C) correlates with the development of the disease. The mutant cannot bind to its natural ligand WBP11, which regulates mRNA processing. In this work we use high-field high-resolution NMR and enhanced sampling molecular dynamics simulations to gain insight into the molecular causes the disease. We find that the wild type protein is partially unfolded exchanging among multiple beta-strand-like conformations in solution. The Y65C mutation further destabilizes the residual fold and primes the protein for the formation of a disulphide bridge, which could be at the origin of the loss of function.


Subject(s)
Carrier Proteins/genetics , Cerebral Palsy/genetics , DNA-Binding Proteins/genetics , Intellectual Disability/genetics , Mental Retardation, X-Linked/genetics , Nuclear Proteins/genetics , Cerebral Palsy/pathology , DNA-Binding Proteins/chemistry , Humans , Intellectual Disability/pathology , Mental Retardation, X-Linked/pathology , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins/chemistry , Point Mutation/genetics , Protein Binding , Protein Conformation, beta-Strand , Protein Folding , RNA Splicing Factors , RNA, Messenger/chemistry , RNA, Messenger/genetics , WW Domains/genetics
8.
PLoS Comput Biol ; 11(11): e1004578, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26606374

ABSTRACT

Due to its inhibition of the Abl kinase domain in the BCR-ABL fusion protein, imatinib is strikingly effective in the initial stage of chronic myeloid leukemia with more than 90% of the patients showing complete remission. However, as in the case of most targeted anti-cancer therapies, the emergence of drug resistance is a serious concern. Several drug-resistant mutations affecting the catalytic domain of Abl and other tyrosine kinases are now known. But, despite their importance and the adverse effect that they have on the prognosis of the cancer patients harboring them, the molecular mechanism of these mutations is still debated. Here by using long molecular dynamics simulations and large-scale free energy calculations complemented by in vitro mutagenesis and microcalorimetry experiments, we model the effect of several widespread drug-resistant mutations of Abl. By comparing the conformational free energy landscape of the mutants with those of the wild-type tyrosine kinases we clarify their mode of action. It involves significant and complex changes in the inactive-to-active dynamics and entropy/enthalpy balance of two functional elements: the activation-loop and the conserved DFG motif. What is more the T315I gatekeeper mutant has a significant impact on the binding mechanism itself and on the binding kinetics.


Subject(s)
Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Fusion Proteins, bcr-abl/chemistry , Fusion Proteins, bcr-abl/genetics , Imatinib Mesylate/pharmacology , Computational Biology , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate/chemistry , Imatinib Mesylate/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Thermodynamics
9.
PLoS One ; 10(6): e0125203, 2015.
Article in English | MEDLINE | ID: mdl-26066800

ABSTRACT

Tumour-associated macrophages (TAMs) have been associated with survival in classic Hodgkin lymphoma (cHL) and other lymphoma types. The maturation and differentiation of tissue macrophages depends upon interactions between colony-stimulating factor 1 receptor (CSF1R) and its ligands. There remains, however, a lack of consistent information on CSF1R expression in TAMs. A new monoclonal antibody, FER216, was generated to investigate CSF1R protein distribution in formalin fixed tissue samples from 24 reactive lymphoid tissues and 187 different lymphoma types. We also analysed the distribution of CSF1R+, CD68+ and CD163+ macrophages by double immunostaining, and studied the relationship between CSF1R expression and survival in an independent series of 249 cHL patients. CSF1R+ TAMs were less frequent in B-cell lymphocytic leukaemia and lymphoblastic B-cell lymphoma than in diffuse large B-cell lymphoma, peripheral T-cell lymphoma, angioimmunoblastic T-cell lymphoma and cHL. HRS cells in cHL and, with the exception of three cases of anaplastic large cell lymphoma, the neoplastic cells in NHLs, lacked detectable CSF1R protein. A CSF1R+ enriched microenvironment in cHL was associated with shorter survival in an independent series of 249 cHL patients. CSF1R pathway activation was evident in the cHL and inactivation of this pathway could be a potential therapeutic target in cHL cases.


Subject(s)
Hodgkin Disease/metabolism , Lymphoid Tissue/metabolism , Lymphoma/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Animals , Cell Line, Tumor , Gene Expression , Hodgkin Disease/genetics , Hodgkin Disease/pathology , Humans , Immunohistochemistry , Immunoprecipitation , Lymphoid Tissue/pathology , Lymphoma/diagnosis , Lymphoma/genetics , Mice , Receptor, Macrophage Colony-Stimulating Factor/genetics , Signal Transduction
10.
J Clin Invest ; 125(2): 824-30, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25574842

ABSTRACT

NOTCH signaling suppresses tumor growth and proliferation in several types of stratified epithelia. Here, we show that missense mutations in NOTCH1 and NOTCH2 found in human bladder cancers result in loss of function. In murine models, genetic ablation of the NOTCH pathway accelerated bladder tumorigenesis and promoted the formation of squamous cell carcinomas, with areas of mesenchymal features. Using bladder cancer cells, we determined that the NOTCH pathway stabilizes the epithelial phenotype through its effector HES1 and, consequently, loss of NOTCH activity favors the process of epithelial-mesenchymal transition. Evaluation of human bladder cancer samples revealed that tumors with low levels of HES1 present mesenchymal features and are more aggressive. Together, our results indicate that NOTCH serves as a tumor suppressor in the bladder and that loss of this pathway promotes mesenchymal and invasive features.


Subject(s)
Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Urinary Bladder Neoplasms/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Transcription Factor HES-1 , Tumor Suppressor Proteins/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
11.
Sci Signal ; 7(321): ra35, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24736456

ABSTRACT

Vav proteins are phosphorylation-dependent guanine nucleotide exchange factors (GEFs) that catalyze the activation of members of the Rho family of guanosine triphosphatases (GTPases). The current regulatory model holds that the nonphosphorylated, catalytically inactive state of these GEFs is maintained by intramolecular interactions among the amino-terminal domains and the central catalytic core, which block the binding of Vav proteins to GTPases. We showed that this autoinhibition is mechanistically more complex, also involving the bivalent association of the carboxyl-terminal Src homology 3 (SH3) region of Vav with its catalytic and pleckstrin homology (PH) domains. Such interactions occurred through proline-rich region-independent mechanisms. Full release from this double-locked state required synergistic weakening effects from multiple phosphorylated tyrosine residues, thus providing an optimized system to generate gradients of Vav GEF activity depending on upstream signaling inputs. This mechanism is shared by mammalian and Drosophila melanogaster Vav proteins, suggesting that it may be a common regulatory feature for this protein family.


Subject(s)
Proto-Oncogene Proteins c-vav/chemistry , src Homology Domains , Animals , COS Cells , Catalytic Domain , Chlorocebus aethiops , Drosophila melanogaster/metabolism , Escherichia coli/metabolism , GTP Phosphohydrolases/metabolism , Humans , Imaging, Three-Dimensional , Jurkat Cells , Mice , Multigene Family , NIH 3T3 Cells , Phosphorylation , Proline/chemistry , Proto-Oncogene Proteins c-vav/metabolism , Signal Transduction , Tyrosine/chemistry
12.
Blood ; 119(19): 4565-76, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22446484

ABSTRACT

Membrane-anchored ephrinB2 and its receptor EphB4 are involved in the formation of blood and lymphatic vessels in normal and pathologic conditions. Eph/ephrin activation requires cell-cell interactions and leads to bidirectional signaling pathways in both ligand- and receptor-expressing cells. To investigate the functional consequences of blocking ephrinB2 activity, 2 highly specific human single-chain Fv (scFv) Ab fragments against ephrinB2 were generated and characterized. Both Ab fragments suppressed endothelial cell migration and tube formation in vitro in response to VEGF and provoked abnormal cell motility and actin cytoskeleton alterations in isolated endothelial cells. As only one of them (B11) competed for binding of ephrinB2 to EphB4, these data suggest an EphB-receptor-independent blocking mechanism. Anti-ephrinB2 therapy reduced VEGF-induced neovascularization in a mouse Matrigel plug assay. Moreover, systemic administration of ephrinB2-blocking Abs caused a drastic reduction in the number of blood and lymphatic vessels in xenografted mice and a concomitant reduction in tumor growth. Our results show for the first time that specific Ab-based ephrinB2 targeting may represent an effective therapeutic strategy to be used as an alternative or in combination with existing antiangiogenic drugs for treating patients with cancer and other angiogenesis-related diseases.


Subject(s)
Antibodies/pharmacology , Ephrin-B2/antagonists & inhibitors , Lymphangiogenesis/drug effects , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibodies/metabolism , Antibodies/therapeutic use , Antibody Specificity , Down-Regulation/drug effects , Ephrin-B2/immunology , Ephrin-B2/metabolism , Female , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Immunotherapy/methods , Lymphangiogenesis/physiology , Mice , Mice, Nude , Mice, SCID , Molecular Targeted Therapy , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Mol Cancer Ther ; 7(4): 862-73, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18413799

ABSTRACT

Human single-chain Fv directed against fibroblast growth factor receptor 3 (FGFR3) have been shown to block proliferation of RT112 bladder carcinoma cells in vitro. Here, we examined the ability of the recombinant gelonin toxin (rGel) to enhance this inhibitory effect in vitro and in vivo on the bladder cancer cell line RT112 and the corresponding xenografts. Immunotoxins were genetically engineered by fusing FGFR3-specific Fv fragments (3C) to the NH(2) terminus of rGel and expressed as a soluble protein in Escherichia coli. The 3C/rGel fusion construct showed an IC(50) of 200 nmol/L against log-phase RT112 cells compared with 1,500 nmol/L for free rGel. Immunofluorescence studies showed that the 3C/rGel construct internalized rapidly into the cytoplasm of RT112 cells within 1 h of exposure. The mechanism of immunotoxin-induced cell death was found to be mediated by apoptosis. RT112 tumor xenografts in severe combined immunodeficient mice treated with 50 mg/kg 3C/rGel exhibited considerable growth delay relative to control tumors and a significant reduction of 55% to 70% in mean tumor size. Immunohistochemical analysis showed that tumors from mice treated with 3C/rGel displayed considerable apoptotic damage compared with control groups. Subcellular location of FGFR3 in immunotoxin-treated tumors indicated a translocation of FGFR3 to the nuclear membrane in contrast to tumors from saline-treated controls. These results show that FGFR3-driven immunotoxins may be an effective therapeutic agent against human bladder and other tumor types overexpressing FGFR3.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Immunotoxins , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Ribosome Inactivating Proteins, Type 1/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Animals , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/pathology , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoglobulin Fragments , Mice , Mice, Nude , Mice, SCID , Protein Transport , RNA, Small Interfering/pharmacology , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/genetics , Recombinant Fusion Proteins/therapeutic use , Surface Plasmon Resonance , Survival Rate , Tumor Cells, Cultured , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
14.
Int J Cancer ; 122(10): 2351-9, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18224686

ABSTRACT

Gastrin and its derivatives are becoming important targets for immunotherapy of pancreatic, gastric and colorectal tumors. This study was conducted to design antibodies able to block gastrin binding to the gastrin/cholecystokinin-2 (CCK-2) receptor in order to delay tumor growth. The authors have used different gastrin molecules, combined with the diphtheria toxoid, to generate and select human single chain variable fragments (scFvs) as well as mouse monoclonal antibodies and scFvs against different regions of gastrin. There was a remarkable conservation in the antibody repertoire against gastrin, independently of the approach and the species. The germlines most frequently used in gastrin antibody formation were identified. Three different epitopes were identified in the gastrin molecule. The resulting mouse monoclonal antibodies and scFvs were analyzed for gastrin neutralization using Colo 320 WT cells, which overexpress the CCK-2 receptor. The gastrin neutralizing activity assay showed that N-terminal specific mouse monoclonal antibodies were more efficient to inhibit proliferation of Colo 320 WT cells than the anti-C terminal antibodies. Moreover, the human antigastrin scFvs obtained in this study inhibited significantly the proliferation of Colo 320 tumoral cells. These findings should contribute to a more rational design of antibody-based antigastrin therapies in cancer, including passive administration of human antibodies with blocking activity.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Colonic Neoplasms/metabolism , Gastrins/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Cell Proliferation , Colonic Neoplasms/pathology , Diphtheria Toxoid/metabolism , Enzyme-Linked Immunosorbent Assay , Gastrins/immunology , Humans , Immunization , Immunoglobulin Variable Region/immunology , Mice , Peptide Library , Receptor, Cholecystokinin B/metabolism , Spleen/immunology , Spleen/metabolism , Surface Plasmon Resonance , Tumor Cells, Cultured
15.
Blood ; 111(1): 351-8, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17898315

ABSTRACT

GCET1 (germinal center B cell-expressed transcript-1) gene codes for a serpin expressed in germinal center (GC) B cells. Following the observation that follicular lymphoma cases exhibit an increased level of Gcet1 expression, compared with follicular hyperplasia, we have characterized Gcet1 protein expression in human tissues, cell lines, and a large series of lymphomas. To this end, we have performed immunohistochemical and Western blot analyses using a newly generated monoclonal antibody that is reactive in paraffin-embedded tissues. Our results demonstrate that Gcet1 is expressed exclusively by neoplasms hypothetically to be arrested at the GC stage of differentiation, including follicular lymphoma, nodular lymphocyte predominant Hodgkin lymphoma, and a subset of diffuse large B-cell lymphoma, T-cell/histiocyte rich B-cell lymphoma, and Burkitt lymphoma. Within these tumors, Gcet-1 protein expression is restricted to a subset of GC B cells, establishing the existence of a distinct heterogeneity among normal and neoplastic GC B cells. None of the other B-cell lymphomas, that is, chronic lymphocytic leukemia, splenic marginal zone lymphoma, and mantle cell lymphoma, was Gcet1(+), which underlines the potential utility of Gcet1 expression in lymphoma diagnosis. The results of RNA and protein expression should prompt further investigation into the role of Gcet1 in regulating B-cell survival.


Subject(s)
Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Germinal Center/pathology , Lymphoma/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Serpins/genetics , Serpins/metabolism , Blotting, Western , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Cell Line, Tumor , Hodgkin Disease/genetics , Hodgkin Disease/metabolism , Hodgkin Disease/pathology , Humans , Immunoenzyme Techniques , Lymphoma/genetics , Lymphoma/metabolism , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Lymphoma, Follicular/genetics , Lymphoma, Follicular/metabolism , Lymphoma, Follicular/pathology , Oligonucleotide Array Sequence Analysis
16.
Eur J Immunol ; 35(6): 1681-91, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15902688

ABSTRACT

The transcription factor FOXP3 plays a key role in CD4(+)CD25(+) regulatory T cell function and represents a specific marker for these cells. Despite its strong association with regulatory T cell function, in humans little is known about the frequency of CD4(+)CD25(+) cells that express FOXP3 protein nor the distribution of these cells in vivo. Here we report the characterization of seven anti-FOXP3 monoclonal antibodies enabling the detection of endogenous human FOXP3 protein by flow cytometry and immunohistochemistry. Flow-cytometric analysis showed that FOXP3 was expressed by the majority of CD4(+)CD25(high) T cells in peripheral blood. By contrast, less than half of the CD4(+)CD25(int) population were FOXP3(+), providing an explanation for observations in human T cells that regulatory activity is enriched within the CD4(+)CD25(high) pool. Although FOXP3 expression was primarily restricted to CD4(+)CD25(+) cells, it was induced following activation of both CD4(+) and CD8(+) T cell clones. These findings indicate that the frequency of FOXP3(+) cells correlates with the level of expression of CD25 in naturally arising regulatory T cells and that FOXP3 protein is expressed by some activated CD4(+) and CD8(+) T cell clones. These reagents represent valuable research tools to further investigate FOXP3 function and are applicable for routine clinical use.


Subject(s)
DNA-Binding Proteins/analysis , Receptors, Interleukin-2/analysis , T-Lymphocytes, Regulatory/chemistry , Animals , Antibodies, Monoclonal/immunology , Cross Reactions , DNA-Binding Proteins/immunology , Forkhead Transcription Factors , Humans , Immune Tolerance , Lymphocyte Activation , Mice , Mice, Inbred BALB C , T-Lymphocytes, Regulatory/physiology
17.
Vaccine ; 21(23): 3342-50, 2003 Jul 04.
Article in English | MEDLINE | ID: mdl-12804866

ABSTRACT

The immunogenicity and protective capability of several baculovirus-expressed infectious bursal disease virus (IBDV)-derived assemblies as VP2 capsids, VPX tubules and polyprotein (PP)-derived mixed structures, were tested. Four-week-old chickens were immunised subcutaneously with one dose of each particulate antigen. VP2 icosahedral capsids induced the highest neutralising response, followed by PP-derived structures and then VPX tubules. All vaccinated animals were protected when challenged with a very virulent IBDV (vvIBDV) isolate, however the degree of protection is directly correlated with the levels of neutralising antibodies. VP2 capsids elicited stronger protective immunity than tubular structures and 3 micrograms of them were sufficient to confer a total protection comparable to that induced by an inactivated vaccine. Therefore, VP2 capsids represent a suitable candidate recombinant vaccine instead of virus-like particles (VLPs) for IBDV infections. Our results also provide clear evidence that the recombinant IBDV-derived antigens are structure-dependent in order to be efficient as vaccine components.


Subject(s)
Chickens/immunology , Infectious bursal disease virus/immunology , Viral Vaccines/immunology , Animals , Birnaviridae Infections/immunology , Birnaviridae Infections/prevention & control , Capsid/immunology , Cells, Cultured , Dose-Response Relationship, Immunologic , Immunization , Infectious bursal disease virus/chemistry , Quality Control , Reverse Transcriptase Polymerase Chain Reaction , Spodoptera , Structure-Activity Relationship , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology , Viral Proteins/biosynthesis , Viral Vaccines/chemistry
18.
Vaccine ; 21(17-18): 1952-60, 2003 May 16.
Article in English | MEDLINE | ID: mdl-12706682

ABSTRACT

The immunogenicity and protective capability of several baculovirus-expressed infectious bursal disease virus (IBDV)-derived assemblies as VP2 capsids, VPX tubules and polyprotein (PP)-derived mixed structures, were tested. Four-week-old chickens were immunised subcutaneously with one dose of each particulate antigen. VP2 icosahedral capsids induced the highest neutralising response, followed by PP-derived structures and then VPX tubules. All vaccinated animals were protected when challenged with a very virulent IBDV (vvIBDV) isolate, however the degree of protection is directly correlated with the levels of neutralising antibodies. VP2 capsids elicited stronger protective immunity than tubular structures and 3& mgr;g of them were sufficient to confer a total protection comparable to that induced by an inactivated vaccine. Therefore, VP2 capsids represent a suitable candidate recombinant vaccine instead of virus-like particles (VLPs) for IBDV infections. Our results also provide clear evidence that the recombinant IBDV-derived antigens are structure-dependent in order to be efficient as vaccine components.


Subject(s)
Chickens/immunology , Animals , Birnaviridae Infections/immunology , Birnaviridae Infections/prevention & control , Capsid/immunology , Cells, Cultured , Dose-Response Relationship, Immunologic , Immunization , Infectious bursal disease virus/chemistry , Infectious bursal disease virus/immunology , Parturition , Quality Control , Reverse Transcriptase Polymerase Chain Reaction , Spodoptera , Structure-Activity Relationship , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology , Viral Proteins/biosynthesis , Viral Vaccines/chemistry , Viral Vaccines/immunology
19.
J Virol ; 76(24): 12646-53, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12438590

ABSTRACT

PEPSCAN analysis has been used to characterize the immunogenic regions of the capsid protein (CP) in virions of plum pox potyvirus (PPV). In addition to the well-known highly immunogenic N- and C-terminal domains of CP, regions within the core domain of the protein have also shown high immunogenicity. Moreover, the N terminus of CP is not homogeneously immunogenic, alternatively showing regions frequently recognized by antibodies and others that are not recognized at all. These results have helped us to design efficient antigen presentation vectors based on PPV. As predicted by PEPSCAN analysis, a small displacement of the insertion site in a previously constructed vector, PPV-gamma, turned the derived chimeras into efficient immunogens. Vectors expressing foreign peptides at different positions within a highly immunogenic region (amino acids 43 to 52) in the N-terminal domain of CP were the most effective at inducing specific antibody responses against the foreign sequence.


Subject(s)
Antigen Presentation , Capsid/immunology , Plum Pox Virus/immunology , Animals , Antibodies, Viral/biosynthesis , Capsid/chemistry , Chimera/immunology , Genetic Vectors , Mice , Mice, Inbred BALB C , Virion/immunology
20.
J Gen Virol ; 82(Pt 10): 2415-2424, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11562535

ABSTRACT

The antigenic structure of African horse sickness virus (AHSV) serotype 4 capsid protein VP2 has been determined at the peptide level by PEPSCAN analysis in combination with a large collection of polyclonal antisera and monoclonal antibodies. VP2, the determinant for the virus serotype and an important target in virus neutralization, was found to contain 15 antigenic sites. A major antigenic region containing 12 of the 15 sites was identified in the region between residues 223 and 400. A second domain between residues 568 and 681 contained the three remaining sites. These sites were used for the synthesis of peptides, which were later tested in rabbits. Of the 15 synthetic peptides, three were able to induce neutralizing antibodies for AHSV-4, defining two neutralizing epitopes, 'a' and 'b', between residues 321 and 339, and 377 and 400, respectively. A combination of peptides representing both sites induced a more effective neutralizing response. Still, the relatively low neutralization titres make the possibility of producing a synthetic vaccine for AHSV unlikely. The complex protein-protein interaction of the outer shell of the viral capsid would probably require the presence of either synthetic peptides in the correct conformation or peptide segments from the different proteins VP2, VP5 and VP7.


Subject(s)
African Horse Sickness Virus/immunology , Capsid/immunology , Peptide Fragments/immunology , Amino Acid Sequence , Animals , Capsid Proteins , Epitopes , Horses , Molecular Sequence Data , Neutralization Tests , Rabbits , Serotyping
SELECTION OF CITATIONS
SEARCH DETAIL
...