Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7(1): 17049, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29213127

ABSTRACT

Engagement of Fcγ-receptors triggers a range of downstream signalling events resulting in a diverse array of immune functions. As a result, blockade of Fc-mediated function is an important strategy for the control of several autoimmune and inflammatory conditions. We have generated a hexameric-Fc fusion protein (hexameric-Fc) and tested the consequences of multi-valent Fcγ-receptor engagement in in vitro and in vivo systems. In vitro engagement of hexameric-Fc with FcγRs showed complex binding interactions that altered with receptor density and triggered the internalisation and degradation of Fcγ-receptors. This caused a disruption of Fc-binding and phagocytosis. In vivo, in a mouse ITP model we observed a short half-life of hexameric-Fc but were nevertheless able to observe inhibition of platelet phagocytosis several days after hexameric-Fc dosing. In cynomolgus monkeys, we again observed a short half-life, but were able to demonstrate effective FcγR blockade. These findings demonstrate the ability of multi-valent Fc-based therapeutics to interfere with FcγR function and a potential mechanism through which they could have a sustained effect; the internalisation and degradation of FcγRs.


Subject(s)
Immunoglobulin Fc Fragments/metabolism , Receptors, IgG/metabolism , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Cytokines/metabolism , Disease Models, Animal , HEK293 Cells , Half-Life , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Macaca fascicularis , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Transgenic , Phagocytosis , Purpura, Thrombocytopenic, Idiopathic/metabolism , Purpura, Thrombocytopenic, Idiopathic/pathology , Receptors, IgG/chemistry , Receptors, IgG/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacokinetics
2.
Regul Toxicol Pharmacol ; 75: 72-80, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26743742

ABSTRACT

Profound immunosuppression (e.g., AIDS, transplant therapy) is epidemiologically associated with an increased cancer risk, and often with oncogenic viruses. It is currently unclear how broadly this association translates to therapeutics that modulate immunity. A workshop co-sponsored by the FDA and HESI examined how perturbing the immune system may contribute to carcinogenesis, and highlighted priorities for improving non-clinical risk assessment of targeted immunomodulatory therapies. Conclusions from the workshop were as follows. 1) While profound altered immunity can promote tumorigenesis, not all components of the immune system are equally important in defense against or promotion of cancer and a similar cancer risk for all immunomodulatory molecules should not be assumed. 2) Rodent carcinogenicity studies have limitations and are generally not reliable predictors of cancer risk associated with immunosuppression. 3) Cancer risk needs to be evaluated based on mechanism-based weight-of-evidence, including data from immune function tests most relevant to tumor immunosurveillance or promotion. 4) Information from nonclinical experiments, clinical epidemiology and immunomodulatory therapeutics show that immunosurveillance involves a complex network of cells and mediators. To support a weight-of-evidence approach, an increased focus on understanding the quantitative relationship between changes in relevant immune function tests and cancer risk is needed.


Subject(s)
Immunologic Factors/adverse effects , Neoplasms/chemically induced , Animals , Humans , Neoplasms/epidemiology , Neoplasms/immunology , Risk Assessment/legislation & jurisprudence , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
3.
MAbs ; 7(5): 829-37, 2015.
Article in English | MEDLINE | ID: mdl-26230385

ABSTRACT

QBP359 is an IgG1 human monoclonal antibody that binds with high affinity to human CCL21, a chemokine hypothesized to play a role in inflammatory disease conditions through activation of resident CCR7-expressing fibroblasts/myofibroblasts. The pharmacokinetics (PK) and pharmacodynamics (PD) of QBP359 in non-human primates were characterized through an integrated approach, combining PK, PD, immunogenicity, immunohistochemistry (IHC) and tissue profiling data from single- and multiple-dose experiments in cynomolgus monkeys. When compared with regular immunoglobulin typical kinetics, faster drug clearance was observed in serum following intravenous administration of 10 mg/kg and 50 mg/kg of QBP359. We have shown by means of PK/PD modeling that clearance of mAb-ligand complex is the most likely explanation for the rapid clearance of QBP359 in cynomolgus monkey. IHC and liquid chromatography mass spectrometry data suggested a high turnover and synthesis rate of CCL21 in tissues. Although lymphoid tissue was expected to accumulate drug due to the high levels of CCL21 present, bioavailability following subcutaneous administration in monkeys was 52%. In human disease states, where CCL21 expression is believed to be expressed at 10-fold higher concentrations compared with cynomolgus monkeys, the PK/PD model of QBP359 and its binding to CCL21 suggested that very large doses requiring frequent administration of mAb would be required to maintain suppression of CCL21 in the clinical setting. This highlights the difficulty in targeting soluble proteins with high synthesis rates.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Chemokine CCL21/antagonists & inhibitors , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Animals , Antibody Affinity , Chromatography, Liquid , Humans , Immunohistochemistry , Macaca fascicularis , Mass Spectrometry
4.
Clin Pharmacol Ther ; 85(3): 247-58, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19177065

ABSTRACT

Modulating immune responses with monoclonal antibodies (mAbs) that target immune molecules has become a promising therapeutic strategy and is under investigation for the treatment of cancer and (auto)-immune diseases. A major hurdle to the development and early clinical investigation of many immunomodulatory mAbs is the inherent risk of adverse immune-mediated drug reactions in humans, such as cytokine storms, autoimmunity, and immunosuppression. Dose selection for first-in-human (FIH) clinical trials involving immunomodulatory mAbs, and mAbs in general, is based on specifically designed preclinical safety studies, primarily in nonhuman primates (NHPs), and on mechanistic ex vivo investigations. Dose selection in such trials is challenging for a number of reasons related to safety. In this context, safety-relevant differences between NHP and human immune systems, species selection/qualification and preclinical study design considerations, the receptor occupancy model and its calculation, the minimal anticipated biological effect level (MABEL) and its use in the selection of a safe starting dose in humans, microdosing and the impact of immunogenicity on safety assessment of mAbs, and safety-relevant formulation properties of therapeutic mAbs are critically reviewed. In addition, the current regulatory requirements are presented and discussed to demonstrate how the TeGenero TGN1412 case is leading to increased regulatory scrutiny regarding dose selection for FIH clinical trials.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Clinical Trials as Topic/standards , Drug-Related Side Effects and Adverse Reactions , Immunologic Factors/administration & dosage , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/physiology , Clinical Trials as Topic/adverse effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/standards , Humans , Immunologic Factors/adverse effects , Immunologic Factors/physiology , Practice Guidelines as Topic
5.
Vaccine ; 21(19-20): 2441-7, 2003 Jun 02.
Article in English | MEDLINE | ID: mdl-12744877

ABSTRACT

Expression of a 17-mer peptide sequence from canine parvovirus expressed on cowpea mosaic virus (CPMV) to form chimaeric virus particles (CVPs) creates vaccine antigens that elicit strong anti-peptide immune responses in mice. Systemic (subcutaneous, s.c.) immunisation and boosting with such CVP constructs produces IgG(2a) serum antibody responses, while mucosal (intranasal, i.n.) immunisation and boosting elicits intestinal IgA responses. Combinations of systemic and mucosal routes for priming and boosting immunisations were used to examine their influence on the level, type and location of immune response generated to one of these constructs (CVP-1). In all cases, s.c. administration, whether for immunisation or boosting, generated a Th1-biased response, reflected in a predominantly IgG(2a) serum antibody isotype and secretion of IFN-gamma from in vitro-stimulated lymphocytes. Serum antibody responses were greatest in animals primed and boosted subcutaneously, and least in mucosally vaccinated mice. The i.n. exposure also led to IFN-gamma release from in vitro-stimulated cells, but serum IgG(2a) was significantly elevated only in mice primed intranasally and boosted subcutaneously. Peptide- and wild-type CPMV-specific IgA responses in gut lavage fluid were greatest in animals exposed mucosally and least in those primed and boosted subcutaneously or primed subcutaneously and boosted orally. Lymphocytes from immunised mice proliferated in response to in vitro stimulation with CPMV but not with peptide. The predominant secretion of IFN-gamma from all immunising/boosting combinations indicates that the route of vaccination and challenge does not alter the Th1 bias of the response to CVP constructs. However, optimal serum and intestinal antibody responses were achieved by combining s.c. and i.n. administration.


Subject(s)
Dog Diseases/immunology , Immunization, Secondary/methods , Parvoviridae Infections/veterinary , Parvovirus/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Antibody Formation , Dogs , Immunity, Mucosal , Mice , Molecular Sequence Data , Parvoviridae Infections/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , Plant Viruses/immunology
6.
Vaccine ; 20(21-22): 2727-34, 2002 Jun 21.
Article in English | MEDLINE | ID: mdl-12034099

ABSTRACT

NIH mice were vaccinated subcutaneously or intranasally with chimaeric cow pea mosaic virus (CPMV) constructs expressing a 17-mer peptide sequence from canine parvovirus (CPV) as monomers or dimers on the small or large protein surface subunits. Responses to the chimaeric virus particles (CVPs) were compared with those of mice immunized with the native virus or with parvovirus peptide conjugated to keyhole limpet haemocyanin (KLH). The characteristics of the immune response to vaccination were examined by measuring serum and mucosal antibody responses in ELISA, in vitro antigen-induced spleen cell proliferation and cytokine responses. Mice made strong antibody responses to the native plant virus and peptide-specific responses to two of the four CVP constructs tested which were approximately 10-fold lower than responses to native plant virus. The immune response generated by the CVP constructs showed a marked TH1 bias, as determined by a predominantly IgG(2a) isotype peptide-specific antibody response and the release of IFN-gamma but not IL-4 or IL-5 from lymphocytes exposed to antigen in vitro. In comparison, parvovirus peptide conjugated to KLH generated an IgG(1)-biased (TH2) response. These data indicate that the presentation of peptides on viral particles could be used to bias the immune response in favor of a TH1 response.Anti-viral and anti-peptide IgA were detected in intestinal and bronchial lavage fluid of immunized mice, demonstrating that a mucosal immune response to CPV can be generated by systemic and mucosal immunization with CVP vaccines. Serum antibody from both subcutaneously-vaccinated and intranasally-vaccinated mice showed neutralizing activity against CPV in vitro.


Subject(s)
Parvovirus, Canine/immunology , Viral Vaccines/administration & dosage , Administration, Intranasal , Amino Acid Sequence , Animals , Antibody Specificity , Comovirus/genetics , Comovirus/immunology , Enzyme-Linked Immunosorbent Assay , Female , Immunity, Mucosal , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Injections, Subcutaneous , Mice , Molecular Sequence Data , Neutralization Tests , Plant Viruses/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Vaccines/immunology
7.
Vaccine ; 19(27): 3661-70, 2001 Jun 14.
Article in English | MEDLINE | ID: mdl-11395200

ABSTRACT

A vaccine based upon a recombinant plant virus (CPMV-PARVO1), displaying a peptide derived from the VP2 capsid protein of canine parvovirus (CPV), has previously been described. To date, studies with the vaccine have utilized viable plant chimaeric particles (CVPs). In this study, CPMV-PARVO1 was inactivated by UV treatment to remove the possibility of replication of the recombinant plant virus in a plant host after manufacture of the vaccine. We show that the inactivated CVP is able to protect dogs from a lethal challenge with CPV following parenteral immunization with the vaccine. Dogs immunized with the inactivated CPMV-PARVO1 in adjuvant displayed no clinical signs of disease and shedding of CPV in faeces was limited following CPV challenge. All immunized dogs elicited high titres of peptide-specific antibody, which neutralized CPV in vitro. Levels of protection, virus shedding and VP2-specific antibody were comparable to those seen in dogs immunized with the same VP2- peptide coupled to keyhole limpet hemocyanin (KLH). Since plant virus-derived vaccines have the potential for cost-effective manufacture and are not known to replicate in mammalian cells, they represent a viable alternative to current replicating vaccine vectors for development of both human and veterinary vaccines.


Subject(s)
Comovirus/immunology , Parvoviridae Infections/prevention & control , Parvovirus, Canine/immunology , Recombinant Proteins/therapeutic use , Viral Proteins/therapeutic use , Viral Vaccines/therapeutic use , Amino Acid Sequence , Animals , Capsid/therapeutic use , Capsid Proteins , Comovirus/radiation effects , Dog Diseases/prevention & control , Dog Diseases/virology , Dogs , Immunization Schedule , Molecular Sequence Data , Parvoviridae Infections/mortality , Parvoviridae Infections/veterinary , Parvovirus, Canine/radiation effects , Ultraviolet Rays , Vaccines, Inactivated/therapeutic use , Vaccines, Synthetic/therapeutic use
8.
Vaccine ; 19(25-26): 3376-83, 2001 May 14.
Article in English | MEDLINE | ID: mdl-11348701

ABSTRACT

Staphylococcus aureus bacteraemia (SAB) originating from local infections can lead to severe secondary infections such as endocarditis. The protective effect of antibodies against secondary infections was studied in a rat model, where a local joint infection leads to bacteraemia and endocarditis on damaged aortic valves. In this study, immunizations with a truncated D2-domain of the S. aureus fibronectin-binding protein displayed on a cow-pea mosaic virus (CPMV-D) carrier induced protection against endocarditis (P < 0.05). Opsonization of S. aureus with antibodies raised against CPMV-D stimulated both neutrophil activity and macrophage phagocytosis in vitro. Furthermore, intravenous administration of these antibodies protected mice from weight loss due to SAB.


Subject(s)
Adhesins, Bacterial , Bacterial Proteins/immunology , Carrier Proteins/immunology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/immunology , Amino Acid Sequence , Animals , Antibodies, Bacterial/biosynthesis , Antibody Specificity , Bacteremia/immunology , Bacteremia/prevention & control , Bacterial Proteins/genetics , Bacterial Vaccines/genetics , Bacterial Vaccines/pharmacology , Carrier Proteins/genetics , Chimera/genetics , Comovirus/genetics , Endocarditis, Bacterial/immunology , Endocarditis, Bacterial/prevention & control , Female , Molecular Sequence Data , Opsonin Proteins/immunology , Peptide Fragments/genetics , Peptide Fragments/immunology , Rats , Rats, Wistar , Staphylococcal Infections/immunology , Staphylococcus aureus/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/pharmacology
9.
Mol Biotechnol ; 17(1): 15-26, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11280928

ABSTRACT

The plant virus, cowpea mosaic virus (CPMV), has been developed as an expression and presentation system to display antigenic epitopes derived from a number of vaccine targets including infectious disease agents and tumors. These chimeric virus particles (CVPs) could represent a cost-effective and safe alternative to live replicating virus and bacterial vaccines. A number of CVPs have now been generated and their immunogenicity examined in a number of animal species. This review details the humoral and cellular immune responses generated by these CVPs following both parenteral and mucosal delivery and highlights the potential of CVPs to elicit protective immunity from both viral and bacterial infection.


Subject(s)
Antigens/metabolism , Comovirus/genetics , Comovirus/immunology , Animals , Capsid/chemistry , Comovirus/chemistry , Epitopes , Humans , Immune System/virology , Peptides/chemistry , Plants/virology , Vaccines/chemistry
10.
J Neuroimmunol ; 106(1-2): 60-8, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10814783

ABSTRACT

Migration of cells into the central nervous system (CNS) is a pivotal step in the pathogenesis of immune-mediated diseases such as multiple sclerosis (MS), experimental allergic encephalomyelitis (EAE) and virus-induced demyelinating diseases. Such migration is dependent on expression of adhesion molecules. The expression of adhesion molecules in the CNS was studied in Biozzi ABH mice infected with Semliki Forest virus (SFV) A7(74) - an important demyelinating model of MS. Expression of LFA-1alpha/CD11a, LFA-1beta/CD18 and ICAM-1/CD56 were rapidly elevated and remained high whereas MAC-1, CD44 and VCAM-1/CD106 were less widely expressed. The alpha4-integrin VLA-4/CD49d was more specifically associated with CNS lesions. To identify the importance of VLA-4, CD44, ICAM-1 and MAC-1 in the pathogenesis of SFV infection, monoclonal antibodies that block these adhesion molecules were administered in vivo during infection. Anti-VLA-4 treatment dramatically reduced the cellular infiltrates and demyelination within the CNS but did not affect the clearance of virus while antibodies to CD44, ICAM and MAC-1 antibody treatment had no effect. This study demonstrates that SFV infection induces the expression of adhesion molecules within the CNS and that VLA-4 plays an important role in the development of inflammation and demyelination in the CNS following SFV infection.


Subject(s)
Alphavirus Infections/physiopathology , Antigens, CD/physiology , Semliki forest virus , Alphavirus Infections/drug therapy , Alphavirus Infections/metabolism , Alphavirus Infections/pathology , Animals , Antibodies/therapeutic use , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Central Nervous System/metabolism , Immunohistochemistry , Integrin alpha4 , Mice , Mice, Inbred Strains , Myelin Sheath/pathology
11.
FEMS Immunol Med Microbiol ; 27(4): 291-7, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10727884

ABSTRACT

Outer membrane protein F of Pseudomonas aeruginosa has vaccine efficacy against infection by P. aeruginosa as demonstrated in a variety of animal models. Through the use of synthetic peptides, three surface-exposed epitopes have been identified. These are called peptides 9 (aa 261-274 in the mature F protein, TDAYNQKLSERRAN), 10 (aa 305-318, NATAEGRAINRRVE), and 18 (aa 282-295, NEYGVEGGRVNAVG). Both the peptide 9 and 10 epitopes are protective when administered as a vaccine. In order to develop a vaccine that is suitable for use in humans, including infants with cystic fibrosis, the use of viral vector systems to present the protective epitopes has been investigated. An 11-amino acid portion of epitope 10 (AEGRAINRRVE) was successfully inserted into the antigenic B site of the hemagglutinin on the surface of influenza virus. This chimeric influenza virus protects against challenge with P. aeruginosa in the mouse model of chronic pulmonary infection. Attempts to derive a chimeric influenza virus carrying epitope 9 have been unsuccessful. A chimeric plant virus, cowpea mosaic virus (CPMV), with epitopes 18 and 10 expressed in tandem on the large coat protein subunit (CPMV-PAE5) was found to elicit antibodies that reacted exclusively with the 10 epitope and not with epitope 18. Use of this chimeric virus as a vaccine afforded protection against challenge with P. aeruginosa in the mouse model of chronic pulmonary infection. Chimeric CPMVs with a single peptide containing epitopes 9 and 18 expressed on either of the coat proteins are in the process of being evaluated. Epitope 9 was successfully expressed on the coat protein of tobacco mosaic virus (TMV), and this chimeric virus is protective when used as a vaccine in the mouse model of chronic pulmonary infection. However, initial attempts to express epitope 10 on the coat protein of TMV have been unsuccessful. Efforts are continuing to construct chimeric viruses that express both the 9 and 10 epitopes in the same virus vector system. Ideally, the use of a vaccine containing two epitopes of protein F is desirable in order to greatly reduce the likelihood of selecting a variant of P. aeruginosa that escapes protective antibodies in immunized humans via a mutation in a single epitope within protein F. When the chimeric influenza virus containing epitope 10 and the chimeric TMV containing epitope 9 were given together as a combined vaccine, the immunized mice produced antibodies directed toward both epitopes 9 and 10. The combined vaccine afforded protection against challenge with P. aeruginosa in the chronic pulmonary infection model at approximately the same level of efficacy as provided by the individual chimeric virus vaccines. These results prove in principle that a combined chimeric viral vaccine presenting both epitopes 9 and 10 of protein F has vaccine potential warranting continued development into a vaccine for use in humans.


Subject(s)
Bacterial Vaccines/immunology , Influenza A virus/genetics , Lung Diseases/prevention & control , Plant Viruses/genetics , Porins/immunology , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/immunology , Animals , Antibodies, Bacterial/blood , Bacterial Vaccines/administration & dosage , Comovirus/genetics , Comovirus/metabolism , Enzyme-Linked Immunosorbent Assay , Epitopes/genetics , Epitopes/immunology , Epitopes/metabolism , Influenza A virus/metabolism , Lung/microbiology , Lung Diseases/microbiology , Mice , Plant Viruses/metabolism , Porins/chemistry , Porins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Tobacco Mosaic Virus/genetics , Tobacco Mosaic Virus/metabolism , Vaccination , Vaccines, Combined/administration & dosage , Vaccines, Combined/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
12.
Immunology ; 98(3): 427-35, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10583604

ABSTRACT

Clinical signs of experimental autoimmune encephalomyelitis (EAE) are associated with the selective recruitment of CD4+ memory (CD45RBlow CD44high) T cells into the central nervous system (CNS). However, we have found that many of these recently recruited memory cells are CD44low, suggesting that the CD44 antigen may be involved in, and transiently lost during, the extravasation process. Indeed, administration of a CD44-specific antibody (IM7.8.1) induced leucocyte CD44 shedding and both prevented the development and ameliorated the severity of established EAE by inhibiting mononuclear cell infiltration into the CNS. Trafficking of cells into lymph nodes, however, a property mainly of naïve cells, was essentially unaffected. In contrast, treatment with antibody to very late activation antigen-4 (VLA-4) prevented homing to both the CNS and to lymph nodes. This study contests previous reports that dismissed a role for CD44 in inflammation of the CNS and, coupled with observations in murine dermatitis and arthritis, suggests that CD44 is involved in the homing of primed lymphocytes to sites of inflammation. CD44 should therefore be considered a target for immunotherapy of T-cell-mediated inflammatory diseases, such as multiple sclerosis.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Hyaluronan Receptors/immunology , Leukocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Central Nervous System/immunology , Chronic Disease , Immunologic Memory , Integrin alpha4beta1 , Integrins/immunology , Lymph Nodes/immunology , Male , Mice , Mice, Inbred BALB C , Receptors, Lymphocyte Homing/immunology , Secondary Prevention , Statistics, Nonparametric
13.
Immunology ; 97(3): 533-9, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10447778

ABSTRACT

The hyaluronic acid binding glycoprotein CD44 is expressed on a wide variety of cells, and by mediating interactions between cells and extracellular matrices promotes the movement of cells from the circulation into organs. Recent reports have described the effects of an antibody specific for CD44 (IM7) that has beneficial effects in two murine models of autoimmune disease. Both experimental allergic encephalomyelitis (EAE) and collagen-induced arthritis were ameliorated by treatment with IM7, which was considered to be acting by preventing the homing of lymphocytes to the relevant inflammatory sites, namely the central nervous system and the synovium, respectively. In this study the same anti-CD44 antibody was used to try to prevent leucocytic infiltration of the thyroid in the murine model of Hashimoto's thyroiditis, experimental autoimmune thyroiditis (EAT). We report that, in contrast to the previous findings, this antibody had an exacerbating effect on thyroiditis induced by immunization of mice with mouse thyroglobulin (MTg) and complete Freund's adjuvant (CFA). Thyroid infiltrates lasted longer and showed increased severity compared with untreated or control antibody-treated mice. Antibody responses to MTg were unaffected by antibody treatment. The data suggest that simple rules cannot be drawn that predict the potential broad therapeutic use of anti-CD44 reagents, presumably due to differences in the cellular phenotypes and the dynamics of their movement into inflammatory sites during different disease processes.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hyaluronan Receptors/immunology , T-Lymphocytes/immunology , Thyroid Gland/immunology , Thyroiditis, Autoimmune/immunology , Animals , Antibodies, Monoclonal/adverse effects , Autoantibodies/blood , Cell Division/immunology , Cell Movement/immunology , Female , Lymph Nodes/immunology , Mice , Mice, Inbred CBA , Spleen/immunology , Thyroglobulin/immunology
14.
Vaccine ; 17(15-16): 1846-57, 1999 Apr 09.
Article in English | MEDLINE | ID: mdl-10217582

ABSTRACT

The D2 peptide derived from an S. aureus fibronectin-binding protein (FnBP) was expressed on the surface of the icosahedral cowpea mosaic virus (amino acids 1-30 of D2) or on the rod-shaped potato virus X (amino acids 1-38 of D2), termed CPMV-MAST1 and PVX-MAST8, respectively. Mice and rats were immunized subcutaneously with CPMV-MAST1 and mice with PVX-MAST8 in adjuvant and high titres of FnBP-specific antibody were obtained. The mouse IgG was predominantly of the IgG2a and IgG2b isotypes, which strongly bound complement component C1q, suggesting a TH1-bias in the peptide-specific responses. Sera from mice and rats immunized with CPMV-MAST1 and from mice immunized with PVX-MAST8 were shown to completely inhibit the binding of fibronectin to immobilised recombinant FnBP and rat sera against CPMV-MAST1 were able to block adherence of S. aureus to fibronectin. These studies demonstrate that the D2 peptide is highly immunogenic when expressed on 2 different plant viruses and highlight the potential of plant virus-based vaccines to protect against S. aureus infections.


Subject(s)
Adhesins, Bacterial , Antibodies, Bacterial/biosynthesis , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Carrier Proteins/immunology , Plant Viruses , Staphylococcus aureus/immunology , Vaccines, Synthetic/immunology , Amino Acid Sequence , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Vaccines/genetics , Binding, Competitive , Carrier Proteins/genetics , Carrier Proteins/metabolism , Comovirus/genetics , Complement C1q/immunology , Complement C1q/metabolism , Female , Fibronectins/metabolism , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/immunology , Plant Viruses/genetics , Potexvirus/genetics , Protein Binding , Rats , Staphylococcus aureus/genetics , Vaccination , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/genetics , Vaccines, Synthetic/isolation & purification
15.
Microbiology (Reading) ; 145 ( Pt 1): 211-220, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10206701

ABSTRACT

A synthetic peptide (peptide 10) representing a surface-exposed, linear B cell epitope from outer-membrane (OM) protein F of Pseudomonas aeruginosa was shown previously to afford protection in mice from P. aeruginosa infection. This peptide was expressed in tandem with the protein F peptide 18 on each of the two coat proteins of cowpea mosaic virus (CPMV). The chimaeric virus particles (CVPs) expressing the peptides on the S (small) coat protein (CPMV-PAE4) and L (large) coat protein (CPMV-PAE5) were used to immunize mice. Following subcutaneous immunization in Freund's and QuilA adjuvants, CPMV-PAE4 induced antibodies predominantly against peptide 18, whereas CPMV-PAE5 produced antibodies exclusively against peptide 10, indicating that the site of peptide expression on CPMV influences its immune recognition. The anti-peptide antibodies elicited by CPMV-PAE5 were predominantly of the IgG2a isotype, indicating a highly polarized TH1-type response. The peptide-specific IgG2a strongly recognized the whole F protein, but more importantly, recognized protein F in all seven Fisher-Devlin immunotypes of P. aeruginosa. Furthermore, the peptide-specific IgG2a in CVP/QS-21 adjuvant-immunized mice was shown to bind complement and to augment phagocytosis of P. aeruginosa by human neutrophils in vitro. The ability of CPMV-PAE5 to induce P. aeruginosa-specific opsonic IgG2a gives it potential for further development as a protective vaccine against P. aeruginosa.


Subject(s)
Antibodies, Bacterial/biosynthesis , Comovirus/genetics , Epitopes/immunology , Porins/immunology , Pseudomonas aeruginosa/immunology , Animals , Capsid/biosynthesis , Capsid/genetics , Comovirus/immunology , Complement System Proteins/immunology , Epitopes/genetics , Genetic Vectors/genetics , Humans , Immunization , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/immunology , Opsonin Proteins/blood , Opsonin Proteins/immunology , Phagocytosis , Porins/genetics , Pseudomonas Infections/classification , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas Infections/prevention & control , Th1 Cells/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
16.
Vaccine ; 17(11-12): 1359-68, 1999 Mar 17.
Article in English | MEDLINE | ID: mdl-10195771

ABSTRACT

The ability of five different adjuvants (alum, complete Freund's adjuvant, Quil A, AdjuPrime and Ribi) to stimulate humoral and T-cell mediated immune responses against a purified chimeric virus particle was investigated. Each adjuvant was administered subcutaneously to adult mice together with 10 microg of wildtype (wt) cowpea mosaic virus (CPMV) or a chimeric CPMV displaying the HIV-1 gp41 peptide, residues 731-752. All preparations elicited strong antibody responses to CPMV, but Quil A elicited the highest and most consistent responses to the HIV-1 peptide. This finding was reflected in both ELISA titres with immobilized peptide and in HIV-1-neutralizing antibody. In addition Quil A was also, the only adjuvant to stimulate an in vitro proliferative T-cell response. Surprisingly with all adjuvant formulations a predominately IgG2a anti-gp41 peptide response was observed, indicating a type 1 T-helper cell-like response. Furthermore, the efficiency of the CPMV display system was demonstrated by its ability to induce good levels of peptide specific antibody in the absence of any adjuvant.


Subject(s)
Adjuvants, Immunologic , Comovirus/immunology , HIV Envelope Protein gp41/immunology , Viral Vaccines/immunology , Alum Compounds , Animals , Antibodies, Viral/biosynthesis , Cell Wall Skeleton/immunology , Chimera , Comovirus/genetics , Cord Factors/immunology , Freund's Adjuvant/immunology , Immunization, Secondary , Lipid A/analogs & derivatives , Lipid A/immunology , Mice , Quillaja Saponins , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Saponins/immunology , T-Lymphocytes/immunology
17.
J Virol ; 73(2): 930-8, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9882293

ABSTRACT

The humoral immune responses to the D2 peptide of fibronectin-binding protein B (FnBP) of Staphylococcus aureus, expressed on the plant virus cowpea mosaic virus (CPMV), were evaluated after mucosal delivery to mice. Intranasal immunization of these chimeric virus particles (CVPs), either alone or in the presence of ISCOM matrix, primed CPMV-specific T cells and generated high titers of CPMV- and FnBP-specific immunoglobulin G (IgG) in sera. Furthermore, CPMV- and FnBP-specific IgA and IgG could also be detected in the bronchial, intestinal, and vaginal lavage fluids, highlighting the ability of CVPs to generate antibody at distant mucosal sites. IgG2a and IgG2b were the dominant IgG subclasses in sera to both CPMV and FnBP, demonstrating a bias in the response toward the T helper 1 type. The sera completely inhibited the binding of human fibronectin to the S. aureus FnBP. Oral immunization of the CVPs also generated CPMV- and FnBP-specific serum IgG; however, these titers were significantly lower and more variable than those generated by the intranasal route, and FnBP-specific intestinal IgA was undetectable. Neither the ISCOM matrix nor cholera toxin enhanced these responses. These studies demonstrate for the first time that recombinant plant viruses have potential as mucosal vaccines without the requirement for adjuvant and that the nasal route is most effective for the delivery of these nonreplicating particles.


Subject(s)
Adhesins, Bacterial , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Carrier Proteins/immunology , Comovirus/immunology , Fibronectins/metabolism , Immunity, Mucosal , Staphylococcus aureus/immunology , Adjuvants, Immunologic , Administration, Intranasal , Administration, Oral , Amino Acid Sequence , Animals , Antibodies, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Vaccines/genetics , Carrier Proteins/genetics , Cholera Toxin/immunology , Comovirus/genetics , Female , Humans , ISCOMs/immunology , Immunoglobulin G/blood , Immunoglobulin G/classification , Immunoglobulin G/immunology , Intestines/immunology , Lung/immunology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Staphylococcus aureus/genetics , T-Lymphocytes/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vagina/immunology , Virion
18.
J Immunol Methods ; 220(1-2): 93-103, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9839930

ABSTRACT

Control of pandemic human immunodeficiency virus type 1 (HIV-1) infection ideally requires specific mucosal immunity to protect the genital regions through which transmission more often occurs. Thus a vaccine that stimulates a disseminated mucosal and systemic protective immune response would be extremely useful. Here we have investigated the ability of a chimeric plant virus, cowpea mosaic virus (CPMV), expressing a 22 amino acid peptide (residues 731-752) of the transmembrane gp41 protein of HIV-1 IIIB (CPMV-HIV/1), to stimulate HIV-1-specific and CPMV-specific mucosal and serum antibody following intranasal or oral immunization together with the widely used mucosal adjuvant, cholera toxin. CPMV-HIV/1 has been shown previously to stimulate HIV-1-specific serum antibody in mice by parenteral immunization. All mice immunized intranasally with two doses of 10 microg of CPMV-HIV/1 produced both HIV-1-specific IgA in faeces as well as higher levels of specific, predominantly IgG2a, serum antibody. Thus there was a predominantly T helper 1 cell response. All mice also responded strongly to CPMV epitopes. Oral immunization of the chimeric cowpea mosaic virus was less effective, even at doses of 500 microg or greater, and stimulated HIV-1-specific serum antibody in only a minority of mice, and no faecal HIV-1 specific IgA.


Subject(s)
AIDS Vaccines/administration & dosage , Comovirus/genetics , Genetic Vectors/genetics , HIV Antibodies/biosynthesis , HIV Envelope Protein gp41/immunology , HIV-1/immunology , Immunization/methods , Immunoglobulin A/biosynthesis , Immunoglobulin G/biosynthesis , Peptide Fragments/immunology , Vaccines, Synthetic/administration & dosage , Adjuvants, Immunologic , Administration, Intranasal , Administration, Oral , Amino Acid Sequence , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Enzyme-Linked Immunosorbent Assay , Feces/chemistry , HIV Antibodies/analysis , HIV Antibodies/blood , HIV Antibodies/immunology , HIV Envelope Protein gp41/genetics , HIV-1/genetics , Immunoglobulin A/analysis , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/analysis , Immunoglobulin G/blood , Immunoglobulin G/immunology , Intestinal Mucosa/immunology , Mice , Mice, Inbred C3H , Molecular Sequence Data , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Recombinant Fusion Proteins/immunology , Vaccines, Synthetic/immunology
19.
Scand J Immunol ; 45(2): 213-20, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9042434

ABSTRACT

Anti-CD44 MoAb IM7 induced the loss of CD44 from mouse leucocytes thereby inhibiting leucocyte migration and joint inflammation in murine arthritis. Thus, targeting CD44 with MoAb may have potential for the treatment of patients with inflammatory joint diseases. Expression of CD44 by peripheral blood (PB) and synovial fluid (SF) leucocytes from rheumatoid arthritis (RA) patients was compared and the ability of IM7 to modulate this expression determined. RASF lymphocytes showed increased CD44 expression compared with those in PB indicative of an activated phenotype. As inflammatory SF did not up-regulate CD44 expression on PB lymphocytes, the increased CD44 expression by SF lymphocytes was a result of the selective homing of CD44(high) cells to the synovium rather than an effect of the synovial environment. RASF granulocytes showed reduced CD44 expression compared with those in PB, again indicative of an activated phenotype. However, this reduction could be induced on PB granulocytes following culture with inflammatory SF and was inhibited by anti-TNF-alpha MoAb, implying that soluble factors in inflammatory SF such as TNF-alpha induced granulocyte activation and CD44 loss. IM7 induced the loss of CD44 from lymphocytes (both from PB and SF) and granulocytes in vitro, but was subsequently re-expressed after 24 h culture in the absence of the MoAb. This loss of CD44 was blocked by serine- and metalloprotease inhibitors implying that IM7 induced the proteolytic cleavage of CD44 by a mechanism similar to that reported for the loss of CD44 from PMA-activated granulocytes. Furthermore, IM7-treated CD44(low) lymphocytes showed reduced adherence to both an endothelial cell line and RA synovial fibroblasts in vitro. The unique ability of IM7 to reduce CD44 expression by lymphocytes suggests that it could prevent lymphocyte extravasation and synovial infiltration in RA as previously reported in murine arthritis.


Subject(s)
Antibodies, Monoclonal/physiology , Arthritis, Rheumatoid/immunology , Granulocytes/immunology , Hyaluronan Receptors/biosynthesis , Lymphocytes/immunology , Synovial Membrane/immunology , Cell Adhesion/immunology , Cells, Cultured , Endothelium/cytology , Endothelium/immunology , Fibroblasts/immunology , Flow Cytometry , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Metalloendopeptidases/antagonists & inhibitors , Serine/antagonists & inhibitors , Synovial Fluid/immunology , Synovial Membrane/cytology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
20.
J Orthop Res ; 15(1): 40-9, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9066525

ABSTRACT

The responses of human peripheral blood monocytes of 10 normal volunteers and 14 patients with total hip replacements to particles of commercially pure titanium and chromium orthophosphate (a corrosion product from cobalt-chromium alloy implants) were studied. In addition, these phagocytosable particles were added to cultured mononuclear cells isolated from the interfacial membrane of 14 patients with failed implants. Peripheral blood monocytes from patients who had had a total hip replacement produced significantly higher levels of interleukin-1 (both interleukin-1 alpha and interleukin-1 beta) and prostaglandin E2 following particulate stimulation than those from normal volunteers. Supernatants from both titanium and chromium orthophosphate-stimulated peripheral blood monocytes from the volunteers and patients with total hip replacement induced bone resorption (assayed in organ cultures of newborn mouse calvariae) and the proliferation of human fibroblasts. The levels of bone resorption were significantly higher (p < 0.05) in patients with implants than in normal volunteers. There were no significant differences in the responses of cells between patients with focal osteolysis and those without osteolysis. Interfacial membrane mononuclear cells also produced high levels of interleukin-1 alpha, interleukin-1 beta, and prostaglandin E2 and expressed bone resorptive activities following stimulation with either titanium or chromium orthophosphate. More importantly, interfacial membrane mononuclear cells "spontaneously" produced high levels of prostaglandin E2 that were comparable with the response of peripheral blood monocytes stimulated with particulate wear debris. The clinical relevance of this study is 2-fold. First, mononuclear cells from patients with total hip replacement were some-how "sensitized" to metal particles in comparison with mononuclear cells from individuals without an implant. Second, the chromium orthophosphate corrosion product was a potent macrophage/monocyte activator and may contribute to macrophage-mediated osteolysis and aseptic loosening.


Subject(s)
Hip Prosthesis , Macrophages/drug effects , Metals/pharmacology , Monocytes/drug effects , Adult , Aged , Aged, 80 and over , Bone Resorption/chemically induced , Bone Resorption/physiopathology , Calcium/metabolism , Cell Division/drug effects , Cell Line/drug effects , Cell Membrane/physiology , Cell Survival/drug effects , Chromium/chemistry , Chromium/immunology , Chromium/pharmacology , Cobalt/chemistry , Cobalt/immunology , Cobalt/pharmacology , Corrosion , Culture Media, Conditioned/pharmacology , Dinoprostone/biosynthesis , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Interleukin-1/biosynthesis , Joint Instability/pathology , Joint Instability/physiopathology , Macrophages/cytology , Macrophages/immunology , Male , Materials Testing , Metals/chemistry , Metals/immunology , Middle Aged , Monocytes/cytology , Monocytes/immunology , Organ Culture Techniques , Osteolysis/pathology , Osteolysis/physiopathology , Titanium/chemistry , Titanium/immunology , Titanium/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...