Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Bioinform ; 3: 1286883, 2023.
Article in English | MEDLINE | ID: mdl-38651055

ABSTRACT

Antibodies are generated with great diversity in nature resulting in a set of molecules, each optimized to bind a specific target. Taking advantage of their diversity and specificity, antibodies make up for a large part of recently developed biologic drugs. For therapeutic use antibodies need to fulfill several criteria to be safe and efficient. Polyspecific antibodies can bind structurally unrelated molecules in addition to their main target, which can lead to side effects and decreased efficacy in a therapeutic setting, for example via reduction of effective drug levels. Therefore, we created a neural-network-based model to predict polyspecificity of antibodies using the heavy chain variable region sequence as input. We devised a strategy for enriching antibodies from an immunization campaign either for antigen-specific or polyspecific binding properties, followed by generation of a large sequencing data set for training and cross-validation of the model. We identified important physico-chemical features influencing polyspecificity by investigating the behaviour of this model. This work is a machine-learning-based approach to polyspecificity prediction and, besides increasing our understanding of polyspecificity, it might contribute to therapeutic antibody development.

2.
MAbs ; 12(1): 1846900, 2020.
Article in English | MEDLINE | ID: mdl-33228444

ABSTRACT

Transgenic animals incorporating human antibody genes are extremely attractive for drug development because they obviate subsequent antibody humanization procedures required for therapeutic translation. Transgenic platforms have previously been established using mice, but also more recently rats, chickens, and cows and are now in abundant use for drug development. However, rabbit-based antibody generation, with a strong track record for specificity and affinity, is able to include gene conversion mediated sequence diversification, thereby enhancing binder maturation and improving the variance/selection of output antibodies in a different way than in rodents. Since it additionally frequently permits good binder generation against antigens that are only weakly immunogenic in other organisms, it is a highly interesting species for therapeutic antibody generation. We report here on the generation, utilization, and analysis of the first transgenic rabbit strain for human antibody production. Through the knockout of endogenous IgM genes and the introduction of human immunoglobulin sequences, this rabbit strain has been engineered to generate a highly diverse human IgG antibody repertoire. We further incorporated human CD79a/b and Bcl2 (B-cell lymphoma 2) genes, which enhance B-cell receptor expression and B-cell survival. Following immunization against the angiogenic factor BMP9 (Bone Morphogenetic Proteins 9), we were able to isolate a set of exquisitely affine and specific neutralizing antibodies from these rabbits. Sequence analysis of these binders revealed that both somatic hypermutation and gene conversion are fully operational in this strain, without compromising the very high degree of humanness. This powerful new transgenic strategy will allow further expansion of the use of endogenous immune mechanisms in drug development.


Subject(s)
Animals, Genetically Modified , Antibody Affinity/immunology , Antibody Specificity/immunology , B-Lymphocytes/immunology , Immunoglobulin G/immunology , Animals , Humans , Immunoglobulin G/genetics , Rabbits
3.
Arthritis Rheumatol ; 67(1): 51-62, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25303306

ABSTRACT

OBJECTIVE: Rheumatoid arthritis therapies that are based on inhibition of a single cytokine, e.g., tumor necrosis factor α (TNFα) or interleukin-6 (IL-6), produce clinically meaningful responses in only about half of the treated patients. This study was undertaken to investigate whether combined inhibition of TNFα and IL-17 has additive or synergistic effects in the suppression of mesenchymal cell activation in vitro and inflammation and tissue destruction in arthritis in vivo. METHODS: Cultures of human fibroblast-like synoviocytes (FLS) were stimulated with TNFα, IL-17, or a combination of both. Single/combined neutralizing antibodies against TNFα and IL-17 were used to examine in vitro cytokine responses and in vivo development of arthritis and bone and cartilage destruction in TNFα-transgenic mice. Bispecific anti-TNFα/IL-17 antibodies were designed, and their potential to block cytokine responses in human FLS was tested. RESULTS: TNFα and IL-17 had additive/synergistic effects in promoting production of IL-6, IL-8, and granulocyte colony-stimulating factor, as well as matrix metalloproteinases, in FLS. Bispecific anti-TNFα/IL-17 antibodies showed superior efficacy in blocking cytokine and chemokine responses in vitro. Furthermore, dual versus single inhibition of both cytokines using neutralizing antibodies was more effective in inhibiting the development of inflammation and bone and cartilage destruction in arthritic mice. CONCLUSION: Combined blockade of TNFα and IL-17 was more effective than single blockade in inhibiting cytokine, chemokine, and matrix enzyme responses from human mesenchymal cells and in blocking tissue destruction associated with arthritis, and additionally showed a positive impact on rebalance of bone homeostasis. Bispecific anti-TNFα/IL-17 antibodies may have superior efficacy in the treatment of arthritis and may overcome the limited therapeutic responses obtained with single cytokine neutralization.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Interleukin-17/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Antibodies, Bispecific/immunology , Antirheumatic Agents/immunology , Arthritis, Rheumatoid/pathology , Cells, Cultured , Disease Models, Animal , Drug Synergism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Granulocyte Colony-Stimulating Factor/metabolism , Humans , In Vitro Techniques , Interleukin-17/immunology , Interleukin-17/pharmacology , Interleukin-8/metabolism , Metalloproteases/metabolism , Mice , Mice, Transgenic , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Synovial Membrane/pathology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
4.
Neuron ; 81(1): 49-60, 2014 Jan 08.
Article in English | MEDLINE | ID: mdl-24411731

ABSTRACT

Although biotherapeutics have vast potential for treating brain disorders, their use has been limited due to low exposure across the blood-brain barrier (BBB). We report that by manipulating the binding mode of an antibody fragment to the transferrin receptor (TfR), we have developed a Brain Shuttle module, which can be engineered into a standard therapeutic antibody for successful BBB transcytosis. Brain Shuttle version of an anti-Aß antibody, which uses a monovalent binding mode to the TfR, increases ß-Amyloid target engagement in a mouse model of Alzheimer's disease by 55-fold compared to the parent antibody. We provide in vitro and in vivo evidence that the monovalent binding mode facilitates transcellular transport, whereas a bivalent binding mode leads to lysosome sorting. Enhanced target engagement of the Brain Shuttle module translates into a significant improvement in amyloid reduction. These findings have major implications for the development of biologics-based treatment of brain disorders.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Protein Transport/physiology , Single-Chain Antibodies/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Animals , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/immunology , Cell Line, Transformed , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Macrolides/pharmacology , Mice , Mice, Transgenic , Models, Immunological , Presenilin-1/genetics , Protein Binding/drug effects , Protein Binding/immunology , Protein Transport/drug effects , Receptors, Transferrin/immunology , Receptors, Transferrin/metabolism , Single-Chain Antibodies/pharmacology , Single-Chain Antibodies/therapeutic use , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Time Factors , Transcytosis/drug effects , Transcytosis/genetics , Transcytosis/immunology
5.
Eur J Immunol ; 43(3): 716-22, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23254454

ABSTRACT

Interleukin (IL)-1α is a potent proinflammatory cytokine that has been implicated in the development of atherosclerosis. We investigated whether a vaccine inducing IL-1α neutralizing antibodies could interfere with disease progression in a murine model of atherosclerosis. We immunized Apolipoprothin E (ApoE)-deficient mice with a vaccine (IL-1α-C-Qß) consisting of full-length, native IL-1α chemically conjugated to virus-like particles derived from the bacteriophage Qß. ApoE(-/-) mice were administered six injections of IL-1α-C-Qß or nonconjugated Qß over a period of 160 days while being maintained on a western diet. Atherosclerosis was measured in the descending aorta and in cross-sections at the aortic root. Macrophage infiltration in the aorta was measured using CD68. Expression levels of VCAM-1, ICAM-1, and MCP-1 were quantified by RT-PCR. Immunization against IL-1α reduced plaque progression in the descending aorta by 50% and at the aortic root by 37%. Macrophage infiltration in the aorta was reduced by 22%. Inflammation was also reduced in the adventitia, with a decrease of 54% in peri-aortic infiltrate score and reduced expression levels of VCAM-1 and ICAM-1. Active immunization targeting IL-1α reduced both the inflammatory reaction in the plaque as well as plaque progression. In summary, vaccination against IL-1α protected ApoE(-/-) mice against disease, suggesting that this may be a potential treatment option for atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Interleukin-1alpha/immunology , Vaccines, Virus-Like Particle/immunology , Animals , Antibodies/immunology , Antibodies, Neutralizing/immunology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Male , Mice , Mice, Knockout , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , Vaccines, Virus-Like Particle/administration & dosage
6.
J Neurosci ; 31(25): 9323-31, 2011 Jun 22.
Article in English | MEDLINE | ID: mdl-21697382

ABSTRACT

Immunization against amyloid-ß (Aß) can reduce amyloid accumulation in vivo and is considered a potential therapeutic approach for Alzheimer's disease. However, it has been associated with meningoencephalitis thought to be mediated by inflammatory T-cells. With the aim of producing an immunogenic vaccine without this side effect, we designed CAD106 comprising Aß1-6 coupled to the virus-like particle Qß. Immunization with this vaccine did not activate Aß-specific T-cells. In APP transgenic mice, CAD106 induced efficacious Aß antibody titers of different IgG subclasses mainly recognizing the Aß3-6 epitope. CAD106 reduced brain amyloid accumulation in two APP transgenic mouse lines. Plaque number was a more sensitive readout than plaque area, followed by Aß42 and Aß40 levels. Studies with very strong overall amyloid reduction showed an increase in vascular Aß, which atypically was nonfibrillar. The efficacy of Aß immunotherapy depended on the Aß levels and thus differed between animal models, brain regions, and stage of amyloid deposition. Therefore, animal studies may not quantitatively predict the effect in human Alzheimer's disease. Our studies provided no evidence for increased microhemorrhages or inflammatory reactions in amyloid-containing brain. In rhesus monkeys, CAD106 induced a similar antibody response as in mice. The antibodies stained amyloid deposits on tissue sections of mouse and human brain but did not label cellular structures containing APP. They reacted with Aß monomers and oligomers and blocked Aß toxicity in cell culture. We conclude that CAD106 immunization is suited to interfere with Aß aggregation and its downstream detrimental effects.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/therapeutic use , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/immunology , Immunotherapy/methods , Alzheimer Disease/immunology , Amyloid beta-Peptides/adverse effects , Animals , Cells, Cultured , Mice , Mice, Transgenic , Treatment Outcome
7.
PLoS One ; 5(3): e9809, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20352110

ABSTRACT

BACKGROUND: Recombinant proteins and in particular single domains or peptides are often poorly immunogenic unless conjugated to a carrier protein. Virus-like-particles are a very efficient means to confer high immunogenicity to antigens. We report here the development of virus-like-particles (VLPs) derived from the RNA bacteriophage AP205 for epitope-based vaccines. METHODOLOGY/PRINCIPAL FINDINGS: Peptides of angiotensin II, S.typhi outer membrane protein (D2), CXCR4 receptor, HIV1 Nef, gonadotropin releasing hormone (GnRH), Influenza A M2-protein were fused to either N- or C-terminus of AP205 coat protein. The A205-peptide fusions assembled into VLPs, and peptides displayed on the VLP were highly immunogenic in mice. GnRH fused to the C-terminus of AP205 induced a strong antibody response that inhibited GnRH function in vivo. Exposure of the M2-protein peptide at the N-terminus of AP205 resulted in a strong M2-specific antibody response upon immunization, protecting 100% of mice from a lethal influenza infection. CONCLUSIONS/SIGNIFICANCE: AP205 VLPs are therefore a very efficient and new vaccine system, suitable for complex and long epitopes, of up to at least 55 amino acid residues in length. AP205 VLPs confer a high immunogenicity to displayed epitopes, as shown by inhibition of endogenous GnRH and protective immunity against influenza infection.


Subject(s)
Epitopes/chemistry , Virion/chemistry , Animals , Cloning, Molecular , Gonadotropin-Releasing Hormone/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptides/chemistry , Protein Structure, Tertiary , RNA Phages/metabolism , Recombinant Proteins/chemistry , Vaccination/instrumentation , Vaccination/methods
8.
Lancet ; 371(9615): 821-7, 2008 Mar 08.
Article in English | MEDLINE | ID: mdl-18328929

ABSTRACT

BACKGROUND: Hypertension can be controlled adequately with existing drugs such as angiotensin-converting enzyme inhibitors or angiotensin II receptor blockers. Nevertheless, treatment success is often restricted by patients not adhering to treatment. Immunisation against angiotensin II could solve this problem. We investigated the safety and efficacy of CYT006-AngQb-a vaccine based on a virus-like particle-that targets angiotensin II to reduce ambulatory blood pressure. METHODS: In this multicentre, double-blind, randomised, placebo-controlled phase IIa trial, 72 patients with mild-to-moderate hypertension were randomly assigned with a computer-generated randomisation list to receive subcutaneous injections of either 100 mug CYT006-AngQb (n=24), 300 mug CYT006-AngQb (24), or placebo (24), at weeks 0, 4, and 12. 24-h ambulatory blood pressure was measured before treatment and at week 14. The primary outcomes were safety and tolerability. Analyses were done by intention to treat. This study is registered with ClinicalTrials.gov, number NCT00500786. FINDINGS: Two patients in the 100 mug group, three in the 300 mug group, and none in the placebo group discontinued study treatment. All patients were included in safety analyses; efficacy analyses did not include the five dropouts, for whom no data were available at week 14. Five serious adverse events were reported (two in the 100 mug group, two in the 300 mug group, and one in the placebo group); none were deemed to be treatment related. Most side-effects were mild, transient reactions at the injection site. Mild, transient influenza-like symptoms were seen in three patients in the 100 mug group, seven in the 300 mug group, and none in the placebo group. In the 300 mug group, there was a reduction from baseline in mean ambulatory daytime blood pressure at week 14 by -9.0/-4.0 mm Hg compared with placebo (p=0.015 for systolic and 0.064 for diastolic). The 300 mug dose reduced the early morning blood-pressure surge compared with placebo (change at 0800 h -25/-13 mm Hg; p<0.0001 for systolic, p=0.0035 for diastolic). INTERPRETATION: Immunisation with CYT006-AngQb was associated with no serious adverse events; most observed adverse events were consistent with local or systemic responses similar to those seen with other vaccines. The 300 mug dose reduced blood pressure in patients with mild-to-moderate hypertension during the daytime, especially in the early morning. FUNDING: Cytos Biotechnology AG.


Subject(s)
Angiotensin II/antagonists & inhibitors , Blood Pressure/drug effects , Hypertension/drug therapy , Oligopeptides/therapeutic use , Vaccines, Synthetic/therapeutic use , Adult , Aged , Angiotensin II/immunology , Antibody Formation/drug effects , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Humans , Hypertension/immunology , Middle Aged , Monitoring, Ambulatory , Oligopeptides/adverse effects , Oligopeptides/immunology , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology
9.
J Hypertens ; 25(1): 63-72, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17143175

ABSTRACT

BACKGROUND: Despite the availability of efficacious drugs, the success of treating hypertension is limited by patients' inconsistent drug intake. Immunization against angiotensin II may offer a valuable alternative to conventional drugs for the treatment of hypertension, because vaccines induce relatively long-lasting effects and do not require daily dosing. Here we describe the preclinical development and the phase I clinical trial testing of a virus-like particle (VLP)-based antihypertensive vaccine. METHODS AND RESULTS: An angiotensin II-derived peptide was conjugated to the VLP Qbeta (AngQb). AngQb was highly immunogenic in mice and rats. To test for efficacy, spontaneously hypertensive rats (SHR) were immunized with 400 microg AngQb or VLP alone. Group mean systolic blood pressure (SBP) was reduced by up to 21 mmHg (159 +/- 2 versus 180 +/- 5 mmHg, P < 0.001), and total angiotensin II levels (antibody-bound and free) were increased ninefold (85 +/- 20 versus 9 +/- 1 pmol/l, P = 0.002) compared with VLP controls. SHR treated with the angiotensin-converting enzyme (ACE) inhibitor ramipril (1 mg/kg per day by mouth) reached an SBP of 155 +/- 2 mmHg. Twelve healthy volunteers of a placebo-controlled randomized phase I trial were injected once with 100 microg AngQb. Angiotensin II-specific antibodies were raised in all subjects (100% responder rate) and AngQb was well tolerated. CONCLUSIONS: AngQb reduces blood pressure in SHR to levels obtained with an ACE inhibitor, and is immunogenic and well tolerated in humans. Therefore, vaccination against angiotensin II has the potential to become a useful antihypertensive treatment providing long-lasting effects and improving patient compliance.


Subject(s)
Angiotensin II/immunology , Antihypertensive Agents/therapeutic use , Hypertension/drug therapy , Vaccines/therapeutic use , Virion/immunology , Adult , Angiotensin II/blood , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Antibody Specificity , Antihypertensive Agents/adverse effects , Antihypertensive Agents/immunology , Antihypertensive Agents/toxicity , Autoantibodies/blood , Blood Pressure/drug effects , Disease Models, Animal , Double-Blind Method , Drug Evaluation, Preclinical , Humans , Hypertension/blood , Hypertension/immunology , Hypertension/physiopathology , Male , Mice , Mice, Inbred BALB C , Middle Aged , Patient Compliance , Ramipril/therapeutic use , Rats , Rats, Inbred SHR , Reference Values , Time Factors , Vaccines/adverse effects , Vaccines/immunology , Vaccines/toxicity
11.
Eur J Immunol ; 35(3): 816-21, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15724244

ABSTRACT

Induction of high frequencies of specific T cells by vaccination requires prime-boost regimens. To reach optimal immune responses, it is necessary to use different vectors for priming and boosting as e.g. DNA vaccination followed by boosting with a recombinant viral vector. Here, we show that vaccines based on virus-like particles (VLP) displaying peptide epitopes are equally effective to induce CTL responses if used in a homologous or heterologous prime-boost setting. Strikingly, high frequencies (>20% of CD8(+) cells) of protective CTL could be induced and maintained by weekly injection of VLP. Thus, the use of VLP may avoid the requirement for complicated heterologous prime-boost regimens, facilitating the development of effective T cell-based vaccines.


Subject(s)
Hepatitis B Core Antigens/immunology , T-Lymphocytes/immunology , Vaccination/methods , Vaccines, DNA , Virion/immunology , Animals , CpG Islands/immunology , Epitopes, T-Lymphocyte/immunology , Female , Mice , Vaccinia/immunology , Vaccinia virus/immunology , Viral Vaccines/immunology
12.
FEBS Lett ; 543(1-3): 11-5, 2003 May 22.
Article in English | MEDLINE | ID: mdl-12753896

ABSTRACT

The decameric peptide SALQNAASIA from the Mycobacterium bovis heat shock protein (hsp) 60 is recognized by the murine T-cell receptor UZ-3-4 in complex with the murine class I major histocompatibility complex molecule H-2D(b). This T-cell receptor cross-reacts with the H-2D(b)-bound non-homologous decameric peptide KDIGNIISDA from the murine hsp60, but does not recognize the nonameric mycobacterial peptide SALQNAASI. Cross-recognition of the KDIGNIISDA/H-2D(b) complex induces autoimmune pathology in immunodeficient mice. We solved the X-ray crystal structure of the SALQNAASIA/H-2D(b) complex at 3.0 A resolution, and we modelled the KDIGNIISDA and SALQNAASI peptides in the H-2D(b) binding site. The structural analysis of the H-2D(b)-bound hsp60 epitopes offers insight into T-cell receptor cross-reactivity.


Subject(s)
Antigens, Bacterial/chemistry , Chaperonin 60/chemistry , Epitopes, T-Lymphocyte/chemistry , H-2 Antigens/chemistry , Mycobacterium bovis/immunology , Peptide Fragments/chemistry , Amino Acid Sequence , Animals , Binding Sites , Chaperonin 60/immunology , Crystallography, X-Ray , H-2 Antigens/metabolism , Histocompatibility Antigen H-2D , Macromolecular Substances , Mice , Models, Molecular , Peptide Fragments/immunology
13.
Intervirology ; 45(4-6): 212-7, 2002.
Article in English | MEDLINE | ID: mdl-12566703

ABSTRACT

Induction of protective immune responses with recombinant antigens is a major challenge for the vaccine industry. Here we present a molecular assembly system that renders antigens of choice highly repetitive. Using this method, efficient antibody responses may be induced in the absence of adjuvants resulting in protection from viral infection and allergic reactions.


Subject(s)
Viral Vaccines/administration & dosage , Virion/immunology , Animals , Antigens/immunology , Humans , Hypersensitivity/therapy , Immunotherapy , Viral Vaccines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...