17 research outputs found

    Characterisation of a Novel Anti-CD52 Antibody with Improved Efficacy and Reduced Immunogenicity

    No full text
    <div><p>Anti-CD52 therapy has been shown to be effective in the treatment of a number of B cell malignancies, hematopoietic disorders and autoimmune diseases (including rheumatoid arthritis and multiple sclerosis); however the current standard of treatment, the humanized monoclonal antibody alemtuzumab, is associated with the development of anti-drug antibodies in a high proportion of patients. In order to address this problem, we have identified a novel murine anti-CD52 antibody which has been humanized using a process that avoids the inclusion within the variable domains of non-human germline MHC class II binding peptides and known CD4+ T cell epitopes, thus reducing its potential for immunogenicity in the clinic. The resultant humanized antibody, ANT1034, was shown to have superior binding to CD52 expressing cells than alemtuzumab and was more effective at directing both antibody dependent and complement dependent cell cytotoxicity. Furthermore, when in the presence of a cross-linking antibody, ANT1034 was more effective at directly inducing apoptosis than alemtuzumab. ANT1034 also showed superior activity in a SCID mouse/human CD52 tumour xenograft model where a single 1 mg/Kg dose of ANT1034 led to increased mouse survival compared to a 10 mg/Kg dose of alemtuzumab. Finally, ANT1034 was compared to alemtuzumab in <i>in vitro</i> T cell assays in order to evaluate its potential to stimulate proliferation of T cells in peripheral blood mononuclear cells derived from a panel of human donors: whereas alemtuzumab stimulated proliferation in a high proportion of the donor cohort, ANT1034 did not stimulate proliferation in any of the donors. Therefore we have developed a candidate therapeutic humanized antibody, ANT1034, that may have the potential to be more efficacious and less immunogenic than the current standard anti-CD52 therapy.</p></div

    Amino acid sequence of murine anti-CD52 antibody 2E8 and lead humanized variant.

    No full text
    <p>Amino acid sequences of the murine antibody 2E8 and lead humanized variant (ANT1034 –composed of VH3 and VK4 variable domains). Sourced human sequence segments together with their GenBank accession numbers are indicated below the amino acid sequences. (a) Heavy chain variable domain; (b) Light chain variable domain. Differences between the murine and humanized sequence are shaded. CDRs (shown in orange text) and numbering are as described by Kabat [<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0138123#pone.0138123.ref041" target="_blank">41</a>]. Sequences are available from GenBank: 2E8_VH—KP877884; 2E8_VK—KP877885; ANT1034_VH—KP877886; ANT1034_VK—KP877887.</p

    Peptide competition ELISA of selected humanized variants.

    No full text
    <p>Titrations of humanized variants were competed against a fixed concentration (35 ng/ml) of biotinylated murine 2E8 for binding to CD52 peptide that was coated directly on an ELISA plate. Binding was detected with streptavidin-HRP and TMB substrate. Antibodies are named in the format ANT10X<sub>1</sub>X<sub>2</sub> where X<sub>1</sub> refers to the VH variant (VH1 to VH5) and X<sub>2</sub> refers to the Vκ variant (Vκ1 to Vκ4).</p

    Characterisation of anti-CD52 humanized antibodies, alemtuzumab and 2E8 chimeric IgG.

    No full text
    <p>Selected humanized antibodies were characterised by: (a) Direct binding of antibodies to REH<sup>++</sup> cells using flow cytometry; (b) Competition binding against alemtuzumab (with murine constant regions) for binding to CD52 on the surface of REH<sup>++</sup> cells using flow cytometry; (c) ADCC using REH<sup>++</sup> target cells (average of 4 PBMC donors) with PBMC effector cells at a target:effector ratio of 50:1, and; (d) CDC using REH<sup>++</sup> target cells and normal human serum as a source of complement. For clarity, error bars are included in (c) and (d) for alemtuzumab and the lead humanized antibody, ANT1034, only.</p

    Selection of high CD52-expressing cell lines.

    No full text
    <p>Raji, REH and CD52-NS0 cells were assessed by flow cytometry for CD52 expression and high CD52 expressing (<sup>++</sup>) clones were identified for the three cell lines. Cells (either parent or <sup>++</sup> lines) were stained with irrelevant human IgG (a) or with alemtuzumab (b) and (c).</p

    Flow cytometry analysis of hybridomas or irrelevant mouse IgG.

    No full text
    <p>Selected hybridoma supernatants or media spiked with irrelevant mouse IgG were assessed for binding to either untransfected or NS0-CD52<sup>++</sup> cells by flow cytometry to identify CD52 binding antibodies.</p

    Characterisation of anti-CD52 antibodies alemtuzumab and chimeric 2E8 IgG.

    No full text
    <p>Antibodies were characterised by: (a) Direct binding of antibodies to HUT78 cells using flow cytometry; (b) Competition against alemtuzumab (with murine constant regions) for binding to CD52 on the surface of HUT78 cells using flow cytometry; (c) ADCC using REH<sup>++</sup> target cells (average of 4 PBMC donors for effector cells).</p

    CD4+ Episcreenâ„¢ T cell proliferation responses to ANT1034, alemtuzumab and chimeric 2E8.

    No full text
    <p>CD4<sup>+</sup> T cells were incubated with autologous mature DC loaded with the samples and assessed for proliferation after 7 days incubation. T cell responses with an SI ≥1.90 (indicated by red dotted line) that were significant (<i>p</i> <0.05) using an unpaired, two sample Student’s <i>t</i>-test were considered positive.</p

    Kaplan-Meier survival curves for the <i>in vivo</i> assessment of ANT1034 and alemtuzumab.

    No full text
    <p>Antibodies were assessed in the Raji human Burkitt lymphoma SCID mouse model. Antibodies were administered on alternate days for seven doses starting 3 days post-injection of tumour cells. Efficacy was determined by comparing the median times to endpoint (either death, or euthanasia for advanced tumour progression).</p

    Chemical Modification by glycation does not enhance the response of PBMC in the IVCIA assay.

    No full text
    <p>Three representative mAbs (Avastin, mAb1, and Humira), that were treated by glycation with different sugars (galactose, glucose and mannose, and a non-glycating sugar, sorbitol), were tested in the IVCIA assay at the early (20 h) (n = 11 donors for most samples) and late (7 day) phases (n = 6 donors). Avastin, mAb1, and Humira have low, medium, and high rates of clinical or predicted immunogenicity, respectively. Heatmaps depict the percentage of donors that responded to the glycated mAbs above the original forms of each molecule (mAb before glycation treatment) and were at least two fold above the background, to highlight the differential response that might be due to glycated mAbs. The percentage of donors with increased secretion of signature cytokines in adherent monocytes at the early phase (red), and in PBMC at the early (yellow) and late (green) phases is highlighted. The percentage of donors with an increase in the number of IFN-γ secreting cells is shown on the far right (blue). The grey boxes show low level responses that were observed in less than or equal to 20% of donors, in contrast to colored boxes (red, yellow, green and blue) that show responses in a greater number of donors (40–100%).</p
    corecore