TGN1412

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 27933 Experts worldwide ranked by ideXlab platform

Richard Stebbings - One of the best experts on this subject based on the ideXlab platform.

  • severity of the TGN1412 trial disaster cytokine storm correlated with il 2 release
    British Journal of Clinical Pharmacology, 2013
    Co-Authors: David Eastwood, Susan J. Thorpe, Robin Thorpe, Lucy Findlay, Chris Bird, Meenu Wadhwa, Stephen Poole, Paula Dilger, Jason Hockley, Richard Stebbings
    Abstract:

    Aim To determine if cytokine release with a solid phase assay is predictive of adverse responses for a range of therapeutic mAbs.

  • after TGN1412 recent developments in cytokine release assays
    Journal of Immunotoxicology, 2013
    Co-Authors: Richard Stebbings, David Eastwood, Stephen Poole, Robin Thorpe
    Abstract:

    The failure of regulatory science to keep pace with and support the development of new biological medicines was very publically highlighted in March 2006 when the first-in-man Phase I clinical trial of the immunomodulatory CD28-specific monoclonal antibody (mAb) TGN1412 ended in disaster when all six volunteers suffered a life-threatening adverse reaction termed a ‘Cytokine Storm’. The poor predictive value of standard pre-clinical safety tests and animal models applied to TGN1412 demonstrated the need for a new generation of immunotoxicity assays and animal models that are both sensitive and predictive of clinical outcome in man. The non-predictive result obtained from pre-clinical safety testing in cynomolgus macaques has now been attributed to a lack of CD28 expression on CD4+ effector memory T-cells that therefore cannot be stimulated by TGN1412. In contrast, high levels of CD28 are expressed on human CD4+ effector memory T-cells, the source of most TGN1412-stimulated pro-inflammatory cytokines. Stand...

  • antibody c region influences TGN1412 like functional activity in vitro
    Journal of Immunology, 2012
    Co-Authors: Christina Ball, David Eastwood, Bernard Fox, Lucy Findlay, Richard Stebbings, Stephen Poole, Giles Sharp, Simon E Hufton, Paul W H I Parren, Robin Thorpe
    Abstract:

    The unexpected outcome of the clinical trial of the superagonistic CD28 mAb TGN1412 (IgG4κ) continues to stimulate interest. We show that TGN1412 binds similarly to human and cynomolgus macaque FcγR, eliminating the possibility that differences in Fc-mediated interactions with FcγR contributed to the failure of preclinical testing in macaques to predict toxicity in humans. The influence of the Fc domain and C region structure on the in vitro functional activity of TGN1412 was investigated using F(ab′)2 and Fab fragments derived from TGN1412 recovered from the trial and recombinant TGN1412 subclass variants and mutants. Superagonistic activity, as measured by cytokine release and proliferation, was assessed by exposing PBMCs to immobilized mAbs/fragments or to aqueous mAbs/fragments in the presence of HUVEC monolayers. Removing the Fc generally curtailed or abolished PBMC activation. However, eliminating detectable FcγR-binding of the IgG4 by mutation (L235E) did not abrogate activity. Stabilizing the “wild-type” IgG4 hinge (S228P) enhanced activity without increasing FcγR binding, which could only partially be explained by inhibition of Fab arm–exchange. Subclass switching the IgG4 mAb to IgG1 decreased activity, whereas switching to IgG2 markedly increased activity. We conclude that the C region strongly influences in vitro CD28-mediated superagonistic signaling. Superagonism requires an intact Fc, as shown by the absence of activity of TGN1412 Fab and F(ab′)2 fragments, but, notably, appears to be relatively independent of FcγR-binding properties. We propose that the Fc, potentially through restricting flexibility, maintains a favorable V region conformation to allow superagonistic activity. These findings have important implications for Ab design strategies.

  • comparison of novel methods for predicting the risk of pro inflammatory clinical infusion reactions during monoclonal antibody therapy
    Journal of Immunological Methods, 2011
    Co-Authors: Lucy Findlay, David Eastwood, Susan J. Thorpe, Robin Thorpe, Christina Ball, Jane C Robinson, Chris Bird, Meenu Wadhwa, Richard Stebbings, Stephen Poole
    Abstract:

    Abstract Two methods for predicting the risk of pro-inflammatory clinical infusion reactions during monoclonal antibody therapy were evaluated. In the first, the antibody of interest is immobilised by air-drying onto 96-well plates prior to the addition of human peripheral blood mononuclear cells (PBMCs). In the second, the antibody is added in aqueous phase to a co-culture of human PBMCs and human endothelium-derived cells. In both methods the cells are incubated with the antibody to allow the accumulation of pro-inflammatory cytokines, quantified by enzyme-linked immunosorbent assay (ELISA). The antibodies associated with clinical infusion reactions, Herceptin, Campath-1H and TGN1412, gave the largest responses taking into account the data for all readouts (tumour necrosis factor-α, TNF, interleukin-6, IL-6, IL-8, IL-2 and cell proliferation) for both methods. Overall, the antibodies tested could be ranked as follows: Tysabri

  • endothelial cells co stimulate peripheral blood mononuclear cell responses to monoclonal antibody TGN1412 in culture
    Cytokine, 2011
    Co-Authors: Lucy Findlay, David Eastwood, Bernard Fox, Christina Ball, Jane C Robinson, Chris Bird, Meenu Wadhwa, Richard Stebbings, Giles Sharp, Stephen Poole
    Abstract:

    The failure of preclinical testing to predict the severity of the cytokine storm experienced by the recipients of the superagonistic anti-CD28 monoclonal antibody (mAb) TGN1412 during its Phase 1 clinical trial prompted the development of new in vitro experimental approaches for mimicking in vivo cytokine release and lymphoproliferation. Peripheral blood mononuclear cells (PBMC) presented to TGN1412 immobilised on plastic has previously been shown to stimulate a pro-inflammatory cytokine response. The aim of the present study was to investigate a 'co-culture' model for the detection of TGN1412-like immunomodulatory activity in which TGN1412 was presented to PBMC in the presence of monolayers of endothelium-derived cells and other cell types, followed by measurement of cytokine levels in the culture supernatants and proliferation of PBMC. Culturing PBMC with TGN1412 over primary human umbilical vein endothelial cells (HUVEC) and HUVEC-derived cell lines retaining classic endothelial markers, but not cell lines of non-endothelial origin, mediated the specific release of IL-6, IL-8 and TNFα, and proliferation of PBMC. Low levels of IL-2 and IFNγ were also detected in supernatants with most donors of PBMC. An anti-CD28 mAb agonist, i.e., not a superagonist like TGN1412, did not stimulate cytokine release or proliferation of PBMC in co-cultures. In conclusion, co-culture experiments for TGN1412-specific cytokine release required cells of endothelial origin. However, the profile of released cytokines in co-cultures did not mirror that in the clinical trial participants or the responses from PBMC exposed to TGN1412 immobilised on plastic, suggesting that TGN1412 stimulation of PBMC can occur through more than one mechanism.

Robin Thorpe - One of the best experts on this subject based on the ideXlab platform.

  • Cytokine release assays for the prediction of therapeutic mAb safety in first-in man trials--Whole blood cytokine release assays are poorly predictive for TGN1412 cytokine storm.
    Journal of immunological methods, 2015
    Co-Authors: Sandrine Vessillier, David Eastwood, Bernard Fox, Jean G. Sathish, Swaminathan Sethu, Thomas Dougall, Susan J. Thorpe, Robin Thorpe, R. Stebbings
    Abstract:

    The therapeutic monoclonal antibody (mAb) TGN1412 (anti-CD28 superagonist) caused near-fatal cytokine release syndrome (CRS) in all six volunteers during a phase-I clinical trial. Several cytokine release assays (CRAs) with reported predictivity for TGN1412-induced CRS have since been developed for the preclinical safety testing of new therapeutic mAbs. The whole blood (WB) CRA is the most widely used, but its sensitivity for TGN1412-like cytokine release was recently criticized. In a comparative study, using group size required for 90% power with 5% significance as a measure of sensitivity, we found that WB and 10% (v/v) WB CRAs were the least sensitive for TGN1412 as these required the largest group sizes (n = 52 and 79, respectively). In contrast, the peripheral blood mononuclear cell (PBMC) solid phase (SP) CRA was the most sensitive for TGN1412 as it required the smallest group size (n = 4). Similarly, the PBMC SP CRA was more sensitive than the WB CRA for muromonab-CD3 (anti-CD3) which stimulates TGN1412-like cytokine release (n = 4 and 4519, respectively). Conversely, the WB CRA was far more sensitive than the PBMC SP CRA for alemtuzumab (anti-CD52) which stimulates FcγRI-mediated cytokine release (n = 8 and 180, respectively). Investigation of potential factors contributing to the different sensitivities revealed that removal of red blood cells (RBCs) from WB permitted PBMC-like TGN1412 responses in a SP CRA, which in turn could be inhibited by the addition of the RBC membrane protein glycophorin A (GYPA); this observation likely underlies, at least in part, the poor sensitivity of WB CRA for TGN1412. The use of PBMC SP CRA for the detection of TGN1412-like cytokine release is recommended in conjunction with adequately powered group sizes for dependable preclinical safety testing of new therapeutic mAbs.

  • severity of the TGN1412 trial disaster cytokine storm correlated with il 2 release
    British Journal of Clinical Pharmacology, 2013
    Co-Authors: David Eastwood, Susan J. Thorpe, Robin Thorpe, Lucy Findlay, Chris Bird, Meenu Wadhwa, Stephen Poole, Paula Dilger, Jason Hockley, Richard Stebbings
    Abstract:

    Aim To determine if cytokine release with a solid phase assay is predictive of adverse responses for a range of therapeutic mAbs.

  • after TGN1412 recent developments in cytokine release assays
    Journal of Immunotoxicology, 2013
    Co-Authors: Richard Stebbings, David Eastwood, Stephen Poole, Robin Thorpe
    Abstract:

    The failure of regulatory science to keep pace with and support the development of new biological medicines was very publically highlighted in March 2006 when the first-in-man Phase I clinical trial of the immunomodulatory CD28-specific monoclonal antibody (mAb) TGN1412 ended in disaster when all six volunteers suffered a life-threatening adverse reaction termed a ‘Cytokine Storm’. The poor predictive value of standard pre-clinical safety tests and animal models applied to TGN1412 demonstrated the need for a new generation of immunotoxicity assays and animal models that are both sensitive and predictive of clinical outcome in man. The non-predictive result obtained from pre-clinical safety testing in cynomolgus macaques has now been attributed to a lack of CD28 expression on CD4+ effector memory T-cells that therefore cannot be stimulated by TGN1412. In contrast, high levels of CD28 are expressed on human CD4+ effector memory T-cells, the source of most TGN1412-stimulated pro-inflammatory cytokines. Stand...

  • this information is current as preclinical testing of immunotherapeutics understanding the causes to improve monoclonal antibody TGN1412 better cytokine storm in the phase i trial of
    2013
    Co-Authors: Stephen Poole, Robin Thorpe, Meenu Wadhwa, Isabelle Cludts, J Thorpe, A F Bristow, Jane Robinson, Tony Meager, Carl Dolman, Bernard Fox
    Abstract:

    and Stephen PooleJ. Thorpe, Adrian Bristow, Meenu Wadhwa, Robin ThorpeTarrant, Jane Robinson, Tony Meager, Carl Dolman, Susan Dilger, Emily Liefooghe, Isabelle Cludts, Bernard Fox, GillEastwood, Chris Bird, David North, Yogesh Mistry, Paula Richard Stebbings, Lucy Findlay, Cherry Edwards, Davidhttp://www.jimmunol.org/content/179/5/3325J Immunol€2007; 179:3325-3331; ;Referenceshttp://www.jimmunol.org/content/179/5/3325.full#ref-list-1This article cites 11 articles, 3 of which you can access for free at: Subscriptionshttp://jimmunol.org/subscriptionsInformation about subscribing to The Journal of Immunology is online at: Permissionshttp://www.aai.org/ji/copyright.htmlSubmit copyright permission requests at: Email Alertshttp://jimmunol.org/cgi/alerts/etocReceive free email-alerts when new articles cite this article. Sign up at:

  • antibody c region influences TGN1412 like functional activity in vitro
    Journal of Immunology, 2012
    Co-Authors: Christina Ball, David Eastwood, Bernard Fox, Lucy Findlay, Richard Stebbings, Stephen Poole, Giles Sharp, Simon E Hufton, Paul W H I Parren, Robin Thorpe
    Abstract:

    The unexpected outcome of the clinical trial of the superagonistic CD28 mAb TGN1412 (IgG4κ) continues to stimulate interest. We show that TGN1412 binds similarly to human and cynomolgus macaque FcγR, eliminating the possibility that differences in Fc-mediated interactions with FcγR contributed to the failure of preclinical testing in macaques to predict toxicity in humans. The influence of the Fc domain and C region structure on the in vitro functional activity of TGN1412 was investigated using F(ab′)2 and Fab fragments derived from TGN1412 recovered from the trial and recombinant TGN1412 subclass variants and mutants. Superagonistic activity, as measured by cytokine release and proliferation, was assessed by exposing PBMCs to immobilized mAbs/fragments or to aqueous mAbs/fragments in the presence of HUVEC monolayers. Removing the Fc generally curtailed or abolished PBMC activation. However, eliminating detectable FcγR-binding of the IgG4 by mutation (L235E) did not abrogate activity. Stabilizing the “wild-type” IgG4 hinge (S228P) enhanced activity without increasing FcγR binding, which could only partially be explained by inhibition of Fab arm–exchange. Subclass switching the IgG4 mAb to IgG1 decreased activity, whereas switching to IgG2 markedly increased activity. We conclude that the C region strongly influences in vitro CD28-mediated superagonistic signaling. Superagonism requires an intact Fc, as shown by the absence of activity of TGN1412 Fab and F(ab′)2 fragments, but, notably, appears to be relatively independent of FcγR-binding properties. We propose that the Fc, potentially through restricting flexibility, maintains a favorable V region conformation to allow superagonistic activity. These findings have important implications for Ab design strategies.

Thomas Hunig - One of the best experts on this subject based on the ideXlab platform.

  • the rise and fall of the cd28 superagonist TGN1412 and its return as tab08 a personal account
    FEBS Journal, 2016
    Co-Authors: Thomas Hunig
    Abstract:

    Two decades ago, we discovered 'superagonistic' monoclonal antibodies specific for the CD28 molecule which are able to polyclonally activate T cells, in particular regulatory T cells, and are therapeutically active in many rodent models of autoimmunity, inflammation, transplantation, and tissue repair. A phase I trial of the human CD28 superagonist TGN1412 failed in 2006 due to an unexpected cytokine release syndrome, but after it became clear that dose-reduction allows to preferentially address regulatory T cells also in humans, clinical development was resumed under the name TAB08. Here, I recount the story of CD28 superagonist development from a personal perspective with an emphasis on the dramatic events during and after the 2006 phase I trial, the reasons for the failure of preclinical research to warn of the impending cytokine storm, and on the research which allowed resumption of clinical development.

  • from TGN1412 to tab08 the return of cd28 superagonist therapy to clinical development for the treatment of rheumatoid arthritis
    Clinical and Experimental Rheumatology, 2016
    Co-Authors: Dmitry Tyrsin, Sergey Chuvpilo, Alexey A Matskevich, Daniil Nemenov, Paula S Romer, Paula Tabares, Thomas Hunig
    Abstract:

    CD28 superagonists (CD28SA) are CD28-specific monoclonal antibodies which are able to activate T-cells without overt TCR engagement. In rodents, CD28SA efficiently activate regulatory T-cells and are therapeutically effective in multiple models of autoimmunity, inflammation and transplantation. However, a phase I study of the human CD28SA TGN1412 in 2006 resulted in a life-threatening cytokine storm. This brief review summarises preclinical work before and since the failed phase I trial with an emphasis on understanding the reasons why there had been no warning of toxicity, and how a novel assay paved the way for a new phase I, phase Ib (both completed), and an ongoing phase II study.

  • in vitro polyclonal activation of conventional t cells with a cd28 superagonist protects mice from acute graft versus host disease
    European Journal of Immunology, 2015
    Co-Authors: Niklas Beyersdorf, Thomas Hunig, Sandra Werner, Nelli Wolf, Thomas Kerkau
    Abstract:

    : Upon transplantation of T cells from a CD28 superagonist (CD28-SA) treated donor into an irradiated allogeneic host, the CD28-SA-induced activation and expansion of Treg cells inhibits acute graft versus host disease (aGvHD), while not abrogating the desired graft versus tumor effect. Human peripheral blood CD4(+) T cells, however, harbor only very few Treg cells. Therefore, we studied whether polyclonal in vitro prestimulation of conventional, that is Treg -cell-depleted, CD4(+) T cells of C57BL/6 mice with CD28-SA-coated paramagnetic beads is sufficient to protect recipient BALB/c mice from aGvHD. CD28-SA prestimulation of conventional CD4(+) T cells efficiently protected BALB/c recipient mice from aGvHD and CD28-SA-stimulated CD4(+) and CD8(+) T cells were capable of mediating long-term protection from the BCL1 lymphoma. The recently completed successful phase I testing of the human CD28-SA TGN1412/TAB08 should greatly facilitate further development of this straightforward method into a novel immunotherapy for patients.

  • human regulatory t cells are selectively activated by low dose application of the cd28 superagonist TGN1412 tab08
    European Journal of Immunology, 2014
    Co-Authors: Paula Tabares, Hans-peter Tony, Dmitry Tyrsin, Sergey Chuvpilo, Alexey A Matskevich, Paula S Romer, Susanne Berr, Yury Fedotov, Hermann Einsele, Thomas Hunig
    Abstract:

    CD28 superagonists (CD28SAs) are potent T-cell-activating monoclonal antibodies (mAbs). In contrast to their benign behavior and marked therapeutic efficacy as activators of regulatory T (Treg) cells in preclinical rodent models, a phase I trial of the human CD28SA TGN1412 (now called TAB08) in 2006 resulted in a life-threatening cytokine release syndrome (CRS). We studied TAB08-mediated Treg-cell activation in a recently developed in vitro system of human PBMCs, which also reproduces the CRS experienced by the healthy volunteers. We show that just as in rodents, CD28SAs are potent activators and expanders of Treg cells from healthy donors and rheumatoid arthritis patients, even under effective blockade of pro-inflammatory cytokine release by a corticosteroid. Moreover, CD28SA titration identifies a dose range where pro-inflammatory cytokine secretion from conventional T cells is absent while appreciable Treg-cell activation is maintained. Finally, we report that low-dose application of TAB08 to healthy volunteers results in dose-dependent systemic release of the Treg-cell signature cytokine IL-10 in the absence of the pro-inflammatory factors associated with the CRS of the 2006 TGN1412 study. These results demonstrate the potential of appropriately dosed CD28SA and corticosteroid comedication to mobilize human Treg cells for the treatment of autoimmune and inflammatory conditions.

  • response to storm forecasting additional lessons from the cd28 superagonist TGN1412 trial
    Nature Reviews Immunology, 2012
    Co-Authors: Thomas Hunig
    Abstract:

    Response to 'Storm forecasting: additional lessons from the CD28 superagonist TGN1412 trial'

David Eastwood - One of the best experts on this subject based on the ideXlab platform.

  • Cytokine release assays for the prediction of therapeutic mAb safety in first-in man trials--Whole blood cytokine release assays are poorly predictive for TGN1412 cytokine storm.
    Journal of immunological methods, 2015
    Co-Authors: Sandrine Vessillier, David Eastwood, Bernard Fox, Jean G. Sathish, Swaminathan Sethu, Thomas Dougall, Susan J. Thorpe, Robin Thorpe, R. Stebbings
    Abstract:

    The therapeutic monoclonal antibody (mAb) TGN1412 (anti-CD28 superagonist) caused near-fatal cytokine release syndrome (CRS) in all six volunteers during a phase-I clinical trial. Several cytokine release assays (CRAs) with reported predictivity for TGN1412-induced CRS have since been developed for the preclinical safety testing of new therapeutic mAbs. The whole blood (WB) CRA is the most widely used, but its sensitivity for TGN1412-like cytokine release was recently criticized. In a comparative study, using group size required for 90% power with 5% significance as a measure of sensitivity, we found that WB and 10% (v/v) WB CRAs were the least sensitive for TGN1412 as these required the largest group sizes (n = 52 and 79, respectively). In contrast, the peripheral blood mononuclear cell (PBMC) solid phase (SP) CRA was the most sensitive for TGN1412 as it required the smallest group size (n = 4). Similarly, the PBMC SP CRA was more sensitive than the WB CRA for muromonab-CD3 (anti-CD3) which stimulates TGN1412-like cytokine release (n = 4 and 4519, respectively). Conversely, the WB CRA was far more sensitive than the PBMC SP CRA for alemtuzumab (anti-CD52) which stimulates FcγRI-mediated cytokine release (n = 8 and 180, respectively). Investigation of potential factors contributing to the different sensitivities revealed that removal of red blood cells (RBCs) from WB permitted PBMC-like TGN1412 responses in a SP CRA, which in turn could be inhibited by the addition of the RBC membrane protein glycophorin A (GYPA); this observation likely underlies, at least in part, the poor sensitivity of WB CRA for TGN1412. The use of PBMC SP CRA for the detection of TGN1412-like cytokine release is recommended in conjunction with adequately powered group sizes for dependable preclinical safety testing of new therapeutic mAbs.

  • severity of the TGN1412 trial disaster cytokine storm correlated with il 2 release
    British Journal of Clinical Pharmacology, 2013
    Co-Authors: David Eastwood, Susan J. Thorpe, Robin Thorpe, Lucy Findlay, Chris Bird, Meenu Wadhwa, Stephen Poole, Paula Dilger, Jason Hockley, Richard Stebbings
    Abstract:

    Aim To determine if cytokine release with a solid phase assay is predictive of adverse responses for a range of therapeutic mAbs.

  • after TGN1412 recent developments in cytokine release assays
    Journal of Immunotoxicology, 2013
    Co-Authors: Richard Stebbings, David Eastwood, Stephen Poole, Robin Thorpe
    Abstract:

    The failure of regulatory science to keep pace with and support the development of new biological medicines was very publically highlighted in March 2006 when the first-in-man Phase I clinical trial of the immunomodulatory CD28-specific monoclonal antibody (mAb) TGN1412 ended in disaster when all six volunteers suffered a life-threatening adverse reaction termed a ‘Cytokine Storm’. The poor predictive value of standard pre-clinical safety tests and animal models applied to TGN1412 demonstrated the need for a new generation of immunotoxicity assays and animal models that are both sensitive and predictive of clinical outcome in man. The non-predictive result obtained from pre-clinical safety testing in cynomolgus macaques has now been attributed to a lack of CD28 expression on CD4+ effector memory T-cells that therefore cannot be stimulated by TGN1412. In contrast, high levels of CD28 are expressed on human CD4+ effector memory T-cells, the source of most TGN1412-stimulated pro-inflammatory cytokines. Stand...

  • antibody c region influences TGN1412 like functional activity in vitro
    Journal of Immunology, 2012
    Co-Authors: Christina Ball, David Eastwood, Bernard Fox, Lucy Findlay, Richard Stebbings, Stephen Poole, Giles Sharp, Simon E Hufton, Paul W H I Parren, Robin Thorpe
    Abstract:

    The unexpected outcome of the clinical trial of the superagonistic CD28 mAb TGN1412 (IgG4κ) continues to stimulate interest. We show that TGN1412 binds similarly to human and cynomolgus macaque FcγR, eliminating the possibility that differences in Fc-mediated interactions with FcγR contributed to the failure of preclinical testing in macaques to predict toxicity in humans. The influence of the Fc domain and C region structure on the in vitro functional activity of TGN1412 was investigated using F(ab′)2 and Fab fragments derived from TGN1412 recovered from the trial and recombinant TGN1412 subclass variants and mutants. Superagonistic activity, as measured by cytokine release and proliferation, was assessed by exposing PBMCs to immobilized mAbs/fragments or to aqueous mAbs/fragments in the presence of HUVEC monolayers. Removing the Fc generally curtailed or abolished PBMC activation. However, eliminating detectable FcγR-binding of the IgG4 by mutation (L235E) did not abrogate activity. Stabilizing the “wild-type” IgG4 hinge (S228P) enhanced activity without increasing FcγR binding, which could only partially be explained by inhibition of Fab arm–exchange. Subclass switching the IgG4 mAb to IgG1 decreased activity, whereas switching to IgG2 markedly increased activity. We conclude that the C region strongly influences in vitro CD28-mediated superagonistic signaling. Superagonism requires an intact Fc, as shown by the absence of activity of TGN1412 Fab and F(ab′)2 fragments, but, notably, appears to be relatively independent of FcγR-binding properties. We propose that the Fc, potentially through restricting flexibility, maintains a favorable V region conformation to allow superagonistic activity. These findings have important implications for Ab design strategies.

  • comparison of novel methods for predicting the risk of pro inflammatory clinical infusion reactions during monoclonal antibody therapy
    Journal of Immunological Methods, 2011
    Co-Authors: Lucy Findlay, David Eastwood, Susan J. Thorpe, Robin Thorpe, Christina Ball, Jane C Robinson, Chris Bird, Meenu Wadhwa, Richard Stebbings, Stephen Poole
    Abstract:

    Abstract Two methods for predicting the risk of pro-inflammatory clinical infusion reactions during monoclonal antibody therapy were evaluated. In the first, the antibody of interest is immobilised by air-drying onto 96-well plates prior to the addition of human peripheral blood mononuclear cells (PBMCs). In the second, the antibody is added in aqueous phase to a co-culture of human PBMCs and human endothelium-derived cells. In both methods the cells are incubated with the antibody to allow the accumulation of pro-inflammatory cytokines, quantified by enzyme-linked immunosorbent assay (ELISA). The antibodies associated with clinical infusion reactions, Herceptin, Campath-1H and TGN1412, gave the largest responses taking into account the data for all readouts (tumour necrosis factor-α, TNF, interleukin-6, IL-6, IL-8, IL-2 and cell proliferation) for both methods. Overall, the antibodies tested could be ranked as follows: Tysabri

Stephen Poole - One of the best experts on this subject based on the ideXlab platform.

  • severity of the TGN1412 trial disaster cytokine storm correlated with il 2 release
    British Journal of Clinical Pharmacology, 2013
    Co-Authors: David Eastwood, Susan J. Thorpe, Robin Thorpe, Lucy Findlay, Chris Bird, Meenu Wadhwa, Stephen Poole, Paula Dilger, Jason Hockley, Richard Stebbings
    Abstract:

    Aim To determine if cytokine release with a solid phase assay is predictive of adverse responses for a range of therapeutic mAbs.

  • after TGN1412 recent developments in cytokine release assays
    Journal of Immunotoxicology, 2013
    Co-Authors: Richard Stebbings, David Eastwood, Stephen Poole, Robin Thorpe
    Abstract:

    The failure of regulatory science to keep pace with and support the development of new biological medicines was very publically highlighted in March 2006 when the first-in-man Phase I clinical trial of the immunomodulatory CD28-specific monoclonal antibody (mAb) TGN1412 ended in disaster when all six volunteers suffered a life-threatening adverse reaction termed a ‘Cytokine Storm’. The poor predictive value of standard pre-clinical safety tests and animal models applied to TGN1412 demonstrated the need for a new generation of immunotoxicity assays and animal models that are both sensitive and predictive of clinical outcome in man. The non-predictive result obtained from pre-clinical safety testing in cynomolgus macaques has now been attributed to a lack of CD28 expression on CD4+ effector memory T-cells that therefore cannot be stimulated by TGN1412. In contrast, high levels of CD28 are expressed on human CD4+ effector memory T-cells, the source of most TGN1412-stimulated pro-inflammatory cytokines. Stand...

  • this information is current as preclinical testing of immunotherapeutics understanding the causes to improve monoclonal antibody TGN1412 better cytokine storm in the phase i trial of
    2013
    Co-Authors: Stephen Poole, Robin Thorpe, Meenu Wadhwa, Isabelle Cludts, J Thorpe, A F Bristow, Jane Robinson, Tony Meager, Carl Dolman, Bernard Fox
    Abstract:

    and Stephen PooleJ. Thorpe, Adrian Bristow, Meenu Wadhwa, Robin ThorpeTarrant, Jane Robinson, Tony Meager, Carl Dolman, Susan Dilger, Emily Liefooghe, Isabelle Cludts, Bernard Fox, GillEastwood, Chris Bird, David North, Yogesh Mistry, Paula Richard Stebbings, Lucy Findlay, Cherry Edwards, Davidhttp://www.jimmunol.org/content/179/5/3325J Immunol€2007; 179:3325-3331; ;Referenceshttp://www.jimmunol.org/content/179/5/3325.full#ref-list-1This article cites 11 articles, 3 of which you can access for free at: Subscriptionshttp://jimmunol.org/subscriptionsInformation about subscribing to The Journal of Immunology is online at: Permissionshttp://www.aai.org/ji/copyright.htmlSubmit copyright permission requests at: Email Alertshttp://jimmunol.org/cgi/alerts/etocReceive free email-alerts when new articles cite this article. Sign up at:

  • antibody c region influences TGN1412 like functional activity in vitro
    Journal of Immunology, 2012
    Co-Authors: Christina Ball, David Eastwood, Bernard Fox, Lucy Findlay, Richard Stebbings, Stephen Poole, Giles Sharp, Simon E Hufton, Paul W H I Parren, Robin Thorpe
    Abstract:

    The unexpected outcome of the clinical trial of the superagonistic CD28 mAb TGN1412 (IgG4κ) continues to stimulate interest. We show that TGN1412 binds similarly to human and cynomolgus macaque FcγR, eliminating the possibility that differences in Fc-mediated interactions with FcγR contributed to the failure of preclinical testing in macaques to predict toxicity in humans. The influence of the Fc domain and C region structure on the in vitro functional activity of TGN1412 was investigated using F(ab′)2 and Fab fragments derived from TGN1412 recovered from the trial and recombinant TGN1412 subclass variants and mutants. Superagonistic activity, as measured by cytokine release and proliferation, was assessed by exposing PBMCs to immobilized mAbs/fragments or to aqueous mAbs/fragments in the presence of HUVEC monolayers. Removing the Fc generally curtailed or abolished PBMC activation. However, eliminating detectable FcγR-binding of the IgG4 by mutation (L235E) did not abrogate activity. Stabilizing the “wild-type” IgG4 hinge (S228P) enhanced activity without increasing FcγR binding, which could only partially be explained by inhibition of Fab arm–exchange. Subclass switching the IgG4 mAb to IgG1 decreased activity, whereas switching to IgG2 markedly increased activity. We conclude that the C region strongly influences in vitro CD28-mediated superagonistic signaling. Superagonism requires an intact Fc, as shown by the absence of activity of TGN1412 Fab and F(ab′)2 fragments, but, notably, appears to be relatively independent of FcγR-binding properties. We propose that the Fc, potentially through restricting flexibility, maintains a favorable V region conformation to allow superagonistic activity. These findings have important implications for Ab design strategies.

  • comparison of novel methods for predicting the risk of pro inflammatory clinical infusion reactions during monoclonal antibody therapy
    Journal of Immunological Methods, 2011
    Co-Authors: Lucy Findlay, David Eastwood, Susan J. Thorpe, Robin Thorpe, Christina Ball, Jane C Robinson, Chris Bird, Meenu Wadhwa, Richard Stebbings, Stephen Poole
    Abstract:

    Abstract Two methods for predicting the risk of pro-inflammatory clinical infusion reactions during monoclonal antibody therapy were evaluated. In the first, the antibody of interest is immobilised by air-drying onto 96-well plates prior to the addition of human peripheral blood mononuclear cells (PBMCs). In the second, the antibody is added in aqueous phase to a co-culture of human PBMCs and human endothelium-derived cells. In both methods the cells are incubated with the antibody to allow the accumulation of pro-inflammatory cytokines, quantified by enzyme-linked immunosorbent assay (ELISA). The antibodies associated with clinical infusion reactions, Herceptin, Campath-1H and TGN1412, gave the largest responses taking into account the data for all readouts (tumour necrosis factor-α, TNF, interleukin-6, IL-6, IL-8, IL-2 and cell proliferation) for both methods. Overall, the antibodies tested could be ranked as follows: Tysabri