Search Results Heading

MBRLSearchResults

mbrl.module.common.modules.added.book.to.shelf
Title added to your shelf!
View what I already have on My Shelf.
Oops! Something went wrong.
Oops! Something went wrong.
While trying to add the title to your shelf something went wrong :( Kindly try again later!
Are you sure you want to remove the book from the shelf?
Oops! Something went wrong.
Oops! Something went wrong.
While trying to remove the title from your shelf something went wrong :( Kindly try again later!
    Done
    Filters
    Reset
  • Language
      Language
      Clear All
      Language
  • Subject
      Subject
      Clear All
      Subject
  • Item Type
      Item Type
      Clear All
      Item Type
  • Discipline
      Discipline
      Clear All
      Discipline
  • Year
      Year
      Clear All
      From:
      -
      To:
  • More Filters
8 result(s) for "Semmrich, Monika"
Sort by:
A bispecific IgG format containing four independent antigen binding sites
Bispecific antibodies come in many different formats, including the particularly interesting two-in-one antibodies, where one conventional IgG binds two different antigens. The IgG format allows these antibodies to mediate Fc-related functionality, and their wild-type structure ensures low immunogenicity and enables standard methods to be used for development. It is however difficult, time-consuming and costly to generate two-in-one antibodies. Herein we demonstrate a new approach to create a similar type of antibody by combining two different variable heavy (VH) domains in each Fab arm of an IgG, a tetra-VH IgG format. The VHs are used as building blocks, where one VH is placed at its usual position, and the second VH replaces the variable light (VL) domain in a conventional IgG. VH domains, binding several different types of antigens, were discovered and could be rearranged in any combination, offering a convenient “plug and play” format. The tetra-VH IgGs were found to be functionally tetravalent, binding two antigens on each arm of the IgG molecule simultaneously. This offers a new strategy to also create monospecific, tetravalent IgGs that, depending on antigen architecture and mode-of-action, may have enhanced efficacy compared to traditional bivalent antibodies.
Vectorized Treg-depleting αCTLA-4 elicits antigen cross-presentation and CD8+ T cell immunity to reject ‘cold’ tumors
BackgroundImmune checkpoint blockade (ICB) is a clinically proven concept to treat cancer. Still, a majority of patients with cancer including those with poorly immune infiltrated ‘cold’ tumors are resistant to currently available ICB therapies. Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) is one of few clinically validated targets for ICB, but toxicities linked to efficacy in approved αCTLA-4 regimens have restricted their use and precluded full therapeutic dosing. At a mechanistic level, accumulating preclinical and clinical data indicate dual mechanisms for αCTLA-4; ICB and regulatory T cell (Treg) depletion are both thought to contribute efficacy and toxicity in available, systemic, αCTLA-4 regimens. Accordingly, strategies to deliver highly effective, yet safe αCTLA-4 therapies have been lacking. Here we assess and identify spatially restricted exposure to a novel strongly Treg-depleting, checkpoint-blocking, vectorized αCTLA-4, as a highly efficacious and potentially safe strategy to target CTLA-4.MethodsA novel human IgG1 CTLA-4 antibody (4-E03) was identified using function-first screening for monoclonal antibodies (mAbs) and targets associated with superior Treg-depleting activity. A tumor-selective oncolytic vaccinia vector was then engineered to encode this novel, strongly Treg-depleting, checkpoint-blocking, αCTLA-4 antibody or a matching surrogate antibody, and Granulocyte-macrophage colony-stimulating factor (GM-CSF) (VVGM-αCTLA-4).ResultsThe identified 4-E03 antibody showed significantly stronger Treg depletion, but equipotent checkpoint blockade, compared with clinically validated αCTLA-4 ipilimumab against CTLA-4-expressing Treg cells in a humanized mouse model in vivo. Intratumoral administration of VVGM-αCTLA-4 achieved tumor-restricted CTLA-4 receptor saturation and Treg depletion, which elicited antigen cross-presentation and stronger systemic expansion of tumor-specific CD8+ T cells and antitumor immunity compared with systemic αCTLA-4 antibody therapy. Efficacy correlated with FcγR-mediated intratumoral Treg depletion. Remarkably, in a clinically relevant mouse model resistant to systemic ICB, intratumoral VVGM-αCTLA-4 synergized with αPD-1 to reject cold tumors.ConclusionOur findings demonstrate in vivo proof of concept for spatial restriction of Treg depletion-optimized immune checkpoint blocking, vectorized αCTLA-4 as a highly effective and safe strategy to target CTLA-4. A clinical trial evaluating intratumoral VVGM-αhCTLA-4 (BT-001) alone and in combination with αPD-1 in metastatic or advanced solid tumors has commenced.
Domain binding and isotype dictate the activity of anti-human OX40 antibodies
BackgroundPrevious data suggests that anti-OX40 mAb can elicit anti-tumor effects in mice through deletion of Tregs. However, OX40 also has powerful costimulatory effects on T cells which could evoke therapeutic responses. Human trials with anti-OX40 antibodies have shown that these entities are well tolerated but to date have delivered disappointing clinical responses, indicating that the rules for the optimal use of anti-human OX40 (hOX40) antibodies is not yet fully understood. Changes to timing and dosages may lead to improved outcomes; however, here we focus on addressing the role of agonism versus depleting activity in determining therapeutic outcomes. We investigated a novel panel of anti-hOX40 mAb to understand how these reagents and mechanisms may be optimized for therapeutic benefit.MethodsThis study examines the binding activity and in vitro activity of a panel of anti-hOX40 antibodies. They were further evaluated in several in vivo models to address how isotype and epitope determine mechanism of action and efficacy of anti-hOX40 mAb.ResultsBinding analysis revealed the antibodies to be high affinity, with epitopes spanning all four cysteine-rich domains of the OX40 extracellular domain. In vivo analysis showed that their activities relate directly to two key properties: (1) isotype—with mIgG1 mAb evoking receptor agonism and CD8+ T-cell expansion and mIgG2a mAb evoking deletion of Treg and (2) epitope—with membrane-proximal mAb delivering more powerful agonism. Intriguingly, both isotypes acted therapeutically in tumor models by engaging these different mechanisms.ConclusionThese findings highlight the significant impact of isotype and epitope on the modulation of anti-hOX40 mAb therapy, and indicate that CD8+ T-cell expansion or Treg depletion might be preferred according to the composition of different tumors. As many of the current clinical trials using OX40 antibodies are now using combination therapies, this understanding of how to manipulate therapeutic activity will be vital in directing new combinations that are more likely to improve efficacy and clinical outcomes.
1368 Preclinical development of an agonistic anti-TNFR2 antibody (BI-1910) for cancer immunotherapy
BackgroundThe pleiotropic TNF-α:TNFR axis plays a central role in the immune system. TNFR2 has been proposed to be both essential for the survival of T regs, as well as providing important co-stimulatory signals for T cell activation and memory generation. In addition, the therapeutic potential of targeting TNFR2 for cancer treatment has been previously indicated. To gain further insight, we characterized the biophysical properties and in vitro and in vivo activities of human and mouse α-TNFR2-specific antibodies designed to agonize the receptor.MethodsA human lead candidate (BI-1910) and a mouse surrogate (mBI-1910) α-TNFR2 were identified. Agonistic activity on T cells were demonstrated for both antibodies in vitro. mBI-1910 showed potent anti-tumor activity both as a single agent and in combination with anti-PD1 in multiple immunocompetent tumor models. The antibody showed co-stimulation through TNFR2, which enhanced T cell activation and induced CD8+ T cell-dependent anti-tumor effects. These findings were confirmed using BI-1910 in human TNFR2 transgenic mice.To address safety, a GLP toxicological study was performed in cynomolgus macaques. Three doses (1, 5, and 25 mg/kg) were given weekly for four consecutive weeks followed by a recovery period of eight weeks. In addition, cytokine release was studied in T cell stimulation assays and in a humanized mouse model. In parallel, multiple immune stimulation assays were studied in vitro using human cells to establish EC50 values and a clear relationship with dose, receptor occupancy and immune cell activation.ResultsFour administrations of BI-1910 to cynomolgus macaques were well tolerated at all doses, with no associated clinical signs and no signs of cytokine release. Pharmacokinetic studies demonstrated an expected human IgG half-life at receptor-saturating doses. Interestingly, there was a clear dose-dependent T cell activation, evidenced by an increase in several T cell activation markers and a shift from naïve to effector memory T cells supporting the proposed mode-of action. Importantly, the nature of BI-1910-induced T cell activation in cynomolgus macaques closely mirrored that in TNFR2 humanized mice, in which clear anti-tumor effects were also demonstrated.ConclusionsThe strong similarities in BI-1910 induced immune response between mice and cynomolgus macaques shows promise that similar T cell activation and following anti-tumor effects will occur also in humans. Collectively, these studies support the upcoming phase I/II study in solid cancer patients planned to start in H2 2023.Ethics ApprovalAll data utilizing human blood or animals was approved by an ethic committé before the experiments were started.Experiments using human blood were approved by the Regional committé for ethichs approval in Lund, Sweden ID numbers 2018/37 and 2010/356The murine experiments were approved by the Ethical committé of animal experiment in Lund and Malmö, Sweden ID numbers 5.8.18–19686/2022; 5.8.18–03333-2020; 5.8.18–02934-2020; 5.8.18–17196-2018The non-human primate experiments were approved by Charles River Laboratories Evreux Ethics Committee (CEC), France, ID number 2850398
725 Pre-clinical development of TNFR2 ligand-blocking BI-1808 for cancer immunotherapy
BackgroundThe pleiotropic TNF-alpha:TNFR axis plays a central role in the immune system. While the cellular expression of TNFR1 is broad, TNFR2 expression is mainly restricted to immune cells. The therapeutic potential of targeting TNFR2 for cancer treatment has been previously indicated and to gain further insight, we characterized a wide panel antibodies, generated from the n-CoDeR F.I.R.S.T™ target and antibody discovery platform. We identified parallel human and mouse TNFR2 specific, complete ligand (TNF-alpha) blocking antibodies and could show potent anti-tumor activity in several immune-competent models, both as single agent and in combination with anti-PD1 using a BI-1808 murine surrogate. The mechanism-of-action was shown to be FcgR dependent and likely mediated through a combination of intra-tumor T reg depletion, CD8+ T cell expansion and modulation of tumor-associated myeloid cells. These findings were confirmed using BI-1808 in a humanized mouse model.MethodsTo address safety of the human lead-candidate BI-1808 two toxicological studies were performed in cynomolgus monkeys. The first study was a dose-range-finding study and the second a GLP study where three doses (2, 20 and 200 mg/kg) were given weekly for four consecutive weeks followed by a recovery period of eight weeks. In addition, cytokine release was further studied in T cell stimulation assays and in a humanized mouse model. Moreover, the BI-1808 murine surrogate was used to study the relationship between dose, receptor occupancy (RO) and efficacy in immune competent mouse cancer experimental models.ResultsFour weekly administrations of BI-1808 to cynomolgus monkeys were well tolerated at all doses, with no associated clinical signs, and no histopathological changes. Non-adverse and reversible increases in neutrophil counts and decreases in T cells were observed at all dose levels. No drug-related adverse events were observed and consequently the NOAEL for BI-1808 was determined to be 200 mg/kg. Pharmacokinetic studies demonstrated an expected half-life of two weeks at receptor saturation. There were no indications of cytokine release in any of the systems tested. Finally, we could show that to achieve max therapeutic effect, sustained RO was needed for approximately two weeks, covering the time it takes to generate a full adaptive Immune response.ConclusionsThere is a clear association between RO and therapeutic effect and BI-1808 is well tolerated at doses associated with high and sustained RO. Collectively, these studies were used to determine the starting dose in upcoming phase I/II study in solid cancer aiming for first-patient in during December 2020.Ethics ApprovalThe study on cynomolgous monkeys was conducted by Citox/Charles River Laboratories in compliance with animal health regulations, in particular: Council Directive No. 2010/63/EU of 22 September 2010 and French decree No. 2013-118 of 01 February 2013 on the protection of animals used for scientific purposes. Studies in mice were approved by the Swedish Animal Experiment Ethics Board, ethical permit/ethical license numbers 5.2.18-17196/2018 and 5.8.18-03333/2020
594 BT-001, an oncolytic vaccinia virus armed with a Treg-depleting human recombinant anti-CTLA4 antibody and GM-CSF to target the tumor microenvironment
BackgroundCheckpoint inhibitor antibodies have improved survival in a variety of cancers, however, a great unmet need remains since only a small fraction of patients responds. Reasons for lack of efficacy are believed to include lack of tumor infiltrating immune cells, a notion supported by improved efficacy observed following combined checkpoint blockade with tumor oncolytic virotherapy which promotes intratumoral T cell infiltration. Oncolytic vaccinia viruses (oVV) also allow genetic encoding of transgenes. This is of special interest for therapeutic proteins exhibiting toxicological limitation or pharmacokinetic issues. Here, BioInvent and Transgene present a potentially safe and more efficacious strategy to combine checkpoint inhibition in the context of oncolytic virotherapy.MethodsUsing the F.I.R.S.T™ discovery platform we have isolated a human recombinant Treg-depleting antibody that has been vectorized alongside GM-CSF into the Invir.IO® oVV. This product named BT-001 consists of a Copenhagen double deleted vaccinia virus encoding the human CTLA4-specific antibody 4-E03 IgG1, which shows improved Treg-depletion compared with ipilimumab in a human PBMC-based NOG/SCID-transfer model. BT-001 also encodes GM-CSF, the cytokine expressed in clinically approved products. A surrogate murine mAb was vectorized into the same oVV (mBT-1) allowing for functional and mechanistic in vivo studies.ResultsOur studies demonstrate that 4-E03 and GM-CSF were expressed as functional molecules after infection by BT-001 of human tumor cell lines in vitro. Moreover, following intratumoral administration in immune competent and immune deficient mice transplanted with mouse or human tumors, transgene expression was sustained at levels associated with receptor saturation for days to weeks. In contrast, and supporting the tumor-selective nature of oVV, blood concentrations of anti-CTLA4 mAb were lower compared to those observed following i.v. administration of therapeutic doses of mAb. The in vivo anti-tumor activity of mBT-1 was assessed in multiple syngeneic mouse tumor models including CT26, EMT6, A20 and C38. Murine surrogate mBT-1 conferred cures in the majority of challenged mice irrespective of tumor origin. The excellent anti-tumoral profile depends on anti-CTLA4 expression and could be boosted by co-administration of anti-PD-1 mAb. Intratumoral treatment with mBT-1 also induces abscopal anti-tumor responses and protects against tumor rechallenge demonstrating a long-lasting systemic anti-tumor activity.ConclusionsA clinical batch of BT-001 has been produced and toxicological evaluation is ongoing. Transgene and BioInvent have applied for a clinical trial targeting injectable superficial tumors. Here, the tumor-localized delivery of anti-CTLA4 may allow a better tolerated and more effective combination therapy with antibodies targeting the PD-1/PDL1 axis.
Directed antigen targeting in vivo identifies a role for CD103+ dendritic cells in both tolerogenic and immunogenic T-cell responses
The αE integrin chain CD103 identifies a subset of migratory dendritic cells (DCs) in the gut, lung, and skin. To gain further understanding of the function of CD103+ DCs in regulating adaptive immunity in vivo, we coupled ovalbumin (OVA) to the CD103 antibody M290 (M290.OVA). Intraperitoneal injection of M290.OVA induced OVA-specific CD8+ and CD4+ T-cell proliferation in lymph nodes (LNs) of wild-type but not CD103−/− mice, or in mice depleted of CD11c+ cells. In the absence of maturation stimuli, systemic antigen targeting to CD103+ DCs led to tolerance of CD8+ T cells, whereas coadministration of adjuvant induced cytotoxic T-lymphocyte (CTL) immunity and antibody production. Mucosal intratracheal application of M290.OVA also induced T-cell proliferation in mediastinal LNs, yet the functional outcome was tolerance that inhibited subsequent development of allergic airway inflammation and immunoglobulin E (IgE) responses to inhaled OVA. These findings identify antigen targeting to CD103+ DCs as a potential strategy to regulate immune responses in nonlymphoid mucosal tissues.
Domain Binding and Isotype Dictate the Activity of Anti-human OX40 Antibodies
Previous data suggests that anti-OX40 mAb can elicit anti-tumour effects in mice through deletion of Tregs. However, OX40 also has powerful costimulatory effects on T cells which could evoke therapeutic responses. The contributions of these different effector mechanisms has not previously been systematically evaluated, particularly for mAb directed to human OX40. Therefore, we generated a novel human OX40 knock-in (KI) mouse to evaluate a panel of anti-hOX40 mAb and show that their activities relate directly to two key properties: 1) isotype; with mIgG1 mAb evoking receptor agonism and CD8+ T cell expansion and mIgG2a mAb evoking deletion of Treg and; 2) epitope; with membrane-proximal mAb delivering more powerful agonism. Intriguingly, both isotypes acted therapeutically in tumour models by engaging these different mechanisms. These findings highlight the significant impact of isotype and epitope on the modulation of anti-hOX40 mAb therapy, and indicate that CD8+ T cell expansion or Treg depletion might be preferable according to the composition of different tumours. Competing Interest Statement MSC is a retained consultant for BioInvent International and has performed educational and advisory roles for Baxalta and Boehringer Ingleheim. He has received research funding from Roche, Gilead, Bioinvent International and GSK. BF, IT, LM and MS are employees of Bioinvent International.