ALBERT

All Library Books, journals and Electronic Records Telegrafenberg

feed icon rss

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    Publication Date: 2015-12-03
    Description: The adoptive transfer of T cells engineered with chimeric antigen receptors (CARs) is currently considered as a highly promising therapeutic option for treatment of otherwise incurable malignant diseases. CARs combine the cellular and humoral arm of the immune response by assembling a single-chain fragment variable (scFv) as binding moiety which provides the antigen-specificity and an activating immune receptor. It has been demonstrated both in vitro and in vivo, that CAR engrafted effector T cells mediate long-lasting anti-tumor responses. Despite encouraging clinical efficacy targeting CD19 in recent clinical trials, the appearance of potentially life-threatening adverse reactions and the lack of control mechanisms once initiated, prevent more widespread application of the CAR technology. To overcome limitations of conventional CAR T cells, a unique chimeric antigen receptor (UniCAR) technology was developed (Fig. 1) which allows precise control of CAR T cell reactivity, thus lowering the risk of side effects while preserving efficacy. Moreover, the UniCAR technology enables the retargeting of engrafted T cells against more than one antigen simultaneously or subsequently, thus reducing the risk for development of antigen-loss tumor variants under treatment. The UniCAR technology splits the signaling and antigen-binding aspects of conventional CAR into two individual components. T cells are engineered to express a universal CAR (UniCAR), which has specificity for a short peptide motif of 10 amino acids derived from a human nuclear protein. Thus, T cells engineered to express UniCAR remain inactivated after re-infusion, since the UniCAR target is not available for binding under physiological conditions. The ultimate antigen-specificity of the system is provided separately by targeting modules (TMs) comprising a binding domain e.g., a tumor-antigen specific scFv, fused to the nuclear antigen motif recognized by the UniCAR binding domain. Here we provide first in vitro and in vivo prove of concept for this new approach. Antigen-specific redirection of T cells armed with the universal CAR in the presence of different targeting modules against various antigens (CD33, CD123, CD19, CD20, PSCA, PSMA,) was effective at femtomolar concentrations of the targeting module both. Taken together, the modular nature of UniCAR technology will allow retargeting of autologous, patient-derived T cells to several antigens under controlled pharmacological conditions and has the potential to become a highly effective treatment option for late stage cancer patients with reduced risks for side effects. Figure 1. Schematic representation of T cell recruitment with the modular UniCAR system. The UniCAR T cell recruitment system consists of two separated units. The first unit is the UniCAR expressed on T cells with a single-chain fragment variable (scFv) specific for a short 10 aa long peptide motif. The intracellular signalling domain of the UniCAR contains a costimulatory domain derived from CD28 and the T cell receptor z chain. The second unit is a targeting molecule (TM) which consists of a scFv fused to the peptide epitope. The cross-linkage of T cell and target cell is mediated by interaction between the UniCAR binding domain on T cells and target cell binding TM. Figure 1. Schematic representation of T cell recruitment with the modular UniCAR system. / The UniCAR T cell recruitment system consists of two separated units. The first unit is the UniCAR expressed on T cells with a single-chain fragment variable (scFv) specific for a short 10 aa long peptide motif. The intracellular signalling domain of the UniCAR contains a costimulatory domain derived from CD28 and the T cell receptor z chain. The second unit is a targeting molecule (TM) which consists of a scFv fused to the peptide epitope. The cross-linkage of T cell and target cell is mediated by interaction between the UniCAR binding domain on T cells and target cell binding TM. Disclosures Cartellieri: Cellex Patient Treatment GmbH: Employment. Loff:GEMoaB Monoclonals GmbH: Employment. Ehninger:GEMoaB Monoclonals GmbH: Employment, Patents & Royalties: related to the UniTARG system. Ehninger:GEMoaB Monoclonals GmbH: Equity Ownership; Cellex Patient Treatment GmbH: Equity Ownership. Bachmann:GEMoaB Monoclonals GmbH: Equity Ownership, Patents & Royalties: related to the UniTARG system.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 2
    Publication Date: 2014-12-06
    Description: Based on compelling evidence from a vast number of in vitro and in vivostudies, Tregs have become an attractive cell population to treat or even prevent auto- and alloimmunity including Graft-versus-Host disease (GvHD). However, several safety concerns still exist as for example the risk of global immunosuppression using polyclonal Tregs. In fact, experiments in mice showed that adoptive transfer or induction of antigen-specific Tregs is more potent regarding suppression of pathogenic immune responses when compared to polyclonal Treg populations. Unfortunately, the isolation and expansion of naturally occurring antigen-specific Tregs is technically difficult, labour-intensive, and time-consuming. An attractive way to overcome these limitations and to endow polyclonal Treg populations with a desired antigen-specificity is their engraftment with chimeric antigen receptors (CARs). In this context, CAR-modification represents a promising approach to redirect polyclonal Tregs in an antigen-specific manner to suppress ongoing self-destructive immune responses at the site of inflammation. Nevertheless, until now redirection of CAR-engineered T cells is limited to a single target antigen, restricting this approach to an unflexible monospecific therapy. Therefore, we developed a more flexible universal CAR (UCAR) platform that allows redirection of T cells to an in principal unrestricted number of surface antigens. T cells are engrafted with UCARs that bind to a small peptide epitope derived from a human nuclear protein. Cross-linkage to target cells is mediated by independent target modules that provide antigen-specificity and comprise the peptide epitope recognized by the UCAR. In order to target different tissue antigens, the target modules can easily be exchanged. Thereby, once established, the treatment strategy can easily be applied to various auto- and alloimmune diseases. At present, the CD45RA+ population is the Treg subset of choice for a clinical application as these cells have the highest capacity to maintain phenotypic and functional Treg properties upon prolonged ex vivo expansion. Here we show that highly pure, sorted CD4+CD25+CD127lowCD45RA+ Tregs can be genetically manipulated using lentiviral gene transfer, resulting in approximately 70 % of UCAR-expressing Treg cells. The transduction procedure itself did not affect the phenotype of UCAR-engineered Tregs as it was similar to non-transduced wildtype cells. Both Treg populations presevered FOXP3 expression even after prolonged in vitro cultivation (〉 95 % FOXP3+). Upon incubation with antigen-positive target cells and a respective target module UCAR-engineered Tregs upregulate the activation markers CD69 and LAP demonstrating that the cells can be restimulated antigen-specifically. Most importantly, UCAR-engrafted Tregs were functionally activated upon antigen encounter, demonstrated by suppression of proliferation and expansion of cocultured autologous T effector cells. Taken together, our results pave the way towards an application of UCAR technology for a site-specific recruitment of CAR-modified Tregs into inflamed tissues aiming at re-establishing immune homeostasis. Due to its high flexibility UCAR-engrafted Tregs can easily and universally be used for treatment of various autoimmune diseases or GvHD just by exchanging the tissue-specific target modules. Disclosures Cartellieri: Cellex Patient Treatment GmbH: Employment. Ehninger:GEMoaB GmbH: Employment, Patents & Royalties. Ehninger:GEMoaB GmbH: Consultancy, Patents & Royalties. Bachmann:GEMoaB GmbH: Consultancy, Patents & Royalties.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 3
    Publication Date: 2011-06-30
    Description: Donor lymphocyte infusions have been effective in patients with chronic myeloid leukemia (CML) relapsing after allogeneic stem cell transplantation, but their use is associated with the risk of graft-versus-host disease. We investigated the effects of prophylactic infusion of in vitro-generated donor T cells reactive against peptides derived from CML-associated antigens. Fourteen CML patients received conditioning therapy followed by CD34+-selected peripheral blood stem cells from matched siblings (n = 7) or unrelated (n = 7) donors. Donor-derived mature dendritic cells generated in vitro from CD14+ monocytes were loaded with human leukocyte Ag-restricted peptides derived from PR1, WT1, and/or B-cell receptor–ABL and used to repetitively stimulate donor CD8+ T cells in the presence of IL-2 and IL-7. Stimulated T cells were infused 28, 56, and 112 days after transplantation. Thirteen patients are alive and 7 remain in molecular remission (median follow-up, 45 months). Interestingly, all 4 patients receiving CD8+ T cells displaying marked cytotoxic activity in vitro and detectable peptide-reactive CD8+ T cells during follow-up have not experienced graft-versus-host disease or relapse. Our study reveals that prophylactic infusion of allogeneic CD8+ T cells reactive against peptides derived from CML-associated antigens is a safe and promising therapeutic strategy. This trial was registered at www.clinicaltrials.gov as #NCT00460629.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 4
    Publication Date: 2014-12-06
    Description: Despite many years of research and great advances in the field, acute myeloid leukemia (AML) still remains one of the most challenging battle fields in the context of hematologic malignancies treatment. Although AML patients initially respond to conventional chemotherapy, a complete remission is rarely achieved and 5-year survival rates remain low especially in elderly patients. Hence, there is a pressing need for novel effective strategies for AML treatment to prevent relapse and treat minimal residual disease (MRD). The use of recombinant bispecific antibodies (bsAbs) for retargeting effector T lymphocytes towards cancer cells is recently emerging as a promising immunotherapeutic approach for tumor treatment. This class of small molecules is designed to bind simultaneously to a pre-defined tumor-associated antigen (TAA) on tumor cells and the activating CD3 complex on T cells. The cross-linkage of immune effector cell and tumor cell leads to a tumor-specific T cell activation and efficient target cell killing independently of the T cell receptor specificity. However, due to their low molecular mass, bsAbs have a short life span in vivo and consequently have to be continuously administrated to patients over prolonged time spans of several weeks to achieve clinical responses. As an alternative to continuous exogenous infusions of short-lived Abs we examined the use of engineered bone marrow-derived human mesenchymal stem cells (hMSCs) as cellular vehicles for the constant production and secretion of a fully humanized anti-CD33-anti-CD3 bsAb that targets the surface molecule CD33, which is widely overexpressed on AML blasts. Our studies demonstrate that gene-modified hMSCs are effective in releasing the bsAb at sufficient amounts to activate and redirect both human primary CD4+ and CD8+T cells from healthy donors against AML cells expressing varying levels of the CD33 antigen, leading to an efficient T cell-mediated tumor cell killing at low effector to target cell ratios and Ab concentrations. Most importantly, we could demonstrate that patient-derived T cells were able to suppress autologous AML blasts upon Ab-mediated cross-linkage over prolonged period of time without being affected by the presence of the modified hMSCs. Additional improvement of this system was achieved by the artificial expression of T cell co-stimulatory 4-1BB ligand (CD137L) on the hMSCs surface. The additional co-stimulatory signal provided by the engineered hMSCs resulted in an enhanced T cell proliferation, a higher pro-inflammatory cytokine release, and consequently in a more pronounced specific tumor cell killing already at earlier time-points. Taken together, our data could demonstrate that continuous in situ delivery of the anti-CD33-anti-CD3 bsAb by genetically modified hMSCs facilitates efficient activation of T cells for specific and efficient killing of AML blasts over prolonged period of time. Furthermore, as promising perspective of this approach for future in vivo application we are currently investigating on the development of biocompatible synthetic scaffolds as transplantable biomaterial-based production platforms for genetically engineered hMSCs as locally confined vehicle of immunotherapeutics. The implantation of these small engineered devices would ensure that the delivery of the anti-cancer agents can be controlled and stopped after tumor clearance by removing the scaffold at a desired time point. In this way, administration of ex vivo gene-modified hMSCs embedded in appropriate scaffolds would result in a continuous in situ production of recombinant Abs for effective and persistent levels of these therapeutic agents over time with low risk of side effects. Disclosures Cartellieri: Cellex Patient Treatment GmbH, Dresden, Germany: Employment. Ehninger:GEMoaB Monoclonals GmbH, Dresden, Germany: Employment, Patents & Royalties. Ehninger:GEMoaB Monoclonals GmbH, Dresden, Germany: Consultancy, Patents & Royalties. Bachmann:GEMoaB Monoclonals GmbH, Dresden, Germany: Consultancy, Patents & Royalties.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 5
    Publication Date: 2015-12-03
    Description: Acute myeloid leukemia (AML) is a hematologic malignancy of the myeloid line with high prevalence in older patients. As complete eradication of metastatic cancer cells is often not achieved by standard therapies, alternative treatment modalities are urgently needed. In recent years, bispecific antibodies (bsAbs) and chimeric antigen receptors (CARs) emerged as promising candidates for an antigen-specific cancer immunotherapy. Both bsAbs and CARs are able to redirect T cells for efficient tumor cell lysis. Nevertheless, the development of a novel TAA specific bsAb or a CAR is a long lasting process. Therefore, we recently introduced a novel antibody-based modular platform (UniTARG) that can be rapidly and easily adapted for redirection of T cells to any TAA in both a bsAb or CAR related manner. The modular UniTARG system distributes the effector arm (the anti-CD3 domain or CAR) and the anti-TAA binding domain to two separate molecules: (I) an exchangeable target module (TM) comprising an anti-TAA binding moiety and a short peptide epitope (E5B9), and (II) a universal effector unit. The effector systems represent either a bsAb with specificity for CD3 and a peptide epitope (E5B9) termed UniMAB or a CAR directed to the E5B9 epitope (UniCAR). Thus, TMs can form a complex with the respective effector system that facilitates the cross-linkage of tumor and T cells similar to conventional bsAbs or CARs. For redirection of T cells to any kind of TAA only the binding moiety of the TM has to be adapted what saves costs and time. To increase tumor specificity and to reduce the risk of tumor escape variants, the modular UniTARG system further offers the possibility to apply simultaneously different monospecific or even bispecific TMs recognizing two TAAs. For proof of concept of a dual targeting using the UniTARG system we selected as TAA on AML blasts the molecules CD33 and CD123. They represent promising target antigens as they are overexpressed on both rapidly proliferating terminal AML blasts and leukemic stem cells which might be responsible for disease relapse after initial chemotherapy. Thus, we generated an anti-CD123 TM and anti-CD33 TM that can be applied within the modular system for single-targeting or that can be combined for dual-targeting of AML blasts. By fusion of the anti-CD123 and anti-CD33 domains via the E5B9 epitope a bispecific TM was further constructed. As revealed by cytotoxicity assays with CD33+ CD123+ AML cell lines, the novel mono- and bispecific TMs can be easily applied to the modular systems to trigger highly potent tumor cell lysis at low E:T ratios and picomolar Ab concentrations. By using the dual-targeting approach we can show that lysis of CD123+ CD33+ AML blasts can be considerably improved in comparison to the mono-specific strategy. Overall, due to the ease and cost-effectiveness of development the UniTARG platform technology represents a promising tool in the field of both bsAbs and CARs with the advantage of simultaneous or consecutive dual or even multispecific targeting. This approach might additionally improve anti-tumor activity by increasing tumor specificity and diminishing off-target effects. Disclosures Cartellieri: Cellex Patient Treatment GmbH: Employment. Ehninger:GEMoaB Monoclonals GmbH: Employment, Patents & Royalties: related to the UniTARG system. Ehninger:GEMoaB Monoclonals GmbH: Equity Ownership, Patents & Royalties: related to the UniTARG system. Bachmann:GEMoaB Monoclonals GmbH: Equity Ownership, Patents & Royalties: related to the UniTARG system.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 6
  • 7
  • 8
    Publication Date: 2021-03-25
    Description: According to the literature, the autoantigen La is involved in Cap-independent translation. It was proposed that one prerequisite for this function is the formation of a protein dimer. However, structural analyses argue against La protein dimers. Noteworthy to mention, these structural analyses were performed under reducing conditions. Here we describe that La protein can undergo redox-dependent structural changes. The oxidized form of La protein can form dimers, oligomers and even polymers stabilized by disulfide bridges. The primary sequence of La protein contains three cysteine residues. Only after mutation of all three cysteine residues to alanine La protein becomes insensitive to oxidation, indicating that all three cysteines are involved in redox-dependent structural changes. Biophysical analyses of the secondary structure of La protein support the redox-dependent conformational changes. Moreover, we identified monoclonal anti-La antibodies (anti-La mAbs) that react with either the reduced or oxidized form of La protein. Differential reactivities to the reduced and oxidized form of La protein were also found in anti-La sera of autoimmune patients.
    Print ISSN: 1661-6596
    Electronic ISSN: 1422-0067
    Topics: Chemistry and Pharmacology
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...