GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: OncoImmunology, Informa UK Limited, Vol. 2, No. 12 ( 2013-12), p. e26770-
    Type of Medium: Online Resource
    ISSN: 2162-402X
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2013
    detail.hit.zdb_id: 2645309-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2618-2618
    Abstract: Abstract 2618 Adoptive transfer of antigen-specific T cells emerged as an attractive strategy to provide cancer patients with immune cells of a desired specificity. The efficacy of such adoptive transfers has been demonstrated in clinical studies. However, enrichment and expansion of tumor-specific T cells are time-consuming and often ineffective, due to the low frequency of tumor-specific precursors in vivo. Alternatively, polyclonal T cells can be genetically modified with chimeric antigen receptors (CARs) to provide these cells with new antigen specificities. CARs consist of a binding moiety specifically recognizing a tumor cell surface antigen fused to a signaling chain derived from a lymphocyte activating receptor. The chimeric receptor approach is able to bypass many of the mechanisms by which tumors avoid immunorecognition, such as MHC down-regulation, lack of expression of costimulatory molecules, and induction of T cell suppression. Acute myeloid leukemia (AML) is an intrinsically resistant disease and even though the majority of the patients initially respond to chemotherapy, the 3-year survival rate is still low. A promising target for immunotherapy of AML is CD33, which is absent on normal pluripotent hematopoietic stem cells and normal tissues, but is present on leukemic blasts in 85–90 % of adult and pediatric AML cases independent of the subtype of AML. Novel human CD33-specific CARs were constructed by fusing a CD33 specific scFv in series with CD3ζ chain plus an additional costimulatory sequence derived from CD28 (Fig. 1A). Both native human CD8+ and CD4+ T cells engrafted with CD33-specific CARs exhibited antigen-specific cytokine secretion, proliferation and target cell lysis (Fig. 1B, C). Moreover, AML blast derived from patients were efficiently killed by allogeneic CAR-engrafted T cells (Fig. 1D). Next, the CD33-specific scFv was fully humanized and afterwards incorporated into the CAR constructs. With this humanized CAR engrafted T cells from AML patients could be redirected against CD33+ cell lines and autologous AML blasts. Upon antigen-recognition, the modified T cells secreted high amount of inflammatory cytokines and efficiently killed the target cells.Fig. 1:Human CAR-engrafted T cells mediate effector functions against CD33+ target cells.A. Schematic representation of the CD33-specific CARs. VL: variable light chain; VH: variable heavy chain; E-Tag: linker epitope in between the VL and the VH chain.Fig. 1:. Human CAR-engrafted T cells mediate effector functions against CD33+ target cells. / A. Schematic representation of the CD33-specific CARs. VL: variable light chain; VH: variable heavy chain; E-Tag: linker epitope in between the VL and the VH chain.B. Cytotoxic effector functions of CAR engrafted human CD8+ and CD4+ T cells against the CD33+ blast line U937 were measured in a standard chromium release assay after 6h of co-incubation.B. Cytotoxic effector functions of CAR engrafted human CD8+ and CD4+ T cells against the CD33+ blast line U937 were measured in a standard chromium release assay after 6h of co-incubation.C. Killing of patient-derived AML blast by allogeneic CAR engrafted T cells was measured in a flow cytometer by exact cell count after 48h of co-cultivation. Three independent donor/patient pairings are shown.C. Killing of patient-derived AML blast by allogeneic CAR engrafted T cells was measured in a flow cytometer by exact cell count after 48h of co-cultivation. Three independent donor/patient pairings are shown. Until now, one major obstacle for an adoptive therapy of genetically modified T cells is the limited amount of T cells that can be isolated from AML patients and modified in vitro. For an efficient in vitro expansion restricted to CAR modified T cells from patients we developed a new method based on a novel CAR-mediated strategy. For this purpose, magnetic beads were coated with an antibody recognizing an epitope (Fig. 1A, E-Tag) which we included in the linker domain between the heavy and the light chain of the scFv portion of the CAR. Adding such magnetic beads to cultures of CAR modified T cells, the CAR engrafted T cells were expanded to similar extends as polyclonal T cell populations with anti-CD3/anti-CD28 coated beads. Furthermore, the antibody-coated beads can be used to isolate the CAR engrafted T cells after expansion and get rid of any contaminating non-modified cells. It may also be useful for elimination of CAR expressing T cells in vivo if necessary. The feasibility of the described method was not limited to CD33 specific CARs but was also functional for CARs equipped with scFvs of other specificities. Therefore, it might be universally applicable for the expansion and preparation of CAR modified T cell grafts in vitro before adoptive transfer back in patients. Taken together, we describe novel humanized CD33-specific CARs that (I) can be specifically expanded, (II) specifically eliminated, if necessary, and (III) may therefore become a novel potent treatment option for a cellular therapy of AML patients. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal of Autoimmunity, Elsevier BV, Vol. 42 ( 2013-5), p. 105-116
    Type of Medium: Online Resource
    ISSN: 0896-8411
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2013
    detail.hit.zdb_id: 1468989-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 1528-1528
    Abstract: Abstract 1528 Acute myeloid leukemia (AML) is the most common diagnosed cancer affecting the myeloid line of immune cells in adults. Although chemotherapy initially leads to a remission of the disease in 60–80% of the cases, an enormous number of these patients (50–80%) relapse. Hence, new adjuvant therapeutic strategies for the elimination of minimal residual disease (MRD) are needed. One novel attractive approach is based on recombinant bispecific antibodies (bsAb). BsAb are able to mediate a direct cross-link between T cells and tumor cells. This in turn leads to a polyclonal, MHC- and TCR-independent activation of CD4+ and CD8+ T cells for the effective killing of the recognized target cells. A front runner in this field is the bsAb blinatumomab with dual specificity for CD3 and CD19 which is already used successfully in first clinical trials for the treatment of B-cell lymphomas. The development of a novel bsAb is usually very time-consuming and includes a series of individual optimization steps to achieve optimal reactivity with lowest possible side effects. The aim of the present work was therefore to establish a novel, more flexible, less time-consuming, modular cell targeting system and to compare its efficiency and sensitivity with classical bsAb. For the immunotargeting of AML by recombinant antibodies we selected the transmembrane glycoprotein CD33, because 90% of the AML patients have CD33-positive blasts irrespective of the underlying AML-subtype. We developed (i) a novel fully humanized bsAb with dual specificity for CD3 and CD33 and in addition (ii) a novel modular cell targeting system which consists of two different recombinant antibody components (figure 1). The first component (targeting module) is an exchangeable linker module composed of an anti-CD33 single-chain fragment variable (scFv) and a peptide epitope (E). The second component (universal effector module) is a bsAb with antigen binding specificity for the CD3-complex on T cells and for the peptide epitope of the scFv-based linker module. Together both molecules are able to form protein complexes similar to bsAb. Figure 1: Schematic representation of (i) the direct cross-linking system and (ii) the modular cell targeting system Figure 1:. Schematic representation of (i) the direct cross-linking system and (ii) the modular cell targeting system The functionality of both immunotargeting systems was examined by performing chromium (51Cr) release assays. As target cells both AML blasts from patients and artificially overexpressing CD33-positive tumor cells were used. Both the direct targeting system consisting of a classical novel humanized anti-CD33-anti-CD3 bsAb and the modular cell targeting system efficiently killed CD33-positive targets cells down to picomolar concentrations (figure 2). Figure 2: Tumor cell lysis mediated by classical bsAb and the modular cell targeting system was determined by performing a chromium release assay. For this purpose, CD33-positive, 51Cr loaded target cells were incubated with PBMCs as effector cells. Co-cultivation occurred either in the presence or absence of recombinant Ab by using an effector to target cell ratio of 10: 1 and different amounts of recombinant Abs. Figure 2:. Tumor cell lysis mediated by classical bsAb and the modular cell targeting system was determined by performing a chromium release assay. For this purpose, CD33-positive, 51Cr loaded target cells were incubated with PBMCs as effector cells. Co-cultivation occurred either in the presence or absence of recombinant Ab by using an effector to target cell ratio of 10: 1 and different amounts of recombinant Abs. Compared to the classical bsAb the novel modular cell targeting has a series of advantages: Just by replacing the targeting module the system can be easily modified for retargeting of T cells against other tumor antigens. Moreover, using linker modules with dual specificities enables a multi-specific targeting which may improve the targeting specificity and thereby might increase the success of an immunotherapy. Taken together, here we present for the first time two novel fully functional humanized, recombinant Ab-based systems for the immunotargeting of CD33+ AML blasts by CD3+ T cells. The obtained data indicate that both the direct cross-linking anti-CD3-anti-CD33 bsAb and the modular cell targeting system hold great potential as an adjuvant cancer immunotherapy for the elimination of MRD in AML-patients. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 4041-4041
    Abstract: Abstract 4041 Hematopoietic stem cell transplantation is a commonly used treatment for various hematological malignancies. However, a life-threatening complication following this therapy is the development of Graft versus Host Disease (GvHD), during which transplanted donor immune cells attack the host and lead to severe inflammatory responses and tissue damage. Given the key role of regulatory T cells (Tregs) in immune homeostasis and peripheral tolerance, the adoptive transfer of these cells may represent a promising therapeutic opportunity for the treatment of GvHD. This approach has been proven successful to enhance graft acceptance and prevent experimental GvHD in several animal models. However, it becomes increasingly evident that antigen-specific Tregs are more efficient than polyclonal Treg populations. The antigen-specificity enables the cells to exert their suppressive effect locally at the appropriate sites of inflammation. Furthermore, the application of antigen-specific Tregs might lower the risk of unfavourable systemic immunosuppression. Nevertheless, one of the main obstacles for their clinical use is the isolation and expansion of therapeutically relevant numbers of antigen-specific Tregs. In light of these arguments, bispecific antibodies (bsAb) could provide a promising tool for a target-dependent tissue specific redirection of polyclonal Tregs. BsAb redirect T cells to target cells by cross-linking their activating CD3 receptor and any chosen antigen on the surface of the target cell. Several studies have proven that CD8+ and CD4+ effector T cells can be successfully activated by bsAb both in vitro and in vivo. However, so far nobody has ever investigated whether Tregs can be redirected with bsAb. Using bsAb against CD3 and two different target antigens we first examined the expression of different activation associated markers on CD4+CD25+ Tregs. We could show that incubation of Tregs with a bsAb in the presence of the respective target antigen leads to the upregulation of CD25, CD69 and LAP (figure 1). Furthermore, we analyzed the cytokine production profile of the bsAb-activated Tregs. Culturing the cells together with target cells and a cross-linking bsAb triggers the release of the immunomodulatory cytokine IL-10. One prerequisite for the treatment of graft rejection and GvHD with bsAb-redirected Tregs is their suppressive efficacy upon antigen-specific activation via bsAb. Therefore we monitored the ability of bsAb-redirected Tregs to inhibit effector mechanisms of autologous T helper cells. Corresponding experiments could demonstrate the striking capacity of redirected Tregs to suppress proliferation, CD25 upregulation and cytokine production of co-cultured effector T cells (figure 2).Figure 1:Expression of different activation associated markers on Tregs incubated without (−) or with a bsAb (+) and the respective target antigen.Figure 1:. Expression of different activation associated markers on Tregs incubated without (−) or with a bsAb (+) and the respective target antigen.Figure 2:A) CFSE-labeled effector T cells (Teff) were cultured together with either unlabeled autologous effector T cells or Tregs at effector:suppressor ratios of 1:1 or 4:1 in the presence (+) or absence (−) of a bsAb and the respective target antigen. B) Cytokine secretion of bsAb-activated Teff in the presence (white bars) or absence (black bars) of autologous bsAb-activated Tregs.Figure 2:. A) CFSE-labeled effector T cells (Teff) were cultured together with either unlabeled autologous effector T cells or Tregs at effector:suppressor ratios of 1:1 or 4:1 in the presence (+) or absence (−) of a bsAb and the respective target antigen. B) Cytokine secretion of bsAb-activated Teff in the presence (white bars) or absence (black bars) of autologous bsAb-activated Tregs. Taken together, we give evidence for the first time that bsAb can redirect Tregs against a surface antigen independently of their T cell receptor specificity. In view of these results, an antigen- and/or site-specific retargeting of Tregs using bsAb may open novel therapeutic approaches for a long-term establishment of tolerance against allogenic transplants and therefore would offer a new treatment option for severe GvHD after allogenic stem cell transplantation. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 121, No. 13 ( 2013-03-28), p. 2462-2473
    Abstract: UTX regulates migration and hematopoiesis. Female UTX-KO mice show key features of myelodysplastic syndrome with chromosomal instability.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 4810-4810
    Abstract: 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.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Antibodies, MDPI AG, Vol. 1, No. 2 ( 2012-07-18), p. 172-198
    Type of Medium: Online Resource
    ISSN: 2073-4468
    Language: English
    Publisher: MDPI AG
    Publication Date: 2012
    detail.hit.zdb_id: 2661514-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: PLoS ONE, Public Library of Science (PLoS), Vol. 9, No. 4 ( 2014-4-3), p. e93745-
    Type of Medium: Online Resource
    ISSN: 1932-6203
    Language: English
    Publisher: Public Library of Science (PLoS)
    Publication Date: 2014
    detail.hit.zdb_id: 2267670-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 188, No. 3 ( 2012-02-01), p. 1551-1558
    Abstract: Bispecific Abs hold great potential for immunotherapy of malignant diseases. Because the first components of this new drug class are now entering clinical trials, all aspects of their mode of action should be well understood. Several studies proved that CD8+ and CD4+ effector T cells can be successfully redirected and activated against tumor cells by bispecific Abs both in vitro and in vivo. To our knowledge, this study provides the first evidence that bispecific Abs can also redirect and activate regulatory T cells against a surface Ag, independently of their TCR specificity. After cross-linking, via a bispecific Ab, redirected regulatory T cells upregulate the activation markers CD69 and CD25, as well as regulatory T cell-associated markers, like CTLA-4 and FOXP3. The activated regulatory T cells secrete the immunosuppressive cytokine IL-10, but, in contrast to CD8+ and CD4+ effector T cells, almost no inflammatory cytokines. In addition, the redirected regulatory T cells are able to suppress effector functions of activated autologous CD4+ T cells both in vitro and in vivo. Therefore, the potential risk for activation of regulatory T cells should be taken into consideration when bispecific Abs are applied for the treatment of malignant diseases. In contrast, an Ag/tissue-specific redirection of regulatory T cells with bispecific Abs holds great potential for the treatment of autoimmune diseases and graft rejection.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2012
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...