Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
Type of Medium
Language
  • 1
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2565-2565
    Abstract: 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.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 137, No. 22 ( 2021-06-3), p. 3145-3148
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: BMC Genomics, Springer Science and Business Media LLC, Vol. 15, No. 1 ( 2014-12)
    Abstract: A close match of the HLA alleles between donor and recipient is an important prerequisite for successful unrelated hematopoietic stem cell transplantation. To increase the chances of finding an unrelated donor, registries recruit many hundred thousands of volunteers each year. Many registries with limited resources have had to find a trade-off between cost and resolution and extent of typing for newly recruited donors in the past. Therefore, we have taken advantage of recent improvements in NGS to develop a workflow for low-cost, high-resolution HLA typing. Results We have established a straightforward three-step workflow for high-throughput HLA typing: Exons 2 and 3 of HLA-A, -B, -C, -DRB1, -DQB1 and -DPB1 are amplified by PCR on Fluidigm Access Array microfluidic chips. Illumina sequencing adapters and sample specific tags are directly incorporated during PCR. Upon pooling and cleanup, 384 samples are sequenced in a single Illumina MiSeq run. We developed “neXtype” for streamlined data analysis and HLA allele assignment. The workflow was validated with 1140 samples typed at 6 loci. All neXtype results were concordant with the Sanger sequences, demonstrating error-free typing of more than 6000 HLA loci. Current capacity in routine operation is 12,000 samples per week. Conclusions The workflow presented proved to be a cost-efficient alternative to Sanger sequencing for high-throughput HLA typing. Despite the focus on cost efficiency, resolution exceeds the current standards of Sanger typing for donor registration.
    Type of Medium: Online Resource
    ISSN: 1471-2164
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2014
    detail.hit.zdb_id: 2041499-7
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: The Prostate, Wiley, Vol. 74, No. 13 ( 2014-09), p. 1335-1346
    Abstract: Recently, we described a novel modular platform technology in which T cell‐recruitment and tumor‐targeting domains of conventional bispecific antibodies are split to independent components, a universal effector module (EM) and replaceable monospecific/monovalent target modules (TMs) that form highly efficient T cell‐retargeting complexes. Theoretically, our unique strategy should allow us to simultaneously retarget T cells to different tumor antigens by combining the EM with two or more different monovalent/monospecific TMs or even with bivalent/bispecific TMs, thereby overcoming limitations of a monospecific treatment such as the selection of target‐negative tumor escape variants. METHODS In order to advance our recently introduced prostate stem cell antigen (PSCA)‐specific modular system for a dual‐targeting of prostate cancer cells, two additional TMs were constructed: a monovalent/monospecific TM directed against the prostate‐specific membrane antigen (PSMA) and a bivalent/bispecific TM (bsTM) with specificity for PSMA and PSCA. The functionality of the novel dual‐targeting strategies was analyzed by performing T cell activation and chromium release assays. RESULTS Similar to the PSCA‐specific modular system, the novel PSMA‐specific modular system mediates an efficient target‐dependent and ‐specific tumor cell lysis at low E:T ratios and picomolar Ab concentrations. Moreover, by combination of the EM with either the bispecific TM directed to PSMA and PSCA or both monospecifc TMs directed to either PSCA or PSMA, dual‐specific targeting complexes were formed which allowed us to kill potential escape variants expressing only one or the other target antigen. CONCLUSIONS Overall, the novel modular system represents a promising tool for multiple tumor targeting. Prostate 74: 1335–1346, 2014 . © 2014 Wiley Periodicals, Inc.
    Type of Medium: Online Resource
    ISSN: 0270-4137 , 1097-0045
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2014
    detail.hit.zdb_id: 1494709-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 2209-2209
    Abstract: Despite remarkable therapeutic success using chimeric antigen receptor modified T-cells (CAR-T) in hematologic malignancies, targeting solid tumors still remains challenging, given their heterogeneity and immunosuppressive milieu, including the expression of inhibitory immune checkpoints such as PD-1/PD-L1. Preclinical models demonstrate that combining CAR-T therapy with checkpoint inhibitors targeting the PD-1/PD-L1 axis could be a promising strategy to enhance anti-cancer activity of CAR-T in solid tumors and on-going phase I trials are clinically evaluating this strategy. However, primary and acquired resistance to checkpoint blockade is frequently observed limiting its broad applicability and efficacy. In addition, lack of specificity results in frequent occurrence of immune-related adverse events affecting several organs. Alternatively, expression of PD-L1 by solid tumor cells could be an attractive target for CAR-T therapy, especially in order to prevent or reverse tumor re-growth and relapse after successful initial tumor cell lysis. The development of a rapidly switchable universal CAR-T platform (UniCAR) with enhanced safety features allowed us to explore the possibility to directly attack PD-L1 expressing solid tumor cells. An inherent key feature of the UniCAR-T platform is a rapid and reversible turn off mechanism (less than 4 hours) determined by the short pharmacokinetic half-life of soluble targeting modules (TMs) and fast internalization of cell-bound TMs. TMs provide the antigen-specificity for UniCAR gene-modified T-cells (UniCAR-T) and consist of an antigen-binding moiety, e.g. an scFv, linked to a small peptide motif recognized by the UniCAR. For redirecting UniCAR-T against the PD-1/PD-L1 axis, a PD-L1 specific TM (TM-PD-L1) was developed and its functionality confirmed in binding assays to human and murine PD-L1 using surface plasmon resonance as well as in cell-based cytotoxicity assays. Half maximal killing efficacy (EC50) mediated by TM-PD-L1 redirected UniCAR-T was in the low picomolar range. As PD-L1 is also expressed on healthy tissue, toxicity studies were performed in a humanized mouse model with human UniCAR-T. Humanized mice treated with TM-PD-L1 twice a day for ten consecutive days did not reveal any signs of toxicity. In a prostate cancer (PCa) xenograft model efficient suppression of tumor growth could be demonstrated by administration of TM-PD-L1. Our data are the first to demonstrate safety and feasibility of active targeting of PD-L1 expressed by solid tumors with CAR-T and that this approach is capable of inducing a significant anti-tumor response. Citation Format: Josephine Dietrich, Simon Loff, Johannes Spehr, Julia Riewaldt, Cordula Gründer, Maria Schreiber, Michael Bachmann, Gerhard Ehninger, Michael Pehl, Marc Cartellieri, Armin Ehninger. Rapidly switchable universal CAR-T cells with improved safety profile allow for active targeting of PD-L1 expressing solid tumors [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 2209.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 4232-4232
    Abstract: Chimeric antigen receptor T-cell (CAR-T) therapy targeting CD19 or BCMA is tremendously effective in treating hematological malignancies. However, the application of CAR-T beyond certain B-cell malignancies such as ALL, MM, lymphoma and CLL remains challenging due to the lack of tumor-specific antigens and the limited ability to control acute and potential long term CAR-T reactivity in patients. Therefore, a rapidly switchable universal CAR-T platform (UniCAR) was developed. In this system, antigen-specificity is provided by soluble adaptors of short pharmacokinetic half-life, termed targeting modules (TM), which redirect T-cells engineered to express a universal CAR against tumors, allowing for excellent controllability of CAR-T reactivity while maintaining the high anti-tumor activity of CAR-T cells. Here we present results from late stage pre-clinical characterization of UniCAR-T targeting CD123 (UniCAR-T-CD123) for treatment of acute leukemia. Pre-clinical characterization confirmed specificity and safety of the approach. UniCAR-T proofed to be per se inert and safe in clinically relevant in vivo toxicity models. Activation of UniCAR-T occurs solely in the presence of the CD123-specific TM (TM123) and upon cross-linkage to CD123 expressing leukemic cells. Specific lysis of leukemic target cell lines and primary patient material is induced in a dose-dependent manner at pico-molar TM123 concentrations. Notably, induction of cytokine release occurs at higher TM doses than on-set of target cell lysis, opening a therapeutic window for clinical application. In vivo efficacy of UniCAR-T re-directed against CD123 was proven in CDX and PDX mouse models. Interestingly, anti-leukemic responses of switch-controllable UniCAR-T were demonstrated to be comparable to conventional CD123-specific CAR-T in vitro at comparable doses. In contrast to conventional CD123 CAR-T, CD123-specific toxicity of TM-activated UniCAR-T towards hematopoietic progenitors was reversible and could be abrogated by withdrawal of TM, allowing for normal development of human hematopoiesis in a xenograft model. In summary, in vitro and in vivo evidence suggests that UniCAR-T-CD123 could provide strong efficacy against CD123 expressing hematological malignancies while providing excellent control and ensuring recovery of normal hematopoiesis post treatment. A phase IA dose-finding study is ongoing. Citation Format: Simon Loff, Jan-Erik Meyer, Johannes Spehr, Julia Riewaldt, Cordula Gründer, Maria Schreiber, Michael Bachmann, Michael Pehl, Gerhard Ehninger, Armin Ehninger, Marc Cartellieri. More than a bridging therapy: Targeting CD123 with rapidly switchable universal CAR-T cells for treatment of acute leukemia [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 4232.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 2176-2176
    Abstract: CAR-T cell therapy holds great promise for treating a wide range of malignancies. Nevertheless, the CAR-T approach faces multiple challenges, including lack of suitable targets, insufficient tumor penetration and a microenvironment hostile to CAR-T cells. Here we provide pre-clinical evidence for using a low-molecular-weight, chemically synthesized compound to re-target CAR-T cells against solid tumors in conjunction with the CD28 co-stimulatory domain to address several of these challenges. PSMA is a validated target for treatment of advanced and metastatic prostate cancer (PCa), but also broadly expressed on tumor neo-vasculature beyond PCa. PSMA is also known to be expressed on a number of normal tissues, albeit to a much lower extent, which necessitates additional safety mechanisms for CAR-T therapy. Therefore, a rapidly switchable universal CAR-T platform (UniCAR) was developed. In this system, antigen-specificity is provided by soluble adaptors termed targeting modules (TM), which redirect T-cells engineered to express a universal CAR in an antigen-specific manner against tumors. The TM explored in the present study incorporates a clinically well-characterized radiotracer peptide motif binding to the enzymatic groove of PSMA. The small-sized TM has favorable pharmacokinetic and pharmacodynamics properties (short half-life of & lt; 30 min; rapid internalization in & lt; 1h), which allows a fast silencing and excellent controllability of UniCAR-T reactivity ( & lt; 4 h). Furthermore, the small molecule TM efficiently penetrates and rapidly accumulates within solid tumors as clinically demonstrated by related radiotracers. Results from our pre-clinical in vivo model demonstrate potent recruitment of UniCAR-T into tumor tissue by the TM and an efficient anti-tumor response. Of note, it was recently shown that the chosen PCa model suppresses CAR-T response by secreting high levels of transforming growth factor beta (TGF-β), creating an immunosuppressive milieu. We could overcome immunosuppression utilizing CD28 compared to 4-1-BB in the intracellular signaling domain of the UniCAR. Administered CD28/zeta UniCAR efficiently suppressed tumor growth in our in vivo model and could be re-activated by consecutive cycles of TM administration even after several weeks. In contrast to CARs with fixed binding moieties, which continuously signal and thereby induce T- cell exhaustion, the discontinuous activation of UniCAR keeps a balanced mix of T-effector and T-memory cells. In summary, our rapidly switchable universal CAR-T platform in combination with a low-molecular-weight TM allows to target less differentially expressed solid tumor antigens. Furthermore, incorporation of the CD28 costimulatory domain overcomes the immunosuppressive tumor microenvironment while discontinuous activation of UniCAR-T prevents exhaustion. Citation Format: Marc Cartellieri, Simon Loff, Johannes Spehr, Mridula Swayampakula, Julia Riewaldt, Cordula Gründer, Maria Schreiber, Michael Bachmann, Anja Feldmann, Michael Pehl, Gerhard Ehninger, Armin Ehninger. Using a PSMA-specific low-molecular-weight compound for prostate cancer treatment with rapidly switchable universal CAR-T cells: Overcoming the challenges of cellular immunotherapies in solid tumors [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 2176.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 1506-1506
    Abstract: Background: Conventional CAR-T cells targeting CD123 in rrAML have achieved objective responses, but led to long-lasting myelosuppression due to expression of CD123 on progenitor cells. UniCAR-T-CD123 is a rapidly switchable two-component CAR-T therapy. An inert universal CAR-T cell (UniCAR-T) is combined with a CD123-specific soluble targeting module with a short half-life (TM123). By administering or withholding the continuous infusion of TM123, the UniCAR-T-cell can be rapidly switched on and off. Within the ongoing Phase IA study in rrAML, we investigated the expansion kinetics of UniCAR-T cells during TM123 administration in peripheral blood and bone marrow as well as cytokine profiles of treated patients. Methods: Prior to administration of autologous UniCAR-T cells, patients received a standard Flu/Cy lymphodepletion regimen at day -5 to -3. TM123 was administered as continuous infusion over 24 days starting at day 0. At day 1 a single dose of UniCAR-T cells was given. Dosing started with 1 x 108 UniCAR-T cells and 0.5 mg TM123 per day in patient 1. Patient 2 received the same TM123 dose and a UniCAR-T dose of 2.5 x 108 cells. Patient 3 received the same cell dose as patient 2 but a higher TM123 dose (1 mg/day). Pharmacokinetic of UniCAR-T and TM123 was determined from peripheral blood and bone marrow by droplet digital PCR and TM123-specific ELISA, respectively. Cytokine levels were measured by microfluidic immunoassay. Results: All 3 patients treated so far achieved an objective response, with one showing a PR and two a CRi. Treatment proved to be tolerable, no DLTs were observed to date and adverse events were mild. Grade 1 CRS (fever) was observed in 2 patients but subsided within 48 h after use of antipyretics. Myelosuppression was observed starting after lymphodepletion, which immediately recovered after TM123 withdrawal on day 24 in all patients, providing evidence for the rapid off-switch of UniCAR-T cells post TM123 administration. UniCAR-T cells expanded in all patients in peripheral blood and bone marrow comparable to data reported for conventional CD123 CAR-T products, and were so far detectable for up to 6 months after administration. Expansion kinetics were TM123-dependent. Patients showed periods of transient increase of IL-6, IFN-γ and TNF-α levels preceding peak expansion and decreasing after termination of TM123 administration. One patient showed additional coinciding momentary elevation of GM-CSF and IL-2. Conclusions: The initial clinical and translational results of UniCAR-T-CD123 represent, to our understanding, a first time evidence for a well-tolerated and effective rapidly switchable CAR-T product. Even though the number of patients treated so far is limited, the data obtained provide clinical proof-of-concept for the opportunity to abrogate side effects by withdrawal of TM123. Enrollment into the Phase IA study is ongoing. Citation Format: Armin Ehninger, Julia Riewaldt, Cordula Gründer, Kristin Franke, Maria Schreiber, Martin Wermke, Sabrina Kraus, Carla Kreissig, Jan Koedam, Michael Pehl, Gerhard Ehninger, Marc Cartellieri. Expansion kinetics and cytokine profiles of UniCAR-T-CD123, a rapidly switchable two-component CAR-T therapy, in patients with relapsed/refractory AML [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1506.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Oncotarget, Impact Journals, LLC, Vol. 9, No. 7 ( 2018-01-26), p. 7487-7500
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2018
    detail.hit.zdb_id: 2560162-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages