1
|
Kubeil M, Neuber C, Starke M, Arndt C, Rodrigues Loureiro L, Hoffmann L, Feldmann A, Bachmann M, Pietzsch J, Comba P, Stephan H. 64Cu tumor labeling with hexadentate picolinic acid-based bispidine immunoconjugates. Chemistry 2024:e202400366. [PMID: 38506263 DOI: 10.1002/chem.202400366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 03/21/2024]
Abstract
Discussed are two picolinate appended bispidine ligands (3,7-diazabicyclo[3.3.1]nonane derivatives) in comparison with an earlier described bis-pyridine derivative, which are all known to strongly bind CuII. The radiopharmacological characterization of the two isomeric bispidine complexes includes quantitative labeling with 64CuII at ambient conditions with high radiochemical purities and yields (molar activities >200 MBq/nmol). Challenge experiments in presence of EDTA, cyclam, human serum and SOD demonstrate high stability and inertness of the 64Cu-bispidine complexes. Biodistribution studies performed in Wistar rats indicate a rapid renal elimination for both 64Cu-labeled chelates. The bispidine ligand with the picolinate group in N7 position was selected for further biological experiments, and its backbone was therefore substituted with a benzyl-NCS group at C9. Two tumor target modules (TMs), targeting prostate stem cell antigen (PSCA), overexpressed in prostate cancer, and the fibroblast activation protein (FAP) in fibrosarcoma, were selected for thiourea coupling with the NCS-functionalized ligand and lysine residues of TMs. Small animal PET experiments on tumor-bearing mice showed specific accumulation of the 64Cu-labeled TMs in PSCA- and FAP-overexpressing tumors (standardized uptake value (SUV) for PC3: 2.7±0.6 and HT1080: 7.2±1.25) with almost no uptake in wild type tumors.
Collapse
Affiliation(s)
- Manja Kubeil
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Christin Neuber
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Miriam Starke
- Universität Heidelberg, Anorganisch-Chemisches, Institut INF 270, 69120, Heidelberg, Germany
| | - Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, Technische Universiät Dresden, 01307, Dresden, Germany
| | - Liliana Rodrigues Loureiro
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Lydia Hoffmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, 01069, Dresden, Germany
| | - Peter Comba
- Universität Heidelberg, Anorganisch-Chemisches, Institut INF 270, 69120, Heidelberg, Germany
- Universität Heidelberg, Interdisciplinary Center for Scientific Computing, INF 205, 69120, Heidelberg, Germany
| | - Holger Stephan
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| |
Collapse
|
2
|
Tomita U, Ishimoto Y, Ri M, Kawase Y, Hizukuri Y, Maru C, Nanai K, Nakamura R, Nakayama M, Oguchi-Oshima K, Sumi H, Ohtsuka T, Iida S, Agatsuma T. A novel T cell-redirecting anti-GPRC5D × CD3 bispecific antibody with potent antitumor activity in multiple myeloma preclinical models. Sci Rep 2024; 14:5135. [PMID: 38429446 PMCID: PMC10907593 DOI: 10.1038/s41598-024-55143-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/20/2024] [Indexed: 03/03/2024] Open
Abstract
G-protein-coupled receptor class 5 member D (GPRC5D) is detected in malignant plasma cells in approximately 90% of patients diagnosed with multiple myeloma (MM). Here, we constructed BsAb5003, a novel humanized bispecific monoclonal antibody targeting CD3 and GPRC5D, and evaluated its therapeutic impact on MM. BsAb5003 induced specific cytotoxicity of GPRC5D-positive MM cells with concomitant T cell activation and cytokine release. The efficacy of BsAb5003 was associated with GPRC5D expression levels in MM cell lines. Flow cytometry analysis of bone marrow mononuclear cells (BMMNCs) from 49 MM patients revealed that GPRC5D was expressed in a wide population of MM patients, including heavily treated and high-risk patients. In ex vivo assays using BMMNCs, BsAb5003 induced potent efficacy against CD138 + MM cells in both newly diagnosed and relapsed/refractory patient samples in a GPRC5D expression-dependent manner. BsAb5003 significantly enhanced T cell activation and cytokine production in combination with immunomodulatory drugs (IMiDs) against MM cell lines. BsAb5003 also demonstrated significant inhibition of in vivo tumor growth by recruiting T cells. Taken together, these results suggest that T cell-redirecting bispecific antibody targeting GPRC5D as monotherapy and combination therapy with IMiDs could be a highly potent and effective treatment approach for a wide population of MM patients.
Collapse
Affiliation(s)
| | | | - Masaki Ri
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | | | | | | | | | | | | | | | | | | | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | | |
Collapse
|
3
|
Logghe T, van Zwol E, Immordino B, Van den Cruys K, Peeters M, Giovannetti E, Bogers J. Hyperthermia in Combination with Emerging Targeted and Immunotherapies as a New Approach in Cancer Treatment. Cancers (Basel) 2024; 16:505. [PMID: 38339258 PMCID: PMC10854776 DOI: 10.3390/cancers16030505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Despite significant advancements in the development of novel therapies, cancer continues to stand as a prominent global cause of death. In many cases, the cornerstone of standard-of-care therapy consists of chemotherapy (CT), radiotherapy (RT), or a combination of both. Notably, hyperthermia (HT), which has been in clinical use in the last four decades, has proven to enhance the effectiveness of CT and RT, owing to its recognized potency as a sensitizer. Furthermore, HT exerts effects on all steps of the cancer-immunity cycle and exerts a significant impact on key oncogenic pathways. Most recently, there has been a noticeable expansion of cancer research related to treatment options involving immunotherapy (IT) and targeted therapy (TT), a trend also visible in the research and development pipelines of pharmaceutical companies. However, the potential results arising from the combination of these innovative therapeutic approaches with HT remain largely unexplored. Therefore, this review aims to explore the oncology pipelines of major pharmaceutical companies, with the primary objective of identifying the principal targets of forthcoming therapies that have the potential to be advantageous for patients by specifically targeting molecular pathways involved in HT. The ultimate goal of this review is to pave the way for future research initiatives and clinical trials that harness the synergy between emerging IT and TT medications when used in conjunction with HT.
Collapse
Affiliation(s)
- Tine Logghe
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
| | - Eke van Zwol
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
| | - Benoît Immordino
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, San Giuliano, 56017 Pisa, Italy
- Institute of Life Sciences, Sant’Anna School of Advanced Studies, 56127 Pisa, Italy
| | | | - Marc Peeters
- Department of Oncology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Elisa Giovannetti
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, San Giuliano, 56017 Pisa, Italy
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Johannes Bogers
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
- Laboratory of Cell Biology and Histology, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
4
|
Loureiro LR, Hoffmann L, Neuber C, Rupp L, Arndt C, Kegler A, Kubeil M, Hagemeyer CE, Stephan H, Schmitz M, Feldmann A, Bachmann M. Immunotheranostic target modules for imaging and navigation of UniCAR T-cells to strike FAP-expressing cells and the tumor microenvironment. J Exp Clin Cancer Res 2023; 42:341. [PMID: 38102692 PMCID: PMC10722841 DOI: 10.1186/s13046-023-02912-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/21/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cells are a promising approach in cancer immunotherapy, particularly for treating hematologic malignancies. Yet, their effectiveness is limited when tackling solid tumors, where immune cell infiltration and immunosuppressive tumor microenvironments (TME) are major hurdles. Fibroblast activation protein (FAP) is highly expressed on cancer-associated fibroblasts (CAFs) and various tumor cells, playing an important role in tumor growth and immunosuppression. Aiming to modulate the TME with increased clinical safety and effectiveness, we developed novel small and size-extended immunotheranostic UniCAR target modules (TMs) targeting FAP. METHODS The specific binding and functionality of the αFAP-scFv TM and the size-extended αFAP-IgG4 TM were assessed using 2D and 3D in vitro models as well as in vivo. Their specific tumor accumulation and diagnostic potential were evaluated using PET studies after functionalization with a chelator and suitable radionuclide. RESULTS The αFAP-scFv and -IgG4 TMs effectively and specifically redirected UniCAR T-cells using 2D, 3D, and in vivo models. Moreover, a remarkably high and specific accumulation of radiolabeled FAP-targeting TMs at the tumor site of xenograft mouse models was observed. CONCLUSIONS These findings demonstrate that the novel αFAP TMs are promising immunotheranostic tools to foster cancer imaging and treatment, paving the way for a more convenient, individualized, and safer treatment of cancer patients.
Collapse
Affiliation(s)
- Liliana R Loureiro
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
| | - Lydia Hoffmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Christin Neuber
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Luise Rupp
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Claudia Arndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Alexandra Kegler
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Manja Kubeil
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Christoph E Hagemeyer
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia
| | - Holger Stephan
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anja Feldmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
5
|
Soto KEG, Loureiro LR, Bartsch T, Arndt C, Kegler A, Mitwasi N, Drewitz L, Hoffmann L, Saleh HA, Crespo E, Mehnert M, Daglar C, Abken H, Momburg F, Bachmann M, Feldmann A. Targeting colorectal cancer cells using AND-gated adaptor RevCAR T-cells. Front Immunol 2023; 14:1302354. [PMID: 38169746 PMCID: PMC10758449 DOI: 10.3389/fimmu.2023.1302354] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024] Open
Abstract
Despite the success of chimeric antigen receptor (CAR) T-cells especially for treating hematological malignancies, critical drawbacks, such as "on-target, off-tumor" toxicities, need to be addressed to improve safety in translating to clinical application. This is especially true, when targeting tumor-associated antigens (TAAs) that are not exclusively expressed by solid tumors but also on hea9lthy tissues. To improve the safety profile, we developed switchable adaptor CAR systems including the RevCAR system. RevCAR T-cells are activated by cross-linking of bifunctional adaptor molecules termed target modules (RevTM). In a further development, we established a Dual-RevCAR system for an AND-gated combinatorial targeting by splitting the stimulatory and co-stimulatory signals of the RevCAR T-cells on two individual CARs. Examples of common markers for colorectal cancer (CRC) are the carcinoembryonic antigen (CEA) and the epithelial cell adhesion molecule (EpCAM), while these antigens are also expressed by healthy cells. Here we describe four novel structurally different RevTMs for targeting of CEA and EpCAM. All anti-CEA and anti-EpCAM RevTMs were validated and the simultaneous targeting of CEA+ and EpCAM+ cancer cells redirected specific in vitro and in vivo killing by Dual-RevCAR T-cells. In summary, we describe the development of CEA and EpCAM specific adaptor RevTMs for monospecific and AND-gated targeting of CRC cells via the RevCAR platform as an improved approach to increase tumor specificity and safety of CAR T-cell therapies.
Collapse
Affiliation(s)
- Karla E. G. Soto
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Liliana R. Loureiro
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Tabea Bartsch
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Claudia Arndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Alexandra Kegler
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Nicola Mitwasi
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Laura Drewitz
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Lydia Hoffmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Haidy A. Saleh
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Eugenia Crespo
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Maria Mehnert
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Cansu Daglar
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Hinrich Abken
- Department of Gene-Immunotherapy, Leibniz-Institute of Immunotherapy, and University Regensburg, Regensburg, Germany
| | - Frank Momburg
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center, Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital, Heidelberg, Germany
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), partner site Dresden, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
| | - Anja Feldmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), partner site Dresden, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
| |
Collapse
|
6
|
Maghsoodi N, Zareinejad M, Golestan A, Mahmoudi Maymand E, Ramezani A. Anti-CD19/CD8 bispecific T cell engager for the potential treatment of B cell malignancies. Cell Immunol 2023; 393-394:104787. [PMID: 37976975 DOI: 10.1016/j.cellimm.2023.104787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023]
Abstract
The administration of blinatumomab was accompanied by several adverse effects, including activation of regulatory T-cells and cytokine storm. The objective of this study was to produce and evaluate a novel αCD8/CD19 BiTE (αCD8/CD19) with the potency to directly target CD8+T-cells. In-silico studies were utilized for determining proper folding, receptor binding, and structural stability of αCD8/CD19 protein. Western blotting and indirect surface staining were used to evaluate the size accuracy and binding potency of the purified protein. Functionality was assessed for granzyme B production, cytotoxicity, and proliferation. TheαCD8/CD19recombinant protein was produced in the CHO-K1 cell line with a final concentration of 1.94 mg/l. The αCD8/CD19 bound to CD8+and CD19+cell lines and induced significant granzyme B production, cytotoxic activity and proliferation potential in the presence of IL-2 and tumor target cells. The maximum CD8+T-cell biological activity was observed on the 10th day with 10:1 effector-to-target ratio.
Collapse
Affiliation(s)
- Nafiseh Maghsoodi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mohammadrasul Zareinejad
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Ali Golestan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Elham Mahmoudi Maymand
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Amin Ramezani
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran.
| |
Collapse
|
7
|
Peschke JC, Bergmann R, Mehnert M, Gonzalez Soto KE, Loureiro LR, Mitwasi N, Kegler A, Altmann H, Wobus M, Máthé D, Szigeti K, Feldmann A, Bornhäuser M, Bachmann M, Fasslrinner F, Arndt C. FLT3-directed UniCAR T-cell therapy of acute myeloid leukaemia. Br J Haematol 2023; 202:1137-1150. [PMID: 37460273 DOI: 10.1111/bjh.18971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/23/2023] [Accepted: 06/28/2023] [Indexed: 09/12/2023]
Abstract
Adaptor chimeric antigen receptor (CAR) T-cell therapy offers solutions for improved safety and antigen escape, which represent main obstacles for the clinical translation of CAR T-cell therapy in myeloid malignancies. The adaptor CAR T-cell platform 'UniCAR' is currently under early clinical investigation. Recently, the first proof of concept of a well-tolerated, rapidly switchable, CD123-directed UniCAR T-cell product treating patients with acute myeloid leukaemia (AML) was reported. Relapsed and refractory AML is prone to high plasticity under therapy pressure targeting one single tumour antigen. Thus, targeting of multiple tumour antigens seems to be required to achieve durable anti-tumour responses, underlining the need to further design alternative AML-specific target modules (TM) for the UniCAR platform. We here present the preclinical development of a novel FMS-like tyrosine kinase 3 (FLT3)-directed UniCAR T-cell therapy, which is highly effective for in vitro killing of both AML cell lines and primary AML samples. Furthermore, we show in vivo functionality in a murine xenograft model. PET analyses further demonstrate a short serum half-life of FLT3 TMs, which will enable a rapid on/off switch of UniCAR T cells. Overall, the presented preclinical data encourage the further development and clinical translation of FLT3-specific UniCAR T cells for the therapy of AML.
Collapse
Affiliation(s)
- J C Peschke
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Partner Site, Dresden, Germany
| | - R Bergmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - M Mehnert
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - K E Gonzalez Soto
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - L R Loureiro
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - N Mitwasi
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - A Kegler
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - H Altmann
- National Center for Tumor Diseases Dresden (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Partner Site, Dresden, Germany
- Medical Clinic and Polyclinic I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - M Wobus
- Medical Clinic and Polyclinic I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - D Máthé
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
- Hungarian Centre of Excellence for Molecular Medicine, In Vivo Imaging Advanced Core Facility, Szeged, Hungary
| | - K Szigeti
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - A Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Partner Site, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - M Bornhäuser
- National Center for Tumor Diseases Dresden (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Partner Site, Dresden, Germany
- Medical Clinic and Polyclinic I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
- School of Cancer and Pharmaceutical Science, King's College, London, UK
| | - M Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC): German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Partner Site, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - F Fasslrinner
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- Medical Clinic and Polyclinic I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - C Arndt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| |
Collapse
|
8
|
Grasso L, Jiang Q, Hassan R, Nicolaides NC, Kline JB. NAV-003, a bispecific antibody targeting a unique mesothelin epitope and CD3ε with improved cytotoxicity against humoral immunosuppressed tumors. Eur J Immunol 2023; 53:e2250309. [PMID: 37146241 PMCID: PMC10524251 DOI: 10.1002/eji.202250309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/07/2023]
Abstract
Mesothelin (MSLN) is a cell surface protein overexpressed in a number of cancer types. Several antibody- and cellular-based MSLN targeting agents have been tested in clinical trials where their therapeutic efficacy has been moderate at best. Previous studies using antibody and Chimeric Antigen Receptor-T cells (CAR-T) strategies have shown the importance of particular MSLN epitopes for optimal therapeutic response, while other studies have found that certain MSLN-positive tumors can produce proteins that can bind to subsets of IgG1-type antibodies and suppress their immune effector activities. In an attempt to develop an improved anti-MSLN targeting agent, we engineered a humanized divalent anti-MSLN/anti-CD3ε bispecific antibody that avoids suppressive factors, can target a MSLN epitope proximal to the tumor cell surface, and is capable of effectively binding, activating, and redirecting T cells to the surface of MSLN-positive tumor cells. NAV-003 has shown significantly improved tumor cell killing against lines producing immunosuppressive proteins in vitro and in vivo. Moreover, NAV-003 demonstrated good tolerability in mice and efficacy against patient-derived mesothelioma xenografts co-engrafted with human peripheral blood mononuclear cells. Together these data support the potential for NAV-003 clinical development and human proof-of-concept studies in patients with MSLN-expressing cancers.
Collapse
Affiliation(s)
- Luigi Grasso
- Navrogen Inc., 1837 University Circle, Cheyney, PA 19319
| | - Qun Jiang
- Thoracic and GI Malignancies Branch, CCR, NCI, NIH, Bethesda, Maryland
| | - Raffit Hassan
- Thoracic and GI Malignancies Branch, CCR, NCI, NIH, Bethesda, Maryland
| | | | | |
Collapse
|
9
|
Striese F, Neuber C, Gräßel S, Arndt C, Ullrich M, Steinbach J, Pietzsch J, Bergmann R, Pietzsch HJ, Sihver W, Frenz M, Feldmann A, Bachmann MP. Preclinical Characterization of the 177Lu-Labeled Prostate Stem Cell Antigen (PSCA)-Specific Monoclonal Antibody 7F5. Int J Mol Sci 2023; 24:ijms24119420. [PMID: 37298374 DOI: 10.3390/ijms24119420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Prostate specific membrane antigen (PSMA) is an excellent target for imaging and treatment of prostate carcinoma (PCa). Unfortunately, not all PCa cells express PSMA. Therefore, alternative theranostic targets are required. The membrane protein prostate stem cell antigen (PSCA) is highly overexpressed in most primary prostate carcinoma (PCa) cells and in metastatic and hormone refractory tumor cells. Moreover, PSCA expression positively correlates with tumor progression. Therefore, it represents a potential alternative theranostic target suitable for imaging and/or radioimmunotherapy. In order to support this working hypothesis, we conjugated our previously described anti-PSCA monoclonal antibody (mAb) 7F5 with the bifunctional chelator CHX-A″-DTPA and subsequently radiolabeled it with the theranostic radionuclide 177Lu. The resulting radiolabeled mAb ([177Lu]Lu-CHX-A″-DTPA-7F5) was characterized both in vitro and in vivo. It showed a high radiochemical purity (>95%) and stability. The labelling did not affect its binding capability. Biodistribution studies showed a high specific tumor uptake compared to most non-targeted tissues in mice bearing PSCA-positive tumors. Accordingly, SPECT/CT images revealed a high tumor-to-background ratios from 16 h to 7 days after administration of [177Lu]Lu-CHX-A″-DTPA-7F5. Consequently, [177Lu]Lu-CHX-A″-DTPA-7F5 represents a promising candidate for imaging and in the future also for radioimmunotherapy.
Collapse
Affiliation(s)
- Franziska Striese
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technical University Dresden, 01062 Dresden, Germany
| | - Christin Neuber
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
| | - Sandy Gräßel
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technical University Dresden, 01062 Dresden, Germany
| | - Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
| | - Martin Ullrich
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technical University Dresden, 01062 Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technical University Dresden, 01062 Dresden, Germany
| | - Ralf Bergmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
- Institute of Biophysics and Radiation Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Hans-Jürgen Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
| | - Wiebke Sihver
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
| | - Marcus Frenz
- Faculty of Informatik and Wirtschaftsinformatik, Provadis School of International Management and Technology AG, 65926 Frankfurt, Germany
| | - Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
| | - Michael P Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany
- National Center for Tumor Diseases (UCC/NCT), Partner Site Dresden, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
10
|
Anderson VE, Brilha SS, Weber AM, Pachnio A, Wiedermann GE, Dauleh S, Ahmed T, Pope GR, Quinn LL, Docta RY, Quattrini A, Masters S, Cartwright N, Viswanathan P, Melchiori L, Rice LV, Sevko A, Gueguen C, Saini M, Tavano B, Abbott RJ, Silk JD, Laugel B, Sanderson JP, Gerry AB. Enhancing Efficacy of TCR-engineered CD4 + T Cells Via Coexpression of CD8α. J Immunother 2023; 46:132-144. [PMID: 36826388 PMCID: PMC10072215 DOI: 10.1097/cji.0000000000000456] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/12/2023] [Indexed: 02/25/2023]
Abstract
Adoptive cell therapy with T cells expressing affinity-enhanced T-cell receptors (TCRs) is a promising treatment for solid tumors. Efforts are ongoing to further engineer these T cells to increase the depth and durability of clinical responses and broaden efficacy toward additional indications. In the present study, we investigated one such approach: T cells were transduced with a lentiviral vector to coexpress an affinity-enhanced HLA class I-restricted TCR directed against MAGE-A4 alongside a CD8α coreceptor. We hypothesized that this approach would enhance CD4 + T-cell helper and effector functions, possibly leading to a more potent antitumor response. Activation of transduced CD4 + T cells was measured by detecting CD40 ligand expression on the surface and cytokine and chemokine secretion from CD4 + T cells and dendritic cells cultured with melanoma-associated antigen A4 + tumor cells. In addition, T-cell cytotoxic activity against 3-dimensional tumor spheroids was measured. Our data demonstrated that CD4 + T cells coexpressing the TCR and CD8α coreceptor displayed enhanced responses, including CD40 ligand expression, interferon-gamma secretion, and cytotoxic activity, along with improved dendritic cell activation. Therefore, our study supports the addition of the CD8α coreceptor to HLA class I-restricted TCR-engineered T cells to enhance CD4 + T-cell functions, which may potentially improve the depth and durability of antitumor responses in patients.
Collapse
|
11
|
Arndt C, Tunger A, Wehner R, Rothe R, Kourtellari E, Luttosch S, Hannemann K, Koristka S, Loureiro LR, Feldmann A, Tonn T, Link T, Kuhlmann JD, Wimberger P, Bachmann MP, Schmitz M. Palbociclib impairs the proliferative capacity of activated T cells while retaining their cytotoxic efficacy. Front Pharmacol 2023; 14:970457. [PMID: 36817127 PMCID: PMC9935825 DOI: 10.3389/fphar.2023.970457] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
The cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitor palbociclib is an emerging cancer therapeutic that just recently gained Food and Drug Administration approval for treatment of estrogen receptor (ER)-positive, human epidermal growth factor receptor (Her)2-negative breast cancer in combination with the ER degrader fulvestrant. However, CDK4/6 inhibitors are not cancer-specific and may affect also other proliferating cells. Given the importance of T cells in antitumor defense, we studied the influence of palbociclib/fulvestrant on human CD3+ T cells and novel emerging T cell-based cancer immunotherapies. Palbociclib considerably inhibited the proliferation of activated T cells by mediating G0/G1 cell cycle arrest. However, after stopping the drug supply this suppression was fully reversible. In light of combination approaches, we further investigated the effect of palbociclib/fulvestrant on T cell-based immunotherapies by using a CD3-PSCA bispecific antibody or universal chimeric antigen receptor (UniCAR) T cells. Thereby, we observed that palbociclib clearly impaired T cell expansion. This effect resulted in a lower total concentration of interferon-γ and tumor necrosis factor, while palbociclib did not inhibit the average cytokine release per cell. In addition, the cytotoxic potential of the redirected T cells was unaffected by palbociclib and fulvestrant. Overall, these novel findings may have implications for the design of treatment modalities combining CDK4/6 inhibition and T cell-based cancer immunotherapeutic strategies.
Collapse
Affiliation(s)
- Claudia Arndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany,Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany,*Correspondence: Claudia Arndt, ; Marc Schmitz,
| | - Antje Tunger
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany,Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Rebekka Wehner
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany,Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany,German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rebecca Rothe
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany,Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Eleni Kourtellari
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Stephanie Luttosch
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Katharina Hannemann
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Stefanie Koristka
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Liliana R. Loureiro
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Anja Feldmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Torsten Tonn
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany,German Red Cross Blood Donation Service North-East, Institute for Transfusion Medicine, Dresden, Germany,Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Theresa Link
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany,German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany,Department of Gynecology and Obstetrics, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Jan Dominik Kuhlmann
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany,German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany,Department of Gynecology and Obstetrics, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Pauline Wimberger
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany,German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany,Department of Gynecology and Obstetrics, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Michael Philipp Bachmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany,National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany,German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany,Tumor Immunology, University Cancer Center (UCC), University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Marc Schmitz
- National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany,Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany,German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Heidelberg, Germany,*Correspondence: Claudia Arndt, ; Marc Schmitz,
| |
Collapse
|
12
|
Saleh HA, Mitwasi N, Ullrich M, Kubeil M, Toussaint M, Deuther-Conrad W, Neuber C, Arndt C, R. Loureiro L, Kegler A, González Soto KE, Belter B, Rössig C, Pietzsch J, Frenz M, Bachmann M, Feldmann A. Specific and safe targeting of glioblastoma using switchable and logic-gated RevCAR T cells. Front Immunol 2023; 14:1166169. [PMID: 37122703 PMCID: PMC10145173 DOI: 10.3389/fimmu.2023.1166169] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
Glioblastoma (GBM) is still an incurable tumor that is associated with high recurrence rate and poor survival despite the current treatment regimes. With the urgent need for novel therapeutic strategies, immunotherapies, especially chimeric antigen receptor (CAR)-expressing T cells, represent a promising approach for specific and effective targeting of GBM. However, CAR T cells can be associated with serious side effects. To overcome such limitation, we applied our switchable RevCAR system to target both the epidermal growth factor receptor (EGFR) and the disialoganglioside GD2, which are expressed in GBM. The RevCAR system is a modular platform that enables controllability, improves safety, specificity and flexibility. Briefly, it consists of RevCAR T cells having a peptide epitope as extracellular domain, and a bispecific target module (RevTM). The RevTM acts as a switch key that recognizes the RevCAR epitope and the tumor-associated antigen, and thereby activating the RevCAR T cells to kill the tumor cells. However, in the absence of the RevTM, the RevCAR T cells are switched off. In this study, we show that the novel EGFR/GD2-specific RevTMs can selectively activate RevCAR T cells to kill GBM cells. Moreover, we show that gated targeting of GBM is possible with our Dual-RevCAR T cells, which have their internal activation and co-stimulatory domains separated into two receptors. Therefore, a full activation of Dual-RevCAR T cells can only be achieved when both receptors recognize EGFR and GD2 simultaneously via RevTMs, leading to a significant killing of GBM cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Haidy A. Saleh
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Nicola Mitwasi
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Martin Ullrich
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Manja Kubeil
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Magali Toussaint
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Christin Neuber
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- Faculty of Medicine Carl Gustav Carus, Mildred Scheel Early Career Center, Technische Universität Dresden, Dresden, Germany
| | - Liliana R. Loureiro
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Alexandra Kegler
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | | | - Birgit Belter
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Claudia Rössig
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Münster, Münster, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Dresden, Germany
| | - Marcus Frenz
- Faculty Informatik and Wirtschaftsinformatik, Provadis School of International Management and Technology AG, Frankfurt, Germany
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site, Dresden, Germany
- *Correspondence: Michael Bachmann,
| | - Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site, Dresden, Germany
| |
Collapse
|
13
|
Yang F, Yan S, Zhu L, Wang FX, Liu F, Cheng L, Yao H, Wu N, Lu R, Wu H. Evaluation of panel of neutralising murine monoclonal antibodies and a humanised bispecific antibody against influenza A(H1N1)pdm09 virus infection in a mouse model. Antiviral Res 2022; 208:105462. [DOI: 10.1016/j.antiviral.2022.105462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/01/2022] [Accepted: 11/06/2022] [Indexed: 11/15/2022]
|
14
|
Gout DY, Groen LS, van Egmond M. The present and future of immunocytokines for cancer treatment. Cell Mol Life Sci 2022; 79:509. [PMID: 36066630 PMCID: PMC9448690 DOI: 10.1007/s00018-022-04514-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/12/2022] [Accepted: 08/02/2022] [Indexed: 12/04/2022]
Abstract
Monoclonal antibody (mAb) therapy has successfully been introduced as treatment of several lymphomas and leukemias. However, solid tumors reduce the efficacy of mAb therapy because of an immune-suppressive tumor micro-environment (TME), which hampers activation of effector immune cells. Pro-inflammatory cytokine therapy may counteract immune suppression in the TME and increase mAb efficacy, but untargeted pro-inflammatory cytokine therapy is limited by severe off-target toxicity and a short half-life of cytokines. Antibody-cytokine fusion proteins, also referred to as immunocytokines, provide a solution to either issue, as the antibody both acts as local delivery platform and increases half-life. The antibody can furthermore bridge local cytotoxic immune cells, like macrophages and natural killer cells with tumor cells, which can be eliminated after effector cells are activated via the cytokine. Currently, a variety of different antibody formats as well as a handful of cytokine payloads are used to generate immunocytokines. However, many potential formats and payloads are still left unexplored. In this review, we describe current antibody formats and cytokine moieties that are used for the development of immunocytokines, and highlight several immunocytokines in (pre-)clinical studies. Furthermore, potential future routes of development are proposed.
Collapse
Affiliation(s)
- Dennis Y Gout
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1108, Amsterdam, The Netherlands.,Cancer Biology and Immunology Program, Cancer Center Amsterdam, Amsterdam, The Netherlands.,Cancer Immunology Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Lotte S Groen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1108, Amsterdam, The Netherlands.,LUMICKS, Paalbergweg 3, 1105 AG, Amsterdam, The Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1108, Amsterdam, The Netherlands. .,Cancer Biology and Immunology Program, Cancer Center Amsterdam, Amsterdam, The Netherlands. .,Cancer Immunology Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands. .,Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
| |
Collapse
|
15
|
Muñoz-López P, Ribas-Aparicio RM, Becerra-Báez EI, Fraga-Pérez K, Flores-Martínez LF, Mateos-Chávez AA, Luria-Pérez R. Single-Chain Fragment Variable: Recent Progress in Cancer Diagnosis and Therapy. Cancers (Basel) 2022; 14:cancers14174206. [PMID: 36077739 PMCID: PMC9455005 DOI: 10.3390/cancers14174206] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Recombinant antibody fragments have shown remarkable potential as diagnostic and therapeutic tools in the fight against cancer. The single-chain fragment variable (scFv) that contains the complete antigen-binding domains of a whole antibody, has several advantages such as a high specificity and affinity for antigens, a low immunogenicity, and the proven ability to penetrate tumor tissues and diffuse. This review provides an overview of the current studies on the principle, generation, and applications of scFvs, particularly in the diagnosis and therapy of cancer, and underscores their potential use in clinical trials. Abstract Cancer remains a public health problem worldwide. Although conventional therapies have led to some excellent outcomes, some patients fail to respond to treatment, they have few therapeutic alternatives and a poor survival prognosis. Several strategies have been proposed to overcome this issue. The most recent approach is immunotherapy, particularly the use of recombinant antibodies and their derivatives, such as the single-chain fragment variable (scFv) containing the complete antigen-binding domains of a whole antibody that successfully targets tumor cells. This review describes the recent progress made with scFvs as a cancer diagnostic and therapeutic tool, with an emphasis on preclinical approaches and their potential use in clinical trials.
Collapse
Affiliation(s)
- Paola Muñoz-López
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Prolongación de Carpio y Plan de Ayala S/N, Mexico City 11340, Mexico
| | - Rosa María Ribas-Aparicio
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Prolongación de Carpio y Plan de Ayala S/N, Mexico City 11340, Mexico
| | - Elayne Irene Becerra-Báez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Prolongación de Carpio y Plan de Ayala S/N, Mexico City 11340, Mexico
| | - Karla Fraga-Pérez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
| | - Luis Fernando Flores-Martínez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
| | - Armando Alfredo Mateos-Chávez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
| | - Rosendo Luria-Pérez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
- Correspondence: ; Tel.: +52-(55)-5228-9917 (ext. 4401)
| |
Collapse
|
16
|
Lindner D, Arndt C, Loureiro LR, Feldmann A, Kegler A, Koristka S, Berndt N, Mitwasi N, Bergmann R, Frenz M, Bachmann MP. Combining Radiation- with Immunotherapy in Prostate Cancer: Influence of Radiation on T Cells. Int J Mol Sci 2022; 23:ijms23147922. [PMID: 35887271 PMCID: PMC9324763 DOI: 10.3390/ijms23147922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 02/06/2023] Open
Abstract
Radiation of tumor cells can lead to the selection and outgrowth of tumor escape variants. As radioresistant tumor cells are still sensitive to retargeting of T cells, it appears promising to combine radio- with immunotherapy keeping in mind that the radiation of tumors favors the local conditions for immunotherapy. However, radiation of solid tumors will not only hit the tumor cells but also the infiltrated immune cells. Therefore, we wanted to learn how radiation influences the functionality of T cells with respect to retargeting to tumor cells via a conventional bispecific T cell engager (BiTE) and our previously described modular BiTE format UNImAb. T cells were irradiated between 2 and 50 Gy. Low dose radiation of T cells up to about 20 Gy caused an increased release of the cytokines IL-2, TNF and interferon-γ and an improved capability to kill target cells. Although radiation with 50 Gy strongly reduced the function of the T cells, it did not completely abrogate the functionality of the T cells.
Collapse
Affiliation(s)
- Diana Lindner
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany; (D.L.); (C.A.); (L.R.L.); (A.F.); (A.K.); (S.K.); (N.B.); (N.M.); (R.B.)
- Tumor Immunology, University Hospital Carl Gustav Carus, University Cancer Center (UCC), Technical University Dresden, 01307 Dresden, Germany
| | - Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany; (D.L.); (C.A.); (L.R.L.); (A.F.); (A.K.); (S.K.); (N.B.); (N.M.); (R.B.)
| | - Liliana Rodrigues Loureiro
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany; (D.L.); (C.A.); (L.R.L.); (A.F.); (A.K.); (S.K.); (N.B.); (N.M.); (R.B.)
| | - Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany; (D.L.); (C.A.); (L.R.L.); (A.F.); (A.K.); (S.K.); (N.B.); (N.M.); (R.B.)
| | - Alexandra Kegler
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany; (D.L.); (C.A.); (L.R.L.); (A.F.); (A.K.); (S.K.); (N.B.); (N.M.); (R.B.)
| | - Stefanie Koristka
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany; (D.L.); (C.A.); (L.R.L.); (A.F.); (A.K.); (S.K.); (N.B.); (N.M.); (R.B.)
| | - Nicole Berndt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany; (D.L.); (C.A.); (L.R.L.); (A.F.); (A.K.); (S.K.); (N.B.); (N.M.); (R.B.)
| | - Nicola Mitwasi
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany; (D.L.); (C.A.); (L.R.L.); (A.F.); (A.K.); (S.K.); (N.B.); (N.M.); (R.B.)
| | - Ralf Bergmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany; (D.L.); (C.A.); (L.R.L.); (A.F.); (A.K.); (S.K.); (N.B.); (N.M.); (R.B.)
- Institute of Biophysics and Radiation Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Marcus Frenz
- Faculty Informatik and Wirtschaftsinformatik, Provadis School of International Management and Technology AG, 65926 Frankfurt, Germany;
| | - Michael P. Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany; (D.L.); (C.A.); (L.R.L.); (A.F.); (A.K.); (S.K.); (N.B.); (N.M.); (R.B.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-351-260-3170
| |
Collapse
|
17
|
Abrams RE, Pierre K, El-Murr N, Seung E, Wu L, Luna E, Mehta R, Li J, Larabi K, Ahmed M, Pelekanou V, Yang ZY, van de Velde H, Stamatelos SK. Quantitative systems pharmacology modeling sheds light into the dose response relationship of a trispecific T cell engager in multiple myeloma. Sci Rep 2022; 12:10976. [PMID: 35768621 PMCID: PMC9243109 DOI: 10.1038/s41598-022-14726-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 06/10/2022] [Indexed: 02/08/2023] Open
Abstract
In relapsed and refractory multiple myeloma (RRMM), there are few treatment options once patients progress from the established standard of care. Several bispecific T-cell engagers (TCE) are in clinical development for multiple myeloma (MM), designed to promote T-cell activation and tumor killing by binding a T-cell receptor and a myeloma target. In this study we employ both computational and experimental tools to investigate how a novel trispecific TCE improves activation, proliferation, and cytolytic activity of T-cells against MM cells. In addition to binding CD3 on T-cells and CD38 on tumor cells, the trispecific binds CD28, which serves as both co-stimulation for T-cell activation and an additional tumor target. We have established a robust rule-based quantitative systems pharmacology (QSP) model trained against T-cell activation, cytotoxicity, and cytokine data, and used it to gain insight into the complex dose response of this drug. We predict that CD3-CD28-CD38 killing capacity increases rapidly in low dose levels, and with higher doses, killing plateaus rather than following the bell-shaped curve typical of bispecific TCEs. We further predict that dose–response curves are driven by the ability of tumor cells to form synapses with activated T-cells. When competition between cells limits tumor engagement with active T-cells, response to therapy may be diminished. We finally suggest a metric related to drug efficacy in our analysis—“effective” receptor occupancy, or the proportion of receptors engaged in synapses. Overall, this study predicts that the CD28 arm on the trispecific antibody improves efficacy, and identifies metrics to inform potency of novel TCEs.
Collapse
Affiliation(s)
- R E Abrams
- Sanofi, 55 Corporate Dr, Bridgewater, NJ, 08807, USA.,Daichi Sankyo, 211 Mt. Airy Rd., Basking Ridge, NJ, 07920, USA
| | - K Pierre
- Sanofi, 55 Corporate Dr, Bridgewater, NJ, 08807, USA.
| | - N El-Murr
- Sanofi, 13 quai Jules Guesde 94403 Cedex, VITRY-SUR-SEINE, Vitry/Alfortville, France
| | - E Seung
- Sanofi, 270 Albany St., Cambridge, MA, 02139, USA.,Modex Therapeutics, 22 Strathmore Road, Natick, MA, 01760, USA
| | - L Wu
- Sanofi, 270 Albany St., Cambridge, MA, 02139, USA.,Modex Therapeutics, 22 Strathmore Road, Natick, MA, 01760, USA
| | | | | | - J Li
- Sanofi, 55 Corporate Dr, Bridgewater, NJ, 08807, USA
| | - K Larabi
- Sanofi, 13 quai Jules Guesde 94403 Cedex, VITRY-SUR-SEINE, Vitry/Alfortville, France
| | - M Ahmed
- Sanofi, 50 Binney St., Cambridge, MA, 02142, USA
| | - V Pelekanou
- Sanofi, 50 Binney St., Cambridge, MA, 02142, USA.,Bayer Pharmaceuticals, Cambridge, MA, 02142, USA
| | - Z-Y Yang
- Sanofi, 270 Albany St., Cambridge, MA, 02139, USA.,Modex Therapeutics, 22 Strathmore Road, Natick, MA, 01760, USA
| | | | - S K Stamatelos
- Sanofi, 55 Corporate Dr, Bridgewater, NJ, 08807, USA. .,Bayer Pharmaceuticals, PH100 Bayer Boulevard, Whippany, NJ, 07981, USA.
| |
Collapse
|
18
|
Mitwasi N, Arndt C, Loureiro LR, Kegler A, Fasslrinner F, Berndt N, Bergmann R, Hořejší V, Rössig C, Bachmann M, Feldmann A. Targeting CD10 on B-Cell Leukemia Using the Universal CAR T-Cell Platform (UniCAR). Int J Mol Sci 2022; 23:4920. [PMID: 35563312 PMCID: PMC9105388 DOI: 10.3390/ijms23094920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 12/10/2022] Open
Abstract
Chimeric antigen receptor (CAR)-expressing T-cells are without a doubt a breakthrough therapy for hematological malignancies. Despite their success, clinical experience has revealed several challenges, which include relapse after targeting single antigens such as CD19 in the case of B-cell acute lymphoblastic leukemia (B-ALL), and the occurrence of side effects that could be severe in some cases. Therefore, it became clear that improved safety approaches, and targeting multiple antigens, should be considered to further improve CAR T-cell therapy for B-ALL. In this paper, we address both issues by investigating the use of CD10 as a therapeutic target for B-ALL with our switchable UniCAR system. The UniCAR platform is a modular platform that depends on the presence of two elements to function. These include UniCAR T-cells and the target modules (TMs), which cross-link the T-cells to their respective targets on tumor cells. The TMs function as keys that control the switchability of UniCAR T-cells. Here, we demonstrate that UniCAR T-cells, armed with anti-CD10 TM, can efficiently kill B-ALL cell lines, as well as patient-derived B-ALL blasts, thereby highlighting the exciting possibility for using CD10 as an emerging therapeutic target for B-cell malignancies.
Collapse
MESH Headings
- Antigens, CD19/metabolism
- Humans
- Immunotherapy, Adoptive
- Leukemia, B-Cell/metabolism
- Leukemia, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Neprilysin/therapeutic use
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes
Collapse
Affiliation(s)
- Nicola Mitwasi
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D-01328 Dresden, Germany; (N.M.); (C.A.); (L.R.L.); (A.K.); (N.B.); (R.B.); (A.F.)
| | - Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D-01328 Dresden, Germany; (N.M.); (C.A.); (L.R.L.); (A.K.); (N.B.); (R.B.); (A.F.)
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, TU Dresden, D-01307 Dresden, Germany;
| | - Liliana R. Loureiro
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D-01328 Dresden, Germany; (N.M.); (C.A.); (L.R.L.); (A.K.); (N.B.); (R.B.); (A.F.)
| | - Alexandra Kegler
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D-01328 Dresden, Germany; (N.M.); (C.A.); (L.R.L.); (A.K.); (N.B.); (R.B.); (A.F.)
| | - Frederick Fasslrinner
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, TU Dresden, D-01307 Dresden, Germany;
- Medical Clinic and Polyclinic I, University Hospital Carl Gustav Carus, TU Dresden, D-01307 Dresden, Germany
| | - Nicole Berndt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D-01328 Dresden, Germany; (N.M.); (C.A.); (L.R.L.); (A.K.); (N.B.); (R.B.); (A.F.)
| | - Ralf Bergmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D-01328 Dresden, Germany; (N.M.); (C.A.); (L.R.L.); (A.K.); (N.B.); (R.B.); (A.F.)
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary
| | - Vaclav Hořejší
- Institute of Molecular Genetics of the Academy of Sciences of the Czech Republic, 142 20 Prague, Czech Republic;
| | - Claudia Rössig
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Münster, 48149 Münster, Germany;
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D-01328 Dresden, Germany; (N.M.); (C.A.); (L.R.L.); (A.K.); (N.B.); (R.B.); (A.F.)
- National Center for Tumor Diseases (NCT), D-01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Tumor Immunology, University Cancer Center (UCC), University Hospital Carl Gustav Carus, TU Dresden, D-01307 Dresden, Germany
| | - Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, D-01328 Dresden, Germany; (N.M.); (C.A.); (L.R.L.); (A.K.); (N.B.); (R.B.); (A.F.)
| |
Collapse
|
19
|
Development and Functional Characterization of a Versatile Radio-/Immunotheranostic Tool for Prostate Cancer Management. Cancers (Basel) 2022; 14:cancers14081996. [PMID: 35454902 PMCID: PMC9027777 DOI: 10.3390/cancers14081996] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In previous studies, we described a modular Chimeric Antigen Receptor (CAR) T cell platform which we termed UniCAR. In contrast to conventional CARs, the interaction of UniCAR T cells does not occur directly between the CAR T cell and the tumor cell but is mediated via bispecific adaptor molecules so-called target modules (TMs). Here we present the development and functional characterization of a novel IgG4-based TM, directed to the tumor-associated antigen (TAA) prostate stem cell antigen (PSCA), which is overexpressed in prostate cancer (PCa). We show that this anti-PSCA IgG4-TM cannot only be used for (i) redirection of UniCAR T cells to PCa cells but also for (ii) positron emission tomography (PET) imaging, and (iii) alpha particle-based endoradiotherapy. For radiolabeling, the anti-PSCA IgG4-TM was conjugated with the chelator DOTAGA. PET imaging was performed using the 64Cu-labeled anti-PSCA IgG4-TM. According to PET imaging, the anti-PSCA IgG4-TM accumulates with high contrast in the PSCA-positive tumors of experimental mice without visible uptake in other organs. For endoradiotherapy the anti-PSCA IgG4-TM-DOTAGA conjugate was labeled with 225Ac3+. Targeted alpha therapy resulted in tumor control over 60 days after a single injection of the 225Ac-labeled TM. The favorable pharmacological profile of the anti-PSCA IgG4-TM, and its usage for (i) imaging, (ii) targeted alpha therapy, and (iii) UniCAR T cell immunotherapy underlines the promising radio-/immunotheranostic capabilities for the diagnostic imaging and treatment of PCa. Abstract Due to its overexpression on the surface of prostate cancer (PCa) cells, the prostate stem cell antigen (PSCA) is a potential target for PCa diagnosis and therapy. Here we describe the development and functional characterization of a novel IgG4-based anti-PSCA antibody (Ab) derivative (anti-PSCA IgG4-TM) that is conjugated with the chelator DOTAGA. The anti-PSCA IgG4-TM represents a multimodal immunotheranostic compound that can be used (i) as a target module (TM) for UniCAR T cell-based immunotherapy, (ii) for diagnostic positron emission tomography (PET) imaging, and (iii) targeted alpha therapy. Cross-linkage of UniCAR T cells and PSCA-positive tumor cells via the anti-PSCA IgG4-TM results in efficient tumor cell lysis both in vitro and in vivo. After radiolabeling with 64Cu2+, the anti-PSCA IgG4-TM was successfully applied for high contrast PET imaging. In a PCa mouse model, it showed specific accumulation in PSCA-expressing tumors, while no uptake in other organs was observed. Additionally, the DOTAGA-conjugated anti-PSCA IgG4-TM was radiolabeled with 225Ac3+ and applied for targeted alpha therapy. A single injection of the 225Ac-labeled anti-PSCA IgG4-TM was able to significantly control tumor growth in experimental mice. Overall, the novel anti-PSCA IgG4-TM represents an attractive first member of a novel group of radio-/immunotheranostics that allows diagnostic imaging, endoradiotherapy, and CAR T cell immunotherapy.
Collapse
|
20
|
Dual-Labelling Strategies for Nuclear and Fluorescence Molecular Imaging: Current Status and Future Perspectives. Pharmaceuticals (Basel) 2022; 15:ph15040432. [PMID: 35455430 PMCID: PMC9028399 DOI: 10.3390/ph15040432] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 12/13/2022] Open
Abstract
Molecular imaging offers the possibility to investigate biological and biochemical processes non-invasively and to obtain information on both anatomy and dysfunctions. Based on the data obtained, a fundamental understanding of various disease processes can be derived and treatment strategies can be planned. In this context, methods that combine several modalities in one probe are increasingly being used. Due to the comparably high sensitivity and provided complementary information, the combination of nuclear and optical probes has taken on a special significance. In this review article, dual-labelled systems for bimodal nuclear and optical imaging based on both modular ligands and nanomaterials are discussed. Particular attention is paid to radiometal-labelled molecules for single-photon emission computed tomography (SPECT) and positron emission tomography (PET) and metal complexes combined with fluorescent dyes for optical imaging. The clinical potential of such probes, especially for fluorescence-guided surgery, is assessed.
Collapse
|
21
|
Köseer AS, Loureiro LR, Jureczek J, Mitwasi N, González Soto KE, Aepler J, Bartsch T, Feldmann A, Kunz-Schughart LA, Linge A, Krause M, Bachmann M, Arndt C, Dubrovska A. Validation of CD98hc as a Therapeutic Target for a Combination of Radiation and Immunotherapies in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:1677. [PMID: 35406454 PMCID: PMC8997111 DOI: 10.3390/cancers14071677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/11/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023] Open
Abstract
Most patients with head and neck squamous cell carcinomas (HNSCC) are diagnosed at a locally advanced stage and show heterogeneous treatment responses. Low SLC3A2 (solute carrier family 3 member 2) mRNA and protein (CD98hc) expression levels are associated with higher locoregional control in HNSCC patients treated with primary radiochemotherapy or postoperative radiochemotherapy, suggesting that CD98hc could be a target for HNSCC radiosensitization. One of the targeted strategies for tumor radiosensitization is precision immunotherapy, e.g., the use of chimeric antigen receptor (CAR) T cells. This study aimed to define the potential clinical value of new treatment approaches combining conventional radiotherapy with CD98hc-targeted immunotherapy. To address this question, we analyzed the antitumor activity of the combination of fractionated irradiation and switchable universal CAR (UniCAR) system against radioresistant HNSCC cells in 3D culture. CD98hc-redirected UniCAR T cells showed the ability to destroy radioresistant HNSCC spheroids. Also, the infiltration rate of the UniCAR T cells was enhanced in the presence of the CD98hc target module. Furthermore, sequential treatment with fractionated irradiation followed by CD98hc-redirected UniCAR T treatment showed a synergistic effect. Taken together, our obtained data underline the improved antitumor effect of the combination of radiotherapy with CD98hc-targeted immunotherapy. Such a combination presents an attractive approach for the treatment of high-risk HNSCC patients.
Collapse
Affiliation(s)
- Ayşe Sedef Köseer
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany; (A.S.K.); (L.R.L.); (A.F.); (L.A.K.-S.); (A.L.); (M.K.); (M.B.)
| | - Liliana R. Loureiro
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany; (A.S.K.); (L.R.L.); (A.F.); (L.A.K.-S.); (A.L.); (M.K.); (M.B.)
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (J.J.); (N.M.); (K.E.G.S.); (J.A.); (T.B.)
| | - Justyna Jureczek
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (J.J.); (N.M.); (K.E.G.S.); (J.A.); (T.B.)
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Tumor Immunology, University Cancer Center (UCC), University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Nicola Mitwasi
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (J.J.); (N.M.); (K.E.G.S.); (J.A.); (T.B.)
| | - Karla Elizabeth González Soto
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (J.J.); (N.M.); (K.E.G.S.); (J.A.); (T.B.)
| | - Julia Aepler
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (J.J.); (N.M.); (K.E.G.S.); (J.A.); (T.B.)
| | - Tabea Bartsch
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (J.J.); (N.M.); (K.E.G.S.); (J.A.); (T.B.)
| | - Anja Feldmann
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany; (A.S.K.); (L.R.L.); (A.F.); (L.A.K.-S.); (A.L.); (M.K.); (M.B.)
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (J.J.); (N.M.); (K.E.G.S.); (J.A.); (T.B.)
| | - Leoni A. Kunz-Schughart
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany; (A.S.K.); (L.R.L.); (A.F.); (L.A.K.-S.); (A.L.); (M.K.); (M.B.)
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany
| | - Annett Linge
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany; (A.S.K.); (L.R.L.); (A.F.); (L.A.K.-S.); (A.L.); (M.K.); (M.B.)
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Mechthild Krause
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany; (A.S.K.); (L.R.L.); (A.F.); (L.A.K.-S.); (A.L.); (M.K.); (M.B.)
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01307 Dresden, Germany
| | - Michael Bachmann
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany; (A.S.K.); (L.R.L.); (A.F.); (L.A.K.-S.); (A.L.); (M.K.); (M.B.)
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (J.J.); (N.M.); (K.E.G.S.); (J.A.); (T.B.)
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Tumor Immunology, University Cancer Center (UCC), University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Claudia Arndt
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany; (A.S.K.); (L.R.L.); (A.F.); (L.A.K.-S.); (A.L.); (M.K.); (M.B.)
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (J.J.); (N.M.); (K.E.G.S.); (J.A.); (T.B.)
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Anna Dubrovska
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01307 Dresden, Germany; (A.S.K.); (L.R.L.); (A.F.); (L.A.K.-S.); (A.L.); (M.K.); (M.B.)
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- OncoRay–National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01307 Dresden, Germany
| |
Collapse
|
22
|
Nguyen-Le TA, Bartsch T, Wodtke R, Brandt F, Arndt C, Feldmann A, Sandoval Bojorquez DI, Roig AP, Ibarlucea B, Lee S, Baek CK, Cuniberti G, Bergmann R, Puentes-Cala E, Soto JA, Kurien BT, Bachmann M, Baraban L. Nanosensors in clinical development of CAR-T cell immunotherapy. Biosens Bioelectron 2022; 206:114124. [PMID: 35272215 DOI: 10.1016/j.bios.2022.114124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/18/2022] [Accepted: 02/20/2022] [Indexed: 11/30/2022]
Abstract
Immunotherapy using CAR-T cells is a new technological paradigm for cancer treatment. To avoid severe side effects and tumor escape variants observed for conventional CAR-T cells approach, adaptor CAR technologies are under development, where intermediate target modules redirect immune cells against cancer. In this work, silicon nanowire field-effect transistors are used to develop target modules for an optimized CAR-T cell operation. Focusing on a library of seven variants of E5B9 peptide that is used as CAR targeting epitope, we performed multiplexed binding tests using nanosensor chips. These peptides had been immobilized onto the sensor to compare the transistor signals upon titration with anti-La 5B9 antibodies. The correlation of binding affinities and sensor sensitivities enabled a selection of candidates for the interaction between CAR and target modules. An extremely low detection limit was observed for the sensor, down to femtomolar concentration, outperforming the current assay of the same purpose. Finally, the CAR T-cells redirection capability of selected peptides in target modules was proven successful in an in-vitro cytotoxicity assay. Our results open the perspective for the nanosensors to go beyond the early diagnostics in clinical cancer research towards developing and monitoring immunotherapeutic treatment, where the quantitative analysis with the standard techniques is limited.
Collapse
Affiliation(s)
- Trang Anh Nguyen-Le
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V. (HZDR), 01328, Dresden, Germany
| | - Tabea Bartsch
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V. (HZDR), 01328, Dresden, Germany
| | - Robert Wodtke
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V. (HZDR), 01328, Dresden, Germany
| | - Florian Brandt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V. (HZDR), 01328, Dresden, Germany; Fakultät Chemie und Lebensmittelchemie, Technische Universität Dresden, Mommsenstraße 4, 01062, Dresden, Germany
| | - Claudia Arndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V. (HZDR), 01328, Dresden, Germany; Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, TU Dresden, 01307, Dresden, Germany
| | - Anja Feldmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V. (HZDR), 01328, Dresden, Germany
| | - Diana Isabel Sandoval Bojorquez
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V. (HZDR), 01328, Dresden, Germany
| | - Arnau Perez Roig
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V. (HZDR), 01328, Dresden, Germany
| | - Bergoi Ibarlucea
- Institute for Materials Science, Max Bergmann Center for Biomaterials, Center for Advancing Electronics Dresden (cfaed), Technische Universität Dresden, 01069, Dresden, Germany
| | - Seungho Lee
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Chan-Ki Baek
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Gianaurelio Cuniberti
- Institute for Materials Science, Max Bergmann Center for Biomaterials, Center for Advancing Electronics Dresden (cfaed), Technische Universität Dresden, 01069, Dresden, Germany
| | - Ralf Bergmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V. (HZDR), 01328, Dresden, Germany; Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Edinson Puentes-Cala
- Corporación para la Investigación de la Corrosión (CIC), Piedecuesta, 681011, Colombia
| | | | - Biji T Kurien
- The Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation and University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V. (HZDR), 01328, Dresden, Germany; Tumor Immunology, University Cancer Center (UCC), University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, 01307, Dresden, Germany; National Center for Tumor Diseases (NCT), Dresden, Germany. Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Dresden, Germany.
| | - Larysa Baraban
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V. (HZDR), 01328, Dresden, Germany.
| |
Collapse
|
23
|
Thoreau F, Chudasama V. Enabling the next steps in cancer immunotherapy: from antibody-based bispecifics to multispecifics, with an evolving role for bioconjugation chemistry. RSC Chem Biol 2022; 3:140-169. [PMID: 35360884 PMCID: PMC8826860 DOI: 10.1039/d1cb00082a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 10/22/2021] [Indexed: 12/02/2022] Open
Abstract
In the past two decades, immunotherapy has established itself as one of the leading strategies for cancer treatment, as illustrated by the exponentially growing number of related clinical trials. This trend was, in part, prompted by the clinical success of both immune checkpoint modulation and immune cell engagement, to restore and/or stimulate the patient's immune system's ability to fight the disease. These strategies were sustained by progress in bispecific antibody production. However, despite the decisive progress made in the treatment of cancer, toxicity and resistance are still observed in some cases. In this review, we initially provide an overview of the monoclonal and bispecific antibodies developed with the objective of restoring immune system functions to treat cancer (cancer immunotherapy), through immune checkpoint modulation, immune cell engagement or a combination of both. Their production, design strategy and impact on the clinical trial landscape are also addressed. In the second part, the concept of multispecific antibody formats, notably MuTICEMs (Multispecific Targeted Immune Cell Engagers & Modulators), as a possible answer to current immunotherapy limitations is investigated. We believe it could be the next step to take for cancer immunotherapy research and expose why bioconjugation chemistry might play a key role in these future developments.
Collapse
Affiliation(s)
- Fabien Thoreau
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street London WC1H 0AJ UK
| |
Collapse
|
24
|
Bartsch T, Arndt C, Loureiro LR, Kegler A, Puentes-Cala E, Soto JA, Kurien BT, Feldmann A, Berndt N, Bachmann MP. A Small Step, a Giant Leap: Somatic Hypermutation of a Single Amino Acid Leads to Anti-La Autoreactivity. Int J Mol Sci 2021; 22:ijms222112046. [PMID: 34769474 PMCID: PMC8584381 DOI: 10.3390/ijms222112046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
The anti-La mab 312B, which was established by hybridoma technology from human-La transgenic mice after adoptive transfer of anti-human La T cells, immunoprecipitates both native eukaryotic human and murine La protein. Therefore, it represents a true anti-La autoantibody. During maturation, the anti-La mab 312B acquired somatic hypermutations (SHMs) which resulted in the replacement of four aa in the complementarity determining regions (CDR) and seven aa in the framework regions. The recombinant derivative of the anti-La mab 312B in which all the SHMs were corrected to the germline sequence failed to recognize the La antigen. We therefore wanted to learn which SHM(s) is (are) responsible for anti-La autoreactivity. Humanization of the 312B ab by grafting its CDR regions to a human Ig backbone confirms that the CDR sequences are mainly responsible for anti-La autoreactivity. Finally, we identified that a single amino acid replacement (D > Y) in the germline sequence of the CDR3 region of the heavy chain of the anti-La mab 312B is sufficient for anti-La autoreactivity.
Collapse
Affiliation(s)
- Tabea Bartsch
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (T.B.); (C.A.); (L.R.L.); (A.K.); (E.P.-C.); (J.A.S.); (A.F.); (N.B.)
| | - Claudia Arndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (T.B.); (C.A.); (L.R.L.); (A.K.); (E.P.-C.); (J.A.S.); (A.F.); (N.B.)
| | - Liliana R. Loureiro
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (T.B.); (C.A.); (L.R.L.); (A.K.); (E.P.-C.); (J.A.S.); (A.F.); (N.B.)
| | - Alexandra Kegler
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (T.B.); (C.A.); (L.R.L.); (A.K.); (E.P.-C.); (J.A.S.); (A.F.); (N.B.)
| | - Edinson Puentes-Cala
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (T.B.); (C.A.); (L.R.L.); (A.K.); (E.P.-C.); (J.A.S.); (A.F.); (N.B.)
- Corporación para la Investigación de la Corrosión (CIC), Piedecuesta 681011, Colombia
| | - Javier Andrés Soto
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (T.B.); (C.A.); (L.R.L.); (A.K.); (E.P.-C.); (J.A.S.); (A.F.); (N.B.)
- BIOGEN Research Group, University of Santander, Faculty of Health Sciences, Cúcuta 540001, Colombia
| | - Biji T. Kurien
- The Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Anja Feldmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (T.B.); (C.A.); (L.R.L.); (A.K.); (E.P.-C.); (J.A.S.); (A.F.); (N.B.)
| | - Nicole Berndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (T.B.); (C.A.); (L.R.L.); (A.K.); (E.P.-C.); (J.A.S.); (A.F.); (N.B.)
| | - Michael P. Bachmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (T.B.); (C.A.); (L.R.L.); (A.K.); (E.P.-C.); (J.A.S.); (A.F.); (N.B.)
- BIOGEN Research Group, University of Santander, Faculty of Health Sciences, Cúcuta 540001, Colombia
- The Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Tumor Immunology, University Cancer Center (UCC), University Hospital Carl Gustav Carus Dresden, TU Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), 03128 Dresden, Germany
- Correspondence: ; Tel.: +49-351-260-3223
| |
Collapse
|
25
|
Berndt N, Bippes CC, Michalk I, Bartsch T, Arndt C, Puentes-Cala E, Soto JA, Loureiro LR, Kegler A, Bachmann D, Gross JK, Gross T, Kurien BT, Scofield RH, Farris AD, James JA, Bergmann R, Schmitz M, Feldmann A, Bachmann MP. And Yet It Moves: Oxidation of the Nuclear Autoantigen La/SS-B Is the Driving Force for Nucleo-Cytoplasmic Shuttling. Int J Mol Sci 2021; 22:9699. [PMID: 34575862 PMCID: PMC8470643 DOI: 10.3390/ijms22189699] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 01/10/2023] Open
Abstract
Decades ago, we and many other groups showed a nucleo-cytoplasmic translocation of La protein in cultured cells. This shuttling of La protein was seen after UV irradiation, virus infections, hydrogen peroxide exposure and the Fenton reaction based on iron or copper ions. All of these conditions are somehow related to oxidative stress. Unfortunately, these harsh conditions could also cause an artificial release of La protein. Even until today, the shuttling and the cytoplasmic function of La/SS-B is controversially discussed. Moreover, the driving mechanism for the shuttling of La protein remains unclear. Recently, we showed that La protein undergoes redox-dependent conformational changes. Moreover, we developed anti-La monoclonal antibodies (anti-La mAbs), which are specific for either the reduced form of La protein or the oxidized form. Using these tools, here we show that redox-dependent conformational changes are the driving force for the shuttling of La protein. Moreover, we show that translocation of La protein to the cytoplasm can be triggered in a ligand/receptor-dependent manner under physiological conditions. We show that ligands of toll-like receptors lead to a redox-dependent shuttling of La protein. The shuttling of La protein depends on the redox status of the respective cell type. Endothelial cells are usually resistant to the shuttling of La protein, while dendritic cells are highly sensitive. However, the deprivation of intracellular reducing agents in endothelial cells makes endothelial cells sensitive to a redox-dependent shuttling of La protein.
Collapse
Affiliation(s)
- Nicole Berndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
| | - Claudia C. Bippes
- Institute of Immunology, Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, 01307 Dresden, Germany; (C.C.B.); (I.M.); (M.S.)
| | - Irene Michalk
- Institute of Immunology, Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, 01307 Dresden, Germany; (C.C.B.); (I.M.); (M.S.)
| | - Tabea Bartsch
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
| | - Claudia Arndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
| | - Edinson Puentes-Cala
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
- Corporación para la Investigación de la Corrosión (CIC), Piedecuesta 681011, Colombia
| | - Javier Andrés Soto
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
- Instituto de Investigación Masira, Facultad de Ciencias Médicas y de la Salud, Universidad de Santander, Cúcuta 540001, Colombia
| | - Liliana R. Loureiro
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
| | - Alexandra Kegler
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
| | - Dominik Bachmann
- Tumor Immunology, University Cancer Center (UCC), University Hospital Carl Gustav Carus Technische Universität Dresden, 01307 Dresden, Germany;
| | - Joanne K. Gross
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - Tim Gross
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - Biji T. Kurien
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - R. Hal Scofield
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - A. Darise Farris
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - Judith A. James
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - Ralf Bergmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
- Department of Biophysics and Radiobiology, Semmelweis University, 1094 Budapest, Hungary
| | - Marc Schmitz
- Institute of Immunology, Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, 01307 Dresden, Germany; (C.C.B.); (I.M.); (M.S.)
- National Center for Tumor Diseases (NCT), 03128 Dresden, Germany
| | - Anja Feldmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
| | - Michael P. Bachmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
- Institute of Immunology, Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, 01307 Dresden, Germany; (C.C.B.); (I.M.); (M.S.)
- National Center for Tumor Diseases (NCT), 03128 Dresden, Germany
| |
Collapse
|
26
|
Belmontes B, Sawant DV, Zhong W, Tan H, Kaul A, Aeffner F, O'Brien SA, Chun M, Noubade R, Eng J, Ma H, Muenz M, Li P, Alba BM, Thomas M, Cook K, Wang X, DeVoss J, Egen JG, Nolan-Stevaux O. Immunotherapy combinations overcome resistance to bispecific T cell engager treatment in T cell-cold solid tumors. Sci Transl Med 2021; 13:13/608/eabd1524. [PMID: 34433637 DOI: 10.1126/scitranslmed.abd1524] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/28/2021] [Indexed: 12/15/2022]
Abstract
Therapeutic approaches are needed to promote T cell-mediated destruction of poorly immunogenic, "cold" tumors typically associated with minimal response to immune checkpoint blockade (ICB) therapy. Bispecific T cell engager (BiTE) molecules induce redirected lysis of cancer cells by polyclonal T cells and have demonstrated promising clinical activity against solid tumors in some patients. However, little is understood about the key factors that govern clinical responses to these therapies. Using an immunocompetent mouse model expressing a humanized CD3ε chain (huCD3e mice) and BiTE molecules directed against mouse CD19, mouse CLDN18.2, or human EPCAM antigens, we investigated the pharmacokinetic and pharmacodynamic parameters and immune correlates associated with BiTE efficacy across multiple syngeneic solid-tumor models. These studies demonstrated that pretreatment tumor-associated T cell density is a critical determinant of response to BiTE therapy, identified CD8+ T cells as important targets and mediators of BiTE activity, and revealed an antagonistic role for CD4+ T cells in BiTE efficacy. We also identified therapeutic combinations, including ICB and 4-1BB agonism, that synergized with BiTE treatment in poorly T cell-infiltrated, immunotherapy-refractory tumors. In these models, BiTE efficacy was dependent on local expansion of tumor-associated CD8+ T cells, rather than their recruitment from circulation. Our findings highlight the relative contributions of baseline T cell infiltration, local T cell proliferation, and peripheral T cell trafficking for BiTE molecule-mediated efficacy, identify combination strategies capable of overcoming resistance to BiTE therapy, and have clinical relevance for the development of BiTE and other T cell engager therapies.
Collapse
Affiliation(s)
- Brian Belmontes
- Amgen Research, Thousand Oaks, CA 91320, USA.,Inflammation and Oncology Therapeutic Area, Amgen, Thousand Oaks, CA 91320, USA
| | - Deepali V Sawant
- Amgen Research, Thousand Oaks, CA 91320, USA.,Inflammation and Oncology Therapeutic Area, Amgen, South San Francisco, CA 94080, USA
| | - Wendy Zhong
- Amgen Research, Thousand Oaks, CA 91320, USA.,Inflammation and Oncology Therapeutic Area, Amgen, South San Francisco, CA 94080, USA
| | - Hong Tan
- Amgen Research, Thousand Oaks, CA 91320, USA.,Inflammation and Oncology Therapeutic Area, Amgen, Thousand Oaks, CA 91320, USA
| | - Anupurna Kaul
- Amgen Research, Thousand Oaks, CA 91320, USA.,Inflammation and Oncology Therapeutic Area, Amgen, South San Francisco, CA 94080, USA
| | - Famke Aeffner
- Amgen Research, Thousand Oaks, CA 91320, USA.,Translational Safety and Bioanalytical Sciences, Amgen, South San Francisco, CA 94080, USA
| | - Sarah A O'Brien
- Amgen Research, Thousand Oaks, CA 91320, USA.,Inflammation and Oncology Therapeutic Area, Amgen, South San Francisco, CA 94080, USA
| | - Matthew Chun
- Amgen Research, Thousand Oaks, CA 91320, USA.,Inflammation and Oncology Therapeutic Area, Amgen, South San Francisco, CA 94080, USA
| | - Rajkumar Noubade
- Amgen Research, Thousand Oaks, CA 91320, USA.,Inflammation and Oncology Therapeutic Area, Amgen, South San Francisco, CA 94080, USA
| | - Jason Eng
- Amgen Research, Thousand Oaks, CA 91320, USA.,Inflammation and Oncology Therapeutic Area, Amgen, South San Francisco, CA 94080, USA
| | - Hayley Ma
- Amgen Research, Thousand Oaks, CA 91320, USA.,Inflammation and Oncology Therapeutic Area, Amgen, Thousand Oaks, CA 91320, USA
| | - Markus Muenz
- Amgen Research, Thousand Oaks, CA 91320, USA.,Amgen Research GmbH, Munich 81477, Germany
| | - Peng Li
- Amgen Research, Thousand Oaks, CA 91320, USA.,Therapeutic Discovery, Amgen, South San Francisco, CA 94080, USA
| | - Benjamin M Alba
- Amgen Research, Thousand Oaks, CA 91320, USA.,Therapeutic Discovery, Amgen, South San Francisco, CA 94080, USA
| | - Melissa Thomas
- Amgen Research, Thousand Oaks, CA 91320, USA.,Therapeutic Discovery, Amgen, South San Francisco, CA 94080, USA
| | - Kevin Cook
- Amgen Research, Thousand Oaks, CA 91320, USA.,Pharmacokinetics and Drug Metabolism, Amgen, South San Francisco, CA 94080, USA
| | - Xiaoting Wang
- Amgen Research, Thousand Oaks, CA 91320, USA.,Translational Safety and Bioanalytical Sciences, Amgen, South San Francisco, CA 94080, USA
| | - Jason DeVoss
- Amgen Research, Thousand Oaks, CA 91320, USA.,Inflammation and Oncology Therapeutic Area, Amgen, South San Francisco, CA 94080, USA
| | - Jackson G Egen
- Amgen Research, Thousand Oaks, CA 91320, USA. .,Inflammation and Oncology Therapeutic Area, Amgen, South San Francisco, CA 94080, USA
| | - Olivier Nolan-Stevaux
- Amgen Research, Thousand Oaks, CA 91320, USA. .,Inflammation and Oncology Therapeutic Area, Amgen, South San Francisco, CA 94080, USA
| |
Collapse
|
27
|
Meyer JE, Loff S, Dietrich J, Spehr J, Jurado Jiménez G, von Bonin M, Ehninger G, Cartellieri M, Ehninger A. Evaluation of switch-mediated costimulation in trans on universal CAR-T cells (UniCAR) targeting CD123-positive AML. Oncoimmunology 2021; 10:1945804. [PMID: 34290907 PMCID: PMC8274446 DOI: 10.1080/2162402x.2021.1945804] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chimeric antigen receptor T cells (CAR-T) targeting CD19 have achieved significant success in patients with B cell malignancies. To date, implementation of CAR-T in other indications remains challenging due to the lack of truly tumor-specific antigens as well as control of CAR-T activity in patients. CD123 is highly expressed in acute myeloid leukemia (AML) blasts including leukemia-initiating cells making it an attractive immunotherapeutic target. However, CD123 expression in normal hematopoietic progenitor cells and endothelia bears the risk of severe toxicities and may limit CAR-T applications lacking fine-tuned control mechanisms. Therefore, we recently developed a rapidly switchable universal CAR-T platform (UniCAR), in which CAR-T activity depends on the presence of a soluble adapter called targeting module (TM), and confirmed clinical proof-of-concept for targeting CD123 in AML with improved safety. As costimulation via 4–1BB ligand (4–1BBL) can enhance CAR-T expansion, persistence, and effector functions, a novel CD123-specific TM variant (TM123-4-1BBL) comprising trimeric single-chain 4–1BBL was developed for transient costimulation of UniCAR-T cells (UniCAR-T) at the leukemic site in trans. TM123-4-1BBL-directed UniCAR-T efficiently eradicated CD123-positive AML cells in vitro and in a CDX in vivo model. Moreover, additional costimulation via TM123-4-1BBL enabled enhanced expansion and persistence with a modulated UniCAR-T phenotype. In addition, the increased hydrodynamic volume of TM123-4-1BBL prolonged terminal plasma half-life and ensured a high total drug exposure in vivo. In conclusion, expanding the soluble adapter optionality for CD123-directed UniCAR-T maintains the platforms high anti-leukemic efficacy and immediate control mechanism for a flexible, safe, and individualized CAR-T therapy of AML patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Malte von Bonin
- Division of Hematology, Oncology and Stem Cell Transplantation, Medical Clinic I, Department of Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | | | | | | |
Collapse
|
28
|
Crawford A, Chiu D. Targeting Solid Tumors Using CD3 Bispecific Antibodies. Mol Cancer Ther 2021; 20:1350-1358. [PMID: 34045228 DOI: 10.1158/1535-7163.mct-21-0073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/06/2021] [Accepted: 05/24/2021] [Indexed: 11/16/2022]
Abstract
Immunotherapies to treat cancer have made tremendous progress over the past decade. In particular, T cell-directed therapies have gained considerable attention with CD3 bispecific antibodies and CAR T cells showing potent responses against hematologic tumors. At present, the ability to adapt these therapeutics to treat solid tumors is less established. Herein, we discuss recent advances in T cell-engaging CD3 bispecific antibodies targeting solid tumors, potential mechanisms of resistance, and future prospects. A better understanding of the mechanisms of immune evasion in solid tumors will enable the development of strategies to overcome this resistance and inform choices of therapeutic combinations.
Collapse
Affiliation(s)
| | - Danica Chiu
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| |
Collapse
|
29
|
Elshiaty M, Schindler H, Christopoulos P. Principles and Current Clinical Landscape of Multispecific Antibodies against Cancer. Int J Mol Sci 2021; 22:5632. [PMID: 34073188 PMCID: PMC8198225 DOI: 10.3390/ijms22115632] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 02/07/2023] Open
Abstract
Building upon the resounding therapeutic success of monoclonal antibodies, and supported by accelerating progress in engineering methods, the field of multispecific therapeutic antibodies is growing rapidly. Over 140 different molecules are currently in clinical testing, with excellent results in recent phase 1-3 clinical trials for several of them. Multivalent bispecific IgG-modified formats predominate today, with a clear tendency for more target antigens and further increased valency in newer constructs. The strategies to augment anticancer efficacy are currently equally divided between disruption of multiple surface antigens, and additional redirection of cytotoxic T or NK lymphocytes against the tumor. Both effects complement other modern modalities, such as tyrosine kinase inhibitors and adoptive cell therapies, with which multispecifics are increasingly applied in combination or merged, for example, in the form of antibody producing CAR-T cells and oncolytics. While mainly focused on B-cell malignancies early on, the contemporary multispecific antibody sector accommodates twice as many trials against solid compared to hematologic cancers. An exciting emerging prospect is the targeting of intracellular neoantigens using T-cell receptor (TCR) fusion proteins or TCR-mimic antibody fragments. Considering the fact that introduction of PD-(L)1 inhibitors only a few years ago has already facilitated 5-year survival rates of 30-50% for per se highly lethal neoplasms, such as metastatic melanoma and non-small-cell lung carcinoma, the upcoming enforcement of current treatments with "next-generation" immunotherapeutics, offers a justified hope for the cure of some advanced cancers in the near future.
Collapse
Affiliation(s)
- Mariam Elshiaty
- Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, 69126 Heidelberg, Germany; (M.E.); (H.S.)
- Translational Lung Cancer Center Heidelberg, Member of the German Center for Lung Research (DZL), 69126 Heidelberg, Germany
| | - Hannah Schindler
- Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, 69126 Heidelberg, Germany; (M.E.); (H.S.)
- Translational Lung Cancer Center Heidelberg, Member of the German Center for Lung Research (DZL), 69126 Heidelberg, Germany
| | - Petros Christopoulos
- Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, 69126 Heidelberg, Germany; (M.E.); (H.S.)
- Translational Lung Cancer Center Heidelberg, Member of the German Center for Lung Research (DZL), 69126 Heidelberg, Germany
| |
Collapse
|
30
|
Singh A, Dees S, Grewal IS. Overcoming the challenges associated with CD3+ T-cell redirection in cancer. Br J Cancer 2021; 124:1037-1048. [PMID: 33469153 PMCID: PMC7960983 DOI: 10.1038/s41416-020-01225-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/24/2020] [Accepted: 11/30/2020] [Indexed: 01/30/2023] Open
Abstract
The development of bispecific antibodies that redirect the cytotoxic activity of CD3+ T cells to tumours is a promising immunotherapeutic strategy for the treatment of haematological malignancies and solid cancers. Since the landmark FDA approval at the end of 2014 of the anti-CD3 × anti-CD19 bispecific antibody blinatumomab (Blincyto®) for the treatment of relapsed/refractory B-cell acute lymphoblastic leukaemia, ~100 clinical trials investigating the safety and efficacy of CD3+ bispecific T-cell redirectors for cancer have been initiated. However, despite early success, numerous challenges pertaining to CD3+ T-cell redirection in the context of cancer exist, including the recruitment of counterproductive CD3+ T-cell subsets, the release of systemic cytokines, the expansion of immune checkpoint molecules, the presence of an immunosuppressive tumour microenvironment, tumour antigen loss/escape, on-target off-tumour toxicity and suboptimal potency. The aim of the present review is to discuss novel approaches to overcome the key challenges associated with CD3+ bispecific T-cell redirection in order to achieve an optimal balance of anti-tumour activity and safety.
Collapse
Affiliation(s)
- Ajit Singh
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sundee Dees
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA, USA
| | - Iqbal S Grewal
- Janssen Biotherapeutics, The Janssen Pharmaceutical Companies of Johnson & Johnson, Spring House, PA, USA.
| |
Collapse
|
31
|
Heitmann JS, Pfluegler M, Jung G, Salih HR. Bispecific Antibodies in Prostate Cancer Therapy: Current Status and Perspectives. Cancers (Basel) 2021; 13:549. [PMID: 33535627 PMCID: PMC7867165 DOI: 10.3390/cancers13030549] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 12/29/2022] Open
Abstract
Prostate carcinoma (PC) is the second most common cancer in men. When the disease becomes unresponsive to androgen deprivation therapy, the remaining treatment options are of limited benefit. Despite intense efforts, none of the T cell-based immunotherapeutic strategies that meanwhile have become a cornerstone for treatment of other malignancies is established in PC. This refers to immune checkpoint inhibition (CI), which generally reinforces T cell immunity as well as chimeric antigen receptor T (CAR-T) cells and bispecific antibodies (bsAbs) that stimulate the T cell receptor/CD3-complex and mobilize T cells in a targeted manner. In general, compared to CAR-T cells, bsAb would have the advantage of being an "off the shelf" reagent associated with less preparative effort, but at present, despite enormous efforts, neither CAR-T cells nor bsAbs are successful in solid tumors. Here, we focus on the various bispecific constructs that are presently in development for treatment of PC, and discuss underlying concepts and the state of clinical evaluation as well as future perspectives.
Collapse
Affiliation(s)
- Jonas S. Heitmann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (J.S.H.); (M.P.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy” (IFIT), University of Tübingen, 72076 Tübingen, Germany;
| | - Martin Pfluegler
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (J.S.H.); (M.P.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy” (IFIT), University of Tübingen, 72076 Tübingen, Germany;
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
| | - Gundram Jung
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy” (IFIT), University of Tübingen, 72076 Tübingen, Germany;
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, 72076 Tübingen, Germany
| | - Helmut R. Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany; (J.S.H.); (M.P.)
- DFG Cluster of Excellence 2180 “Image-Guided and Functional Instructed Tumor Therapy” (IFIT), University of Tübingen, 72076 Tübingen, Germany;
| |
Collapse
|
32
|
Adamaki M, Zoumpourlis V. Immunotherapy as a Precision Medicine Tool for the Treatment of Prostate Cancer. Cancers (Basel) 2021; 13:E173. [PMID: 33419051 PMCID: PMC7825410 DOI: 10.3390/cancers13020173] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the most frequently diagnosed type of cancer among Caucasian males over the age of 60 and is characterized by remarkable heterogeneity and clinical behavior, ranging from decades of indolence to highly lethal disease. Despite the significant progress in PCa systemic therapy, therapeutic response is usually transient, and invasive disease is associated with high mortality rates. Immunotherapy has emerged as an efficacious and non-toxic treatment alternative that perfectly fits the rationale of precision medicine, as it aims to treat patients on the basis of patient-specific, immune-targeted molecular traits, so as to achieve the maximum clinical benefit. Antibodies acting as immune checkpoint inhibitors and vaccines entailing tumor-specific antigens seem to be the most promising immunotherapeutic strategies in offering a significant survival advantage. Even though patients with localized disease and favorable prognostic characteristics seem to be the ones that markedly benefit from such interventions, there is substantial evidence to suggest that the survival benefit may also be extended to patients with more advanced disease. The identification of biomarkers that can be immunologically targeted in patients with disease progression is potentially amenable in this process and in achieving significant advances in the decision for precision treatment of PCa.
Collapse
Affiliation(s)
- Maria Adamaki
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 48 Vassileos Constantinou Avenue, 11635 Athens, Greece;
| | | |
Collapse
|
33
|
Feldmann A, Hoffmann A, Bergmann R, Koristka S, Berndt N, Arndt C, Rodrigues Loureiro L, Kittel-Boselli E, Mitwasi N, Kegler A, Lamprecht C, González Soto KE, Bachmann M. Versatile chimeric antigen receptor platform for controllable and combinatorial T cell therapy. Oncoimmunology 2020; 9:1785608. [PMID: 32923149 PMCID: PMC7458653 DOI: 10.1080/2162402x.2020.1785608] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells show remarkable therapeutic effects in some hematological malignancies. However, CAR T cells can also cause life-threatening side effects. In order to minimize off-target and on-target/off-tumor reactions, improve safety, enable controllability, provide high flexibility, and increase tumor specificity, we established a novel humanized artificial receptor platform termed RevCARs. RevCAR genes encode for small surface receptors lacking any antigen-binding moiety. Steering of RevCAR T cells occurs via bispecific targeting molecules (TMs). The small size of RevCAR-encoding genes allows the construction of polycistronic vectors. Here, we demonstrate that RevCAR T cells efficiently kill tumor cells, can be steered by TMs, flexibly redirected against multiple targets, and used for combinatorial targeting following the "OR" and "AND" gate logic.
Collapse
Affiliation(s)
- Anja Feldmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Anja Hoffmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Ralf Bergmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Stefanie Koristka
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Nicole Berndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Claudia Arndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Liliana Rodrigues Loureiro
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Enrico Kittel-Boselli
- Tumor Immunology, University Cancer Center (UCC), University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Nicola Mitwasi
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Alexandra Kegler
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Chris Lamprecht
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,Department of Neurology, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Karla Elizabeth González Soto
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Michael Bachmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,Tumor Immunology, University Cancer Center (UCC), University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Dresden, Germany
| |
Collapse
|
34
|
Loureiro LR, Feldmann A, Bergmann R, Koristka S, Berndt N, Máthé D, Hegedüs N, Szigeti K, Videira PA, Bachmann M, Arndt C. Extended half-life target module for sustainable UniCAR T-cell treatment of STn-expressing cancers. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:77. [PMID: 32370811 PMCID: PMC7201957 DOI: 10.1186/s13046-020-01572-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/15/2020] [Indexed: 12/11/2022]
Abstract
Background Adapter chimeric antigen receptor (CAR) approaches have emerged has promising strategies to increase clinical safety of CAR T-cell therapy. In the UniCAR system, the safety switch is controlled via a target module (TM) which is characterized by a small-size and short half-life. The rapid clearance of these TMs from the blood allows a quick steering and self-limiting safety switch of UniCAR T-cells by TM dosing. This is mainly important during onset of therapy when tumor burden and the risk for severe side effects are high. For long-term UniCAR therapy, the continuous infusion of TMs may not be an optimal setting for the patients. Thus, in later stages of treatment, single infusions of TMs with an increased half-life might play an important role in long-term surveillance and eradication of residual tumor cells. Given this, we aimed to develop and characterize a novel TM with extended half-life targeting the tumor-associated carbohydrate sialyl-Tn (STn). Methods The extended half-life TM is composed of the STn-specific single-chain variable fragment (scFv) and the UniCAR epitope, fused to the hinge region and Fc domain of a human immunoglobulin 4 (IgG4) antibody. Specific binding and functionality of the αSTn-IgG4 TM as well as pharmacokinetic features were assessed using in vitro and in vivo assays and compared to the already established small-sized αSTn TM. Results The novel αSTn-IgG4 TM efficiently activates and redirects UniCAR T-cells to STn-expressing tumors in a target-specific and TM-dependent manner, thereby promoting the secretion of proinflammatory cytokines and tumor cell lysis in vitro and in experimental mice. Moreover, PET-imaging results demonstrate the specific enrichment of the αSTn-IgG4 TM at the tumor site, while presenting a prolonged serum half-life compared to the short-lived αSTn TM. Conclusion In a clinical setting, the combination of TMs with different formats and pharmacokinetics may represent a promising strategy for retargeting of UniCAR T-cells in a flexible, individualized and safe manner at particular stages of therapy. Furthermore, as these molecules can be used for in vivo imaging, they pose as attractive candidates for theranostic approaches.
Collapse
Affiliation(s)
- Liliana R Loureiro
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01328, Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Ralf Bergmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01328, Dresden, Germany.,Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Stefanie Koristka
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Nicole Berndt
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Nikolett Hegedüs
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Krisztián Szigeti
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Paula A Videira
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01328, Dresden, Germany. .,National Center for Tumor Diseases (NCT), Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany. .,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Tumor Immunology, University CancerCenter (UCC), University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany.
| | - Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01328, Dresden, Germany
| |
Collapse
|
35
|
Arndt C, Loureiro LR, Feldmann A, Jureczek J, Bergmann R, Máthé D, Hegedüs N, Berndt N, Koristka S, Mitwasi N, Fasslrinner F, Lamprecht C, Kegler A, Hoffmann A, Bartsch T, Köseer AS, Egan G, Schmitz M, Hořejší V, Krause M, Dubrovska A, Bachmann M. UniCAR T cell immunotherapy enables efficient elimination of radioresistant cancer cells. Oncoimmunology 2020; 9:1743036. [PMID: 32426176 PMCID: PMC7219270 DOI: 10.1080/2162402x.2020.1743036] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 01/30/2023] Open
Abstract
Induction or selection of radioresistant cancer (stem) cells following standard radiotherapy is presumably one of the major causes for recurrence of metastatic disease. One possibility to prevent tumor relapse is the application of targeted immunotherapies including, e.g., chimeric antigen receptor (CAR) T cells. In light of long-term remissions, it is highly relevant to clarify whether radioresistant cancer cells are susceptible to CAR T cell-mediated killing. To answer this question, we evaluated the anti-tumor activity of the switchable universal chimeric antigen receptor (UniCAR) system against highly radioresistant head and neck squamous cell carcinoma cells both in vitro and in vivo. Following specific UniCAR T cell engagement via EGFR or CD98 target modules, T cell effector mechanisms were induced including secretion of pro-inflammatory cytokines, up-regulation of granzyme B and perforin, as well as T cell proliferation. CD98- or EGFR-redirected UniCAR T cells further possess the capability to efficiently lyse radioresistant tumor cells. Observed anti-tumor effects were comparable to those against the radiosensitive parental cell lines. Finally, redirected UniCAR T cells significantly inhibited the growth of radioresistant cancer cells in immunodeficient mice. Taken together, our obtained data underline that the UniCAR system is able to overcome radioresistance. Thus, it represents an attractive technology for the development of combined radioimmunotherapeutic approaches that might improve the outcome of patients with metastatic radioresistant tumor diseases.
Collapse
Affiliation(s)
- Claudia Arndt
- Department of Radioimmunology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Liliana R Loureiro
- Department of Radioimmunology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Anja Feldmann
- Department of Radioimmunology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Justyna Jureczek
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,UniversityCancerCenter (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Ralf Bergmann
- Department of Radioimmunology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Semmelweis University, Department of Biophysics and Radiation Biology, Budapest, Hungary
| | - Domokos Máthé
- Semmelweis University, Department of Biophysics and Radiation Biology, Budapest, Hungary.,CROmed Translational Research Centers, Budapest, Hungary
| | - Nikolett Hegedüs
- Semmelweis University, Department of Biophysics and Radiation Biology, Budapest, Hungary
| | - Nicole Berndt
- Department of Radioimmunology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefanie Koristka
- Department of Radioimmunology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Nicola Mitwasi
- Department of Radioimmunology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Frederick Fasslrinner
- Medical Clinic and Polyclinic I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Chris Lamprecht
- Department of Neurology, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Alexandra Kegler
- Department of Radioimmunology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anja Hoffmann
- Department of Radioimmunology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Tabea Bartsch
- Department of Radioimmunology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Ayşe Sedef Köseer
- National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Gary Egan
- Monash Biomedical Imaging, School of Psychological Sciences, and Australian Research Council Centre of Excellence for Integrative Brain Function, Monash University, Melbourne, Australia
| | - Marc Schmitz
- National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Vaclav Hořejší
- Institute of Molecular Genetics of the Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Mechthild Krause
- National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany.,Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anna Dubrovska
- National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Michael Bachmann
- Department of Radioimmunology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,UniversityCancerCenter (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| |
Collapse
|
36
|
Mitwasi N, Feldmann A, Arndt C, Koristka S, Berndt N, Jureczek J, Loureiro LR, Bergmann R, Máthé D, Hegedüs N, Kovács T, Zhang C, Oberoi P, Jäger E, Seliger B, Rössig C, Temme A, Eitler J, Tonn T, Schmitz M, Hassel JC, Jäger D, Wels WS, Bachmann M. "UniCAR"-modified off-the-shelf NK-92 cells for targeting of GD2-expressing tumour cells. Sci Rep 2020; 10:2141. [PMID: 32034289 PMCID: PMC7005792 DOI: 10.1038/s41598-020-59082-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/20/2020] [Indexed: 12/13/2022] Open
Abstract
Antigen-specific redirection of immune effector cells with chimeric antigen receptors (CARs) demonstrated high therapeutic potential for targeting cancers of different origins. Beside CAR-T cells, natural killer (NK) cells represent promising alternative effectors that can be combined with CAR technology. Unlike T cells, primary NK cells and the NK cell line NK-92 can be applied as allogeneic off-the-shelf products with a reduced risk of toxicities. We previously established a modular universal CAR (UniCAR) platform which consists of UniCAR-expressing immune cells that cannot recognize target antigens directly but are redirected by a tumour-specific target module (TM). The TM contains an antigen-binding moiety fused to a peptide epitope which is recognized by the UniCAR molecule, thereby allowing an on/off switch of CAR activity, and facilitating flexible targeting of various tumour antigens depending on the presence and specificity of the TM. Here, we provide proof of concept that it is feasible to generate a universal off-the-shelf cellular therapeutic based on UniCAR NK-92 cells targeted to tumours expressing the disialoganglioside GD2 by GD2-specific TMs that are either based on an antibody-derived single-chain fragment variable (scFv) or an IgG4 backbone. Redirected UniCAR NK-92 cells induced specific killing of GD2-expressing cells in vitro and in vivo, associated with enhanced production of interferon-γ. Analysis of radiolabelled proteins demonstrated that the IgG4-based format increased the in vivo half-life of the TM markedly in comparison to the scFv-based molecule. In summary, UniCAR NK-92 cells represent a universal off-the-shelf platform that is highly effective and flexible, allowing the use of different TM formats for specific tumour targeting.
Collapse
Affiliation(s)
- Nicola Mitwasi
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefanie Koristka
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Nicole Berndt
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Justyna Jureczek
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Liliana R Loureiro
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,National Center for Tumor Diseases (NCT), University Hospital 'Carl Gustav Carus', TU Dresden, Dresden, Germany
| | - Ralf Bergmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Semmelweis University, Department of Biophysics and Radiation Biology, Budapest, Hungary
| | - Domokos Máthé
- Semmelweis University, Department of Biophysics and Radiation Biology, Budapest, Hungary
| | - Nikolett Hegedüs
- Semmelweis University, Department of Biophysics and Radiation Biology, Budapest, Hungary
| | | | - Congcong Zhang
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pranav Oberoi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elke Jäger
- Department of Hematology and Oncology, Krankenhaus Nordwest, Frankfurt am Main, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Claudia Rössig
- Department of Pediatric Hematology and Oncology, University Children´s Hospital Münster, Münster, Germany
| | - Achim Temme
- German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), University Hospital 'Carl Gustav Carus', TU Dresden, Dresden, Germany.,Department of Neurosurgery, Section Experimental Neurosurgery and Tumor Immunology, University Hospital 'Carl Gustav Carus', TU Dresden, Dresden, Germany
| | - Jiri Eitler
- Expermintal Transfusion Medicine, Medical Faculty 'Carl Gustav Carus', TU Dresden, Dresden, Germany
| | - Torsten Tonn
- German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Expermintal Transfusion Medicine, Medical Faculty 'Carl Gustav Carus', TU Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Marc Schmitz
- German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), University Hospital 'Carl Gustav Carus', TU Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, Dresden, Germany.,Institute of Immunology, Medical Faculty 'Carl Gustav Carus', TU Dresden, Dresden, Germany
| | - Jessica C Hassel
- Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Medical Center Heidelberg, Heidelberg, Germany
| | - Winfried S Wels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany. .,German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany. .,National Center for Tumor Diseases (NCT), University Hospital 'Carl Gustav Carus', TU Dresden, Dresden, Germany. .,Tumor Immunology, University Cancer Center (UCC) 'Carl Gustav Carus', TU Dresden, Dresden, Germany.
| |
Collapse
|
37
|
Frerichs KA, Broekmans MEC, Marin Soto JA, van Kessel B, Heymans MW, Holthof LC, Verkleij CPM, Boominathan R, Vaidya B, Sendecki J, Axel A, Gaudet F, Pillarisetti K, Zweegman S, Adams HC, Mutis T, van de Donk NWCJ. Preclinical Activity of JNJ-7957, a Novel BCMA×CD3 Bispecific Antibody for the Treatment of Multiple Myeloma, Is Potentiated by Daratumumab. Clin Cancer Res 2020; 26:2203-2215. [PMID: 31969333 DOI: 10.1158/1078-0432.ccr-19-2299] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 12/03/2019] [Accepted: 01/17/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Multiple myeloma (MM) patients with disease refractory to all available drugs have a poor outcome, indicating the need for new agents with novel mechanisms of action. EXPERIMENTAL DESIGN We evaluated the anti-MM activity of the fully human BCMA×CD3 bispecific antibody JNJ-7957 in cell lines and bone marrow (BM) samples. The impact of several tumor- and host-related factors on sensitivity to JNJ-7957 therapy was also evaluated. RESULTS We show that JNJ-7957 has potent activity against 4 MM cell lines, against tumor cells in 48 of 49 BM samples obtained from MM patients, and in 5 of 6 BM samples obtained from primary plasma cell leukemia patients. JNJ-7957 activity was significantly enhanced in patients with prior daratumumab treatment, which was partially due to enhanced killing capacity of daratumumab-exposed effector cells. BCMA expression did not affect activity of JNJ-7957. High T-cell frequencies and high effector:target ratios were associated with improved JNJ-7957-mediated lysis of MM cells. The PD-1/PD-L1 axis had a modest negative impact on JNJ-7957 activity against tumor cells from daratumumab-naïve MM patients. Soluble BCMA impaired the ability of JNJ-7957 to kill MM cells, although higher concentrations were able to overcome this negative effect. CONCLUSIONS JNJ-7957 effectively kills MM cells ex vivo, including those from heavily pretreated MM patients, whereby several components of the immunosuppressive BM microenvironment had only modest effects on its killing capacity. Our findings support the ongoing trial with JNJ-7957 as single agent and provide the preclinical rationale for evaluating JNJ-7957 in combination with daratumumab in MM.
Collapse
Affiliation(s)
- Kristine A Frerichs
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Marloes E C Broekmans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Jhon A Marin Soto
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Berris van Kessel
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Martijn W Heymans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Biostatistics, Amsterdam, the Netherlands
| | - Lisa C Holthof
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Christie P M Verkleij
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | | | - Bhavesh Vaidya
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | - Jocelyn Sendecki
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | - Amy Axel
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | - Francois Gaudet
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | | | - Sonja Zweegman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Homer C Adams
- Janssen Research and Development, LLC, Spring House, Pennsylvania
| | - Tuna Mutis
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Niels W C J van de Donk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
38
|
Therapeutic Mesenchymal Stromal Cells for Immunotherapy and for Gene and Drug Delivery. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 16:204-224. [PMID: 32071924 PMCID: PMC7012781 DOI: 10.1016/j.omtm.2020.01.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mesenchymal stromal cells (MSCs) possess several fairly unique properties that, when combined, make them ideally suited for cellular-based immunotherapy and as vehicles for gene and drug delivery for a wide range of diseases and disorders. Key among these are: (1) their relative ease of isolation from a variety of tissues; (2) the ability to be expanded in culture without a loss of functionality, a property that varies to some degree with tissue source; (3) they are relatively immune-inert, perhaps obviating the need for precise donor/recipient matching; (4) they possess potent immunomodulatory functions that can be tailored by so-called licensing in vitro and in vivo; (5) the efficiency with which they can be modified with viral-based vectors; and (6) their almost uncanny ability to selectively home to damaged tissues, tumors, and metastases following systemic administration. In this review, we summarize the latest research in the immunological properties of MSCs, their use as immunomodulatory/anti-inflammatory agents, methods for licensing MSCs to customize their immunological profile, and their use as vehicles for transferring both therapeutic genes in genetic disease and drugs and genes designed to destroy tumor cells.
Collapse
|
39
|
Feldmann A, Arndt C, Koristka S, Berndt N, Bergmann R, Bachmann MP. Conventional CARs versus modular CARs. Cancer Immunol Immunother 2019; 68:1713-1719. [PMID: 31542798 PMCID: PMC6805801 DOI: 10.1007/s00262-019-02399-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 09/16/2019] [Indexed: 01/23/2023]
Abstract
The clinical application of immune effector cells genetically modified to express chimeric antigen receptors (CARs) has shown impressive results including complete remissions of certain malignant hematological diseases. However, their application can also cause severe side effects such as cytokine release syndrome (CRS) or tumor lysis syndrome (TLS). One limitation of currently applied CAR T cells is their lack of regulation. Especially, an emergency shutdown of CAR T cells in case of life-threatening side effects is missing. Moreover, targeting of tumor-associated antigens (TAAs) that are not only expressed on tumor cells but also on vital tissues requires the possibility of a switch allowing to repeatedly turn the activity of CAR T cells on and off. Here we summarize the development of a modular CAR variant termed universal CAR (UniCAR) system that promises to overcome these limitations of conventional CARs.
Collapse
Affiliation(s)
- Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Stefanie Koristka
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Nicole Berndt
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ralf Bergmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Michael P Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany.
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- University Cancer Center (UCC) Dresden, Tumor Immunology, Carl Gustav Carus' Technische Universität Dresden, Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.
| |
Collapse
|
40
|
Arndt C, Feldmann A, Koristka S, Schäfer M, Bergmann R, Mitwasi N, Berndt N, Bachmann D, Kegler A, Schmitz M, Puentes-Cala E, Soto JA, Ehninger G, Pietzsch J, Liolios C, Wunderlich G, Kotzerke J, Kopka K, Bachmann M. A theranostic PSMA ligand for PET imaging and retargeting of T cells expressing the universal chimeric antigen receptor UniCAR. Oncoimmunology 2019; 8:1659095. [PMID: 31646084 PMCID: PMC6791425 DOI: 10.1080/2162402x.2019.1659095] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/19/2019] [Accepted: 08/18/2019] [Indexed: 01/26/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells have shown impressive therapeutic potential. Due to the lack of direct control mechanisms, therapy-related adverse reactions including cytokine release- and tumor lysis syndrome can even become life-threatening. In case of target antigen expression on non-malignant cells, CAR T cells can also attack healthy tissues. To overcome such side effects, we have established a modular CAR platform termed UniCAR: UniCAR T cells per se are inert as they recognize a peptide epitope (UniCAR epitope) that is not accessible on the surface of living cells. Bifunctional adapter molecules termed target modules (TM) can cross-link UniCAR T cells with target cells. In the absence of TMs, UniCAR T cells automatically turn off. Until now, all UniCAR TMs were constructed by fusion of the UniCAR epitope to an antibody domain. To open up the wide field of low-molecular-weight compounds for retargeting of UniCAR T cells to tumor cells, and to follow in parallel the progress of UniCAR T cell therapy by PET imaging we challenged the idea to convert a PET tracer into a UniCAR-TM. For proof of concept, we selected the clinically used PET tracer PSMA-11, which binds to the prostate-specific membrane antigen overexpressed in prostate carcinoma. Here we show that fusion of the UniCAR epitope to PSMA-11 results in a low-molecular-weight theranostic compound that can be used for both retargeting of UniCAR T cells to tumor cells, and for non-invasive PET imaging and thus represents a member of a novel class of theranostics.
Collapse
Affiliation(s)
- Claudia Arndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Anja Feldmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Stefanie Koristka
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Martin Schäfer
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ralf Bergmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Nicola Mitwasi
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Nicole Berndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Dominik Bachmann
- UniversityCancerCenter (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Alexandra Kegler
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Marc Schmitz
- Institute of Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | | | | | | | - Jens Pietzsch
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,Department of Chemistry and Food Chemistry, School of Science, TU Dresden, Dresden, Germany
| | - Christos Liolios
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gerd Wunderlich
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Jörg Kotzerke
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,Department of Nuclear Medicine, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), partner site Dresden, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Klaus Kopka
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Michael Bachmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,UniversityCancerCenter (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
41
|
Koristka S, Ziller-Walter P, Bergmann R, Arndt C, Feldmann A, Kegler A, Cartellieri M, Ehninger A, Ehninger G, Bornhäuser M, Bachmann MP. Anti-CAR-engineered T cells for epitope-based elimination of autologous CAR T cells. Cancer Immunol Immunother 2019; 68:1401-1415. [PMID: 31414180 PMCID: PMC6768917 DOI: 10.1007/s00262-019-02376-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 08/07/2019] [Indexed: 12/16/2022]
Abstract
Although CAR T-cell therapy has demonstrated tremendous clinical efficacy especially in hematological malignancies, severe treatment-associated toxicities still compromise the widespread application of this innovative technology. Therefore, developing novel approaches to abrogate CAR T-cell-mediated side effects is of great relevance. Several promising strategies pursue the selective antibody-based depletion of adoptively transferred T cells via elimination markers. However, given the limited half-life and tissue penetration, dependence on the patients’ immune system and on-target/off-side effects of proposed monoclonal antibodies, we sought to exploit αCAR-engineered T cells to efficiently eliminate CAR T cells. For comprehensive and specific recognition, a small peptide epitope (E-tag) was incorporated into the extracellular spacer region of CAR constructs. We provide first proof-of-concept for targeting this epitope by αE-tag CAR T cells, allowing an effective killing of autologous E-tagged CAR T cells both in vitro and in vivo whilst sparing cells lacking the E-tag. In addition to CAR T-cell cytotoxicity, the αE-tag-specific T cells can be empowered with cancer-fighting ability in case of relapse, hence, have versatile utility. Our proposed methodology can most probably be implemented in CAR T-cell therapies regardless of the targeted tumor antigen aiding in improving overall safety and survival control of highly potent gene-modified cells.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Autoantigens/immunology
- Cytotoxicity, Immunologic
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Genetic Engineering
- Humans
- Immunotherapy, Adoptive/methods
- Male
- Mice
- Neoplasm Recurrence, Local
- PC-3 Cells
- Peptide Fragments/genetics
- Prostatic Neoplasms/immunology
- Prostatic Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Chimeric Antigen/genetics
- T-Lymphocytes, Cytotoxic/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Stefanie Koristka
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Pauline Ziller-Walter
- Tumor Immunology, University Cancer Center (UCC), 'Carl Gustav Carus' Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Ralf Bergmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Claudia Arndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Anja Feldmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Alexandra Kegler
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Marc Cartellieri
- Cellex Patient Treatment GmbH, Tatzberg 47, 01307, Dresden, Germany
| | - Armin Ehninger
- GEMoaB Monoclonals GmbH, Tatzberg 47, 01307, Dresden, Germany
| | - Gerhard Ehninger
- Cellex Patient Treatment GmbH, Tatzberg 47, 01307, Dresden, Germany
- GEMoaB Monoclonals GmbH, Tatzberg 47, 01307, Dresden, Germany
| | - Martin Bornhäuser
- Medical Clinic and Policlinic I, University Hospital 'Carl Gustav Carus' Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 'Carl Gustav Carus' Technische Universität Dresden, Dresden, Germany
| | - Michael P Bachmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328, Dresden, Germany.
- Tumor Immunology, University Cancer Center (UCC), 'Carl Gustav Carus' Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
- German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), 'Carl Gustav Carus' Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
42
|
Jureczek J, Bergmann R, Berndt N, Koristka S, Kegler A, Puentes-Cala E, Soto JA, Arndt C, Bachmann M, Feldmann A. An oligo-His-tag of a targeting module does not influence its biodistribution and the retargeting capabilities of UniCAR T cells. Sci Rep 2019; 9:10547. [PMID: 31332252 PMCID: PMC6646371 DOI: 10.1038/s41598-019-47044-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022] Open
Abstract
Recently, we established the controllable modular UniCAR platform technology to advance the efficacy and safety of CAR T cell therapy. The UniCAR system is composed of (i) target modules (TMs) and (ii) UniCAR armed T cells. TMs are bispecific molecules that are able to bind to the tumor cell surface and simultaneously to UniCAR T cells. For interaction with UniCAR T cells, TMs contain a peptide epitope sequence which is recognised by UniCAR T cells. So far, a series of TMs against a variety of tumor targets including against the prostate stem cell antigen (PSCA) were constructed and functionally characterised. In order to facilitate their purification all these TMs are expressed as recombinant proteins equipped with an oligo-His-tag. The aim of the here presented manuscript was to learn whether or not the oligo-His-tag of the TM influences the UniCAR system. For this purpose, we constructed TMs against PSCA equipped with or lacking an oligo-His-tag. Both TMs were compared side by side including for functionality and biodistribution. According to our data, an oligo-His-tag of a UniCAR TM has only little if any effect on its binding affinity, in vitro and in vivo killing capability and in vivo biodistribution.
Collapse
Affiliation(s)
- Justyna Jureczek
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Ralf Bergmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Nicole Berndt
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Stefanie Koristka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Alexandra Kegler
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | | | | | - Claudia Arndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Michael Bachmann
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany. .,University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany. .,Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany. .,National Center for Tumor Diseases (NCT), University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.
| | - Anja Feldmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| |
Collapse
|
43
|
Trabolsi A, Arumov A, Schatz JH. T Cell–Activating Bispecific Antibodies in Cancer Therapy. THE JOURNAL OF IMMUNOLOGY 2019; 203:585-592. [DOI: 10.4049/jimmunol.1900496] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/03/2019] [Indexed: 01/13/2023]
|
44
|
Arndt C, Bachmann M, Bergmann R, Berndt N, Feldmann A, Koristka S. Theranostic CAR T cell targeting: A brief review. J Labelled Comp Radiopharm 2019; 62:533-540. [PMID: 30889625 DOI: 10.1002/jlcr.3727] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/13/2019] [Indexed: 12/21/2022]
Abstract
More than hundred years ago, Paul Ehrlich postulated that our immune system should be able to recognize tumor cells. Just recently, the development of check point inhibitors, bispecific antibodies, and T cells genetically modified to express chimeric antigen receptors (CARs) underlines the true power of our immune system. T cells genetically modified with CARs can lead to complete remission of malignant hematologic diseases. However, they can also cause life-threatening side effects. In case of cytokine release syndrome, tumor lysis syndrome, or deadly side effects on the central nervous system, an emergency shut down of CAR T cells is needed. Targeting of tumor-associated antigens that are also expressed on vital tissues require a possibility to repeatedly switch the activity of CAR T cells on and off on demand and to follow the treatment by imaging. Theranostic, modular CARs such as the UniCAR system may help to overcome these problems.
Collapse
Affiliation(s)
- Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.,UniversityCancerCenter (UCC) Dresden, Tumor Immunology, 'Carl Gustav Carus' Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), partner site Dresden, Dresden, Germany
| | - Ralf Bergmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Nicole Berndt
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefanie Koristka
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| |
Collapse
|
45
|
The UniCAR system: A modular CAR T cell approach to improve the safety of CAR T cells. Immunol Lett 2019; 211:13-22. [PMID: 31091431 DOI: 10.1016/j.imlet.2019.05.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 12/19/2022]
Abstract
The idea to eliminate tumor cells via our own immune system is more than a hundred years old. However, a real break through came first with the development of check point inhibitors, bispecific antibodies (bsAbs) and T cells genetically modified to express Chimeric Antigen Receptors (CARs). Eventhough the clinical application of T cells equipped with CARs can lead to a complete remission, unfortunately, their application can also cause severe or even life threatening side effects as their activity can no more be adjusted once given to the patient. For targeting of tumor cells expressing tumor associated antigens (TAAs) which are not limited to tumor cells but also accessible on healthy tissues CAR T cells should not be permanently in a killing mode but be equipped with some kind of a switch whereby the activity of CAR T cells can reversely be turned "on and off ". Moreover, in case of cytokine release syndrome (CRS), tumor lysis syndrome (TLS), or other deadly side effects the possibility of an emergency shut down of the CAR T cell activity should exist. Modular CAR variants such as the UniCAR system may fulfill these requirements.
Collapse
|
46
|
Albert S, Koristka S, Gerbaulet A, Cartellieri M, Arndt C, Feldmann A, Berndt N, Loureiro LR, von Bonin M, Ehninger G, Eugster A, Bonifacio E, Bornhäuser M, Bachmann MP, Ehninger A. Tonic Signaling and Its Effects on Lymphopoiesis of CAR-Armed Hematopoietic Stem and Progenitor Cells. THE JOURNAL OF IMMUNOLOGY 2019; 202:1735-1746. [PMID: 30728213 DOI: 10.4049/jimmunol.1801004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/04/2019] [Indexed: 01/01/2023]
Abstract
Long-term survival of adoptively transferred chimeric Ag receptor (CAR) T cells is often limited. Transplantation of hematopoietic stem cells (HSCs) transduced to express CARs could help to overcome this problem as CAR-armed HSCs can continuously deliver CAR+ multicell lineages (e.g., T cells, NK cells). In dependence on the CAR construct, a variable extent of tonic signaling in CAR T cells was reported; thus, effects of CAR-mediated tonic signaling on the hematopoiesis of CAR-armed HSCs is unclear. To assess the effects of tonic signaling, two CAR constructs were established and analyzed 1) a signaling CAR inducing a solid Ag-independent tonic signaling termed CAR-28/ζ and 2) a nonstimulating control CAR construct lacking intracellular signaling domains termed CAR-Stop. Bone marrow cells from immunocompetent mice were isolated, purified for HSC-containing Lin-cKit+ cells or the Lin-cKit+ Sca-1+ subpopulation (Lin-Sca-1+cKit+), and transduced with both CAR constructs. Subsequently, modified bone marrow cells were transferred into irradiated mice, in which they successfully engrafted and differentiated into hematopoietic progenitors. HSCs expressing the CAR-Stop sustained normal hematopoiesis. In contrast, expression of the CAR-28/ζ led to elimination of mature CAR+ T and B cells, suggesting that the CAR-mediated tonic signaling mimics autorecognition via the newly recombined immune receptors in the developing lymphocytes.
Collapse
Affiliation(s)
- Susann Albert
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany.,University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Stefanie Koristka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Alexander Gerbaulet
- Institute of Immunology, Medical Faculty Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | | | - Claudia Arndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Anja Feldmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Nicole Berndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Liliana R Loureiro
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Malte von Bonin
- German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Gerhard Ehninger
- University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,National Center for Tumor Diseases (NCT), partner site Dresden, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Anne Eugster
- Center for Regenerative Therapies Dresden (CRTD), Technical University Dresden, 01307 Dresden, Germany; and
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden (CRTD), Technical University Dresden, 01307 Dresden, Germany; and
| | - Martin Bornhäuser
- University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,National Center for Tumor Diseases (NCT), partner site Dresden, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Michael P Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; .,University Cancer Center (UCC), Tumor Immunology, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,National Center for Tumor Diseases (NCT), partner site Dresden, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany
| | - Armin Ehninger
- Medical Clinic and Policlinic I, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany.,GEMoaB Monoclonals GmbH, 01307 Dresden, Germany
| |
Collapse
|
47
|
David T, Hlinová V, Kubíček V, Bergmann R, Striese F, Berndt N, Szöllősi D, Kovács T, Máthé D, Bachmann M, Pietzsch HJ, Hermann P. Improved Conjugation, 64-Cu Radiolabeling, in Vivo Stability, and Imaging Using Nonprotected Bifunctional Macrocyclic Ligands: Bis(Phosphinate) Cyclam (BPC) Chelators. J Med Chem 2018; 61:8774-8796. [PMID: 30180567 DOI: 10.1021/acs.jmedchem.8b00932] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Bifunctional derivatives of bis(phosphinate)-bearing cyclam (BPC) chelators bearing a carboxylate, amine, isothiocyanate, azide, or cyclooctyne in the BP side chain were synthesized. Conjugations required no protection of phosphinate or ring secondary amine groups. The ring amines were not reactive (proton protected) at pH < ∼8. For isothiocyanate coupling, oligopeptide N-terminal α-amines were more suitable than alkyl amines, e.g., Lys ω-amine (p Ka ∼7.5-8.5 and ∼10-11, respectively) due to lower basicity. The Cu-64 labeling was efficient at room temperature (specific activity ∼100 GBq/μmol; 25 °C, pH 6.2, ∼100 ligand equiv, 10 min). A representative Cu-64-BPC was tested in vivo showing fast clearance and no nonspecific radioactivity deposition. The monoclonal anti-PSCA antibody 7F5 conjugates with thiocyanate BPC derivative or NODAGA were radiolabeled and studied in PC3-PSCA tumor bearing mice by PET. The radiolabeled BPC conjugate was accumulated in the prostate tumor with a low off-target uptake, unlike Cu-64-labeled NODAGA-antibody conjugate. The BPC chelators have a great potential for theranostic applications of the Cu-64/Cu-67 matched pair.
Collapse
Affiliation(s)
- Tomáš David
- Department of Inorganic Chemistry, Faculty of Science , Charles University , Hlavova 2030 , 128 40 Prague , Czech Republic.,Institute of Radiopharmaceutical Cancer Research , Helmholtz-Zentrum Dresden-Rossendorf , Bautzner Landstrasse 400 , 01328 Dresden , Germany
| | - Veronika Hlinová
- Department of Inorganic Chemistry, Faculty of Science , Charles University , Hlavova 2030 , 128 40 Prague , Czech Republic
| | - Vojtěch Kubíček
- Department of Inorganic Chemistry, Faculty of Science , Charles University , Hlavova 2030 , 128 40 Prague , Czech Republic
| | - Ralf Bergmann
- Institute of Radiopharmaceutical Cancer Research , Helmholtz-Zentrum Dresden-Rossendorf , Bautzner Landstrasse 400 , 01328 Dresden , Germany
| | - Franziska Striese
- Institute of Radiopharmaceutical Cancer Research , Helmholtz-Zentrum Dresden-Rossendorf , Bautzner Landstrasse 400 , 01328 Dresden , Germany
| | - Nicole Berndt
- Partner Site Dresden , German Cancer Consortium (DKTK) , Fetscherstrasse 74 , 01307 Dresden , Germany.,German Cancer Research Center (DKFZ) , Im Neuenheimer Feld 280 , 69120 Heidelberg , Germany
| | - Dávid Szöllősi
- Department of Biophysics and Radiation Biology , Semmelweis University , Tűzoltó utca 37-47 , H-1094 Budapest , Hungary
| | - Tibor Kovács
- Institute of Radiochemistry and Radioecology , University of Pannonia , Egyetem St. 10 , H-8200 Veszprém , Hungary.,Social Organization for Radioecological Cleanliness , P.O. Box 158, H-8200 Veszprém , Hungary
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology , Semmelweis University , Tűzoltó utca 37-47 , H-1094 Budapest , Hungary
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research , Helmholtz-Zentrum Dresden-Rossendorf , Bautzner Landstrasse 400 , 01328 Dresden , Germany.,Tumor Immunology, University Cancer Center (UCC) , "Carl Gustav Carus" Technische Universität Dresden , Fetscherstrasse 74 , 01307 Dresden , Germany.,National Center for Tumor Diseases (NCT) , "Carl Gustav Carus" Technische Universität Dresden , Fetscherstrasse 74 , 01307 Dresden , Germany
| | - Hans-Jürgen Pietzsch
- Institute of Radiopharmaceutical Cancer Research , Helmholtz-Zentrum Dresden-Rossendorf , Bautzner Landstrasse 400 , 01328 Dresden , Germany
| | - Petr Hermann
- Department of Inorganic Chemistry, Faculty of Science , Charles University , Hlavova 2030 , 128 40 Prague , Czech Republic
| |
Collapse
|
48
|
Loureiro LR, Feldmann A, Bergmann R, Koristka S, Berndt N, Arndt C, Pietzsch J, Novo C, Videira P, Bachmann M. Development of a novel target module redirecting UniCAR T cells to Sialyl Tn-expressing tumor cells. Blood Cancer J 2018; 8:81. [PMID: 30190468 PMCID: PMC6127150 DOI: 10.1038/s41408-018-0113-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 06/08/2018] [Accepted: 07/10/2018] [Indexed: 01/21/2023] Open
Affiliation(s)
- L R Loureiro
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Lisboa, Portugal.,Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,UniversityCancerCenter (UCC) Dresden, Tumor Immunology, 'Carl Gustav Carus' Technische Universität Dresden, Dresden, Germany
| | - A Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - R Bergmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - S Koristka
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - N Berndt
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), partner site Dresden, Heidelberg, Germany
| | - C Arndt
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - J Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Dresden, Germany
| | - C Novo
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Lisboa, Portugal.,UEIPM, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, Lisboa, Portugal.,CDG & Allies, Professional and Patient Association International Network (PPAIN), Caparica, Portugal
| | - P Videira
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Lisboa, Portugal.,UEIPM, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, Lisboa, Portugal.,CDG & Allies, Professional and Patient Association International Network (PPAIN), Caparica, Portugal
| | - M Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany. .,UniversityCancerCenter (UCC) Dresden, Tumor Immunology, 'Carl Gustav Carus' Technische Universität Dresden, Dresden, Germany. .,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), partner site Dresden, Heidelberg, Germany. .,National Center of Tumor Diseases (NCT), partner site Dresden, Dresden, Germany.
| |
Collapse
|
49
|
Albert S, Arndt C, Koristka S, Berndt N, Bergmann R, Feldmann A, Schmitz M, Pietzsch J, Steinbach J, Bachmann M. From mono- to bivalent: improving theranostic properties of target modules for redirection of UniCAR T cells against EGFR-expressing tumor cells in vitro and in vivo. Oncotarget 2018; 9:25597-25616. [PMID: 29876011 PMCID: PMC5986651 DOI: 10.18632/oncotarget.25390] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/28/2018] [Indexed: 02/06/2023] Open
Abstract
CAR-modified T cells show impressive results in clinical trials. However, cytokine release syndrome and "on-target, off-tumor" reactions represent most concerning side effects. To improve the safety of CAR-T cell therapy, we established a switchable CAR platform termed UniCAR system consisting of two components: UniCAR-modified T cells and tumor-specific target modules (TM). For treatment of EGFR+ epithelial tumors, we recently described a monovalent nanobody-based α-EGFR TM, either expressed in bacteria or eukaryotic cells. In spite of the identical primary sequence the eukaryotic TM showed a reduced killing capability and affinity. Here we describe a novel bivalent α-EGFR-EGFR TM. As expected, the avidity of the bivalent TM is higher than that of its monovalent counterpart. Binding of neither the monovalent α-EGFR TM nor the bivalent α-EGFR-EGFR TM to EGFR effected the EGF-mediated signaling. While the monovalent α-EGFR TM could only mediate the killing of tumor cells expressing high levels of EGFR, the bivalent α-EGFR-EGFR TM could redirect UniCAR T cells to tumor cells expressing low levels of EGFR. According to PET experiments in vivo, the increased avidity of the bivalent α-EGFR-EGFR TM improves the enrichment at the tumor site and its use for PET imaging.
Collapse
Affiliation(s)
- Susann Albert
- UniversityCancerCenter (UCC) Dresden, Tumor Immunology, ‘Carl Gustav Carus’ Technische Universität Dresden, Dresden, Germany
| | - Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefanie Koristka
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Nicole Berndt
- German Cancer Consortium (DKTK), part\ner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ralf Bergmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Marc Schmitz
- German Cancer Consortium (DKTK), part\ner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Immunology, Medical Faculty, ‘Carl Gustav Carus’ Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), partner site Dresden, Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Germany
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- National Center for Tumor Diseases (NCT), partner site Dresden, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Germany
| | - Michael Bachmann
- UniversityCancerCenter (UCC) Dresden, Tumor Immunology, ‘Carl Gustav Carus’ Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- German Cancer Consortium (DKTK), part\ner site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), partner site Dresden, Dresden, Germany
| |
Collapse
|
50
|
Feldmann A, Arndt C, Bergmann R, Loff S, Cartellieri M, Bachmann D, Aliperta R, Hetzenecker M, Ludwig F, Albert S, Ziller-Walter P, Kegler A, Koristka S, Gärtner S, Schmitz M, Ehninger A, Ehninger G, Pietzsch J, Steinbach J, Bachmann M. Retargeting of T lymphocytes to PSCA- or PSMA positive prostate cancer cells using the novel modular chimeric antigen receptor platform technology "UniCAR". Oncotarget 2018; 8:31368-31385. [PMID: 28404896 PMCID: PMC5458214 DOI: 10.18632/oncotarget.15572] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 01/04/2017] [Indexed: 11/25/2022] Open
Abstract
New treatment options especially of solid tumors including for metastasized prostate cancer (PCa) are urgently needed. Recent treatments of leukemias with chimeric antigen receptors (CARs) underline their impressive therapeutic potential. However CARs currently applied in the clinics cannot be repeatedly turned on and off potentially leading to severe life threatening side effects. To overcome these problems, we recently described a modular CAR technology termed UniCAR: UniCAR T cells are inert but can be turned on by application of one or multiple target modules (TMs). Here we present preclinical data summarizing the retargeting of UniCAR T cells to PCa cells using TMs directed to prostate stem cell- (PSCA) or/and prostate specific membrane antigen (PSMA). In the presence of the respective TM(s), we see a highly efficient target-specific and target-dependent activation of UniCAR T cells, secretion of pro-inflammatory cytokines, and PCa cell lysis both in vitro and experimental mice.
Collapse
Affiliation(s)
- Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Ralf Bergmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Simon Loff
- UniversityCancerCenter (UCC) 'Carl Gustav Carus' TU Dresden, Tumor Immunology, Dresden, Germany.,GEMoaB Monoclonals GmbH, Dresden, Germany
| | - Marc Cartellieri
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Cellex Patient Treatment GmbH, Dresden, Germany
| | - Dominik Bachmann
- UniversityCancerCenter (UCC) 'Carl Gustav Carus' TU Dresden, Tumor Immunology, Dresden, Germany
| | - Roberta Aliperta
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Mirjam Hetzenecker
- UniversityCancerCenter (UCC) 'Carl Gustav Carus' TU Dresden, Tumor Immunology, Dresden, Germany
| | - Florian Ludwig
- UniversityCancerCenter (UCC) 'Carl Gustav Carus' TU Dresden, Tumor Immunology, Dresden, Germany
| | - Susann Albert
- UniversityCancerCenter (UCC) 'Carl Gustav Carus' TU Dresden, Tumor Immunology, Dresden, Germany
| | - Pauline Ziller-Walter
- UniversityCancerCenter (UCC) 'Carl Gustav Carus' TU Dresden, Tumor Immunology, Dresden, Germany
| | - Alexandra Kegler
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefanie Koristka
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sebastian Gärtner
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Marc Schmitz
- Institute of Immunology, 'Carl Gustav Carus', TU Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden; and German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Dresden, 'Carl Gustav Carus' TU Dresden, Dresden, Germany
| | | | - Gerhard Ehninger
- UniversityCancerCenter (UCC) 'Carl Gustav Carus' TU Dresden, Tumor Immunology, Dresden, Germany.,Medical Clinic and Policlinic I, University Hospital 'Carl Gustav Carus', TU Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden; and German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Dresden, 'Carl Gustav Carus' TU Dresden, Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Department of Chemistry and Food Chemistry, School of Science, TU Dresden, Dresden, Germany
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden; and German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Dresden, 'Carl Gustav Carus' TU Dresden, Dresden, Germany.,Department of Chemistry and Food Chemistry, School of Science, TU Dresden, Dresden, Germany
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,UniversityCancerCenter (UCC) 'Carl Gustav Carus' TU Dresden, Tumor Immunology, Dresden, Germany.,Medical Clinic and Policlinic I, University Hospital 'Carl Gustav Carus', TU Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), partner site Dresden; and German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Dresden, 'Carl Gustav Carus' TU Dresden, Dresden, Germany
| |
Collapse
|