30 results on '"Constanze Heise"'
Search Results
2. Microphthalmia-Associated Transcription Factor (MITF) Regulates Immune Cell Migration into Melanoma
- Author
-
Gabriela M Wiedemann, Celina Aithal, Angelina Kraechan, Constanze Heise, Bruno L Cadilha, Jin Zhang, Peter Duewell, Robert Ballotti, Stefan Endres, Corine Bertolotto, and Sebastian Kobold
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Microphthalmia-associated transcription factor (MITF) is a key transcription factor in melanoma development and progression. MITF amplification and downregulation have been observed in a significant proportion of melanoma patients and correlate with clinical outcomes. Here, we have investigated the effect of MITF on melanoma chemokine expression and immune cell attraction. In B16F10 melanoma cells, MITF knockdown reduced expression of CXCL10, with concomitantly decreased attraction of immune cells and accelerated tumor outgrowth. Conversely, overexpression of MITF in YUMM1.1 melanoma cells also led to an increased immune cell attraction in vitro. Subcutaneous YUMM1.1 melanomas overexpressing MITF however showed a reduced immune infiltration of lymphocytes and an increased tumor growth. In human melanoma cell lines, silencing of MITF enhanced chemokine production and immune cell attraction, while overexpression of MITF led to lower immune cell attraction. In summary, our results show that MITF regulates chemokine expression in murine and in human melanoma cells, and affects in vivo immune cell attraction and tumor growth. These results reveal a functional relationship between MITF and immune cell infiltration, which may be exploited for cancer therapy.
- Published
- 2019
- Full Text
- View/download PDF
3. PD1-CD28 Fusion Protein Enables CD4+ T Cell Help for Adoptive T Cell Therapy in Models of Pancreatic Cancer and Non-hodgkin Lymphoma
- Author
-
Felicitas Rataj, Fabian B. T. Kraus, Michael Chaloupka, Simon Grassmann, Constanze Heise, Bruno L. Cadilha, Peter Duewell, Stefan Endres, and Sebastian Kobold
- Subjects
adoptive T cell transfer ,cancer immunotherapy ,costimulation ,PD-1-CD28 fusion protein ,CD4+ T cells ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Background: Interaction of the programmed death receptor 1 (PD-1) and its ligand, PD-L1, suppresses T cell activity and permits tumors to evade T cell-mediated immune surveillance. We have recently demonstrated that antigen-specific CD8+ T cells transduced with a PD1-CD28 fusion protein are protected from PD-1-mediated inhibition. We have now investigated the potential of PD1-CD28 fusion protein-transduced CD4+ T cells alone or in combination with CD8+ T cells for immunotherapy of pancreatic cancer and non-Hodgkin lymphoma.Methods: OVA-specific CD4+ and CD8+ were retrovirally transduced with the PD1-CD28 fusion protein. Cytokine release, proliferation, cytotoxic activity, and phenotype of transduced T cells were assessed in the context of Panc02-OVA (murine pancreatic cancer model) and E.G7-PD-L1 (murine T cell lymphoma model) cells.Results: Stimulation of PD1-CD28 fusion protein-transduced CD4+ T cells with anti-CD3 and recombinant PD-L1 induced specific T cell activation, as measured by IFN-y release and T cell proliferation. Coculture with Panc02-OVA or E.G7-PD-L1 tumor cells also led to specific activation of CD4+ T cells. Cytokine release and T cell proliferation was most effective when tumor cells simultaneously encountered genetically engineered CD4+ and CD8+ T cells. Synergy between both cell populations was also observed for specific tumor cell lysis. T cell cytotoxicity was mediated via granzyme B release and mediated enhanced tumor control in vivo. Transduced CD4+ and CD8+ T cells in co-culture with tumor cells developed a predominant central memory phenotype over time. Different ratios of CD4+ and CD8+ transduced T cells led to a significant increase of IFN-y and IL-2 secretion positively correlating with CD4+ T cell numbers used. Mechanistically, IL-2 and MHC-I were central to the synergistic activity of CD4+ and CD8+ T cells, since neutralization of IL-2 prevented the crosstalk between these cell populations.Conclusion: PD1-CD28 fusion protein-transduced CD4+ T cells significantly improved anti-tumoral effect of fusion protein-transduced CD8+ T cells. Thus, our results indicate that PD1-CD28 fusion protein-transduced CD4+ T cells have the potential to overcome the PD-1-PD-L1 immunosuppressive axis in pancreatic cancer and non-Hodgkin lymphoma.
- Published
- 2018
- Full Text
- View/download PDF
4. Cyclic Dichalcogenides Extend the Reach of Bioreductive Prodrugs to Harness Thiol/Disulfide Oxidoreductases: Applications to seco-Duocarmycins Targeting the Thioredoxin System
- Author
-
Jan G. Felber, Annabel Kitowski, Lukas Zeisel, Martin S. Maier, Constanze Heise, Julia Thorn-Seshold, and Oliver Thorn-Seshold
- Subjects
General Chemical Engineering ,General Chemistry - Published
- 2023
- Full Text
- View/download PDF
5. Supplementary figure legend from Bispecific Antibodies Enable Synthetic Agonistic Receptor-Transduced T Cells for Tumor Immunotherapy
- Author
-
Sebastian Kobold, Christian Klein, Stefan Endres, Claudio Sustmann, Gerhard Niederfellner, Peter Duewell, Jian Ding, Daniel Lamp, Martin Jastroch, Johannes vom Berg, Ramona Murr, Constanze Heise, Stefanie Lesch, Bruno L. Cadilha, Felicitas Rataj, Martina Geiger, Richard Klaus, Mathias Kurzay, Moritz L. Schmidbauer, Mohamed-Reda Benmebarek, and Clara H. Karches
- Abstract
Supplementary figure legend
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 1 from Bispecific Antibodies Enable Synthetic Agonistic Receptor-Transduced T Cells for Tumor Immunotherapy
- Author
-
Sebastian Kobold, Christian Klein, Stefan Endres, Claudio Sustmann, Gerhard Niederfellner, Peter Duewell, Jian Ding, Daniel Lamp, Martin Jastroch, Johannes vom Berg, Ramona Murr, Constanze Heise, Stefanie Lesch, Bruno L. Cadilha, Felicitas Rataj, Martina Geiger, Richard Klaus, Mathias Kurzay, Moritz L. Schmidbauer, Mohamed-Reda Benmebarek, and Clara H. Karches
- Abstract
Supplementary Figure 1: Murine SAR expression and immunophenotyping of transduced T cells
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 5 from Bispecific Antibodies Enable Synthetic Agonistic Receptor-Transduced T Cells for Tumor Immunotherapy
- Author
-
Sebastian Kobold, Christian Klein, Stefan Endres, Claudio Sustmann, Gerhard Niederfellner, Peter Duewell, Jian Ding, Daniel Lamp, Martin Jastroch, Johannes vom Berg, Ramona Murr, Constanze Heise, Stefanie Lesch, Bruno L. Cadilha, Felicitas Rataj, Martina Geiger, Richard Klaus, Mathias Kurzay, Moritz L. Schmidbauer, Mohamed-Reda Benmebarek, and Clara H. Karches
- Abstract
Supplementary Figure 5: Effect of pre-activated SAR T cells on hepatocellular carcinoma and glioblastoma cells

- Published
- 2023
- Full Text
- View/download PDF
8. Data from Bispecific Antibodies Enable Synthetic Agonistic Receptor-Transduced T Cells for Tumor Immunotherapy
- Author
-
Sebastian Kobold, Christian Klein, Stefan Endres, Claudio Sustmann, Gerhard Niederfellner, Peter Duewell, Jian Ding, Daniel Lamp, Martin Jastroch, Johannes vom Berg, Ramona Murr, Constanze Heise, Stefanie Lesch, Bruno L. Cadilha, Felicitas Rataj, Martina Geiger, Richard Klaus, Mathias Kurzay, Moritz L. Schmidbauer, Mohamed-Reda Benmebarek, and Clara H. Karches
- Abstract
Purpose:Genetically engineered T cells are powerful anticancer treatments but are limited by safety and specificity issues. We herein describe an MHC-unrestricted modular platform combining autologous T cells, transduced with a targetable synthetic agonistic receptor (SAR), with bispecific antibodies (BiAb) that specifically recruit and activate T cells for tumor killing.Experimental Design:BiAbs of different formats were generated by recombinant expression. T cells were retrovirally transduced with SARs. T-cell activation, proliferation, differentiation, and T-cell–induced lysis were characterized in three murine and human tumor models in vitro and in vivo.Results:Murine T cells transduced with SAR composed of an extracellular domain EGFRvIII fused to CD28 and CD3ζ signaling domains could be specifically recruited toward murine tumor cells expressing EpCAM by anti-EGFRvIII × anti-EpCAM BiAb. BiAb induced selective antigen-dependent activation, proliferation of SAR T cells, and redirected tumor cell lysis. Selectivity was dependent on the monovalency of the antibody for EGFRvIII. We identified FAS ligand as a major mediator of killing utilized by the T cells. Similarly, human SAR T cells could be specifically redirected toward mesothelin-expressing human pancreatic cancer cells. In vivo, treatment with SAR T cells and BiAb mediated antitumoral activity in three human pancreatic cancer cell xenograft models. Importantly, SAR activity, unlike CAR activity, was reversible in vitro and in vivo.Conclusions:We describe a novel ACT platform with antitumor activity in murine and human tumor models with a distinct mode of action that combines adoptive T-cell therapy with bispecific antibodies.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 4 from Bispecific Antibodies Enable Synthetic Agonistic Receptor-Transduced T Cells for Tumor Immunotherapy
- Author
-
Sebastian Kobold, Christian Klein, Stefan Endres, Claudio Sustmann, Gerhard Niederfellner, Peter Duewell, Jian Ding, Daniel Lamp, Martin Jastroch, Johannes vom Berg, Ramona Murr, Constanze Heise, Stefanie Lesch, Bruno L. Cadilha, Felicitas Rataj, Martina Geiger, Richard Klaus, Mathias Kurzay, Moritz L. Schmidbauer, Mohamed-Reda Benmebarek, and Clara H. Karches
- Abstract
Supplementary Figure 4: Human SAR T cells are specifically stimulated by anti-human mesothelin x anti-EGFRv3 2 + 1 BiAb and redirect lysis to mesothelin+ target cells in vitro and in vivo
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure 3 from Bispecific Antibodies Enable Synthetic Agonistic Receptor-Transduced T Cells for Tumor Immunotherapy
- Author
-
Sebastian Kobold, Christian Klein, Stefan Endres, Claudio Sustmann, Gerhard Niederfellner, Peter Duewell, Jian Ding, Daniel Lamp, Martin Jastroch, Johannes vom Berg, Ramona Murr, Constanze Heise, Stefanie Lesch, Bruno L. Cadilha, Felicitas Rataj, Martina Geiger, Richard Klaus, Mathias Kurzay, Moritz L. Schmidbauer, Mohamed-Reda Benmebarek, and Clara H. Karches
- Abstract
Supplementary Figure 3: In contrast to SAR T cells, CAR T cells rely on the release of perforin for efficient killing.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 2 from Bispecific Antibodies Enable Synthetic Agonistic Receptor-Transduced T Cells for Tumor Immunotherapy
- Author
-
Sebastian Kobold, Christian Klein, Stefan Endres, Claudio Sustmann, Gerhard Niederfellner, Peter Duewell, Jian Ding, Daniel Lamp, Martin Jastroch, Johannes vom Berg, Ramona Murr, Constanze Heise, Stefanie Lesch, Bruno L. Cadilha, Felicitas Rataj, Martina Geiger, Richard Klaus, Mathias Kurzay, Moritz L. Schmidbauer, Mohamed-Reda Benmebarek, and Clara H. Karches
- Abstract
Supplementary Figure 2: Soluble and bound EGFR ligands do not trigger unwanted SAR T cell activation, while antibodies monovalent and bivalent for SAR can trigger T cell activation to the same extent.
- Published
- 2023
- Full Text
- View/download PDF
12. Cyclic dichalcogenides extend the reach of bioreductive prodrugs to harness the thioredoxin system: applications toseco-duocarmycins
- Author
-
Jan G. Felber, Annabel Kitowski, Lukas Zeisel, Martin S. Maier, Constanze Heise, Julia Thorn-Seshold, and Oliver Thorn-Seshold
- Abstract
Small molecule prodrug approaches that can activate cancer therapeutics selectively in tumors are urgently needed. Here, we developed the first antitumor prodrugs designed for activation by the thioredoxin (Trx) oxidoreductase system. This critical cellular disulfide redox axis is tightly linked to dysregulated redox/metabolic states in cancer, yet it cannot be addressed by current bioreductive prodrugs, which mainly cluster around oxidised nitrogen species. We instead harnessed Trx/TrxR-specific artificial dichalcogenides to gate the bioactivity of a series of 10 “off-to-on” reduction-activated duocarmycin prodrugs. The prodrugs were tested for cell-free and cellular activity dependent on reducing enzyme systems in 177 cell lines, to establish broad trends for redox-based cellular bioactivity of the dichalcogenides. They were well toleratedin vivoin mice, indicating low systemic release of their duocarmycin cargo, andin vivoanti-tumor efficacy trials in mouse models of breast and pancreatic cancer gave promising initial results indicating effective tumoral drug release, presumably byin situbioreductive activation. This work therefore presents a chemically novel class of bioreductive prodrugs against a previously unaddressed reductase type, validates its ability to accessin vivocompatible small-molecule prodrugs even of potently cumulative toxins, and so introduces carefully tuned dichalcogenides as a platform strategy for specific bioreduction-based release.
- Published
- 2022
- Full Text
- View/download PDF
13. Self-reporting styrylthiazolium photopharmaceuticals: mitochondrial localisation as well as SAR drive biological activity
- Author
-
Li Gao, Yvonne Kraus, Andrea Stegner, Thomas Wein, Constanze Heise, Leonie von Brunn, Elena Fajardo-Ruiz, Julia Thorn-Seshold, and Oliver Thorn-Seshold
- Subjects
Cell Death ,Organic Chemistry ,Physical and Theoretical Chemistry ,Coloring Agents ,Biochemistry ,Azo Compounds ,Mitochondria - Abstract
Novel photoswitches with features complementary to the well-established azobenzenes are increasingly driving high-precision research in photopharmacology in cells and in vivo. Styrylthiazolium (StyTz) and styrylbenzothiazolium (StyBtz) are cellularly untested E/Z-isomerisation photoswitches which are nearly isosteric to azobenzenes, but have distinct photophysical and physicochemical properties: including ca. 60 nm red-shifted π→π* absorption, self-reporting fluorescence, Z→E relaxation that matches typical biological timescales, and good solubility due to their permanent positive charge. We here tested StyTz and StyBtz for their potential as photopharmaceutical scaffolds, by applying them to photocontrol the dynamics of the microtubule cytoskeleton. We observed light-specific disruption of microtubule network architecture and antiproliferative activity, with a structure-activity relationship matching expectations for tubulin inhibitors. However, while testing the lead StyBtz2 for its molecular mechanism of action, we found that it did not inhibit microtubule dynamics. We tracked its localisation in live cells by relying on its self-reporting fluorescence, observing accumulation of E-StyBtz2 into mitochondria; after several minutes of illumination it was then released into the cytosol concomitant with blebbing and cell death. We interpret this as light-dependent catastrophic rupturing of mitochondria on acute timescales. We conclude that StyTz/StyBtz can be interesting photopharmaceutical scaffolds for addressing mitochondrial, rather than cytosolic, targets.
- Published
- 2022
14. [5]-Helistatins: Tubulin binding helicenes with antimitotic activity
- Author
-
James Rushworth, Aditya Thawani, Elena Fajardo-Ruiz, Joyce Meiring, Constanze Heise, Andrew White, Anna Akhmanova, Jochen Brandt, Oliver Thorn-Seshold, and Matthew Fuchter
- Abstract
Helicenes are high interest synthetic targets with unique conjugated helical structures that have found important technological applications. Despite this interest, helicenes have had limited impact in chemical biology. Herein, we disclose a first-in-class antimitotic helicene, helistatin 1 (HA-1), where the helicene scaffold acts as a structural mimic of colchicine, a known antimitotic drug. The synthesis proceeds via sequential Pd-catalyzed coupling reactions and a π-Lewis acid cycloisomerization mediated by PtCl2. HA-1 was found to block microtubule polymerisation in both cell-free and live cell assays. Not only does this demonstrate the feasibility of using helicenes as bioactive scaffolds against protein targets, but also suggests wider potential for the use of helicenes as isosteres of biaryls or cis-stilbenes - themselves common drug and natural product scaffolds. Overall, this study further supports future opportunities for helicenes for a range of chemical biological applications.
- Published
- 2022
- Full Text
- View/download PDF
15. PD-1-CD28 fusion protein strengthens mesothelin-specific TRuC T cells in preclinical solid tumor models
- Author
-
Stefanie Lesch, Alessia Nottebrock, Felicitas Rataj, Constanze Heise, Stefan Endres, and Sebastian Kobold
- Subjects
Cancer Research ,Oncology ,Adoptive Cell Transfer ,Immunosuppression ,Pd-1-cd28 Fusion Protein ,T Cell Hypofunction ,Truc T Cells ,Molecular Medicine ,General Medicine - Abstract
Background T cell receptor fusion constructs (TRuC) consist of an antibody-based single chain variable fragment (scFv) fused to a T cell receptor chain (TCR) and allow recognition of cancer cells in an HLA-independent manner. Unlike chimeric antigen receptors (CAR), TRuC are integrated into the TCR complex resulting in a functional chimera with novel specificity, whilst retaining TCR signaling. To further enhance anti-tumor function, we expressed a PD-1-CD28 fusion receptor in TRuC T cells aiming to prevent tumor-induced immune suppression and T cell anergy. Methods The activation level of engineered T cells was investigated in co-culture experiments with tumor cells followed by quantification of released cytokines using ELISA. To study T cell-mediated tumor cell lysis in vitro, impedance-based real-time tumor cell killing and LDH release was measured. Finally, two xenograft mouse cancer models were employed to explore the therapeutic potential of engineered T cells. Results In co-culture assays, co-expression of PD-1-CD28 enhanced cytokine production of TRuC T cells. This effect was dependent on PD-L1 to PD-1-CD28 interactions, as blockade of PD-L1 amplified IFN-γ production in unmodified TRuC T cells to a greater level compared to TRuC-PD-1-CD28 T cells. In vivo, PD-1-CD28 co-expression supported the anti-tumor efficacy of TRuC T cells in two xenograft mouse cancer models. Conclusion Together, these results demonstrate the therapeutic potential of PD-1-CD28 co-expression in TRuC T cells to prevent PD-L1-induced T cell hypofunction.
- Published
- 2022
- Full Text
- View/download PDF
16. In vivo photocontrol of microtubule dynamics and integrity, migration and mitosis, by the potent GFP-imaging-compatible photoswitchable reagents SBTubA4P and SBTub2M
- Author
-
Anna Akhmanova, Constanze Heise, M. Wranik, Oliver Thorn-Seshold, Clemens Cabernard, Li Gao, Adam Varady, Jörg Standfuss, Jennifer A. Taylor, Joyce C.M. Meiring, Andreas R. Bausch, Beatrice Terni, Iris E. Ruider, Artur Llobet, Martin Distel, Cecilia D. Velasco, Michel O. Steinmetz, and Julia Thorn-Seshold
- Subjects
Cell division ,Photoswitch ,Microtubule ,Chemistry ,In vivo ,ved/biology ,ved/biology.organism_classification_rank.species ,Biophysics ,Cytoskeleton ,Model organism ,Mitosis ,Green fluorescent protein - Abstract
Photoswitchable reagents to modulate microtubule stability and dynamics are an exciting tool approach towards micron- and millisecond-scale control over endogenous cytoskeleton-dependent processes. When these reagents are globally administered yet locally photoactivated in 2D cell culture, they can exert precise biological control that would have great potential forin vivotranslation across a variety of research fields and for all eukaryotes. However, photopharmacology’s reliance on the azobenzene photoswitch scaffold has been accompanied by a failure to translate this temporally- and cellularly-resolved control to 3D models or toin vivoapplications in multi-organ animals, which we attribute substantially to the metabolic liabilities of azobenzenes.Here, we optimised the potency and solubility of metabolically stable, druglike colchicinoid microtubule inhibitors based instead on the styrylbenzothiazole (SBT) photoswitch scaffold, that are non-responsive to the major fluorescent protein imaging channels and so enable multiplexed imaging studies. We applied these reagents to 3D systems (organoids, tissue explants) and classic model organisms (zebrafish, clawed frog) with one- and two-protein imaging experiments. We successfully used systemic treatment plus spatiotemporally-localised illuminationsin vivoto photocontrol microtubule dynamics, network architecture, and microtubule-dependent processes in these systems with cellular precision and second-level resolution. These nanomolar,in vivo-capable photoswitchable reagents can prove a game-changer for high-precision cytoskeleton research in cargo transport, cell motility, cell division and development. More broadly, their straightforward design can also inspire the development of similarly capable optical reagents for a range of protein targets, so bringing generalin vivophotopharmacology one step closer to productive realisation.
- Published
- 2021
- Full Text
- View/download PDF
17. Pyrrole Hemithioindigo Antimitotics with Near-Quantitative Bidirectional Photoswitching Photocontrol Cellular Microtubule Dynamics with Single-Cell Precision
- Author
-
Alexander Sailer, Joyce Meiring, Constanze Heise, Linda Pettersson, Anna Akhmanova, Julia Thorn-Seshold, and Oliver Thorn-Seshold
- Abstract
Photoswitchably bioactive reagents, known as “photopharmaceuticals”, promise powerful applications in high-precision biological research. Yet most photoswitch scaffolds cannot be quantitatively bidirectionally photoisomerised, so they suffer from residual background activity that can confound experiments. We rationally designed photopharmaceuticals using the emerging near-quantitative photoswitch pyrrole hemithioindigo (PHTubs), to isomer-specifically inhibit the cytoskeletal protein tubulin. These PHTub reagents allow simultaneous visible-light imaging and photoswitching in live cells, where they could be used for cell-precise photomodulation of microtubule dynamics, and photocontrol over cell cycle progression and cell death. This is, as far as we know, the first use of a hemithioindigo photopharmaceutical for high-spatiotemporal-resolution biological control in live cells. This work opens up new horizons for high-precision microtubule research using PHTubs; and shows the cellular applicability of the near-quantitative photoswitch pyrrole hemithioindigo as a valuable scaffold for photocontrol of a range of other biological targets.
- Published
- 2021
- Full Text
- View/download PDF
18. T cells armed with C-X-C chemokine receptor type 6 enhance adoptive cell therapy for pancreatic tumours
- Author
-
Alessia Nottebrock, Eric Tartour, Klaus-Peter Janssen, Nicholas Tokarew, Christine Hoerth, Moritz Thomas, Johannes Lutz, Niels Halama, Martin Jastroch, Öykü Umut, Simon Grassmann, Andrew S. Liss, Maximilian Reichert, Stefanie Lesch, Philipp Metzger, S Stoiber, Benjamin Larimer, Justyna Ogonek, Bruno L. Cadilha, M. Kurzay, Jasper N. Pruessmann, Felicitas Rataj, Mauro Di Pilato, Max Schnurr, Viktoria Blumenberg, Stephan Kruger, Sebastian Kobold, Matthias Seifert, Lene Vimeux, Zahra Dantes, Carsten Marr, Klara Dorman, M. Benmebarek, C. Karches, Moritz Rapp, Duc Huynh, Stefan Endres, Lars M. König, Thi Anh-Dao Tran, Constanze Heise, Dario Dhoqina, Daniel Lamp, Marion Subklewe, Thorsten R. Mempel, Emmanuel Donnadieu, Ruth Grünmeier, Anna Reischer, Svenja Ruehland, Michael Hristov, Adrian Gottschlich, Taisuke Baba, Peter Duewell, Steffen Ormanns, Sandy Joaquina, Simon Rothenfusser, Thomas Hank, Arman Oener, Remco T. A. Megens, Biomedische Technologie, and RS: Carim - B01 Blood proteins & engineering
- Subjects
0301 basic medicine ,RECRUITMENT ,EXPRESSION ,INFILTRATING LYMPHOCYTES ,medicine.medical_treatment ,T-Lymphocytes ,Cell- and Tissue-Based Therapy ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,CXC CHEMOKINE ,Immunotherapy, Adoptive ,TRANSDUCTION ,Article ,Cell therapy ,Mice ,03 medical and health sciences ,Chemokine receptor ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Antigen ,Pancreatic cancer ,medicine ,Animals ,Humans ,IMMUNOTHERAPY ,Receptors, CXCR6 ,Chemistry ,LOCALIZATION ,medicine.disease ,CHIMERIC ANTIGEN RECEPTOR ,CANCER ,C-X-C Chemokine Receptor Type 6 ,Chimeric antigen receptor ,Computer Science Applications ,Pancreatic Neoplasms ,030104 developmental biology ,IMMUNE CELLS ,Mesothelin ,Cancer research ,Receptors, Chemokine ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Forced expression of C-X-C chemokine receptor type 6 in antigen-specific T cells enhanced the recognition and lysis of pancreatic cancer cells and the efficacy of adoptive cell therapy for pancreatic cancer.The efficacy of adoptive cell therapy for solid tumours is hampered by the poor accumulation of the transferred T cells in tumour tissue. Here, we show that forced expression of C-X-C chemokine receptor type 6 (whose ligand is highly expressed by human and murine pancreatic cancer cells and tumour-infiltrating immune cells) in antigen-specific T cells enhanced the recognition and lysis of pancreatic cancer cells and the efficacy of adoptive cell therapy for pancreatic cancer. In mice with subcutaneous pancreatic tumours treated with T cells with either a transgenic T-cell receptor or a murine chimeric antigen receptor targeting the tumour-associated antigen epithelial cell adhesion molecule, and in mice with orthotopic pancreatic tumours or patient-derived xenografts treated with T cells expressing a chimeric antigen receptor targeting mesothelin, the T cells exhibited enhanced intratumoral accumulation, exerted sustained anti-tumoral activity and prolonged animal survival only when co-expressing C-X-C chemokine receptor type 6. Arming tumour-specific T cells with tumour-specific chemokine receptors may represent a promising strategy for the realization of adoptive cell therapy for solid tumours.
- Published
- 2021
- Full Text
- View/download PDF
19. Bispecific antibodies enable synthetic agonistic receptor-transduced T cells for tumor immunotherapy
- Author
-
C. Karches, Felicitas Rataj, Constanze Heise, Johannes vom Berg, Bruno L. Cadilha, M. Benmebarek, Peter Duewell, Gerhard Niederfellner, Jian Ding, M. Kurzay, Stefanie Lesch, Stefan Endres, Daniel Lamp, Martin Jastroch, Sebastian Kobold, Moritz L. Schmidbauer, Martina Geiger, Ramona Murr, Christian Klein, R. Klaus, Claudio Sustmann, University of Zurich, and Kobold, Sebastian
- Subjects
0301 basic medicine ,Cancer Research ,CD3 Complex ,T cell ,medicine.medical_treatment ,T-Lymphocytes ,Melanoma, Experimental ,Receptors, Antigen, T-Cell ,Mice, Transgenic ,610 Medicine & health ,Mice, SCID ,Immunotherapy, Adoptive ,Fas ligand ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,CD28 Antigens ,Mice, Inbred NOD ,Antibodies, Bispecific ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,10239 Institute of Laboratory Animal Science ,1306 Cancer Research ,Receptor ,biology ,Chemistry ,CD28 ,Immunotherapy ,Epithelial Cell Adhesion Molecule ,Xenograft Model Antitumor Assays ,In vitro ,3. Good health ,ErbB Receptors ,Mice, Inbred C57BL ,Pancreatic Neoplasms ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Mesothelin ,biology.protein ,Cancer research ,570 Life sciences ,2730 Oncology ,Antibody - Abstract
Purpose: Genetically engineered T cells are powerful anticancer treatments but are limited by safety and specificity issues. We herein describe an MHC-unrestricted modular platform combining autologous T cells, transduced with a targetable synthetic agonistic receptor (SAR), with bispecific antibodies (BiAb) that specifically recruit and activate T cells for tumor killing. Experimental Design: BiAbs of different formats were generated by recombinant expression. T cells were retrovirally transduced with SARs. T-cell activation, proliferation, differentiation, and T-cell–induced lysis were characterized in three murine and human tumor models in vitro and in vivo. Results: Murine T cells transduced with SAR composed of an extracellular domain EGFRvIII fused to CD28 and CD3ζ signaling domains could be specifically recruited toward murine tumor cells expressing EpCAM by anti-EGFRvIII × anti-EpCAM BiAb. BiAb induced selective antigen-dependent activation, proliferation of SAR T cells, and redirected tumor cell lysis. Selectivity was dependent on the monovalency of the antibody for EGFRvIII. We identified FAS ligand as a major mediator of killing utilized by the T cells. Similarly, human SAR T cells could be specifically redirected toward mesothelin-expressing human pancreatic cancer cells. In vivo, treatment with SAR T cells and BiAb mediated antitumoral activity in three human pancreatic cancer cell xenograft models. Importantly, SAR activity, unlike CAR activity, was reversible in vitro and in vivo. Conclusions: We describe a novel ACT platform with antitumor activity in murine and human tumor models with a distinct mode of action that combines adoptive T-cell therapy with bispecific antibodies.
- Published
- 2019
20. Photoswitchable microtubule stabilisers optically control tubulin cytoskeleton structure and function
- Author
-
Rebekkah Bingham, Dirk Trauner, Adrian Müller-Deku, Constanze Heise, Kristina Loy, Oliver Thorn-Seshold, Julia Ahlfeld, and Yvonne Kraus
- Subjects
0303 health sciences ,biology ,010405 organic chemistry ,Chemistry ,Neurodegeneration ,Motility ,Cell cycle ,medicine.disease ,01 natural sciences ,Small molecule ,0104 chemical sciences ,Structure and function ,03 medical and health sciences ,Tubulin ,Microtubule ,biology.protein ,medicine ,Biophysics ,Cytoskeleton ,030304 developmental biology - Abstract
Small molecule inhibitors provide a versatile method for studies in microtubule cytoskeleton research, since tubulin is not readily amenable to functional control using genetics. However, traditional chemical inhibitors do not allow spatiotemporally precise applications on the length and time scales appropriate for selectively modulating microtubule-dependent processes. We have synthesised a panel of taxane-based light-responsive microtubule stabilisers, whose tubulin hyperpolymerisation activity can be induced by photoisomerisation to their thermodynamically metastable state. These reagents can be isomerised in live cells, optically controlling microtubule network integrity, cell cycle repartition, and cell survival, and offering biological response on the timescale of seconds and spatial precision to the level of individual cells. These azobenzene-based microtubule stabilisers offer the possibility of noninvasive, highly spatiotemporally precise modulation of the microtubule cytoskeleton in live cells, and can prove powerful reagents for studies of intracellular transport, cell motility, and neurodegeneration.Abstract Figure
- Published
- 2019
- Full Text
- View/download PDF
21. Isoquinoline-Based Biaryls as a Robust Scaffold for Microtubule Inhibitors
- Author
-
Benedikt Melzer, Oliver Thorn-Seshold, Monique Preuße, Constanze Heise, Franz Bracher, Yvonne Kraus, Li Gao, Carina Glas, and Julia Ahlfeld
- Subjects
Scaffold ,Microtubule dynamics ,Cancer therapy ,Antineoplastic Agents ,HL-60 Cells ,01 natural sciences ,Microtubules ,Polymerization ,03 medical and health sciences ,chemistry.chemical_compound ,Structure-Activity Relationship ,Microtubule ,Drug Discovery ,Humans ,Isoquinoline ,Cells, Cultured ,030304 developmental biology ,Cell Proliferation ,Pharmacology ,0303 health sciences ,Microscopy, Confocal ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Organic Chemistry ,Cell Cycle ,General Medicine ,Prodrug ,Druglikeness ,Isoquinolines ,Combinatorial chemistry ,Tubulin Modulators ,0104 chemical sciences ,Tubulin Inhibitors ,chemistry ,Drug Screening Assays, Antitumor ,HeLa Cells - Abstract
We here report the discovery of isoquinoline-based biaryls as a new scaffold for colchicine domain tubulin inhibitors. Colchicine domain inhibitors comprise a structurally diverse class of compounds offering highly desirable cytotoxic and vascular disrupting bioactivities. Current research on colchicine domain inhibitors is focused on improving in vivo robustness and tolerability: properties that are inextricably linked to the scaffold structure employed. The isoquinoline-based biaryl scaffold we now report offers high-potency tubulin inhibition with excellent robustness and druglikeness, allowing solubility, in vivo tolerability and facile late-stage structural diversification through a tolerant synthetic route. We have confirmed the tubulin-binding properties and mechanism of these isoquinoline-based biaryls through a series of cellular tests and established safe in vivo dosing parameters in mice. By addressing several problems facing the current families of inhibitors, we thus expect that this new scaffold will find a range of powerful in vivo applications towards translational use in cancer therapy.
- Published
- 2019
- Full Text
- View/download PDF
22. High-affinity CD16-polymorphism and Fc-engineered antibodies enable activity of CD16-chimeric antigen receptor-modified T cells for cancer therapy
- Author
-
Justyna Ogonek, Constanze Heise, Stefan Endres, Christian Klein, Felicitas Rataj, S Stoiber, Bruno L. Cadilha, Sebastian Kobold, Erwin van Puijenbroek, Florian Asang, Peter Duewell, Severin Johannes Jacobi, and Nicholas Tokarew
- Subjects
Cancer Research ,medicine.medical_treatment ,T cell ,T-Lymphocytes ,chemical and pharmacologic phenomena ,Lymphocyte Activation ,Immunotherapy, Adoptive ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,immune system diseases ,Cell Line, Tumor ,Neoplasms ,medicine ,Cytotoxic T cell ,Animals ,Humans ,Cell Proliferation ,CD20 ,Polymorphism, Genetic ,Receptors, Chimeric Antigen ,biology ,Chemistry ,Receptors, IgG ,Antibody-Dependent Cell Cytotoxicity ,virus diseases ,Antibodies, Monoclonal ,hemic and immune systems ,Immunotherapy ,Chimeric antigen receptor ,biological factors ,3. Good health ,Immunoglobulin Fc Fragments ,ErbB Receptors ,Killer Cells, Natural ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Cancer research ,Tumour immunology ,Antibody ,Rituximab - Abstract
BACKGROUND CD16-chimeric antigen receptors (CAR) T cells recognise the Fc-portion of therapeutic antibodies, which can enable the selective targeting of different antigens. Limited evidence exists as to which CD16-CAR design and antibody partner might be most effective. We have hypothesised that the use of high-affinity CD16 variants, with increased Fc-terminus antibody affinity, combined with Fc-engineered antibodies, would provide superior CD16-CAR T cell efficacy. METHODS CD16-CAR T (wild-type or variants) cells were co-cultured with Panc-1 pancreatic cancer, Raji lymphoma or A375 melanoma cells in the presence or absence of anti-CD20, anti-MCSP, wild-type or the glycoengineered antibody variants. The endpoints were proliferation, activation, and cytotoxicity in vitro. RESULTS The CD16 158 V variant of CD16-CAR T cells showed increased cytotoxic activity against all the tested cancer cells in the presence of the wild-type antibody directed against MCSP or CD20. Glycoengineered antibodies enhanced CD16-CAR T cell activity irrespective of CD16 polymorphisms as compared with the wild-type antibody. The combination of the glycoengineered antibodies with the CD16-CAR 158 V variant synergised as seen by the increase in all endpoints. CONCLUSION These results indicate that CD16-CAR with the high-affinity CD16 variant 158 V, combined with Fc-engineered antibodies, have high anti-tumour efficacy.
- Published
- 2019
- Full Text
- View/download PDF
23. Arming T cells with C-X-C-motive receptor 6 enables adoptive T cell therapy of pancreatic cancer
- Author
-
Bruno L. Cadilha, Stefanie Lesch, M. Reichert, Marion Subklewe, J. Lutz, Constanze Heise, Thorsten R. Mempel, Steffen Ormanns, Z. Dantes, Anna Reischer, Peter Duewell, S Stoiber, S. Endres, S. Kobold, R. Megens, Justyna Ogonek, Jasper N. Pruessmann, C. Karches, Simon Grassmann, Felicitas Rataj, V. Blumenberg, M. Di Pilato, Max Schnurr, S. Ruehland, and Klara Dorman
- Subjects
Cancer Research ,medicine.anatomical_structure ,Oncology ,business.industry ,T cell ,Pancreatic cancer ,Cancer research ,medicine ,Receptor ,medicine.disease ,business - Published
- 2019
- Full Text
- View/download PDF
24. Cancer cells induce interleukin-22 production from memory CD4
- Author
-
Cornelia, Voigt, Peter, May, Adrian, Gottschlich, Anamarija, Markota, Daniel, Wenk, Inga, Gerlach, Sebastian, Voigt, Georgios T, Stathopoulos, Kristina A M, Arendt, Constanze, Heise, Felicitas, Rataj, Klaus-Peter, Janssen, Melanie, Königshoff, Hauke, Winter, Isabelle, Himsl, Wolfgang E, Thasler, Max, Schnurr, Simon, Rothenfußer, Stefan, Endres, and Sebastian, Kobold
- Subjects
CD4-Positive T-Lymphocytes ,Mice, Inbred BALB C ,Lung Neoplasms ,Inflammasomes ,Interleukins ,Interleukin-1beta ,Breast Neoplasms ,Biological Sciences ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Mice ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Culture Media, Conditioned ,NLR Family, Pyrin Domain-Containing 3 Protein ,Leukocytes, Mononuclear ,Animals ,Humans ,Female ,Neoplasm Transplantation ,Cell Proliferation ,Signal Transduction - Abstract
IL-22 has been identified as a cancer-promoting cytokine, but its regulation in cancer tissue has not been addressed. Using both murine and human models, we demonstrate that cancer cells directly induce IL-22 production. We prove that interleukin-1β induced by inflammasome activation is critical for IL-22 production. IL-1β increased the activity of the IL-22 transcription factors in lineage-committed T cells. We show the existence of IL-22–producing Th1, Th17, and Th22 cells in tumor tissue of patients. Use of the clinically approved IL-1 receptor antagonist anakinra in vivo reduced IL-22 production and reduced tumor growth in a breast cancer model. These data provide the basis for therapeutic interventions, particularly using anakinra, aiming at limiting IL-22 production in patients with cancer.
- Published
- 2017
25. A novel modular platform for adoptive T cell therapy combining bispecific antibodies with synthetic agonistic receptors
- Author
-
Stefanie Lesch, C. Sustmann, Felicitas Rataj, M. Benmebarek, Bruno L. Cadilha, Ramona Murr, S. Kobold, Daniel Lamp, Moritz L. Schmidbauer, S. Endres, Christian Klein, C. Karches, Martina Geiger, Gerhard Niederfellner, J. vom Berg, Constanze Heise, M. Kurzay, Martin Jastroch, Peter Duewell, and R. Klaus
- Subjects
Cancer Research ,Bispecific antibody ,medicine.anatomical_structure ,Oncology ,Chemistry ,business.industry ,T cell ,Agonistic behaviour ,medicine ,Modular design ,business ,Receptor ,Cell biology - Published
- 2019
- Full Text
- View/download PDF
26. Characterization of bispecific antibodies that drive synthetic agonistic receptor - transduced T cells to mediate specific and conditional therapy in human pancreatic cancer models
- Author
-
Moritz L. Schmidbauer, S. Endres, Daniel Lamp, Martin Jastroch, Stefanie Lesch, C. Sustmann, Felicitas Rataj, Bruno L. Cadilha, M. Benmebarek, Gerhard Niederfellner, Christian Klein, Ramona Murr, R. Klaus, Constanze Heise, C. Karches, S. Kobold, J. vom Berg, Martina Geiger, and M. Kurzay
- Subjects
Cancer Research ,business.industry ,T cell ,CD28 ,Cancer ,Context (language use) ,medicine.disease ,Chimeric antigen receptor ,Cytokine release syndrome ,medicine.anatomical_structure ,Oncology ,Antigen ,Cancer research ,medicine ,Receptor ,business - Abstract
Adoptive T cell therapy, namely chimeric antigen receptor (CAR) T cell therapy has been a groundbreaking and effective treatment of relapsed or refractory haematological malignancies. Still, many patients do not respond or relapse with treatment-resistant disease. Additionally, toxicities such as cytokine release syndrome remain problematic. Cancer heterogeneity, beyond cancer types and inter-patient differences, is present within every individual patient. This heterogeneity, especially in the context of solid tumors, has meant that targeted immunotherapies have fared relatively poorly, creating a need for a modular platform with a capacity to target multiple antigens simultaneously and/or sequentially. Likewise, treatment-related toxicities have limited the therapeutic efficacy and breadth of patient selection. To tackle these caveats through a modular and controllable approach, we equipped T cells with synthetic agonistic receptors (SARs) that are only activated when a tumor-associated antigen and a cross-linking bispecific antibody (BiAb) specific for both SAR T cell and tumor cell are also present. The SAR itself is constituted of an inert extracellular domain in the form of EGFRvIII, that is fused to the T cell activating domains CD28 and CD3ζ. The BiAb employed is a trivalent CrossMab, with two binding arms (2 x Fab) for the tumor-associated antigen (mesothelin), and one binding arm for the SAR receptor (EGFRvIII). We showed that BiAb triggering of the SAR is conditional upon the binding of the second BiAb specificity. What is particularly advantageous with this approach is that T cell activation may only occur when the BiAb is present and in proximity to the antibody-targeted tumor cell. This conditional T cell activation is an inherent safety feature of the platform, whereby if unwanted levels of T cell activation are observed, depletion of the BiAb from the system could result in the reversal of said activation, thus managing the potential toxicity. This work was able to determine the validity and efficacy of the approach. Through the generation of several human pancreatic cancer models, and extensive in vitro and in vivo testing, the platform could be characterized. Its translational relevance and significance as a next-generation adoptive T cell therapy with the potential to plug some gaping pitfalls of current ACT approaches were also shown.
- Published
- 2019
- Full Text
- View/download PDF
27. Mesothelin-targeted bispecific antibodies drive synthetic agonistic receptor – Transduced T cells to mediate specific and conditional therapy of human pancreatic cancer models
- Author
-
Daniel Lamp, Ramona Murr, Martin Jastroch, Gerhard Niederfellner, Stefanie Lesch, Bruno L. Cadilha, C. Karches, Claudio Sustmann, Christian Klein, Martina Geiger, Felicitas Rataj, Moritz L. Schmidbauer, J. Keyl, Sebastian Kobold, M. Benmebarek, Constanze Heise, R. Klaus, M. Kurzay, J. vom Berg, and Stefan Endres
- Subjects
Cancer Research ,Bispecific antibody ,biology ,business.industry ,medicine.disease ,Oncology ,Pancreatic cancer ,Agonistic behaviour ,Cancer research ,biology.protein ,Medicine ,Mesothelin ,business ,Receptor - Published
- 2018
- Full Text
- View/download PDF
28. Cancer cells regulate Interleukin-22 production to promote tumor growth
- Author
-
C. Voigt, S. Endres, T. Ruzicka, Anamarija Markota, Harland S. Winter, K. Arendt, S. Kobold, I. Himsl, A. Kraechan, A. Gottschlich, Felicitas Rataj, G. Stathopoulos, P. May, M. Königshoff, W. Thasler, S. Rothenfußer, S. Voigt, I. Gerlach, D. Wenk, Klaus-Peter Janssen, Constanze Heise, Max Schnurr, and J. Suárez Gosálvez
- Subjects
Interleukin 22 ,Cancer Research ,Oncology ,Cancer cell ,Cancer research ,Tumor growth ,Biology - Published
- 2018
- Full Text
- View/download PDF
29. Proof of concept and mode of action of a novel modular platform for adoptive T cell therapy combining bispecific antibodies with synthetic agonistic receptors
- Author
-
Felicitas Rataj, Bruno L. Cadilha, Martina Geiger, Martin Jastroch, M. Benmebarek, Daniel Lamp, S. Kobold, C. Karches, Peter Duewell, Ramona Murr, Stefanie Lesch, Constanze Heise, Moritz L. Schmidbauer, S. Endres, C. Sustmann, Gerhard Niederfellner, Christian Klein, R. Klaus, J. vom Berg, and M. Kurzay
- Subjects
Cancer Research ,Bispecific antibody ,Computer science ,business.industry ,T cell ,Modular design ,medicine.anatomical_structure ,Oncology ,Proof of concept ,Agonistic behaviour ,medicine ,business ,Mode of action ,Receptor ,Neuroscience - Published
- 2018
- Full Text
- View/download PDF
30. Combined tumor-directed recruitment and protection from immune suppression enable CAR T cell efficacy in solid tumors
- Author
-
Bruno L. Cadilha, Klara Dorman, Marion Subklewe, C. Karches, Jasper N. Pruessmann, Stefanie Lesch, Moritz Thomas, Stefan Endres, Florian Märkl, Matthias Seifert, Sebastian Kobold, Carsten Marr, Jin Zhang, Duc Huynh, Mauro Di Pilato, Theo Lorenzini, Javier Suarez-Gosalvez, Mira Vänttinen, Dharmendra Pandey, Yi Zeng, Katrin Manske, S Stoiber, M. Benmebarek, Constanze Heise, Thorsten R. Mempel, A Öner, Hannah Obeck, Tobias Feuchtinger, and Adrian Gottschlich
- Subjects
T cell ,CCL1 ,CCR8 ,T-Lymphocytes, Regulatory ,03 medical and health sciences ,Chemokine receptor ,0302 clinical medicine ,Immune system ,Transforming Growth Factor beta ,Neoplasms ,medicine ,Humans ,Receptor ,Research Articles ,Cancer ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Chemistry ,Effector ,SciAdv r-articles ,ddc ,3. Good health ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Research Article ,Transforming growth factor - Abstract
The combination of directed migration and immunosuppressive shielding enables effective T cell therapy in solid tumors., CAR T cell therapy remains ineffective in solid tumors, due largely to poor infiltration and T cell suppression at the tumor site. T regulatory (Treg) cells suppress the immune response via inhibitory factors such as transforming growth factor–β (TGF-β). Treg cells expressing the C-C chemokine receptor 8 (CCR8) have been associated with poor prognosis in solid tumors. We postulated that CCR8 could be exploited to redirect effector T cells to the tumor site while a dominant-negative TGF-β receptor 2 (DNR) can simultaneously shield them from TGF-β. We identified that CCL1 from activated T cells potentiates a feedback loop for CCR8+ T cell recruitment to the tumor site. This sustained and improved infiltration of engineered T cells synergized with TGF-β shielding for improved therapeutic efficacy. Our results demonstrate that addition of CCR8 and DNR into CAR T cells can render them effective in solid tumors.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.