71 results on '"Ralph Mocikat"'
Search Results
2. Immune checkpoint blockade impairs immunosuppressive mechanisms of regulatory T cells in B-cell lymphoma
- Author
-
Vera Bauer, Fatima Ahmetlić, Nadine Hömberg, Albert Geishauser, Martin Röcken, and Ralph Mocikat
- Subjects
Foxp3 ,CTLA-4 ,PD-1 ,IL-10 ,MYC lymphoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
In malignant disease, CD4+Foxp3+ regulatory T cells (Tregs) hamper antitumor immune responses and may provide a target for immunotherapy. Although immune checkpoint blockade (ICB) has become an established therapy for several cancer entities including lymphoma, its mechanisms have not been entirely uncovered. Using endogenously arising λ-MYC-transgenic mouse B-cell lymphomas, which can effectively be suppressed by either Treg ablation or ICB, we investigated which mechanisms are used by Tregs to suppress antitumor responses and how ICB affects these pathways. During tumor development, Tregs up-regulated Foxp3, CD25, CTLA-4 and IL-10, which correlated with enhanced immunosuppressive functions. Thus, in contrast to other tumors, Tregs did not become dysfunctional despite chronic stimulation in the tumor microenvironment and progressive up-regulation of PD-1. Immunosuppression was mediated by direct contacts between Tregs and effector T cells and by IL-10. When λ-MYC mice were treated with ICB antibodies, Tregs revealed a less profound up-regulation of Foxp3, CD25 and IL-10 and a decreased suppressive capacity. This may be due to the shift towards a pro-inflammatory milieu fostered by ICB. In summary, an ICB-induced interference with Treg-dependent immunosuppression may contribute to the success of ICB.
- Published
- 2021
- Full Text
- View/download PDF
3. Therapy of lymphoma by immune checkpoint inhibitors: the role of T cells, NK cells and cytokine-induced tumor senescence
- Author
-
Vera Bauer, Tanja Riedel, Ralph Mocikat, Fatima Ahmetlic, Josia Fauser, Carolin Flessner, Nadine Hömberg, Roman Hennel, Ellen Brenner, and Kirsten Lauber
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Although antibodies blocking immune checkpoints have already been approved for clinical cancer treatment, the mechanisms involved are not yet completely elucidated. Here we used a λ-MYC transgenic model of endogenously growing B-cell lymphoma to analyze the requirements for effective therapy with immune checkpoint inhibitors.Methods Growth of spontaneous lymphoma was monitored in mice that received antibodies targeting programmed cell death protein 1 and cytotoxic T lymphocyte-associated protein-4, and the role of different immune cell compartments and cytokines was studied by in vivo depletion experiments. Activation of T and natural killer cells and the induction of tumor senescence were analyzed by flow cytometry.Results On immune checkpoint blockade, visible lymphomas developed at later time points than in untreated controls, indicating an enhanced tumor control. Importantly, 20% to 30% of mice were even long-term protected and did never develop clinical signs of tumor growth. The therapeutic effect was dependent on cytokine-induced senescence in malignant B cells. The proinflammatory cytokines interferon-γ (IFN-γ) and tumor necrosis factor (TNF) were necessary for the survival benefit as well as for senescence induction in the λ-MYC model. Antibody therapy improved T-cell functions such as cytokine production, and long-time survivors were only observed in the presence of T cells. Yet, NK cells also had a pronounced effect on therapy-induced delay of tumor growth. Antibody treatment enhanced numbers, proliferation and IFN-γ expression of NK cells in developing tumors. The therapeutic effect was fully abrogated only after depletion of both, T cells and NK cells, or after ablation of either IFN-γ or TNF.Conclusions Tumor cell senescence may explain why patients responding to immune checkpoint blockade frequently show stable growth arrest of tumors rather than complete tumor regression. In the lymphoma model studied, successful therapy required both, tumor-directed T-cell responses and NK cells, which control, at least partly, tumor development through cytokine-induced tumor senescence.
- Published
- 2021
- Full Text
- View/download PDF
4. 837 Interleukin-10 drives the development of T regulatory type 1 (Tr1) cells and is a target for immunotherapy
- Author
-
Yanchun Ma, Vera Bauer, Tanja Riedel, Thomas Hofer, Martin Roecken, and Ralph Mocikat
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
5. Potential of the Trifunctional Bispecific Antibody Surek Depends on Dendritic Cells: Rationale for a New Approach of Tumor Immunotherapy
- Author
-
Nina Eissler, Josef Mysliwietz, Nina Deppisch, Peter Ruf, Horst Lindhofer, and Ralph Mocikat
- Subjects
Trifunctional Bispecific Antibody ,FACS Staining ,Induce Tumor Cell Killing ,Catumaxomab ,Kill Tumor Cells ,Therapeutics. Pharmacology ,RM1-950 ,Biochemistry ,QD415-436 - Abstract
Abstract Trifunctional bispecific antibodies (trAbs) used in tumor immunotherapy have the unique ability to recruit T cells toward antigens on the tumor cell surface and, moreover, to activate accessory cells through their immunoglobulin Fc region interacting with activating Fcγ receptors. This scenario gives rise to additional costimulatory signals required for T cell-mediated tumor cell destruction and induction of an immunologic memory. Here we show in an in vitro system that most effective trAb-dependent T-cell activation and tumor cell elimination are achieved in the presence of dendritic cells (DCs). On the basis of these findings, we devise a novel approach of cancer immunotherapy that combines the specific advantages of trAbs with those of DC-based vaccination. Simultaneous delivery of trAbs and in vitro differentiated DCs resulted in a markedly improved tumor rejection in a murine melanoma model compared with monotherapy.
- Published
- 2013
- Full Text
- View/download PDF
6. A novel CXCL10-based GPI-anchored fusion protein as adjuvant in NK-based tumor therapy.
- Author
-
Niklas Muenchmeier, Sophia Boecker, Lorenz Bankel, Laura Hinz, Nicole Rieth, Constantin Lapa, Anna N Mendler, Elfriede Noessner, Ralph Mocikat, and Peter J Nelson
- Subjects
Medicine ,Science - Abstract
BACKGROUND: Cellular therapy is a promising therapeutic strategy for malignant diseases. The efficacy of this therapy can be limited by poor infiltration of the tumor by immune effector cells. In particular, NK cell infiltration is often reduced relative to T cells. A novel class of fusion proteins was designed to enhance the recruitment of specific leukocyte subsets based on their expression of a given chemokine receptor. The proteins are composed of an N-terminal chemokine head, the mucin domain taken from the membrane-anchored chemokine CX3CL1, and a C-terminal glycosylphosphatidylinositol (GPI) membrane anchor replacing the normal transmembrane domain allowing integration of the proteins into cell membranes when injected into a solid tumor. The mucin domain in conjunction with the chemokine head acts to specifically recruit leukocytes expressing the corresponding chemokine receptor. METHODOLOGY/PRINCIPAL FINDINGS: A fusion protein comprising a CXCL10 chemokine head (CXCL10-mucin-GPI) was used for proof of concept for this approach and expressed constitutively in Chinese Hamster Ovary cells. FPLC was used to purify proteins. The recombinant proteins efficiently integrated into cell membranes in a process dependent upon the GPI anchor and were able to activate the CXCR3 receptor on lymphocytes. Endothelial cells incubated with CXCL10-mucin-GPI efficiently recruited NK cells in vitro under conditions of physiologic flow, which was shown to be dependent on the presence of the mucin domain. Experiments conducted in vivo using established tumors in mice suggested a positive effect of CXCL10-mucin-GPI on the recruitment of NK cells. CONCLUSIONS: The results suggest enhanced recruitment of NK cells by CXCL10-mucin-GPI. This class of fusion proteins represents a novel adjuvant in cellular immunotherapy. The underlying concept of a chemokine head fused to the mucin domain and a GPI anchor signal sequence may be expanded into a broader family of reagents that will allow targeted recruitment of cells in various settings.
- Published
- 2013
- Full Text
- View/download PDF
7. EBV-gp350 confers B-cell tropism to tailored exosomes and is a neo-antigen in normal and malignant B cells--a new option for the treatment of B-CLL.
- Author
-
Romana Ruiss, Simon Jochum, Ralph Mocikat, Wolfgang Hammerschmidt, and Reinhard Zeidler
- Subjects
Medicine ,Science - Abstract
gp350, the major envelope protein of Epstein-Barr-Virus, confers B-cell tropism to the virus by interacting with the B lineage marker CD21. Here we utilize gp350 to generate tailored exosomes with an identical tropism. These exosomes can be used for the targeted co-transfer of functional proteins to normal and malignant human B cells. We demonstrate here the co-transfer of functional CD154 protein on tailored gp350+ exosomes to malignant B blasts from patients with B chronic lymphocytic leukemia (B-CLL), rendering B blasts immunogenic to tumor-reactive autologous T cells. Intriguingly, engulfment of gp350+ exosomes by B-CLL cells and presentation of gp350-derived peptides also re-stimulated EBV-specific T cells and redirected the strong antiviral cellular immune response in patients to leukemic B cells. In essence, we show that gp350 alone confers B-cell tropism to exosomes and that these exosomes can be further engineered to simultaneously trigger virus- and tumor-specific immune responses. The simultaneous exploitation of gp350 as a tropism molecule for tailored exosomes and as a neo-antigen in malignant B cells provides a novel attractive strategy for immunotherapy of B-CLL and other B-cell malignancies.
- Published
- 2011
- Full Text
- View/download PDF
8. Data from Efficacy and Tolerability of a GD2-Directed Trifunctional Bispecific Antibody in a Preclinical Model: Subcutaneous Administration Is Superior to Intravenous Delivery
- Author
-
Ralph Mocikat, Horst Lindhofer, Raymund Buhmann, Frauke Neff, Nina Eissler, Peter Ruf, and Nina Deppisch
- Abstract
Trifunctional bispecific antibodies (trAb) are novel anticancer drugs that recruit and activate different types of immune effector cells at the targeted tumor. Thus, tumor cells are effectively eliminated and a long-lasting tumor-specific T-cell memory is induced. The trAb Ektomab is directed against human CD3 on T cells and the tumor-associated ganglioside GD2, which is an attractive target for immunotherapy of melanoma in humans. To optimize clinical applicability, we studied different application routes with respect to therapeutic efficacy and tolerability by using the surrogate trAb Surek (anti-GD2 × anti-murine CD3) and a murine melanoma engineered to express GD2. We show that subcutaneous injection of the trAb is superior to the intravenous delivery pathway, which is the standard application route for therapeutic antibodies. Despite lower plasma levels after subcutaneous administration, the same tumor-protective potential was observed in vivo compared with intravenous administration of Surek. However, subcutaneously delivered Surek showed better tolerability. This could be explained by a continuous release of the antibody leading to constant plasma levels and a delayed induction of proinflammatory cytokines. Importantly, the induction of counter-regulatory mechanisms was reduced after subcutaneous application. These findings are relevant for the clinical application of trifunctional bispecific antibodies and, possibly, also other immunoglobulin constructs. Mol Cancer Ther; 14(8); 1877–83. ©2015 AACR.
- Published
- 2023
9. Supplementary Figures S1-S4 from Efficacy and Tolerability of a GD2-Directed Trifunctional Bispecific Antibody in a Preclinical Model: Subcutaneous Administration Is Superior to Intravenous Delivery
- Author
-
Ralph Mocikat, Horst Lindhofer, Raymund Buhmann, Frauke Neff, Nina Eissler, Peter Ruf, and Nina Deppisch
- Abstract
Supplementary Figures S1-S4. Supplementary Figure S1. Development of anti-antibodies following Surek treatment; Supplementary Figure S2. Activation and proliferation of T cells after treatment with trAb Surek alone; Supplementary Figure S3. Weight loss of mice after unspecific immune activation induced by CD3 binding of Surek; Supplementary Figure S4. Cytokine milieu induced by Surek s.c.
- Published
- 2023
10. Data from Regulatory T Cells in an Endogenous Mouse Lymphoma Recognize Specific Antigen Peptides and Contribute to Immune Escape
- Author
-
Ralph Mocikat, Martin Röcken, Stefan Stevanović, Tim Sparwasser, Albert Geishauser, Nico Trautwein, Vera Bauer, Nadine Hömberg, Tanja Riedel, and Fatima Ahmetlić
- Abstract
Foxp3+ regulatory T cells (Tregs) sustain immune homeostasis and may contribute to immune escape in malignant disease. As a prerequisite for developing immunologic approaches in cancer therapy, it is necessary to understand the ontogeny and the antigenic specificities of tumor-infiltrating Tregs. We addressed this question by using a λ-MYC transgenic mouse model of endogenously arising B-cell lymphoma, which mirrors key features of human Burkitt lymphoma. We show that Foxp3+ Tregs suppress antitumor responses in endogenous lymphoma. Ablation of Foxp3+ Tregs significantly delayed tumor development. The ratio of Treg to effector T cells was elevated in growing tumors, which could be ascribed to differential proliferation. The Tregs detected were mainly natural Tregs that apparently recognized self-antigens. We identified MHC class II–restricted nonmutated self-epitopes, which were more prevalent in lymphoma than in normal B cells and could be recognized by Tregs. These epitopes were derived from proteins that are associated with cellular processes related to malignancy and may be overexpressed in the tumor.
- Published
- 2023
11. Supplementary Data from Regulatory T Cells in an Endogenous Mouse Lymphoma Recognize Specific Antigen Peptides and Contribute to Immune Escape
- Author
-
Ralph Mocikat, Martin Röcken, Stefan Stevanović, Tim Sparwasser, Albert Geishauser, Nico Trautwein, Vera Bauer, Nadine Hömberg, Tanja Riedel, and Fatima Ahmetlić
- Abstract
Supplementary figures and table
- Published
- 2023
12. Bifunctional PD-1 × αCD3 × αCD33 fusion protein reverses adaptive immune escape in acute myeloid leukemia
- Author
-
Marion Subklewe, Bettina Brauchle, Christina Krupka, Nadja C. Fenn, Ana Ogrinc Wagner, Felicitas Rataj, Katrin Deiser, Sebastian Kobold, Karsten Spiekermann, Monika Herrmann, Anetta Marcinek, Klaus H. Metzeler, Ralph Mocikat, and Karl-Peter Hopfner
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,Myeloid ,CD3 Complex ,Recombinant Fusion Proteins ,Programmed Cell Death 1 Receptor ,Sialic Acid Binding Ig-like Lectin 3 ,Immunology ,CD33 ,Adaptive Immunity ,Biochemistry ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Mice, Inbred NOD ,Cell Line, Tumor ,medicine ,Animals ,Humans ,biology ,Chemistry ,Myeloid leukemia ,Cell Biology ,Hematology ,medicine.disease ,Xenograft Model Antitumor Assays ,Immune checkpoint ,Neoplasm Proteins ,Leukemia, Myeloid, Acute ,Cytolysis ,Leukemia ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Antibody ,Single-Chain Antibodies - Abstract
The CD33-targeting bispecific T-cell engager (BiTE) AMG 330 proved to be highly efficient in mediating cytolysis of acute myeloid leukemia (AML) cells in vitro and in mouse models. Yet, T-cell activation is correlated with upregulation of programmed cell death-ligand 1 (PD-L1) and other inhibitory checkpoints on AML cells that confer adaptive immune resistance. PD-1 and PD-L1 blocking agents may counteract T-cell dysfunction, however, at the expense of broadly distributed immune-related adverse events (irAEs). We developed a bifunctional checkpoint inhibitory T cell–engaging (CiTE) antibody that combines T-cell redirection to CD33 on AML cells with locally restricted immune checkpoint blockade. This is accomplished by fusing the extracellular domain of PD-1 (PD-1ex), which naturally holds a low affinity to PD-L1, to an αCD3.αCD33 BiTE-like scaffold. By a synergistic effect of checkpoint blockade and avidity-dependent binding, the PD-1ex attachment increases T-cell activation (3.3-fold elevation of interferon-γ) and leads to efficient and highly selective cytotoxicity against CD33+PD-L1+ cell lines (50% effective concentration = 2.3-26.9 pM) as well as patient-derived AML cells (n = 8). In a murine xenograft model, the CiTE induces complete AML eradication without initial signs of irAEs as measured by body weight loss. We conclude that our molecule preferentially targets AML cells, whereas high-affinity blockers, such as clinically approved anticancer agents, also address PD-L1+ non-AML cells. By combining the high efficacy of T-cell engagers with immune checkpoint blockade in a single molecule, we expect to minimize irAEs associated with the systemic application of immune checkpoint inhibitors and suggest high therapeutic potential, particularly for patients with relapsed/ refractory AML.
- Published
- 2018
13. Immune checkpoint blockade impairs immunosuppressive mechanisms of regulatory T cells in B-cell lymphoma
- Author
-
Martin Röcken, Nadine Hömberg, Fatima Ahmetlić, Albert Geishauser, Vera Bauer, and Ralph Mocikat
- Subjects
Cancer Research ,medicine.medical_treatment ,chemical and pharmacologic phenomena ,MYC lymphoma ,Immune system ,hemic and lymphatic diseases ,PD-1 ,Medicine ,IL-2 receptor ,RC254-282 ,Original Research ,Tumor microenvironment ,business.industry ,FOXP3 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunosuppression ,hemic and immune systems ,Immunotherapy ,Immune checkpoint ,Oncology ,CTLA-4 ,Ctla-4 ,Foxp3 ,Il-10 ,Myc Lymphoma ,Pd-1 ,IL-10 ,Cancer research ,business - Abstract
Highlights • During lymphoma growth, Tregs evolve an increasingly suppressive phenotype. • Lymphoma-infiltrating Tregs show an enhanced immunosuppressive function. • Cell contacts and IL-10 are required for Treg-mediated immunosuppression. • Alterations of intratumoral Tregs are partly abrogated by immune checkpoint blockade., In malignant disease, CD4+Foxp3+ regulatory T cells (Tregs) hamper antitumor immune responses and may provide a target for immunotherapy. Although immune checkpoint blockade (ICB) has become an established therapy for several cancer entities including lymphoma, its mechanisms have not been entirely uncovered. Using endogenously arising λ-MYC-transgenic mouse B-cell lymphomas, which can effectively be suppressed by either Treg ablation or ICB, we investigated which mechanisms are used by Tregs to suppress antitumor responses and how ICB affects these pathways. During tumor development, Tregs up-regulated Foxp3, CD25, CTLA-4 and IL-10, which correlated with enhanced immunosuppressive functions. Thus, in contrast to other tumors, Tregs did not become dysfunctional despite chronic stimulation in the tumor microenvironment and progressive up-regulation of PD-1. Immunosuppression was mediated by direct contacts between Tregs and effector T cells and by IL-10. When λ-MYC mice were treated with ICB antibodies, Tregs revealed a less profound up-regulation of Foxp3, CD25 and IL-10 and a decreased suppressive capacity. This may be due to the shift towards a pro-inflammatory milieu fostered by ICB. In summary, an ICB-induced interference with Treg-dependent immunosuppression may contribute to the success of ICB.
- Published
- 2021
14. Interleukin-10 counteracts T-helper type 1 responses in B-cell lymphoma and is a target for tumor immunotherapy
- Author
-
Ralph Mocikat, Nadine Hömberg, Tanja Riedel, Fatima Ahmetlić, Martin Röcken, Vera Bauer, Thomas Höfer, and Yanchun Ma
- Subjects
0301 basic medicine ,Cancer Research ,Lymphoma, B-Cell ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Biology ,T-Lymphocytes, Regulatory ,03 medical and health sciences ,Interferon-gamma ,Mice ,0302 clinical medicine ,Immune system ,Downregulation and upregulation ,medicine ,Tumor Microenvironment ,Animals ,Humans ,B-cell lymphoma ,Il-10 ,Lymphoma ,Th1 Cells ,Tr1 Cells ,Tumor Escape ,C-myc ,Cancer ,Cell Differentiation ,Immunotherapy ,Dendritic Cells ,medicine.disease ,Interleukin-10 ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Interleukin 10 ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research - Abstract
To establish strategies for immunotherapy of B-cell lymphoma, it is mandatory to gain deeper insights into the mechanisms of tumor immune escape. In a mouse model of endogenously arising lymphoma, we investigated the impact of IL-10 on the regulation of antitumor responses. Despite progressive functional impairment of NK cells and lack of IFN-γ in the tumor milieu, we found an augmented fraction of T helper type 1 (Th1) cells, which continued to express IFN-γ but also upregulated IL-10 during disease development. Using a lymphoma microenvironment in vitro, we showed that Th1 cells were converted to Foxp3-negative T regulatory type 1 (Tr1) cells, which coexpressed IFN-γ and IL-10 and upregulated PD-1. This differentiation required pre-existing IL-10, which was primarily provided by malignant B cells and dendritic cells. IFN-γ only declined in cells with the uppermost PD-1 levels. Importantly, antibody-mediated IL-10 ablation in vivo improved effector cell functions and significantly suppressed tumor development. While the contribution of IL-10 to cancer immune escape has been controversially discussed in the past, we show that IL-10 suppresses ongoing, potentially protective immune responses in lymphoma and might be a target for immunotherapy.
- Published
- 2021
15. The role of dendritic cells for therapy of B-cell lymphoma with immune checkpoint inhibitors
- Author
-
Martin Röcken, Ralph Mocikat, Vera Bauer, Tanja Riedel, Anne Scheuerpflug, and Fatima Ahmetlić
- Subjects
0301 basic medicine ,Cancer Research ,Lymphoma, B-Cell ,Lymphoma ,Immune checkpoint inhibitors ,Immunology ,Programmed Cell Death 1 Receptor ,Genes, myc ,Mice, Transgenic ,Immune Checkpoint Blocking ,Tumor-infiltrating Dendritic Cells ,Interferon-γ ,λ ,myc Mouse ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,λ-MYC mouse ,medicine ,Immunology and Allergy ,Animals ,Humans ,CTLA-4 Antigen ,Tumor-infiltrating dendritic cells ,B-cell lymphoma ,Immune Checkpoint Inhibitors ,Cells, Cultured ,Mice, Inbred BALB C ,biology ,Chemistry ,Dendritic Cells ,medicine.disease ,In vitro ,Immune checkpoint ,Cancer treatment ,Mice, Inbred C57BL ,Disease Models, Animal ,Immune checkpoint blocking ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Drug Therapy, Combination ,Original Article ,Antibody - Abstract
Immune checkpoint blocking (ICB) is a promising new tool of cancer treatment. Yet, the underlying therapeutic mechanisms are not fully understood. Here we investigated the role of dendritic cells (DCs) for the therapeutic effect of ICB in a λ-MYC-transgenic mouse model of endogenously arising B-cell lymphoma. The growth of these tumors can be effectively delayed by antibodies against CTLA-4 and PD-1. Tumor-infiltrating DCs from mice having received therapy showed an upregulation of costimulatory molecules as well as an augmented IL-12/IL-10 ratio as compared to untreated controls. Both alterations seemed to be induced by interferon-γ (IFN-γ), which is upregulated in T cells and natural killer cells upon ICB. Furthermore, the enhanced IL-12/IL-10 ratio, which favors Th1-prone antitumor T-cell responses, was a consequence of direct interaction of ICB antibodies with DCs. Importantly, the capability of tumor-infiltrating DCs of stimulating peptide-specific or allogeneic T-cell responses in vitro was improved when DCs were derived from ICB-treated mice. The data indicate that ICB therapy is not only effective by directly activating T cells, but also by triggering a complex network, in which DCs play a pivotal role at the interface between innate and adaptive antitumor responses. Electronic supplementary material The online version of this article (10.1007/s00262-020-02767-6) contains supplementary material, which is available to authorized users.
- Published
- 2020
16. Cancer immune control needs senescence induction by interferon-dependent cell cycle regulator pathways in tumours
- Author
-
Albert Geishauser, Andreas Beilhack, Tanja Riedel, Bernd J. Pichler, Jürgen Bauer, Nadine Simon, Fatima Ahmetlić, Heidi Braumüller, Saskia Biskup, Nadine Hömberg, Martin Schaller, Dennis Döcker, Christopher Schroeder, Lars Zender, Andrea Forschner, Franz J. Hilke, Manfred Kneilling, Thomas Eigentler, Katharina Böhm, Ellen Brenner, Dirk Schadendorf, Birgit Fehrenbacher, Stefan Zwirner, B.F. Schörg, German Demidov, Martin Eichner, Thomas Wieder, Heike Niessner, Leticia Quintanilla-Martinez, Tobias Sinnberg, Ralph Mocikat, Martin Röcken, Dominik Sonanini, Daniel Dauch, and Katja J. Jarick
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,0301 basic medicine ,Science ,medicine.medical_treatment ,Medizin ,General Physics and Astronomy ,Cancer immunotherapy ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigens, Neoplasm ,CDKN2A ,Cell Line, Tumor ,medicine ,Animals ,Humans ,RNA, Messenger ,lcsh:Science ,Tumour-suppressor proteins ,Melanoma ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,Multidisciplinary ,Cell Cycle ,Cancer ,General Chemistry ,Immunotherapy ,Cell cycle ,medicine.disease ,Survival Analysis ,Immune checkpoint ,Tumor Burden ,Mice, Inbred C57BL ,Ki-67 Antigen ,STAT1 Transcription Factor ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,lcsh:Q ,Interferons ,Lymph Nodes - Abstract
Immune checkpoint blockade (ICB)-based or natural cancer immune responses largely eliminate tumours. Yet, they require additional mechanisms to arrest those cancer cells that are not rejected. Cytokine-induced senescence (CIS) can stably arrest cancer cells, suggesting that interferon-dependent induction of senescence-inducing cell cycle regulators is needed to control those cancer cells that escape from killing. Here we report in two different cancers sensitive to T cell-mediated rejection, that deletion of the senescence-inducing cell cycle regulators p16Ink4a/p19Arf (Cdkn2a) or p21Cip1 (Cdkn1a) in the tumour cells abrogates both the natural and the ICB-induced cancer immune control. Also in humans, melanoma metastases that progressed rapidly during ICB have losses of senescence-inducing genes and amplifications of senescence inhibitors. Metastatic cells also resist CIS. Such genetic and functional alterations are infrequent in metastatic melanomas regressing during ICB. Thus, activation of tumour-intrinsic, senescence-inducing cell cycle regulators is required to stably arrest cancer cells that escape from eradication., The growth of cancer cells can be stably arrested by cytokine-induced senescence. Here, the authors show that cancers with defects in senescence-inducing cell cycle regulator pathways are resistant to immune checkpoint blockade.
- Published
- 2020
17. Internationalisierung und Wissenschaftssprache – eine Einführung
- Author
-
Ralph Mocikat
- Published
- 2020
18. Die Anglophonisierung der Wissenschaften als Folge ihrer Ökonomisierung?
- Author
-
Ralph Mocikat
- Published
- 2020
19. Reversal of tumor acidosis by systemic buffering reactivates NK cells to express IFN-γ and induces NK cell-dependent lymphoma control without other immunotherapies
- Author
-
Christoph D. Brenner, Ralph Mocikat, Johann Pötzl, Martin Röcken, Albert Geishauser, David Roser, Nadine Hömberg, Michael Weigand, and Lorenz Bankel
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,Chemistry ,medicine.medical_treatment ,Cell ,Immunotherapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Lymphatic system ,Immune system ,Oncology ,Downregulation and upregulation ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Interferon gamma ,Cytotoxicity ,medicine.drug - Abstract
Like other immune cells, natural killer (NK) cells show impaired effector functions in the microenvironment of tumors, but little is known on the underlying mechanisms. Since lactate acidosis, a hallmark of malignant tissue, was shown to contribute to suppression of effective antitumor immune responses, we investigated the impact of tissue pH and lactate concentration on NK-cell functions in an aggressive model of endogenously arising B-cell lymphoma. The progressive loss of IFN-γ production by NK cells observed during development of this disease could be ascribed to decreased pH values and lactate accumulation in the microenvironment of growing tumors. Interestingly, IFN-γ expression by lymphoma-derived NK cells could be restored by transfer of these cells into a normal micromilieu. Likewise, systemic alkalization by oral delivery of bicarbonate to lymphoma-developing mice was capable of enhancing IFN-γ expression in NK cells and increasing the NK-cell numbers in the lymphoid organs where tumors were growing. By contrast, NK-cell cytotoxicity was dampened in vivo by tumor-dependent mechanisms that seemed to be different from lactate acidosis and could not be restored in a normal milieu. Most importantly, alkalization and the concomitant IFN-γ upregulation in NK cells were sufficient to significantly delay tumor growth without any other immunotherapy. This effect was strictly dependent on NK cells.
- Published
- 2017
20. Therapy of lymphoma by immune checkpoint inhibitors: the role of T cells, NK cells and cytokine-induced tumor senescence
- Author
-
Carolin Flessner, Ellen Brenner, Tanja Riedel, Roman Hennel, Vera Bauer, Nadine Hömberg, Fatima Ahmetlić, Josia Fauser, Ralph Mocikat, Martin Röcken, and Kirsten Lauber
- Subjects
CTLA-4 antigen ,Cancer Research ,Lymphoma ,T-Lymphocytes ,medicine.medical_treatment ,Immunology ,Antibodies, Monoclonal, Humanized ,programmed cell death 1 receptor ,Interferon-gamma ,Mice ,Immune system ,Interferon ,medicine ,Animals ,Humans ,tumor microenvironment ,Ctla-4 Antigen ,Hematologic Neoplasms ,Immunotherapy ,Programmed Cell Death 1 Receptor ,Tumor Microenvironment ,Immunology and Allergy ,Cytotoxic T cell ,hematologic neoplasms ,Immune Checkpoint Inhibitors ,Cellular Senescence ,RC254-282 ,Cell Proliferation ,Pharmacology ,Tumor microenvironment ,Tumor Necrosis Factor-alpha ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Basic Tumor Immunology ,Xenograft Model Antitumor Assays ,Immune checkpoint ,Killer Cells, Natural ,Nivolumab ,Treatment Outcome ,Cytokine ,Oncology ,Cancer research ,Cytokines ,Molecular Medicine ,Tumor necrosis factor alpha ,immunotherapy ,business ,medicine.drug - Abstract
BackgroundAlthough antibodies blocking immune checkpoints have already been approved for clinical cancer treatment, the mechanisms involved are not yet completely elucidated. Here we used a λ-MYC transgenic model of endogenously growing B-cell lymphoma to analyze the requirements for effective therapy with immune checkpoint inhibitors.MethodsGrowth of spontaneous lymphoma was monitored in mice that received antibodies targeting programmed cell death protein 1 and cytotoxic T lymphocyte-associated protein-4, and the role of different immune cell compartments and cytokines was studied by in vivo depletion experiments. Activation of T and natural killer cells and the induction of tumor senescence were analyzed by flow cytometry.ResultsOn immune checkpoint blockade, visible lymphomas developed at later time points than in untreated controls, indicating an enhanced tumor control. Importantly, 20% to 30% of mice were even long-term protected and did never develop clinical signs of tumor growth. The therapeutic effect was dependent on cytokine-induced senescence in malignant B cells. The proinflammatory cytokines interferon-γ (IFN-γ) and tumor necrosis factor (TNF) were necessary for the survival benefit as well as for senescence induction in the λ-MYC model. Antibody therapy improved T-cell functions such as cytokine production, and long-time survivors were only observed in the presence of T cells. Yet, NK cells also had a pronounced effect on therapy-induced delay of tumor growth. Antibody treatment enhanced numbers, proliferation and IFN-γ expression of NK cells in developing tumors. The therapeutic effect was fully abrogated only after depletion of both, T cells and NK cells, or after ablation of either IFN-γ or TNF.ConclusionsTumor cell senescence may explain why patients responding to immune checkpoint blockade frequently show stable growth arrest of tumors rather than complete tumor regression. In the lymphoma model studied, successful therapy required both, tumor-directed T-cell responses and NK cells, which control, at least partly, tumor development through cytokine-induced tumor senescence.
- Published
- 2021
21. Der Wert der Mehrsprachigkeit in den Naturwissenschaften und der Medizin
- Author
-
Ralph Mocikat
- Published
- 2018
22. Efficacy and Tolerability of a GD2-Directed Trifunctional Bispecific Antibody in a Preclinical Model: Subcutaneous Administration Is Superior to Intravenous Delivery
- Author
-
Nina Eissler, Ralph Mocikat, Raymund Buhmann, Horst Lindhofer, Peter Ruf, Frauke Neff, and Nina Deppisch
- Subjects
Cancer Research ,Injections, Subcutaneous ,T-Lymphocytes ,medicine.medical_treatment ,Melanoma, Experimental ,Biological Availability ,Antineoplastic Agents ,Trab ,Pharmacology ,Lymphocyte Activation ,Proinflammatory cytokine ,Mice ,Subcutaneous injection ,In vivo ,Cell Line, Tumor ,Gangliosides ,Antibodies, Bispecific ,Animals ,Medicine ,biology ,business.industry ,Melanoma ,Immunotherapy ,medicine.disease ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,Oncology ,Tolerability ,biology.protein ,Cytokines ,Administration, Intravenous ,Female ,Antibody ,business - Abstract
Trifunctional bispecific antibodies (trAb) are novel anticancer drugs that recruit and activate different types of immune effector cells at the targeted tumor. Thus, tumor cells are effectively eliminated and a long-lasting tumor-specific T-cell memory is induced. The trAb Ektomab is directed against human CD3 on T cells and the tumor-associated ganglioside GD2, which is an attractive target for immunotherapy of melanoma in humans. To optimize clinical applicability, we studied different application routes with respect to therapeutic efficacy and tolerability by using the surrogate trAb Surek (anti-GD2 × anti-murine CD3) and a murine melanoma engineered to express GD2. We show that subcutaneous injection of the trAb is superior to the intravenous delivery pathway, which is the standard application route for therapeutic antibodies. Despite lower plasma levels after subcutaneous administration, the same tumor-protective potential was observed in vivo compared with intravenous administration of Surek. However, subcutaneously delivered Surek showed better tolerability. This could be explained by a continuous release of the antibody leading to constant plasma levels and a delayed induction of proinflammatory cytokines. Importantly, the induction of counter-regulatory mechanisms was reduced after subcutaneous application. These findings are relevant for the clinical application of trifunctional bispecific antibodies and, possibly, also other immunoglobulin constructs. Mol Cancer Ther; 14(8); 1877–83. ©2015 AACR.
- Published
- 2015
23. Critical role of the NKG2D receptor for NK cell-mediated control and immune escape of B-cell lymphoma
- Author
-
Dirk H. Busch, Andrew Flatley, Lena Belting, Ralph Mocikat, Margarethe Przewoznik, Bojan Polić, Martin Röcken, Christoph D. Brenner, Nadine Hömberg, and Tanja Riedel
- Subjects
Immunology ,hemic and immune systems ,chemical and pharmacologic phenomena ,Biology ,medicine.disease ,NKG2D ,biological factors ,Lymphoma ,Immunosurveillance ,Interleukin 21 ,NK-92 ,Tumor Escape ,medicine ,Interleukin 12 ,Cancer research ,Immunology and Allergy ,B-cell lymphoma - Abstract
Little is known on the control of lymphomas by NK cells. Here, we study the role of the NK group 2D (NKG2D) receptor for the immunosurveillance of lymphoma. By using transplantable tumors as well as a λ-myc-transgenic model of endogenously arising lymphoma and NKG2D-deficient mice, we show that NK cells eliminate tumor cells in vivo after receiving two signals. One step involved the activation of NK cells giving rise to IFN-γ expression, which was effected by MHCI(low) tumor cells or DCs. However, this was necessary but not sufficient to mediate cytotoxicity. Triggering cytotoxicity additionally required a second step, which could be mediated by engagement of the NKG2D receptor. Thus, NKG2D-deficient NK cells could become activated in vivo, but they were not able to reject transplanted lymphomas or to degranulate in animals bearing autochthonous lymphomas. Tumor growth in NKG2D-deficient λ-myc-transgenic mice was significantly accelerated compared to NKG2D-competent animals. Whereas the latter developed tumors that lost expression of NKG2D ligands (NKG2D-L) in late disease stages, this did not occur in NKG2D-deficient mice. This indicates that NK cells and the NKG2D receptor play a role for control of lymphomas and that selection for NKG2D-L loss mutants provides a mechanism of tumor escape.
- Published
- 2015
24. Alterations of costimulatory molecules and instructive cytokines expressed by dendritic cells in the microenvironment of an endogenous mouse lymphoma
- Author
-
Jakob Weiß, Ralph Mocikat, Martin Röcken, Margarethe Przewoznik, Elfriede Noessner, Marcella Naujoks, Tanja Riedel, and Nadine Hömberg
- Subjects
Cancer Research ,BALB 3T3 Cells ,Lymphoma, B-Cell ,medicine.medical_treatment ,T cell ,Immunology ,Genes, myc ,Mice, Transgenic ,Biology ,Flow cytometry ,Mice ,Immune system ,Downregulation and upregulation ,Tumor Microenvironment ,medicine ,Animals ,Immunology and Allergy ,B-cell lymphoma ,Tumor microenvironment ,medicine.diagnostic_test ,Dendritic Cells ,Oncogenes ,Flow Cytometry ,medicine.disease ,Lymphoma ,Killer Cells, Natural ,Mice, Inbred C57BL ,Cytokine ,medicine.anatomical_structure ,Oncology ,Cancer research ,Cytokines - Abstract
Costimulatory surface molecules and instructive cytokines expressed by dendritic cells (DCs) determine the outcome of an immune response. In malignant disease, DCs are often functionally compromised. In most tumors studied so far, the deficient induction of effective T cell responses has been associated with a blockade of DC maturation, but little has been known on DCs infiltrating malignant B cell lymphoma. Here, we investigated for the first time the phenotypic and functional status of DCs in B cell lymphoma, and we analyzed the network of DCs, tumor cells, natural killer (NK) cells and cytokines present in the tumor micromilieu. Therefor, we used an endogenous myc-transgenic mouse lymphoma model, because transplanted tumor cells foster an IFN-γ-driven Th1 antitumor response rather than an immunosuppressive environment, which is observed in autochthonous neoplasias. Lymphoma-infiltrating DCs showed a mature phenotype and a Th2-inducing cytokine pattern. This situation is in contrast to most human malignancies and mouse models described. Cellular contacts between DCs and tumor cells, which involved CD62L on the lymphoma, caused upregulation of costimulatory molecules, whereas IL-10 primarily derived from lymphoma cells induced an IL-12/IL-10 shift in DCs. Thus, alteration of costimulatory molecules and instructive cytokines was mediated by distinct mechanisms. Normal NK cells were able to additionally modulate DC maturation but this effect was absent in the lymphoma environment where IFN-γ production by NK cells was severely impaired. These data are relevant for establishing novel immunotherapeutic approaches against B cell lymphoma.
- Published
- 2014
25. Potential of the Trifunctional Bispecific Antibody Surek Depends on Dendritic Cells: Rationale for a New Approach of Tumor Immunotherapy
- Author
-
Josef Mysliwietz, Horst Lindhofer, Ralph Mocikat, Peter Ruf, Nina Deppisch, and Nina Eissler
- Subjects
T-Lymphocytes ,medicine.medical_treatment ,Immunoglobulin Fc ,Biology ,Mice ,Antigen ,Cancer immunotherapy ,Cell Line, Tumor ,Antibodies, Bispecific ,Genetics ,medicine ,Animals ,Receptor ,Melanoma ,Molecular Biology ,Cells, Cultured ,Genetics (clinical) ,Articles ,Dendritic Cells ,Immunotherapy ,medicine.disease ,Molecular medicine ,Coculture Techniques ,In vitro ,Mice, Inbred C57BL ,Immunology ,Molecular Medicine ,Female - Abstract
Trifunctional bispecific antibodies (trAbs) used in tumor immunotherapy have the unique ability to recruit T cells toward antigens on the tumor cell surface and, moreover, to activate accessory cells through their immunoglobulin Fc region interacting with activating Fcγ receptors. This scenario gives rise to additional costimulatory signals required for T cell–mediated tumor cell destruction and induction of an immunologic memory. Here we show in an in vitro system that most effective trAb-dependent T-cell activation and tumor cell elimination are achieved in the presence of dendritic cells (DCs). On the basis of these findings, we devise a novel approach of cancer immunotherapy that combines the specific advantages of trAbs with those of DC-based vaccination. Simultaneous delivery of trAbs and in vitro differentiated DCs resulted in a markedly improved tumor rejection in a murine melanoma model compared with monotherapy.
- Published
- 2013
26. Potent CD4+ T cell-associated antitumor memory responses induced by trifunctional bispecific antibodies in combination with immune checkpoint inhibition
- Author
-
Nina Eißler, Peter Ruf, Horst Lindhofer, Nina Deppisch, and Ralph Mocikat
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,medicine.medical_treatment ,Catumaxomab ,Melanoma, Experimental ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cancer immunotherapy ,Cell Line, Tumor ,Antibodies, Bispecific ,T-cell activation ,medicine ,melanoma ,Animals ,CTLA-4 Antigen ,cancer immunotherapy ,biology ,business.industry ,Melanoma ,Immunotherapy ,medicine.disease ,Ipilimumab ,Xenograft Model Antitumor Assays ,Immune checkpoint ,Tumor antigen ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,CTLA-4 ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,tumor antigen ,Antibody ,business ,Immunologic Memory ,Spleen ,medicine.drug ,Research Paper - Abstract
// Nina Deppisch 1 , Peter Ruf 2 , Nina Eisler 1 , Horst Lindhofer 2 , Ralph Mocikat 1, 3 1 Institut fur Molekulare Immunologie, Helmholtz-Zentrum Munchen, Germany 2 Trion Research GmbH, Martinsried, Germany 3 AG Translationale Molekulare Immunologie, Helmholtz-Zentrum Munchen, Germany Correspondence to: Ralph Mocikat, email: Mocikat@helmholtz-muenchen.de Keywords: tumor antigen, T-cell activation, melanoma, CTLA-4, cancer immunotherapy Received: July 25, 2016 Accepted: December 05, 2016 Published: December 10, 2016 ABSTRACT Combinatorial approaches of immunotherapy hold great promise for the treatment of malignant disease. Here, we examined the potential of combining an immune checkpoint inhibitor and trifunctional bispecific antibodies (trAbs) in a preclinical melanoma mouse model using surrogate antibodies of Ipilimumab and Catumaxomab, both of which have already been approved for clinical use. The specific binding arms of trAbs redirect T cells to tumor cells and trigger direct cytotoxicity, while the Fc region activates accessory cells eventually giving rise to a long-lasting immunologic memory. We show here that T cells redirected to tumor cells by trAbs strongly upregulate CTLA-4 expression in vitro and in vivo . This suggested that blocking of CTLA-4 in combination with trAb treatment enhances T-cell activation in a tumor-selective manner. However, when mice were challenged with melanoma cells and subsequently treated with antibodies, there was only a moderate beneficial effect of the combinatorial approach in vivo with regard to direct tumor destruction in comparison to trAb therapy alone. By contrast, a significantly improved vaccination effect was obtained by CTLA-4 blocking during trAb-dependent immunization. This resulted in enhanced rejection of melanoma cells given after pre-immunization. The improved immunologic memory induced by the combinatorial approach correlated with an increased humoral antitumor response as measured in the sera and an expansion of CD4 + memory T cells found in the spleens.
- Published
- 2016
27. Development of a Bifunctional Checkpoint Inhibitory T Cell Engager (CiTE) to Reverse Adaptive Immune Escape in AML
- Author
-
Nadja C. Fenn, Christina Krupka, Monika Herrmann, Karl-Peter Hopfner, Sebastian Kobold, Katrin Deiser, Bettina Lindl, Ralph Mocikat, Klaus H. Metzeler, Karsten Spiekermann, and Marion Subklewe
- Subjects
0301 basic medicine ,Severe combined immunodeficiency ,Cell cycle checkpoint ,biology ,Chemistry ,T cell ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,biology.protein ,IL-2 receptor ,Antibody ,Cytotoxicity - Abstract
The CD33-targeting bispecific T cell engager (BiTE®) AMG 330 proved to be highly efficient in mediating cytotoxicity of AML cells in vitro and in mouse models (Krupka et al, Blood 2014). Yet, T cell activation is correlated with the upregulation of PD-L1 and other inhibitory checkpoint molecules on AML cells that confer adaptive immune resistance (Krupka et al, Leukemia 2016). PD-1/PD-L1 blocking agents may counteract T cell dysfunction, however, at the expense of broadly distributed immune-related adverse events (irAEs). We developed a checkpoint inhibitory T cell engaging (CiTE) antibody that combines T cell redirection to CD33 on AML cells with locally restricted immune checkpoint blockade. CiTE constructs were generated by first fusing a high-affinity CD33 single-chain variable fragment (scFv) to a CD3ε scFv in one polypeptide chain. Next, this single-chain chain was fused to the extracellular domain of PD-1 (PD-1ex), which naturally holds a low affinity to PD-L1. Antigen binding of CiTE constructs as well as CiTE mediated cytotoxicity of AML cell lines and primary AML cells were done using multiparameter flow cytometry. T cell activation and cytotoxicity assays were complemented by cytometric bead arrays. Murine AML xenograft studies using non-obese diabetic (NOD) scid gamma mice were used for engraftment of primary AML cells and assessment of CiTE mediated cytotoxicity in vivo. CiTE antibody constructs were successfully generated by fusing the bispecific CD33-CD3ε scFv to the endogenous extracellular domain of human PD-1 (PD-1ex). The CiTE was compared to a single chain triplebody (sctb), in which PD-1ex was replaced by a high-affinity PD-L1 scFv. The BiTE-like molecule, PD-1ex.αCD3 and αPD-L1.αCD3, as well as a non-targeting molecule served as controls. When investigating CiTE and sctb as whole molecules, both bound with similar affinities to CD33+PD-L1+ AML cell lines and HD T cells. CiTE- and sctb-induced upregulation of CD69 and CD25 on healthy donor T cells in the presence of MOLM-13-PD-L1 cells. By a synergistic effect of checkpoint blockade and avidity-dependent binding, the PD-1ex attachment increased T cell activation (3.3-fold elevation of IFN-γ release) and lead to efficient and highly selective cytotoxicity of CD33+PD-L1+ cells (EC50 = 2.3 pM to 26.9 pM) as well as primary AML patient samples (n=8). CiTE induced preferential lysis of CD33+PD-L1+ cells and had no activity against CD33-PD-L1+ cells. This was supported by the observation that the CiTE molecule was able to selectively induce elimination of CD33+PD-L1+ cells in the presence of PD-L1+ cells. In a murine xenograft model, the CiTE induced complete AML eradication without causing leukemia-unrelated T cell activation or body weight loss. Notably, murine and human PD-L1 bind with similar affinities to PD-1. We conclude that our molecule preferentially targets CD33+PD-L1+ AML cells, whereas high-affinity blocking agents also address PD-L1+ non-AML cells. Based on these findings, we expect to reverse adaptive immune escape mechanisms of T cell recruiting antibody formats and avoid irAEs associated with systemic checkpoint blockade, suggesting efficient therapeutic potential particularly for patients with relapsed or refractory AML. Future studies will need to further examine efficiency and tolerance in advanced in vivo models before applying the CiTE format into a clinical setting. Disclosures Lindl: Amgen: Research Funding. Metzeler:Celgene: Consultancy, Research Funding; Novartis: Consultancy. Subklewe:Gilead: Consultancy, Honoraria, Research Funding; Amgen: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria; Roche: Consultancy, Research Funding; Celgene: Consultancy, Honoraria.
- Published
- 2018
28. Immunostimulatory RNA Oligonucleotides Induce an Effective Antitumoral NK Cell Response through the TLR7
- Author
-
Carole Bourquin, Gunther Hartmann, Bettina Storch, Stefan Endres, Veit Hornung, Nadja Sandholzer, Cornelia Wurzenberger, Anna-Lisa Lanz, Hendrik Poeck, Michael Berger, Ralph Mocikat, David Anz, and Laura S. Schmidt
- Subjects
Immunology ,Biology ,Lymphocyte Activation ,Interferon-gamma ,Mice ,Adjuvants, Immunologic ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Cytotoxicity ,B cell ,Mice, Knockout ,Oligoribonucleotides ,Oligonucleotide ,Effector ,RNA ,Cancer ,Dendritic Cells ,TLR7 ,medicine.disease ,Molecular biology ,Cell biology ,Killer Cells, Natural ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Oligodeoxyribonucleotides ,Toll-Like Receptor 7 ,Interferon Type I ,Female ,Poly A - Abstract
RNA oligonucleotides containing immune-activating sequences promote the development of cytotoxic T cell and B cell responses to Ag. In this study, we show for the first time that immunostimulatory RNA oligonucleotides induce a NK cell response that prevents growth of NK-sensitive tumors. Treatment of mice with immunostimulatory RNA oligonucleotides activates NK cells in a sequence-dependent manner, leading to enhanced IFN-γ production and increased cytotoxicity. Use of gene-deficient mice showed that NK activation is entirely TLR7-dependent. We further demonstrate that NK activation is indirectly induced through IL-12 and type I IFN production by dendritic cells. Reconstitution of TLR7-deficient mice with wild-type dendritic cells restores NK activation upon treatment with immunostimulatory RNA oligonucleotides. Thus, by activating both NK cells and CTLs, RNA oligonucleotides stimulate two major cellular effectors of antitumor immunity. This dual activation may enhance the efficacy of immunotherapeutic strategies against cancer by preventing the development of tumor immune escape variants.
- Published
- 2009
29. Die Diktatur der Zitatenindizes: Folgen für die WissenskulturThe Tyranny of Citation Indices: Implications for the Culture of Knowledge
- Author
-
Ralph Mocikat
- Subjects
Political science ,Economics, Econometrics and Finance (miscellaneous) ,Environmental Science (miscellaneous) ,Citation ,Humanities ,Management - Published
- 2009
30. EpCAM, a human tumor-associated antigen promotes Th2 development and tumor immune evasion
- Author
-
Ralph Mocikat, Regina Heidenreich, Alexandra I. Ziegler, Martin Röcken, Susanne Weidemann, Heidi Braumüller, and Hartwig Wolburg
- Subjects
medicine.medical_treatment ,Immunology ,In Vitro Techniques ,Biology ,medicine.disease_cause ,Cancer Vaccines ,Biochemistry ,Mice ,chemistry.chemical_compound ,Th2 Cells ,Immune system ,Antigen ,Antigens, Neoplasm ,Cell Line, Tumor ,medicine ,Animals ,Cells, Cultured ,Interleukin 4 ,Mice, Knockout ,Mice, Inbred BALB C ,Cell Death ,Cell Differentiation ,Epithelial cell adhesion molecule ,Cell Biology ,Hematology ,Th1 Cells ,Epithelial Cell Adhesion Molecule ,Adoptive Transfer ,Gene Expression Regulation, Neoplastic ,Immunosurveillance ,Cytokine ,chemistry ,Tumor progression ,Colonic Neoplasms ,Female ,Interleukin-4 ,Carcinogenesis ,Cell Adhesion Molecules ,Cell Division ,Neoplasm Transplantation - Abstract
Experimental tumor vaccination and adoptive T-cell therapies show that interferon-γ (IFN-γ)–producing CD4+ T helper cells (Th1) can be highly effective in tumor prevention and therapy. Unexpectedly, first vaccine trials in humans revealed that tumor immune therapy may not only be protective, but, on the contrary, even promote tumor progression. Here, we analyzed T-cell immune responses to the epithelial cell adhesion molecule (EpCAM), one of the most common tumor-associated antigens (TAA) serving as immune target in colon cancer patients. Th-cell priming against EpCAM inevitably resulted in interleukin-4 (IL-4)–dominated Th2 responses, even under most stringent Th1-inducing conditions. These EpCAM-reactive Th2 cells rather promoted growth of EpCAM-expressing tumors. To analyze the role of IL-4 in tumor immune evasion, we generated EpCAM-reactive Th1 cells from IL-4.ko mice. These Th1 cells provided tumor-specific protection and established highly protective Th1 memory responses, even in naive BALB/c mice. Inhibition of tumor growth by Th1 cells resulted in intra-tumoral expression of cytokines of the IL-12 family and of IFN-γ. Preventing activation-associated death of Th1 cells further increased intratumoral IFN-γ expression and improved therapeutic efficacy. Thus, human TAA may promote tumor immune evasion by strongly favoring Th2 development.
- Published
- 2009
31. A Polyvalent Cellular Vaccine Induces T-cell Responses Against Specific Self-antigens Overexpressed in Chronic Lymphocytic B-cell Leukemia
- Author
-
Elfriede Nöner, Ralph Mocikat, Martin Dreyling, Bernhard Frankenberger, Ulrich Wahl, Michael Hallek, and Konrad Kronenberger
- Subjects
Male ,Cancer Research ,medicine.medical_treatment ,T cell ,Chronic lymphocytic leukemia ,Immunology ,Autoantigens ,Cancer Vaccines ,Immune system ,Antigen ,Antigens, Neoplasm ,Humans ,Immunology and Allergy ,Medicine ,Antigen-presenting cell ,Aged ,Aged, 80 and over ,Pharmacology ,B-Lymphocytes ,business.industry ,Immunotherapy, Active ,Proto-Oncogene Proteins c-mdm2 ,Dendritic Cells ,Immunotherapy ,Dendritic cell ,Middle Aged ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,DNA-Binding Proteins ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-2 ,B-cell leukemia ,Female ,business ,T-Lymphocytes, Cytotoxic ,Transcription Factors - Abstract
B cell-derived chronic lymphocytic leukemia (CLL) is an incurable disease that requires innovative therapeutic regimens. Experimental approaches of immunotherapy aiming at induction of systemic T-cell responses have been developed. Trioma cells provide a potent vaccine derived from malignant B cells that allows multiple antigens (Ags) from the parental tumor to be ingested by Ag-presenting cells. Like other strategies using modified whole tumor cells, this approach induces polyvalent responses. Using trioma cell-pulsed dendritic cells (DCs) for T-cell activation in vitro, we asked whether specific Ags overexpressed by CLL can be identified as target structures of such responses and what is the nature of these Ags. Expression levels of several genes in CLL samples were quantitated by reverse transcriptase-polymerase chain reaction. T lymphocytes were polyvalently stimulated by trioma-pulsed DCs and specificities were tested by determining cytokine secretion in the presence of target cells transfected with RNA coding for those Ags that were found to be overexpressed. We demonstrate that DCs pulsed with the modified tumor cells efficiently activate T lymphocytes against CLL and that overexpressed Ags related to leukemogenesis, such as BCL-2, MDM2, and ETV5, serve as targets for those T cells. Immune escape by Ag loss or mutation is less likely to occur if immunity is directed against altered self-proteins that are involved in malignant transformation. Therefore, vaccines based on modified tumor cells such as triomas hold promise for immunotherapy of CLL and other malignancies. Polyvalent vaccines originally designed as individualized therapeutics may be more broadly applicable, at least in patients showing similar Ag patterns.
- Published
- 2008
32. Antitumor effector functions of T cells are dependent on in vivo priming and restricted T-cell receptor expression
- Author
-
Martin Röcken, Elfriede Nößner, Carolin Lüking, Konrad Kronenberger, Ralph Mocikat, and Bernhard Frankenberger
- Subjects
CD4-Positive T-Lymphocytes ,Cancer Research ,Adoptive cell transfer ,Lymphoma, B-Cell ,Receptors, Antigen, T-Cell, alpha-beta ,T-Lymphocytes ,Priming (immunology) ,CD8-Positive T-Lymphocytes ,Biology ,Lymphocyte Activation ,Interferon-gamma ,Mice ,Interleukin 21 ,Antigen ,In vivo ,Animals ,Cytotoxic T cell ,RNA, Messenger ,IL-2 receptor ,Mice, Inbred BALB C ,Reverse Transcriptase Polymerase Chain Reaction ,Vaccination ,Flow Cytometry ,Adoptive Transfer ,Cell biology ,Oncology ,Immunology ,Female ,Immunologic Memory ,Ex vivo ,T-Lymphocytes, Cytotoxic - Abstract
Tumor-specific T cells are crucial for immunologic control of malignant disease. T cells can be induced in vivo by vaccination or adoptively transferred after activation ex vivo. We investigated the requirements for generating T cells with optimal antitumor effector functions in a murine lymphoma model. Using adoptive transfer, we show that in vivo efficacy of T cells cannot be predicted by tumor reactivity in vitro. A restricted T-cell receptor β chain repertoire of T-cell populations stimulated ex vivo against tumor cells was necessary but not sufficient for tumor protectivity. Tumor elimination furthermore required vaccination of donor mice, hence in vivo priming. The in vivo priming step may allow tumor-specific T cells to accumulate in vitro more rapidly and to survive for longer periods after withdrawal of the antigenic stimulus and adoptive transfer. A possible survival benefit of in vivo induced T cells may be ascribed to the responsiveness to homeostatic cytokines and to unique cytokine milieus encountered in vivo. Most importantly, monoclonal T cells cannot inhibit tumor growth. A prerequisite of tumor rejection was the expression of at least 2 T-cell receptor β chains by transferred T-cell populations. This finding has implications for designing adoptive transfer strategies for the clinic. © 2008 Wiley-Liss, Inc.
- Published
- 2008
33. Die Sprachenfrage in den Naturwissenschaften
- Author
-
Ralph Mocikat
- Published
- 2015
34. Persistence of xenogenized vaccine cellsin vivo
- Author
-
Ralph Mocikat, Nicolas Graf, and Christian Adam
- Subjects
Cancer Research ,business.industry ,medicine.medical_treatment ,Cell ,Antigen presentation ,Immunotherapy ,Cell cycle ,Suicide gene ,Vaccination ,medicine.anatomical_structure ,Oncology ,Antigen ,Immunology ,medicine ,business ,B cell - Abstract
Trioma cell vaccination is a potent new immunotherapeutic strategy for the treatment of B cell neoplasias. It is based on the specific redirection of tumor antigens against surface receptors on professional antigen-presenting cells (APC) that internalize antigens and present immunogenic peptides to T-lymphocytes. Tumor cells are converted to trioma cells by fusion with xenogeneic hybridomas expressing an anti-APC specificity. The trioma cell is a polyvalent vaccine that contains potentially all lymphoma-derived antigens. Apart from the expression of the APC-binding arm by the trioma cell, another requirement for successful tumor protection is the xenogeneic moiety of the trioma cells. We show that, despite their xenogenicity, trioma cells persist for extended periods in vaccinated animals. Trioma cells could be identified in spleens as long as about 12 weeks post vaccination. By using a suicide gene approach, trioma cells could partly be eliminated from immunized mice, whereby the antitumor effect was partly abrogated. We argue that not all trioma cells are immediately lysed in vivo and that the cell cycle of the remaining cells is arrested after having undergone few divisions. Trioma cells surviving in vivo may be instrumental for efficient induction of tumor immunity.
- Published
- 2003
35. CD40-independent NK-cell help promotes dendritic cell vaccine-induced T-cell immunity against endogenous B-cell lymphoma
- Author
-
Nadine, Hömberg, Christian, Adam, Tanja, Riedel, Christoph, Brenner, Andrew, Flatley, Martin, Röcken, and Ralph, Mocikat
- Subjects
Mice, Inbred BALB C ,Lymphoma, B-Cell ,Genes, myc ,Mice, Transgenic ,chemical and pharmacologic phenomena ,Dendritic Cells ,Cancer Vaccines ,Killer Cells, Natural ,Mice, Inbred C57BL ,Interferon-gamma ,Mice ,Cell Line, Tumor ,Ctl Response ,Dc Maturation ,Adaptive Antitumor Immunity ,Interferon-γ ,λ-myc Mouse ,Animals ,Cytokines ,Female ,CD40 Antigens ,Neoplasm Transplantation ,T-Lymphocytes, Cytotoxic - Abstract
It is well established that an interplay between natural killer (NK) cells and dendritic cells (DCs) gives rise to their reciprocal activation and provides a Th1-biased cytokine milieu that fosters antitumor T-cell responses. Ex vivo-differentiated DCs transferred into mice strongly stimulate endogenous NK cells to produce interferon (IFN)-γ and initiate a cascade that eventually leads to cytotoxic T-lymphocyte responses. We show that the ability of exogenous DCs to trigger this pathway obviates CD40 signaling and CD4(+) T-cell help and depends on a preceding maturation step. Importantly, this mechanism was also effective in endogenously arising tumors where IFN-γ production is compromised in contrast to transplantable tumors. In c-myc-transgenic mice developing spontaneous lymphomas, injection of unpulsed DCs caused NK-cell activation and induced CD8(+) T cells capable of recognizing the lymphoma cells. Animals treated with unpulsed DCs showed a survival benefit compared to untreated myc mice. Hence, tumor immunity induced by DC-based vaccines not only depends on specific antigens loaded on the DCs. Rather, DC vaccines generate broader immune responses, because endogenous DCs presenting tumor antigens may also become stimulated by NK cells that were activated by exogenous DCs. Thus, the DC/NK-cell/cytotoxic T lymphocyte axis may commonly have relevance for DC-based vaccination protocols in clinical settings.
- Published
- 2014
36. B-cell lymphoma idiotypes chimerized by gene targeting can induce tumor immunity
- Author
-
Jean-Pierre Kremer, Helge Menzel, Stefan Thierfelder, Ralph Mocikat, and Michael Selmayr
- Subjects
Cancer Research ,Lymphoma, B-Cell ,medicine.medical_treatment ,Gene Targeting ,Idiotype ,Immunoglobulin Heavy Chain ,Lymphoma Immunization ,Antigen presentation ,DNA vaccination ,Mice ,Immunoglobulin Idiotypes ,Tumor Cells, Cultured ,Vaccines, DNA ,medicine ,Animals ,Humans ,B-cell lymphoma ,Molecular Biology ,Mice, Inbred BALB C ,Mice, Inbred C3H ,biology ,Chimera ,Gene targeting ,Immunotherapy ,medicine.disease ,Adoptive Transfer ,Fusion protein ,Antibodies, Anti-Idiotypic ,Immunization ,Immunology ,biology.protein ,Molecular Medicine ,Antibody ,Injections, Intraperitoneal - Abstract
Immunization with modified immunoglobulin (Ig) idiotypes (Ids) of B-cell lymphomas is an attractive approach of experimental tumor immunotherapy. We show here that B-lymphoma cells can be gene-modified by homologous recombination at the Ig heavy chain locus. Although it has been demonstrated previously that a protein vaccine containing a mouse/human chimeric Ig had no immunostimulatory effect, we show that a xenogeneic Fc segment attached to the Id by gene targeting in autologous murine tumor cells can serve as an immunogenic carrier and is capable of inducing tumor protection. A prerequisite for successful vaccination is the delivery of tumor cells that have been engineered to express the Id in the chimeric form rather than administration of the soluble chimeric protein. Also DNA vaccination with plasmids encoding chimeric Ids was reported to induce an anti-idiotypic response, suggesting that there might be related mechanisms such as enhanced antigen presentation. Immunization with engineered lymphoma cells is a very potent protocol: in the cell-based setting, minute levels of expression in the gene-targeted cells are sufficient to confer tumor immunity. Because the titers of anti-Id antibodies induced do not reflect the degree of tumor protection, the immune mechanisms responsible for tumor rejection cannot be ascribed exclusively to a humoral response.
- Published
- 2000
37. Gene therapy of B-cell lymphoma with cytokine gene-modified trioma cells
- Author
-
Horst Lindhofer, Michael Selmayr, Jean‐P. Kremer, Ralph Mocikat, Lothar Hültner, and John Strehl
- Subjects
Idiotype ,Cancer Research ,Lymphoma, B-Cell ,Time Factors ,Genetic enhancement ,medicine.medical_treatment ,Antigen-Presenting Cells ,Enzyme-Linked Immunosorbent Assay ,Biology ,Cancer Vaccines ,Mice ,Antibodies, Bispecific ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Cloning, Molecular ,Antigen-presenting cell ,B-cell lymphoma ,Mice, Inbred BALB C ,Genetic transfer ,Granulocyte-Macrophage Colony-Stimulating Factor ,Genetic Therapy ,Immunotherapy ,medicine.disease ,Lymphoma ,Cytokine ,Oncology ,Immunology ,Cytokines ,Female ,Interleukin-4 - Abstract
The trioma approach is a new immunotherapeutic strategy for treating B-cell lymphomas. It is based on converting the tumour idiotype to a bispecific immunoglobulin that redirects the idiotype to antigen-presenting cells. We show here that even pre-existing tumours can be eradicated by trioma vaccination, that the trioma approach is superior to vaccination with cytokine gene-modified autologous tumour cells and that there is a synergism between trioma immunisation and GM-CSF gene transfer. Furthermore, we show that the immunising potential of GM-CSF gene-modified autologous lymphoma cells is not as dependent on the cytokine expression level as described for other tumour models, such that even minute expression rates are effective. IL-4 gene transfer in the lymphoma model is considerably less efficient or even ineffective when more sensitive systems are used. Remarkably, trioma-mediated effects are extinguished when IL-4 is expressed by the trioma cell.
- Published
- 1999
38. Improving the expression of chimeric antibodies following homologous recombination in hybridoma cells
- Author
-
Ralph Mocikat
- Subjects
Hybridomas ,Genes, Immunoglobulin ,biology ,Recombinant Fusion Proteins ,Immunology ,food and beverages ,Gene targeting ,Immunoglobulin E ,Molecular biology ,Recombinant Proteins ,law.invention ,Mice ,law ,Sequence Homology, Nucleic Acid ,biology.protein ,Recombinant DNA ,Animals ,Humans ,Immunology and Allergy ,Immunoglobulin heavy chain ,Antibody ,Homologous recombination ,Gene ,Recombination - Abstract
Mouse/human chimeric antibodies can easily be generated by exchanging the immunoglobulin constant gene segments with human sequences in mouse hybridoma cells by gene targeting. This obviates the need to isolate the variable gene regions from the hybridoma and permits high-level expression of chimeric antibodies, because the production rate of the hybridoma is often maintained. Here we show that the efficiency of this strategy can be further improved by increasing the number of high-producer clones arising from a recombination experiment. In principle, antibody production can be enhanced by removing the selection marker genes from the targeted cells.
- Published
- 1999
39. A novel CXCL10-based GPI-anchored fusion protein as adjuvant in NK-based tumor therapy
- Author
-
Laura Hinz, Niklas Muenchmeier, Nicole Rieth, Sophia Boecker, Peter J. Nelson, Elfriede Noessner, Anna N. Mendler, Lorenz Bankel, Constantin Lapa, and Ralph Mocikat
- Subjects
Glycosylphosphatidylinositols ,Recombinant Fusion Proteins ,lcsh:Medicine ,CHO Cells ,C-C chemokine receptor type 6 ,Biology ,Protein Engineering ,Cell Line ,Mice ,Chemokine receptor ,Cricetulus ,Adjuvants, Immunologic ,Cricetinae ,Neoplasms ,Cell Adhesion ,Animals ,Humans ,CXCL10 ,ddc:610 ,lcsh:Science ,CXCL14 ,CXCL16 ,Multidisciplinary ,Cell Membrane ,lcsh:R ,Mucins ,Endothelial Cells ,Endocytosis ,Cell biology ,Chemokine CXCL10 ,Killer Cells, Natural ,Mice, Inbred C57BL ,XCL2 ,Female ,lcsh:Q ,Immunotherapy ,Rheology ,CC chemokine receptors ,Neoplasm Transplantation ,Research Article ,CCL21 - Abstract
Background Cellular therapy is a promising therapeutic strategy for malignant diseases. The efficacy of this therapy can be limited by poor infiltration of the tumor by immune effector cells. In particular, NK cell infiltration is often reduced relative to T cells. A novel class of fusion proteins was designed to enhance the recruitment of specific leukocyte subsets based on their expression of a given chemokine receptor. The proteins are composed of an N-terminal chemokine head, the mucin domain taken from the membrane-anchored chemokine CX3CL1, and a C-terminal glycosylphosphatidylinositol (GPI) membrane anchor replacing the normal transmembrane domain allowing integration of the proteins into cell membranes when injected into a solid tumor. The mucin domain in conjunction with the chemokine head acts to specifically recruit leukocytes expressing the corresponding chemokine receptor. Methodology/Principal Findings A fusion protein comprising a CXCL10 chemokine head (CXCL10-mucin-GPI) was used for proof of concept for this approach and expressed constitutively in Chinese Hamster Ovary cells. FPLC was used to purify proteins. The recombinant proteins efficiently integrated into cell membranes in a process dependent upon the GPI anchor and were able to activate the CXCR3 receptor on lymphocytes. Endothelial cells incubated with CXCL10-mucin-GPI efficiently recruited NK cells in vitro under conditions of physiologic flow, which was shown to be dependent on the presence of the mucin domain. Experiments conducted in vivo using established tumors in mice suggested a positive effect of CXCL10-mucin-GPI on the recruitment of NK cells. Conclusions The results suggest enhanced recruitment of NK cells by CXCL10-mucin-GPI. This class of fusion proteins represents a novel adjuvant in cellular immunotherapy. The underlying concept of a chemokine head fused to the mucin domain and a GPI anchor signal sequence may be expanded into a broader family of reagents that will allow targeted recruitment of cells in various settings.
- Published
- 2013
40. T-helper-1-cell cytokines drive cancer into senescence
- Author
-
Friedegund Meier, Rupert Handgretinger, Maries van den Broek, Marina Pesic, Manfred Kneilling, Martin Röcken, Martin Schaller, Tarun Mehra, Heike Niessner, Falko Fend, Matthias Hahn, Julia Berdel, Susanne Ullrich, Karin Schilbach, Hans-Ulrich Häring, Klaus Schulze-Osthoff, Sonja Aßmann, Heidi Braumüller, Jürgen Bauer, Birgit Fehrenbacher, Ralph Mocikat, Christoph M. Griessinger, Frank Essmann, Ellen Brenner, Lars Zender, Felicia Ranta, Thomas Wieder, Kilian Braungart, Mohammed Alkhaled, Leticia Quintanilla-Martinez, University of Zurich, and Braumüller, Heidi
- Subjects
Senescence ,Male ,Time Factors ,Antigens, Polyomavirus Transforming ,Mice, Transgenic ,610 Medicine & health ,Mice, SCID ,Biology ,10263 Institute of Experimental Immunology ,Retinoblastoma Protein ,Interferon-gamma ,Mice ,Phosphoserine ,CDKN2A ,Mice, Inbred NOD ,Neoplasms ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,HSPA1L ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,Cell Proliferation ,1000 Multidisciplinary ,Multidisciplinary ,Cell growth ,Tumor Necrosis Factor-alpha ,Cell Cycle ,Cancer ,Oncogenes ,Cell cycle ,Th1 Cells ,Acquired immune system ,medicine.disease ,Disease Models, Animal ,STAT1 Transcription Factor ,Receptors, Tumor Necrosis Factor, Type I ,Immunology ,Cancer research ,Disease Progression ,Cytokines ,570 Life sciences ,biology ,Tumor necrosis factor alpha ,Female ,Tumor Suppressor Protein p53 - Abstract
Cancer control by adaptive immunity involves a number of defined death and clearance mechanisms. However, efficient inhibition of exponential cancer growth by T cells and interferon-γ (IFN-γ) requires additional undefined mechanisms that arrest cancer cell proliferation. Here we show that the combined action of the T-helper-1-cell cytokines IFN-γ and tumour necrosis factor (TNF) directly induces permanent growth arrest in cancers. To safely separate senescence induced by tumour immunity from oncogene-induced senescence, we used a mouse model in which the Simian virus 40 large T antigen (Tag) expressed under the control of the rat insulin promoter creates tumours by attenuating p53- and Rb-mediated cell cycle control. When combined, IFN-γ and TNF drive Tag-expressing cancers into senescence by inducing permanent growth arrest in G1/G0, activation of p16INK4a (also known as CDKN2A), and downstream Rb hypophosphorylation at serine 795. This cytokine-induced senescence strictly requires STAT1 and TNFR1 (also known as TNFRSF1A) signalling in addition to p16INK4a. In vivo, Tag-specific T-helper 1 cells permanently arrest Tag-expressing cancers by inducing IFN-γ- and TNFR1-dependent senescence. Conversely, Tnfr1(-/-)Tag-expressing cancers resist cytokine-induced senescence and grow aggressively, even in TNFR1-expressing hosts. Finally, as IFN-γ and TNF induce senescence in numerous murine and human cancers, this may be a general mechanism for arresting cancer progression.
- Published
- 2013
41. Immunosuppression by Fc region-mismatched anti-T cell antibody treatment
- Author
-
Josef Mysliwietz, Horst Dr Lindhofer, Elisabeth Kremmer, Ralph Mocikat, and Stefan Thierfelder
- Subjects
Graft Rejection ,Male ,Idiotype ,medicine.drug_class ,T-Lymphocytes ,medicine.medical_treatment ,Immunology ,Priming (immunology) ,Receptors, Fc ,Monoclonal antibody ,Mice ,Antibodies, Bispecific ,Immune Tolerance ,medicine ,Animals ,Immunology and Allergy ,Immunosuppression Therapy ,biology ,Antibodies, Monoclonal ,Anti-antibodies ,Anti-t Cell Mab ,Skin Grafts ,Skin Transplantation ,Immunotherapy ,Fragment crystallizable region ,Antibodies, Anti-Idiotypic ,Mice, Inbred C57BL ,Polyclonal antibodies ,Monoclonal ,Mice, Inbred CBA ,biology.protein ,Female ,Antibody - Abstract
Formation of anti-antibodies (anti-Ab) is known to counteract immunotherapy with anti-T cell antibodies. Our previously described immunological approach prevented anti-Ab with the consequence of prolonged survival of fully mismatched skin grafts in C57BL/6 mice. These mice were treated with a single priming injection of a monoclonal anti-T cell Ab followed by repeated injections of anti-T cell mAb differing in species origin from the priming mAb. We now show prolonged tolerance to discordant xenogeneic, to bispecific, and even to polyclonal Ab, and demonstrate that the underlying immunosuppressive principle is due to a difference in heavy chain constant region between first and second antibodies, independent of whether or not they share the same idiotype. To examine this phenomenon, a panel of mAb was generated which share the same mouse anti-Thy-1.2 idiotype, but carry a human IgG1(T23), IgG3(T212C8), or mouse IgG2a(MmT1) constant heavy chain region. We found that sequential injection of MmT1 and T23 according to the above treatment schedule induced huIgG1 isotype-specific tolerance to T23, which was similar to that seen when using a primary mAb (MmT5) that was, instead, fully mismatched with T23 in both idiotype and constant region. Thus, differences of idiotype between primary and booster Ab were inconsequential for their ability to inhibit anti-Ab formation. This novel form of induced specific tolerance to anti-T cell Ig survived graft rejection and was still evident 230 days after termination of the T cell depletion protocol. Taken together, these results demonstrate that rechallenge with Fc region-mismatched Ab opens an immunological window that allows for induction of tolerance to immunogenic anti-T cell Ab and prolonged immunosuppression.
- Published
- 1995
42. Integration vectors for antibody chimerization by homologous recombination in hybridoma cells
- Author
-
Christian Kardinal, Reinhard Zeidler, Erik Hooijberg, Ralph Mocikat, and Peter Lang
- Subjects
Recombinant Fusion Proteins ,Genetic Vectors ,Immunology ,Computational biology ,Homology (biology) ,Mice ,Mice, Inbred AKR ,Nucleic acid thermodynamics ,Animals ,Humans ,Immunology and Allergy ,Genetics ,Hybridomas ,biology ,Nucleic Acid Hybridization ,Gene targeting ,Isotype ,Immunoglobulin Isotypes ,Gene Targeting ,biology.protein ,Immunoglobulin heavy chain ,Chimeric Antibody ,Gene Expression ,Homologous Recombination ,Igh Locus ,Integration Vector ,Antibody ,Immunoglobulin Heavy Chains ,Homologous recombination ,Recombination - Abstract
Gene targeting in hybridoma cells provides a tool for generating chimeric antibodies with great ease and at high yield. We present an evaluation of integration vectors for the chimerization of the immunoglobulin heavy chain locus which are universally applicable to hybridomas of different isotypes and mouse strains. There are three problems arising with vector integration: (i) the frequent persistence of the parental isotype; (ii) an isotype-dependent aberrant replacement-like recombination giving rise to antibodies devoid of the CH1 domain; and (iii) secondary recombinations leading to excision of the integrated sequence. To overcome these problems, we have systematically evaluated the consequences of extending the vector flank. Although the homology length clearly determines the recombination frequency, this effect is counteracted by the secondary recombination, which also correlates to the homology length. In contrast, the truncating recombination events are not dependent on the homology length and never lead to re-excision of the construct. To take advantage of the increased genetic stability obtained with short flanks, we constructed an enrichment vector which yields high recombination efficiencies despite using a short flanking sequence. In addition, irradiation of the cells enhanced homologous recombination. The problem of the co-production of two isotypes was overcome by a two-step targeting reaction.
- Published
- 1995
43. Ganglioside GD2-specific trifunctional surrogate antibody Surek demonstrates therapeutic activity in a mouse melanoma model
- Author
-
Horst Lindhofer, Peter Ruf, Christine Zehetmeier, Beatrix Schäfer, Matthias Plöscher, Ivonne Suckstorff, Juergen Hess, Ralph Mocikat, Nina Eissler, and Susanne Wosch
- Subjects
Cytotoxicity, Immunologic ,Adoptive cell transfer ,Skin Neoplasms ,Time Factors ,Antibodies, Neoplasm ,T-Lymphocytes ,medicine.medical_treatment ,CD3 ,Dose-Response Relationship, Immunologic ,lcsh:Medicine ,Trab ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Immune system ,Antigen ,Antibody Specificity ,Gangliosides ,Antibodies, Bispecific ,medicine ,Animals ,Humans ,Melanoma ,Medicine(all) ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Research ,Immune Sera ,lcsh:R ,General Medicine ,Immunotherapy ,Trifunctional bispecific asntibody ,Adoptive Transfer ,Survival Analysis ,Molecular biology ,Immunity, Humoral ,Treatment Outcome ,Immunoglobulin G ,biology.protein ,Ganglioside GD2 ,Immunization ,Antibody ,Trifunctional Bispecific Antibody ,Ganglioside Gd2 ,CD8 - Abstract
Background Trifunctional bispecific antibodies (trAb) are a special class of bispecific molecules recruiting and activating T cells and accessory immune cells simultaneously at the targeted tumor. The new trAb Ektomab that targets the melanoma-associated ganglioside antigen GD2 and the signaling molecule human CD3 (hCD3) on T cells demonstrated potent T-cell activation and tumor cell destruction in vitro. However, the relatively low affinity for the GD2 antigen raised the question of its therapeutic capability. To further evaluate its efficacy in vivo it was necessary to establish a mouse model. Methods We generated the surrogate trAb Surek, which possesses the identical anti-GD2 binding arm as Ektomab, but targets mouse CD3 (mCD3) instead of hCD3, and evaluated its chemical and functional quality as a therapeutic antibody homologue. The therapeutic and immunizing potential of Surek was investigated using B78-D14, a B16 melanoma transfected with GD2 and GD3 synthases and showing strong GD2 surface expression. The induction of tumor-associated and autoreactive antibodies was evaluated. Results Despite its low affinity of approximately 107 M-1 for GD2, Surek exerted efficient tumor cell destruction in vitro at an EC50 of 70ng/ml [0.47nM]. Furthermore, Surek showed strong therapeutic efficacy in a dose-dependent manner and is superior to the parental GD2 mono-specific antibody, while the use of a control trAb with irrelevant target specificity had no effect. The therapeutic activity of Surek was strictly dependent on CD4+ and CD8+ T cells, and cured mice developed a long-term memory response against a second challenge even with GD2-negative B16 melanoma cells. Moreover, tumor protection was associated with humoral immune responses dominated by IgG2a and IgG3 tumor-reactive antibodies indicating a Th1-biased immune response. Autoreactive antibodies against the GD2 target antigen were not induced. Conclusion Our data suggest that Surek revealed strong tumor elimination and anti-tumor immunization capabilities. The results warrant further clinical development of the human therapeutic equivalent antibody Ektomab.
- Published
- 2012
44. Recruitment of natural killer cells in advanced stages of endogenously arising B-cell lymphoma: implications for therapeutic cell transfer
- Author
-
Niklas Münchmeier, David Anz, Martin Röcken, Johann Pötzl, Marcella Naujoks, Peter J. Nelson, Ralph Mocikat, Nadine Hömberg, Carole Bourquin, Christoph D. Brenner, and Margarethe Przewoznik
- Subjects
Cancer Research ,Adoptive cell transfer ,Lymphoma, B-Cell ,Receptors, CXCR3 ,Immunology ,Biology ,Chemokine CXCL9 ,Proinflammatory cytokine ,Cell Line ,Interferon-gamma ,Mice ,Immune system ,Interferon ,medicine ,Immunology and Allergy ,CXCL10 ,Cytotoxic T cell ,Animals ,B-cell lymphoma ,Neoplasm Staging ,Pharmacology ,Mice, Knockout ,medicine.disease ,Adoptive Transfer ,Chemokine CXCL10 ,Killer Cells, Natural ,Mice, Inbred C57BL ,Chemotaxis, Leukocyte ,Tumor progression ,medicine.drug - Abstract
During inflammation and in transplantable tumor models, natural killer (NK) cells are recruited to pathologic tissues and activated to produce proinflammatory cytokines favoring adaptive immune responses of the T-helper type 1 (Th1) type. Interferon (IFN)-γ is needed to induce chemokines that attract NK cells in transplanted tumors. Nothing, however, is known on NK-cell migration in spontaneous tumors. As effective recruitment is a prerequisite for therapeutic NK-cell transfer, we investigated the cytokine milieu and the mechanisms that are instrumental for NK-cell accumulation in an endogenous tumor model. We make use of λ-myc transgenic mice that harbor the c-myc oncogene and develop spontaneous B-cell lymphoma. In contrast to lymphomas induced by tumor cell injection, virtually no IFN-γ produced by NK or by other cells was present in the tumor environment, particularly in advanced stages. Dendritic cells showed an impaired expression of interleukin-12, which is suggestive of deficient Th1 priming. The IFN-γ-dependent chemokines CXCL9 and CXCL10 were pivotal for NK-cell migration in the endogenous lymphoma model. Although IFN-γ was absent in late tumor stages, there was still expression of CXCL9 and CXCL10 with an ongoing influx of NK cells. The results demonstrate that transplantable tumor models do not reflect the situation as found in endogenously arising neoplasia, because in the latter, effective Th1 and cytotoxic T-lymphocyte responses are presumably not induced because of impaired IFN-γ production. The data also suggest that CXCL9 and CXCL10 production and NK-cell migration become independent of IFN-γ during tumor progression, and therefore support approaches of adoptive NK-cell transfer that hold promise for treatment of cancer.
- Published
- 2012
45. Trifunctional bispecific antibodies induce tumor-specific T cells and elicit a vaccination effect
- Author
-
Peter Ruf, Josef Mysliwietz, Nina Eissler, Ralph Mocikat, and Horst Lindhofer
- Subjects
Cancer Research ,CD3 Complex ,T-Lymphocytes ,medicine.medical_treatment ,CD3 ,Melanoma, Experimental ,Epitopes, T-Lymphocyte ,Lymphocyte Activation ,Cancer Vaccines ,Epitope ,Mice ,Immune system ,Antigen ,Gangliosides ,Antibodies, Bispecific ,medicine ,Animals ,Lymph node ,biology ,Chemistry ,Immunotherapy ,Fragment crystallizable region ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Oncology ,Immunology ,biology.protein ,Female ,Antibody - Abstract
A major goal of tumor immunotherapy is the induction of long-lasting systemic T-cell immunity. Bispecific antibodies (bsAbs) that lack the immunoglobulin Fc region confer T–cell-mediated killing of tumor cells but do not induce long-term memory. In contrast, trifunctional bsAbs comprise an appropriate Fc region and, therefore, not only recruit T cells but also accessory cells that bear activating Fcγ receptors (FcγR), providing additional T–cell-activating signals and securing presentation of tumor-derived antigens to T cells. In this study, we show that trifunctional bsAbs induce a polyvalent T-cell response and, therefore, a vaccination effect. Mice were treated with melanoma cells and with a trifunctional bsAb directed against the melanoma target antigen ganglioside GD2 in addition to murine CD3. The trifunctional bsAb activated dendritic cells and induced a systemic immune response that was not replicated by treatment with the F(ab′)2-counterpart lacking the Fc region. Restimulation of spleen and lymph node cells in vitro yielded T-cell lines that specifically produced interferon-γ in response to tumor. In addition, trifunctional bsAb-induced T cells recognized various specific peptides derived from melanoma-associated antigens. Moreover, these polyvalent responses proved to be tumor-suppressive and could not be induced by the corresponding bsF(ab′)2-fragment. Taken together, our findings provide preclinical proof of concept that trifunctional bsAbs can induce tumor-specific T cells with defined antigen specificity. Cancer Res; 72(16); 3958–66. ©2012 AACR.
- Published
- 2012
46. Immunological approach to inhibit formation of anti-antibodies to allo- and xenogeneic anti-T cell immunoglobulin
- Author
-
Josef Mysliwietz, Stefan Thierfelder, Elisabeth Kremmer, and Ralph Mocikat
- Subjects
Antigens, Differentiation, T-Lymphocyte ,Male ,Idiotype ,medicine.drug_class ,T-Lymphocytes ,T cell ,Immunology ,Priming (immunology) ,Biology ,Monoclonal antibody ,Mice ,Immunoglobulin Idiotypes ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Antilymphocyte Serum ,Immunosuppression Therapy ,Graft Survival ,Skin Transplantation ,Molecular biology ,Antibodies, Anti-Idiotypic ,Rats ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Mice, Inbred CBA ,biology.protein ,Female ,Antibody ,Hapten ,CD8 - Abstract
Inhibitory anti-antibodies induced in patients by xenogeneic or even by humanized anti-T cell antibodies remain an unresolved problem. Mice also produce anti-antibodies following injection of xeno- or allogeneic anti-T cell antibodies. Here we report a principle based on sequentially applied anti-T cell antibodies generated in different species, which results in suppressed anti-antibody formation and prolonged immunosuppression. Thus, a single priming injection in mice of mouse (MmT1 or MmT5 differing by idiotype only) or of rat (RmT1) anti-mouse Thy-1 monoclonal antibodies (mAb) or of rat anti-mouse L3T4 + Ly-2 (RmCD4 + CD8) mAb suppressed anti-antibody formation against subsequent booster injections of one of the above antibodies, provided that they differed in species origin from the priming antibody. Correspondingly, a sixfold and longer prolongation of 50% survival of fully mismatched skin grafts was observed. Less or no anti-antibody suppression and little prolongation of graft survival was obtained if the 'first' and the 'second' (and following) antibody injections were of the same species, differing by iso- or idiotype only. Finally, the suppressive principle did not manifest itself at all if the initial antibody injection included both the first and second antibody. These findings are discussed with reference to earlier studies on hapten/carrier effects as well as on immunosuppression attributed to 'non-depleting' rat anti-CD4/CD8 T cell antibodies.
- Published
- 1994
47. Replacement-like recombination induced by an integration vector with a murine homology flank at the immunoglobulin heavy-chain locus in mouse and rat hybridoma cells
- Author
-
Peter Lang and Ralph Mocikat
- Subjects
Blotting, Western ,Genetic Vectors ,Molecular Sequence Data ,Enzyme-Linked Immunosorbent Assay ,Biology ,Polymerase Chain Reaction ,Genetic recombination ,Homology (biology) ,Cell Line ,Mice ,Genetics ,Animals ,Humans ,Ectopic recombination ,Molecular Biology ,Recombination, Genetic ,Hybridomas ,Base Sequence ,Gene targeting ,Isotype ,Rats ,Non-homologous end joining ,Blotting, Southern ,Immunoglobulin Constant Regions ,Immunoglobulin Heavy Chains ,Homologous recombination ,Recombination - Abstract
Vectors for homologous recombination are commonly designed as replacement or integration constructs. We have evaluated integration vectors for the substitution of the immunoglobulin heavy-chain constant region by various human isotypes in mouse and rat hybridomas. It is known that under certain circumstances replacement vectors exhibit a lower target efficiency and can be incorporated by integration events. Conversely, we show here that an integration vector can undergo a replacement event despite having free homologous adjacent DNA ends, which would be expected to initiate integration according to the double-strand break repair model. Moreover, in cases of replacement recombination the 5' crossover is not necessarily located within the homology region, thereby giving rise to a truncated gene product. Whether or not the replacement leads to such deletions is clearly dependent on the isotypes involved in the targeting reaction. The fact that the vector is correctly targeted to the heavy-chain locus, but that the homology region is not always the site of recombination, points to a novel recombination mechanism that may be specific for the immunoglobulin loci and that seems to be predominant even in the presence of the free homologous adjacent ends of an integration vector. Furthermore we demonstrate that homologous recombination at the heavy-chain locus is also possible between sequences from different species. The implications of our findings for the production of chimeric antibodies are discussed.
- Published
- 1994
48. Brauchen wir noch deutschsprachige Fachzeitschriften?
- Author
-
Wolfgang Haße and Ralph Mocikat
- Published
- 2014
49. Targeting membrane heat-shock protein 70 (Hsp70) on tumors by cmHsp70.1 antibody
- Author
-
Kathrin Hube, László Vígh, Wolfgang Sievert, Ralf Dressel, Ralph Mocikat, Julia Riegger, Arne Skerra, Gabriele Multhoff, Alan Graham Pockley, Isabelle Riederer, Lars Friedrich, Stefan Stangl, Elisabeth Kremmer, Kristin Kuhs, and Mathias Gehrmann
- Subjects
Interleukin 2 ,medicine.drug_class ,medicine.medical_treatment ,Cell ,Biology ,Monoclonal antibody ,Epitope ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,HSP70 Heat-Shock Proteins ,030304 developmental biology ,Antibody-dependent cell-mediated cytotoxicity ,0303 health sciences ,Mice, Inbred BALB C ,Multidisciplinary ,Macrophages ,Antibodies, Monoclonal ,Immunotherapy ,Biological Sciences ,Molecular biology ,3. Good health ,Protein Structure, Tertiary ,Killer Cells, Natural ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Epitope mapping ,Cholesterol ,030220 oncology & carcinogenesis ,biology.protein ,Interleukin-2 ,Cellular Cytotoxicity ,Epitope Mapping ,Surface Antigen ,Syngeneic Tumor Model ,Tumor Antibody Dependent ,Antibody ,Neoplasm Transplantation ,medicine.drug ,Granulocytes ,Protein Binding - Abstract
Immunization of mice with a 14-mer peptide TKDNNLLGRFELSG, termed “TKD,” comprising amino acids 450–461 (aa 450–461 ) in the C terminus of inducible Hsp70, resulted in the generation of an IgG1 mouse mAb cmHsp70.1. The epitope recognized by cmHsp70.1 mAb, which has been confirmed to be located in the TKD sequence by SPOT analysis, is frequently detectable on the cell surface of human and mouse tumors, but not on isogenic cells and normal tissues, and membrane Hsp70 might thus serve as a tumor-specific target structure. As shown for human tumors, Hsp70 is associated with cholesterol-rich microdomains in the plasma membrane of mouse tumors. Herein, we show that the cmHsp70.1 mAb can selectively induce antibody-dependent cellular cytotoxicity (ADCC) of membrane Hsp70 + mouse tumor cells by unstimulated mouse spleen cells. Tumor killing could be further enhanced by activating the effector cells with TKD and IL-2. Three consecutive injections of the cmHsp70.1 mAb into mice bearing CT26 tumors significantly inhibited tumor growth and enhanced the overall survival. These effects were associated with infiltrations of NK cells, macrophages, and granulocytes. The Hsp70 specificity of the ADCC response was confirmed by preventing the antitumor response in tumor-bearing mice by coinjecting the cognate TKD peptide with the cmHsp70.1 mAb, and by blocking the binding of cmHsp70.1 mAb to CT26 tumor cells using either TKD peptide or the C-terminal substrate-binding domain of Hsp70.
- Published
- 2010
50. Rapid Isolation of Immunoglobulin Variable Genes from Cell Lysates of Rat Hybridomas by Polymerase Chain Reaction
- Author
-
Ralph Mocikat, Christiane Harloff, and Gertrud Kütemeier
- Subjects
medicine.drug_class ,Molecular Sequence Data ,Immunology ,Immunoglobulin Variable Region ,DNA-Directed DNA Polymerase ,Biology ,Monoclonal antibody ,Polymerase Chain Reaction ,law.invention ,Mice ,chemistry.chemical_compound ,law ,Complementary DNA ,Genetics ,medicine ,Animals ,Taq Polymerase ,Amino Acid Sequence ,Cloning, Molecular ,Alleles ,Polymerase chain reaction ,Cloning ,Hybridomas ,Base Sequence ,Nucleic acid sequence ,Antibodies, Monoclonal ,DNA ,Molecular biology ,Rats ,chemistry ,Biochemistry ,Recombinant DNA ,biology.protein ,Antibody ,Taq polymerase - Abstract
The isolation of the rearranged immunoglobulin genes from a hybridoma cell line, which is a prerequisite for the construction of a recombinant antibody, can easily be achieved by polymerase chain reaction. Here we demonstrate that this method, which was originally described for cloning murine immunoglobulin genes from cDNA, is also applicable for rat genes. We show that the procedure also works with crude cell lysates as starting material, thereby greatly reducing the time required for sample preparation. In addition we have sequenced the nonfunctional heavy chain variable gene of the fusion partner X63Ag8.653, which was readily amplified from our hybridoma cells, and whose sequence has been so far unknown.
- Published
- 1992
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.