27 results on '"Vesna Pulko"'
Search Results
2. Src/lck inhibitor dasatinib reversibly switches off cytokine release and T cell cytotoxicity following stimulation with T cell bispecific antibodies
- Author
-
Christian Klein, Marina Bacac, Pablo Umana, Gabrielle Leclercq, Hélène Haegel, Anneliese Schneider, Anna Maria Giusti, Estelle Marrer-Berger, Christophe Boetsch, Antje-Christine Walz, Vesna Pulko, Johannes Sam, John Challier, Cristiano Ferlini, and Alex Odermatt
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background T cell engagers are bispecific antibodies recognizing, with one moiety, the CD3ε chain of the T cell receptor and, with the other moiety, specific tumor surface antigens. Crosslinking of CD3 upon simultaneous binding to tumor antigens triggers T cell activation, proliferation and cytokine release, leading to tumor cell killing. Treatment with T cell engagers can be associated with safety liabilities due to on-target on-tumor, on-target off-tumor cytotoxic activity and cytokine release syndrome (CRS). Tyrosine kinases such as SRC, LCK or ZAP70 are involved in downstream signaling pathways after engagement of the T cell receptor and blocking these kinases might serve to abrogate T cell activation when required (online supplemental material 1). Dasatinib was previously identified as a potent kinase inhibitor that switches off CAR T cell functionality.Methods Using an in vitro model of target cell killing by human peripheral blood mononuclear cells, we assessed the effects of dasatinib combined with 2+1 T cell bispecific antibodies (TCBs) including CEA-TCB, CD19-TCB or HLA-A2 WT1-TCB on T cell activation, proliferation and target cell killing measured by flow cytometry and cytokine release measured by Luminex. To determine the effective dose of dasatinib, the Incucyte system was used to monitor the kinetics of TCB-mediated target cell killing in the presence of escalating concentrations of dasatinib. Last, the effects of dasatinib were evaluated in vivo in humanized NSG mice co-treated with CD19-TCB. The count of CD20+ blood B cells was used as a readout of efficacy of TCB-mediated killing and cytokine levels were measured in the serum.Results Dasatinib concentrations above 50 nM prevented cytokine release and switched off-target cell killing, which were subsequently restored on removal of dasatinib. In addition, dasatinib prevented CD19-TCB-mediated B cell depletion in humanized NSG mice. These data confirm that dasatinib can act as a rapid and reversible on/off switch for activated T cells at pharmacologically relevant doses as they are applied in patients according to the label.Conclusion Taken together, we provide evidence for the use of dasatinib as a pharmacological on/off switch to mitigate off-tumor toxicities or CRS by T cell bispecific antibodies.
- Published
- 2021
- Full Text
- View/download PDF
3. CAR-J cells for antibody discovery and lead optimization of TCR-like immunoglobulins
- Author
-
Christian Jost, Diana Darowski, John Challier, Vesna Pulko, Lydia J Hanisch, Wei Xu, Ekkehard Mössner, Alexander Bujotzek, Stefan Klostermann, Pablo Umana, Roland E. Kontermann, and Christian Klein
- Subjects
Lead identification ,T-cell receptor (TCR) ,T-cell bispecific antibodies (TCB) ,TCR-like antibodies (TCRL) ,chimeric antigen receptor (CAR) ,major histocompatibility complex (MHC) ,Therapeutics. Pharmacology ,RM1-950 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
T-cell bispecific antibodies (TCBs) are a novel class of engineered immunoglobulins that unite monovalent binding to the T-cell receptor (TCR) CD3e chain and bivalent binding to tumor-associated antigens in order to recruit and activate T-cells for tumor cell killing. In vivo, T-cell activation is usually initiated via the interaction of the TCR with the peptide-HLA complex formed by the human leukocyte antigen (HLA) and peptides derived from intracellular proteins. TCR-like antibodies (TCRLs) that recognize pHLA-epitopes extend the target space of TCBs to peptides derived from intracellular proteins, such as those overexpressed during oncogenesis or created via mutations found in cancer. One challenge during lead identification of TCRL-TCBs is to identify TCRLs that specifically, and ideally exclusively, recognize the desired pHLA, but not unrelated pHLAs. In order to identify TCRLs suitable for TCRL-TCBs, large numbers of TCRLs have to be tested in the TCB format. Here, we propose a novel approach using chimeric antigen receptors (CARs) to facilitate the identification of highly selective TCRLs. In this new so-called TCRL-CAR-J approach, TCRL-candidates are transduced as CARs into Jurkat reporter-cells, and subsequently assessed for their specificity profile. This work demonstrates that the CAR-J reporter-cell assay can be applied to predict the profile of TCRL-TCBs without the need to produce each candidate in the final TCB format. It is therefore useful in streamlining the identification of TCRL-TCBs.
- Published
- 2020
- Full Text
- View/download PDF
4. Dysregulated TGF-β Production Underlies the Age-Related Vulnerability to Chikungunya Virus.
- Author
-
Jennifer L Uhrlaub, Vesna Pulko, Victor R DeFilippis, Rebecca Broeckel, Daniel N Streblow, Gary D Coleman, Byung S Park, John F Lindo, Ivan Vickers, Joshua J Anzinger, and Janko Nikolich-Žugich
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Chikungunya virus (CHIKV) is a re-emerging global pathogen with pandemic potential, which causes fever, rash and debilitating arthralgia. Older adults over 65 years are particularly susceptible to severe and chronic CHIKV disease (CHIKVD), accounting for >90% of all CHIKV-related deaths. There are currently no approved vaccines or antiviral treatments available to limit chronic CHIKVD. Here we show that in old mice excessive, dysregulated TGFβ production during acute infection leads to a reduced immune response and subsequent chronic disease. Humans suffering from CHIKV infection also exhibited high TGFβ levels and a pronounced age-related defect in neutralizing anti-CHIKV antibody production. In vivo reduction of TGFβ levels minimized acute joint swelling, restored neutralizing antibody production and diminished chronic joint pathology in old mice. This study identifies increased and dysregulated TGFβ secretion as one key mechanism contributing to the age-related loss of protective anti-CHIKV-immunity leading to chronic CHIKVD.
- Published
- 2016
- Full Text
- View/download PDF
5. A de-risking experimental framework defines the physiological interactome of TCR-based therapeutics in human tissues
- Author
-
Estelle Marrer-Berger, Annalisa Nicastri, Angelique Augustin, Vesna Pulko, Hanqing Liao, Lydia Duerner, Tina Wienzierl, Lauriane Cabon, Emmanuelle Lezan, Christian Klein, Pablo Umaña, Isaac Woodhouse, Alexander Bujotzek, and Nicola Ternette
- Abstract
Selective binding of TCR-based therapies that target a single tumour-specific peptide epitope presented by human leukocyte antigens (HLA) is the absolute prerequisite for their therapeutic suitability and patient safety. To date, selectivity assessment has been limited to peptide library screening and predictive computational modeling. We developed the first experimental platform to de novo identify interactomes of TCR-like molecules directly in human tissues using mass spectrometry. As proof of concept, we confirm the target epitope of a novel MAGE-A4-specific TCR-like antibody. We further determine 16 cross-reactive sequences for ESK1, a TCR-like bispecific antibody recognizing WT-1 with known off-target activity, in healthy liver tissue. We observe strong, off-target-induced T cell activation for 8/16 sequences, demonstrating the high specificity of the approach. Off-target sequences define a previously missed amino acid compensation motif that structurally mimics the target peptide groove coordination and allows for peptide interaction with the engager molecule. We establish the importance of the identified off-target activity by demonstrating 3D liver spheroid killing in the presence of ESK1 and healthy donor PBMC. Finally, we utilize our approach to de-risk our novel MAGE-A4 targeting TCR-like bispecific antibody prior to entering now ongoing clinical trials. We conclude that our strategy offers an accurate, scalable route for de-risking TCR-based therapeutics prior to first-in-human clinical application.
- Published
- 2022
- Full Text
- View/download PDF
6. JAK and mTOR inhibitors prevent cytokine release while retaining T cell bispecific antibody in vivo efficacy
- Author
-
Gabrielle Leclercq, Hélène Haegel, Alberto Toso, Tina Zimmermann, Luke Green, Nathalie Steinhoff, Johannes Sam, Vesna Pulko, Anneliese Schneider, Anna Maria Giusti, John Challier, Anne Freimoser-Grundschober, Laurent Larivière, Alex Odermatt, Martin Stern, Pablo Umana, Marina Bacac, and Christian Klein
- Subjects
Pharmacology ,Clinical/Translational Cancer Immunotherapy ,Cancer Research ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,MTOR Inhibitors ,cytokines ,Mice ,Oncology ,Antibodies, Bispecific ,Molecular Medicine ,Immunology and Allergy ,Animals ,Humans ,Janus Kinase Inhibitors ,immunotherapy ,RC254-282 ,T-lymphocytes - Abstract
BackgroundT cell engaging therapies, like chimeric antigen receptor T cells and T cell bispecific antibodies (TCBs), efficiently redirect T cells towards tumor cells, facilitating the formation of a cytotoxic synapse and resulting in subsequent tumor cell killing, a process that is accompanied by the release of cytokines. Despite their promising efficacy in the clinic, treatment with TCBs is associated with a risk of cytokine release syndrome (CRS). The aim of this study was to identify small molecules able to mitigate cytokine release while retaining T cell-mediated tumor killing.MethodsBy screening a library of 52 Food and Drug Administration approved kinase inhibitors for their impact on T cell proliferation and cytokine release after CD3 stimulation, we identified mTOR, JAK and Src kinases inhibitors as potential candidates to modulate TCB-mediated cytokine release at pharmacologically active doses. Using an in vitro model of target cell killing by human peripheral blood mononuclear cells, we assessed the effects of mTOR, JAK and Src kinase inhibitors combined with 2+1 T cell bispecific antibodies (TCBs) including CEA-TCB and CD19-TCB on T cell activation, proliferation and target cell killing measured by flow cytometry and cytokine release measured by Luminex. The combination of mTOR, JAK and Src kinase inhibitors together with CD19-TCB was evaluated in vivo in non-tumor bearing stem cell humanized NSG mice in terms of B cell depletion and in a lymphoma patient-derived xenograft (PDX) model in humanized NSG mice in terms of antitumor efficacy.ResultsThe effect of Src inhibitors differed from those of mTOR and JAK inhibitors with the suppression of CD19-TCB-induced tumor cell lysis in vitro, whereas mTOR and JAK inhibitors primarily affected TCB-mediated cytokine release. Importantly, we confirmed in vivo that Src, JAK and mTOR inhibitors strongly reduced CD19-TCB-induced cytokine release. In humanized NSG mice, continuous treatment with a Src inhibitor prevented CD19-TCB-mediated B cell depletion in contrast to mTOR and JAK inhibitors, which retained CD19-TCB efficacy. Ultimately, transient treatment with Src, mTOR and JAK inhibitors minimally interfered with antitumor efficacy in a lymphoma PDX model.ConclusionsTaken together, these data support further evaluation of the use of Src, JAK and mTOR inhibitors as prophylactic treatment to prevent occurrence of CRS.
- Published
- 2021
7. P09.01 The use of FDA approved JAK, mTOR and Src inhibitors to regulate T cell-bispecific antibody-induced cytokine release while not preventing T cell cytotoxicity
- Author
-
Anna Maria Giusti, A Toso, Helene Haegel, Nathalie Steinhoff, T Zimmermann, Luke Green, Gabrielle Leclercq, Marina Bacac, Christian Klein, P. Umana, Anneliese Schneider, Vesna Pulko, and Johannes Sam
- Subjects
business.industry ,medicine.medical_treatment ,T cell ,medicine.disease ,Dasatinib ,Cytokine release syndrome ,Cell killing ,Cytokine ,medicine.anatomical_structure ,Sirolimus ,medicine ,Cancer research ,business ,Tyrosine kinase ,Proto-oncogene tyrosine-protein kinase Src ,medicine.drug - Abstract
Background T cell bispecific antibodies (TCBs) are potent T cell engagers, harboring a 2+1 format with one binder to the CD3e chain and two binders to specific tumor antigens. Crosslinking of CD3 with tumor antigens triggers T cell activation and proliferation, cytokine release and tumor cell killing. TCB treatment is sometimes associated with safety liabilities due to on-target on-tumor or on-target off-tumor cytotoxicity and cytokine release. Off-tumor activity of the TCB may occur if the targeted tumor antigens are expressed on healthy cells, which may potentially result in tissue damages and compromise the patient’s safety. Patients treated with TCBs may also experience a Cytokine Release Syndrome (CRS), characterized by fever, hypotension and respiratory deficiency and associated with the release of pro-inflammatory cytokines such as IL-6, TNF-α, IFN-γ, and IL-1β. Tyrosine kinases such as Src, mTOR and JAK1/2 are involved in downstream signaling pathways after engagement of the T cell receptor. Materials and Methods 52 FDA approved kinase inhibitors were screened in the presence of T cells activated on CD3 coated plates, mimicking TCB stimulation. Src, mTOR and JAK inhibitors were selected based on their capacity to prevent both, cytokine release and T cell proliferation. Using an in vitro model of target cell killing by human peripheral blood mononuclear cells stimulated with TCBs, we validated the effects of mTOR, JAK and Src kinase inhibitors on TCB-induced T cell activation, tumor cell killing and cytokine release. In vivo, the effect of mTOR, JAK and Src kinase inhibitors on TCB-induced cytokine release was confirmed in humanized NOD scid gamma (NSG) mice engrafted with human hematopoietic stem cells and treated with CD19-TCB. Results In line with previous reports for CAR-T cells, dasatinib (a src inhibitor) was found to fully switch off TCB-induced T cell functionality as well as the other src inhibitors bosutinib and ponatinib. In contrast, temsirolimus, sirolimus and everolimus (mTOR inhibitors) and ruxolitinib, baricitinib, tofacitinib, and fedratinib (JAK1/2 inhibitors) were found to more potently prevent TCB-induced cytokine release without blocking TCB-mediated target cell killing. Conclusions These results provide evidence that the mechanisms of TCB-dependent cytokine release and tumor cell killing can be uncoupled. The FDA-approved mTOR and JAK1/2 inhibitors could potentially be used to mitigate CRS whereas the Src inhibitor dasatinib could rather stand as a potential antidote for on-target off-tumor activity or high-grade CRS. Disclosure Information G. Leclercq: A. Employment (full or part-time); Modest; Roche. E. Ownership Interest (stock, stock options, patent or other intellectual property); Modest; Roche. H. Haegel: A. Employment (full or part-time); Modest; Roche. E. Ownership Interest (stock, stock options, patent or other intellectual property); Modest; Roche. A. Schneider: A. Employment (full or part-time); Modest; Roche. A. Giusti: A. Employment (full or part-time); Modest; Roche. V. Pulko: A. Employment (full or part-time); Modest; Roche. A. Toso: A. Employment (full or part-time); Modest; Roche. T. Zimmermann: A. Employment (full or part-time); Modest; Roche. L. Green: A. Employment (full or part-time); Modest; Roche. N. Steinhoff: A. Employment (full or part-time); Modest; Roche. J. Sam: A. Employment (full or part-time); Modest; Roche. M. Bacac: A. Employment (full or part-time); Modest; Roche. E. Ownership Interest (stock, stock options, patent or other intellectual property); Modest; Roche. P. Umana: A. Employment (full or part-time); Modest; Roche. E. Ownership Interest (stock, stock options, patent or other intellectual property); Modest; Roche. C. Klein: A. Employment (full or part-time); Modest; Roche. E. Ownership Interest (stock, stock options, patent or other intellectual property); Modest; Roche.
- Published
- 2021
- Full Text
- View/download PDF
8. Src/lck inhibitor dasatinib reversibly switches off cytokine release and T cell cytotoxicity following stimulation with T cell bispecific antibodies
- Author
-
Anna Maria Giusti, Johannes Sam, Marina Bacac, Vesna Pulko, Cristiano Ferlini, Helene Haegel, Anneliese Schneider, Gabrielle Leclercq, John Challier, Estelle Marrer-Berger, Pablo Umana, Antje-Christine Walz, Christian Klein, Christophe Boetsch, and Alex Odermatt
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Dasatinib ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,Antibodies, Bispecific ,preclinical ,Immunology and Allergy ,Cytotoxic T cell ,immunologic ,RC254-282 ,Clinical/Translational Cancer Immunotherapy ,Chemistry ,ZAP70 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,drug therapy ,Cytokine release syndrome ,Cytokine ,medicine.anatomical_structure ,Cell killing ,Oncology ,030220 oncology & carcinogenesis ,Molecular Medicine ,cytotoxicity ,immunotherapy ,Tyrosine kinase ,medicine.drug ,T cell ,Immunology ,Receptors, Antigen, T-Cell ,Antineoplastic Agents ,03 medical and health sciences ,drug evaluation ,medicine ,Animals ,Humans ,t-lymphocytes ,lymphocyte activation ,Pharmacology ,combination ,medicine.disease ,cytokines ,030104 developmental biology ,inflammation ,Cancer research - Abstract
BackgroundT cell engagers are bispecific antibodies recognizing, with one moiety, the CD3ε chain of the T cell receptor and, with the other moiety, specific tumor surface antigens. Crosslinking of CD3 upon simultaneous binding to tumor antigens triggers T cell activation, proliferation and cytokine release, leading to tumor cell killing. Treatment with T cell engagers can be associated with safety liabilities due to on-target on-tumor, on-target off-tumor cytotoxic activity and cytokine release syndrome (CRS). Tyrosine kinases such as SRC, LCK or ZAP70 are involved in downstream signaling pathways after engagement of the T cell receptor and blocking these kinases might serve to abrogate T cell activation when required (online supplemental material 1). Dasatinib was previously identified as a potent kinase inhibitor that switches off CAR T cell functionality.MethodsUsing an in vitro model of target cell killing by human peripheral blood mononuclear cells, we assessed the effects of dasatinib combined with 2+1 T cell bispecific antibodies (TCBs) including CEA-TCB, CD19-TCB or HLA-A2 WT1-TCB on T cell activation, proliferation and target cell killing measured by flow cytometry and cytokine release measured by Luminex. To determine the effective dose of dasatinib, the Incucyte system was used to monitor the kinetics of TCB-mediated target cell killing in the presence of escalating concentrations of dasatinib. Last, the effects of dasatinib were evaluated in vivo in humanized NSG mice co-treated with CD19-TCB. The count of CD20+ blood B cells was used as a readout of efficacy of TCB-mediated killing and cytokine levels were measured in the serum.ResultsDasatinib concentrations above 50 nM prevented cytokine release and switched off-target cell killing, which were subsequently restored on removal of dasatinib. In addition, dasatinib prevented CD19-TCB-mediated B cell depletion in humanized NSG mice. These data confirm that dasatinib can act as a rapid and reversible on/off switch for activated T cells at pharmacologically relevant doses as they are applied in patients according to the label.ConclusionTaken together, we provide evidence for the use of dasatinib as a pharmacological on/off switch to mitigate off-tumor toxicities or CRS by T cell bispecific antibodies.
- Published
- 2021
9. CAR-J cells for antibody discovery and lead optimization of TCR-like immunoglobulins
- Author
-
John Challier, Lydia Jasmin Hanisch, Ekkehard Mössner, Diana Darowski, Wei Xu, Christian Jost, Alexander Bujotzek, Roland E. Kontermann, Pablo Umana, Christian Klein, Vesna Pulko, and Stefan Klostermann
- Subjects
major histocompatibility complex (MHC) ,Immunology ,Epitopes, T-Lymphocyte ,Human leukocyte antigen ,Jurkat cells ,Immunotherapy, Adoptive ,03 medical and health sciences ,Jurkat Cells ,0302 clinical medicine ,Antigen ,In vivo ,Antigens, Neoplasm ,Report ,Antibodies, Bispecific ,Immunology and Allergy ,Humans ,Receptor ,chimeric antigen receptor (CAR) ,030304 developmental biology ,Immunoassay ,0303 health sciences ,Receptors, Chimeric Antigen ,biology ,Chemistry ,T-cell receptor ,T-cell receptor (TCR) ,cellular assay ,Lead identification ,Chimeric antigen receptor ,Cell biology ,TCR-like antibodies (TCRL) ,human leukocyte antigen (HLA) ,T-cell bispecific antibodies (TCB) ,030220 oncology & carcinogenesis ,biology.protein ,Wilms tumor protein (WT1) ,Antibody - Abstract
T-cell bispecific antibodies (TCBs) are a novel class of engineered immunoglobulins that unite monovalent binding to the T-cell receptor (TCR) CD3e chain and bivalent binding to tumor-associated antigens in order to recruit and activate T-cells for tumor cell killing. In vivo, T-cell activation is usually initiated via the interaction of the TCR with the peptide-HLA complex formed by the human leukocyte antigen (HLA) and peptides derived from intracellular proteins. TCR-like antibodies (TCRLs) that recognize pHLA-epitopes extend the target space of TCBs to peptides derived from intracellular proteins, such as those overexpressed during oncogenesis or created via mutations found in cancer. One challenge during lead identification of TCRL-TCBs is to identify TCRLs that specifically, and ideally exclusively, recognize the desired pHLA, but not unrelated pHLAs. In order to identify TCRLs suitable for TCRL-TCBs, large numbers of TCRLs have to be tested in the TCB format. Here, we propose a novel approach using chimeric antigen receptors (CARs) to facilitate the identification of highly selective TCRLs. In this new so-called TCRL-CAR-J approach, TCRL-candidates are transduced as CARs into Jurkat reporter-cells, and subsequently assessed for their specificity profile. This work demonstrates that the CAR-J reporter-cell assay can be applied to predict the profile of TCRL-TCBs without the need to produce each candidate in the final TCB format. It is therefore useful in streamlining the identification of TCRL-TCBs.
- Published
- 2020
10. 653 Dasatinib as a rapid pharmacological ON/OFF switch for T cell bispecific antibody-induced T cell activation and cytokine release
- Author
-
Christophe Boetsch, Estelle Marrer Berger, Anneliese Schneider, Gabrielle Leclercq, Vesna Pulko, Antje Walz, Cristiano Ferlini, Christian Klein, and Helene Haegel
- Subjects
Chemistry ,medicine.medical_treatment ,ZAP70 ,T cell ,medicine.disease ,Dasatinib ,Cytokine release syndrome ,Cell killing ,Cytokine ,medicine.anatomical_structure ,Cancer research ,medicine ,Cytotoxic T cell ,Tyrosine kinase ,medicine.drug - Abstract
Background T cell bispecific antibodies (TCBs) are extremely potent T cell engagers, harboring a 2+1 format with one binder to the CD3e chain and two binders to specific tumor antigens. Crosslinking of CD3 with tumor antigens triggers T cell activation, proliferation and cytokine release, leading to tumor cell killing.1 2 TCB treatment is sometimes associated with safety liabilities due to on-target on-tumor, on-target off-tumor cytotoxic activity and cytokine release. Patients treated with TCBs may experience a Cytokine Release Syndrome (CRS), characterized by fever, hypotension and respiratory deficiency and associated with the release of pro-inflammatory cytokines such as IL-6, TNF-α, IFN-γ, and IL-1β.3 Off-tumor toxicity may occur if target antigens are expressed in healthy cells, which may potentially result in tissue damages and compromise the patient‘s safety. Rapid pharmacological blockade of T cell activation and proliferation is a promising approach to mitigate these life-threatening toxicities. Tyrosine kinases such as SRC, LCK or ZAP70 are involved in downstream signaling pathways after engagement of the T cell receptor and blocking these kinases might serve to abrogate T cell activation when required. Dasatinib was identified as a potent candidate that switches off CAR T cell functionality.4 5 Methods Using an in vitro model of target cell killing by human peripheral blood mononuclear cells, we assessed the reversible effects of dasatinib combined with CEA-TCB or HLA-A2-WT1-TCB on T cell activation and proliferation, target cell killing and cytokine release. At assay endpoints, T cell phenotype and target cell killing were measured by flow cytometry and supernatants were analyzed by Luminex to assess cytokine release. To determine the effective dose of dasatinib, the Incucyte system was used to follow kinetics of target cells killing by TCB in the presence of a dose response of dasatinib concentrations. Results 100 nM dasatinib prevented TCB-mediated target cell killing when added in the system upon restimulation of activated T cells (figure 1). Dasatinib concentrations above 50 nM fully switched off target cell killing (figure 2) which was restored upon removal of dasatinib. These data confirm that dasatinib act as a potent and reversible on/off switch for activated T cells at pharmacologically relevant doses as they are applied in patients according to the label.6 Conclusions Taken together, we provide evidence for the use of dasatinib as a pharmacological on/off switch to mitigate off-tumor toxicities or CRS by T cell engaging therapies. These data are being validated in vivo. References Bacac M, Fauti T, Sam J, Colombetti S, Weinzierl T, Ouaret D, et al. A novel carcinoembryonic antigen T-Cell Bispecific Antibody [CEA TCB] for the treatment of solid tumors. Clin Cancer Res 2016;22(13):3286–97. Bacac M, Klein C, Umana P. CEA TCB: A novel head-to-tail 2:1 T cell bispecific antibody for treatment of CEA-positive solid tumors. Oncoimmunology 2016;5(8):e1203498. Shimabukuro-Vornhagen A, Godel P, Subklewe M, Stemmler HJ, Schloser HA, Schlaak M, et al. Cytokine release syndrome. J Immunother Cancer 2018;6(1):56. Weber EW, Lynn RC, Sotillo E, Lattin J, Xu P, Mackall CL. Pharmacologic control of CAR-T cell function using dasatinib. Blood Advances 2019;3(5):711–7. Mestermann K, Giavridis T, Weber J, Rydzek J, Frenz S, Nerreter T, et al. The tyrosine kinase inhibitor dasatinib acts as a pharmacologic on/off switch for CAR T cells. Science Translational Medicine 2019;11(499):eaau5907. Wang X, Roy A, Hochhaus A, Kantarjian HM, Chen TT, Shah NP. Differential effects of dosing regimen on the safety and efficacy of dasatinib: retrospective exposure-response analysis of a Phase III study. Clinical pharmacology : advances and applications 2013;5:85–97.
- Published
- 2020
- Full Text
- View/download PDF
11. Dendritic cells dictate responses to PD-L1 blockade cancer immunotherapy
- Author
-
Mario Perro, Vaios Karanikas, Marcin Kowanetz, Andreas Roller, Christian Klein, Marieke F. Fransen, Ines Matos, Stefano Sammicheli, Stanford Chen, Eva Sum, Pablo Umana, Vesna Pulko, Christian Jost, Regula B. Buser, Daniel S. Chen, Priti S. Hegde, Ferry Ossendorp, Sara Colombetti, Maud Léa Mayoux, Wei Xu, Anton Belousov, Karolin Rommel, Pulmonary medicine, and CCA - Cancer biology and immunology
- Subjects
Lung Neoplasms ,biology ,business.industry ,medicine.medical_treatment ,T cell ,CD28 ,General Medicine ,Immunotherapy ,Dendritic Cells ,Antibodies, Monoclonal, Humanized ,B7-H1 Antigen ,medicine.anatomical_structure ,Cancer immunotherapy ,Atezolizumab ,PD-L1 ,Carcinoma, Non-Small-Cell Lung ,Blocking antibody ,biology.protein ,Cancer research ,Medicine ,Humans ,business ,CD80 - Abstract
PD-L1/PD-1 blocking antibodies have demonstrated therapeutic efficacy across a range of human cancers. Extending this benefit to a greater number of patients, however, will require a better understanding of how these therapies instigate anticancer immunity. Although the PD-L1/PD-1 axis is typically associated with T cell function, we demonstrate here that dendritic cells (DCs) are an important target of PD-L1 blocking antibody. PD-L1 binds two receptors, PD-1 and B7.1 (CD80). PD-L1 is expressed much more abundantly than B7.1 on peripheral and tumor-associated DCs in patients with cancer. Blocking PD-L1 on DCs relieves B7.1 sequestration in cis by PD-L1, which allows the B7.1/CD28 interaction to enhance T cell priming. In line with this, in patients with renal cell carcinoma or non-small cell lung cancer treated with atezolizumab (PD-L1 blockade), a DC gene signature is strongly associated with improved overall survival. These data suggest that PD-L1 blockade reinvigorates DC function to generate potent anticancer T cell immunity.
- Published
- 2020
- Full Text
- View/download PDF
12. Augmenting Efficacy of T-Cell Bispecific Antibodies in AML through a Tumor Stroma-Targeted 4-1BB Agonist
- Author
-
Nora Zieger, Anne-Sophie Neumann, Marion Subklewe, Alexandra Leutbecher, Vesna Pulko, Christina Claus, Anetta Marcinek, Pablo Umana, Veit Buecklein, Christian Klein, and Gerulf Hänel
- Subjects
Agonist ,Bispecific antibody ,medicine.anatomical_structure ,medicine.drug_class ,Chemistry ,T cell ,Immunology ,medicine ,Cancer research ,Cell Biology ,Hematology ,Tumor stroma ,Biochemistry - Abstract
Bispecific antibodies represent a promising treatment option for acute myeloid leukemia (AML). We have recently described a novel T-cell bispecific antibody (TCB) targeting the intracellular tumor antigen Wilms tumor 1 (WT1) in the context of HLA-A*02 (Augsberger et al. Blood 2021). Based on these findings a multicenter first-in-human clinical trial was initiated in relapse/refractory AML (NCT04580121). Possible immune escape mechanisms against T-cell based immunotherapy are provided by the tumor microenvironment (TME) of the bone marrow by co-inhibition of T cells or stromal cells shielding leukemic cells from immune effector cells. To overcome the immunosuppressive effect of the TME and to enhance T-cell responses, we evaluated the combination of the WT1-TCB with an antibody fusion protein that targets a stromal antigen (Fibroblast-activation protein; FAP) and provides a positive costimulatory signal (4-1BBL) to T cells. FAP is upregulated on cancer-associated fibroblasts after remodulation of the bone marrow niche by leukemic cells, and the FAP specificity of the molecule therefore provides T-cell co-stimulation tightly restricted to the tumor niche. Efficacy of the combination (WT1-TCB + FAP-4-1BBL antibody fusion protein) was evaluated in co-culture assays over 4 days with primary HLA-A*02 + AML cells, healthy donor (HD) T cells and three NIH-3T3 fibroblast cell lines. NIH-3T3 cell lines were genetically modified to express low and high levels of FAP, respectively. Wild-type NIH-3T3 cells were included as control. Additionally, a control (Ctrl)-TCB and a Ctrl-4-1BBL antibody fusion protein recognizing a non-tumor target derived from the human germline repertoire were included. Enhancement of T-cell mediated cytotoxicity by the FAP-4-1BBL antibody fusion protein was evaluated by (1) specific lysis of primary AML cells, (2) upregulation of the T-cell activation markers CD25 and 4-1BB, (3) T-cell expansion calculated as fold change compared to day 0, and (4) Granzyme B-expression which was evaluated by intracellular staining. After 4 days of co-culture, with an E:T ratio of 1:2, we observed a mean specific lysis of 55.1±8.2% (±SEM; n=4) of primary AML cells mediated by HD T cells and WT1-TCB. Notably, this was reduced to 19.4±5.9% (±SEM; n=4) in the presence of NIH-3T3 cells. However, AML cell lysis was restored by the addition of the FAP-4-1BBL antibody fusion protein in the presence of high FAP expressing NIH-3T3 cells (mean specific lysis: 62.8±7.3%; ±SEM; n=4). Concomitantly, the FAP-4-1BBL antibody fusion protein led to increased expression of the activation molecules CD25 (MFI ratio: 22.1±5.3 vs. 10.4±1.3; ±SEM; n=4) and 4-1BB (MFI ratio: 10.4±6.0 vs. 2.1±0.3; ±SEM; n=4) on CD3 + T cells. Furthermore, lysis was accompanied by increased frequencies of granzyme B expressing T cells (45.0±2.5% vs. 16.1±5.3%; n=3). Importantly, the FAP-4-1BBL antibody fusion protein led to improved T-cell proliferation, especially of CD8 + T cells (fold change on day 4 vs day 0: 5.7±2.2 vs. 1.0±0.3; ±SEM; n=4). Overall similar observations were made in the presence of low FAP expressing NIH-3T3 cells. Taken together, we have established an in vitro model system mimicking the immunoprotective bone marrow TME using NIH-3T3 cells resulting in impaired AML cell lysis. Providing additional T-cell co-stimulation by a tumor-stroma targeted 4-1BB agonist, however, restored WT1-TCB-mediated cytotoxicity of primary AML cells in the presence of FAP expressing cell lines. Importantly, the combination overcame the immunosuppressive effect of the NIH-3T3 cells on T cells as further demonstrated by improved T-cell activation and expansion. The tumor-stroma targeted 4-1BB agonist therefore represents a promising combinatorial approach to enhance T-cell activity at the local tumor site and warrants further investigations in an in vivo model system. Disclosures Pulko: Roche: Current Employment, Current equity holder in publicly-traded company, Patents & Royalties. Claus: Roche: Current Employment, Current equity holder in publicly-traded company, Patents & Royalties. Buecklein: Pfizer: Consultancy, Honoraria, Speakers Bureau; Kite/Gilead: Consultancy, Honoraria, Other: Congress and travel support, Research Funding; Novartis: Consultancy, Other: congress and travel support, Research Funding, Speakers Bureau; Miltenyi: Research Funding; BMS/Celgene: Consultancy, Research Funding; Amgen: Consultancy, Honoraria. Umana: Roche: Current Employment, Current equity holder in publicly-traded company, Patents & Royalties. Klein: Roche: Current Employment, Current equity holder in publicly-traded company, Patents & Royalties. Subklewe: Novartis: Consultancy, Research Funding, Speakers Bureau; Klinikum der Universität München: Current Employment; Roche: Research Funding; Seattle Genetics: Consultancy, Research Funding; Pfizer: Consultancy, Speakers Bureau; Janssen: Consultancy; Takeda: Speakers Bureau; MorphoSys: Research Funding; Miltenyi: Research Funding; Gilead: Consultancy, Research Funding, Speakers Bureau; Amgen: Consultancy, Research Funding, Speakers Bureau; BMS/Celgene: Consultancy, Research Funding, Speakers Bureau.
- Published
- 2021
- Full Text
- View/download PDF
13. Human memory T cells with a naive phenotype accumulate with aging and respond to persistent viruses
- Author
-
Michael S. Diamond, Kristy O. Murray, Carmine Martinez, Shripad Sinari, Erin C. Bush, Janko Nikolich-Žugich, Kenneth S. Knox, John Davies, Elias K. Haddad, Vesna Pulko, Peter A. Sims, Marion C. Lanteri, Michael P. Busch, Dean Billheimer, and Anne M. Wertheimer
- Subjects
Adult ,0301 basic medicine ,Aging ,Immunosenescence ,Immunology ,CD8-Positive T-Lymphocytes ,Biology ,Lymphocyte Activation ,Article ,Immunophenotyping ,Young Adult ,03 medical and health sciences ,Interleukin 21 ,0302 clinical medicine ,Immune system ,Antigen ,T-Lymphocyte Subsets ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Antigen-presenting cell ,Cells, Cultured ,Aged ,Aged, 80 and over ,CD28 ,Middle Aged ,Natural killer T cell ,Virology ,3. Good health ,Phenotype ,030104 developmental biology ,Virus Diseases ,Acute Disease ,Transcriptome ,Immunologic Memory ,CD8 ,030215 immunology - Abstract
The number of naive T cells decreases and susceptibility to new microbial infections increases with age. Here we describe a previously unknown subset of phenotypically naive human CD8(+) T cells that rapidly secreted multiple cytokines in response to persistent viral antigens but differed transcriptionally from memory and effector T cells. The frequency of these CD8(+) T cells, called 'memory T cells with a naive phenotype' (TMNP cells), increased with age and after severe acute infection and inversely correlated with the residual capacity of the immune system to respond to new infections with age. CD8(+) TMNP cells represent a potential new target for the immunotherapy of persistent infections and should be accounted for and subtracted from the naive pool if truly naive T cells are needed to respond to antigens.
- Published
- 2016
- Full Text
- View/download PDF
14. Role of Cell-Intrinsic and Environmental Age-Related Changes in Altered Maintenance of Murine T Cells in Lymphoid Organs
- Author
-
John Davies, Vesna Pulko, Janko Nikolich-Žugich, Jose Padilla Torres, and Heather L. Thompson
- Subjects
0301 basic medicine ,Male ,Aging ,Stromal cell ,Parabiosis ,Lymphoid Tissue ,T cell ,T-Lymphocytes ,Spleen ,03 medical and health sciences ,Mice ,medicine ,Cytotoxic T cell ,Animals ,Humans ,business.industry ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Lymphatic system ,The Journal of Gerontology: Biological Sciences ,Bone marrow ,Lymph Nodes ,Geriatrics and Gerontology ,business ,Peripheral lymph - Abstract
Age-related changes in primary lymphoid organs are well described. Less is known about age-related changes affecting peripheral lymphoid organs, although defects in old peripheral lymph nodes (pLNs) were recently described in both steady state and during viral infection. To address whether such pLN defects were intrinsic to old T cells or extrinsic (due to aging microenvironment), we employed heterochronic parabiosis. We found no age-related intrinsic or extrinsic barriers to T cell circulation and seeding of pLN, spleen, and bone marrow. However, heterochronic parabiosis failed to improve cellularity of old pLN, suggesting an environment-based limit on pLN cellularity. Furthermore, upon parabiosis, pLN of the adult partner exhibited reduced, old-like stromal and T cell cellularity, which was restored following separation of parabionts. Decay measurement of adult and old T cell subsets following separation of heterochronic parabionts delineated both T cell-intrinsic and environmental changes in T cell maintenance. Moreover, parabiotic separation revealed differences between CD4 and CD8 T cell subset maintenance with aging, the basis of which will require further investigation. Reasons for this asymmetric and subset-specific pattern of differential maintenance are discussed in light of possible age-related changes in lymph nodes as the key sites for peripheral T cell maintenance.
- Published
- 2017
15. Aging and Cytomegalovirus Infection Differentially and Jointly Affect Distinct Circulating T Cell Subsets in Humans
- Author
-
Dragana Nikolich-Žugich, Anne M. Wertheimer, Noreen Currier, Carmine Martinez, Jennifer L. Uhrlaub, Jeffrey Kaye, Byung Park, Janko Nikolich-Žugich, Vesna Pulko, and Michael S. Bennett
- Subjects
Adult ,CD4-Positive T-Lymphocytes ,Male ,Aging ,T cell ,Immunology ,Cytomegalovirus ,CD8-Positive T-Lymphocytes ,Biology ,medicine.disease_cause ,Article ,medicine ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Aged ,Aged, 80 and over ,Effector ,Middle Aged ,Intrinsic and extrinsic aging ,Titer ,medicine.anatomical_structure ,Cytomegalovirus Infections ,Female ,Immunologic Memory ,Memory T cell ,CD8 - Abstract
The impact of intrinsic aging upon human peripheral blood T cell subsets remains incompletely quantified and understood. This impact must be distinguished from the influence of latent persistent microorganisms, particularly CMV, which has been associated with age-related changes in the T cell pool. In a cross-sectional cohort of 152 CMV-negative individuals, aged 21–101 y, we found that aging correlated strictly to an absolute loss of naive CD8, but not CD4, T cells but, contrary to many reports, did not lead to an increase in memory T cell numbers. The loss of naive CD8 T cells was not altered by CMV in 239 subjects (range 21–96 y), but the decline in CD4+ naive cells showed significance in CMV+ individuals. These individuals also exhibited an absolute increase in the effector/effector memory CD4+ and CD8+ cells with age. That increase was seen mainly, if not exclusively, in older subjects with elevated anti-CMV Ab titers, suggesting that efficacy of viral control over time may determine the magnitude of CMV impact upon T cell memory, and perhaps upon immune defense. These findings provide important new insights into the age-related changes in the peripheral blood pool of older adults, demonstrating that aging and CMV exert both distinct and joint influence upon blood T cell homeostasis in humans.
- Published
- 2014
- Full Text
- View/download PDF
16. Targeting Intracellular WT1 in AML Utilizing a T Cell Bispecific Antibody Construct: Augmenting Efficacy through Combination with Lenalidomide
- Author
-
Johannes Sam, Wei Xu, Nikola P. Konstandin, Lydia Jasmin Hanisch, Marion Subklewe, Marc Schmitz, John Challier, Veit Bücklein, Vesna Pulko, Christian Augsberger, Christina Heitmüller, Michael von Bergwelt, Maurine Rothe, Anne Schönle, Antje Tunger, Karsten Spiekermann, Christian Klein, Gerulf Hänel, Alejandro Carpy, Felix S. Lichtenegger, and Pablo Umana
- Subjects
biology ,business.industry ,medicine.medical_treatment ,T cell ,Immunology ,Cell Biology ,Hematology ,Immunotherapy ,Biochemistry ,Jurkat cells ,Tumor antigen ,medicine.anatomical_structure ,Cancer immunotherapy ,Antigen ,Aldesleukin ,medicine ,biology.protein ,Cancer research ,Antibody ,business - Abstract
Antibody-based immunotherapy represents a promising strategy to target chemo-resistant leukemic cells. However, current antibody-based approaches are restricted to cell lineage surface antigens. Targeting intracellular antigens enables to enlarge the number of suitable tumor-associated target antigens with a more restricted expression profile. In this study we evaluated a 2+1 T Cell Bispecific (TCB) antibody for immunotherapy of acute myeloid leukemia (AML). The T cell receptor (TCR)-like TCB targets the intracellular tumor antigen Wilms tumor 1 (WT1) by bivalent recognition of the peptide RMFPNAPYL in the context of human leukocyte antigen allele A*02 (HLA-A2). Complementary binding to CD3ε recruits T cells irrespective of their TCR-specificity. We further analyzed enhancement of TCB-mediated T cell cytotoxicity through combination with the immune-modulatory drug lenalidomide. WT1 expression levels in cancer cell lines and primary AML patient samples at different time points during course of the disease were determined by quantitative real-time PCR, western blot and immunohistochemical staining. WT1-TCB-mediated cytotoxicity was analyzed by co-cultivation of WT1-expressing HLA-A2+ cancer cell lines with T cells from healthy donors. Specific lysis was assessed by flow cytometry. TCR downstream signaling was measured by co-cultivation of primary AML cells with NFAT Luciferase Reporter Jurkat cells. WT1-TCB-mediated cytotoxicity against primary AML cells and combination with 10 μM lenalidomide was evaluated in our pre-established feeder layer-based ex vivo long-term culture system. For in vivo testing, NSG mice (NOD.Cg-Prkdcscid-Il2rgtm1Wjl/SzJ) were humanized with human HLA-A2+ CD34+ cord blood cells. After successful engraftment and development of human T cells, WT1-expressing HLA-A2+ SKM-1 tumor cells were subcutaneously inoculated followed by weekly administration of the WT1-TCB. In accordance with previous reports, we observed WT1 expression in 79% (n=38) of cancer cell lines and in 92% (n=65) of AML patient samples at the time of initial diagnosis. Moreover, WT1 expression levels correlated with the percentage of AML blasts: no significant WT1 expression was observed at time of CR (n=26), whereas WT1 was expressed again at time of relapse (n=21). WT1-TCBs elicited antibody-mediated T cell cytotoxicity against peptide-pulsed T2 cells and AML cell lines in a WT1 and HLA-restricted manner. Equally, TCR downstream signaling was observed in a WT1-restrictive manner by co-cultivation of primary AML cells with NFAT Luciferase Reporter Jurkat cells. WT1-TCBs further mediated specific lysis of primary AML cells upon addition of allogenic T cells from healthy donors (mean specific lysis: 67±6% after 13-14 days; ±SEM; n=18). Correspondingly, up-regulation of T cell activation and surrogate exhaustion markers was observed (MFI fold change CD69: 9.3±1.5, PD-1: 5.1±0.7, TIM-3: 4.7±0.6; ±SEM; n=22). WT1-TCBs also mediated killing of primary AML cells in an autologous setting (mean specific lysis: 38±13% after 13-14 days; ±SEM; n=5). In comparison with WT1RMF-specific T cells, only bivalent binding by WT1-TCB induced efficient lysis of primary AML cells. Interestingly, combination of WT1-TCB with lenalidomide further enhanced antibody-mediated T-cell cytotoxicity against primary AML cells (mean specific lysis on day 3-4: 32±10% vs 59±9%; p=0.0017; ±SEM; n=13). This was accompanied by an increased secretion of the proinflammatory cytokines IL-2, IFN-γ and TNF-α and promoted the differentiation of naïve T cells towards a memory phenotype characterized by a downregulation of CD45RA. Furthermore, WT1-TCB-treated humanized mice bearing SKM-1 tumors showed a dose dependent and significant reduction in tumor growth resulting in tumor control. TCR-like TCBs targeting intracellular tumor antigens are a promising tool for cancer immunotherapy. Notably, the 2+1 TCB molecular format for bivalent binding facilitates potent in vitro, ex vivo and in vivo killing of AML cell lines and primary AML samples which present low numbers of the RMF peptide-MHC complex on the cell surface validating WT1-TCB as a promising therapeutic agent for the treatment of AML. Our results further indicate that the combinatorial approach with lenalidomide leads to increased TCB-mediated T cell cytotoxicity. Disclosures Klein: Roche: Employment, Equity Ownership, Patents & Royalties. Xu:Roche: Employment, Equity Ownership, Patents & Royalties. Heitmüller:Roche: Employment. Hanisch:Roche: Employment, Equity Ownership, Patents & Royalties. Sam:Roche: Employment, Equity Ownership, Patents & Royalties. Pulko:Roche: Employment, Equity Ownership, Patents & Royalties. Schönle:Roche: Employment, Equity Ownership, Patents & Royalties. Challier:Roche: Employment, Equity Ownership, Patents & Royalties. Carpy:Roche: Employment, Equity Ownership, Patents & Royalties. Lichtenegger:Roche: Employment. Umana:Roche: Employment, Equity Ownership, Patents & Royalties. Subklewe:Roche: Consultancy, Research Funding; Miltenyi: Research Funding; Oxford Biotherapeutics: Research Funding; Morphosys: Research Funding; Gilead: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria; AMGEN: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria; Janssen: Consultancy.
- Published
- 2019
- Full Text
- View/download PDF
17. PF207 TARGETING WILMS TUMOR 1 WITH A T CELL BISPECIFIC ANTIBODY (WT1-TCB): EX VIVO AND IN VIVO POTENCY BY BIVALENT RECOGNITION OF PEPTIDE-MHC COMPLEXES FROM AN INTRACELLULAR TUMOR ANTIGEN
- Author
-
Pablo Umana, Marc Schmitz, C. Heitmüller, Nikola P. Konstandin, John Challier, Karsten Spiekermann, Vesna Pulko, Christian Klein, Lydia Jasmin Hanisch, Christian Augsberger, M. Subklewe, Johannes Sam, Veit Bücklein, Antje Tunger, Alejandro Carpy, Wei Xu, M. von Bergwelt, Maurine Rothe, and Anne Schönle
- Subjects
Chemistry ,T cell ,Wilms' tumor ,Hematology ,medicine.disease ,Tumor antigen ,Bivalent (genetics) ,medicine.anatomical_structure ,In vivo ,medicine ,Cancer research ,Potency ,Ex vivo ,Intracellular - Published
- 2019
- Full Text
- View/download PDF
18. B7-H1 limits the entry of effector CD8+T cells to the memory pool by upregulating Bim
- Author
-
Eugene D. Kwon, Rachel M. Gibbons, Vesna Pulko, Christopher J. Krco, Susan M. Harrington, Xin Liu, and Haidong Dong
- Subjects
Programmed cell death ,Effector ,business.industry ,Immunology ,apoptosis ,tumor immunotherapy ,3. Good health ,Cell biology ,B7-H1 ,Oncology ,Downregulation and upregulation ,Apoptosis ,PD-1 ,metastasis ,Immunology and Allergy ,Cytotoxic T cell ,Medicine ,Receptor ,business ,CD80 ,CD8 ,Research Paper - Abstract
Protective T‑cell immunity against cancer and infections is dependent on the generation of a durable effector and memory T‑cell pool. Studies from cancer and chronic infections reveal that B7-H1 (PD-L1) engagement with its receptor PD-1 promotes apoptosis of effector T cells. It is not clear how B7-H1 regulates T‑cell apoptosis and the subsequent impact of B7-H1 on the generation of memory T cells. In immunized B7-H1-deficient mice, we detected an increased expansion of effector CD8(+) T cells and a delayed T‑cell contraction followed by the emergence of a protective CD8(+) T‑cell memory capable of completely rejecting tumor metastases in the lung. Intracellular staining revealed that antigen-primed CD8(+) T cells in B7-H1-deficient mice express lower levels of the pro-apoptotic molecule Bim. The engagement of activated CD8(+) T cells by a plate-bound B7-H1 fusion protein led to the upregulation of Bim and increased cell death. Assays based on blocking antibodies determined that both PD-1 and CD80 are involved in the B7-H1-mediated regulation of Bim in activated CD8(+) T cells. Our results suggest that B7-H1 may negatively regulate CD8(+) T‑cell memory by enhancing the depletion of effector CD8(+) T cells through the upregulation of Bim. Our findings may provide a new strategy for targeting B7-H1 signaling in effector CD8(+) T cells to achieve protective antitumor memory responses.
- Published
- 2012
- Full Text
- View/download PDF
19. B7-H1 Expressed by Activated CD8 T Cells Is Essential for Their Survival
- Author
-
Susan M. Harrington, Kimberley J. Harris, Rachel M. Gibbons, Vesna Pulko, Xin Liu, Haidong Dong, Christopher J. Krco, and Eugene D. Kwon
- Subjects
Cell Survival ,Immunology ,Apoptosis ,Mice, Transgenic ,Cell Separation ,CD8-Positive T-Lymphocytes ,Biology ,Lymphocyte Activation ,Article ,B7-H1 Antigen ,Mice ,Interleukin 21 ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Interleukin 3 ,ZAP70 ,CD28 ,Flow Cytometry ,Natural killer T cell ,Adoptive Transfer ,Molecular biology ,Mice, Inbred C57BL ,Female - Abstract
An immunoinhibitory role of B7 homologue 1 (B7-H1) expressed by non-T cells has been established; however, the function of B7-H1 expressed by T cells is not clear. Peak expression of B7-H1 on Ag-primed CD8 T cells was observed during the contraction phase of an immune response. Unexpectedly, B7-H1 blockade at this stage reduced the numbers of effector CD8 T cells, suggesting B7-H1 blocking Ab may disturb an unknown function of B7-H1 expressed by CD8 T cells. To exclusively examine the role of B7-H1 expressed by T cells, we introduced B7-H1 deficiency into TCR transgenic (OT-1) mice. Naive B7-H1–deficient CD8 T cells proliferated normally following Ag stimulation; however, once activated, they underwent more robust contraction in vivo and more apoptosis in vitro. In addition, B7-H1–deficient CD8 T cells were more sensitive to Ca-dependent and Fas ligand-dependent killing by cytotoxic T lymphocytes. Activation-induced Bcl-xL expression was lower in activated B7-H1–deficient CD8 T cells, whereas Bcl-2 and Bim expression were comparable to the wild type. Transfer of effector B7-H1–deficient CD8 T cells failed to suppress tumor growth in vivo. Thus, upregulation of B7-H1 on primed T cells helps effector T cells survive the contraction phase and consequently generate optimal protective immunity.
- Published
- 2011
- Full Text
- View/download PDF
20. TLR3-Stimulated Dendritic Cells Up-regulate B7-H1 Expression and Influence the Magnitude of CD8 T Cell Responses to Tumor Vaccination
- Author
-
Eugene D. Kwon, Xin Liu, Haidong Dong, Xavier Frigola, Christopher J. Krco, Vesna Pulko, and Kimberley J. Harris
- Subjects
Effector ,medicine.medical_treatment ,T cell ,Immunology ,Priming (immunology) ,Immunotherapy ,Biology ,Blockade ,medicine.anatomical_structure ,TLR3 ,medicine ,Cancer research ,Immunology and Allergy ,Cytotoxic T cell ,B7-H1 Antigen - Abstract
Agonists of TLR have been explored as vaccine adjuvants for tumor immunotherapy. However, their immunological consequences are not fully understood. Although TLR signaling increases the functional potential of dendritic cells (DCs) for priming T cells, coinduction of potentially negative immunoregulatory capacities may impair effector T cell generation. We examined the expression and function of B7 family costimulatory molecules on DCs after activation with the TLR3 agonist, polyinosinic:polycytidylic acid. We demonstrated that polyinosinic:polycytidylic acid consistently up-regulated both B7-2 and B7-H1 molecules on resident, migratory DCs from spleen and lymph nodes. Depletion or blockade of B7-H1 on activated DCs increased the magnitude of effector CD8 T cell expansion. DC-based or protein-based tumor vaccines, in combination with B7-H1 blockade, induced strong effector CD8 T cell responses, resulting in protective immunity against newly established tumors. Our studies suggest that TLR3 signaling has the potential to up-regulate both positive and negative coregulatory molecules on APCs. Selective blockade of negative regulatory molecules in combination with TLR3 agonist may be an effective strategy for increasing the efficacy of tumor vaccines.
- Published
- 2009
- Full Text
- View/download PDF
21. Abstract 3658: Dendritic cells dictate the responsiveness of PD-L1 blockade in cancer
- Author
-
Christian Klein, Ines Matos, Maud Léa Mayoux, Wei Xu, Ferry Ossendorp, Vesna Pulko, Marieke F. Fransen, Pablo Umana, Vaios Karanikas, and Andreas Roller
- Subjects
0301 basic medicine ,Cancer Research ,biology ,business.industry ,T cell ,Priming (immunology) ,CD11c ,Blockade ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Immune system ,Oncology ,Atezolizumab ,PD-L1 ,Immunology ,medicine ,biology.protein ,business ,Lymph node - Abstract
Recent advances in cancer immunotherapies with PD-1/PD-L1 pathway blockade have transformed the way that cancer is being treated, leading to durable responses and prolonged overall survival. The general thinking is that PD-1/PD-L1 blockade reinvigorates tumor-infiltrating PD-1+ T cells with exhausted phenotypes. However, a mechanistic understanding on why only a subset of patients (10-30%) responds to checkpoint inhibition remains largely unknown, as does the exact immune mechanism of PD-1/PD-L1 blockade. Here, we discovered that PD-L1 blockade mediates anti-tumor immunity via dendritic cells. In human blood, in vitro and ex vivo dendritic cells (DCs) express both PD-1 and PD-L1, and an activation signal via TLR agonists triggers downregulation of PD-1 to empower the T cell stimulatory capability of DCs. In contrast, tolerogenic DCs remain PD-1 positive, correlating to T cell-unresponsiveness. Anti-PD-L1 Ab directly induces maturation of DCs and renders them capable of stimulating T cell proliferation. Similarly, anti-PD-L1 Ab treatment in tumor-bearing mice induces massive infiltration and activation of CD11c+ DCs in the spleen and draining lymph node, indicating that PD-1 is a negative regulator in DCs. Furthermore, ablation of DCs prior to anti-PD-L1 Ab treatment in an established MC38 tumor model in CD11c-DTR mice suggests a crucial role of DCs in mediating response to PD-L1 treatment. In support of the preclinical evidence that DCs are the primary target of anti-PD-L1 Ab, we analyzed RNA-seq data from tumor biopsies at baseline in patients with renal cell carcinoma prior to treatment with atezolizumab and found that the abundance of genes related to cross-presenting DC subsets (such as XCR1) correlates with a survival advantage in response to atezolizumab (HR=0.13, median OS is 16.2 months in patients with DC gene score 50%). In conclusion, we discovered abundance of tumor-infiltrating DCs as a novel biomarker that can predict the clinical response of PD-L1 blockade. We postulate that checkpoint inhibition directly on tumor-infiltrating DCs likely contributes to amplification of Ag-specific priming of tumor-specific T cells. Citation Format: Maud Mayoux, Marieke F. Fransen, Andreas Roller, Ines Matos, Vesna Pulko, Vaios Karanikas, Pablo Umana, Christian Klein, Ferry A. Ossendorp, Wei Xu. Dendritic cells dictate the responsiveness of PD-L1 blockade in cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3658. doi:10.1158/1538-7445.AM2017-3658
- Published
- 2017
- Full Text
- View/download PDF
22. Induction of a Th1 response from Th2-polarized T cells by activated dendritic cells: dependence on TCR:peptide-MHC interaction, ICAM-1, IL-12, and IFN-gamma
- Author
-
William A. Faubion, Karla R. Wiehagen, Suresh Radhakrishnan, Larry R. Pease, Virginia P. Van Keulen, Vesna Pulko, and Keith L. Knutson
- Subjects
T cell ,Immunology ,Receptors, Antigen, T-Cell ,Receptors, Cell Surface ,Biology ,Interferon-gamma ,Mice ,Signaling lymphocytic activation molecule ,Th2 Cells ,Adjuvants, Immunologic ,Signaling Lymphocytic Activation Molecule Family Member 1 ,Gardiquimod ,Antigens, CD ,Histocompatibility Antigens ,medicine ,Immunology and Allergy ,Animals ,Secretion ,Immunologic Capping ,Cells, Cultured ,Mice, Knockout ,ICAM-1 ,Antigen Presentation ,Effector ,T-cell receptor ,Toll-Like Receptors ,Antibodies, Monoclonal ,Dendritic Cells ,Th1 Cells ,Intercellular Adhesion Molecule-1 ,Interleukin-12 ,Cell biology ,medicine.anatomical_structure ,Immune System Diseases ,Interleukin 12 ,B7-1 Antigen ,CpG Islands ,Peptides ,Protein Binding - Abstract
Dendritic cells (DC) are important regulators of T cell immunity. The degree of stimulation, the pattern of costimulatory molecules expressed, and the cytokines secreted by DC dictate the nature of the effector and memory cells generated, particularly with respect to their Th1 or Th2 phenotypes. In this study, we demonstrate that the addition of activated DC to spleen cultures containing established Th2-polarized CD4+ T cells was sufficient to suppress Th2 and induce Th1 cytokines in a recall response, a phenomenon referred to as phenotype reversal. The ability of activated DC to induce phenotype reversal displayed exquisite Ag specificity. The DC activator B7-DC cross-linking Ab (XAb) was >10,000-fold more efficient at inducing phenotype reversal than the TLR agonists CpG-oligodeoxynucleotide and Gardiquimod. Characterization of the mechanisms governing phenotype reversal revealed the requirement for cognate interaction between the TCR:peptide-MHC complex, the expression of the costimulation/adhesion molecule ICAM-1, and secretion of IL-12 and IFN-γ by the activated DC. The requirement for the costimulation/adhesion molecule SLAM (signaling lymphocytic activation molecule) was found to be quantitative. Thus, activation of DC, particularly by crosslinking B7-DC, can modulate well-established Th2 T cell responses in an Ag-specific manner. Because the regulation of mouse and human DC by B7-DC XAb overlaps in several significant ways, immune modulation with B7-DC XAb is a potential strategy for treating Th2-mediated diseases.
- Published
- 2007
23. Heterochronic parabiosis: allowing the dissection of the aged immune system (LYM2P.723)
- Author
-
John Davies, Vesna Pulko, Heather Thompson, and Janko Nikolich-Zugich
- Subjects
Immunology ,Immunology and Allergy - Abstract
Parabiosis is the surgical union of two organisms resulting in the development of a single, shared circulatory system. When animals of different ages are conjoined (i.e. heterochronic parabiosis), blood-borne factors from the younger animal can often beneficially affect the older animal and recapitulate the youthful phenotype & function in target tissues. However, the effects of heterochronic parabiosis on the aged immune system remain unexplored. An important question to be answered is whether the cellular defects involved in the aged immune system are due to intrinsic deficiencies or if they can be rescued by extrinsic factors. Also, T cell ontogenesis is known to diminish with age due to thymic involution as well as defects in lymphopoiesis that affect pre-T cells. The heterochronic parabiosis model allows testing of contributions of the aged thymus, pre-T cells and of systemic environment in immune aging. The preliminary data demonstrate differential migration patterns of T cells, B cells & dendritic cells as a function of age. Of interest, pre-T cells from either animal adequately seed the opposing thymus and undergo thymopoiesis and other data suggest that there are no intrinsic defects in old thymocytes, but rather of the old thymic environment. That latter defect may not be corrected by heterochronic parabiosis.
- Published
- 2015
- Full Text
- View/download PDF
24. Retraction: Induction of a Th1 Response from Th2-Polarized T Cells by Activated Dendritic Cells: Dependence on TCR:Peptide-MHC Interaction, ICAM-1, IL-12, and IFN-γ
- Author
-
Keith L. Knutson, William A. Faubion, Virginia P. Van Keulen, Karla R. Wiehagen, Vesna Pulko, and Larry R. Pease
- Subjects
ICAM-1 ,Chemistry ,Immunology ,T-cell receptor ,Interleukin 12 ,Immunology and Allergy ,Peptide-MHC ,Th1 response ,Molecular biology ,humanities - Abstract
We wish to retract the article titled “Induction of a Th1 Response from Th2-Polarized T Cells by Activated Dendritic Cells: Dependence on TCR:Peptide-MHC Interaction, ICAM-1, IL-12, and IFN-γ,” by Suresh Radhakrishnan, Karla R. Wiehagen, Vesna Pulko, Virginia Van Keulen, William A. Faubion
- Published
- 2010
- Full Text
- View/download PDF
25. T cell-intrinsic B7-homolog-1 (B7-H1) regulates effector CD8 T cell survival and memory cell generation (83.13)
- Author
-
Haidong Dong, Vesna Pulko, Xin Liu, and Kimberley Harris
- Subjects
Immunology ,Immunology and Allergy - Abstract
B7 homolog-1 ( B7-H1) expressed by antigen-presenting cells or tumor cells is a negative regulator of T cell responses, however its function on T cells is not clear. Here we show that B7-H1 was upregulated in CD8 T cells during immunization and its expression was maintained on memory CD8 T cells. Although B7-H1 deficient naïve CD8 T cells responded normally to antigen stimulation and underwent normal antigen-independent homeostatic proliferation, the maximal expansion of CD8 T cells was blocked due to their high susceptibility to apoptosis. B7-H1-deficient effector CD8 T cells showed weaker effector function and cytotoxicity against tumor cells compared to that of normal CD8 T cells. Once activated, B7-H1-deficient CD8 T cells generated less memory T cells. This effect was independent of the primary expansion of the antigen-specific CD8 T cell population. Our results revealed previously unknown function of B7-H1 on CD8 T cells in mediating survival and differentiation of effector CD8 T cells into memory T cells.
- Published
- 2009
- Full Text
- View/download PDF
26. Induction of a Th1 Response from Th2 Polarized T Cells by Activated Dendritic Cells: Dependence on TCR:pMHC Interaction, ICAM-1, IL-12 (92.7)
- Author
-
Suresh Radhakrishnan, Karla R Wiehagen, Vesna Pulko, Keith Knutson, Faubion WIlliam, and Larry R Pease
- Subjects
Immunology ,Immunology and Allergy - Abstract
Dendritic cells (DC) are important regulators of T cell immunity. The degree of stimulation, the pattern of co-stimulatory molecules expressed, and the cytokines secreted by DC dictate the nature of the effector and memory cells generated, particularly with respect to their Th1 or Th2 phenotypes. In this report we demonstrate that addition of activated DC to spleen cultures containing established Th2 polarized CD4+ T cells was sufficient to suppress Th2 and to induce Th1 cytokines in a recall response, a phenomenon referred to as phenotype reversal. The ability of activated DC to induce phenotype reversal displayed exquisite antigen specificity. The DC activator B7-DC XAb was more than 10,000-fold more efficient at inducing phenotype reversal than the TLR agonists, CpG-ODN and Gardiquimod. Characterization of the mechanisms governing phenotype reversal revealed the requirement for cognate interaction between the TCR: pMHC complex, the expression of the co-stimulation/adhesion molecule ICAM-1, and secretion of IL-12 and IFNƒ×ƒnby the activated DC. Requirement for the co-stimulation/adhesion molecule SLAM was found to be quantitative. Thus, activation of DC, particularly by cross linking B7-DC, can modulate well established Th2 T cell responses in an antigen specific manner.
- Published
- 2007
- Full Text
- View/download PDF
27. Targeting intracellular WT1 in AML with a novel RMF-peptide-MHC specific T-cell bispecific antibody
- Author
-
Pier Edoardo Rovatti, Antje-Christine Walz, Christian Gassner, Pablo Umana, Michael von Bergwelt-Baildon, Nikola P. Konstandin, Angélique Augustin, Binje Vick, John Challier, Estelle Marrer-Berger, Wei Xu, Jigar Patel, Marc Schmitz, Christina Krupka, Jörg Benz, Lydia Jasmin Hanisch, Karsten Spiekermann, Gerulf Hänel, Christian Klein, Maurine Rothe, Vesna Pulko, Antje Tunger, Felix S Lichtenegger, Katharina Hunt, Alejandro Carpy, Emmanuelle Lezan, Axel Ducret, Alexander Bujotzek, Christian Augsberger, Marion Subklewe, Daniela Ortiz-Franyuti, Anne Schönle, Victor Lyamichev, Luca Vago, Irmela Jeremias, Johannes Sam, Augsberger, C., Hanel, G., Xu, W., Pulko, V., Hanisch, L. J., Augustin, A., Challier, J., Hunt, K., Vick, B., Rovatti, P. E., Krupka, C., Rothe, M., Schonle, A., Sam, J., Lezan, E., Ducret, A., Ortiz-Franyuti, D., Walz, A. -C., Benz, J., Bujotzek, A., Lichtenegger, F. S., Gassner, C., Carpy, A., Lyamichev, V., Patel, J., Konstandin, N., Tunger, A., Schmitz, M., von Bergwelt-Baildon, M., Spiekermann, K., Vago, L., Jeremias, I., Marrer-Berger, E., Umana, P., Klein, C., and Subklewe, M.
- Subjects
medicine.medical_treatment ,T cell ,Immunology ,Receptors, Antigen, T-Cell ,Biochemistry ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Antigen ,Cell Line, Tumor ,Antibodies, Bispecific ,HLA-A2 Antigen ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,Cytotoxicity ,WT1 Proteins ,030304 developmental biology ,0303 health sciences ,biology ,Chemistry ,Cell Biology ,Hematology ,Immunotherapy ,Tumor antigen ,3. Good health ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Antibody ,Clone (B-cell biology) ,Peptides ,Ex vivo ,T-Lymphocytes, Cytotoxic - Abstract
Antibody-based immunotherapy is a promising strategy for targeting chemoresistant leukemic cells. However, classical antibody-based approaches are restricted to targeting lineage-specific cell surface antigens. By targeting intracellular antigens, a large number of other leukemia-associated targets would become accessible. In this study, we evaluated a novel T-cell bispecific (TCB) antibody, generated by using CrossMAb and knob-into-holes technology, containing a bivalent T-cell receptor–like binding domain that recognizes the RMFPNAPYL peptide derived from the intracellular tumor antigen Wilms tumor protein (WT1) in the context of HLA-A*02. Binding to CD3ε recruits T cells irrespective of their T-cell receptor specificity. WT1-TCB elicited antibody-mediated T-cell cytotoxicity against AML cell lines in a WT1- and HLA-restricted manner. Specific lysis of primary acute myeloid leukemia (AML) cells was mediated in ex vivo long-term cocultures by using allogeneic (mean ± standard error of the mean [SEM] specific lysis, 67 ± 6% after 13-14 days; n = 18) or autologous, patient-derived T cells (mean ± SEM specific lysis, 54 ± 12% after 11-14 days; n = 8). WT1-TCB–treated T cells exhibited higher cytotoxicity against primary AML cells than an HLA-A*02 RMF-specific T-cell clone. Combining WT1-TCB with the immunomodulatory drug lenalidomide further enhanced antibody-mediated T-cell cytotoxicity against primary AML cells (mean ± SEM specific lysis on days 3-4, 45.4 ± 9.0% vs 70.8 ± 8.3%; P = .015; n = 9-10). In vivo, WT1-TCB–treated humanized mice bearing SKM-1 tumors exhibited a significant and dose-dependent reduction in tumor growth. In summary, we show that WT1-TCB facilitates potent in vitro, ex vivo, and in vivo killing of AML cell lines and primary AML cells; these results led to the initiation of a phase 1 trial in patients with relapsed/refractory AML (#NCT04580121).
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.