26 results on '"Nicole Kirchhammer"'
Search Results
2. Deletion of SNX9 alleviates CD8 T cell exhaustion for effective cellular cancer immunotherapy
- Author
-
Marcel P. Trefny, Nicole Kirchhammer, Priska Auf der Maur, Marina Natoli, Dominic Schmid, Markus Germann, Laura Fernandez Rodriguez, Petra Herzig, Jonas Lötscher, Maryam Akrami, Jane C. Stinchcombe, Michal A. Stanczak, Andreas Zingg, Melanie Buchi, Julien Roux, Romina Marone, Leyla Don, Didier Lardinois, Mark Wiese, Lukas T. Jeker, Mohamed Bentires-Alj, Jérémie Rossy, Daniela S. Thommen, Gillian M. Griffiths, Heinz Läubli, Christoph Hess, and Alfred Zippelius
- Subjects
Science - Abstract
The efficacy of T-cell-based cancer immunotherapies can be compromised by T cell exhaustion. Here the authors develop a human ex vivo exhaustion model and, based on a CRISPR-Cas9 screen, identify SNX9 as a regulator of T cell exhaustion, showing that SNX9 knockout is associated with improved T cell function and anti-tumor activity in preclinical cancer models.
- Published
- 2023
- Full Text
- View/download PDF
3. Dual TLR9 and PD-L1 targeting unleashes dendritic cells to induce durable antitumor immunity
- Author
-
Alfred Zippelius, Mélanie Buchi, Richard Klar, Julia Festag, Sven Michel, Frank Jaschinski, Sebastian Kobold, Abhishek S Kashyap, Johannes vom Berg, Laura Fernandez-Rodriguez, Chiara Cianciaruso, Ruben Bill, Marcel P Trefny, Nicole Kirchhammer, Rainer H Kohler, Elham Jones, Andre Maaske, Karen O Dixon, and Mikael J Pittet
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Although immune checkpoint inhibitors have been a breakthrough in clinical oncology, these therapies fail to produce durable responses in a significant fraction of patients. This lack of long-term efficacy may be due to a poor pre-existing network linking innate and adaptive immunity. Here, we present an antisense oligonucleotide (ASO)-based strategy that dually targets toll-like receptor 9 (TLR9) and programmed cell death ligand 1 (PD-L1), aiming to overcome resistance to anti-PD-L1 monoclonal therapy.Methods We designed a high-affinity immunomodulatory IM-TLR9:PD-L1-ASO antisense oligonucleotide (hereafter, IM-T9P1-ASO) targeting mouse PD-L1 messenger RNA and activating TLR9. Then, we performed in vitro and in vivo studies to validate the IM-T9P1-ASO activity, efficacy, and biological effects in tumors and draining lymph nodes. We also performed intravital imaging to study IM-T9P1-ASO pharmacokinetics in the tumor.Results IM-T9P1-ASO therapy, unlike PD-L1 antibody therapy, results in durable antitumor responses in multiple mouse cancer models. Mechanistically, IM-T9P1-ASO activates a state of tumor-associated dendritic cells (DCs), referred to here as DC3s, which have potent antitumor potential but express the PD-L1 checkpoint. IM-T9P1-ASO has two roles: it triggers the expansion of DC3s by engaging with TLR9 and downregulates PD-L1, thereby unleashing the antitumor functions of DC3s. This dual action leads to tumor rejection by T cells. The antitumor efficacy of IM-T9P1-ASO depends on the antitumor cytokine interleukin-12 (IL-12), produced by DC3s, and Batf3, a transcription factor required for DC development.Conclusions By simultaneously targeting TLR9 and PD-L1, IM-T9P1-ASO amplifies antitumor responses via DC activation, leading to sustained therapeutic efficacy in mice. By highlighting differences and similarities between mouse and human DCs, this study could serve to develop similar therapeutic strategies for patients with cancer.
- Published
- 2023
- Full Text
- View/download PDF
4. iMATCH: an integrated modular assembly system for therapeutic combination high-capacity adenovirus gene therapy
- Author
-
Dominik Brücher, Nicole Kirchhammer, Sheena N. Smith, Jatina Schumacher, Nina Schumacher, Jonas Kolibius, Patrick C. Freitag, Markus Schmid, Fabian Weiss, Corina Keller, Melanie Grove, Urs F. Greber, Alfred Zippelius, and Andreas Plückthun
- Subjects
high-capacity vectors ,helper-dependent adenovirus ,gutless adenovirus ,gene therapy ,cancer therapy ,immunotherapy ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Adenovirus-mediated combination gene therapies have shown promising results in vaccination or treating malignant and genetic diseases. Nevertheless, an efficient system for the rapid assembly and incorporation of therapeutic genes into high-capacity adenoviral vectors (HCAdVs) is still missing. In this study, we developed the iMATCH (integrated modular assembly for therapeutic combination HCAdVs) platform, which enables the generation and production of HCAdVs encoding therapeutic combinations in high quantity and purity within 3 weeks. Our modular cloning system facilitates the efficient combination of up to four expression cassettes and the rapid integration into HCAdV genomes with defined sizes. Helper viruses (HVs) and purification protocols were optimized to produce HCAdVs with distinct capsid modifications and unprecedented purity (0.1 ppm HVs). The constitution of HCAdVs, with adapters for targeting and a shield of trimerized single-chain variable fragment (scFv) for reduced liver clearance, mediated cell- and organ-specific targeting of HCAdVs. As proof of concept, we show that a single HCAdV encoding an anti PD-1 antibody, interleukin (IL)-12, and IL-2 produced all proteins, and it led to tumor regression and prolonged survival in tumor models, comparable to a mixture of single payload HCAdVs in vitro and in vivo. Therefore, the iMATCH system provides a versatile platform for the generation of high-capacity gene therapy vectors with a high potential for clinical development.
- Published
- 2021
- Full Text
- View/download PDF
5. A novel rabbit derived anti-HER2 antibody with pronounced therapeutic effectiveness on HER2-positive breast cancer cells in vitro and in humanized tumor mice (HTM)
- Author
-
Anja Kathrin Wege, Nicole Kirchhammer, Linda Veronique Kazandjian, Sandra Prassl, Michael Brandt, Gerhard Piendl, Olaf Ortmann, Stephan Fischer, and Gero Brockhoff
- Subjects
Anti-HER2 antibody ,Breast cancer ,Treatment efficiency ,Humanized tumor mice ,Monoclonal antibody ,Medicine - Abstract
Abstract Background Antibody based cancer therapies have achieved convincing success rates combining enhanced tumor specificity and reduced side effects in patients. Trastuzumab that targets the human epidermal growth factor related receptor 2 (HER2) is one of the greatest success stories in this field. For decades, trastuzumab based treatment regimens are significantly improving the prognosis of HER2-positive breast cancer patients both in the metastatic and the (neo-) adjuvant setting. Nevertheless, ≥ 50% of trastuzumab treated patients experience de-novo or acquired resistance. Therefore, an enhanced anti-HER2 targeting with improved treatment efficiency is still aspired. Methods Here, we determined cellular and molecular mechanisms involved in the treatment of HER2-positive BC cells with a new rabbit derived HER2 specific chimeric monoclonal antibody called “B100″. We evaluated the B100 treatment efficiency of HER2-positive BC cells with different sensitivity to trastuzumab both in vitro and in the presence of a human immune system in humanized tumor mice. Results B100 not only efficiently blocks cell proliferation but more importantly induces apoptotic tumor cell death. Detailed in vitro analyses of B100 in comparison to trastuzumab (and pertuzumab) revealed equivalent HER2 internalization and recycling capacity, similar Fc receptor signaling, but different HER2 epitope recognition with high binding and treatment efficiency. In trastuzumab resistant SK-BR-3 based humanized tumor mice the B100 treatment eliminated the primary tumor but even more importantly eradicated metastasized tumor cells in lung, liver, brain, and bone marrow. Conclusion Overall, B100 demonstrated an enhanced anti-tumor activity both in vitro and in an enhanced preclinical HTM in vivo model compared to trastuzumab or pertuzumab. Thus, the use of B100 is a promising option to complement and to enhance established treatment regimens for HER2-positive (breast) cancer and to overcome trastuzumab resistance. Extended preclinical analyses using appropriate models and clinical investigations are warranted.
- Published
- 2020
- Full Text
- View/download PDF
6. Siglec Receptors Modulate Dendritic Cell Activation and Antigen Presentation to T Cells in Cancer
- Author
-
Jinyu Wang, Michela Manni, Anne Bärenwaldt, Ronja Wieboldt, Nicole Kirchhammer, Robert Ivanek, Michal Stanczak, Alfred Zippelius, David König, Natalia Rodrigues Manutano, and Heinz Läubli
- Subjects
sialic acid ,tumor immunology ,glyco-immune checkpoint ,antigen ,antigen processing ,Biology (General) ,QH301-705.5 - Abstract
Interactions between sialylated glycans and sialic acid-binding immunoglobulin-like lectin (Siglec) receptors have been recently described as potential new immune checkpoint that can be targeted to improve anticancer immunity. Myeloid cells have been reported to express a wide range of different Siglecs; however, their expression and functions on cancer-associated dendritic cells (DCs) were not fully characterized. We found that classical conventional DCs (cDCs) from cancer patient samples have a high expression of several inhibitory Siglecs including Siglec-7, Siglec-9, and Siglec-10. In subcutaneous murine tumor models, we also found an upregulation of the inhibitory Siglec-E receptor on cancer-associated cDCs. DC lines and bone marrow-derived DCs (BMDCs) with expression of these inhibitory Siglecs showed impaired maturation states on transcriptome and protein level. Furthermore, ablation of these inhibitory Siglecs from DCs enhanced their capability to prime antigen-specific T cells and induce proliferation. Our work provides a deeper understanding of the influence of inhibitory Siglecs on DCs and reveals a potential new target to improve cancer immunotherapy.
- Published
- 2022
- Full Text
- View/download PDF
7. GEF-H1 Signaling upon Microtubule Destabilization Is Required for Dendritic Cell Activation and Specific Anti-tumor Responses
- Author
-
Abhishek S. Kashyap, Laura Fernandez-Rodriguez, Yun Zhao, Gianni Monaco, Marcel P. Trefny, Naohiro Yoshida, Kea Martin, Ashwani Sharma, Natacha Olieric, Pankaj Shah, Michal Stanczak, Nicole Kirchhammer, Sung-Moo Park, Sebastien Wieckowski, Heinz Laubli, Rachid Zagani, Benjamin Kasenda, Michel O. Steinmetz, Hans-Christian Reinecker, and Alfred Zippelius
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Dendritic cell (DC) activation is a critical step for anti-tumor T cell responses. Certain chemotherapeutics can influence DC function. Here we demonstrate that chemotherapy capable of microtubule destabilization has direct effects on DC function; namely, it induces potent DC maturation and elicits anti-tumor immunity. Guanine nucleotide exchange factor-H1 (GEF-H1) is specifically released upon microtubule destabilization and is required for DC activation. In response to chemotherapy, GEF-H1 drives a distinct cell signaling program in DCs dominated by the c-Jun N-terminal kinase (JNK) pathway and AP-1/ATF transcriptional response for control of innate and adaptive immune responses. Microtubule destabilization, and subsequent GEF-H1 signaling, enhances cross-presentation of tumor antigens to CD8 T cells. In absence of GEF-H1, anti-tumor immunity is hampered. In cancer patients, high expression of the GEF-H1 immune gene signature is associated with prolonged survival. Our study identifies an alternate intracellular axis in DCs induced upon microtubule destabilization in which GEF-H1 promotes protective anti-tumor immunity. : Certain chemotherapeutics elicit potent anti-tumor immunity. Kashyap et al. demonstrate that microtubule-destabilizing chemotherapeutics induce maturation of dendritic cells through activation of microtubule-associated protein GEF-H1. This leads to effective priming of CD8 T cells against tumor antigens. GEF-H1 is critical for anti-tumor immunity of microtubule-targeting chemotherapy. Keywords: GEF-H1, lfc, dendritic cells, microtubule-targeting agents, cross presentation, JNK pathway, ansamitocin-P3, plinabulin, immunotherapy, chemotherapy
- Published
- 2019
- Full Text
- View/download PDF
8. Supplementary Figures from Hyperglycemia Enhances Cancer Immune Evasion by Inducing Alternative Macrophage Polarization through Increased O-GlcNAcylation
- Author
-
Heinz Läubli, Adriane R. Todeschini, Alfred Zippelius, Wagner B. Dias, César de Souza Bastos, Miguel Fontes, Agatha Carlos Fonseca, Ronan Christian Santos, Gianni Monaco, Alessandra A. Filardy, Nicole Kirchhammer, Isadora de Araújo Oliveira, Michal A. Stanczak, and Natália Rodrigues Mantuano
- Abstract
Supplementary Figures 1-8
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Hyperglycemia Enhances Cancer Immune Evasion by Inducing Alternative Macrophage Polarization through Increased O-GlcNAcylation
- Author
-
Heinz Läubli, Adriane R. Todeschini, Alfred Zippelius, Wagner B. Dias, César de Souza Bastos, Miguel Fontes, Agatha Carlos Fonseca, Ronan Christian Santos, Gianni Monaco, Alessandra A. Filardy, Nicole Kirchhammer, Isadora de Araújo Oliveira, Michal A. Stanczak, and Natália Rodrigues Mantuano
- Abstract
Diabetes mellitus (DM) significantly increases the risk for cancer and cancer progression. Hyperglycemia is the defining characteristic of DM and tightly correlates with a poor prognosis in patients with cancer. The hexosamine biosynthetic pathway (HBP) is emerging as a pivotal cascade linking high glucose, tumor progression, and impaired immune function. Here we show that enhanced glucose flow through the HBP drives cancer progression and immune evasion by increasing O-GlcNAcylation in tumor-associated macrophages (TAM). Increased O-GlcNAc skewed macrophage polarization to a M2-like phenotype supporting tumor progression. Finally, we found an upregulation of M2 markers on TAMs in DM2 patients with colorectal cancer compared with nondiabetic normoglycemic patients. Our results provide evidence for a new and targetable mechanism of cancer immune evasion in patients with hyperglycemia, advocating for strict control of hyperglycemia in patients with cancer.
- Published
- 2023
- Full Text
- View/download PDF
10. Combination cancer immunotherapies: Emerging treatment strategies adapted to the tumor microenvironment
- Author
-
Priska Auf der Maur, Heinz Laubli, Alfred Zippelius, Nicole Kirchhammer, and Marcel Philipp Trefny
- Subjects
Neoplasms ,Tumor Microenvironment ,Humans ,General Medicine ,Immunotherapy - Abstract
Immune checkpoint blockade (ICB) has revolutionized cancer treatment. However, resistance to ICB occurs frequently due to tumor-intrinsic alterations or extrinsic factors in the tumor microenvironment. This Viewpoint aims to give an update on recent developments in immunotherapy for solid tumors and highlights progress in translational research and clinical practice.
- Published
- 2022
11. Targeting cancer glycosylation repolarizes tumor-associated macrophages allowing effective immune checkpoint blockade
- Author
-
Michal A. Stanczak, Natalia Rodrigues Mantuano, Nicole Kirchhammer, David E. Sanin, Francis Jacob, Ricardo Coelho, Arun V. Everest-Dass, Jinyu Wang, Marcel P. Trefny, Gianni Monaco, Anne Bärenwaldt, Melissa A. Gray, Adam Petrone, Abhishek S. Kashyap, Katharina Glatz, Benjamin Kasenda, Karl Normington, James Broderick, Li Peng, Oliver M.T. Pearce, Erika L. Pearce, Carolyn R. Bertozzi, Alfred Zippelius, and Heinz Läubli
- Subjects
Sialic Acid Binding Immunoglobulin-like Lectins ,Mice ,Glycosylation ,Neoplasms ,Tumor-Associated Macrophages ,Tumor Microenvironment ,Humans ,Animals ,General Medicine ,Immune Checkpoint Inhibitors - Abstract
Immune checkpoint blockade (ICB) has substantially improved the prognosis of patients with cancer, but the majority experiences limited benefit, supporting the need for new therapeutic approaches. Up-regulation of sialic acid–containing glycans, termed hypersialylation, is a common feature of cancer-associated glycosylation, driving disease progression and immune escape through the engagement of Siglec receptors on tumor-infiltrating immune cells. Here, we show that tumor sialylation correlates with distinct immune states and reduced survival in human cancers. The targeted removal of Siglec ligands in the tumor microenvironment, using an antibody-sialidase conjugate, enhanced antitumor immunity and halted tumor progression in several murine models. Using single-cell RNA sequencing, we revealed that desialylation repolarized tumor-associated macrophages (TAMs). We also identified Siglec-E as the main receptor for hypersialylation on TAMs. Last, we found that genetic and therapeutic desialylation, as well as loss of Siglec-E, enhanced the efficacy of ICB. Thus, therapeutic desialylation represents an immunotherapeutic approach to reshape macrophage phenotypes and augment the adaptive antitumor immune response.
- Published
- 2022
12. 434 Targeting the expression of Neuropilin-1 by locked nucleic acid modified antisense oligonucleotides results in potent anti-tumor activity in vivo
- Author
-
Richard Klar, Clara Seger, Nicole Kirchhammer, André Maaske, Julia Festag, Laura Fernandez Rodriguez, Mélanie Buchi, Monika Schell, Stefanie Raith, Sven Michel, Alfred Zippelius, and Frank Jaschinski
- Published
- 2022
- Full Text
- View/download PDF
13. iMATCH: an integrated modular assembly system for therapeutic combination high-capacity adenovirus gene therapy
- Author
-
Alfred Zippelius, Nicole Kirchhammer, Andreas Plückthun, Melanie Grove, Fabian Weiss, Markus Schmid, Patrick C. Freitag, Nina Schumacher, Dominik Brücher, Urs F. Greber, Corina Keller, Jatina Schumacher, Jonas Kolibius, Sheena N. Smith, University of Zurich, and Plückthun, Andreas
- Subjects
0301 basic medicine ,gutless adenovirus ,lcsh:QH426-470 ,medicine.medical_treatment ,Genetic enhancement ,helper-dependent adenovirus ,Computational biology ,Vectors in gene therapy ,Biology ,Genome ,03 medical and health sciences ,0302 clinical medicine ,gene vectors ,1311 Genetics ,PD-1 ,10019 Department of Biochemistry ,1312 Molecular Biology ,Genetics ,medicine ,lcsh:QH573-671 ,Molecular Biology ,Gene ,Cloning ,lcsh:Cytology ,IL-2 ,Immunotherapy ,gene therapy ,10124 Institute of Molecular Life Sciences ,lcsh:Genetics ,030104 developmental biology ,Capsid ,IL-12 ,1313 Molecular Medicine ,030220 oncology & carcinogenesis ,Helper virus ,high-capacity vectors ,570 Life sciences ,biology ,Molecular Medicine ,cancer therapy ,Original Article ,immunotherapy - Abstract
Adenovirus-mediated combination gene therapies have shown promising results in vaccination or treating malignant and genetic diseases. Nevertheless, an efficient system for the rapid assembly and incorporation of therapeutic genes into high-capacity adenoviral vectors (HCAdVs) is still missing. In this study, we developed the iMATCH (integrated modular assembly for therapeutic combination HCAdVs) platform, which enables the generation and production of HCAdVs encoding therapeutic combinations in high quantity and purity within 3 weeks. Our modular cloning system facilitates the efficient combination of up to four expression cassettes and the rapid integration into HCAdV genomes with defined sizes. Helper viruses (HVs) and purification protocols were optimized to produce HCAdVs with distinct capsid modifications and unprecedented purity (0.1 ppm HVs). The constitution of HCAdVs, with adapters for targeting and a shield of trimerized single-chain variable fragment (scFv) for reduced liver clearance, mediated cell- and organ-specific targeting of HCAdVs. As proof of concept, we show that a single HCAdV encoding an anti PD-1 antibody, interleukin (IL)-12, and IL-2 produced all proteins, and it led to tumor regression and prolonged survival in tumor models, comparable to a mixture of single payload HCAdVs in vitro and in vivo. Therefore, the iMATCH system provides a versatile platform for the generation of high-capacity gene therapy vectors with a high potential for clinical development., Graphical Abstract, The iMATCH platform enables an efficient assembly of up to four orthogonal expression cassettes on a single plasmid from which high-capacity adenoviral vectors can be generated within 3 weeks in unprecedented purity. iMATCH vectors encoding immunotherapeutic combinations led to tumor regression and prolonged survival, comparable to mixtures of single payload HCAdVs.
- Published
- 2021
14. NK cells with tissue-resident traits shape response to immunotherapy by inducing adaptive antitumor immunity
- Author
-
Nicole Kirchhammer, Marcel P. Trefny, Marina Natoli, Dominik Brücher, Sheena N. Smith, Franziska Werner, Victoria Koch, David Schreiner, Ewelina Bartoszek, Mélanie Buchi, Markus Schmid, Daniel Breu, K. Patricia Hartmann, Polina Zaytseva, Daniela S. Thommen, Heinz Läubli, Jan P. Böttcher, Michal A. Stanczak, Abhishek S. Kashyap, Andreas Plückthun, Alfred Zippelius, University of Zurich, Kirchhammer, Nicole, and Zippelius, Alfred
- Subjects
Integrin alpha1 ,Programmed Cell Death 1 Receptor ,610 Medicine & health ,General Medicine ,2700 General Medicine ,Dendritic Cells ,Interleukin-12 ,Killer Cells, Natural ,Mice ,Neoplasms ,10019 Department of Biochemistry ,Tumor Microenvironment ,570 Life sciences ,biology ,Animals ,Immunotherapy - Abstract
T cell–directed cancer immunotherapy often fails to generate lasting tumor control. Harnessing additional effectors of the immune response against tumors may strengthen the clinical benefit of immunotherapies. Here, we demonstrate that therapeutic targeting of the interferon-γ (IFN-γ)–interleukin-12 (IL-12) pathway relies on the ability of a population of natural killer (NK) cells with tissue-resident traits to orchestrate an antitumor microenvironment. In particular, we used an engineered adenoviral platform as a tool for intratumoral IL-12 immunotherapy (AdV5–IL-12) to generate adaptive antitumor immunity. Mechanistically, we demonstrate that AdV5–IL-12 is capable of inducing the expression of CC-chemokine ligand 5 (CCL5) in CD49a + NK cells both in tumor mouse models and tumor specimens from patients with cancer. AdV5–IL-12 imposed CCL5-induced type I conventional dendritic cell (cDC1) infiltration and thus increased DC-CD8 T cell interactions. A similar observation was made for other IFN-γ–inducing therapies such as Programmed cell death 1 (PD-1) blockade. Conversely, failure to respond to IL-12 and PD-1 blockade in tumor models with low CD49a + CXCR6 + NK cell infiltration could be overcome by intratumoral delivery of CCL5. Thus, therapeutic efficacy depends on the abundance of NK cells with tissue-resident traits and, specifically, their capacity to produce the DC chemoattractant CCL5. Our findings reveal a barrier for T cell–focused therapies and offer mechanistic insights into how T cell–NK cell–DC cross-talk can be enhanced to promote antitumor immunity and overcome resistance.
- Published
- 2022
- Full Text
- View/download PDF
15. Adaptive anti-tumor immunity is orchestrated by a population of CCL5-producing tissue-resident NK cells
- Author
-
Nicole Kirchhammer, Dominik Brücher, K Patricia Hartmann, Heinz Läubli, Michal A. Stanczak, Andreas Plückthun, Daniela S. Thommen, Ewelina Bartoszek, Markus Schmid, Polina Zaytseva, Jan P. Böttcher, David Schreiner, Melanie Buchi, Sheena N. Smith, Abhishek S. Kashyap, Franziska Werner, Marcel P. Trefny, Daniel Breu, Alfred Zippelius, Marina Natoli, and Victoria Koch
- Subjects
education.field_of_study ,medicine.medical_treatment ,T cell ,Population ,Biology ,CCL5 ,Immune checkpoint ,Crosstalk (biology) ,Paracrine signalling ,medicine.anatomical_structure ,Immune system ,Cancer immunotherapy ,medicine ,Cancer research ,education - Abstract
T cell-directed cancer immunotherapy often fails to generate lasting tumor control. Harnessing additional effectors of the immune response against tumors may strengthen the clinical benefit of immunotherapies. Here, we demonstrate that therapeutic targeting of the IFNγ-IL-12 pathway relies on the ability of a population of tissue-resident NK (trNK) cells to orchestrate an anti-tumor microenvironment. Particularly, utilizing an engineered adenoviral platform, we show that paracrine IL-12 enhances functional DC-CD8 T cell interactions to generate adaptive anti-tumor immunity. This effect depends on the abundance of trNK cells and specifically their capacity to produce the cDC1-chemoattractant CCL5. Failure to respond to IL-12 and other IFNγ-inducing therapies such as immune checkpoint blockade in tumors with low trNK cell infiltration could be overcome by intra-tumoral delivery of CCL5. Our findings reveal a novel barrier for T cell-focused therapies and offer mechanistic insights into how T cell-NK cell-DC crosstalk can be enhanced to promote anti-tumor immunity and overcome resistance.SignificanceWe identified the lack of CCL5-producing, tissue-resident NK (trNK) cells as a barrier to T cell-focused therapies. While IL-12 induces anti-tumoral DC-T cell crosstalk in trNK cellrichtumors, resistance to IL-12 or anti-PD-1 in trNK cellpoortumors can be overcome by the additional delivery of CCL5.
- Published
- 2021
- Full Text
- View/download PDF
16. Targeting cancer glycosylation repolarizes tumor-associated macrophages allowing effective immune checkpoint blockade
- Author
-
Marcel P. Trefny, A. Petrone, K. Normington, Jing Wang, L. Peng, J. Broderick, Katharina Glatz, Alfred Zippelius, Erika L. Pearce, N. R. Mantuano, Nicole Kirchhammer, Melissa A. Gray, Abhishek S. Kashyap, Gianni Monaco, David E. Sanin, Oliver M. T. Pearce, Carolyn R. Bertozzi, H. Laeubli, Benjamin Kasenda, and Michal A. Stanczak
- Subjects
Tumor microenvironment ,Immune system ,Downregulation and upregulation ,Immunity ,Tumor progression ,business.industry ,Cancer research ,medicine ,Macrophage ,Cancer ,medicine.disease ,business ,Immune checkpoint - Abstract
Immune checkpoint blockade (ICB) has significantly improved the prognosis of cancer patients, but the majority experience limited benefit, evidencing the need for new therapeutic approaches. Upregulation of sialic acid-containing glycans, termed hypersialylation, is a common feature of cancer-associated glycosylation, driving disease progression and immune escape via the engagement of Siglec-receptors on tumor-infiltrating immune cells. Here, we show that tumor sialylation correlates with distinct immune states and reduced survival in human cancers. The targeted removal of Siglec-ligands in the tumor microenvironment, using an antibody-sialidase conjugate, enhances anti-tumor immunity and halts tumor progression in several mouse tumor models. Using single-cell RNA sequencing, we reveal desialylation mechanistically to repolarize tumor-associated macrophages (TAMs) and identify Siglec-E on TAMs as the main receptor for hypersialylation. Finally, we show genetic and therapeutic desialylation, as well as loss of Siglec-E, to synergize with ICB. Thus, therapeutic desialylation represents a novel immunotherapeutic approach, shaping macrophage phenotypes and augmenting the adaptive anti-tumor immune response.
- Published
- 2021
- Full Text
- View/download PDF
17. Magnesium sensing via LFA-1 regulates CD8
- Author
-
Jonas, Lötscher, Adrià-Arnau, Martí I Líndez, Nicole, Kirchhammer, Elisabetta, Cribioli, Greta Maria Paola, Giordano Attianese, Marcel P, Trefny, Markus, Lenz, Sacha I, Rothschild, Paolo, Strati, Marco, Künzli, Claudia, Lotter, Susanne H, Schenk, Philippe, Dehio, Jordan, Löliger, Ludivine, Litzler, David, Schreiner, Victoria, Koch, Nicolas, Page, Dahye, Lee, Jasmin, Grählert, Dmitry, Kuzmin, Anne-Valérie, Burgener, Doron, Merkler, Miklos, Pless, Maria L, Balmer, Walter, Reith, Jörg, Huwyler, Melita, Irving, Carolyn G, King, Alfred, Zippelius, and Christoph, Hess
- Subjects
Cytotoxicity, Immunologic ,Male ,Immunological Synapses ,MAP Kinase Signaling System ,Proto-Oncogene Proteins c-jun ,Bacterial Infections ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Lymphocyte Function-Associated Antigen-1 ,Mice, Inbred C57BL ,HEK293 Cells ,Phenotype ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,Magnesium ,Immunotherapy ,Phosphorylation ,Immunologic Memory ,Caloric Restriction - Abstract
The relevance of extracellular magnesium in cellular immunity remains largely unknown. Here, we show that the co-stimulatory cell-surface molecule LFA-1 requires magnesium to adopt its active conformation on CD8
- Published
- 2021
18. Abstract 4149: Knock-down of Neuropilin-1 by locked nucleic acid antisense oligonucleotides facilitates cancer immune control
- Author
-
Andre Maaske, Nicole Kirchhammer, Julia Festag, Laura Fernandez Rodriguez, Mélanie Buchi, Monika Schell, Stefanie Raith, Sven Michel, Richard Klar, Alfred Zippelius, and Frank Jaschinski
- Subjects
Cancer Research ,Oncology - Abstract
Immune checkpoint inhibitors have become an important tool for the treatment of cancer. However, the majority of patients does not respond to the currently employed antibody-based treatments, indicating a need for additional targets and new treatment modalities. The invention of 3rd generation chemistries like locked nucleic acids (LNAs) has allowed the development of highly specific and efficacious antisense oligonucleotides (ASOs) that allow target knockdown in vivo without the need for complex delivery formulations. The membrane-bound protein neuropilin-1 (NRP1) was initially identified as a factor involved in cell migration, -survival, and neoangiogenesis making it an attractive target for cancer therapy. It was subsequently shown that it has additional immune-mediated pro-tumorigenic roles. It can suppress anti-tumor immune activity via regulation of trafficking of tumor-associated macrophages, phenotypic stability of Tregs, and exhaustion of effector T cells. Considering that these effects are mediated by multiple domains of NRP1, there is a conceptual advantage to down-regulate the expression of the whole protein over the functional or steric blockade of individual domains. We used our in-house Oligofyer™ bioinformatics system to design both human NRP1 specific LNAplus™ ASOs, as well as murine surrogates, which both achieved target knock-down of more than 85% in vitro. Furthermore, the murine surrogate down regulated NRP1 in tumor mouse models. Systemic application of mouse-specific ASOs without additional carriers or adjuvants has led to a strong delay of tumor growth or complete eradication in syngeneic mouse models. This effect was almost completely abrogated in immune-compromised NSG mice. Furthermore, re-challenge of immune-competent mice after tumor eradication did not result in new tumor growth, suggesting induction of a long-lasting immunity by LNAplus™ ASO-mediated knockdown of NRP1. Taken together, these encouraging results indicate that the ASO-mediated down-regulation of NRP1 has the potential to become a promising treatment option for patients in need. We are currently performing further experiments to fully elucidate the mechanisms that underlie the observed anti-tumor efficacy. Citation Format: Andre Maaske, Nicole Kirchhammer, Julia Festag, Laura Fernandez Rodriguez, Mélanie Buchi, Monika Schell, Stefanie Raith, Sven Michel, Richard Klar, Alfred Zippelius, Frank Jaschinski. Knock-down of Neuropilin-1 by locked nucleic acid antisense oligonucleotides facilitates cancer immune control [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 4149.
- Published
- 2022
- Full Text
- View/download PDF
19. A novel rabbit derived anti-HER2 antibody with pronounced therapeutic effectiveness on HER2-positive breast cancer cells in vitro and in humanized tumor mice (HTM)
- Author
-
Michael Brandt, Sandra Prassl, Nicole Kirchhammer, Anja K. Wege, Gerhard Piendl, Stephan Fischer, Linda Veronique Kazandjian, Gero Brockhoff, and Olaf Ortmann
- Subjects
Monoclonal antibody ,0301 basic medicine ,Anti-HER2 antibody, Breast cancer, Treatment efficiency, Humanized tumor mice, Monoclonal antibody ,Receptor, ErbB-2 ,medicine.drug_class ,Anti-HER2 antibody ,610 Medizin ,lcsh:Medicine ,Apoptosis ,Breast Neoplasms ,Treatment efficiency ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,Breast cancer ,0302 clinical medicine ,Immune system ,Trastuzumab ,Cell Line, Tumor ,medicine ,Animals ,Humans ,skin and connective tissue diseases ,neoplasms ,Cell Proliferation ,ddc:610 ,biology ,business.industry ,Research ,lcsh:R ,Cancer ,General Medicine ,medicine.disease ,Primary tumor ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Rabbits ,Pertuzumab ,Antibody ,business ,Humanized tumor mice ,medicine.drug - Abstract
Background Antibody based cancer therapies have achieved convincing success rates combining enhanced tumor specificity and reduced side effects in patients. Trastuzumab that targets the human epidermal growth factor related receptor 2 (HER2) is one of the greatest success stories in this field. For decades, trastuzumab based treatment regimens are significantly improving the prognosis of HER2-positive breast cancer patients both in the metastatic and the (neo-) adjuvant setting. Nevertheless, ≥ 50% of trastuzumab treated patients experience de-novo or acquired resistance. Therefore, an enhanced anti-HER2 targeting with improved treatment efficiency is still aspired. Methods Here, we determined cellular and molecular mechanisms involved in the treatment of HER2-positive BC cells with a new rabbit derived HER2 specific chimeric monoclonal antibody called “B100″. We evaluated the B100 treatment efficiency of HER2-positive BC cells with different sensitivity to trastuzumab both in vitro and in the presence of a human immune system in humanized tumor mice. Results B100 not only efficiently blocks cell proliferation but more importantly induces apoptotic tumor cell death. Detailed in vitro analyses of B100 in comparison to trastuzumab (and pertuzumab) revealed equivalent HER2 internalization and recycling capacity, similar Fc receptor signaling, but different HER2 epitope recognition with high binding and treatment efficiency. In trastuzumab resistant SK-BR-3 based humanized tumor mice the B100 treatment eliminated the primary tumor but even more importantly eradicated metastasized tumor cells in lung, liver, brain, and bone marrow. Conclusion Overall, B100 demonstrated an enhanced anti-tumor activity both in vitro and in an enhanced preclinical HTM in vivo model compared to trastuzumab or pertuzumab. Thus, the use of B100 is a promising option to complement and to enhance established treatment regimens for HER2-positive (breast) cancer and to overcome trastuzumab resistance. Extended preclinical analyses using appropriate models and clinical investigations are warranted.
- Published
- 2020
20. Magnesium sensing via LFA-1 regulates CD8+ T cell effector function
- Author
-
Jonas Lötscher, Adrià-Arnau Martí i Líndez, Nicole Kirchhammer, Elisabetta Cribioli, Greta Maria Paola Giordano Attianese, Marcel P. Trefny, Markus Lenz, Sacha I. Rothschild, Paolo Strati, Marco Künzli, Claudia Lotter, Susanne H. Schenk, Philippe Dehio, Jordan Löliger, Ludivine Litzler, David Schreiner, Victoria Koch, Nicolas Page, Dahye Lee, Jasmin Grählert, Dmitry Kuzmin, Anne-Valérie Burgener, Doron Merkler, Miklos Pless, Maria L. Balmer, Walter Reith, Jörg Huwyler, Melita Irving, Carolyn G. King, Alfred Zippelius, and Christoph Hess
- Subjects
610 Medicine & health ,General Biochemistry, Genetics and Molecular Biology - Abstract
The relevance of extracellular magnesium in cellular immunity remains largely unknown. Here, we show that the co-stimulatory cell-surface molecule LFA-1 requires magnesium to adopt its active conformation on CD8+ T��cells, thereby augmenting calcium flux, signal transduction, metabolic reprogramming, immune synapse formation, and, as a consequence, specific cytotoxicity. Accordingly, magnesium-sufficiency sensed via LFA-1 translated to the superior performance of pathogen- and tumor-specific T��cells, enhanced effectiveness of bi-specific T��cell engaging antibodies, and improved CAR T��cell function. Clinically, low serum magnesium levels were associated with more rapid disease progression and shorter overall survival in CAR T��cell and immune checkpoint antibody-treated patients. LFA-1 thus directly incorporates information on the composition of the microenvironment as a determinant of outside-in signaling activity. These findings conceptually link co-stimulation and nutrient sensing and point to the magnesium-LFA-1 axis as a therapeutically amenable biologic system.
- Published
- 2022
- Full Text
- View/download PDF
21. Hyperglycemia Enhances Cancer Immune Evasion by Inducing Alternative Macrophage Polarization through Increased O-GlcNAcylation
- Author
-
Isadora A. Oliveira, Alessandra A. Filardy, Alfred Zippelius, Heinz Läubli, César de Souza Bastos, Nicole Kirchhammer, Michal A. Stanczak, Agatha Carlos Fonseca, Ronan Christian Santos, Natalia Rodrigues Mantuano, Gianni Monaco, Adriane R. Todeschini, Wagner B. Dias, and Miguel Fontes
- Subjects
Male ,Cancer Research ,Glycosylation ,Colorectal cancer ,Immunology ,Macrophage polarization ,Mice, SCID ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Downregulation and upregulation ,Diabetes mellitus ,medicine ,Animals ,Humans ,Immune Evasion ,business.industry ,Macrophages ,Cancer ,medicine.disease ,Phenotype ,Disease Models, Animal ,Tumor progression ,030220 oncology & carcinogenesis ,Hyperglycemia ,Cancer research ,business ,030215 immunology - Abstract
SummaryDiabetes mellitus (DM) significantly increases the risk for cancer and cancer progression. Hyperglycemia is the defining characteristic of DM and tightly correlates with a poor prognosis in cancer patients. The hexosamine biosynthetic pathway (HBP) is emerging as a pivotal cascade linking high glucose, tumor progression and impaired immune function. Here we show that enhanced glucose flow through the HBP drives cancer progression and immune evasion by increasing O-GlcNAcylation in tumor-associated macrophages (TAMs). Increased O-GlcNAc skewed macrophage polarization to a M2-like phenotype. HBP or O-GlcNAcylation inhibition reprogrammed TAMs to an anti-tumoral phenotype. Finally, we found an upregulation of M2 markers on TAMs in DM2 patients with colorectal cancer compared to non-diabetic normoglycemic patients. Our results provide evidence for a new and targetable mechanism of cancer immune evasion in patients with hyperglycemia, advocating for strict control of hyperglycemia in cancer patients.SignificanceHyperglycemia increases O-GlcNAc levels in TAMs, programing them to a pro-tumorigenic phenotype (M2-like), contributing to cancer progression. Inhibition of O-GlcNAcylation could therefore be used to reprogram intratumoral macrophages to an anti-tumoral phenotype.
- Published
- 2019
22. Heterologous arenavirus vector prime-boost overrules self-tolerance for efficient tumor-specific CD8 T cell attack
- Author
-
Ilena Vincenti, Sabine Hoepner, Alfred Zippelius, Ursula Berka, Stephanie Darbre, Magdalena A. Krzyzaniak, Stephan Günther, Nicole Kirchhammer, Carsten Magnus, Romy Kerber, Daniel D. Pinschewer, Weldy V. Bonilla, Josipa Raguz, Sarah Schmidt, Min Lu, Klaus Orlinger, Anna-Friederike Marx, Sandra M. Kallert, Mindaugas Pauzuolis, and Doron Merkler
- Subjects
medicine.medical_treatment ,Genetic Vectors ,Guinea Pigs ,Immunization, Secondary ,Gene Expression ,pre-existing immunity ,Heterologous ,CD8 T cells ,Immunodominance ,ddc:616.07 ,Lymphocytic choriomeningitis ,Cancer Vaccines ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,therapeutic tumor vaccine ,medicine ,Alarmins ,Animals ,Lymphocytic choriomeningitis virus ,Cytotoxic T cell ,Vector (molecular biology) ,arenavirus ,Pichinde virus ,Phylogeny ,tumor control ,Arenavirus ,biology ,Vaccination ,anti-vector immunity ,Immunotherapy ,Mastocytoma ,biology.organism_classification ,medicine.disease ,Antibodies, Neutralizing ,Survival Analysis ,Virology ,viral genealogy ,Mice, Inbred C57BL ,CTL ,Self Tolerance ,Female ,Genetic Engineering ,T-Lymphocytes, Cytotoxic - Abstract
Summary Therapeutic vaccination regimens inducing clinically effective tumor-specific CD8+ T lymphocyte (CTL) responses are an unmet medical need. We engineer two distantly related arenaviruses, Pichinde virus and lymphocytic choriomeningitis virus, for therapeutic cancer vaccination. In mice, life-replicating vector formats of these two viruses delivering a self-antigen in a heterologous prime-boost regimen induce tumor-specific CTL responses up to 50% of the circulating CD8 T cell pool. This CTL attack eliminates established solid tumors in a significant proportion of animals, accompanied by protection against tumor rechallenge. The magnitude of CTL responses is alarmin driven and requires combining two genealogically distantly related arenaviruses. Vector-neutralizing antibodies do not inhibit booster immunizations by the same vector or by closely related vectors. Rather, CTL immunodominance hierarchies favor vector backbone-targeted responses at the expense of self-reactive CTLs. These findings establish an arenavirus-based immunotherapy regimen that allows reshuffling of immunodominance hierarchies and breaking self-directed tolerance for efficient tumor control., Graphical abstract, Highlights Engineered arenaviruses induce potent tumor self-specific CD8 T cell (CTL) response Combinations of distantly but not closely related arenavirus vectors eliminate tumors Vector backbone-targeted CTL responses compete against tumor self-reactive CTLs Optimized vector combinations reshuffle immunodominance to break self-tolerance, Therapeutic tumor vaccination should break self-tolerance. Assessing combinations of engineered arenavirus vectors, Bonilla et al. find that distantly related vector combinations reshuffle T cell immunodominance hierarchies to break self-tolerance and eliminate established solid tumors, whereas closely related vectors interfere because of immunodominance of anti-vector CD8 T cells rather than antibodies.
- Published
- 2021
- Full Text
- View/download PDF
23. Arenavirus vectors for massive tumor self-antigen-specific CD8 T cell attack
- Author
-
Daniel D. Pinschewer, Weldy V. Bonilla, Sandra Kallert, Nicole Kirchhammer, Anna-Friederike Marx, Magdalena Krzyzaniak, Sarah Schmidt, Josipa Raguz, Ursula Berka, Stephan Günther, Carsten Magnus, Klaus Orlinger, and Alfred Zippelius
- Subjects
Immunology ,Immunology and Allergy - Abstract
Therapeutic vaccination regimens inducing high frequencies of clinically effective tumor-targeting CD8 T cells represent an unmet need in cancer immunotherapy. We exploit cutting-edge viral reverse genetics to harness the immunostimulatory properties of the arenaviruses Pichinde virus and lymphocytic choriomeningitis virus for therapeutic cancer vaccination. When administering to mice life-replicating vector formats of these two viruses, both delivering the same cancer testis self-antigen in a heterologous prime-boost regimen, tumor-specific CD8 T cell responses reached up to 50% of the circulating CD8 T cell pool. This massive CD8 T cell attack eliminated established solid tumors in a significant proportion of animals, accompanied by complete protection against tumor re-challenge. The magnitude of CD8 T cell responses was driven by alarmin signals and depended on the combined use of two genealogically distantly related arenaviruses. Combinations of closely related vectors or the repeated administration of the same vector were not inhibited by vector-neutralizing antibodies, but CD8 T cell immunodominance hierarchies favored vector backbone-targeted responses at the expense of self-reactive T cell specificities. These findings establish a powerful arenavirus-based tumor immunotherapy regimen exploiting key innate signaling pathways and reshuffling immunodominance hierarchies to break tumor self-directed tolerance
- Published
- 2020
- Full Text
- View/download PDF
24. GEF-H1 Signaling upon Microtubule Destabilization Is Required for Dendritic Cell Activation and Specific Anti-tumor Responses
- Author
-
Michal A. Stanczak, Pankaj Shah, Naohiro Yoshida, Benjamin Kasenda, Nicole Kirchhammer, Abhishek S. Kashyap, Sung-Moo Park, Rachid Zagani, Sébastien Wieckowski, Laura Fernandez-Rodriguez, Gianni Monaco, Alfred Zippelius, Yun Zhao, Marcel P. Trefny, Hans Christian Reinecker, Ashwani Sharma, Michel O. Steinmetz, Kea Martin, Heinz Läubli, and Natacha Olieric
- Subjects
0301 basic medicine ,Cell signaling ,medicine.medical_treatment ,T cell ,Microtubules ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Neoplasms ,medicine ,Cytotoxic T cell ,Humans ,lcsh:QH301-705.5 ,Chemistry ,fungi ,Cross-presentation ,Cell Differentiation ,Immunotherapy ,Dendritic cell ,Dendritic Cells ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,030217 neurology & neurosurgery ,Intracellular ,Rho Guanine Nucleotide Exchange Factors ,Signal Transduction - Abstract
SUMMARY Dendritic cell (DC) activation is a critical step for anti-tumor T cell responses. Certain chemotherapeutics can influence DC function. Here we demonstrate that chemotherapy capable of microtubule destabilization has direct effects on DC function; namely, it induces potent DC maturation and elicits anti-tumor immunity. Guanine nucleotide exchange factor-H1 (GEF-H1) is specifically released upon microtubule destabilization and is required for DC activation. In response to chemotherapy, GEF-H1 drives a distinct cell signaling program in DCs dominated by the c-Jun N-terminal kinase (JNK) pathway and AP-1/ATF transcriptional response for control of innate and adaptive immune responses. Microtubule destabilization, and subsequent GEF-H1 signaling, enhances cross-presentation of tumor antigens to CD8 T cells. In absence of GEF-H1, anti-tumor immunity is hampered. In cancer patients, high expression of the GEF-H1 immune gene signature is associated with prolonged survival. Our study identifies an alternate intracellular axis in DCs induced upon microtubule destabilization in which GEF-H1 promotes protective anti-tumor immunity., Graphical Abstract, In Brief Certain chemotherapeutics elicit potent anti-tumor immunity. Kashyap et al. demonstrate that microtubule-destabilizing chemotherapeutics induce maturation of dendritic cells through activation of microtubule-associated protein GEF-H1. This leads to effective priming of CD8 T cells against tumor antigens. GEF-H1 is critical for anti-tumor immunity of microtubule-targeting chemotherapy.
- Published
- 2019
25. Regulation of Programmed Death Ligand 1 (PD-L1) Expression in Breast Cancer Cell Lines In Vitro and in Immunodeficient and Humanized Tumor Mice
- Author
-
Brockhoff, Eva-Maria Rom-Jurek, Nicole Kirchhammer, Peter Ugocsai, Olaf Ortmann, Anja Wege, and Gero
- Subjects
breast cancer cell lines ,PD-(L)1 ,humanized tumor mice ,Epirubicin ,Paclitaxel ,skin and connective tissue diseases - Abstract
Programmed death ligand 1 (PD-L1) expression is an efficient strategy of tumor cells to escape immunological eradiation. However, only little is known about the factors that affect the cellular expression levels. Here we assessed the PD-L1 expression on different breast cancer cell lines under standard in vitro culture conditions and as a function of Epirubicin or Paclitaxel treatment. Moreover, we evaluated the expression in immunodeficient tumor mice as well as in humanized tumor mice (i.e., in the presence of a human immune system). We found highest PD-L1 levels in JIMT-1 and MDA-MB-231 cells. Epirubicin treatment caused a decrease and Paclitaxel treatment an increased PD-L1 expression in MDA-MB-231 cells. In addition, we identified nuclear PD-L1 in MDA-MB-231 cells. All in vivo transplanted breast cancer cell lines downregulated PD-L1 expression compared to their in vitro counterpart. Neither the gene copy number nor the presence of human immune system in humanized tumor mice had an effect on the PD-L1 content. We demonstrate that the degree of PD-L1 expression amongst breast cancer cell lines varies considerably. In addition, cytotoxic treatments and other extrinsic parameters differentially affect the expression. Hence, further investigations including in vivo evaluations are necessary to understand PD-L1 regulation for advanced breast cancer stratification.
- Published
- 2018
- Full Text
- View/download PDF
26. Arenavirus-based vector platform for massive tumor self-antigen-specific CD8 T cell immunity
- Author
-
Nicole Kirchhammer, Alfred Zippelius, Ursula Berka, Sandra M. Kallert, Daniel D. Pinschewer, Sarah Schmidt, Weldy V. Bonilla, Magdalena A. Krzyzaniak, S. Guenther, Anna-Friederike Marx, Klaus Orlinger, and Josipa Raguz
- Subjects
Cancer Research ,Arenavirus ,biology ,business.industry ,viruses ,biology.organism_classification ,complex mixtures ,Virology ,Oncology ,Immunity ,Antigen specific ,T cell immunity ,Cytotoxic T cell ,Medicine ,Vector (molecular biology) ,business ,CD8 - Abstract
e14297 Background: The induction of powerful CD8+ T cell immunity to tumor associated self-antigens (TAAs) represents a critical yet challenging goal. Here we report on the development of an arenavirus-based delivery platform meeting this challenge. Previously we have shown that genetically engineered replication-attenuated lymphocytic choriomeningitis virus (LCMV) vectors, TheraT(LCMV), induce strong TAA-specific CD8 T cell immunity, but these responses can not be substantially augmented upon TheraT(LCMV) readministration. Counter to expectations, vector-neutralizing antibodies were not accountable for limited homologous prime-boosting capacity. Instead, dominant viral backbone-reactive CD8+ T cells competed against subdominant TAA-specific responses, limiting their magnitude. Methods: Herein we engineered and characterized delivery systems based on the arenaviruses Mopeia, Candid#1 and Pichinde (TheraT(MOP), TheraT(CAND), TheraT(PIC)). Results: We demonstrate that heterologous TheraT(CAND) – TheraT(LCMV) and TheraT(PIC)-TheraT(LCMV) prime-boost substantially augment TAA-specific CD8 T cell responses by rendering them immunodominant. Accordingly, intravenous administration of mice triggered up to 50% TAA epitope-specific CD8+ T cells and cured established tumors. Conversely, TheraT(MOP) – TheraT(LCMV) prime-boost was poorly immunogenic owing to cross-reactive T cell epitopes in the respective viral backbones. Conclusions: These findings establish heterologous arenavirus prime-boost combinations as a powerful new modality in tumor immunotherapy and highlight CD8 T cell epitope dominance as a significant hurdle to overcome in the vectored delivery of TAAs.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.