24 results on '"Marcel P. Trefny"'
Search Results
2. 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
3. 13C tracer analysis reveals the landscape of metabolic checkpoints in human CD8+T cell differentiation
- Author
-
Alexander Kirchmair, Niloofar Nemati, Giorgia Lamberti, Marcel P. Trefny, Anne Krogsdam, Anita Siller, Paul Hörtnagl, Petra Schumacher, Sieghart Sopper, Adolf M. Sandbichler, Alfred Zippelius, Bart Ghesquière, and Zlatko Trajanoski
- Abstract
Naïve T cells remain in an actively maintained state of quiescence until activation by antigenic signals, upon which they start proliferation and generation of effector cells to initiate a functional immune response. Metabolic reprogramming is essential to meet the biosynthetic demands of the differentiation process, and failure to do so can promote the development of hypofunctional exhausted T cells. Here we used13C metabolomics and transcriptomics to study the metabolic dynamics of CD8+T cells in their complete course of differentiation from naïve over stem-like memory to effector cells. The quiescence of naïve T cells was evident in a profound suppression of glucose oxidation and a decreased expression ofENO1, downstream of which no glycolytic flux was detectable. Moreover, TCA cycle activity was low in naïve T cells and associated with a downregulation of SDH subunits. Upon stimulation and exit from quiescence, the initiation of cell growth and proliferation was accompanied by differential expression of T cell regulatory genes and metabolic reprogramming towards aerobic glycolysis with high rates of nutrient uptake, respiration and lactate production. High flux in anabolic pathways imposed a strain on NADH homeostasis, which coincided with engagement of the proline cycle for mitochondrial redox shuttling. With acquisition of effector functions, cells increasingly relied on glycolysis as opposed to oxidative phosphorylation, which paradoxically was not linked to changes in mitochondrial abundance. We further investigated the metabolic phenotype of exhausted T cells, finding that decreased effector function concurred with a reduction in mitochondrial metabolism, glycolysis and amino acid import, and an upregulation of suppressive and quiescence-associated genes, includingTXNIPandKLF2. Thus, these results identify multiple features critical for the metabolic reprogramming that supports quiescence, proliferation and effector function of CD8+T cells during differentiation. Further, an impairment of the same processes in exhaustion suggests that targeting these control points may be useful for both modulation of differentiation and prevention of exhaustion.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Data from A Variant of a Killer Cell Immunoglobulin-like Receptor Is Associated with Resistance to PD-1 Blockade in Lung Cancer
- Author
-
Heinz Läubli, Alfred Zippelius, Kirsten D. Mertz, Lukas Flatz, Zlatko Trajanoski, Martin Früh, Frank Stenner, Stefan Schaub, Ilaria Alborelli, Philip Jermann, Spasenija Savic, Florian Geier, Daniela S. Thommen, Severin Poechtrager, Petra Herzig, Monika Kaiser, Abhishek S. Kashyap, Fiamma Berner, Michal A. Stanczak, Benjamin Kasenda, Dietmar Rieder, Franziska Uhlenbrock, Sacha I. Rothschild, and Marcel P. Trefny
- Abstract
Supplementary Methods and Data
- Published
- 2023
- Full Text
- View/download PDF
5. Data from A Variant of a Killer Cell Immunoglobulin-like Receptor Is Associated with Resistance to PD-1 Blockade in Lung Cancer
- Author
-
Heinz Läubli, Alfred Zippelius, Kirsten D. Mertz, Lukas Flatz, Zlatko Trajanoski, Martin Früh, Frank Stenner, Stefan Schaub, Ilaria Alborelli, Philip Jermann, Spasenija Savic, Florian Geier, Daniela S. Thommen, Severin Poechtrager, Petra Herzig, Monika Kaiser, Abhishek S. Kashyap, Fiamma Berner, Michal A. Stanczak, Benjamin Kasenda, Dietmar Rieder, Franziska Uhlenbrock, Sacha I. Rothschild, and Marcel P. Trefny
- Abstract
Purpose:PD-(L)1–blocking antibodies have clinical activity in metastatic non–small cell lung cancer (NSCLC) and mediate durable tumor remissions. However, the majority of patients are resistant to PD-(L)1 blockade. Understanding mechanisms of primary resistance may allow prediction of clinical response and identification of new targetable pathways.Experimental Design:Peripheral blood mononuclear cells were collected from 35 patients with NSCLC receiving nivolumab monotherapy. Cellular changes, cytokine levels, gene expression, and polymorphisms were compared between responders and nonresponders to treatment. Findings were confirmed in additional cohorts of patients with NSCLC receiving immune checkpoint blockade.Results:We identified a genetic variant of a killer cell immunoglobulin-like receptor (KIR) KIR3DS1 that is associated with primary resistance to PD-1 blockade in patients with NSCLC. This association could be confirmed in independent cohorts of patients with NSCLC. In a multivariate analysis of the pooled cohort of 135 patients, the progression-free survival was significantly associated with presence of the KIR3DS1 allele (HR, 1.72; 95% confidence interval, 1.10–2.68; P = 0.017). No relationship was seen in cohorts of patients with NSCLC who did not receive immunotherapy. Cellular assays from patients before and during PD-1 blockade showed that resistance may be due to NK-cell dysfunction.Conclusions:We identified an association of the KIR3DS1 allelic variant with response to PD-1–targeted immunotherapy in patients with NSCLC. This finding links NK cells with response to PD-1 therapy. Although the findings are interesting, a larger analysis in a randomized trial will be needed to confirm KIRs as predictive markers for response to PD-1–targeted immunotherapy.
- Published
- 2023
- Full Text
- View/download PDF
6. 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
7. Dual TLR9 and PD-L1 targeting unleashes dendritic cells to induce durable antitumor immunity
- Author
-
Laura Fernandez-Rodriguez, Chiara Cianciaruso, Ruben Bill, Marcel P Trefny, Richard Klar, Nicole Kirchhammer, Mélanie Buchi, Julia Festag, Sven Michel, Rainer H Kohler, Elham Jones, Andre Maaske, Abhishek S Kashyap, Frank Jaschinski, Karen O Dixon, Mikael J Pittet, and Alfred Zippelius
- Subjects
Pharmacology ,Cancer Research ,Oncology ,Immunology ,Molecular Medicine ,Immunology and Allergy - Abstract
BackgroundAlthough 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.MethodsWe 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 performedin vitroandin vivostudies 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.ResultsIM-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, andBatf3, a transcription factor required for DC development.ConclusionsBy 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
8. 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
9. A Variant of a Killer Cell Immunoglobulin-like Receptor Is Associated with Resistance to PD-1 Blockade in Lung Cancer
- Author
-
Marcel P. Trefny, Zlatko Trajanoski, Spasenija Savic, Petra Herzig, Sacha I. Rothschild, Kirsten D. Mertz, Stefan Schaub, Fiamma Berner, Franziska Uhlenbrock, Martin Früh, Benjamin Kasenda, Ilaria Alborelli, Lukas Flatz, Dietmar Rieder, Florian Geier, Severin Poechtrager, Michal A. Stanczak, Philip Jermann, Heinz Läubli, Monika Kaiser, Alfred Zippelius, Abhishek S. Kashyap, Daniela S. Thommen, and Frank Stenner
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Receptors, KIR3DS1 ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Killer-cell immunoglobulin-like receptor ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,medicine ,Humans ,Lung cancer ,Aged ,Aged, 80 and over ,biology ,business.industry ,Genetic Variation ,Immunotherapy ,Middle Aged ,medicine.disease ,Immune checkpoint ,Blockade ,Treatment Outcome ,030104 developmental biology ,Cytokine ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Nivolumab ,Antibody ,business - Abstract
Purpose: PD-(L)1–blocking antibodies have clinical activity in metastatic non–small cell lung cancer (NSCLC) and mediate durable tumor remissions. However, the majority of patients are resistant to PD-(L)1 blockade. Understanding mechanisms of primary resistance may allow prediction of clinical response and identification of new targetable pathways. Experimental Design: Peripheral blood mononuclear cells were collected from 35 patients with NSCLC receiving nivolumab monotherapy. Cellular changes, cytokine levels, gene expression, and polymorphisms were compared between responders and nonresponders to treatment. Findings were confirmed in additional cohorts of patients with NSCLC receiving immune checkpoint blockade. Results: We identified a genetic variant of a killer cell immunoglobulin-like receptor (KIR) KIR3DS1 that is associated with primary resistance to PD-1 blockade in patients with NSCLC. This association could be confirmed in independent cohorts of patients with NSCLC. In a multivariate analysis of the pooled cohort of 135 patients, the progression-free survival was significantly associated with presence of the KIR3DS1 allele (HR, 1.72; 95% confidence interval, 1.10–2.68; P = 0.017). No relationship was seen in cohorts of patients with NSCLC who did not receive immunotherapy. Cellular assays from patients before and during PD-1 blockade showed that resistance may be due to NK-cell dysfunction. Conclusions: We identified an association of the KIR3DS1 allelic variant with response to PD-1–targeted immunotherapy in patients with NSCLC. This finding links NK cells with response to PD-1 therapy. Although the findings are interesting, a larger analysis in a randomized trial will be needed to confirm KIRs as predictive markers for response to PD-1–targeted immunotherapy.
- Published
- 2019
- Full Text
- View/download PDF
10. 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
11. 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
12. 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
13. Hepatic stellate cells suppress NK cell-sustained breast cancer dormancy
- Author
-
Ryoko Okamoto, Luigi Terracciano, Mohamed Bentires-Alj, Marcel P. Trefny, Marcus Vetter, Alfred Zippelius, Joao C. Guimaraes, Walter P. Weber, Katrin Volkmann, Christian Kurzeder, Duvini De Silva, Ana Luísa Correia, Alexander Schmidt, Kirsten D. Mertz, Sandro Bruno, and Priska Auf der Maur
- Subjects
0301 basic medicine ,Chemokine ,Multidisciplinary ,medicine.medical_treatment ,Cancer ,Immunotherapy ,Biology ,medicine.disease ,CXCR4 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cell quiescence ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,biology.protein ,Hepatic stellate cell ,Dormancy ,Cancer dormancy - Abstract
The persistence of undetectable disseminated tumour cells (DTCs) after primary tumour resection poses a major challenge to effective cancer treatment1–3. These enduring dormant DTCs are seeds of future metastases, and the mechanisms that switch them from dormancy to outgrowth require definition. Because cancer dormancy provides a unique therapeutic window for preventing metastatic disease, a comprehensive understanding of the distribution, composition and dynamics of reservoirs of dormant DTCs is imperative. Here we show that different tissue-specific microenvironments restrain or allow the progression of breast cancer in the liver—a frequent site of metastasis4 that is often associated with a poor prognosis5. Using mouse models, we show that there is a selective increase in natural killer (NK) cells in the dormant milieu. Adjuvant interleukin-15-based immunotherapy ensures an abundant pool of NK cells that sustains dormancy through interferon-γ signalling, thereby preventing hepatic metastases and prolonging survival. Exit from dormancy follows a marked contraction of the NK cell compartment and the concurrent accumulation of activated hepatic stellate cells (aHSCs). Our proteomics studies on liver co-cultures implicate the aHSC-secreted chemokine CXCL12 in the induction of NK cell quiescence through its cognate receptor CXCR4. CXCL12 expression and aHSC abundance are closely correlated in patients with liver metastases. Our data identify the interplay between NK cells and aHSCs as a master switch of cancer dormancy, and suggest that therapies aimed at normalizing the NK cell pool might succeed in preventing metastatic outgrowth. Liver-resident natural killer (NK) cells sustain the dormancy of disseminated breast cancer cells, and a decrease in NK cells and increase in activated hepatic stellate cells is associated with the formation of liver metastases.
- Published
- 2021
14. 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
15. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small cell lung cancer treated with PD-1 blockade
- Author
-
Viktor H. Koelzer, Didier Lardinois, Kirsten D. Mertz, Spasenija Savic Prince, Ping-Chih Ho, Jonathan C Hanhart, Daniela S. Thommen, Alfred Zippelius, Catherine Schill, Marcel P. Trefny, Vaios Karanikas, Sarah Dimeloe, Ton N. Schumacher, Mark Wiese, Andreas Roller, Petra Herzig, Christoph Hess, Christian Klein, Anna Kiialainen, University of Zurich, and Thommen, Daniela S
- Subjects
0301 basic medicine ,Lung Neoplasms ,Transcription, Genetic ,medicine.medical_treatment ,T cell ,Programmed Cell Death 1 Receptor ,610 Medicine & health ,Biology ,CD8-Positive T-Lymphocytes ,NSCLC ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Lymphocytes, Tumor-Infiltrating ,1300 General Biochemistry, Genetics and Molecular Biology ,T-Lymphocyte Subsets ,10049 Institute of Pathology and Molecular Pathology ,Carcinoma, Non-Small-Cell Lung ,medicine ,Cytotoxic T cell ,Humans ,Lung cancer ,T cell exhaustion ,chemokine ,General Medicine ,T lymphocyte ,Immunotherapy ,CXCL13 ,immune checkpoint blockade ,medicine.disease ,Lipid Metabolism ,Chemokine CXCL13 ,3. Good health ,Mitochondria ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,Glucose ,Phenotype ,Virus Diseases ,030220 oncology & carcinogenesis ,IL-10 ,Chronic Disease ,Cancer research ,CD8-Positive T-Lymphocytes/immunology ,CD8-Positive T-Lymphocytes/ultrastructure ,Carcinoma, Non-Small-Cell Lung/genetics ,Carcinoma, Non-Small-Cell Lung/immunology ,Chemokine CXCL13/metabolism ,Glucose/metabolism ,Lung Neoplasms/genetics ,Lung Neoplasms/immunology ,Lymphocytes, Tumor-Infiltrating/immunology ,Mitochondria/metabolism ,Mitochondria/ultrastructure ,Programmed Cell Death 1 Receptor/metabolism ,T-Lymphocyte Subsets/immunology ,Virus Diseases/immunology ,CD8 - Abstract
Evidence from mouse chronic viral infection models suggests that CD8 + T cell subsets characterized by distinct expression levels of the receptor PD-1 diverge in their state of exhaustion and potential for reinvigoration by PD-1 blockade. However, it remains unknown whether T cells in human cancer adopt a similar spectrum of exhausted states based on PD-1 expression levels. We compared transcriptional, metabolic and functional signatures of intratumoral CD8 + T lymphocyte populations with high (PD-1 T ), intermediate (PD-1 N ) and no PD-1 expression (PD-1 - ) from non-small-cell lung cancer patients. PD-1 T T cells showed a markedly different transcriptional and metabolic profile from PD-1 N and PD-1 - lymphocytes, as well as an intrinsically high capacity for tumor recognition. Furthermore, while PD-1 T lymphocytes were impaired in classical effector cytokine production, they produced CXCL13, which mediates immune cell recruitment to tertiary lymphoid structures. Strikingly, the presence of PD-1 T cells was strongly predictive for both response and survival in a small cohort of non-small-cell lung cancer patients treated with PD-1 blockade. The characterization of a distinct state of tumor-reactive, PD-1-bright lymphocytes in human cancer, which only partially resembles that seen in chronic infection, provides potential avenues for therapeutic intervention.
- Published
- 2018
16. Author Correction: Hepatic stellate cells suppress NK cell-sustained breast cancer dormancy
- Author
-
Alexander Schmidt, Joao C. Guimaraes, Christian Kurzeder, Marcus Vetter, Mohamed Bentires-Alj, Alfred Zippelius, Duvini De Silva, Kirsten D. Mertz, Ana Luísa Correia, Walter P. Weber, Marcel P. Trefny, Sandro Bruno, Luigi Terracciano, Katrin Volkmann, Ryoko Okamoto, and Priska Auf der Maur
- Subjects
Multidisciplinary ,Breast cancer ,medicine.anatomical_structure ,business.industry ,Cell ,Hepatic stellate cell ,Cancer research ,Medicine ,Dormancy ,Cancer Microenvironment ,business ,medicine.disease - Published
- 2021
- Full Text
- View/download PDF
17. PD-1+ natural killer cells in human non-small cell lung cancer can be activated by PD-1/PD-L1 blockade
- Author
-
Michal A. Stanczak, Franziska Uhlenbrock, Monika Kaiser, Heinz Läubli, Spasenija Savic, Petra Herzig, Mark Wiese, Marcel P. Trefny, Alfred Zippelius, and Didier Lardinois
- Subjects
0303 health sciences ,Cancer Research ,Innate immune system ,biology ,Chemistry ,medicine.medical_treatment ,Immunology ,Cell ,Immune checkpoint ,3. Good health ,Blockade ,Immunological synapse ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,Cancer immunotherapy ,030220 oncology & carcinogenesis ,PD-L1 ,medicine ,biology.protein ,Cancer research ,Immunology and Allergy ,Receptor ,030304 developmental biology - Abstract
Natural killer (NK) cells are critically involved in anti-tumor immunity by targeting tumor cells. In this study, we show that intratumoral NK cells from NSCLC patients expressed elevated levels of the immune checkpoint receptor PD-1 on their cell surface. In contrast to the expression of activating receptors, PD-1; +; NK cells co-expressed more inhibitory receptors compared to PD-1; -; NK cells. Intratumoral NK cells were less functional compared to peripheral NK cells, and this dysfunction correlated with PD-1 expression. Tumor cells expressing PD-L1 inhibited the functionality of PD-1; +; NK cells in ex vivo models and induced PD-1 clustering at the immunological synapse between NK cells and tumor cells. Notably, treatment with PD-1 blockade was able to reverse PD-L1-mediated inhibition of PD-1; +; NK cells. Our findings highlight the therapeutic potential of PD-1; +; NK cells in immune checkpoint blockade and could guide the development of NK cell-stimulating agents in combination with PD-1 blockade.
- Published
- 2020
18. Hepatic stellate cells suppress NK cell-sustained breast cancer dormancy
- Author
-
Ana Luísa, Correia, Joao C, Guimaraes, Priska, Auf der Maur, Duvini, De Silva, Marcel P, Trefny, Ryoko, Okamoto, Sandro, Bruno, Alexander, Schmidt, Kirsten, Mertz, Katrin, Volkmann, Luigi, Terracciano, Alfred, Zippelius, Marcus, Vetter, Christian, Kurzeder, Walter Paul, Weber, and Mohamed, Bentires-Alj
- Subjects
Proteomics ,Mice, Inbred BALB C ,Liver Neoplasms ,Breast Neoplasms ,Mice, SCID ,Neoplasms, Experimental ,Chemokine CXCL12 ,Coculture Techniques ,Killer Cells, Natural ,Interferon-gamma ,Mice ,Mice, Inbred NOD ,Cell Line, Tumor ,Hepatic Stellate Cells ,Tumor Microenvironment ,Animals ,Humans ,Female ,Immunotherapy ,Neoplasm Metastasis ,Transcriptome - Abstract
The persistence of undetectable disseminated tumour cells (DTCs) after primary tumour resection poses a major challenge to effective cancer treatment
- Published
- 2019
19. PD-1
- Author
-
Marcel P, Trefny, Monika, Kaiser, Michal A, Stanczak, Petra, Herzig, Spasenija, Savic, Mark, Wiese, Didier, Lardinois, Heinz, Läubli, Franziska, Uhlenbrock, and Alfred, Zippelius
- Subjects
Adult ,Aged, 80 and over ,Male ,Lung Neoplasms ,Programmed Cell Death 1 Receptor ,Antibodies, Monoclonal ,Middle Aged ,Prognosis ,B7-H1 Antigen ,Killer Cells, Natural ,Carcinoma, Non-Small-Cell Lung ,Case-Control Studies ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Humans ,Female ,Immunotherapy ,Aged ,Follow-Up Studies - Abstract
Natural killer (NK) cells are critically involved in anti-tumor immunity by targeting tumor cells. In this study, we show that intratumoral NK cells from NSCLC patients expressed elevated levels of the immune checkpoint receptor PD-1 on their cell surface. In contrast to the expression of activating receptors, PD-1
- Published
- 2019
20. CD36-mediated metabolic adaptation supports regulatory T cell survival and function in tumors
- Author
-
Haiping Wang, Taha Merghoub, Sarah-Maria Fendt, Camilla Jandus, Ping-Chih Ho, Xin Xie, Fabien Franco, Juan Fernández-García, Etienne Meylan, Ira Goldberg, Chin Hsien Tsai, Yao Chen Tsui, Isabell Schulze, Roy L. Silverstein, Roberta Zappasodi, Jedd D. Wolchok, Marcel P. Trefny, Alfred Zippelius, Florence Picard, and Syed Raza Mohmood
- Subjects
0301 basic medicine ,CD36 Antigens ,Male ,FITNESS ,muscle ,medicine.medical_treatment ,Apoptosis ,medicine.disease_cause ,THERAPY ,T-Lymphocytes, Regulatory ,Autoimmunity ,Mice ,0302 clinical medicine ,Cancer immunotherapy ,Neoplasms ,foxp3 ,Tumor Microenvironment ,Immunology and Allergy ,Homeostasis ,Mice, Knockout ,depletion ,hemic and immune systems ,MUSCLE ,fitness ,medicine.anatomical_structure ,Female ,Immunotherapy ,DEPLETION ,Life Sciences & Biomedicine ,Reprogramming ,Signal Transduction ,FOXP3 ,Regulatory T cell ,REPROGRAMS ,Immunology ,chemical and pharmacologic phenomena ,macrophage ,Biology ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Animals ,Apoptosis/immunology ,CD36 Antigens/deficiency ,CD36 Antigens/genetics ,CD36 Antigens/immunology ,Homeostasis/immunology ,Humans ,Lipid Metabolism/genetics ,Lymphocytes, Tumor-Infiltrating/immunology ,Mice, Inbred C57BL ,Neoplasms/immunology ,Neoplasms/metabolism ,Neoplasms/pathology ,PPAR-beta/immunology ,Signal Transduction/immunology ,T-Lymphocytes, Regulatory/immunology ,T-Lymphocytes, Regulatory/metabolism ,T-Lymphocytes, Regulatory/pathology ,Tumor Microenvironment/immunology ,PPAR-beta ,therapy ,Tumor microenvironment ,Science & Technology ,tregs ,Lipid Metabolism ,reprograms ,030104 developmental biology ,Cell culture ,Cancer research ,MACROPHAGE ,TREGS ,030215 immunology - Abstract
Depleting regulatory T cells (T-reg cells) to counteract immunosuppressive features of the tumor microenvironment (TME) is an attractive strategy for cancer treatment; however, autoimmunity due to systemic impairment of their suppressive function limits its therapeutic potential. Elucidating approaches that specifically disrupt intratumoral T-reg cells is direly needed for cancer immunotherapy. We found that CD36 was selectively upregulated in intrautumoral T-reg cells as a central metabolic modulator. CD36 fine-tuned mitochondrial fitness via peroxisome proliferator-activated receptor-beta signaling, programming T-reg cells to adapt to a lactic acid-enriched TME. Genetic ablation of Cd36 in T-reg cells suppressed tumor growth accompanied by a decrease in intratumoral T-reg cells and enhancement of antitumor activity in tumor-infiltrating lymphocytes without disrupting immune homeostasis. Furthermore, CD36 targeting elicited additive antitumor responses with anti-programmed cell death protein 1 therapy. Our findings uncover the unexplored metabolic adaptation that orchestrates the survival and functions of intratumoral T-reg cells, and the therapeutic potential of targeting this pathway for reprogramming the TME., Tumor environments are highly acidic due to high concentrations of lactic acid. Ho and colleagues report that tumor-infiltrating regulatory T cells adapt to this tumor environment by upregulating expression of CD36, which allows them to use fatty acids to fuel their metabolism.
- Published
- 2019
21. 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
22. Successful anti-PD-1 cancer immunotherapy requires T cell-dendritic cell crosstalk involving the cytokines IFN-γ and IL-12
- Author
-
Christopher G. Twitty, Sarah Warren, Robert H. Pierce, Mai H. Le, Christopher Garris, Adil Daud, Alfred Zippelius, Erica Browning, Ralph Weissleder, Mikael J. Pittet, Alain Algazi, Gordon J. Freeman, Sean P. Arlauckas, Cecile Piot, Sara I. Pai, Rainer H. Kohler, Camilla Engblom, SuFey Ong, Seth B. Garren, Christina Pfirschke, Marcel P. Trefny, Jeremy Gungabeesoon, and Marie Siwicki
- Subjects
0301 basic medicine ,Male ,Monoclonal/administration & dosage/immunology ,medicine.medical_treatment ,T-Lymphocytes ,Programmed Cell Death 1 Receptor ,Inbred C57BL ,Transgenic ,Mice ,checkpoint ,0302 clinical medicine ,Cancer immunotherapy ,Neoplasms ,NF-kappa B/immunology/metabolism ,Monoclonal ,80 and over ,2.1 Biological and endogenous factors ,Immunology and Allergy ,Aetiology ,IFN-γ ,Aged, 80 and over ,NF-kappa B ,Antibodies, Monoclonal ,Middle Aged ,Interleukin-12 ,Crosstalk (biology) ,medicine.anatomical_structure ,Infectious Diseases ,IL-12 ,030220 oncology & carcinogenesis ,Interleukin 12 ,Immunotherapy/methods ,Interleukin-12/administration & dosage/immunology/metabolism ,Female ,immunotherapy ,Immunotherapy ,Signal Transduction ,Adult ,dendritic cell ,T cell ,Immunology ,Interferon-gamma/immunology/metabolism ,Mice, Transgenic ,Biology ,Signal Transduction/drug effects/immunology ,Antibodies ,Article ,Dendritic Cells/immunology/metabolism ,Vaccine Related ,03 medical and health sciences ,Interferon-gamma ,Programmed Cell Death 1 Receptor/antagonists & inhibitors/immunology/metabolism ,T-Lymphocytes/drug effects/immunology/metabolism ,medicine ,cancer ,Animals ,Humans ,Aged ,Inflammatory and immune system ,Dendritic cell ,Dendritic Cells ,non-canonical NF-κB ,NFKB1 ,Immune checkpoint ,Mice, Inbred C57BL ,030104 developmental biology ,Cancer research ,anti-PD-1 ,Immunization ,Neoplasms/immunology/metabolism/therapy - Abstract
Anti-PD-1 immune checkpoint blockers can induce sustained clinical responses in cancer but how they function invivo remains incompletely understood. Here, we combined intravital real-time imaging with single-cell RNA sequencing analysis and mouse models to uncover anti-PD-1 pharmacodynamics directly within tumors. We showed that effective antitumor responses required a subset of tumor-infiltrating dendritic cells (DCs), which produced interleukin 12 (IL-12). These DCs did not bind anti-PD-1 but produced IL-12 upon sensing interferon γ (IFN-γ) that was released from neighboring Tcells. In turn, DC-derived IL-12 stimulated antitumor Tcell immunity. These findings suggest that full-fledged activation of antitumor Tcells by anti-PD-1 is not direct, but rather involves Tcell:DC crosstalk and is licensed by IFN-γ and IL-12. Furthermore, we found that activating the non-canonical NF-κB transcription factor pathway amplified IL-12-producing DCs and sensitized tumors to anti-PD-1 treatment, suggesting a therapeutic strategy to improve responses to checkpoint blockade.
- Published
- 2018
23. Self-associated molecular patterns mediate cancer immune evasion by engaging Siglecs on T cells
- Author
-
Didier Lardinois, Daniela S. Thommen, Lothar Tietze, Stephan von Gunten, Kayluz Frias Boligan, Heinz-Josef Lenz, Mohammedyaseen Syedbasha, Younghan Han, Ajit Varki, Heinz Läubli, Frank Stenner, Christopher I. Amos, Daniel E. Speiser, Marcel P. Trefny, Viola Heinzelmann-Schwarz, Michael von Bergwelt-Baildon, Michal A. Stanczak, Adrian Egli, Wu Zhang, Alfred Zippelius, Alexandar Tzankov, and Shoib S. Siddiqui
- Subjects
Male ,0301 basic medicine ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,610 Medicine & health ,03 medical and health sciences ,Immune system ,Cancer immunotherapy ,Antigens, CD ,Cell Line, Tumor ,Neoplasms ,medicine ,Humans ,Receptor ,Sialic Acid Binding Immunoglobulin-like Lectins ,Polymorphism, Genetic ,biology ,Cancer ,SIGLEC ,General Medicine ,Immunotherapy ,respiratory system ,medicine.disease ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Cancer research ,Female ,Antibody ,Research Article - Abstract
First-generation immune checkpoint inhibitors, including anti–CTLA-4 and anti–programmed death 1 (anti–PD-1) antibodies, have led to major clinical progress, yet resistance frequently leads to treatment failure. Thus, new targets acting on T cells are needed. CD33-related sialic acid–binding immunoglobulin-like lectins (Siglecs) are pattern-recognition immune receptors binding to a range of sialoglycan ligands, which appear to function as self-associated molecular patterns (SAMPs) that suppress autoimmune responses. Siglecs are expressed at very low levels on normal T cells, and these receptors were not until recently considered as interesting targets on T cells for cancer immunotherapy. Here, we show an upregulation of Siglecs, including Siglec-9, on tumor-infiltrating T cells from non–small cell lung cancer (NSCLC), colorectal, and ovarian cancer patients. Siglec-9–expressing T cells coexpressed several inhibitory receptors, including PD-1. Targeting of the sialoglycan-SAMP/Siglec pathway in vitro and in vivo resulted in increased anticancer immunity. T cell expression of Siglec-9 in NSCLC patients correlated with reduced survival, and Siglec-9 polymorphisms showed association with the risk of developing lung and colorectal cancer. Our data identify the sialoglycan-SAMP/Siglec pathway as a potential target for improving T cell activation for immunotherapy.
- Published
- 2018
- Full Text
- View/download PDF
24. 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
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.