1
|
Gao F, Wang X, Qin N, Zhang M, Liao M, Zeng M, Lu D, Sha O. The analysis of cathepsin L that mediates cellular SARS-CoV-2 infection leading to COVID-19 in head and neck squamous cell carcinoma. Front Immunol 2023; 14:1156038. [PMID: 37292206 PMCID: PMC10246451 DOI: 10.3389/fimmu.2023.1156038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/09/2023] [Indexed: 06/10/2023] Open
Abstract
The vulnerability of the oral cavity to SARS-CoV-2 infection is well-known, and cancer patients are at a higher risk of COVID-19, emphasizing the need to prioritize this patient population. Head and neck squamous cell carcinoma (HNSCC) is one of the most common malignant cancers associated with early metastasis and poor prognosis. It has been established that cancerous tissues express Cathepsin L (CTSL), a proteinase that regulates cancer progression and SARS-CoV-2 entry. Therefore, it is essential to evaluate the correlation between disease outcomes and CTSL expression in cancer tissues and predict the susceptibility of cancer patients to SARS-CoV-2. In this study, we used transcriptomic and genomic data to profile CTSL expression in HNSCC and developed a CTSL signature that could reflect the response of HNSCC patients to chemotherapy and immunotherapy. Additionally, we investigated the relationship between CTSL expression and immune cell infiltration and established CTSL as a potential carcinogenic factor for HNSCC patients. These findings could aid in understanding the mechanisms underlying the increased susceptibility of HNSCC patients to SARS-CoV-2 and contribute to the development of therapy for both HNSCC and COVID-19.
Collapse
Affiliation(s)
- Feng Gao
- School of Dentistry, Institute of Stomatological Research, Medical School, Shenzhen University, Shenzhen, China
| | - Xia Wang
- Medical School, Shenzhen University, Shenzhen, China
| | - Nianhong Qin
- Medical School, Shenzhen University, Shenzhen, China
| | - Mingxia Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Mingfeng Liao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, China
| | - Meiqi Zeng
- School of Dentistry, Institute of Stomatological Research, Medical School, Shenzhen University, Shenzhen, China
| | - Desheng Lu
- Medical School, Shenzhen University, Shenzhen, China
| | - Ou Sha
- School of Dentistry, Institute of Stomatological Research, Medical School, Shenzhen University, Shenzhen, China
| |
Collapse
|
2
|
Li Q, Chen W, Li Q, Mao J, Chen X. A novel neutrophil extracellular trap signature to predict prognosis and immunotherapy response in head and neck squamous cell carcinoma. Front Immunol 2022; 13:1019967. [PMID: 36225931 PMCID: PMC9549764 DOI: 10.3389/fimmu.2022.1019967] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/06/2022] [Indexed: 12/02/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most common malignant cancers, and patients with HNSCC possess early metastases and poor prognosis. Systematic therapies (including chemotherapy, targeted therapy, and immunotherapy) are generally applied in the advanced/late stages of HNSCC, but primary and acquired resistance eventually occurs. At present, reliable biomarkers to predict the prognosis of HNSCC have not been completely identified. Recent studies have shown that neutrophil extracellular traps (NETs) are implicated in cancer progression, metastasis and cancer immune response, and NET-related gene signatures are associated with the prognosis of patients with several human cancers. To explore whether NET-related genes play crucial roles in HNSCC, we have performed systematic analysis and reported several findings in the current study. Firstly, we identified seven novel NET-related genes and developed a NET-score signature, which was highly associated with the clinicopathological and immune traits of the HNSCC patients. Then, we, for the first time, found that NIFK was significantly upregulated in HNSCC patient samples, and its levels were significantly linked to tumor malignancy and immune status. Moreover, functional experiments confirmed that NIFK was required for HNSCC cell proliferation and metastasis. Altogether, this study has identified a novel NET-score signature based on seven novel NET-related genes to predict the prognosis of HNSCC and NIFK has also explored a new method for personalized chemo-/immuno-therapy of HNSCC.
Collapse
Affiliation(s)
- Qilin Li
- Department of Stomatology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Weimin Chen
- Department of Stomatology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Qiuhui Li
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- *Correspondence: Xin Chen, ; Qiuhui Li, ; Jing Mao,
| | - Jing Mao
- Department of Stomatology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
- *Correspondence: Xin Chen, ; Qiuhui Li, ; Jing Mao,
| | - Xin Chen
- Department of Oncology, Tongji Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xin Chen, ; Qiuhui Li, ; Jing Mao,
| |
Collapse
|
3
|
Rexach JE, Polioudakis D, Yin A, Swarup V, Chang TS, Nguyen T, Sarkar A, Chen L, Huang J, Lin LC, Seeley W, Trojanowski JQ, Malhotra D, Geschwind DH. Tau Pathology Drives Dementia Risk-Associated Gene Networks toward Chronic Inflammatory States and Immunosuppression. Cell Rep 2020; 33:108398. [PMID: 33207193 PMCID: PMC7842189 DOI: 10.1016/j.celrep.2020.108398] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 06/29/2020] [Accepted: 10/26/2020] [Indexed: 12/31/2022] Open
Abstract
To understand how neural-immune-associated genes and pathways contribute to neurodegenerative disease pathophysiology, we performed a systematic functional genomic analysis in purified microglia and bulk tissue from mouse and human AD, FTD, and PSP. We uncover a complex temporal trajectory of microglial-immune pathways involving the type 1 interferon response associated with tau pathology in the early stages, followed by later signatures of partial immune suppression and, subsequently, the type 2 interferon response. We find that genetic risk for dementias shows disease-specific patterns of pathway enrichment. We identify drivers of two gene co-expression modules conserved from mouse to human, representing competing arms of microglial-immune activation (NAct) and suppression (NSupp) in neurodegeneration. We validate our findings by using chemogenetics, experimental perturbation data, and single-cell sequencing in post-mortem brains. Our results refine the understanding of stage- and disease-specific microglial responses, implicate microglial viral defense pathways in dementia pathophysiology, and highlight therapeutic windows. Rexach et al. use transcriptional network analysis to define dynamic microglial transitions across neurodegeneration, discovering that three dementias with tau pathology involve dysregulated microglial viral and antiviral pathways. Bio-informatics coupled with experimental validation identifies regulatory drivers, implicating double-stranded RNA and interferon-response genes as drivers of early immune suppression in disease.
Collapse
Affiliation(s)
- Jessica E Rexach
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Damon Polioudakis
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Anna Yin
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Vivek Swarup
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Timothy S Chang
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tam Nguyen
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Arjun Sarkar
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lawrence Chen
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jerry Huang
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Li-Chun Lin
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - William Seeley
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - John Q Trojanowski
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dheeraj Malhotra
- Neuroscience and Rare Diseases, Roche Pharma Research and Early Development, F. Hoffman-LaRoche, Basel, Switzerland
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute of Precision Health, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
4
|
Socodato R, Henriques JF, Portugal CC, Almeida TO, Tedim-Moreira J, Alves RL, Canedo T, Silva C, Magalhães A, Summavielle T, Relvas JB. Daily alcohol intake triggers aberrant synaptic pruning leading to synapse loss and anxiety-like behavior. Sci Signal 2020; 13:13/650/eaba5754. [PMID: 32963013 DOI: 10.1126/scisignal.aba5754] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alcohol abuse adversely affects the lives of millions of people worldwide. Deficits in synaptic transmission and in microglial function are commonly found in human alcohol abusers and in animal models of alcohol intoxication. Here, we found that a protocol simulating chronic binge drinking in male mice resulted in aberrant synaptic pruning and substantial loss of excitatory synapses in the prefrontal cortex, which resulted in increased anxiety-like behavior. Mechanistically, alcohol intake increased the engulfment capacity of microglia in a manner dependent on the kinase Src, the subsequent activation of the transcription factor NF-κB, and the consequent production of the proinflammatory cytokine TNF. Pharmacological blockade of Src activation or of TNF production in microglia, genetic ablation of Tnf, or conditional ablation of microglia attenuated aberrant synaptic pruning, thereby preventing the neuronal and behavioral effects of the alcohol. Our data suggest that aberrant pruning of excitatory synapses by microglia may disrupt synaptic transmission in response to alcohol abuse.
Collapse
Affiliation(s)
- Renato Socodato
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Joana F Henriques
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Camila C Portugal
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Tiago O Almeida
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Joana Tedim-Moreira
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Renata L Alves
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Teresa Canedo
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.,Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Cátia Silva
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Teresa Summavielle
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal. .,Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
5
|
Yang Z, Liao J, Cullen KJ, Dan H. Inhibition of IKKβ/NF-κB signaling pathway to improve Dasatinib efficacy in suppression of cisplatin-resistant head and neck squamous cell carcinoma. Cell Death Discov 2020; 6:36. [PMID: 32435511 PMCID: PMC7229171 DOI: 10.1038/s41420-020-0270-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 01/17/2020] [Accepted: 02/04/2020] [Indexed: 02/06/2023] Open
Abstract
Proto-oncogene tyrosine-protein kinase Src plays an important role in Head and Neck Squamous Cell Carcinoma (HNSCC). However, the FDA-approved SRC inhibitor Dasatinib shows very limited efficacy in HNSCC clinical trials, even though Dasatinib can completely inhibit SRC in the laboratory setting. These results suggest that SRC inhibition can cause compensatory up-regulation and/or activation of other survival pathways, which suggests that co-targeting of SRC and the potential signaling pathways may improve the Dasatinib efficacy. In this study, we investigated the role of IKKβ/NF-κB in regulation of the sensitivity of cisplatin-resistant HNSCC to Dasatinib. Additionally, we wished to determine whether inhibition of the IKKβ/NF-κB signaling pathway could enhance Dasatinib efficacy to inhibit cisplatin-resistant HNSCC without the use of cisplatin. Previous studies have shown that ETS-1 is a crucial SRC effector protein that regulates cancer cell proliferation, anti-apoptosis, and metastasis. We found that SRC kinase inhibition by Dasatinib decreased ETS-1 expression but caused elevation of IKKβ/NF-κB signaling in multiple cisplatin-resistant HNSCC. Interestingly, inhibition of IKKβ/NF-κB by CmpdA (Bay65-1942), a recently identified IKKβ inhibitor, also led to a decrease in ETS-1 levels. Moreover, the knockdown of IKK, but not NF-κB, dramatically decreased ETS-1 expression. In addition, IKKβ and ETS-1 interacted in cisplatin-resistant HNSCC. These data demonstrated cross-talk between SRC and IKK to regulate NF-κB and ETS-1. Furthermore, we found that simultaneous inhibition of SRC and IKKβ through a Dasatinib and CmpdA combination synergistically inhibited NF-κB activation and ETS-1expression, suppressed cell proliferation, and induced apoptosis. Taken together, our data indicate that SRC and IKKβ play crucial roles in cisplatin-resistant HNSCCC and co-targeting SRC and IKKβ could be an effective strategy to treat cisplatin-resistant HNSCC.
Collapse
Affiliation(s)
- Zejia Yang
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - Jipei Liao
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - Kevin J. Cullen
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - Hancai Dan
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD USA
| |
Collapse
|
6
|
Adhikaree J, Franks HA, Televantos C, Vaghela P, Kaur AP, Walker D, Schmitz M, Jackson AM, Patel PM. Impaired circulating myeloid CD1c+ dendritic cell function in human glioblastoma is restored by p38 inhibition - implications for the next generation of DC vaccines. Oncoimmunology 2019; 8:1593803. [PMID: 31143512 PMCID: PMC6527277 DOI: 10.1080/2162402x.2019.1593803] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 02/22/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023] Open
Abstract
Current treatments for glioblastoma (GBM) have limited efficacy and significant morbidity and therefore new strategies are urgently needed. Dendritic cells have the power to create anti-tumor immune responses. The greater potency of circulating dendritic cells (DC) over laboratory-generated monocyte-derived DC makes them exciting new immunotherapeutic candidates. To determine the immune status of GBM patients we initially investigated the frequency and function of circulating DC subsets. Furthermore, we tested the therapeutic potential of inhibiting the p38 mitogen-activated protein kinase pathway (p38i) in circulating DC to overcome DC dysfunction. GBM patients (n = 16) had significantly reduced numbers of the major myeloid circulating dendritic cell (cDC2) and plasmacytoid DC vs healthy controls; 1736 vs 4975 (p = 0.028) and 893 vs 2287 cells/mL (P = <0.001) respectively. This inversely correlated with dexamethasone (Dex) dose in a log-linear model, and disease status. Patients' cDC2 were immature with impaired interleukin (IL)-12 secretion, reduced IL-12:IL-10 ratio, and low HLA-DR and CD86 expression. Exposure of healthy donor cDC2 to Dex or GBM cell lysate resulted in a similar low IL-12:IL-10 ratio. Inhibition of p38 restored the IL-12:IL-10 balance in Dex or tumor lysate-conditioned healthy cDC2 and enhanced T-cell proliferation and interferon-gamma (IFNγ) production. Importantly, patient-derived cDC2 showed a similar reversal of DC dysfunction with p38i. This study demonstrates the therapeutic potential of developing the next generation of DC vaccines using enhanced p38i-conditioned cDC2. We will therefore shortly embark on a clinical trial of adoptively transferred, p38 MAPK-inhibited cDC2 in adults with GBM.
Collapse
Affiliation(s)
- Jason Adhikaree
- Division of Cancer and Stem Cells, Host-Tumour Interactions Group, UK
| | - Hester Ann Franks
- Division of Cancer and Stem Cells, Host-Tumour Interactions Group, UK
| | | | - Poonam Vaghela
- Division of Cancer and Stem Cells, Host-Tumour Interactions Group, UK
| | | | - David Walker
- Children’s Brain Tumour Research Centre, University of Nottingham, Nottingham, UK
| | - Marc Schmitz
- Institute of Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
- National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | |
Collapse
|
7
|
Src promotes anti-inflammatory (M2) macrophage generation via the IL-4/STAT6 pathway. Cytokine 2018; 111:209-215. [DOI: 10.1016/j.cyto.2018.08.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/19/2018] [Accepted: 08/28/2018] [Indexed: 01/12/2023]
|
8
|
Wehrstedt S, Kubis J, Zimmermann A, Bruns H, Mayer D, Grieshober M, Stenger S. The tyrosine kinase inhibitor dasatinib reduces the growth of intracellular Mycobacterium tuberculosis despite impairing T-cell function. Eur J Immunol 2018; 48:1892-1903. [PMID: 30242834 DOI: 10.1002/eji.201847656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/10/2018] [Accepted: 09/20/2018] [Indexed: 12/31/2022]
Abstract
Tyrosine kinases are checkpoints for multiple cellular pathways and dysregulation induces malignancies, most notably chronic myeloid leukemia (CML). Inhibition of Abl-tyrosine kinases has evolved as a new concept for the treatment of CML and other malignant diseases. Due to the multiple immune-modulatory pathways controlled by tyrosine kinases, treatment with tyrosine kinase inhibitors (TKIs) will not only affect the biology of malignant cells but also modulate physiological immune functions. To understand the effects of TKIs on host defense against intracellular bacteria, we investigated the immunological impact of the dual Abl/Src TKI dasatinib on the cellular immune response to Mycobacterium tuberculosis (Mtb). Our results demonstrate that dasatinib impaired proliferation, cytokine release (IFN-γ, TNF-α, GM-CSF), expression of granulysin and degranulation of cytotoxic effector molecules of human Mtb-specific T-lymphocytes by inhibition of lymphocyte-specific protein tyrosine kinase (Lck) phosphorylation. Despite this profound inhibition of T-cell function, dasatinib suppressed growth of virulent Mtb in human macrophages co-cultured with autologous Mtb-specific T-cells (49±15%). Functional analysis suggested that growth inhibition is due to dasatinib-triggered lysosomal acidification in Mtb-infected macrophages. These results highlight the significance of innate immune responses, i.e. acidification of lysosomes, which control the multiplication of intracellular bacteria despite the lack of efficient T-cell support.
Collapse
Affiliation(s)
- Stephanie Wehrstedt
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Germany
| | - Jan Kubis
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Germany
| | - Andreas Zimmermann
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Germany
| | - Heiko Bruns
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, Germany
| | - Daniel Mayer
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Germany
| | - Mark Grieshober
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Germany
| | - Steffen Stenger
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, Germany
| |
Collapse
|
9
|
Gonçalves-de-Albuquerque CF, Rohwedder I, Silva AR, Ferreira AS, Kurz ARM, Cougoule C, Klapproth S, Eggersmann T, Silva JD, de Oliveira GP, Capelozzi VL, Schlesinger GG, Costa ER, Estrela Marins RDCE, Mócsai A, Maridonneau-Parini I, Walzog B, Macedo Rocco PR, Sperandio M, de Castro-Faria-Neto HC. The Yin and Yang of Tyrosine Kinase Inhibition During Experimental Polymicrobial Sepsis. Front Immunol 2018; 9:901. [PMID: 29760707 PMCID: PMC5936983 DOI: 10.3389/fimmu.2018.00901] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/11/2018] [Indexed: 12/29/2022] Open
Abstract
Neutrophils are the first cells of our immune system to arrive at the site of inflammation. They release cytokines, e.g., chemokines, to attract further immune cells, but also actively start to phagocytose and kill pathogens. In the case of sepsis, this tightly regulated host defense mechanism can become uncontrolled and hyperactive resulting in severe organ damage. Currently, no effective therapy is available to fight sepsis; therefore, novel treatment targets that could prevent excessive inflammatory responses are warranted. Src Family tyrosine Kinases (SFK), a group of tyrosine kinases, have been shown to play a major role in regulating immune cell recruitment and host defense. Leukocytes with SFK depletion display severe spreading and migration defects along with reduced cytokine production. Thus, we investigated the effects of dasatinib, a tyrosine kinase inhibitor, with a strong inhibitory capacity on SFKs during sterile inflammation and polymicrobial sepsis in mice. We found that dasatinib-treated mice displayed diminished leukocyte adhesion and extravasation in tumor necrosis factor-α-stimulated cremaster muscle venules in vivo. In polymicrobial sepsis, sepsis severity, organ damage, and clinical outcome improved in a dose-dependent fashion pointing toward an optimal therapeutic window for dasatinib dosage during polymicrobial sepsis. Dasatinib treatment may, therefore, provide a balanced immune response by preventing an overshooting inflammatory reaction on the one side and bacterial overgrowth on the other side.
Collapse
Affiliation(s)
- Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.,Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany.,Laboratório de Imunofarmacologia, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ina Rohwedder
- Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | - Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | | | - Angela R M Kurz
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.,Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | - Céline Cougoule
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Sarah Klapproth
- Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | - Tanja Eggersmann
- Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | - Johnatas D Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gisele Pena de Oliveira
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vera Luiza Capelozzi
- Laboratório de Genômica Pulmonar, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Edlaine Rijo Costa
- Laboratorio de Farmacologia, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rita de Cassia Elias Estrela Marins
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.,Laboratório de Pesquisa Clínica em DST e AIDS, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Attila Mócsai
- MTA-SE "Lendület" Inflammation Physiology Research Group, Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Isabelle Maridonneau-Parini
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Barbara Walzog
- Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Markus Sperandio
- Walter Brendel Centre, Department of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, Ludwig Maximilians University München, Munich, Germany
| | | |
Collapse
|
10
|
Song N, Guo H, Ren J, Hao S, Wang X. Synergistic anti-tumor effects of dasatinib and dendritic cell vaccine on metastatic breast cancer in a mouse model. Oncol Lett 2018; 15:6831-6838. [PMID: 29731862 PMCID: PMC5920940 DOI: 10.3892/ol.2018.8188] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 07/03/2017] [Indexed: 11/17/2022] Open
Abstract
Immunotherapy is currently considered as one of the major anti-tumor modalities, but its efficacy is limited. Dasatinib could improve the expansion and recruitment of cluster of differentiation (CD) 8+T cells and natural killer (NK) cells to the tumor microenvironment. The present study aimed to evaluate the synergistic anti-tumor effects of dasatinib with dendritic cell (DC) vaccine in metastatic breast cancer. Dasatinib with DC vaccine was administered to mice inoculated with 4T1 breast cancer cells. Thereafter, tumor volume was measured every other day. On day 34, lung metastasis was assessed with a stereomicroscope. Tumor proliferation and angiogenesis were determined by immunohistochemistry. Apoptosis in tumor tissues was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling. The results showed that although there were no significant differences in tumor volumes between the untreated control, DC vaccine and dasatinib groups, the tumor volume was significantly decreased in the combined treatment group compared to the other three groups. Mice in the combined treatment group showed the longest survival time, while mice treated with either single treatment had a slightly increased survival time compared to the untreated control mice. Additionally, the number of metastatic lung nodules was significantly decreased in combined treatment group compared with the dasatinib alone, DC vaccine alone and untreated control groups. Furthermore, the combined treatment group showed significantly reduced intratumoral microvessel density compared to the other three groups. In addition, the ratios of CD8+ T and NK cells were significantly increased in the combined treatment group compared with the other three groups. These results suggest that dasatinib combined with the DC vaccine is a possible modality for the treatment of metastatic breast cancer.
Collapse
Affiliation(s)
- Ningning Song
- Department of Thyroid and Breast Surgery, Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, P.R. China
| | - Hulin Guo
- Department of Integrated Traditional Chinese and Western Medicine, The Fifth People's Hospital of Qinghai Province, Xining, Qinghai 810007, P.R. China
| | - Jia Ren
- Department of Oncology, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Suhong Hao
- Department of Oncology, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Xinchao Wang
- Department of Thyroid and Breast Surgery, Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, P.R. China
| |
Collapse
|
11
|
Nishi H, Furuhashi K, Cullere X, Saggu G, Miller MJ, Chen Y, Rosetti F, Hamilton SL, Yang L, Pittman SP, Liao J, Herter JM, Berry JC, DeAngelo DJ, Zhu C, Tsokos GC, Mayadas TN. Neutrophil FcγRIIA promotes IgG-mediated glomerular neutrophil capture via Abl/Src kinases. J Clin Invest 2017; 127:3810-3826. [PMID: 28891817 DOI: 10.1172/jci94039] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/18/2017] [Indexed: 12/30/2022] Open
Abstract
The kidney glomerular capillaries are frequent sites of immune complex deposition and subsequent neutrophil accumulation in post-infectious and rapidly progressive glomerulonephritis. However, the mechanisms of neutrophil recruitment remain enigmatic, and there is no targeted therapeutic to avert this proximal event in glomerular inflammation. The uniquely human activating Fc receptor FcγRIIA promotes glomerular neutrophil accumulation and damage in anti-glomerular basement membrane-induced (anti-GBM-induced) glomerulonephritis when expressed on murine neutrophils. Here, we found that neutrophils are directly captured by immobilized IgG antibodies under physiological flow conditions in vitro through FcγRIIA-dependent, Abl/Src tyrosine kinase-mediated F-actin polymerization. Biophysical measurements showed that the lifetime of FcγRIIA-IgG bonds increased under mechanical force in an F-actin-dependent manner, which could enable the capture of neutrophils under physiological flow. Kidney intravital microscopy revealed that circulating neutrophils, which were similar in diameter to glomerular capillaries, abruptly arrested following anti-GBM antibody deposition via neutrophil FcγRIIA and Abl/Src kinases. Accordingly, inhibition of Abl/Src with bosutinib reduced FcγRIIA-mediated glomerular neutrophil accumulation and renal injury in experimental, crescentic anti-GBM nephritis. These data identify a pathway of neutrophil recruitment within glomerular capillaries following IgG deposition that may be targeted by bosutinib to avert glomerular injury.
Collapse
Affiliation(s)
- Hiroshi Nishi
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kazuhiro Furuhashi
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Xavier Cullere
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gurpanna Saggu
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mark J Miller
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yunfeng Chen
- Woodruff School of Mechanical Engineering and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Florencia Rosetti
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Samantha L Hamilton
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lihua Yang
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Spencer P Pittman
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jiexi Liao
- Woodruff School of Mechanical Engineering and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Jan M Herter
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey C Berry
- Donald Danforth Plant Science Center, St. Louis, Missouri, USA
| | - Daniel J DeAngelo
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Cheng Zhu
- Woodruff School of Mechanical Engineering and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Tanya N Mayadas
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Hu X, Han C, Jin J, Qin K, Zhang H, Li T, Li N, Cao X. Integrin CD11b attenuates colitis by strengthening Src-Akt pathway to polarize anti-inflammatory IL-10 expression. Sci Rep 2016; 6:26252. [PMID: 27188220 PMCID: PMC4870583 DOI: 10.1038/srep26252] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/28/2016] [Indexed: 12/20/2022] Open
Abstract
Interleukin-10 (IL-10) plays a central role in regulation of intestinal mucosal homeostasis and prevention of inflammatory bowel disease (IBD). We previously reported that CD11b(hi) regulatory dendritic cells (DCs) can produce more IL-10, and CD11b can negatively regulate Toll-like receptors (TLRs)-induced inflammatory responses in macrophages. However whether CD11b and its signaling can control autoimmunity via IL-10 production remains unclear. Here we found that CD11b deficient (Itgam(-/-)) mice were more susceptible to dextran sulfate sodium (DSS)-induced colitis, with more tumor necrosis factor α (TNF-α) while less IL-10 production. CD11b inhibited nuclear factor-kappa B (NF-κB) while promoted activator protein 1 (AP-1) activation through activating sarcoma oncogene (Src), leading to decreased TNF-α while increased IL-10 production. Src interacted with and promoted c-casitas B lineage lymphoma proto-oncogene (c-Cbl)-mediated degradation of the inhibitory subunit p85 of phosphatidylinositol 3-kinase (PI3K). Importantly, Src inhibitor dasatinib aggravated DSS-induced colitis by decreasing IL-10 while increasing TNF-α in vivo. Therefore, CD11b promotes IL-10 production by activating Src-Akt signal pathway. An axis of CD11b-Src pathway is important in balancing homeostasis of TLR-induced pro-inflammatory and anti-inflammatory responses.
Collapse
Affiliation(s)
- Xiang Hu
- National Key Laboratory of Medical Molecular Biology &Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Chaofeng Han
- National Key Laboratory of Medical Immunology &Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Jing Jin
- National Key Laboratory of Medical Immunology &Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Kewei Qin
- National Key Laboratory of Medical Immunology &Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Hua Zhang
- National Key Laboratory of Medical Immunology &Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Tianliang Li
- National Key Laboratory of Medical Immunology &Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Nan Li
- National Key Laboratory of Medical Immunology &Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Xuetao Cao
- National Key Laboratory of Medical Molecular Biology &Department of Immunology, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.,National Key Laboratory of Medical Immunology &Institute of Immunology, Second Military Medical University, Shanghai, China
| |
Collapse
|
13
|
Elias D, Ditzel HJ. Fyn is an important molecule in cancer pathogenesis and drug resistance. Pharmacol Res 2015; 100:250-4. [DOI: 10.1016/j.phrs.2015.08.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 08/14/2015] [Indexed: 01/06/2023]
|
14
|
Nerreter T, Köchel C, Jesper D, Eichelbrönner I, Putz E, Einsele H, Seggewiss-Bernhardt R. Dasatinib enhances migration of monocyte-derived dendritic cells by reducing phosphorylation of inhibitory immune receptors Siglec-9 and Siglec-3. Exp Hematol 2014; 42:773-82.e1-3. [PMID: 24882272 DOI: 10.1016/j.exphem.2014.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 04/18/2014] [Accepted: 05/19/2014] [Indexed: 11/19/2022]
Abstract
The SRC family of kinases (SFKs) is crucial to malignant growth, but also important for signaling in immune cells such as dendritic cells (DCs). These specialized antigen-presenting cells are essential for inducing and boosting specific T-cell responses against pathogens and malignancies. Targeted therapy with SFK inhibitors holds great promise as a direct anti-cancer treatment, but potentially also as an indirect treatment via immunomodulation. Here, we investigated whether the BCR-ABL/SRC inhibitor dasatinib would modulate the major effector functions of DCs, especially their migration, a prerequisite to interaction with lymphocytes in secondary lymphoid organs. We report for the first time that dasatinib more than doubled the number of mature human monocyte-derived DCs (moDCs) migrating toward a CCL19 gradient despite unchanged CCR7 expression when used for pretreatment. These effects were caused by dephosphorylation of SFKs, as confirmed by the specific SFK inhibitor SRC inhibitor 1, leading to dephosphorylation of the inhibitory immunoreceptors Siglec-9 and Siglec-3. The specific blocking of the latter also enhanced migration and underlined the importance of these SFK-dependent receptor systems for migration of moDCs. Dasatinib hampered the secretion of interleukin-12 by moDCs at clinically relevant concentrations. In contrast, endocytosis or boosting of cytomegalovirus-specific CD8(+) T-cell responses remained unaltered when applying dasatinib-pretreated moDCs, in line with minor effects on the expression of co-stimulatory molecules essential for DC-T cell interaction. The induction of enhanced migration of moDCs may potentially be useful in chemo-immunotherapeutic applications. Thus, the use of dasatinib or blocking Siglec antibodies as adjuvants in this setting to induce stronger immune responses is worthy of further study.
Collapse
Affiliation(s)
- Thomas Nerreter
- Immune Recovery Section, Division of Hematology, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany; Comprehensive Cancer Center Mainfranken (CCC MF), University Hospital of Würzburg, Würzburg, Germany
| | - Christoph Köchel
- Immune Recovery Section, Division of Hematology, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Daniel Jesper
- Immune Recovery Section, Division of Hematology, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Irina Eichelbrönner
- Immune Recovery Section, Division of Hematology, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Evelyn Putz
- Department of Transfusion Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Immune Recovery Section, Division of Hematology, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Ruth Seggewiss-Bernhardt
- Comprehensive Cancer Center Mainfranken (CCC MF), University Hospital of Würzburg, Würzburg, Germany.
| |
Collapse
|
15
|
Wölfl M, Greenberg PD. Antigen-specific activation and cytokine-facilitated expansion of naive, human CD8+ T cells. Nat Protoc 2014; 9:950-66. [PMID: 24675735 PMCID: PMC4312138 DOI: 10.1038/nprot.2014.064] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antigen-specific priming of human, naive T cells has been difficult to assess. Owing to the low initial frequency in the naive cell pool of specific T cell precursors, such an analysis has been obscured by the requirements for repeated stimulations and prolonged culture time. In this protocol, we describe how to evaluate antigen-specific priming of CD8(+) cells 10 d after a single specific stimulation. The assay provides reference conditions, which result in the expansion of a substantial population of antigen-specific T cells from the naive repertoire. Various conditions and modifications during the priming process (e.g., testing new cytokines, co-stimulators and so on) can now be directly compared with the reference conditions. Factors relevant to achieving effective priming include the dendritic cell preparation, the T cell preparation, the cell ratio at the time of priming, the serum source used for the experiment and the timing of addition and concentration of the cytokines used for expansion. This protocol is relevant for human immunology, vaccine biology and drug development.
Collapse
Affiliation(s)
- Matthias Wölfl
- Children's Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg, Würzburg, Germany
| | - Philip D Greenberg
- Fred Hutchinson Cancer Research Center and the Departments of Immunology and Medicine, University of Washington, Seattle, WA
| |
Collapse
|
16
|
Wolleschak D, Mack TS, Perner F, Frey S, Schnöder TM, Wagner MC, Höding C, Pils MC, Parkner A, Kliche S, Schraven B, Hebel K, Brunner-Weinzierl M, Ranjan S, Isermann B, Lipka DB, Fischer T, Heidel FH. Clinically relevant doses of FLT3-kinase inhibitors quizartinib and midostaurin do not impair T-cell reactivity and function. Haematologica 2014; 99:e90-3. [PMID: 24633870 DOI: 10.3324/haematol.2014.104331] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Denise Wolleschak
- Department of Hematology and Oncology, Center of Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Thomas S Mack
- Department of Hematology and Oncology, Center of Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Florian Perner
- Department of Hematology and Oncology, Center of Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Stephanie Frey
- Department of Hematology and Oncology, Center of Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Tina M Schnöder
- Department of Hematology and Oncology, Center of Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Marie-Christine Wagner
- Department of Hematology and Oncology, Center of Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Christine Höding
- Department of Hematology and Oncology, Center of Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Marina C Pils
- Mousepathology, Animal Experimental Unit, Helmholtz-Center for Infection Research, Braunschweig, Germany
| | - Andreas Parkner
- Department of Transfusion Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Stefanie Kliche
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Medical Center, Magdeburg, Germany Department of Immune Control, Helmholtz Center for Infection Research, Braunschwei, Germany
| | - Katrin Hebel
- Department of Experimental Pediatrics, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Monika Brunner-Weinzierl
- Department of Experimental Pediatrics, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Satish Ranjan
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Berend Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Daniel B Lipka
- Department of Hematology and Oncology, Center of Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany German Cancer Research Center (DKFZ), Division of Epigenomics and Cancer Risk Factors, Heidelberg, Germany
| | - Thomas Fischer
- Department of Hematology and Oncology, Center of Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Florian H Heidel
- Department of Hematology and Oncology, Center of Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| |
Collapse
|
17
|
Lowe DB, Bose A, Taylor JL, Tawbi H, Lin Y, Kirkwood JM, Storkus WJ. Dasatinib promotes the expansion of a therapeutically superior T-cell repertoire in response to dendritic cell vaccination against melanoma. Oncoimmunology 2014; 3:e27589. [PMID: 24734217 PMCID: PMC3984268 DOI: 10.4161/onci.27589] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 12/18/2013] [Accepted: 12/18/2013] [Indexed: 12/16/2022] Open
Abstract
Dasatinib (DAS) is a potent inhibitor of the BCR-ABL, SRC, c-KIT, PDGFR, and ephrin tyrosine kinases that has demonstrated only modest clinical efficacy in melanoma patients. Given reports suggesting that DAS enhances T cell infiltration into the tumor microenvironment, we analyzed whether therapy employing the combination of DAS plus dendritic cell (DC) vaccination would promote superior immunotherapeutic benefit against melanoma. Using a M05 (B16.OVA) melanoma mouse model, we observed that a 7-day course of orally-administered DAS (0.1 mg/day) combined with a DC-based vaccine (VAC) against the OVA257–264 peptide epitope more potently inhibited tumor growth and extended overall survival as compared with treatment with either single modality. The superior efficacy of the combinatorial treatment regimen included a reduction in hypoxic-signaling associated with reduced levels of immunosuppressive CD11b+Gr1+ myeloid-derived suppressor cells (MDSC) and CD4+Foxp3+ regulatory T (Treg) populations in the melanoma microenvironment. Furthermore, DAS + VAC combined therapy upregulated expression of Type-1 T cell recruiting CXCR3 ligand chemokines in the tumor stroma correlating with activation and recruitment of Type-1, vaccine-induced CXCR3+CD8+ tumor-infiltrating lymphocytes (TILs) and CD11c+ DC into the tumor microenvironment. The culmination of this bimodal approach was a profound “spreading” in the repertoire of tumor-associated antigens recognized by CD8+ TILs, in support of the therapeutic superiority of combined DAS + VAC immunotherapy in the melanoma setting.
Collapse
Affiliation(s)
- Devin B Lowe
- Department of Dermatology; University of Pittsburgh School of Medicine; Pittsburgh, PA USA
| | - Anamika Bose
- Department of Dermatology; University of Pittsburgh School of Medicine; Pittsburgh, PA USA
| | - Jennifer L Taylor
- Department of Dermatology; University of Pittsburgh School of Medicine; Pittsburgh, PA USA
| | - Hussein Tawbi
- Department of Medicine; University of Pittsburgh School of Medicine; Pittsburgh, PA USA ; University of Pittsburgh Cancer Institute; Pittsburgh, PA USA
| | - Yan Lin
- Department of Biostatistics; University of Pittsburgh School of Medicine; Pittsburgh, PA USA ; University of Pittsburgh Cancer Institute; Pittsburgh, PA USA
| | - John M Kirkwood
- Department of Medicine; University of Pittsburgh School of Medicine; Pittsburgh, PA USA ; University of Pittsburgh Cancer Institute; Pittsburgh, PA USA
| | - Walter J Storkus
- Department of Dermatology; University of Pittsburgh School of Medicine; Pittsburgh, PA USA ; Department of Immunology; University of Pittsburgh School of Medicine; Pittsburgh, PA USA ; University of Pittsburgh Cancer Institute; Pittsburgh, PA USA
| |
Collapse
|