51
|
Yinhang W, Wei W, Jing Z, Qing Z, Yani Z, Yangyanqiu W, Shuwen H. Biological roles of toll-like receptors and gut microbiota in colorectal cancer. Future Microbiol 2022; 17:1071-1089. [PMID: 35916158 DOI: 10.2217/fmb-2021-0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most considerably common malignancies of the alimentary system, with high mortality and incidence rates. The present study suggested that the occurrence of CRC is closely related to bacteria, as the large intestine is a gathering place for human micro-organisms. However, the nosogenesis of bacteria leading to tumorigenesis is still obscure. Recently, many studies have reported that toll-like receptors and their related molecular pathways are involved in the process of gut micro-organisms generating CRC. Gut micro-organisms can promote or inhibit the development of CRC via binding to special toll-like receptors. In this paper, the authors review the relationship among toll-like receptors, gut micro-organisms and CRC in order to provide a reference for future tumor immunotherapy and targeted therapy.
Collapse
Affiliation(s)
- Wu Yinhang
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,The Second School of Clinical Medicine, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang Province, 310053, China.,Key Laboratory of Multiomics Research & Clinical Transformation of Digestive Cancer of Huzhou,1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| | - Wu Wei
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,Key Laboratory of Multiomics Research & Clinical Transformation of Digestive Cancer of Huzhou,1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| | - Zhuang Jing
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,Key Laboratory of Multiomics Research & Clinical Transformation of Digestive Cancer of Huzhou,1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| | - Zhou Qing
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,Key Laboratory of Multiomics Research & Clinical Transformation of Digestive Cancer of Huzhou,1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| | - Zhou Yani
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,Graduate School of Medicine Faculty, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou, Zhejiang Province, 310058, China
| | - Wang Yangyanqiu
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,Graduate School of Medicine Faculty, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou, Zhejiang Province, 310058, China
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,Key Laboratory of Multiomics Research & Clinical Transformation of Digestive Cancer of Huzhou,1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| |
Collapse
|
52
|
Timperi E, Gueguen P, Molgora M, Magagna I, Kieffer Y, Lopez-Lastra S, Sirven P, Baudrin LG, Baulande S, Nicolas A, Champenois G, Meseure D, Vincent-Salomon A, Tardivon A, Laas E, Soumelis V, Colonna M, Mechta-Grigoriou F, Amigorena S, Romano E. Lipid-associated macrophages are induced by cancer-associated fibroblasts and mediate immune suppression in breast cancer. Cancer Res 2022; 82:3291-3306. [PMID: 35862581 DOI: 10.1158/0008-5472.can-22-1427] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/29/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022]
Abstract
Tumor-associated macrophages (TAM) play a detrimental role in triple-negative breast cancer (TNBC). In-depth analysis of TAM characteristics and interactions with stromal cells, such as cancer-associated fibroblast (CAF), could provide important biological and therapeutic insights. Here we identify at the single-cell level a monocyte-derived-STAB1+TREM2high lipid-associated macrophage (LAM) subpopulation with immune suppressive capacities that is expanded in patients resistant to immune checkpoint blockade (ICB). Genetic depletion of this LAM subset in mice suppressed TNBC tumor growth. Flow cytometry and bulk-RNA sequencing data demonstrated that co-culture with TNBC-derived CAFs led to reprogramming of blood monocytes towards immune suppressive STAB1+TREM2high LAMs, which inhibit T cell activation and proliferation. Cell-to-cell interaction modeling and assays in vitro demonstrated the role of the inflammatory CXCL12-CXCR4 axis in CAF-myeloid cell crosstalk and recruitment of monocytes in tumor sites. Altogether, these data suggest an inflammation model whereby monocytes recruited to the tumor via the CAF-driven CXCL12-CXCR4 axis acquire pro-tumorigenic LAM capacities to support an immunosuppressive microenvironment.
Collapse
Affiliation(s)
| | | | - Martina Molgora
- Washington University School of Medicine, St Louis, United States
| | | | | | | | | | | | | | | | | | | | | | - Anne Tardivon
- Institut Curie, Service de Radiologie, Paris, France
| | - Enora Laas
- PSL Research University, Institut Curie, Paris, F-75248, France, Paris, France, France
| | | | - Marco Colonna
- Washington University School of Medicine, St Louis, MO, USA, St Louis, United States
| | | | | | - Emanuela Romano
- Institut Curie, Center of Cancer Immunotherapy, Paris, France
| |
Collapse
|
53
|
Crame EE, Nourmohammadi S, Wardill HR, Coller JK, Bowen JM. Contribution of TLR4 to colorectal tumor microenvironment, etiology and prognosis. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04199-4. [PMID: 35841426 DOI: 10.1007/s00432-022-04199-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/07/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Toll-like receptor 4 (TLR4) is increasingly recognized for its ability to govern the etiology and prognostic outcomes of colorectal cancer (CRC) due to its profound immunomodulatory capacity. Despite widespread interest in TLR4 and CRC, no clear analysis of current literature and data exists. Therefore, translational advances have failed to move beyond conceptual ideas and suggestions. METHODS We aimed to determine the relationship between TLR4 and CRC through a systematic review and analysis of published literature and datasets. Data were extracted from nine studies that reported survival, CRC staging and tumor progression data in relation to TLR4 expression. Primary and metastatic tumor samples with associated clinical data were identified through the Cancer Genome Atlas (TCGA) database. RESULTS Systematic review identified heterogeneous relationships between TLR4 and CRC traits, with no clear theme evident across studies. A total of 448 datasets were identified through the TCGA database. Analysis of TCGA datasets revealed TLR4 mRNA expression is decreased in advanced CRC stages (P < 0.05 for normal vs Stage II, Stage III and Stage IV). Stage-dependent impact of TLR4 expression on survival outcomes were also found, with high TLR4 expression associated with poorer prognosis (stage I vs III (HR = 4.2, P = 0.008) and stage I vs IV (HR = 11.3, P < 0.001)). CONCLUSION While TLR4 mRNA expression aligned with CRC staging, it appeared to heterogeneously regulate survival outcomes depending on the stage of disease. This underscores the complex relationship between TLR4 and CRC, with unique impacts dependent on disease stage.
Collapse
Affiliation(s)
- Elise E Crame
- Discipline of Physiology, School of Biomedicine, The University of Adelaide, Level 2 Helen Mayo South, North Terrace, Adelaide, SA, 5000, Australia.
| | - Saeed Nourmohammadi
- Discipline of Physiology, School of Biomedicine, The University of Adelaide, Level 2 Helen Mayo South, North Terrace, Adelaide, SA, 5000, Australia
| | - Hannah R Wardill
- Discipline of Physiology, School of Biomedicine, The University of Adelaide, Level 2 Helen Mayo South, North Terrace, Adelaide, SA, 5000, Australia.,Precision Medicine (Cancer), The South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Janet K Coller
- Discipline of Pharmacology, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Joanne M Bowen
- Discipline of Physiology, School of Biomedicine, The University of Adelaide, Level 2 Helen Mayo South, North Terrace, Adelaide, SA, 5000, Australia
| |
Collapse
|
54
|
Hubbard JM, Tőke ER, Moretto R, Graham RP, Youssoufian H, Lőrincz O, Molnár L, Csiszovszki Z, Mitchell JL, Wessling J, Tóth J, Cremolini C. Safety and Activity of PolyPEPI1018 Combined with Maintenance Therapy in Metastatic Colorectal Cancer: an Open-Label, Multicenter, Phase Ib Study. Clin Cancer Res 2022; 28:2818-2829. [PMID: 35472243 PMCID: PMC9365360 DOI: 10.1158/1078-0432.ccr-22-0112] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/11/2022] [Accepted: 04/25/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE Although chemotherapy is standard of care for metastatic colorectal cancer (mCRC), immunotherapy has no role in microsatellite stable (MSS) mCRC, a "cold" tumor. PolyPEPI1018 is an off-the-shelf, multi-peptide vaccine derived from 7 tumor-associated antigens (TAA) frequently expressed in mCRC. This study assessed PolyPEPI1018 combined with first-line maintenance therapy in patients with MSS mCRC. PATIENTS AND METHODS Eleven patients with MSS mCRC received PolyPEPI1018 and Montanide ISA51VG adjuvant subcutaneously, combined with fluoropyrimidine/biologic following first-line induction with chemotherapy and a biologic (NCT03391232). In Part A of the study, 5 patients received a single dose; in Part B, 6 patients received up to three doses of PolyPEPI1018 every 12 weeks. The primary objective was safety; secondary objectives were preliminary efficacy, immunogenicity at peripheral and tumor level, and immune correlates. RESULTS PolyPEPI1018 vaccination was safe and well tolerated. No vaccine-related serious adverse event occurred. Eighty percent of patients had CD8+ T-cell responses against ≥3 TAAs. Increased density of tumor-infiltrating lymphocytes were detected post-treatment for 3 of 4 patients' liver biopsies, combined with increased expression of immune-related gene signatures. Three patients had objective response according to RECISTv1.1, and 2 patients qualified for curative surgery. Longer median progression-free survival for patients receiving multiple doses compared with a single dose (12.5 vs. 4.6 months; P = 0.017) suggested a dose-efficacy correlation. The host HLA genotype predicted multi-antigen-specific T-cell responses (P = 0.01) indicative of clinical outcome. CONCLUSIONS PolyPEPI1018 added to maintenance chemotherapy for patients with unresectable, MSS mCRC was safe and associated with specific immune responses and antitumor activity warranting further confirmation in a randomized, controlled setting.
Collapse
Affiliation(s)
| | - Enikő R Tőke
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Roberto Moretto
- Department of Translational Research and New Technologies in Medicine and Surgery, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | | | | | - Orsolya Lőrincz
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Levente Molnár
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Zsolt Csiszovszki
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | | | | | - József Tóth
- Treos Bio Ltd, London, United Kingdom
- Treos Bio Zrt, Veszprém, Hungary
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| |
Collapse
|
55
|
Yu G, Wang W, He X, Xu J, Xu R, Wan T, Wu Y. Synergistic Therapeutic Effects of Low Dose Decitabine and NY-ESO-1 Specific TCR-T Cells for the Colorectal Cancer With Microsatellite Stability. Front Oncol 2022; 12:895103. [PMID: 35774131 PMCID: PMC9239344 DOI: 10.3389/fonc.2022.895103] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 05/17/2022] [Indexed: 12/26/2022] Open
Abstract
Patients of colorectal cancer (CRC) with microsatellite stability (MSS) show poor clinical response and little beneficial result from the immune-checkpoint inhibitors, due to the ‘cold’ tumor microenvironment. Meanwhile, decitabine can drive the ‘cold’ microenvironment towards ‘hot’ in multiple ways, such as upregulating the tumor associated antigen (TAA) and human leukocyte antigen (HLA) molecular. NY-ESO-1, one of the most important TAAs, can be observably induced in tumors by low dose decitabine, and present itself as ideal targets for antigen specific T cell receptor engineered T (TCR-T) cells. We innovatively used a synergistic tactic, combining decitabine and NY-ESO-1 specific TCR-T cells, for fighting the MSS CRC. Firstly, we confirmed the lysing effect of the NY-ESO-1 TCR-T cells on the NY-ESO-1+ and HLA-A2+ cells in vitro and in vivo. In A375 tumor-bearing mice, the results showed that NY-ESO-1 TCR-T cell therapy could inhibit A375 tumor growth and prolonged the survival time. Furthermore, the synergistic effect of decitabine and NY-ESO-1 TCR-T cells was shown to induce an even higher percentage of tumor cells being lysed in vitro than other control groups, and more potent tumor inhibition and longer survival time were observed in vivo. The innovative synergistic therapeutic strategy of decitabine and TCR-T cells for the CRC with MSS may be also effective in the treatment of other epithelial malignancies. Decitabine may likewise be adopted in combination with other cellular immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Tao Wan
- *Correspondence: Tao Wan, ; Yanfeng Wu,
| | | |
Collapse
|
56
|
Rostamizadeh L, Molavi O, Rashid M, Ramazani F, Baradaran B, Lavasanaifar A, Lai R. Recent advances in cancer immunotherapy: Modulation of tumor microenvironment by Toll-like receptor ligands. BIOIMPACTS : BI 2022; 12:261-290. [PMID: 35677663 PMCID: PMC9124882 DOI: 10.34172/bi.2022.23896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 12/18/2022]
Abstract
![]()
Immunotherapy is considered a promising approach for cancer treatment. An important strategy for cancer immunotherapy is the use of cancer vaccines, which have been widely used for cancer treatment. Despite the great potential of cancer vaccines for cancer treatment, their therapeutic effects in clinical settings have been limited. The main reason behind the lack of significant therapeutic outcomes for cancer vaccines is believed to be the immunosuppressive tumor microenvironment (TME). The TME counteracts the therapeutic effects of immunotherapy and provides a favorable environment for tumor growth and progression. Therefore, overcoming the immunosuppressive TME can potentially augment the therapeutic effects of cancer immunotherapy in general and therapeutic cancer vaccines in particular. Among the strategies developed for overcoming immunosuppression in TME, the use of toll-like receptor (TLR) agonists has been suggested as a promising approach to reverse immunosuppression. In this paper, we will review the application of the four most widely studied TLR agonists including agonists of TLR3, 4, 7, and 9 in cancer immunotherapy.
Collapse
Affiliation(s)
- Leila Rostamizadeh
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Biotechnology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Rashid
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Ramazani
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afsaneh Lavasanaifar
- Faculty of Pharmacy and Pharmaceutical Science, University of Alberta, Edmonton, Canada
| | - Raymond Lai
- Department of Laboratory Medicine & Pathology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
57
|
5-Fluorouracil Treatment of CT26 Colon Cancer Is Compromised by Combined Therapy with IMMODIN. Int J Mol Sci 2022; 23:ijms23126374. [PMID: 35742825 PMCID: PMC9223647 DOI: 10.3390/ijms23126374] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/03/2022] [Accepted: 06/05/2022] [Indexed: 02/01/2023] Open
Abstract
Due to the physiological complexity of the tumour, a single drug therapeutic strategy may not be sufficient for effective treatment. Emerging evidence suggests that combination strategies may be important to achieve more efficient tumour responses. Different immunomodulators are frequently tested to reverse the situation for the purpose of improving immune response and minimizing chemotherapy side effects. Immodin (IM) represents an attractive alternative to complement chemotherapy, which can be used to enhance the immune system after disturbances resulting from the side effects of chemotherapy. In the presented study, a model of CT26 tumor-bearing mice was used to investigate the effect of single IM or its combination with 5-fluorouracil (5-FU) on colon cancer cells. Our results highlight that the beneficial role of IM claimed in previous studies cannot be generalised to all chemotherapeutic drugs, as 5-FU toxicity was not increased. On the contrary, the chemotherapeutic anti-cancer efficacy of 5-FU was greatly compromised when combined with IM. Indeed, the combined treatment was significantly less effective regarding the tumour growth and animal survival, most probably due to the increased number of tumour-associated macrophages, and increased 5-FU cytotoxic effect related to kidneys and the liver.
Collapse
|
58
|
Zhang J, Li Z, Chandrasekar A, Li S, Ludolph A, Boeckers TM, Huber-Lang M, Roselli F, Olde Heuvel F. Fast Maturation of Splenic Dendritic Cells Upon TBI Is Associated With FLT3/FLT3L Signaling. Front Immunol 2022; 13:824459. [PMID: 35281004 PMCID: PMC8907149 DOI: 10.3389/fimmu.2022.824459] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/31/2022] [Indexed: 12/29/2022] Open
Abstract
The consequences of systemic inflammation are a significant burden after traumatic brain injury (TBI), with almost all organs affected. This response consists of inflammation and concurrent immunosuppression after injury. One of the main immune regulatory organs, the spleen, is highly interactive with the brain. Along this brain–spleen axis, both nerve fibers as well as brain-derived circulating mediators have been shown to interact directly with splenic immune cells. One of the most significant comorbidities in TBI is acute ethanol intoxication (EI), with almost 40% of patients showing a positive blood alcohol level (BAL) upon injury. EI by itself has been shown to reduce proinflammatory mediators dose-dependently and enhance anti-inflammatory mediators in the spleen. However, how the splenic immune modulatory effect reacts to EI in TBI remains unclear. Therefore, we investigated early splenic immune responses after TBI with and without EI, using gene expression screening of cytokines and chemokines and fluorescence staining of thin spleen sections to investigate cellular mechanisms in immune cells. We found a strong FLT3/FLT3L induction 3 h after TBI, which was enhanced by EI. The FLT3L induction resulted in phosphorylation of FLT3 in CD11c+ dendritic cells, which enhanced protein synthesis, maturation process, and the immunity of dendritic cells, shown by pS6, peIF2A, MHC-II, LAMP1, and CD68 by immunostaining and TNF-α expression by in-situ hybridization. In conclusion, these data indicate that TBI induces a fast maturation and immunity of dendritic cells which is associated with FLT3/FLT3L signaling and which is enhanced by EI prior to TBI.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany
| | - Zhenghui Li
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany.,Department of Neurosurgery, Kaifeng Central Hospital, Kaifeng, China
| | - Akila Chandrasekar
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany
| | - Shun Li
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany
| | - Albert Ludolph
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE) , Ulm, Germany
| | - Tobias Maria Boeckers
- German Center for Neurodegenerative Diseases (DZNE) , Ulm, Germany.,Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital, Ulm, Germany
| | - Francesco Roselli
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE) , Ulm, Germany.,Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Florian Olde Heuvel
- Department of Neurology, Center for Biomedical Research (ZBMF), Ulm University, Ulm, Germany
| |
Collapse
|
59
|
Pretre V, Papadopoulos D, Regard J, Pelletier M, Woo J. Interleukin-1 (IL-1) and the inflammasome in cancer. Cytokine 2022; 153:155850. [PMID: 35279620 DOI: 10.1016/j.cyto.2022.155850] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/13/2022] [Accepted: 03/03/2022] [Indexed: 12/14/2022]
Abstract
Numerous preclinical and clinical studies have demonstrated the significant contribution of inflammation to the development and progression of various types of cancer. Inflammation in the tumor microenvironment mediates complex interactions between innate immunity, adaptive immunity, microbiomes and stroma, and ultimately alters the overall fitness of tumor cells at multiple stages of carcinogenesis. Malignancies are known to arise in areas of chronic inflammation and inflammation in the tumor microenvironment (often called tumor-promoting inflammation) is believed to allow cancer cells to evade immunosurveillance while promoting genetic instability, survival and progression. Among the strongest data suggesting a causal role for inflammation in cancer come from the recent CANTOS trial which demonstrated that interleukin-1β (IL-1β) inhibition with canakinumab leads to a significant, dose-dependent decrease in incident lung cancer. This observation has launched a series of additional clinical studies to understand the role of IL-1β and the inflammasome in cancer, and the clinical utility of IL-1β inhibition in different stages of lung cancer. In this article we will review recent data implicating IL-1β signaling and its upstream regulator NLRP3 in both solid tumor and hematologic malignancies. We will discuss the key preclinical observations and the current clinical landscape, and describe the pharmacologic tools which will be used to evaluate the effects of blocking tumor-promoting inflammation clinically.
Collapse
|
60
|
Le Naour J, Sztupinszki Z, Carbonnier V, Casiraghi O, Marty V, Galluzzi L, Szallasi Z, Kroemer G, Vacchelli E. A loss-of-function polymorphism in ATG16L1 compromises therapeutic outcome in head and neck carcinoma patients. Oncoimmunology 2022; 11:2059878. [PMID: 35481288 PMCID: PMC9037530 DOI: 10.1080/2162402x.2022.2059878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The anticancer immune response is shaped by immunogenic cell stress and death pathways. Thus, cancer cells can release danger-associated molecular patterns that act on pattern recognition receptors expressed by dendritic cells and their precursors to elicit an antitumor immune response. Here, we investigated the impact of single nucleotide polymorphisms (SNPs) in genes affecting this cancer-immunity dialogue in the context of head and neck squamous cell carcinoma (HNSCC). We observed that homozygosity for a loss-of-function SNP (rs2241880, leading to the substitution of a threonine residue in position 300 by an alanine) affecting autophagy related 16 like 1 (ATG16L1) is coupled to poor progression-free survival in platinum-treated HNSCC patients. This result was obtained on a cohort of patients enrolled at the Gustave Roussy Cancer Campus and was validated on an independent cohort of The Cancer Genome Atlas (TCGA). Homozygosity in rs2241880 is well known to predispose to Crohn’s disease, and epidemiological associations between Crohn’s disease and HNSCC have been reported at the levels of cancer incidence and prognosis. We speculate that rs2241880 might be partially responsible for this association.
Collapse
Affiliation(s)
- Julie Le Naour
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Université Paris Sud, Paris Saclay, Faculty of Medicine Kremlin Bicêtre, France
| | - Zsofia Sztupinszki
- Computational Health Informatics Program (CHIP), Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Vincent Carbonnier
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Université Paris Sud, Paris Saclay, Faculty of Medicine Kremlin Bicêtre, France
| | - Odile Casiraghi
- Department of Head and Neck Surgical and Medical Oncology, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - Virginie Marty
- Experimental and Translational Pathology Platform (PETRA), AMMICa Inserm US23/UMS CNRS3655, Gustave Roussy Cancer Campus, Villejuif, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Zoltan Szallasi
- Computational Health Informatics Program (CHIP), Boston Children’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Institut du Cancer Paris CARPEMAP-HP, Hôpital Européen Georges Pompidou, Pôle de Biologie, Paris, France
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Erika Vacchelli
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
61
|
Abstract
A favorable outcome of the COVID-19 crisis might be achieved with massive vaccination. The proposed vaccines contain several different vaccine active principles (VAP), such as inactivated virus, antigen, mRNA, and DNA, which are associated with either standard adjuvants or nanomaterials (NM) such as liposomes in Moderna's and BioNTech/Pfizer's vaccines. COVID-19 vaccine adjuvants may be chosen among liposomes or other types of NM composed for example of graphene oxide, carbon nanotubes, micelles, exosomes, membrane vesicles, polymers, or metallic NM, taking inspiration from cancer nano-vaccines, whose adjuvants may share some of their properties with those of viral vaccines. The mechanisms of action of nano-adjuvants are based on the facilitation by NM of targeting certain regions of immune interest such as the mucus, lymph nodes, and zones of infection or blood irrigation, the possible modulation of the type of attachment of the VAP to NM, in particular VAP positioning on the NM external surface to favor VAP presentation to antigen presenting cells (APC) or VAP encapsulation within NM to prevent VAP degradation, and the possibility to adjust the nature of the immune response by tuning the physico-chemical properties of NM such as their size, surface charge, or composition. The use of NM as adjuvants or the presence of nano-dimensions in COVID-19 vaccines does not only have the potential to improve the vaccine benefit/risk ratio, but also to reduce the dose of vaccine necessary to reach full efficacy. It could therefore ease the overall spread of COVID-19 vaccines within a sufficiently large portion of the world population to exit the current crisis.
Collapse
Affiliation(s)
- Edouard Alphandéry
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, 75005 Paris, France. .,Nanobacterie SARL, 36 Boulevard Flandrin, 75116, Paris, France.,Institute of Anatomy, UZH University of Zurich, Instiute of Anatomy, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| |
Collapse
|
62
|
Chand S, Gowen A, Savine M, Moore D, Clark A, Huynh W, Wu N, Odegaard K, Weyrich L, Bevins RA, Fox HS, Pendyala G, Yelamanchili SV. A comprehensive study to delineate the role of an extracellular vesicle-associated microRNA-29a in chronic methamphetamine use disorder. J Extracell Vesicles 2021; 10:e12177. [PMID: 34913274 PMCID: PMC8674191 DOI: 10.1002/jev2.12177] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 10/27/2021] [Accepted: 11/25/2021] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs), which express a repertoire of cargo molecules (cf. proteins, microRNA, lipids, etc.), have been garnering a prominent role in the modulation of several cellular processes. Here, using both non-human primate and rodent model systems, we provide evidence that brain-derived EV (BDE) miRNA, miR-29a-3p (mir-29a), is significantly increased during chronic methamphetamine (MA) exposure. Further, miR-29a levels show significant increase both with drug-seeking and reinstatement in a rat MA self-administration model. We also show that EV-associated miR-29a is enriched in EV pool comprising of small EVs and exomeres and further plays a critical role in MA-induced inflammation and synaptodendritic damage. Furthermore, treatment with the anti-inflammatory drug ibudilast (AV411), which is known to reduce MA relapse, decreased the expression of miR-29a and subsequently attenuated inflammation and rescued synaptodendritic injury. Finally, using plasma from MUD subjects, we provide translational evidence that EV-miR29a could potentially serve as a biomarker to detect neuronal damage in humans diagnosed with MA use disorder (MUD). In summary, our work suggests that EV-associated miR-29a-3p plays a crucial role in MUD and might be used as a potential blood-based biomarker for detecting chronic inflammation and synaptic damage.
Collapse
Affiliation(s)
- Subhash Chand
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Austin Gowen
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Mason Savine
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Dalia Moore
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Alexander Clark
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Wendy Huynh
- Department of PsychologyUniversity of Nebraska–Lincoln (UNL)LincolnNebraskaUSA
| | - Niming Wu
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Katherine Odegaard
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | | | - Rick A. Bevins
- Department of PsychologyUniversity of Nebraska–Lincoln (UNL)LincolnNebraskaUSA
| | - Howard S. Fox
- Department of Neurological SciencesUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Gurudutt Pendyala
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Sowmya V. Yelamanchili
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| |
Collapse
|
63
|
Yin H, Pu N, Chen Q, Zhang J, Zhao G, Xu X, Wang D, Kuang T, Jin D, Lou W, Wu W. Gut-derived lipopolysaccharide remodels tumoral microenvironment and synergizes with PD-L1 checkpoint blockade via TLR4/MyD88/AKT/NF-κB pathway in pancreatic cancer. Cell Death Dis 2021; 12:1033. [PMID: 34718325 PMCID: PMC8557215 DOI: 10.1038/s41419-021-04293-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 09/05/2021] [Accepted: 09/24/2021] [Indexed: 02/07/2023]
Abstract
Lipopolysaccharide (LPS) as an important inflammatory mediator activates the innate/adaptive immune system. The existence of LPS in pancreatic ductal adenocarcinoma (PDAC) has been reported, however, its biological function in PDAC remains unclear. Here, we demonstrated that circulating and tumoral LPS was significantly increased by intestinal leakage in the orthotopic murine PDAC model, and LPS administration promoted T cell infiltration but exhaustion paradoxically in the subcutaneous murine PDAC model. By bioinformatic analysis, Toll-like receptor 4 (TLR4), LPS receptor, was further found to enrich in immune tolerance signaling in PDAC tissues. Then, a significant positive correlation was found between TLR4 and programmed death ligand-1 (PD-L1) in clinical PDAC tissues, as well as serum LPS and tumoral PD-L1. Meanwhile, LPS stimulation in vitro and in vivo obviously upregulated tumor PD-L1 expression, and effectively promoted cancer cells resistance to T cell cytotoxicity. Mechanistically, the activation of TLR4/MyD88/AKT/NF-κB cascade was found to participate in LPS mediated PD-L1 transcription via binding to its promoter regions, which was enhanced by crosstalk between NF-κB and AKT pathways. Finally, PD-L1 blockade could significantly reverse LPS-induced immune escape, and synergized with LPS treatment. Taken together, LPS can remodel tumor microenvironment, and synergize with PD-L1 blockade to suppress tumor growth, which may be a promising comprehensive strategy for PDAC.
Collapse
MESH Headings
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Aged
- Animals
- B7-H1 Antigen/genetics
- B7-H1 Antigen/metabolism
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Disease Models, Animal
- Female
- Gastrointestinal Tract/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Immune Checkpoint Inhibitors/pharmacology
- Immune Evasion/drug effects
- Immune Tolerance/drug effects
- Lipopolysaccharides
- Lymphocytes, Tumor-Infiltrating/drug effects
- Male
- Mice, Inbred BALB C
- Models, Biological
- Myeloid Differentiation Factor 88/metabolism
- NF-kappa B/metabolism
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction
- Toll-Like Receptor 4/metabolism
- Transcription, Genetic/drug effects
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
- Mice
Collapse
Affiliation(s)
- Hanlin Yin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ning Pu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qiangda Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jicheng Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guochao Zhao
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xuefeng Xu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dansong Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Tiantao Kuang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dayong Jin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenhui Lou
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Wenchuan Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
64
|
Firmal P, Shah VK, Pant R, Chattopadhyay S. RING finger protein TOPORS modulates the expression of tumor suppressor SMAR1 in colorectal cancer via the TLR4-TRIF pathway. Mol Oncol 2021; 16:1523-1540. [PMID: 34689394 PMCID: PMC8978522 DOI: 10.1002/1878-0261.13126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/22/2021] [Indexed: 11/13/2022] Open
Abstract
TOP1‐binding arginine/serine‐rich protein (TOPORS), a really interesting new gene finger protein, has the ability to bind to a palindromic consensus DNA sequence that enables it to function as a potential transcriptional regulator. However, its role in regulating the transcription of cancer‐associated genes is yet to be explored. As Toll‐like receptor 4 (TLR4) agonists are known to regress solid tumors, we observed that lipopolysaccharide (LPS) induces TOPORS via a TLR4‐TIR domain‐containing adapter‐inducing interferon‐β‐dependent pathway, which in turn modulates the transcription of tumor suppressor scaffold/matrix attachment region‐binding protein 1 (SMAR1, also known as BANP). ChIP analysis showed that TOPORS binds on the SMAR1 promoter and its occupancy increases upon LPS treatment. A previous study from our laboratory revealed that SMAR1 acts as a repressor of signal transducer and activator of transcription 3 (STAT3) transcription. Tumor growth, as well as tumor‐associated macrophage polarization, depends on the status of the STAT1:STAT3 ratio. LPS‐induced SMAR1 expression decreases STAT3 expression and also skews the macrophage polarization toward M1 phenotype. In contrast, LPS failed to polarize tumor‐associated macrophages to M1 phenotype in a SMAR1‐silenced condition, which shows the involvement of SMAR1 in dictating the fate of colorectal cancer progression. Identification of the molecular mechanism behind LPS‐mediated tumor regression would be crucial for designing cancer treatment strategies involving bacterial components.
Collapse
Affiliation(s)
- Priyanka Firmal
- National Centre for Cell Science, S.P. Pune University Campus, Ganeshkhind, Pune, 411007
| | - Vibhuti Kumar Shah
- National Centre for Cell Science, S.P. Pune University Campus, Ganeshkhind, Pune, 411007
| | - Richa Pant
- National Centre for Cell Science, S.P. Pune University Campus, Ganeshkhind, Pune, 411007
| | - Samit Chattopadhyay
- National Centre for Cell Science, S.P. Pune University Campus, Ganeshkhind, Pune, 411007.,Department of Biological Sciences, BITS Pilani, K. K. Birla Goa Campus, NH 17B, Zuarinagar, Goa, 403726.,Indian Institute of Chemical Biology, Raja S C Mullick Road, Jadavpur, Kolkata, 700032
| |
Collapse
|
65
|
Liang F, Rezapour A, Szeponik L, Alsén S, Wettergren Y, Bexe Lindskog E, Quiding-Järbrink M, Yrlid U. Antigen Presenting Cells from Tumor and Colon of Colorectal Cancer Patients Are Distinct in Activation and Functional Status, but Comparably Responsive to Activated T Cells. Cancers (Basel) 2021; 13:cancers13205247. [PMID: 34680397 PMCID: PMC8533845 DOI: 10.3390/cancers13205247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/15/2021] [Accepted: 10/17/2021] [Indexed: 12/24/2022] Open
Abstract
Although mouse models of CRC treatments have demonstrated robust immune activation, it remains unclear to what extent CRC patients' APCs and TILs interact to fuel or quench treatment-induced immune responses. Our ex vivo characterization of tumor and adjacent colon cell suspensions suggest that contrasting environments in these tissues promoted inversed expression of T cell co-stimulatory CD80, and co-inhibitory programmed death (PD)-ligand1 (PD-L1) on intratumoral vs. colonic APCs. While putative tumor-specific CD103+CD39+CD8+ TILs expressed lower CD69 (early activation marker) and higher PD-1 (extended activation/exhaustion marker) than colonic counterparts, the latter had instead higher CD69 and lower PD-1 levels. Functional comparisons showed that intratumoral APCs were inferior to colonic APCs regarding protein uptake and upregulation of CD80 and PD-L1 after protein degradation. Our attempt to model CRC treatment-induced T cell activation in vitro showed less interferon (IFN)-γ production by TILs than colonic T cells. In this model, we also measured APCs' CD80 and PD-L1 expression in response to activated co-residing T cells. These markers were comparable in the two tissues, despite higher IFN- γ exposure for colonic APCs. Thus, APCs within distinct intratumoral and colonic milieus showed different activation and functional status, but were similarly responsive to signals from induced T cell activation.
Collapse
Affiliation(s)
- Frank Liang
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (A.R.); (L.S.); (S.A.); (M.Q.-J.)
- Correspondence: (F.L.); (U.Y.)
| | - Azar Rezapour
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (A.R.); (L.S.); (S.A.); (M.Q.-J.)
| | - Louis Szeponik
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (A.R.); (L.S.); (S.A.); (M.Q.-J.)
| | - Samuel Alsén
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (A.R.); (L.S.); (S.A.); (M.Q.-J.)
| | - Yvonne Wettergren
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska University Hospital, University of Gothenburg, 413 45 Gothenburg, Sweden; (Y.W.); (E.B.L.)
| | - Elinor Bexe Lindskog
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska University Hospital, University of Gothenburg, 413 45 Gothenburg, Sweden; (Y.W.); (E.B.L.)
| | - Marianne Quiding-Järbrink
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (A.R.); (L.S.); (S.A.); (M.Q.-J.)
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (A.R.); (L.S.); (S.A.); (M.Q.-J.)
- Correspondence: (F.L.); (U.Y.)
| |
Collapse
|
66
|
Hyaluronic Acid-Glycine-Cholesterol Conjugate-Based Nanoemulsion as a Potent Vaccine Adjuvant for T Cell-Mediated Immunity. Pharmaceutics 2021; 13:pharmaceutics13101569. [PMID: 34683862 PMCID: PMC8539354 DOI: 10.3390/pharmaceutics13101569] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/09/2021] [Accepted: 09/18/2021] [Indexed: 01/05/2023] Open
Abstract
Clinical cases of allergic reaction that are due to excipients containing polyethylene glycol (PEG), a hydrophilic molecule commonly used in drug/vaccine formulations, has attracted much attention in recent years. In order to develop PEG-free adjuvants, we investigated the feasibility of natural ingredients in the human body such as hyaluronic acid in the form of hyaluronic acid-glycine cholesterol (HACH) conjugate as an excipient for vaccine formulation. Interestingly, HACH grafted with ~13 wt.% cholesterol has good water dispersity and can serve as an emulsifier to stabilize the squalene/water interfaces, yielding a milky white and isotropic emulsion (SQ@HACH) after being passed through a high-shear microfluidizer. Our results show that SQ@HACH particles possessed a unimodal average hydrodynamic diameter of approximately 190 nm measured by dynamic light scattering and exhibited good stability upon storage at 4 °C and 37 °C for over 20 weeks. The results of immunogenicity using a mouse model with ovalbumin (OVA) as the antigen revealed that SQ@HACH significantly enhanced antigen-specific immune responses, including the polarization of IgG antibodies, the cytokine secretions of T cells, and enhancement of cytotoxic T lymphocyte (CTL) activation. Moreover, SQ@HACH revealed lower local inflammation and rapidly absorbing properties compared with AlPO4 after intramuscular injection in vivo, indicating the potential functions of the HA-derived conjugate as an excipient in vaccine formulations for enhancement of T cell-mediated immunity.
Collapse
|
67
|
Dark Side of Cancer Therapy: Cancer Treatment-Induced Cardiopulmonary Inflammation, Fibrosis, and Immune Modulation. Int J Mol Sci 2021; 22:ijms221810126. [PMID: 34576287 PMCID: PMC8465322 DOI: 10.3390/ijms221810126] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/07/2021] [Accepted: 09/15/2021] [Indexed: 12/15/2022] Open
Abstract
Advancements in cancer therapy increased the cancer free survival rates and reduced the malignant related deaths. Therapeutic options for patients with thoracic cancers include surgical intervention and the application of chemotherapy with ionizing radiation. Despite these advances, cancer therapy-related cardiopulmonary dysfunction (CTRCPD) is one of the most undesirable side effects of cancer therapy and leads to limitations to cancer treatment. Chemoradiation therapy or immunotherapy promote acute and chronic cardiopulmonary damage by inducing reactive oxygen species, DNA damage, inflammation, fibrosis, deregulation of cellular immunity, cardiopulmonary failure, and non-malignant related deaths among cancer-free patients who received cancer therapy. CTRCPD is a complex entity with multiple factors involved in this pathogenesis. Although the mechanisms of cancer therapy-induced toxicities are multifactorial, damage to the cardiac and pulmonary tissue as well as subsequent fibrosis and organ failure seem to be the underlying events. The available biomarkers and treatment options are not sufficient and efficient to detect cancer therapy-induced early asymptomatic cell fate cardiopulmonary toxicity. Therefore, application of cutting-edge multi-omics technology, such us whole-exome sequencing, DNA methylation, whole-genome sequencing, metabolomics, protein mass spectrometry and single cell transcriptomics, and 10 X spatial genomics, are warranted to identify early and late toxicity, inflammation-induced carcinogenesis response biomarkers, and cancer relapse response biomarkers. In this review, we summarize the current state of knowledge on cancer therapy-induced cardiopulmonary complications and our current understanding of the pathological and molecular consequences of cancer therapy-induced cardiopulmonary fibrosis, inflammation, immune suppression, and tumor recurrence, and possible treatment options for cancer therapy-induced cardiopulmonary toxicity.
Collapse
|
68
|
Liu N, Shan F, Ma M. Strategic enhancement of immune checkpoint inhibition in refractory Colorectal Cancer: Trends and future prospective. Int Immunopharmacol 2021; 99:108017. [PMID: 34352568 DOI: 10.1016/j.intimp.2021.108017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/17/2021] [Accepted: 07/21/2021] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC), known as a frequently fatal disease, ranking as the third most common malignancy, is the second leading cause of cancer related mortality worldwide. Metastases are common in CRC patients which account for approximately 25% of the patients at diagnosis, 50% of patients during treatment which is associated closely with CRC mortality. Conventional therapies such as surgery, chemotherapy, and radiotherapy are standards of care for the treatment of CRC patients. However, primary tumor recurrence and secondary disease in patients receiving standard of care treatment modalities occur in 50% of patients so that new treatment modalities are needed. Immune checkpoint inhibition (ICI) has transformed the management of patients suffered from metastatic CRC (mCRC) with mismatch repair deficiency (dMMR) and microsatellite instability (MSI) -high (MSI-H) while manifests ineffectiveness in preserved mismatch repair (pMMR) or microsatellite stable (MSS) "cold" tumors which makes up the majority (95%) of mCRC. In this review, we mainly lay emphasis on the development of combinations in therapy strategies with ICIs with other immune based treatment approaches to increase the intra-tumoral immune response and render tumors 'immune-reactive', thereby increasing the efficacy of tumor immunotherapy.
Collapse
Affiliation(s)
- Ning Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Fengping Shan
- Department of Immunology, College of Basic Medical Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning Province, China
| | - Mingxing Ma
- Department of Colorectal Cancer Surgery, Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
69
|
Inflammation and tumor progression: signaling pathways and targeted intervention. Signal Transduct Target Ther 2021; 6:263. [PMID: 34248142 PMCID: PMC8273155 DOI: 10.1038/s41392-021-00658-5] [Citation(s) in RCA: 881] [Impact Index Per Article: 293.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/11/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer development and its response to therapy are regulated by inflammation, which either promotes or suppresses tumor progression, potentially displaying opposing effects on therapeutic outcomes. Chronic inflammation facilitates tumor progression and treatment resistance, whereas induction of acute inflammatory reactions often stimulates the maturation of dendritic cells (DCs) and antigen presentation, leading to anti-tumor immune responses. In addition, multiple signaling pathways, such as nuclear factor kappa B (NF-kB), Janus kinase/signal transducers and activators of transcription (JAK-STAT), toll-like receptor (TLR) pathways, cGAS/STING, and mitogen-activated protein kinase (MAPK); inflammatory factors, including cytokines (e.g., interleukin (IL), interferon (IFN), and tumor necrosis factor (TNF)-α), chemokines (e.g., C-C motif chemokine ligands (CCLs) and C-X-C motif chemokine ligands (CXCLs)), growth factors (e.g., vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-β), and inflammasome; as well as inflammatory metabolites including prostaglandins, leukotrienes, thromboxane, and specialized proresolving mediators (SPM), have been identified as pivotal regulators of the initiation and resolution of inflammation. Nowadays, local irradiation, recombinant cytokines, neutralizing antibodies, small-molecule inhibitors, DC vaccines, oncolytic viruses, TLR agonists, and SPM have been developed to specifically modulate inflammation in cancer therapy, with some of these factors already undergoing clinical trials. Herein, we discuss the initiation and resolution of inflammation, the crosstalk between tumor development and inflammatory processes. We also highlight potential targets for harnessing inflammation in the treatment of cancer.
Collapse
|
70
|
Stephens AJ, Burgess-Brown NA, Jiang S. Beyond Just Peptide Antigens: The Complex World of Peptide-Based Cancer Vaccines. Front Immunol 2021; 12:696791. [PMID: 34276688 PMCID: PMC8279810 DOI: 10.3389/fimmu.2021.696791] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/17/2021] [Indexed: 12/14/2022] Open
Abstract
Peptide-based cancer vaccines rely upon the strong activation of the adaptive immune response to elicit its effector function. They have shown to be highly specific and safe, but have yet to prove themselves as an efficacious treatment for cancer in the clinic. This is for a variety of reasons, including tumour heterogeneity, self-tolerance, and immune suppression. Importance has been placed on the overall design of peptide-based cancer vaccines, which have evolved from simple peptide derivatives of a cancer antigen, to complex drugs; incorporating overlapping regions, conjugates, and delivery systems to target and stimulate different components of antigen presenting cells, and to bolster antigen cross-presentation. Peptide-based cancer vaccines are increasingly becoming more personalised to an individual's tumour antigen repertoire and are often combined with existing cancer treatments. This strategy ultimately aids in combating the shortcomings of a more generalised vaccine strategy and provides a comprehensive treatment, taking into consideration cancer cell variability and its ability to avoid immune interrogation.
Collapse
Affiliation(s)
- Alexander J Stephens
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom.,Centre for Medicines Discovery, Nuffield Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Nicola A Burgess-Brown
- Centre for Medicines Discovery, Nuffield Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Shisong Jiang
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
71
|
Nishimura J, Deguchi S, Tanaka H, Yamakoshi Y, Yoshii M, Tamura T, Toyokawa T, Lee S, Muguruma K, Ohira M. Induction of Immunogenic Cell Death of Esophageal Squamous Cell Carcinoma by 5-Fluorouracil and Cisplatin. In Vivo 2021; 35:743-752. [PMID: 33622867 DOI: 10.21873/invivo.12315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Neoadjuvant chemotherapy (NAC) using 5-FU (5-fluorouracil)/CDDP (cisplatin) is a standard therapy for stage II/III thoracic esophageal squamous cell carcinoma (ESCC) in Japan. The aim of this study was to investigate whether 5-FU/CDDP could induce immunogenic cell death in ESCC cell lines. MATERIALS AND METHODS Tumor samples for immunohistochemistry were obtained from 50 patients (mean age=63.1 years) with pathological stage 0-IVa ESCC who underwent NAC followed by surgery. Cell lines T.T and KYSE30 were used for the in vitro experiments. RESULTS The concentrations of HMGB1 were elevated in the cell line supernatants treated with 5-FU/CDDP. 5-FU/CDDP treated dendritic cells (DCs) showed a mature phenotype, and enhanced T cell proliferation capacity. In addition, mature DCs were observed in surgical specimens with a histological response after treatment with 5-FU/CDDP chemotherapy. CONCLUSION 5-FU/CDDP could induce immunogenic cell death in the tumor microenvironment of ESCC.
Collapse
Affiliation(s)
- Junya Nishimura
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Sota Deguchi
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Tanaka
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yoshihito Yamakoshi
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Mami Yoshii
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tatsuro Tamura
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Takahiro Toyokawa
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shigeru Lee
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kazuya Muguruma
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masaichi Ohira
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
72
|
Eberle J, Wiehe RS, Gole B, Mattis LJ, Palmer A, Ständker L, Forssmann WG, Münch J, Gebhardt JCM, Wiesmüller L. A Fibrinogen Alpha Fragment Mitigates Chemotherapy-Induced MLL Rearrangements. Front Oncol 2021; 11:689063. [PMID: 34222016 PMCID: PMC8249925 DOI: 10.3389/fonc.2021.689063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/03/2021] [Indexed: 11/25/2022] Open
Abstract
Rearrangements in the Mixed Lineage Leukemia breakpoint cluster region (MLLbcr) are frequently involved in therapy-induced leukemia, a severe side effect of anti-cancer therapies. Previous work unraveled Endonuclease G as the critical nuclease causing initial breakage in the MLLbcr in response to different types of chemotherapeutic treatment. To identify peptides protecting against therapy-induced leukemia, we screened a hemofiltrate-derived peptide library by use of an enhanced green fluorescent protein (EGFP)-based chromosomal reporter of MLLbcr rearrangements. Chromatographic purification of one active fraction and subsequent mass spectrometry allowed to isolate a C-terminal 27-mer of fibrinogen α encompassing amino acids 603 to 629. The chemically synthesized peptide, termed Fα27, inhibited MLLbcr rearrangements in immortalized hematopoietic cells following treatment with the cytostatics etoposide or doxorubicin. We also provide evidence for protection of primary human hematopoietic stem and progenitor cells from therapy-induced MLLbcr breakage. Of note, fibrinogen has been described to activate toll-like receptor 4 (TLR4). Dissecting the Fα27 mode-of action revealed association of the peptide with TLR4 in an antagonistic fashion affecting downstream NFκB signaling and pro-inflammatory cytokine production. In conclusion, we identified a hemofiltrate-derived peptide inhibitor of the genome destabilizing events causing secondary leukemia in patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Julia Eberle
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| | | | - Boris Gole
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| | - Liska Jule Mattis
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| | - Anja Palmer
- Department of Physics, Institute of Biophysics, Ulm University, Ulm, Germany
| | - Ludger Ständker
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
| | - Wolf-Georg Forssmann
- Pharis Biotec GmbH and Peptide Research Group, Institute of Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Jan Münch
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | | - Lisa Wiesmüller
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| |
Collapse
|
73
|
Fourie C, Shridas P, Davis T, de Villiers WJ, Engelbrecht AM. Serum amyloid A and inflammasome activation: A link to breast cancer progression? Cytokine Growth Factor Rev 2021; 59:62-70. [DOI: 10.1016/j.cytogfr.2020.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022]
|
74
|
Pahlavanneshan S, Sayadmanesh A, Ebrahimiyan H, Basiri M. Toll-Like Receptor-Based Strategies for Cancer Immunotherapy. J Immunol Res 2021; 2021:9912188. [PMID: 34124272 PMCID: PMC8166496 DOI: 10.1155/2021/9912188] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/28/2021] [Accepted: 05/09/2021] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptors (TLRs) are expressed and play multiple functional roles in a variety of immune cell types involved in tumor immunity. There are plenty of data on the pharmacological targeting of TLR signaling using agonist molecules that boost the antitumor immune response. A recent body of research has also demonstrated promising strategies for improving the cell-based immunotherapy methods by inducing TLR signaling. These strategies include systemic administration of TLR antagonist along with immune cell transfer and also genetic engineering of the immune cells using TLR signaling components to improve the function of genetically engineered immune cells such as chimeric antigen receptor-modified T cells. Here, we explore the current status of the cancer immunotherapy approaches based on manipulation of TLR signaling to provide a perspective of the underlying rationales and potential clinical applications. Altogether, reviewed publications suggest that TLRs make a potential target for the immunotherapy of cancer.
Collapse
Affiliation(s)
- Saghar Pahlavanneshan
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Sayadmanesh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamidreza Ebrahimiyan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
75
|
Lurje I, Werner W, Mohr R, Roderburg C, Tacke F, Hammerich L. In Situ Vaccination as a Strategy to Modulate the Immune Microenvironment of Hepatocellular Carcinoma. Front Immunol 2021; 12:650486. [PMID: 34025657 PMCID: PMC8137829 DOI: 10.3389/fimmu.2021.650486] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/22/2021] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular Carcinoma (HCC) is a highly prevalent malignancy that develops in patients with chronic liver diseases and dysregulated systemic and hepatic immunity. The tumor microenvironment (TME) contains tumor-associated macrophages (TAM), cancer-associated fibroblasts (CAF), regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSC) and is central to mediating immune evasion and resistance to therapy. The interplay between these cells types often leads to insufficient antigen presentation, preventing effective anti-tumor immune responses. In situ vaccines harness the tumor as the source of antigens and implement sequential immunomodulation to generate systemic and lasting antitumor immunity. Thus, in situ vaccines hold the promise to induce a switch from an immunosuppressive environment where HCC cells evade antigen presentation and suppress T cell responses towards an immunostimulatory environment enriched for activated cytotoxic cells. Pivotal steps of in situ vaccination include the induction of immunogenic cell death of tumor cells, a recruitment of antigen-presenting cells with a focus on dendritic cells, their loading and maturation and a subsequent cross-priming of CD8+ T cells to ensure cytotoxic activity against tumor cells. Several in situ vaccine approaches have been suggested, with vaccine regimens including oncolytic viruses, Flt3L, GM-CSF and TLR agonists. Moreover, combinations with checkpoint inhibitors have been suggested in HCC and other tumor entities. This review will give an overview of various in situ vaccine strategies for HCC, highlighting the potentials and pitfalls of in situ vaccines to treat liver cancer.
Collapse
Affiliation(s)
- Isabella Lurje
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| | - Wiebke Werner
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| | - Raphael Mohr
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| | - Christoph Roderburg
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| | - Linda Hammerich
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
76
|
Sudo T, Motomura Y, Okuzaki D, Hasegawa T, Yokota T, Kikuta J, Ao T, Mizuno H, Matsui T, Motooka D, Yoshizawa R, Nagasawa T, Kanakura Y, Moro K, Ishii M. Group 2 innate lymphoid cells support hematopoietic recovery under stress conditions. J Exp Med 2021; 218:e20200817. [PMID: 33666647 PMCID: PMC7941180 DOI: 10.1084/jem.20200817] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 10/29/2020] [Accepted: 02/02/2021] [Indexed: 12/18/2022] Open
Abstract
The cell-cycle status of hematopoietic stem and progenitor cells (HSPCs) becomes activated following chemotherapy-induced stress, promoting bone marrow (BM) regeneration; however, the underlying molecular mechanism remains elusive. Here we show that BM-resident group 2 innate lymphoid cells (ILC2s) support the recovery of HSPCs from 5-fluorouracil (5-FU)-induced stress by secreting granulocyte-macrophage colony-stimulating factor (GM-CSF). Mechanistically, IL-33 released from chemo-sensitive B cell progenitors activates MyD88-mediated secretion of GM-CSF in ILC2, suggesting the existence of a B cell-ILC2 axis for maintaining hematopoietic homeostasis. GM-CSF knockout mice treated with 5-FU showed severe loss of myeloid lineage cells, causing lethality, which was rescued by transferring BM ILC2s from wild-type mice. Further, the adoptive transfer of ILC2s to 5-FU-treated mice accelerates hematopoietic recovery, while the reduction of ILC2s results in the opposite effect. Thus, ILC2s may function by "sensing" the damaged BM spaces and subsequently support hematopoietic recovery under stress conditions.
Collapse
Affiliation(s)
- Takao Sudo
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasutaka Motomura
- World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory for Innate Immune Systems, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Daisuke Okuzaki
- Single Cell Genomics, Human Immunology, World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tetsuo Hasegawa
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
| | - Takafumi Yokota
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Tomoka Ao
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Hiroki Mizuno
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Takahiro Matsui
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Daisuke Motooka
- Single Cell Genomics, Human Immunology, World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Ryosuke Yoshizawa
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
| | - Takashi Nagasawa
- World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
| | - Yuzuru Kanakura
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuyo Moro
- World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory for Innate Immune Systems, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| |
Collapse
|
77
|
Laan M, Salumets A, Klein A, Reintamm K, Bichele R, Peterson H, Peterson P. Post-Aire Medullary Thymic Epithelial Cells and Hassall's Corpuscles as Inducers of Tonic Pro-Inflammatory Microenvironment. Front Immunol 2021; 12:635569. [PMID: 33868260 PMCID: PMC8050345 DOI: 10.3389/fimmu.2021.635569] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/05/2021] [Indexed: 11/13/2022] Open
Abstract
While there is convincing evidence on the role of Aire-positive medullary thymic epithelial cells (mTEC) in the induction of central tolerance, the nature and function of post-Aire mTECs and Hassall's corpuscles have remained enigmatic. Here we summarize the existing data on these late stages of mTEC differentiation with special focus on their potential to contribute to central tolerance induction by triggering the unique pro-inflammatory microenvironment in the thymus. In order to complement the existing evidence that has been obtained from mouse models, we performed proteomic analysis on microdissected samples from human thymic medullary areas at different differentiation stages. The analysis confirms that at the post-Aire stages, the mTECs lose their nuclei but maintain machinery required for translation and exocytosis and also upregulate proteins specific to keratinocyte differentiation and cornification. In addition, at the late stages of differentiation, the human mTECs display a distinct pro-inflammatory signature, including upregulation of the potent endogenous TLR4 agonist S100A8/S100A9. Collectively, the study suggests a novel mechanism by which the post-Aire mTECs and Hassall's corpuscles contribute to the thymic microenvironment with potential cues on the induction of central tolerance.
Collapse
Affiliation(s)
- Martti Laan
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Ahto Salumets
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- Institute of Computer Science, Faculty of Science and Technology, University of Tartu, Tartu, Estonia
| | - Annabel Klein
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Kerli Reintamm
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Rudolf Bichele
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Hedi Peterson
- Institute of Computer Science, Faculty of Science and Technology, University of Tartu, Tartu, Estonia
| | - Pärt Peterson
- Molecular Pathology Research Group, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
78
|
Yang S, Shim MK, Kim WJ, Choi J, Nam GH, Kim J, Kim J, Moon Y, Kim HY, Park J, Park Y, Kim IS, Ryu JH, Kim K. Cancer-activated doxorubicin prodrug nanoparticles induce preferential immune response with minimal doxorubicin-related toxicity. Biomaterials 2021; 272:120791. [PMID: 33831739 DOI: 10.1016/j.biomaterials.2021.120791] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/29/2021] [Indexed: 12/27/2022]
Abstract
The effective chemotherapeutic drug, doxorubicin (DOX), elicits immunogenic cell death (ICD) and additional anticancer immune responses during chemotherapy. However, it also induces severe side effects and systemic immunosuppression, hampering its wide clinical application. Herein, we constructed cancer-activated DOX prodrug by conjugating the cathepsin B-cleavable peptide (Phe-Arg-Arg-Gly, FRRG) to a doxorubicin (DOX), resulting in FRRG-DOX that self-assembled into cancer-activated DOX prodrug nanoparticles (CAP-NPs). The resulting CAP-NPs were further stabilized with the FDA-approved compound, Pluronic F68. CAP-NPs formed stable prodrug nanoparticles and they were specifically cleaved to cytotoxic DOX molecules only in cathepsin B-overexpressing cancer cells, inducing a cancer cell-specific cytotoxicity. In particular, the CAP-NPs induced ICD through cathepsin B-cleavage mechanism only in targeted cancer cells in vitro. In colon tumor-bearing mice, selectively accumulated CAP-NPs at tumors enhanced antitumor immunity without DOX-related severe toxicity, inflammatory response and systemic immunosuppression. Moreover, cytotoxicity against immune cells infiltrated into tumor microenvironment was significantly reduced compared to free DOX, leading to increased response to checkpoint inhibitor immunotherapy. The combinatorial treatment of CAP-NPs with anti-PD-L1 exhibited high rate of complete tumor regression (50%) compared to free DOX with anti-PD-L1. Concurrently, DOX-related side effects were greatly reduced during chemoimmunotherapy. Collectively, our results suggest that cancer-activated DOX prodrug nanoparticles provide a promising approach to increase clinical benefit by inducing an immune response preferentially only to targeted cancer cells, not to normal cells and immune cells, and potentiates checkpoint inhibitor immunotherapy.
Collapse
Affiliation(s)
- Suah Yang
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Man Kyu Shim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Woo Jun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Jiwoong Choi
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Gi-Hoon Nam
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Jeongrae Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Jinseong Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Yujeong Moon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea; Department of Bioengineering, Korea University, Seoul, 02841, Republic of Korea
| | - Han Young Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Jooho Park
- Department of Biomedical & Health Science, Konkuk University, Chungju, 27478, Republic of Korea
| | - Yoon Park
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Ju Hee Ryu
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea.
| | - Kwangmeyung Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea.
| |
Collapse
|
79
|
Keshavarz-Fathi M, Rezaei N. Cancer Immunoprevention: Current Status and Future Directions. Arch Immunol Ther Exp (Warsz) 2021; 69:3. [PMID: 33638703 DOI: 10.1007/s00005-021-00604-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 02/06/2021] [Indexed: 12/24/2022]
Abstract
Cancer is one of the most serious diseases affecting health and the second leading cause of death worldwide. Despite the development of various therapeutic modalities to deal with cancer, limited improvement in overall survival of patients has been yielded. Since there is no certain cure for cancer, detection of premalignant lesions, and prevention of their progression are vital to the decline of high morbidity and mortality of cancer. Among approaches to cancer prevention, immunoprevention has gained further attention in recent years. Deep understanding of the tumor/immune system interplay and successful prevention of virally-induced malignancies by vaccines have paved the way toward broadening cancer immunoprevention application. The identification of tumor antigens in premalignant lesions was the turning point in cancer immunoprevention that led to designing preventive vaccines for various malignancies including multiple myeloma, colorectal, and breast cancer. In addition to vaccines, immune checkpoint inhibitors are also being tested for the prevention of oral squamous cell carcinoma (SCC), and imiquimod which is an established drug for the prevention of skin SCC, is a non-specific immunomodulator. Herein, to provide a bench-to-bedside understanding of cancer immunoprevention, we will review the role of the immune system in suppression and promotion of tumors, immunoprevention of virally-induced cancers, identification of tumor antigens in premalignant lesions, and clinical advances of cancer immunoprevention.
Collapse
Affiliation(s)
- Mahsa Keshavarz-Fathi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, 14194, Tehran, Iran.
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, 14194, Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
80
|
Li J, Wu H, Gao H, Kou R, Xie Y, Zhang Z, Zhang X. TLR4 promoter rs1927914 variant contributes to the susceptibility of esophageal squamous cell carcinoma in the Chinese population. PeerJ 2021; 9:e10754. [PMID: 33585082 PMCID: PMC7860108 DOI: 10.7717/peerj.10754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/21/2020] [Indexed: 12/13/2022] Open
Abstract
Background Toll-like receptor 4 (TLR4), as a key regulator of both innate and acquired immunity, has been linked with the development of various cancers, including esophageal cancer. This study aims to analyze the association of potential functional genetic polymorphisms in TLR4 with the risk of esophageal cancer. Methods This case-control study involved in 480 ESCC patients and 480 health controls. Polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) was used to genotype TLR4 rs1927914 polymorphism. Taqman probe method was used to determine the genotypes of TLR4 rs11536891 and rs7873784 variants. The relationship between TLR4 genetic variation and ESCC risk was analyzed by Logistic regression model by calculating the odds ratio (OR) and 95% confidence interval (95% CI). Results Compared with TLR4 rs1927914 AA genotype carriers, GG carriers had a lower ESCC risk (OR = 0.59, 95% CI [0.38–0.93], P = 0.023). Stratification analysis by age showed that TLR4 rs1927914 GG could affect the risk of ESCC in elderly people (OR = 0.59, 95% CI [0.36–0.97]). Smoking stratification analysis indicated that rs1927914 GG carriers were related to ESCC susceptibility among non-smokers (OR = 0.36, 95% CI [0.18–0.73]). Dual luciferase reporter assay suggested that rs1927914 G-containing TLR4 promoter displayed a 1.76-fold higher luciferase activity than rs1927914 A-containing counterpart in KYSE30 cells. Electrophoretic mobility shift assay (EMSA) showed the KYSE30 cell nuclear extract was able to bind the probe with rs1927914 G allele and this DNA-protein interaction could be eliminated by competition assays with unlabeled rs1927914 G probe, which indicating that the binding is sequence-specific. Our results also showed that TLR4 rs7873784 (G>C) and rs11536891 (T>C) conformed to complete genetic linkage. The genotype distributions of TLR4 rs11536891 variant among ESCC patients and normal controls have no statistical significance. Conclusion The TLR4 rs1927914 variant contributes to the ESCC risk by effecting the promoter activity.
Collapse
Affiliation(s)
- Jiaying Li
- School of Public Health, North China University of Science and Technology, Tangshan, China.,College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Hongjiao Wu
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Hui Gao
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Ruihuan Kou
- Affliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, China
| | - Yuning Xie
- School of Public Health, North China University of Science and Technology, Tangshan, China.,College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Zhi Zhang
- Affliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, China
| | - Xuemei Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, China.,College of Life Science, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
81
|
Deng Y, Govers C, Ter Beest E, van Dijk AJ, Hettinga K, Wichers HJ. A THP-1 Cell Line-Based Exploration of Immune Responses Toward Heat-Treated BLG. Front Nutr 2021; 7:612397. [PMID: 33521038 PMCID: PMC7838438 DOI: 10.3389/fnut.2020.612397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Allergen recognition and processing by antigen presenting cells is essential for the sensitization step of food allergy. Macrophages and dendritic cells are both phagocytic antigen presenting cells and play important roles in innate immune responses and signaling between the innate and adaptive immune system. To obtain a model system with a homogeneous genetic background, we derived macrophages and dendritic cells from THP-1 monocytes. The difference between macrophages and dendritic cells was clearly shown by differences in their transcription response (microarray) and protein expression levels. Their resemblance to primary cells was analyzed by comparison to properties as described in literature. The uptake of β-lactoglobulin after wet-heating (60°C in solution) by THP-1 derived macrophages was earlier reported to be significantly increased. To analyse the subsequent immune response, we incubated THP-1 derived macrophages and dendritic cells with native and differently processed β-lactoglobulin and determined the transcription and cytokine expression levels of the cells. A stronger transcriptional response was found in macrophages than in dendritic cells, while severely structurally modified β-lactoglobulin induced a more limited transcriptional response, especially when compared to native and limitedly modified β-lactoglobulin. These results show that processing is relevant for the transcriptional response toward β-lactoglobulin of innate immune cells.
Collapse
Affiliation(s)
- Ying Deng
- Food and Biobased Research, Wageningen University and Research, Wageningen, Netherlands.,Food Chemistry, Department of Agrotechnology and Food Sciences, Wageningen University and Research, Wageningen, Netherlands
| | - Coen Govers
- Food and Biobased Research, Wageningen University and Research, Wageningen, Netherlands
| | - Ellen Ter Beest
- Food and Biobased Research, Wageningen University and Research, Wageningen, Netherlands.,Food Chemistry, Department of Agrotechnology and Food Sciences, Wageningen University and Research, Wageningen, Netherlands
| | - Aalt-Jan van Dijk
- Bioinformatics Group, Department of Plant Sciences, Wageningen University and Research, Wageningen, Netherlands
| | - Kasper Hettinga
- Food Quality and Design, Department of Agrotechnology and Food Sciences, Wageningen University and Research, Wageningen, Netherlands
| | - Harry J Wichers
- Food and Biobased Research, Wageningen University and Research, Wageningen, Netherlands.,Food Chemistry, Department of Agrotechnology and Food Sciences, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
82
|
Cui L, Wang X, Zhang D. TLRs as a Promise Target Along With Immune Checkpoint Against Gastric Cancer. Front Cell Dev Biol 2021; 8:611444. [PMID: 33469538 PMCID: PMC7813757 DOI: 10.3389/fcell.2020.611444] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer (GC) is one of the most common cancers in the world, and the incidence of gastric cancer in Asia appears to increase in recent years. Although there is a lot of improvement in treatment approaches, the prognosis of GC is poor. So it is urgent to search for a novel and more effective treatment to improve the survival rate of patients. Both innate immunity and adaptive immunity are important in cancer. In the innate immune system, pattern recognition receptors (PRRs) activate immune responses by recognizing pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). Toll-like receptors (TLRs) are a class of pattern recognition receptors (PRRs). Many studies have reported that TLRs are involved in the occurrence, development, and treatment of GC. Therefore, TLRs are potential targets for immunotherapy to gastric cancer. However, gastric cancer is a heterogeneous disorder, and TLRs function in GC is complex. TLRs agonists can be potentially used not only as therapeutic agents to treat gastric cancer but also as adjuvants in conjunction with other immunotherapies. They might provide a promising new target for GC treatment. In the review, we sort out the mechanism of TLRs involved in tumor immunity and summarize the current progress in TLRs-based therapeutic approaches and other immunotherapies in the treatment of GC.
Collapse
Affiliation(s)
- Lin Cui
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xiuqing Wang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Dekai Zhang
- Center for Infectious and Inflammatory Diseases, Texas A&M University, Houston, TX, United States
| |
Collapse
|
83
|
Xu D, Wang Y, Wu J, Zhang Z, Chen J, Xie M, Tang R, Cheng C, Chen L, Lin S, Luo X, Zheng J. MTIF2 impairs 5 fluorouracil-mediated immunogenic cell death in hepatocellular carcinoma in vivo: Molecular mechanisms and therapeutic significance. Pharmacol Res 2021; 163:105265. [PMID: 33129983 DOI: 10.1016/j.phrs.2020.105265] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related morbidity and mortality; it has been reported that immune cell infiltration is a prognosis factor. Here we identified genes that associated with tumor immune cell infiltrate; the underlying mechanism was verified by in vivo and in vitro experiment. In this study, Weighted correlation network analysis (WGCNA) and CIBERSORT tool were used to identify MTIF2 as the hub tumor immune infiltrating gene in HCC. To investigate the underlying role played by MTIF2, MTIF2 was knocked down by transfection of shRNA targeting MTIF2, CCK8, and EdU incorporation assay was used to evaluate the effect of MTIF2 on proliferation, wound heal assay and transwell assay was used to confirm its effect on cell migration. Ecto-calreticulin on the cell surface was evaluated by flow cytometry, ATP, and HMGB1 secretion were tested to the investigated effect of MTIF2 on the immunogenic cell death (ICD) process. We found that down-regulation of MTIF2 impaired proliferation and migration capacity of HCC cells, chemoresistance to 5-Fluorouracil (5-FU) weakened after MTIF2 was knocked down. Reduced release of damage-associated molecular patterns (DAMP) was observed after MTIF2 was overexpressed, which subsequently impaired dendritic cell (DC) maturation and proliferation of CD8 + T cells. Mechanically, the co-IP experiment confirmed that MTIF2 could interact with AIFM1, prevents AIFM1 induced transcription of caspase3, and finally suppress apoptosis. In vivo experiment also used to confirm our previously conclusion, our result indicated that MTIF2 overexpression suppresses tumor apoptosis and immune cell activity in the 5-FU therapy in vivo model, by suppression maturation of tumor-infiltrated DC. Collectively, our study confirmed that MTIF2 impair drug-induced immunogenic cell death in hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Dafeng Xu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Yu Wang
- Geriatric Medicine Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Jincai Wu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Zhensheng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Jiacheng Chen
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Mingwei Xie
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Rong Tang
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Chen Cheng
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Liang Chen
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Shiyun Lin
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Xiangxiang Luo
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Jinfang Zheng
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China.
| |
Collapse
|
84
|
Hayashi K, Nikolos F, Lee YC, Jain A, Tsouko E, Gao H, Kasabyan A, Leung HE, Osipov A, Jung SY, Kurtova AV, Chan KS. Tipping the immunostimulatory and inhibitory DAMP balance to harness immunogenic cell death. Nat Commun 2020; 11:6299. [PMID: 33288764 PMCID: PMC7721802 DOI: 10.1038/s41467-020-19970-9] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
Induction of tumor cell death is the therapeutic goal for most anticancer drugs. Yet, a mode of drug-induced cell death, known as immunogenic cell death (ICD), can propagate antitumoral immunity to augment therapeutic efficacy. Currently, the molecular hallmark of ICD features the release of damage-associated molecular patterns (DAMPs) by dying cancer cells. Here, we show that gemcitabine, a standard chemotherapy for various solid tumors, triggers hallmark immunostimualtory DAMP release (e.g., calreticulin, HSP70, and HMGB1); however, is unable to induce ICD. Mechanistic studies reveal gemcitabine concurrently triggers prostaglandin E2 release as an inhibitory DAMP to counterpoise the adjuvanticity of immunostimulatory DAMPs. Pharmacological blockade of prostaglandin E2 biosythesis favors CD103+ dendritic cell activation that primes a Tc1-polarized CD8+ T cell response to bolster tumor rejection. Herein, we postulate that an intricate balance between immunostimulatory and inhibitory DAMPs could determine the outcome of drug-induced ICD and pose COX-2/prostaglandin E2 blockade as a strategy to harness ICD.
Collapse
Affiliation(s)
- K Hayashi
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - F Nikolos
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Y C Lee
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei City, Taiwan
| | - A Jain
- Alkek Center for Molecular Discovery, Proteomics Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - E Tsouko
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - H Gao
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - A Kasabyan
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - H E Leung
- Alkek Center for Molecular Discovery, Proteomics Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - A Osipov
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - S Y Jung
- Alkek Center for Molecular Discovery, Proteomics Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - A V Kurtova
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - K S Chan
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
| |
Collapse
|
85
|
Sato Y, Motoyama S, Wakita A, Kawakita Y, Liu J, Nagaki Y, Nanjo H, Ito S, Terata K, Imai K, Minamiya Y. High TLR4 expression predicts a poor prognosis after esophagectomy for advanced thoracic esophageal squamous cell carcinoma. Esophagus 2020; 17:408-416. [PMID: 32170544 DOI: 10.1007/s10388-020-00732-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 03/04/2020] [Indexed: 02/03/2023]
Abstract
BACKGROUND Poor oral health is an independent risk factor for upper aerodigestive tract cancers, including esophageal squamous cell carcinoma (ESCC). The pattern recognition receptor Toll-like receptor 4 (TLR4) recognizes lipopolysaccharide in the cell walls of Gram-negative periodontal pathogens associated with the development and progression of ESCC. It is, therefore, plausible that TLR4 plays a crucial role in the pathogenesis of ESCC. METHODS We used an ESCC tissue microarray to confirm expression of TLR4 in patients with ESCC and to determine whether TLR4 expression status correlates with the clinicopathological features of these patients or their prognosis after esophagectomy. We also tested whether the combined expression statuses of TLR4 and TLR3 better correlate with prognosis in these patients than either parameter alone. RESULTS Clinical ESCC samples from all 177 patients tested showed expression of TLR4. Moreover, high TLR4 expression (3 + and 2 +) correlated with poorer 5-year overall survival after esophagectomy than lower TLR4 expression (1 +) (p = 0.0491). Patients showing high TLR4 expression tended to have a poorer prognosis whether treated with surgery alone or with surgery and adjuvant chemotherapy. Univariate and multivariate analyses showed TLR4 expression status to be an independent prognostic factor affecting 5-year overall survival. Patients exhibiting high TLR4 expression with low TLR3 expression had a much poorer prognosis than other patients (p = < 0.0001). CONCLUSION High TLR4 expression predicts a poor prognosis in advanced thoracic ESCC patients after esophagectomy.
Collapse
Affiliation(s)
- Yusuke Sato
- Department of Surgery, Akita University Graduate School of Medicine, Akita, 010-8543, Japan.
| | - Satoru Motoyama
- Department of Surgery, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Akiyuki Wakita
- Department of Surgery, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Yuta Kawakita
- Department of Surgery, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Jiajia Liu
- Department of Surgery, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Yushi Nagaki
- Department of Surgery, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Hiroshi Nanjo
- Department of Pathology, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Satoru Ito
- Department of Pathology, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Kaori Terata
- Department of Surgery, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Kazuhiro Imai
- Department of Surgery, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Yoshihiro Minamiya
- Department of Surgery, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| |
Collapse
|
86
|
Feng X, Yu W, Cao L, Meng F, Cong M. A novel chrysin thiazole derivative polarizes macrophages to an M1 phenotype via targeting TLR4. Int Immunopharmacol 2020; 88:106986. [PMID: 33182070 DOI: 10.1016/j.intimp.2020.106986] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022]
Abstract
Tumor-associated macrophages (TAMs) are an important cause of tumorigenesis and tumor development. M2 macrophages can promote tumor growth while M1 macrophages kill tumor cells, therefore, polarizing macrophages to achieve a functional M1 phenotype could effectively play its anti-tumor role. In the current study, we synthesized a novel chrysin derivative which is termed as ChR-TD. And we found ChR-TD might be a ligand of TLR4 that polarized the TAMs towards M1 phenotype and played its anti-tumor role. Further study indicated that ChR-TD reprogrammed the macrophages into an M1 phenotype via TLR4 activation. Moreover, ChR-TD activated TLR4/NF-κB signaling pathway and promoted the NF-κB/p65 translocated into the nuclear, leading to the activation of NF-κB and proinflammatory cytokines release. In addition, type I interferon signaling was also activated by ChR-TD, leading to the expressions of IFN-α and IFN-β and its targeted genes NOS2, MCP-1 and IP-10 were significantly increased in macrophages. Importantly, these effects were disturbed in TLR4-/- macrophages, which are constructed by using CRISPR/Cas9 system. And the molecule docking simulation further indicated that ChR-TD could bind to TLR4 and might be a ligand of TLR4. Hence, these findings suggested that ChR-TD might be a ligand of TLR4 and can be used as a potential lead compound for tumors treatment.
Collapse
Affiliation(s)
- Xiujing Feng
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan 250021, China.
| | - Wen Yu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210046, China
| | - Lingsen Cao
- Key Lab of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
| | - Fanda Meng
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan 250021, China
| | - Mulin Cong
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan 250021, China
| |
Collapse
|
87
|
Kulshrestha A, Katara GK, Ibrahim SA, Riehl VE, Schneiderman S, Bilal M, Young AN, Levine S, Fleetwood S, Dolan J, Gilman-Sachs A, Beaman KD. In vivo anti-V-ATPase antibody treatment delays ovarian tumor growth by increasing antitumor immune responses. Mol Oncol 2020; 14:2436-2454. [PMID: 32797726 PMCID: PMC7530789 DOI: 10.1002/1878-0261.12782] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/19/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022] Open
Abstract
Tumor acidity is the key metabolic feature promoting cancer progression and is modulated by pH regulators on a cancer cell's surface that pump out excess protons/lactic acid for cancer cell survival. Neutralizing tumor acidity improves the therapeutic efficacy of current treatments including immunotherapies. Vacuolar-ATPase (V-ATPase) proton pumps encompass unique plasma membrane-associated subunit isoforms, making this molecule an important target for anticancer therapy. Here, we examined the in vivo therapeutic efficacy of an antibody (a2v-mAB) targeting specific V-ATPase-'V0a2' surface isoform in controlling ovarian tumor growth. In vitro a2v-mAb treatment inhibited the proton pump activity in ovarian cancer (OVCA) cells. In vivo intraperitoneal a2v-mAb treatment drastically delayed ovarian tumor growth with no measurable in vivo toxicity in a transplant tumor model. To explore the possible mechanism causing delayed tumor growth, histochemical analysis of the a2v-mAb-treated tumor tissues displayed high immune cell infiltration (M1-macrophages, neutrophils, CD103+ cells, and NK cells) and an enhanced antitumor response (iNOS, IFN-y, IL-1α) compared to control. There was marked decrease in CA-125-positive cancer cells and an enhanced active caspase-3 expression in a2v-mAb-treated tumors. RNA-seq analysis of a2v-mAb tumor tissues further revealed upregulation of apoptosis-related and toll-like receptor pathway-related genes. Indirect coculture of a2v-mAb-treated OVCA cells with human PBMCs in an unbuffered medium led to an enhanced gene expression of antitumor molecules IFN-y, IL-17, and IL-12-A in PBMCs, further validating the in vivo antitumor responses. In conclusion, V-ATPase inhibition using a monoclonal antibody directed against the V0a2 isoform increases antitumor immune responses and could therefore constitute an effective treatment strategy in OVCA.
Collapse
Affiliation(s)
- Arpita Kulshrestha
- Department of Microbiology and Immunology, Center for Cancer cell biology, Immunology and infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Center for Cancer cell biology, Immunology and infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Safaa A Ibrahim
- Department of Microbiology and Immunology, Center for Cancer cell biology, Immunology and infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.,Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Valerie E Riehl
- Department of Microbiology and Immunology, Center for Cancer cell biology, Immunology and infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Sylvia Schneiderman
- Department of Microbiology and Immunology, Center for Cancer cell biology, Immunology and infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Mahmood Bilal
- Department of Microbiology and Immunology, Center for Cancer cell biology, Immunology and infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Alexandria N Young
- Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, IL, USA
| | - Shayna Levine
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Sara Fleetwood
- Department of Microbiology and Immunology, Center for Cancer cell biology, Immunology and infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - James Dolan
- Department of Obstetrics & Gynecology, Advocate Lutheran General Hospital, Park Ridge, IL, USA
| | - Alice Gilman-Sachs
- Department of Microbiology and Immunology, Center for Cancer cell biology, Immunology and infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Center for Cancer cell biology, Immunology and infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.,Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
88
|
Bai L, Li W, Zheng W, Xu D, Chen N, Cui J. Promising targets based on pattern recognition receptors for cancer immunotherapy. Pharmacol Res 2020; 159:105017. [DOI: 10.1016/j.phrs.2020.105017] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023]
|
89
|
Moeng S, Son SW, Lee JS, Lee HY, Kim TH, Choi SY, Kuh HJ, Park JK. Extracellular Vesicles (EVs) and Pancreatic Cancer: From the Role of EVs to the Interference with EV-Mediated Reciprocal Communication. Biomedicines 2020; 8:biomedicines8080267. [PMID: 32756339 PMCID: PMC7459718 DOI: 10.3390/biomedicines8080267] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/25/2020] [Accepted: 08/01/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is malignant and the seventh leading cause of cancer-related deaths worldwide. However, chemotherapy and radiotherapy are—at most—moderately effective, indicating the need for new and different kinds of therapies to manage this disease. It has been proposed that the biologic properties of pancreatic cancer cells are finely tuned by the dynamic microenvironment, which includes extracellular matrix, cancer-associated cells, and diverse immune cells. Accumulating evidence has demonstrated that extracellular vesicles (EVs) play an essential role in communication between heterogeneous subpopulations of cells by transmitting multiplex biomolecules. EV-mediated cell–cell communication ultimately contributes to several aspects of pancreatic cancer, such as growth, angiogenesis, metastasis and therapeutic resistance. In this review, we discuss the role of extracellular vesicles and their cargo molecules in pancreatic cancer. We also present the feasibility of the inhibition of extracellular biosynthesis and their itinerary (release and uptake) for a new attractive therapeutic strategy against pancreatic cancer.
Collapse
Affiliation(s)
- Sokviseth Moeng
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
| | - Seung Wan Son
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
| | - Jong Sun Lee
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
| | - Han Yeoung Lee
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
| | - Tae Hee Kim
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
| | - Hyo Jeong Kuh
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Jong Kook Park
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
- Correspondence: ; Tel.: +82-33-248-2114
| |
Collapse
|
90
|
Crestani A, Benoit L, Touboul C, Pasquier J. Hyperthermic intraperitoneal chemotherapy (HIPEC): Should we look closer at the microenvironment? Gynecol Oncol 2020; 159:285-294. [PMID: 32732012 DOI: 10.1016/j.ygyno.2020.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/11/2020] [Indexed: 10/23/2022]
Abstract
The age of cancer as an isolated single-cell concept is now behind us. It is now established that epithelial ovarian cancer, like other cancers, interacts with the healthy bystander cells to influence them and takes advantage of their nutritional, immunological, disseminating and other capacities. This interaction has become a therapeutic target, as shown by the numerous studies on this subject. Intraperitoneal chemo-hyperthermia has been part of the therapeutic armamentarium for some time yet its efficiency in ovarian cancer has only been recently proven in a randomized controlled trial. However, its therapeutic performance is not revolutionary and epithelial ovarian cancer maintains a high mortality. In this review, we studied the impact of HIPEC on the microenvironment and vice versa to determine whether it could be the key to this lukewarm efficacy. We began by exploring the modalities of HIPEC and establishing the reasons that make this treatment topical. Then, we examined its impact on each element of the tumor environment to obtain a global view of the resistance mechanisms at work in HIPEC.
Collapse
Affiliation(s)
- Adrien Crestani
- INSERM UMRS 938, Centre de recherche Saint Antoine, Team Cancer Biology and Therapeutics, Institut Universitaire de Cancérologie, Sorbonne Université, F-75012 Paris, France; Service de chirurgie gynécologique, hôpital Tenon, 4, rue de la Chine, 75012 Paris, France.
| | - Louise Benoit
- INSERM UMRS 938, Centre de recherche Saint Antoine, Team Cancer Biology and Therapeutics, Institut Universitaire de Cancérologie, Sorbonne Université, F-75012 Paris, France; Service de chirurgie gynécologique, hôpital Tenon, 4, rue de la Chine, 75012 Paris, France
| | - Cyril Touboul
- INSERM UMRS 938, Centre de recherche Saint Antoine, Team Cancer Biology and Therapeutics, Institut Universitaire de Cancérologie, Sorbonne Université, F-75012 Paris, France; Service de chirurgie gynécologique, hôpital Tenon, 4, rue de la Chine, 75012 Paris, France
| | - Jennifer Pasquier
- INSERM UMRS 938, Centre de recherche Saint Antoine, Team Cancer Biology and Therapeutics, Institut Universitaire de Cancérologie, Sorbonne Université, F-75012 Paris, France; Department of Genetic Medicine, Weill Cornell Medicine, Qatar
| |
Collapse
|
91
|
Rébé C, Ghiringhelli F. Interleukin-1β and Cancer. Cancers (Basel) 2020; 12:E1791. [PMID: 32635472 PMCID: PMC7408158 DOI: 10.3390/cancers12071791] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Within a tumor, IL-1β is produced and secreted by various cell types, such as immune cells, fibroblasts, or cancer cells. The IL1B gene is induced after "priming" of the cells and a second signal is required to allow IL-1β maturation by inflammasome-activated caspase-1. IL-1β is then released and leads to transcription of target genes through its ligation with IL-1R1 on target cells. IL-1β expression and maturation are guided by gene polymorphisms and by the cellular context. In cancer, IL-1β has pleiotropic effects on immune cells, angiogenesis, cancer cell proliferation, migration, and metastasis. Moreover, anti-cancer treatments are able to promote IL-1β production by cancer or immune cells, with opposite effects on cancer progression. This raises the question of whether or not to use IL-1β inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Cédric Rébé
- Platform of Transfer in Cancer Biology, Centre Georges François Leclerc, INSERM LNC UMR1231, University of Bourgogne Franche-Comté, F-21000 Dijon, France
| | - François Ghiringhelli
- Platform of Transfer in Cancer Biology, Centre Georges François Leclerc, INSERM LNC UMR1231, University of Bourgogne Franche-Comté, F-21000 Dijon, France
| |
Collapse
|
92
|
Fluoropyrimidine Modulation of the Anti-Tumor Immune Response-Prospects for Improved Colorectal Cancer Treatment. Cancers (Basel) 2020; 12:cancers12061641. [PMID: 32575843 PMCID: PMC7352193 DOI: 10.3390/cancers12061641] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023] Open
Abstract
Chemotherapy modulates the anti-tumor immune response and outcomes depend on the balance of favorable and unfavorable effects of drugs on anti-tumor immunity. 5-Florouracil (5-FU) is widely used in adjuvant chemotherapy regimens to treat colorectal cancer (CRC) and provides a survival benefit. However, survival remains poor for CRC patients with advanced and metastatic disease and immune checkpoint blockade therapy benefits only a sub-set of CRC patients. Here we discuss the effects of 5-FU-based chemotherapy regimens to the anti-tumor immune response. We consider how different aspects of 5-FU's multi-factorial mechanism differentially affect malignant and immune cell populations. We summarize recent studies with polymeric fluoropyrimidines (e.g., F10, CF10) that enhance DNA-directed effects and discuss how such approaches may be used to enhance the anti-tumor immune response and improve outcomes.
Collapse
|
93
|
Li Z, He C, Zhang J, Zhang H, Wei H, Wu S, Jiang W. P2Y6 Deficiency Enhances Dendritic Cell–Mediated Th1/Th17 Differentiation and Aggravates Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2020; 205:387-397. [DOI: 10.4049/jimmunol.1900916] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 05/14/2020] [Indexed: 01/16/2023]
|
94
|
Javaid N, Choi S. Toll-like Receptors from the Perspective of Cancer Treatment. Cancers (Basel) 2020; 12:E297. [PMID: 32012718 PMCID: PMC7072551 DOI: 10.3390/cancers12020297] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/22/2020] [Accepted: 01/25/2020] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors (TLRs) represent a family of pattern recognition receptors that recognize certain pathogen-associated molecular patterns and damage-associated molecular patterns. TLRs are highly interesting to researchers including immunologists because of the involvement in various diseases including cancers, allergies, autoimmunity, infections, and inflammation. After ligand engagement, TLRs trigger multiple signaling pathways involving nuclear factor-κB (NF-κB), interferon-regulatory factors (IRFs), and mitogen-activated protein kinases (MAPKs) for the production of various cytokines that play an important role in diseases like cancer. TLR activation in immune as well as cancer cells may prevent the formation and growth of a tumor. Nonetheless, under certain conditions, either hyperactivation or hypoactivation of TLRs supports the survival and metastasis of a tumor. Therefore, the design of TLR-targeting agonists as well as antagonists is a promising immunotherapeutic approach to cancer. In this review, we mainly describe TLRs, their involvement in cancer, and their promising properties for anticancer drug discovery.
Collapse
Affiliation(s)
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea;
| |
Collapse
|
95
|
STAT3 inhibitory stattic enhances immunogenic cell death induced by chemotherapy in cancer cells. ACTA ACUST UNITED AC 2020; 28:159-169. [PMID: 31942696 DOI: 10.1007/s40199-020-00326-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/07/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Induction of immunogenic cell death (ICD) is considered a promising strategy for cancer immunotherapy. Stattic is an inhibitor of STAT3, which is found constitutively active in many cancers and plays a major role in cancer progression. OBJECTIVES In the present study, we proposed to evaluate whether stattic can enhance the effects of chemotherapy in the induction of ICD in cancer cells harboring hyperactive STAT3. METHODS The growth inhibitory effects of stattic and chemo agents including doxorubicin (DOX) and oxaliplatin (OXP) were evaluated using MTT assay in B16F10 and CT26 cell lines. Flow cytometry was applied to study cell apoptosis and calreticulin (CRT) surface exposure. The levels of high mobility group box 1 (HGMB1), heat shock protein70 (HSP70) and interleukin-12 (IL-12) were measured using ELISA. RESULTS Treatment of B16F10 and CT26 cells with stattic in combination with DOX resulted in synergistic antitumor effects with combination index being 0.82 and 0.87, respectively. Interestingly, we found a higher level of ICD markers including CRT expression as well as HMGB1 and HSP70 secretion in the cells received combination therapy of stattic and DOX as compared with monotherapies. Moreover, exposure of dendritic cells (DCs) to conditioned media (CM) from cancer cells treated with stattic and/or DOX resulted in secretion of IL-12, which is an indicator of DCs maturation and induction of Th1 response. OXP and stattic monotherapy induced ICD in CT26 cells and stimulated IL-12 secretion by DCs; however, we did not observe a significant increase in the level of ICD in CT26 cells and IL-12 secretion by DCs when CT26 cells were treated with stattic and OXP combination as compared with monotherapy groups. CONCLUSION These findings indicate that STAT3 inhibitory stattic can increase ICD induced by DOX. Graphical abstract.
Collapse
|
96
|
Wang Y, Hays E, Rama M, Bonavida B. Cell-mediated immune resistance in cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:232-251. [PMID: 35310881 PMCID: PMC8932590 DOI: 10.20517/cdr.2019.98] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/13/2019] [Accepted: 12/19/2019] [Indexed: 11/23/2022]
Abstract
The genetic and epigenetic aberrations that underlie immune resistance lead to tumors that are refractory to clinically established and experimental immunotherapies, including monoclonal antibodies and T cell-based therapies. From various forms of cytotoxic T cells to small molecule inhibitors that revamp the tumor microenvironment, these therapies have demonstrated notable responses in cancer models and a resistant subset of cancer patients, used both alone and in combination. However, even current approaches, such as those targeting checkpoint molecules, tumor ligands, and involving gene-related therapies, present a challenge in non-responding patients. In this perspective, we discuss the most common mechanisms of immune resistance, including tumor heterogeneity, tumor ligand and major histocompatibility complex modulation, anti-apoptotic pathways, checkpoint inhibitory ligands, immunosuppressive cells and factors in the tumor microenvironment, and activation-induced cell death. In addition, we discuss the strategies designed to circumvent these resistance pathways to showcase the potential of emerging technologies in battling the rise of resistance.
Collapse
Affiliation(s)
- Yuhao Wang
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, The University of California, Los Angeles, Los Angeles, CA 90025-1747, USA
| | - Emily Hays
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, The University of California, Los Angeles, Los Angeles, CA 90025-1747, USA
| | - Martina Rama
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, The University of California, Los Angeles, Los Angeles, CA 90025-1747, USA
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, The University of California, Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
97
|
Bains SJ, Abrahamsson H, Flatmark K, Dueland S, Hole KH, Seierstad T, Redalen KR, Meltzer S, Ree AH. Immunogenic cell death by neoadjuvant oxaliplatin and radiation protects against metastatic failure in high-risk rectal cancer. Cancer Immunol Immunother 2019; 69:355-364. [PMID: 31893287 PMCID: PMC7044156 DOI: 10.1007/s00262-019-02458-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/18/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVE High rates of systemic failure in locally advanced rectal cancer call for a rational use of conventional therapies to foster tumor-defeating immunity. METHODS We analyzed the high-mobility group box-1 (HMGB1) protein, a measure of immunogenic cell death (ICD), in plasma sampled from 50 patients at the time of diagnosis and following 4 weeks of induction chemotherapy and 5 weeks of sequential chemoradiotherapy, both neoadjuvant modalities containing oxaliplatin. The patients had the residual tumor resected and were followed for long-term outcome. RESULTS Patients who met the main study end point-freedom from distant recurrence-showed a significant rise in HMGB1 during the induction chemotherapy and consolidation over the chemoradiotherapy. The higher the ICD increase, the lower was the metastatic failure risk (hazard ratio 0.26, 95% confidence interval 0.11-0.62, P = 0.002). However, patients who received the full-planned oxaliplatin dose of the chemoradiotherapy regimen had poorer metastasis-free survival (P = 0.020) than those who had the oxaliplatin dose reduced to avert breach of the radiation delivery, which is critical to maintain efficient tumor cell kill and in the present case, probably also protected the ongoing radiation-dependent ICD response from systemic oxaliplatin toxicity. CONCLUSION The findings indicated that full-dose induction oxaliplatin followed by an adapted oxaliplatin dose that was compliant with full-intensity radiation caused induction and maintenance of ICD and as a result, durable disease-free outcome for a patient population prone to metastatic progression.
Collapse
Affiliation(s)
- Simer J Bains
- Department of Oncology, Akershus University Hospital, P.O. Box 1000, 1478, Lørenskog, Norway
| | - Hanna Abrahamsson
- Department of Oncology, Akershus University Hospital, P.O. Box 1000, 1478, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kjersti Flatmark
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Tumor Biology, Oslo University Hospital, Oslo, Norway.,Department of Gastroenterological Surgery, Oslo University Hospital, Oslo, Norway
| | - Svein Dueland
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Knut H Hole
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Radiology, Oslo University Hospital, Oslo, Norway
| | - Therese Seierstad
- Division of Radiology and Nuclear Medicine, Department of Research and Development, Oslo University Hospital, Oslo, Norway
| | - Kathrine Røe Redalen
- Department of Oncology, Akershus University Hospital, P.O. Box 1000, 1478, Lørenskog, Norway.,Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sebastian Meltzer
- Department of Oncology, Akershus University Hospital, P.O. Box 1000, 1478, Lørenskog, Norway
| | - Anne Hansen Ree
- Department of Oncology, Akershus University Hospital, P.O. Box 1000, 1478, Lørenskog, Norway. .,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
98
|
BET bromodomain inhibitor JQ1 promotes immunogenic cell death in tongue squamous cell carcinoma. Int Immunopharmacol 2019; 76:105921. [DOI: 10.1016/j.intimp.2019.105921] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 09/15/2019] [Accepted: 09/15/2019] [Indexed: 02/08/2023]
|
99
|
Braunstein MJ, Kucharczyk J, Adams S. Targeting Toll-Like Receptors for Cancer Therapy. Target Oncol 2019; 13:583-598. [PMID: 30229471 DOI: 10.1007/s11523-018-0589-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The immune system encompasses a broad array of defense mechanisms against foreign threats, including invading pathogens and transformed neoplastic cells. Toll-like receptors (TLRs) are critically involved in innate immunity, serving as pattern recognition receptors whose stimulation leads to additional innate and adaptive immune responses. Malignant cells exploit the natural immunomodulatory functions of TLRs, expressed mainly by infiltrating immune cells but also aberrantly by tumor cells, to foster their survival, invasion, and evasion of anti-tumor immune responses. An extensive body of research has demonstrated context-specific roles for TLR activation in different malignancies, promoting disease progression in certain instances while limiting cancer growth in others. Despite these conflicting roles, TLR agonists have established therapeutic benefits as anti-cancer agents that activate immune cells in the tumor microenvironment and facilitate the expression of cytokines that allow for infiltration of anti-tumor lymphocytes and the suppression of oncogenic signaling pathways. This review focuses on the clinical application of TLR agonists for cancer treatment. We also highlight agents that are undergoing development in clinical trials, including investigations of TLR agonists in combination with other immunotherapies.
Collapse
Affiliation(s)
- Marc J Braunstein
- Department of Medicine, NYU Winthrop Hospital, 120 Mineola Blvd. Suite 500, Mineola, 11501, NY, USA
| | - John Kucharczyk
- Department of Medicine, NYU Winthrop Hospital, 120 Mineola Blvd. Suite 500, Mineola, 11501, NY, USA
| | - Sylvia Adams
- Department of Medicine, NYU Langone Medical Center, Laura and Isaac Perlmutter Cancer Center, 160 East 34th Street, 4th Floor, New York, 10016, NY, USA.
| |
Collapse
|
100
|
Andersohn A, Garcia MI, Fan Y, Thompson MC, Akimzhanov AM, Abdullahi A, Jeschke MG, Boehning D. Aggregated and Hyperstable Damage-Associated Molecular Patterns Are Released During ER Stress to Modulate Immune Function. Front Cell Dev Biol 2019; 7:198. [PMID: 31620439 PMCID: PMC6759876 DOI: 10.3389/fcell.2019.00198] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 09/02/2019] [Indexed: 12/23/2022] Open
Abstract
Chronic ER stress occurs when protein misfolding in the Endoplasmic reticulum (ER) lumen remains unresolved despite activation of the unfolded protein response. We have shown that traumatic injury such as a severe burn leads to chronic ER stress in vivo leading to systemic inflammation which can last for more than a year. The mechanisms linking chronic ER stress to systemic inflammatory responses are not clear. Here we show that induction of chronic ER stress leads to the release of known and novel damage-associated molecular patterns (DAMPs). The secreted DAMPs are aggregated and markedly protease resistant. ER stress-derived DAMPs activate dendritic cells (DCs) which are then capable of polarizing naïve T cells. Our findings indicate that induction of chronic ER stress may lead to the release of hyperstable DAMPs into the circulation resulting in persistent systemic inflammation and adverse outcomes.
Collapse
Affiliation(s)
- Alexander Andersohn
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, TX, United States
| | - M Iveth Garcia
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, TX, United States
| | - Ying Fan
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, TX, United States
| | - Max C Thompson
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, TX, United States
| | - Askar M Akimzhanov
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, TX, United States
| | - Abdikarim Abdullahi
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Marc G Jeschke
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Darren Boehning
- Department of Biochemistry and Molecular Biology, McGovern Medical School at UTHealth, Houston, TX, United States.,Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| |
Collapse
|