101
|
Yu G, Gao J, Hu W, Hu D, Wang W, Yang S, Gao J. ORM1 promotes tumor progression of kidney renal clear cell carcinoma (KIRC) through CALR-mediated apoptosis. Sci Rep 2023; 13:15687. [PMID: 37735575 PMCID: PMC10514263 DOI: 10.1038/s41598-023-42962-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 09/17/2023] [Indexed: 09/23/2023] Open
Abstract
Kidney renal clear cell carcinoma (KIRC) is the most prevalent type of kidney cancer and causes thousands of deaths each year. The prognosis for KIRC is poor. One critical factor is that the mechanism beneath KIRC is unclear. ORM1 is a reactant to acute inflammation. In this study, we demonstrated that methylation of ORM1 promoter was low and ORM1 was expressed significantly higher in KIRC. KIRC with higher ORM1 expression exhibited worse survival probability. Meanwhile, ORM1 was expressed higher in KIRC cell lines. When ORM1 was knocked down, cell proliferation ability was inhibited potently compared to the NC control. Cell migration as well as invasion ability were also suppressed dramatically. At molecular level, the expression of active caspase-3 and Bax was upregulated in ORM1-KD group while Bcl-2 downregulated. Moreover, CALR decreased following ORM1-KD and rescued expression of CALR increased Bcl-2 level but reduced the level of cleaved caspase-3 and Bax. Consistently, the apoptotic rate of 786-O and Caki-2 cells was upregulated in ORM1-KD but downregulated after CALR overexpression. The activity of caspase-3 was also regulated by ORM1-KD. In addition, the inhibition rate of sorafenib was enhanced in ORM1 KD group but reduced after overexpression of ORM1. Conclusively, ORM1 is clinically associated with progression of KIRC and regulates cell proliferation, migration, invasion, and apoptosis in KIRC. Moreover, ORM1 affects the efficiency of sorafenib in KIRC and regulates caspase-3 mediated cascades response through CALR molecule. This study provides us a new way to recognize the development and progression in KIRC.
Collapse
Affiliation(s)
- Gang Yu
- Department of Nephrology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, 200233, China
| | - Juan Gao
- Physical Examination Center, Shanghai Lung Hospital Affiliated to Tongji University, Shanghai, 200433, China
| | - Weifeng Hu
- Department of Nephrology, PLA Naval Medical Center, Shanghai, 200052, China
| | - Dayong Hu
- Department of Nephrology, The Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| | - Weibing Wang
- Department of Epidemiology, School of Public Health of Fudan University, Shanghai, 200032, China
| | - Shiquan Yang
- Department of General Practice, Xujiahui Community Healthcare Center of Xuhui District of Shanghai, West Guangyuan Road No.349, Shanghai, 200030, China.
| | - Jing Gao
- Department of General Practice, Xujiahui Community Healthcare Center of Xuhui District of Shanghai, West Guangyuan Road No.349, Shanghai, 200030, China.
| |
Collapse
|
102
|
Elgohary S, Eissa RA, El Tayebi HM. Thymoquinone, a Novel Multi-Strike Inhibitor of Pro-Tumorigenic Breast Cancer (BC) Markers: CALR, NLRP3 Pathway and sPD-L1 in PBMCs of HR+ and TNBC Patients. Int J Mol Sci 2023; 24:14254. [PMID: 37762557 PMCID: PMC10531892 DOI: 10.3390/ijms241814254] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/19/2023] [Accepted: 06/25/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer (BC) is not only a mass of malignant cells but also a systemic inflammatory disease. BC pro-tumorigenic inflammation has been shown to promote immune evasion and provoke BC progression. The NOD-like receptor (NLR) family pyrin domain-containing protein 3 (NLRP3) inflammasome is activated when pattern recognition receptors (PRRs) sense danger signals such as calreticulin (CALR) from damaged/dying cells, leading to the secretion of interleukin-1β (IL-1β). CALR is a novel BC biological marker, and its high levels are associated with advanced tumors. NLRP3 expression is strongly correlated with an elevated proliferative index Ki67, BC progression, metastasis, and recurrence in patients with hormone receptor-positive (HR+) and triple-negative BC (TNBC). Tumor-associated macrophages (TAMs) secrete high levels of IL-1β promoting endocrine resistance in HR+ BC. Recently, an immunosuppressive soluble form of programmed death ligand 1 (sPD-L1) has been identified as a novel prognostic biomarker in triple-negative breast cancer (TNBC) patients. Interestingly, IL-1β induces sPD-L1 release. BC Patients with elevated IL-1β and sPD-L1 levels show significantly short progression-free survival. For the first time, this study aims to investigate the inhibitory impact of thymoquinone (TQ) on CALR, the NLRP3 pathway and sPD-L1 in HR+ and TNBC. Blood samples were collected from 45 patients with BC. The effect of differing TQ concentrations for different durations on the expression of CALR, NLRP3 complex components and IL-1β as well as the protein levels of sPD-L1 and IL-1β were investigated in the peripheral blood mononuclear cells (PBMCs) and TAMs of TNBC and HR+ BC patients, respectively. The findings showed that TQ significantly downregulated the expression of CALR, NLRP3 components and IL-1β together with the protein levels of secreted IL-1β and sPD-L1. The current findings demonstrated novel immunomodulatory effects of TQ, highlighting its potential role not only as an excellent adjuvant but also as a possible immunotherapeutic agent in HR+ and TNBC patients.
Collapse
Affiliation(s)
- Sawsan Elgohary
- Clinical Pharmacology and Pharmacogenomics Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt;
| | - Reda A. Eissa
- Department of Surgery, Faculty of Medicine, Ain Shams University, Cairo 11591, Egypt;
| | - Hend M. El Tayebi
- Clinical Pharmacology and Pharmacogenomics Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt;
| |
Collapse
|
103
|
Pace A, Scirocchi F, Napoletano C, Zizzari IG, Po A, Megiorni F, Asquino A, Pontecorvi P, Rahimi H, Marchese C, Ferretti E, Nuti M, Rughetti A. Targeting FGFRs by pemigatinib induces G1 phase cell cycle arrest, cellular stress and upregulation of tumor suppressor microRNAs. J Transl Med 2023; 21:626. [PMID: 37715207 PMCID: PMC10504800 DOI: 10.1186/s12967-023-04450-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/18/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Fibroblast growth factor receptor (FGFR) gene family alterations are found in several cancers, indicating their importance as potential therapeutic targets. The FGFR-tyrosine kinase inhibitor (TKI) pemigatinib has been introduced in the treatment of advanced cholangiocarcinoma and more recently for relapsed or refractory myeloid/lymphoid neoplasms with FGFR2 and FGFR1 rearrangements, respectively. Several clinical trials are currently investigating the possible combination of pemigatinib with immunotherapy. In this study, we analyzed the biological and molecular effects of pemigatinib on different cancer cell models (lung, bladder, and gastric), which are currently objective of clinical trial investigations. METHODS NCI-H1581 lung, KATO III gastric and RT-112 bladder cancer cell lines were evaluated for FGFR expression by qRT-PCR and Western blot. Cell lines were treated with Pem and then characterized for cell proliferation, apoptosis, production of intracellular reactive oxygen species (ROS), and induction of senescence. The expression of microRNAs with tumor suppressor functions was analyzed by qRT-PCR, while modulation of the proteins coded by their target genes was evaluated by Western blot and mRNA. Descriptive statistics was used to analyze the various data and student's t test to compare the analysis of two groups. RESULTS Pemigatinib exposure triggered distinct signaling pathways and reduced the proliferative ability of all cancer cells, inducing G1 phase cell cycle arrest and strong intracellular stress resulting in ROS production, senescence and apoptosis. Pemigatinib treatment also caused the upregulation of microRNAs (miR-133b, miR-139, miR-186, miR-195) with tumor suppressor functions, along with the downregulation of validated protein targets with oncogenic roles (c-Myc, c-MET, CDK6, EGFR). CONCLUSIONS These results contribute to clarifying the biological effects and molecular mechanisms mediated by the anti-FGFR TKI pemigatinib in distinct tumor settings and support its exploitation for combined therapies.
Collapse
Affiliation(s)
- Angelica Pace
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Fabio Scirocchi
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Chiara Napoletano
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | | | - Agnese Po
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Francesca Megiorni
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Angela Asquino
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Paola Pontecorvi
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Hassan Rahimi
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Elisabetta Ferretti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Marianna Nuti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Aurelia Rughetti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
104
|
Palacon MP, Ferrisse TM, Barbeiro CDO, Massucato EMS, Bufalino A. Calreticulin Expression in Human Carcinomas: A Systematic Review and Meta-Analysis. Asian Pac J Cancer Prev 2023; 24:2929-2940. [PMID: 37774043 PMCID: PMC10762747 DOI: 10.31557/apjcp.2023.24.9.2929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023] Open
Abstract
OBJECTIVE The present study performed a systematic review and meta-analysis of observational studies on whether calreticulin levels could represent a prognostic factor in carcinoma patients. Calreticulin (CRT) is a multifunctional protein in the endoplasmic reticulum that can play distinct roles in different cancers. METHODS The search was performed in PubMed, Scopus, the Cochrane Library, Web of Science, Lilacs, Science Direct, Embase, Bireme, and SciELO databases. After a full-text evaluation, only 14 articles remained. The RoBANS tool assessed the risk of bias. The meta-analysis was performed with R software, and the odds ratio (OR) was the effect measure. The random effects model was chosen, and the quality of evidence was evaluated according to GRADE. RESULT The most frequent carcinomas were in the breasts and the colon. CRT expression varied according to carcinoma origin and type, but these diseases had a prevalence of high CRT levels, indicating tumor progression. The high CRT levels were associated with lymph node metastasis (OR = 3.06 [1.71; 5.48]/p = 0.0002/I2 = 0%). All included articles had a blinding bias. CONCLUSION High CRT levels may represent a prognostic factor for metastatic lymph nodes in carcinoma patients.
Collapse
Affiliation(s)
- Mariana Paravani Palacon
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| | - Tulio Morandin Ferrisse
- Department of Dental Prothesis, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| | - Camila de Oliveira Barbeiro
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| | - Elaine Maria Sgavioli Massucato
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| | - Andreia Bufalino
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| |
Collapse
|
105
|
Zhang M, Xiao J, Liu J, Bai X, Zeng X, Zhang Z, Liu F. Calreticulin as a marker and therapeutic target for cancer. Clin Exp Med 2023; 23:1393-1404. [PMID: 36335525 DOI: 10.1007/s10238-022-00937-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022]
Abstract
Calreticulin (CRT) is a multifunctional protein found within the endoplasmic reticulum (ER). In addition, CRT participates in the formation and development of tumors and promotes the proliferation and migration of tumor cells. When a malignant tumor occurs in the human body, cancer cells that die from immunogenic cell death (ICD) expose CRT on their surface, and CRT that is transferred to the cell surface represents an "eat me" signal, which promotes dendritic cells to phagocytose the tumor cells, thereby increasing the sensitivity of tumors to anticancer immunotherapy. Expression of CRT in tumor tissues is higher than in normal tissues and is associated with disease progression in many malignant tumors. Thus, the dysfunctional production of CRT can promote tumorigenesis because it disturbs not only the balance of healthy cells but also the body's immune surveillance. CRT may be a diagnostic marker and a therapeutic target for cancer, which is discussed extensively in this review.
Collapse
Affiliation(s)
- Meilan Zhang
- Department of Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Juan Xiao
- Department of Otolaryngology, Hengyang Medical School, The Second Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Jiangrong Liu
- Department of Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xue Bai
- Department of Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xuemei Zeng
- Department of Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhiwei Zhang
- Department of Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Feng Liu
- Department of Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
106
|
Qin Y, Zhang H, Li Y, Xie T, Yan S, Wang J, Qu J, Ouyang F, Lv S, Guo Z, Wei H, Yu CY. Promotion of ICD via Nanotechnology. Macromol Biosci 2023; 23:e2300093. [PMID: 37114599 DOI: 10.1002/mabi.202300093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/17/2023] [Indexed: 04/29/2023]
Abstract
Immunotherapy represents the most promising treatment strategy for cancer, but suffers from compromised therapeutic efficiency due to low immune activity of tumor cells and an immunosuppressive microenvironment, which significantly hampers the clinical translations of this treatment strategy. To promote immunotherapy with desired therapeutic efficiency, immunogenic cell death (ICD), a particular type of death capable of reshaping body's antitumor immune activity, has drawn considerable attention due to the potential to stimulate a potent immune response. Still, the potential of ICD effect remains unsatisfactory because of the intricate tumor microenvironment and multiple drawbacks of the used inducing agents. ICD has been thoroughly reviewed so far with a general classification of ICD as a kind of immunotherapy strategy and repeated discussion of the related mechanism. However, there are no published reviews, to the authors' knowledge, providing a systematic summarization on the enhancement of ICD via nanotechnology. For this purpose, this review first discusses the four stages of ICD according to the development mechanisms, followed by a comprehensive description on the use of nanotechnology to enhance ICD in the corresponding four stages. The challenges of ICD inducers and possible solutions are finally summarized for future ICD-based enhanced immunotherapy.
Collapse
Affiliation(s)
- Yang Qin
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Haitao Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yunxian Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ting Xie
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Shuang Yan
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jiaqi Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jun Qu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Feijun Ouyang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Shaoyang Lv
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zifen Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|
107
|
Fan P, Guan Y, Zhang X, Wang J, Xu Y, Song B, Zhang S, Wang H, Liu Y, Qiao ZY. Cell membrane-specific self-assembly of peptide nanomedicine induces tumor immunogenic death to enhance cancer therapy. NANOSCALE HORIZONS 2023; 8:1226-1234. [PMID: 37366007 DOI: 10.1039/d3nh00173c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Immunogenic cell death (ICD), as an unusual cell death pattern, mediates cancer cells to release a series of damage-associated molecular patterns (DAMPs), and is widely used in the field of cancer immunotherapy. Injuring the cell membrane can serve as a novel ICD initiation strategy. In this study, a peptide nanomedicine (PNpC) is designed using the fragment CM11 of cecropin, which is effective in disrupting cell membranes because of its α-helical structure. PNpC self-assembles in situ in the presence of high levels of alkaline phosphatase (ALP) on the tumor cell membrane, transforming from nanoparticles to nanofibers, which reduces the cellular internalization of the nanomedicine and increases the interaction between CM11 and tumor cell membranes. Both in vitro and in vivo results indicate that PNpC plays a significant role in killing tumor cells by triggering ICD. The ICD induced by the destruction of the cancer cell membrane is accompanied by the release of DAMPs, which promotes the maturation of DCs and facilitates the presentation of tumor-associated antigens (TAA), resulting in the infiltration of CD8+ T cells. We believe that PNpC can trigger ICD while killing cancer cells, providing a new reference for cancer immunotherapy.
Collapse
Affiliation(s)
- Pengsheng Fan
- College of Marine Life Science, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, China.
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Yinghua Guan
- College of Marine Life Science, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, China.
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Xiaoying Zhang
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Jiaqi Wang
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Yinsheng Xu
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Benli Song
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Suling Zhang
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Hao Wang
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Ya Liu
- College of Marine Life Science, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, China.
| | - Zeng-Ying Qiao
- Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| |
Collapse
|
108
|
SUN DONGJIE, ZHANG CHI. Immunogenic cell death-related long noncoding RNA influences immunotherapy against lung adenocarcinoma. Oncol Res 2023; 31:753-767. [PMID: 37547766 PMCID: PMC10398397 DOI: 10.32604/or.2023.029287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/06/2023] [Indexed: 08/08/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is the leading cause of cancer-related deaths, accounting for over a million deaths worldwide annually. Immunogenic cell death (ICD) elicits an adaptive immune response. However, the role of ICD-related long noncoding RNAs (lncRNAs) in LUAD is unknown. In this study, we investigated the characteristics of the tumor microenvironment in LUAD, the prognostic significance of ICD-related lncRNAs, and the half-maximal inhibitory concentration (IC50) of possible chemotherapeutic drugs. We sorted prognostic lncRNAs using univariate Cox regression and constructed a risk signature based on them. We then confirmed the model's accuracy and generated a nomogram. Additionally, we performed immune microenvironment analysis, somatic mutation calculation, Tumor Immune Dysfunction and Exclusion (TIDE) analysis, and anticancer pharmaceutical IC50 prediction. Least absolute shrinkage and selection operator Cox regression identified 27 prognostic lncRNAs related to ICD, and a unique risk signature using 10 ICD-related lncRNAs was constructed. The risk score was confirmed to be a reliable predictor of survival, with the highest c-index score. The signature had a remarkable predictive performance with clinical applicability and could accurately predict the overall survival in LUAD. Furthermore, the lncRNA signature was closely associated with immunocyte invasion. We also analyzed the correlation between the risk score, tumor-infiltrating immune cells, and prognosis and identified high immune and ESTIMATE scores in low-risk patients. Moreover, we observed elevated checkpoint gene expression and low TIDE scores in high-risk patients, indicating a good immunotherapy response. Finally, high-risk patients were shown to be susceptible to anticancer medications. Therefore, our unique risk signature comprising 10 ICD-related lncRNAs was demonstrated to indicate the characteristics of the tumor-immune microenvironment in LUAD, predict patients' overall survival, and guide individualized treatment.
Collapse
Affiliation(s)
- DONGJIE SUN
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, China
- College of Basic Medical Sciences, Jilin University, Changchun, China
| | - CHI ZHANG
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
109
|
Xu K, Ge Q, Zhang Y, Ouyang G, Yan X. Expression properties, structural features and functional analysis of CALR E381A in MPN patients. Am J Transl Res 2023; 15:4718-4726. [PMID: 37560236 PMCID: PMC10408505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/24/2023] [Indexed: 08/11/2023]
Abstract
OBJECTIVE To investigate the expression properties, structural features and function of CALR E381A in myeloproliferative neoplasms (MPN) patients. METHODS In this retrospective study, 435 MPN patients admitted to the Department of Hematology, Ningbo First Hospital from July 2015 to July 2021 were selected as the study subjects. Mutations in CALR exon 9 from genomic DNA samples were identified by PCR, followed by Sanger sequencing. The physicochemical properties of the wild-type calreticulin and the p.E381A variant, and the structural information of the p.E381A variant were analyzed by using the bioinformatics databases. Growth assay of UT-7/mpl cells with CALR E381A was used for the functional analysis of CALR E381A. RESULTS The predominant types of CALR variants were identified as follows: p.L367fs*46 (38.1%), p.K385fs*47 (25.8%) and p.E381A (19.6%). Notably, the frequency of the p.E381A variant (c.1142A >C) in polycythemia vera or essential thrombocythemia was significantly higher than the frequency of that as a single nucleotide polymorphism (SNP) in the East Asian population. Furthermore, CALR E381A coexisted with other genetic variants, of which JAK2 V617F was more common. Bioinformatics analysis confirmed that CALR E381A did not change the physicochemical properties of the calreticulin protein, but did change the electrical charge, energy state and steric hindrance of amino acid residues at site 381. UT-7/mpl cells harboring CALR E381A overexpression did not exhibit altered cell growth, which is distinctly different from the stereotypical frameshift mutation. CONCLUSION CALR E381A is not a driver mutation for the development of MPN but may be a risk SNP implying an inherited predisposition for MPN disease in East Asian populations.
Collapse
Affiliation(s)
- Kaihong Xu
- Department of Hematology, Ningbo First Hospital Ningbo, Zhejiang, China
| | - Qunfang Ge
- Department of Hematology, Ningbo First Hospital Ningbo, Zhejiang, China
| | - Yanli Zhang
- Department of Hematology, Ningbo First Hospital Ningbo, Zhejiang, China
| | - Guifang Ouyang
- Department of Hematology, Ningbo First Hospital Ningbo, Zhejiang, China
| | - Xiao Yan
- Department of Hematology, Ningbo First Hospital Ningbo, Zhejiang, China
| |
Collapse
|
110
|
Wang Y, Wang W, Gu R, Chen J, Chen Q, Lin T, Wu J, Hu Y, Yuan A. In Situ Vaccination with Mitochondria-Targeting Immunogenic Death Inducer Elicits CD8 + T Cell-Dependent Antitumor Immunity to Boost Tumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300286. [PMID: 37127892 PMCID: PMC10369267 DOI: 10.1002/advs.202300286] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/28/2023] [Indexed: 05/03/2023]
Abstract
In situ vaccination can elicit systemic antitumor immunity to potentiate immune checkpoint blockade (ICB) in poorly immunogenic tumors. Herein, an immunogenic cell death (ICD) inducer for in situ vaccination, which is based on a mitochondria-targeting modification of fenofibric acid (FFa), a lipid-lowering drug with potential inhibitory efficacy of respiratory complex I is developed. Mitochondria-targeting FFa (Mito-FFa) inhibits complex I efficiently and increases mitochondrial ROS (mtROS) generation, which further triggers endoplasmic reticulum (ER) stress with unprecedented calreticulin (CRT) exposure on tumor cellular membranes. Moreover, the generated mtROS also oxidizes mitochondrial DNA (mtDNA) and promotes it leakage into the cytoplasm for cGAS-STING-dependent type I interferon (IFN-I) secretion. The synchronous CRT exposure and IFN-I secretion successively improve the uptake of tumor antigens, maturation of dendritic cells (DCs) and cross-priming of CD8+ T cells. In a poorly immunogenic 4T1 tumor model, a single intratumoral (i.t.) Mito-FFa injection turns immune-cold tumors into hot ones and elicits systemic tumor-specific CD8+ T cells responses against primary and metastatic tumors. Furthermore, the synergistic effect with PD-L1 blockade and good bio-safety of i.t. Mito-FFa administration suggest the great translational potential of Mito-FFa in tumor immunotherapy.
Collapse
Affiliation(s)
- Yuxiang Wang
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
| | - Weiran Wang
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
| | - Rong Gu
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
| | - Jing Chen
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
| | - Qian Chen
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
| | - Tingsheng Lin
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
- Department of UrologyNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing210093P. R. China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
- Jiangsu Key Laboratory for Nano TechnologyNanjing UniversityNanjing210093P. R. China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
- Jiangsu Key Laboratory for Nano TechnologyNanjing UniversityNanjing210093P. R. China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
- Jiangsu Key Laboratory for Nano TechnologyNanjing UniversityNanjing210093P. R. China
| |
Collapse
|
111
|
Chen Q, Chen J, Zhang Q, Yang P, Gu R, Ren H, Dai Y, Huang S, Wu J, Wu X, Hu Y, Yuan A. Combining High-Z Sensitized Radiotherapy with CD73 Blockade to Boost Tumor Immunotherapy. ACS NANO 2023. [PMID: 37327456 DOI: 10.1021/acsnano.2c11403] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Radiation therapy (RT) has the capacity to induce immunogenic death in tumor cells, thereby potentially inducing in situ vaccination (ISV) to prime systemic antitumor immune responses. However, RT alone is often faced with various limitations during ISV induction, such as insufficient X-ray deposition and an immunosuppressive microenvironment. To overcome these limitations, we constructed nanoscale coordination particles AmGd-NPs by self-assembling high-Z metal gadolinium (Gd) and small molecular CD73 inhibitor AmPCP. Then, AmGd-NPs could synergize with RT to enhance immunogenic cell death, improve phagocytosis, and promote antigen presentation. Additionally, AmGd-NPs could also gradually release AmPCP to inhibit CD73's enzymatic activity and prevent the conversion of extracellular ATP to adenosine (Ado), thereby driving a proinflammatory tumor microenvironment that promotes DC maturation. As a result, AmGd-NPs sensitized RT induced potent in situ vaccination and boosted CD8+ T cell-dependent antitumor immune responses against both primary and metastatic tumors, which could also be potentiated by immune checkpoint inhibitory therapy.
Collapse
Affiliation(s)
- Qian Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing 210093, China
| | - Jing Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing 210093, China
| | - Qingqing Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing 210093, China
| | - Peizheng Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing 210093, China
| | - Rong Gu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing 210093, China
| | - Hao Ren
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing 210093, China
| | - Yue Dai
- Evaluation Center of Jiangsu Medical Products Administration, Nanjing 210093, China
| | - Shiqian Huang
- State Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing 210093, China
| | - Xudong Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing 210093, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing 210093, China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Science, Nanjing University, Nanjing 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing 210093, China
| |
Collapse
|
112
|
Gu Y, Zhou G, Tang X, Shen F, Ding J, Hua K. The biological roles of CD24 in ovarian cancer: old story, but new tales. Front Immunol 2023; 14:1183285. [PMID: 37359556 PMCID: PMC10288981 DOI: 10.3389/fimmu.2023.1183285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
CD24 is a glycosylphosphatidylinositol linked molecular which expressed in diverse malignant tumor cells, particular in ovarian carcinoma cells and ovarian carcinoma stem cells. The CD24 expression is associated with increased metastatic potential and poor prognosis of malignancies. CD24 on the surface of tumor cells could interact with Siglec-10 on the surface of immune cells, to mediate the immune escape of tumor cells. Nowadays, CD24 has been identified as a promising focus for targeting therapy of ovarian cancer. However, the roles of CD24 in tumorigenesis, metastasis, and immune escape are still not clearly demonstrated systematically. In this review, we i) summarized the existing studies on CD24 in diverse cancers including ovarian cancer, ii) illustrated the role of CD24-siglec10 signaling pathway in immune escape, iii) reviewed the existing immunotherapeutic strategies (targeting the CD24 to restore the phagocytic effect of Siglec-10 expressing immune cells) based on the above mechanisms and evaluated the priorities in the future research. These results might provide support for guiding the CD24 immunotherapy as the intervention upon solid tumors.
Collapse
Affiliation(s)
- Yuanyuan Gu
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Guannan Zhou
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, China
| | - Xue Tang
- Department of Laboratory Medicine, Division of Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fang Shen
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jingxin Ding
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Keqin Hua
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
113
|
Chandrasekar SV, Singh A, Ranjan A. Overcoming Resistance to Immune Checkpoint Inhibitor Therapy Using Calreticulin-Inducing Nanoparticle. Pharmaceutics 2023; 15:1693. [PMID: 37376141 PMCID: PMC10302072 DOI: 10.3390/pharmaceutics15061693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/31/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Nanoparticles (NPs) have the ability to transform poorly immunogenic tumors into activated 'hot' targets. In this study, we investigated the potential of a liposome-based nanoparticle (CRT-NP) expressing calreticulin as an in-situ vaccine to restore sensitivity to anti-CTLA4 immune checkpoint inhibitor (ICI) in CT26 colon tumors. We found that a CRT-NP with a hydrodynamic diameter of approximately 300 nm and a zeta potential of approximately +20 mV induced immunogenic cell death (ICD) in CT-26 cells in a dose-dependent manner. In the mouse model of CT26 xenograft tumors, both CRT-NP and ICI monotherapy caused moderate reductions in tumor growth compared to the untreated control group. However, the combination therapy of CRT-NP and anti-CTLA4 ICI resulted in remarkable suppression of tumor growth rates (>70%) compared to untreated mice. This combination therapy also reshaped the tumor microenvironment (TME), achieving the increased infiltration of antigen-presenting cells (APCs) such as dendritic cells and M1 macrophages, as well as an abundance of T cells expressing granzyme B and a reduction in the population of CD4+ Foxp3 regulatory cells. Our findings indicate that CRT-NPs can effectively reverse immune resistance to anti-CTLA4 ICI therapy in mice, thereby improving the immunotherapeutic outcome in the mouse model.
Collapse
Affiliation(s)
| | | | - Ashish Ranjan
- Department of Physiological Sciences, College of Veterinary, Oklahoma State University, Stillwater, OK 74078, USA; (S.V.C.)
| |
Collapse
|
114
|
Liu X, Chen Y, Fu Y, Jiang D, Gao F, Tang Z, Bian X, Wu S, Yu Y, Wang X, Shen J, Li C. Breaking Spatiotemporal Barriers of Immunogenic Chemotherapy via an Endoplasmic Reticulum Membrane-Assisted Liposomal Drug Delivery. ACS NANO 2023. [PMID: 37207349 DOI: 10.1021/acsnano.3c01446] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Immunogenic chemotherapy is a promising approach in cancer treatment, but the number of drugs capable of inducing immunogenic cell death is limited, and chronic immunogenic exposure can delay antitumor immune response and be counteracted by immunosuppressive factors. In this study, we used single-cell and multilevel analyses to highlight the critical importance of the first exposure to calreticulin (CRT) in eliciting immunogenicity. We then developed the ERASION (endoplasmic reticulum (ER) membrane to assist (AS) the presentation of intrinsic onco-immunogenicity (ION)) strategy, leveraging the high expression of functional proteins, including CRT, on the ER membrane. ER membrane-coated liposome (ER@PLip) was able to target the tumor and immune effectors and promoted dendritic cell maturation and T cell infiltration. This enabled eliciting an immunogenic effect from a nonimmunogenic chemotherapeutic drug. By utilizing the ER membrane-associated STING protein, ERASION enabled activating the STING pathway and the generation of adaptive antitumor immunity. This study presents a potential universal platform for integrating traditional chemotherapy and therapeutic modalities.
Collapse
Affiliation(s)
- Xinlong Liu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Yujuan Chen
- Department of Breast Surgery, Clinical Center for Breast, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yu Fu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Dingxi Jiang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Feiyan Gao
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Zhongjie Tang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Xufei Bian
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Shuang Wu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Yang Yu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Xiaoyou Wang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| | - Jie Shen
- Departments of Biomedical and Pharmaceutical Sciences and Chemical Engineering, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, People's Republic of China
| |
Collapse
|
115
|
Dong Y, Xu C, Su G, Li Y, Yan B, Liu Y, Yin T, Mou S, Mei H. Clinical value of anoikis-related genes and molecular subtypes identification in bladder urothelial carcinoma and in vitro validation. Front Immunol 2023; 14:1122570. [PMID: 37275895 PMCID: PMC10232821 DOI: 10.3389/fimmu.2023.1122570] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/03/2023] [Indexed: 06/07/2023] Open
Abstract
Background Anoikis is a programmed cell death process that was proven to be associated with cancer. Uroepithelial carcinoma of the bladder (BLCA) is a malignant disease of the urinary tract and has a strong metastatic potential. To determine whether anoikis-associated genes can predict the prognosis of BLCA accurately, we evaluated the prognostic value of anoikis-associated genes in BLCA and constructed the best model to predict prognosis. Method The BLCA transcriptome data were downloaded from TCGA and GEO databases, and genes with differential expression were selected and then clustered using non-negative matrix factorization (NMF). The genes with the most correlation with anoikis were screened and identified using univariate Cox regression, lasso regression, and multivariate Cox regression. The GEO dataset was used for external validation. Nomograms were created based on risk characteristics in combination with clinical variants and the performance of the model was validated with receiver operating characteristic (ROC) curves. The immunotherapeutic significance of this risk score was assessed using the immune phenomenon score (IPS). IC50 values of predictive chemotherapeutic agents were calculated. Finally, we used RT-qPCR to determine the mRNA expression of four genes, CALR, FASN, CASP6, and RAD9A. Result We screened 406 tumor samples and 19 normal tissue samples from the TCGA database. Based on anoikis-associated genes, we classified patients into two subtypes (C1 and C2) using NMF method. Subsequently, nine core genes were screened by multiple methods after analysis, which were used to construct risk profiles. The design of nomograms based on risk profiles and clinical variables, ROC, and calibration curves confirmed that the model could well have the ability to predict the survival of BLCA patients at 1, 3, and 5 years. By predicting the IC50 values of chemotherapeutic drugs, it was learned that the high-risk group (HRG) was more susceptible to paclitaxel, gemcitabine, and cisplatin, and the low-risk group (LRG) was more susceptible to veriparib and afatinib. Conclusion In summary, the risk score of anoikis-associated genes can be applied as a predictor to predict the prognosis of BLCA in clinical practice.
Collapse
Affiliation(s)
- Ying Dong
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Key Laboratory of Medical Reprogramming Technology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Chaojie Xu
- Department of Urology, Peking University First Hospital, Institution of Urology, Peking University, Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, National Urological Cancer Center, Beijing, China
| | - Ganglin Su
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Yanfeng Li
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Key Laboratory of Medical Reprogramming Technology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Bing Yan
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Key Laboratory of Medical Reprogramming Technology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yuhan Liu
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Key Laboratory of Medical Reprogramming Technology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Tao Yin
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Key Laboratory of Medical Reprogramming Technology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Shuanzhu Mou
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Key Laboratory of Medical Reprogramming Technology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Hongbing Mei
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, China
- Key Laboratory of Medical Reprogramming Technology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| |
Collapse
|
116
|
Guilbaud E, Kroemer G, Galluzzi L. Calreticulin exposure orchestrates innate immunosurveillance. Cancer Cell 2023:S1535-6108(23)00141-1. [PMID: 37207656 DOI: 10.1016/j.ccell.2023.04.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/21/2023]
Abstract
Calreticulin (CALR) exposure on the cell surface is known to deliver robust pro-phagocytic signals to myeloid cells. In Nature, Sen Santara et al. demonstrate that surface-exposed CALR also operates as an endogenous activator of natural killer (NK) cells. Collectively, these findings suggest that CALR exposure orchestrates multiple facets of innate immunosurveillance.
Collapse
Affiliation(s)
- Emma Guilbaud
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, 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.
| |
Collapse
|
117
|
Liu Z, Xu X, Liu K, Zhang J, Ding D, Fu R. Immunogenic Cell Death in Hematological Malignancy Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207475. [PMID: 36815385 PMCID: PMC10161053 DOI: 10.1002/advs.202207475] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/09/2023] [Indexed: 05/06/2023]
Abstract
Although the curative effect of hematological malignancies has been improved in recent years, relapse or drug resistance of hematological malignancies will eventually recur. Furthermore, the microenvironment disorder is an important mechanism in the pathogenesis of hematological malignancies. Immunogenic cell death (ICD) is a unique mechanism of regulated cell death (RCD) that triggers an intact antigen-specific adaptive immune response by firing a set of danger signals or damage-associated molecular patterns (DAMPs), which is an immunotherapeutic modality with the potential for the treatment of hematological malignancies. This review summarizes the existing knowledge about the induction of ICD in hematological malignancies and the current research on combining ICD inducers with other treatment strategies for hematological malignancies.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of HematologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Xintong Xu
- Department of HematologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Kaining Liu
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive, Materials, Ministry of Education and College of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Jingtian Zhang
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive, Materials, Ministry of Education and College of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive, Materials, Ministry of Education and College of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Rong Fu
- Department of HematologyTianjin Medical University General HospitalTianjin300052P. R. China
| |
Collapse
|
118
|
Chen Y, Zeng L, Zhu H, Wu Q, Liu R, Liang Q, Chen B, Dai H, Tang K, Liao C, Huang Y, Yan X, Fan K, Du JZ, Lin R, Wang J. Ferritin Nanocaged Doxorubicin Potentiates Chemo-Immunotherapy against Hepatocellular Carcinoma via Immunogenic Cell Death. SMALL METHODS 2023; 7:e2201086. [PMID: 36446639 DOI: 10.1002/smtd.202201086] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/05/2022] [Indexed: 05/17/2023]
Abstract
Although immunotherapy of hepatocellular carcinoma using immune checkpoint inhibitors has achieved certain success, only a subset of patients benefits from this therapeutic strategy. The combination of immunostimulatory chemotherapeutics represents a promising strategy to enhance the effectiveness of immunotherapy. However, it is hampered by the poor delivery of conventional chemotherapeutics. Here, it is shown that H-ferritin nanocages loaded with doxorubicin (DOX@HFn) show potent chemo-immunotherapy in hepatocellular carcinoma tumor models. DOX@HFn is constructed with uniform size, high stability, favorable drug loading, and intracellular acidity-driven drug release. The receptor-mediated targeting of DOX@HFn to liver cancer cells promote cellular uptake and tumor penetration in vitro and in vivo. DOX@HFn triggers immunogenic cell death to tumor cells and promotes the subsequent activation and maturation of dendritic cells. In vivo studies in H22 subcutaneous hepatoma demonstrate that DOX@HFn significantly inhibits the tumor growth with >30% tumors completely eliminated, while alleviating the systemic toxicity of free DOX. DOX@HFn also exhibits robust antitumor immune response and tumoricidal effect in a more aggressive Hepa1-6 orthotopic liver tumor model, which is confirmed by the in situ magnetic resonance imaging and transcriptome sequencing. This study provides a facile and robust strategy to improve therapeutic efficacy of liver cancer.
Collapse
Affiliation(s)
- Yang Chen
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Linyuan Zeng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Center for Precision Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Hongzhang Zhu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Qifei Wu
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Rong Liu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Qian Liang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bin Chen
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Haitao Dai
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Keyu Tang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Changli Liao
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yonghui Huang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xiyun Yan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jin-Zhi Du
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Run Lin
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Center for Precision Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, 511442, China
| |
Collapse
|
119
|
Zhu H, Deng J, Yuan M, Rong X, Xiang X, Du F, Luo X, Cheng C, Qiu L. Semiconducting Titanate Supported Ruthenium Clusterzymes for Ultrasound-Amplified Biocatalytic Tumor Nanotherapies. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206911. [PMID: 36765452 DOI: 10.1002/smll.202206911] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/15/2023] [Indexed: 05/04/2023]
Abstract
The external-stimulation-induced reactive-oxygen-species (ROS) generation has attracted increasing attention in therapeutics for malignant tumors. However, engineering a nanoplatform that integrates with efficient biocatalytic ROS generation, ultrasound-amplified ROS production, and simultaneous relief of tumor hypoxia is still a great challenge. Here, we create new semiconducting titanate-supported Ru clusterzymes (RuNC/BTO) for ultrasound-amplified biocatalytic tumor nanotherapies. The morphology and chemical/electronic structure analysis prove that the biocatalyst consists of Ru nanoclusters that are tightly stabilized by Ru-O coordination on BaTiO3 . The peroxidase (POD)- and halogenperoxidase-like biocatalysis reveals that the RuNC/BTO can produce abundant •O2 - radicals. Notably, the RuNC/BTO exhibits the highest turnover number (63.29 × 10-3 s-1 ) among the state-of-the-art POD-mimics. Moreover, the catalase-like activity of the RuNC/BTO facilitates the decomposition of H2 O2 to produce O2 for relieving the hypoxia of the tumor and amplifying the ROS level via ultrasound irradiation. Finally, the systematic cellular and animal experiments have validated that the multi-modal strategy presents superior tumor cell-killing effects and suppression abilities. We believe that this work will offer an effective clusterzyme that can adapt to the tumor microenvironment-specific catalytic therapy and also provide a new pathway for engineering high-performance ROS production materials across broad therapeutics and biomedical fields.
Collapse
Affiliation(s)
- Huang Zhu
- College of Polymer Science and Engineering, Med-X Center for Materials, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Jiuhong Deng
- West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Minjia Yuan
- College of Polymer Science and Engineering, Med-X Center for Materials, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Xiao Rong
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xi Xiang
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fangxue Du
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xianglin Luo
- College of Polymer Science and Engineering, Med-X Center for Materials, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Chong Cheng
- College of Polymer Science and Engineering, Med-X Center for Materials, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Li Qiu
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
120
|
Lim JU, Lee E, Lee SY, Cho HJ, Ahn DH, Hwang Y, Choi JY, Yeo CD, Park CK, Kim SJ. Current literature review on the tumor immune micro-environment, its heterogeneity and future perspectives in treatment of advanced non-small cell lung cancer. Transl Lung Cancer Res 2023; 12:857-876. [PMID: 37197639 PMCID: PMC10183402 DOI: 10.21037/tlcr-22-633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 02/07/2023] [Indexed: 03/21/2023]
Abstract
Background and Objective Immune checkpoint inhibitors (ICI) were a major clinical advancement that provided an opportunity to improve the prognosis of patients with non-small cell lung cancer (NSCLC). However, programmed death-ligand-1 (PD-L1) expression does not sufficiently predict ICI efficacy in NSCLC patients. In recent studies, the tumor immune microenvironment (TIME) was shown to have a central role in lung cancer progression and to affect clinical outcome of patients diagnosed with lung cancer. As development of new therapeutic targets to overcome ICI resistance is a priority, understanding the TIME is important. Recently, a series of studies was conducted to target each component of TIME to improve efficacy of cancer treatment. In this review, important features related to TIME, its heterogeneity and current trends in treatment targeting the component of TIME are discussed. Methods PubMed and PMC were searched from January 1st, 2012 to August 16th, 2022 using the following key words: "NSCLC", "Tumor microenvironment", "Immune", "Metastasis" and "Heterogeneity". Key Content and Findings Heterogeneity in the TIME can be either spatial or temporal. Subsequent to heterogeneous changes in the TIME, treatment of lung cancer can be more challenging because drug resistance is more likely to occur. In terms of the TIME, the main concept for increasing the chance of successful NSCLC treatment is to activate immune responses against tumor cells and inhibit immunosuppressive activities. In addition, relevant research is focused on normalizing an otherwise aberrant TIME in NSCLC patients. Potential therapeutic targets include immune cells, cytokine interactions, and non-immune cells such as fibroblasts or vessels. Conclusions In management of lung cancer, understanding TIME and its heterogeneity is significant to treatment outcomes. Ongoing trials including various treatment modalities such as radiotherapy, cytotoxic chemotherapy, and anti-angiogenic treatment and regimens inhibiting other immunoinhibitory molecules are promising.
Collapse
Affiliation(s)
- Jeong Uk Lim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eunyoung Lee
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang-Yun Lee
- Department of Biomedical Engineering, Gachon University, Seongnam, Republic of Korea
| | - Hyeong Jun Cho
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong Hyuck Ahn
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yongki Hwang
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joon Young Choi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chang Dong Yeo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chan Kwon Park
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Joon Kim
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
121
|
Ahmadzada T, Vijayan A, Vafaee F, Azimi A, Reid G, Clarke S, Kao S, Grau GE, Hosseini-Beheshti E. Small and Large Extracellular Vesicles Derived from Pleural Mesothelioma Cell Lines Offer Biomarker Potential. Cancers (Basel) 2023; 15:cancers15082364. [PMID: 37190292 DOI: 10.3390/cancers15082364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 05/17/2023] Open
Abstract
Pleural mesothelioma, previously known as malignant pleural mesothelioma, is an aggressive and fatal cancer of the pleura, with one of the poorest survival rates. Pleural mesothelioma is in urgent clinical need for biomarkers to aid early diagnosis, improve prognostication, and stratify patients for treatment. Extracellular vesicles (EVs) have great potential as biomarkers; however, there are limited studies to date on their role in pleural mesothelioma. We conducted a comprehensive proteomic analysis on different EV populations derived from five pleural mesothelioma cell lines and an immortalized control cell line. We characterized three subtypes of EVs (10 K, 18 K, and 100 K), and identified a total of 4054 unique proteins. Major differences were found in the cargo between the three EV subtypes. We show that 10 K EVs were enriched in mitochondrial components and metabolic processes, while 18 K and 100 K EVs were enriched in endoplasmic reticulum stress. We found 46 new cancer-associated proteins for pleural mesothelioma, and the presence of mesothelin and PD-L1/PD-L2 enriched in 100 K and 10 K EV, respectively. We demonstrate that different EV populations derived from pleural mesothelioma cells have unique cancer-specific proteomes and carry oncogenic cargo, which could offer a novel means to extract biomarkers of interest for pleural mesothelioma from liquid biopsies.
Collapse
Affiliation(s)
- Tamkin Ahmadzada
- School of Medical Sciences, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Abhishek Vijayan
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Fatemeh Vafaee
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, NSW 2052, Australia
- UNSW Data Science Hub, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ali Azimi
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
- Department of Dermatology, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Glen Reid
- Department of Pathology, University of Otago, Dunedin 9016, New Zealand
| | - Stephen Clarke
- School of Medical Sciences, The University of Sydney, Camperdown, NSW 2006, Australia
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW 2065, Australia
| | - Steven Kao
- School of Medical Sciences, The University of Sydney, Camperdown, NSW 2006, Australia
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, NSW 2050, Australia
- Asbestos Diseases Research Institute, Sydney, NSW 2139, Australia
| | - Georges E Grau
- School of Medical Sciences, The University of Sydney, Camperdown, NSW 2006, Australia
- The Sydney Nano Institute, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Elham Hosseini-Beheshti
- School of Medical Sciences, The University of Sydney, Camperdown, NSW 2006, Australia
- The Sydney Nano Institute, The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
122
|
Zhang D, Jiang C, Zheng X, Lin Z, Zhuang Q, Xie H, Liang Y, Xu Y, Cui L, Liu X, Zeng Y. Normalization of Tumor Vessels by Lenvatinib-Based Metallo-Nanodrugs Alleviates Hypoxia and Enhances Calreticulin-Mediated Immune Responses in Orthotopic HCC and Organoids. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2207786. [PMID: 37052507 DOI: 10.1002/smll.202207786] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/22/2023] [Indexed: 06/19/2023]
Abstract
Immunocheckpoint inhibitors combined with Lenvatinib is the first line treatment for hepatocellular carcinoma (HCC), but their potency is hampered by the low response rate and adverse events. Herein, a targeted therapeutic strategy through the coassembly of Lenvatinib, Adriamycin, Fe3+ ion, and a natural polyphenol (metallo-nanodrugs) is presented by coordination effect for potentiating tumor vascular normalization and systematic chemo-immunotherapy to effectively inhibit the progression of HCC in both orthotopic model and patients-derived organoids. In mice with orthotopic HCC, the obtained metallo-nanodrugs efficiently increase the drug accumulation in orthotopic tumors and can respond to acidic tumor environment. The promotion of tumor vascular normalization by metallo-nanodrugs is observed, which enhances the infiltrating T lymphocytes in tumor, and reinforces the calreticulin-mediated antitumor immunity through alleviating hypoxia, reducing regulatory T cells, and down-regulating PDL1 expression of tumors. The excellent therapeutic efficiency with complete remission of orthotopic tumors (3/6) and long-term survival of mice (4/6, 42 days) are also achieved. Furthermore, the excellent therapeutic effect of metallo-nanodrugs is also validated in 5 patient-derived organoids, and hence can provide a marvelous systemic chemo-immunotherapy strategy for enhancing HCC treatment.
Collapse
Affiliation(s)
- Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Chenwei Jiang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Xiaoyuan Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Zhiwen Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, P. R. China
| | - Qiuyu Zhuang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Huanzhang Xie
- Fujian Key Laboratory of Functional Marine Sensing Materials, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, 350108, P. R. China
| | - Yuzhi Liang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
| | - Yu Xu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Linsheng Cui
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, P. R. China
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350002, P. R. China
| | - Yongyi Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, P. R. China
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, P. R. China
| |
Collapse
|
123
|
Shi F, Huang X, Hong Z, Lu N, Huang X, Liu L, Liang T, Bai X. Improvement strategy for immune checkpoint blockade: A focus on the combination with immunogenic cell death inducers. Cancer Lett 2023; 562:216167. [PMID: 37031916 DOI: 10.1016/j.canlet.2023.216167] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/19/2023] [Accepted: 04/03/2023] [Indexed: 04/11/2023]
Abstract
Cancer immunotherapies have yielded promising outcomes in various malignant tumors by blocking specific immune checkpoint molecules, such as programmed cell death 1 and cytotoxic T lymphocyte antigen 4. However, only a few patients respond to immune checkpoint blockade therapy because of the poor immunogenicity of tumor cells and immune-suppressive tumor microenvironment. Accumulating evidence suggests that chemotherapeutic agents, including oxaliplatin and doxorubicin, not only mediate direct cytotoxicity in tumor cells but also induce immunogenic cancer cell death to stimulate a powerful anti-cancer immune response in the tumor microenvironment. In this review, we summarize the recent advances in cancer combination therapy based on immune checkpoint inhibitors plus immunogenic cell death inducers. Despite some clinical failures and challenges, immunogenic cell death inducers have displayed great potential when combined with immune checkpoint inhibitors for anti-cancer treatment in both preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Fukang Shi
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Xing Huang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Zhengtao Hong
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Na Lu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Xin Huang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Lingyue Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
124
|
Zhao YY, Lian JX, Lan Z, Zou KL, Wang WM, Yu GT. Ferroptosis promotes anti-tumor immune response by inducing immunogenic exposure in HNSCC. Oral Dis 2023; 29:933-941. [PMID: 34773344 DOI: 10.1111/odi.14077] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 11/03/2021] [Accepted: 11/07/2021] [Indexed: 01/04/2023]
Abstract
Accumulated evidence indicates that immune cell populations play pivotal roles in the process of tumor initiation, progression, recurrence, metastasis, and immune escape. Ferroptosis is a form of regulating cell death in the nexus between metabolism, redox biology, and human health. Ferroptosis is considered as a vital important event in HNSCC, but the underling mechanism of regulating immune cell populations remains poorly understood. Our tissue microarray study showed that patients with high expression of GPX4 were related to poor survival. Moreover, the expression of GPX4 has been negatively associated with immunogenic cell death-related protein calreticulin in HNSCC tissue cohort. Further, RSL3 was used to induce ferroptosis in HNSCC xenograft of C3H/He mouse. We found that the occurrence of ferroptosis had significantly reduced the number of myeloid-derived suppressor cells (MDSCs) and tumor-associated M2-like macrophages (M2 TAMs) in tumor microenvironment. Meanwhile, the tumor-infiltrating CD4+ and CD8+ T cells were increased. And the calreticulin and HMGB1 may be potential candidate proteins improving the immunosuppressive tumor microenvironment. Taken together, our project suggests that ferroptosis can promote anti-tumor immune response by reversing immunosuppressive microenvironment, indicating that ferroptosis inducer is a promising therapeutic strategy in HNSCC.
Collapse
Affiliation(s)
- Yu-Yue Zhao
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Jun-Xiang Lian
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Zhou Lan
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Ke-Long Zou
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Wei-Ming Wang
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital of Central South University, Changsha, China
| | - Guang-Tao Yu
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
125
|
Liu S, Liang H, Lv L, Hu F, Liu Q, Wang Y, Zhu J, Chen Z, Li J, Wang Z, Chang YN, Li J, Ma X, Chen K, Xing G. 3D culture boosting fullerenol nanoparticles to induce calreticulin exposure on MCF-7 cells for enhanced macrophage-mediated cell removal. Colloids Surf B Biointerfaces 2023; 224:113204. [PMID: 36801743 DOI: 10.1016/j.colsurfb.2023.113204] [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: 12/24/2022] [Revised: 02/05/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023]
Abstract
Calreticulin (CRT) on the cell surface that acts as an "eat me" signal is vital for macrophage-mediated programmed cell removal. The polyhydroxylated fullerenol nanoparticle (FNP) has appeared as an effective inducer to cause CRT exposure on cancer cell surface, but it failed in treating some cancer cells such as MCF-7 cells based on previous findings. Here, we carried out the 3D culture of MCF-7 cells, and interestingly found that the FNP induced CRT exposure on cells in 3D spheres via re-distributing CRT from endoplasmic reticulum (ER) to cell surface. Phagocytosis experiments in vitro and in vivo illustrated the combination of FNP and anti-CD47 monoclonal antibody (mAb) further enhanced macrophage-mediated phagocytosis to cancer cells. The maximal phagocytic index in vivo was about three times higher than that of the control group. Moreover, in vivo tumorigenesis experiments in mice proved that FNP could regulate the progress of MCF-7 cancer stem-like cells (CSCs). These findings expand the application of FNP in tumor therapy of anti-CD47 mAb and 3D culture can be used as a screening tool for nanomedicine.
Collapse
Affiliation(s)
- Sen Liu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China; College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China; Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China
| | - Haojun Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China
| | - Linwen Lv
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China
| | - Fan Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China
| | - Qiuyang Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China
| | - Yujiao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China
| | - Junyu Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China
| | - Ziteng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China
| | - Jiacheng Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China
| | - Zhijie Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China
| | - Ya-Nan Chang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China
| | - Juan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China
| | - Xiancai Ma
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou 510005, China.
| | - Kui Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China.
| | - Gengmei Xing
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Science (CAS), Beijing 100049, China.
| |
Collapse
|
126
|
Kyllesbech C, Trier N, Mughal F, Hansen P, Holmström M, El Fassi D, Hasselbalch H, Skov V, Kjær L, Andersen M, Ciplys E, Slibinskas R, Frederiksen J, Højrup P, Houen G. Antibodies to calnexin and mutated calreticulin are common in human sera. Curr Res Transl Med 2023; 71:103380. [PMID: 36738659 DOI: 10.1016/j.retram.2023.103380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/12/2023] [Accepted: 01/24/2023] [Indexed: 01/30/2023]
Abstract
PURPOSE OF THE STUDY Calreticulin is an endoplasmic reticulum chaperone protein, which is involved in protein folding and in peptide loading of major histocompatibility complex class I molecules together with its homolog calnexin. Mutated calreticulin is associated with a group of hemopoietic disorders, especially myeloproliferative neoplasms. Currently only the cellular immune response to mutated calreticulin has been described, although preliminary findings have indicated that antibodies to mutated calreticulin are not specific for myeloproliferative disorders. These findings have prompted us to characterize the humoral immune response to mutated calreticulin and its chaperone homologue calnexin. PATIENTS AND METHODS We analyzed sera from myeloproliferative neoplasm patients, healthy donors and relapsing-remitting multiple sclerosis patients for the occurrence of autoantibodies to wild type and mutated calreticulin forms and to calnexin by enzyme-linked immunosorbent assay. RESULTS Antibodies to mutated calreticulin and calnexin were present at similar levels in serum samples of myeloproliferative neoplasm and multiple sclerosis patients as well as healthy donors. Moreover, a high correlation between antibodies to mutated calreticulin and calnexin was seen for all patient and control groups. Epitope binding studies indicated that cross-reactive antibodies bound to a three-dimensional epitope encompassing a short linear sequence in the C-terminal of mutated calreticulin and calnexin. CONCLUSION Collectively, these findings indicate that calreticulin mutations may be common and not necessarily lead to onset of myeloproliferative neoplasm, possibly due to elimination of cells with mutations. This, in turn, may suggest that additional molecular changes may be required for development of myeloproliferative neoplasm.
Collapse
Affiliation(s)
- C Kyllesbech
- Department of Neurology, Valdemar Hansens vej 23, Rigshospitalet, Glostrup, Denmark; Institute of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M, Denmark
| | - N Trier
- Department of Neurology, Valdemar Hansens vej 23, Rigshospitalet, Glostrup, Denmark
| | - F Mughal
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen Ø, Denmark
| | - P Hansen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, Copenhagen Ø, Denmark
| | - M Holmström
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Borgmester Ib Juuls Vej 25C, Copenhagen University Hospital, Herlev, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, Copenhagen, Denmark
| | - D El Fassi
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, Copenhagen N, Denmark
| | - H Hasselbalch
- Department of Hematology, Zealand University Hospital Roskilde, Sygehusvej 10, Roskilde, Denmark
| | - V Skov
- Department of Hematology, Zealand University Hospital Roskilde, Sygehusvej 10, Roskilde, Denmark
| | - L Kjær
- Department of Hematology, Zealand University Hospital Roskilde, Sygehusvej 10, Roskilde, Denmark
| | - M Andersen
- Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, Copenhagen, Denmark
| | - E Ciplys
- Institute of Biotechnology, University of Vilnius, Sauletékio al. 7, Vilnius, Lithuania
| | - R Slibinskas
- Institute of Biotechnology, University of Vilnius, Sauletékio al. 7, Vilnius, Lithuania
| | - J Frederiksen
- Department of Neurology, Valdemar Hansens vej 23, Rigshospitalet, Glostrup, Denmark
| | - P Højrup
- Institute of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M, Denmark
| | - G Houen
- Department of Neurology, Valdemar Hansens vej 23, Rigshospitalet, Glostrup, Denmark; Institute of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M, Denmark.
| |
Collapse
|
127
|
Preet Kaur A, Alice A, Crittenden MR, Gough MJ. The role of dendritic cells in radiation-induced immune responses. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:61-104. [PMID: 37438021 DOI: 10.1016/bs.ircmb.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Dendritic cells perform critical functions in bridging innate and adaptive immunity. Their ability to sense adjuvant signals in their environment, migrate on maturation, and cross-present cell-associated antigens enables these cells to carry antigen from tissue sites to lymph nodes, and thereby prime naïve T cells that cannot enter tissues. Despite being an infrequent cell type in tumors, we discuss how dendritic cells impact the immune environment of tumors and their response to cancer therapies. We review how radiation therapy of tumors can impact dendritic cells, through transfer of cell associated antigens to dendritic cells and the release of endogenous adjuvants, resulting in increased antigen presentation in the tumor-draining lymph nodes. We explore how tumor specific factors can result in negative regulation of dendritic cell function in the tumor, and the impact of direct radiation exposure to dendritic cells in the treatment field. These data suggest an important role for dendritic cell subpopulations in activating new T cell responses and boosting existing T cell responses to tumor associated antigens in tumor draining lymph nodes following radiation therapy. It further justifies a focus on the needs of the lymph node T cells to improve systemic anti-immunity following radiation therapy.
Collapse
Affiliation(s)
- Aanchal Preet Kaur
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Alejandro Alice
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States; The Oregon Clinic, Portland, OR, United States
| | - Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States.
| |
Collapse
|
128
|
Li K, Huo Q, Dimmitt NH, Qu G, Bao J, Pandya PH, Saadatzadeh MR, Bijangi-Vishehsaraei K, Kacena MA, Pollok KE, Lin CC, Li BY, Yokota H. Osteosarcoma-enriched transcripts paradoxically generate osteosarcoma-suppressing extracellular proteins. eLife 2023; 12:e83768. [PMID: 36943734 PMCID: PMC10030111 DOI: 10.7554/elife.83768] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 03/01/2023] [Indexed: 03/23/2023] Open
Abstract
Osteosarcoma (OS) is the common primary bone cancer that affects mostly children and young adults. To augment the standard-of-care chemotherapy, we examined the possibility of protein-based therapy using mesenchymal stem cells (MSCs)-derived proteomes and OS-elevated proteins. While a conditioned medium (CM), collected from MSCs, did not present tumor-suppressing ability, the activation of PKA converted MSCs into induced tumor-suppressing cells (iTSCs). In a mouse model, the direct and hydrogel-assisted administration of CM inhibited tumor-induced bone destruction, and its effect was additive with cisplatin. CM was enriched with proteins such as calreticulin, which acted as an extracellular tumor suppressor by interacting with CD47. Notably, the level of CALR transcripts was elevated in OS tissues, together with other tumor-suppressing proteins, including histone H4, and PCOLCE. PCOLCE acted as an extracellular tumor-suppressing protein by interacting with amyloid precursor protein, a prognostic OS marker with poor survival. The results supported the possibility of employing a paradoxical strategy of utilizing OS transcriptomes for the treatment of OS.
Collapse
Affiliation(s)
- Kexin Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical UniversityHarbinChina
- Department of Biomedical Engineering, Indiana University Purdue University IndianapolisIndianapolisUnited States
| | - Qingji Huo
- Department of Pharmacology, School of Pharmacy, Harbin Medical UniversityHarbinChina
- Department of Biomedical Engineering, Indiana University Purdue University IndianapolisIndianapolisUnited States
| | - Nathan H Dimmitt
- Department of Biomedical Engineering, Indiana University Purdue University IndianapolisIndianapolisUnited States
| | - Guofan Qu
- Department of Orthopedic Surgery, Harbin Medical University Cancer HospitalHarbinChina
| | - Junjie Bao
- Department of Orthopedic Surgery, Harbin Medical University Cancer HospitalHarbinChina
| | - Pankita H Pandya
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of MedicineIndianapolisUnited States
- Department of Pediatrics, Indiana University School of MedicineIndianapolisUnited States
| | - M Reza Saadatzadeh
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of MedicineIndianapolisUnited States
- Department of Pediatrics, Indiana University School of MedicineIndianapolisUnited States
| | | | - Melissa A Kacena
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of MedicineIndianapolisUnited States
- Department of Orthopaedic Surgery, Indiana University School of MedicineIndianapolisUnited States
- Indiana Center for Musculoskeletal Health, Indiana University School of MedicineIndianapolisUnited States
| | - Karen E Pollok
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of MedicineIndianapolisUnited States
- Department of Pediatrics, Indiana University School of MedicineIndianapolisUnited States
| | - Chien-Chi Lin
- Department of Biomedical Engineering, Indiana University Purdue University IndianapolisIndianapolisUnited States
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of MedicineIndianapolisUnited States
| | - Bai-Yan Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical UniversityHarbinChina
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University IndianapolisIndianapolisUnited States
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of MedicineIndianapolisUnited States
- Indiana Center for Musculoskeletal Health, Indiana University School of MedicineIndianapolisUnited States
| |
Collapse
|
129
|
Jiménez-Fernández M, de la Fuente H, Martín P, Cibrián D, Sánchez-Madrid F. Unraveling CD69 signaling pathways, ligands and laterally associated molecules. EXCLI JOURNAL 2023; 22:334-351. [PMID: 37223078 PMCID: PMC10201016 DOI: 10.17179/excli2022-5751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/09/2023] [Indexed: 05/25/2023]
Abstract
CD69 is an early leukocyte activation marker involved in the regulation of the immune response. Initial in vitro studies evaluated its function using monoclonal antibodies until knock-out mice were developed. Subsequently, four ligands for CD69 have been identified, namely galectin-1, S100A8/S100A9 complex, myosin light chains 9 and 12, and oxidized low-density lipoproteins. In addition, several molecules are laterally associated with and regulated by CD69, including calreticulin and two transmembrane receptors, sphingosine-1-phosphate receptor (S1P1) and the heterodimeric amino acid transporter complex SLC7A5-SLC3A2 (LAT1-CD98). Recently, CD69 engagement has been shown to induce the expression of the immunoregulatory receptor programmed cell death-1 (PD-1) in T cells. The molecular signaling induced by CD69 has been explored in different scenarios and cell types. This review provides a perspective on the molecular pathways, ligands and cellular functions known to be regulated by CD69.
Collapse
Affiliation(s)
- María Jiménez-Fernández
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), 28006 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 29029 Madrid, Spain
| | - Hortensia de la Fuente
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), 28006 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 29029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pilar Martín
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 29029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Danay Cibrián
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), 28006 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 29029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco Sánchez-Madrid
- Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), 28006 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 29029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
130
|
Structural and Dynamic Differences between Calreticulin Mutants Associated with Essential Thrombocythemia. Biomolecules 2023; 13:biom13030509. [PMID: 36979444 PMCID: PMC10046389 DOI: 10.3390/biom13030509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/12/2023] Open
Abstract
Essential thrombocythemia (ET) is a blood cancer. ET is characterized by an overproduction of platelets that can lead to thrombosis formation. Platelet overproduction occurs in megakaryocytes through a signaling pathway that could involve JAK2, MPL, or CALR proteins. CALR mutations are associated with 25–30% of ET patients; CALR variants must be dimerized to induce ET. We classified these variants into five classes named A to E; classes A and B are the most frequent classes in patients with ET. The dynamic properties of these five classes using structural models of CALR’s C-domain were analyzed using molecular dynamics simulations. Classes A, B, and C are associated with frameshifts in the C-domain. Their dimers can be stable only if a disulfide bond is formed; otherwise, the two monomers repulse each other. Classes D and E cannot be stable as dimers due to the absence of disulfide bonds. Class E and wild-type CALR have similar dynamic properties. These results suggest that the disulfide bond newly formed in classes A, B, and C may be essential for the pathogenicity of these variants. They also underline that class E cannot be directly related to ET but corresponds to human polymorphisms.
Collapse
|
131
|
Li W, Jiang Y, Lu J. Nanotechnology-enabled immunogenic cell death for improved cancer immunotherapy. Int J Pharm 2023; 634:122655. [PMID: 36720448 PMCID: PMC9975075 DOI: 10.1016/j.ijpharm.2023.122655] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023]
Abstract
Tumor immunotherapy has revolutionized the field of oncology treatments in recent years. As one of the promising strategies of cancer immunotherapy, tumor immunogenic cell death (ICD) has shown significant potential for tumor therapy. Nanoparticles are widely used for drug delivery due to their versatile characteristics, such as stability, slow blood elimination, and tumor-targeting ability. To increase the specificity of ICD inducers and improve the efficiency of ICD induction, functionally specific nanoparticles, such as liposomes, nanostructured lipid carriers, micelles, nanodiscs, biomembrane-coated nanoparticles and inorganic nanoparticles have been widely reported as the vehicles to deliver ICD inducers in vivo. In this review, we summarized the strategies of different nanoparticles for ICD-induced cancer immunotherapy, and systematically discussed their advantages and disadvantages as well as provided feasible strategies for solving these problems. We believe that this review will offer some insights into the design of effective nanoparticulate systems for the therapeutic delivery of ICD inducers, thus, promoting the development of ICD-mediated cancer immunotherapy.
Collapse
Affiliation(s)
- Wenpan Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Yanhao Jiang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States; NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, AZ 85721, United States; BIO5 Institute, The University of Arizona, Tucson, AZ 85721, United States; Southwest Environmental Health Sciences Center, The University of Arizona, Tucson 85721, United States.
| |
Collapse
|
132
|
Galluzzi L, Kepp O, Hett E, Kroemer G, Marincola FM. Immunogenic cell death in cancer: concept and therapeutic implications. J Transl Med 2023; 21:162. [PMID: 36864446 PMCID: PMC9979428 DOI: 10.1186/s12967-023-04017-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/08/2023] [Indexed: 03/04/2023] Open
Abstract
Mammalian cells responding to specific perturbations of homeostasis can undergo a regulated variant of cell death that elicits adaptive immune responses. As immunogenic cell death (ICD) can only occur in a precise cellular and organismal context, it should be conceptually differentiated from instances of immunostimulation or inflammatory responses that do not mechanistically depend on cellular demise. Here, we critically discuss key conceptual and mechanistic aspects of ICD and its implications for cancer (immuno)therapy.
Collapse
Affiliation(s)
- 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.
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Erik Hett
- Sonata Therapeutics, Boston, MA, USA
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.,Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France.,Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | | |
Collapse
|
133
|
Marshall K, Twum Y, Gao W. Proteome derangement in malignant epithelial cells and its stroma following exposure to 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Arch Toxicol 2023; 97:711-720. [PMID: 36434399 PMCID: PMC10071504 DOI: 10.1007/s00204-022-03426-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
Discovering novel changes in the proteome of malignant lung epithelial cells and/or the tumor-microenvironment is paramount for diagnostic, prognostic, and/or therapy development. A time-dependent 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced mouse lung tumor model was used to screen the proteome of lung tumors. NNK-transformed human lung epithelial BEAS-2B cells were then established to evaluate the epithelial cell-specific protein changes. A duration-dependent increase of tumor burden was observed in NNK-treated mice, 2/12 (17%), 8/12 (67%), 9/12 (75%), and 10/10 (100%) at weeks 8, 12, 16, and 20 after the NNK exposure, respectively. A total of 25 differentially expressed proteins (≥ twofold change), predominantly structural, signaling, and metabolic proteins, were detected by two-dimensional difference gel electrophoresis and identified by mass spectrometry. Calregulin, ezrin, histamine releasing factor (HRF), and inorganic pyrophosphatase 1 (PPA1) exhibited changes and were further confirmed via immunoblotting. In addition, immunohistochemistry (IHC) analysis indicated upregulated E-cadherin and decreased vimentin expression in epithelial cells of tumor tissues. Acquisition of a neoplastic phenotype in NNK-transformed BEAS-2B cells was demonstrated by enhanced wound closure and increased anchorage independent colony formation. In transformed BEAS-2B cells, protein expression of E-cadherin, ezrin, and PPA1 (but not calregulin and HRF) was upregulated, as was observed in tumor tissues IHC staining using mouse lung tumor tissues further revealed that HRF upregulation was not lung epithelial cell specific. Altogether, tumorigenesis after NNK exposure may be initiated by protein dysregulation in lung epithelial cells together with proteome derangement derived from other cell types existing in the tumor-microenvironment.
Collapse
Affiliation(s)
- Kent Marshall
- Department of Occupational and Environmental Health Sciences, West Virginia University, School of Public Health, 64 Medical Center Drive, Morgantown, WV, 26506, USA
- West Virginia University, School of Medicine, 1 Medical Center Drive, Morgantown, WV, 26505, USA
- West Virginia Clinical and Translational Science Institute, Morgantown, WV, USA
| | - Yaw Twum
- Department of Occupational and Environmental Health Sciences, West Virginia University, School of Public Health, 64 Medical Center Drive, Morgantown, WV, 26506, USA
| | - Weimin Gao
- Department of Occupational and Environmental Health Sciences, West Virginia University, School of Public Health, 64 Medical Center Drive, Morgantown, WV, 26506, USA.
- West Virginia Clinical and Translational Science Institute, Morgantown, WV, USA.
| |
Collapse
|
134
|
Karami Fath M, Garousi S, Mottahedi M, Ghasemzadeh N, Salmani K, Olfati F, Beit Saeed M, Sotoudeh S, Barati G. The role of hypoxia-inducible factors in breast cancer stem cell specification. Pathol Res Pract 2023; 243:154349. [PMID: 36791562 DOI: 10.1016/j.prp.2023.154349] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023]
Abstract
Breast tumor is heterogeneous cancer with high morbidity and mortality rates, particularly in developing countries. Despite new efforts to reduce the breast cancer implications, the number of newly diagnosed cases is increasing worldwide. It is believed that cancer stem cells (CSCs) are responsible for the implication of cancers including breast cancer. Although CSCs compose a small population in tumor bulks, they play a crucial role in tumor initiation, progression, metastasis, and chemotherapeutic resistance. These events are mediated by the hypoxia-inducible factor (HIF) pathway which regulates the transcription of genes involved in CSC maintenance and tumorigenesis. In this review, we discussed the mechanisms by which hypoxia- or chemotherapy-induced HIFs promote breast CSC specification.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Setareh Garousi
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehran Mottahedi
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Kiana Salmani
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Olfati
- Department of Reproductive Health, Faculty of Nursing and Midwifery, Tehran University of Medical Sciences, Tehran, Iran
| | - Miad Beit Saeed
- Faculty of Nursing and Midwifery, Abadan Islamic Azad University, Abadan, Iran
| | - Sina Sotoudeh
- Faculty of Nursing and Midwifery, Guilan University of Medical Sciences, Guilan, Iran
| | | |
Collapse
|
135
|
Su W, Qiu W, Li SJ, Wang S, Xie J, Yang QC, Xu J, Zhang J, Xu Z, Sun ZJ. A Dual-Responsive STAT3 Inhibitor Nanoprodrug Combined with Oncolytic Virus Elicits Synergistic Antitumor Immune Responses by Igniting Pyroptosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209379. [PMID: 36545949 DOI: 10.1002/adma.202209379] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/14/2022] [Indexed: 06/17/2023]
Abstract
Immune checkpoint blockade (ICB) therapy shows excellent efficacy against malignancies; however, insufficient tumor immunogenicity and the immunosuppressive tumor microenvironment (TME) are considered as the two major stumbling blocks to a broad ICB response. Here, a combinational therapeutic strategy is reported, wherein TME-reactive oxygen species/pH dual-responsive signal transducers and activators of transcription 3 inhibitor nanoprodrugs MPNPs are combined with oncolytic herpes simplex virus 1 virotherapy to synergistically ignite pyroptosis for enhancing immunotherapy. MPNPs exhibit a certain level of tumor accumulation, reduce tumor cell stemness, and enhance antitumor immune responses. Furthermore, the simultaneous application of oncolytic viruses (OVs) confers MPNPs with higher tumor penetration capacity and remarkable gasdermin-E-mediated pyroptosis, thereby reshaping the TME and transforming "cold" tumors into "hot" ones. This "fire of immunity" strategy successfully activates robust T-cell-dependent antitumor responses, potentiating ICB effects against local recurrence and pulmonary metastasis in preclinical "cold" murine triple-negative breast cancer and syngeneic oral cancer models. Collectively, this work may pave a new way and offer an unprecedented opportunity for the combination of OVs with nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Wen Su
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Wei Qiu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Shu-Jin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Shuo Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Jun Xie
- State Key Laboratory of Virology, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430079, P. R. China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, P. R. China
| | - Qi-Chao Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Jiming Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Junjie Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
- State Key Laboratory of Virology, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430079, P. R. China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, P. R. China
| | - Zhigang Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy & Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| |
Collapse
|
136
|
Passively-targeted mitochondrial tungsten-based nanodots for efficient acute kidney injury treatment. Bioact Mater 2023; 21:381-393. [PMID: 36185743 PMCID: PMC9483742 DOI: 10.1016/j.bioactmat.2022.08.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/05/2022] [Accepted: 08/20/2022] [Indexed: 12/25/2022] Open
Abstract
Acute kidney injury (AKI) can lead to loss of kidney function and a substantial increase in mortality. The burst of reactive oxygen species (ROS) plays a key role in the pathological progression of AKI. Mitochondrial-targeted antioxidant therapy is very promising because mitochondria are the main source of ROS in AKI. Antioxidant nanodrugs with actively targeted mitochondria have achieved encouraging success in many oxidative stress-induced diseases. However, most strategies to actively target mitochondria make the size of nanodrugs too large to pass through the glomerular system to reach the renal tubules, the main damage site of AKI. Here, an ultra-small Tungsten-based nanodots (TWNDs) with strong ROS scavenging can be very effective for treatment of AKI. TWNDs can reach the tubular site after crossing the glomerular barrier, and enter the mitochondria of the renal tubule without resorting to complex active targeting strategies. To our knowledge, this is the first time that ultra-small negatively charged nanodots can be used to passively target mitochondrial therapy for AKI. Through in-depth study of the therapeutic mechanism, such passive mitochondria-targeted TWNDs are highly effective in protecting mitochondria by reducing mitochondrial ROS and increasing mitophagy. In addition, TWNDs can also reduce the infiltration of inflammatory cells. This work provides a new way to passively target mitochondria for AKI, and give inspiration for the treatment of many major diseases closely related to mitochondria, such as myocardial infarction and cerebral infarction. Mitochondrial-targeted antioxidant therapy is a very promising treatment for AKI. TWNDs have a strong ability to scavenge ROS, and their small size allows them to cross the glomerular filtration barrier. TWNDs passively target the mitochondria of renal tubular cells and are highly effective in restoring renal function.
Collapse
|
137
|
Sun Z, Zhao M, Wang W, Hong L, Wu Z, Luo G, Lu S, Tang Y, Li J, Wang J, Zhang Y, Zhang L. 5-ALA mediated photodynamic therapy with combined treatment improves anti-tumor efficacy of immunotherapy through boosting immunogenic cell death. Cancer Lett 2023; 554:216032. [PMID: 36493899 DOI: 10.1016/j.canlet.2022.216032] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/04/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
Photodynamic therapy (PDT) is clinically promising in destructing primary tumors and immunotherapy awakes host immunity to control distant metastases. 5-aminolevulinic acid (5-ALA), a smart photosensitizer, converts into a physiological PDT agent with no dark toxicity in vivo. In this study, we found for the first time 5-ALA-PDT induced colorectal cancer (CRC) cells death by immunogenic cell death (ICD) upon AKT inhibition. Dying cancer cells induced by 5-ALA-PDT efficiently activated bone-marrow derived dendritic cells (BMDCs). Simultaneously, autophagy was observed after AKT inhibition by 5-ALA-PDT. Besides, we found cells died more remarkable by ICD under a circumstance of low occurrence of autophagy. To evaluate the effects of 5-ALA-PDT in vivo, we applied subcutaneous tumor mouse model and delightedly found 5-ALA-PDT induced a systemic antitumor immune response to control both primary tumors and distant metastases. Meanwhile, 5-ALA-PDT enhanced Th1 immunity, leading cytotoxic T lymphocyte response, and raised tumor-specific T cells. Combining with Chloroquine (CQ), 5-ALA-PDT further augmented tumor-specific immunity effects indicating protective role of autophagy. Together, the combination therapy of 5-ALA-PDT and autophagy inhibitor synergistically led to a novel clinical approach and potential ICD-based tumor vaccine for CRC patients.
Collapse
Affiliation(s)
- Zhuoran Sun
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, 410013, PR China; Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China
| | - Weibi Wang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, 410013, PR China
| | - Lanhui Hong
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, 410013, PR China
| | - Zhongguang Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, 410013, PR China
| | - Guang Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, 410013, PR China
| | - Siyao Lu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, 410013, PR China
| | - Yueyue Tang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, 410013, PR China
| | - Jiehan Li
- School of Biomedical Sciences, Hunan University, Changsha, 410082, PR China
| | - Jiangang Wang
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China
| | - Yingjie Zhang
- School of Biomedical Sciences, Hunan University, Changsha, 410082, PR China.
| | - Lingling Zhang
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, 410013, PR China.
| |
Collapse
|
138
|
Assessment of immunological memory formation in vivo. Methods Cell Biol 2023; 174:65-73. [PMID: 36710052 DOI: 10.1016/bs.mcb.2021.12.029] [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: 02/01/2023]
Abstract
The reinstatement of cancer immunosurveillance during cancer therapy is the sole means of achieving long term success with durable disease control and sometimes even cure. Induction of immunogenic cell death (ICD) by pharmacological or physical interventions such as anthracycline-based chemotherapy or ionizing irradiation, respectively, is a potent strategy for triggering immunological memory in immunocompetent mice. Thus, mice that were cured from syngeneic transplanted cancers were able to reject the same tumor cells several weeks after the eradication of the initial tumor, contrasting with the fact that antigenically distinct cells readily formed tumors. Here, we show how to harness sequential injections of antigenically identical or distinct cancer cells in immunocompetent animals to evaluate the generation of immunological memory.
Collapse
|
139
|
Liu XZ, Wen ZJ, Li YM, Sun WR, Hu XQ, Zhu JZ, Li XY, Wang PY, Pedraz JL, Lee JH, Kim HW, Ramalingam M, Xie S, Wang R. Bioengineered Bacterial Membrane Vesicles with Multifunctional Nanoparticles as a Versatile Platform for Cancer Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:3744-3759. [PMID: 36630299 DOI: 10.1021/acsami.2c18244] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Inducing immunogenic cell death (ICD) is a critical strategy for enhancing cancer immunotherapy. However, inefficient and risky ICD inducers along with a tumor hypoxia microenvironment seriously limit the immunotherapy efficacy. Non-specific delivery is also responsible for this inefficiency. In this work, we report a drug-free bacteria-derived outer membrane vesicle (OMV)-functionalized Fe3O4-MnO2 (FMO) nanoplatform that realized neutrophil-mediated targeted delivery and photothermally enhanced cancer immunotherapy. In this system, modification of OMVs derived from Escherichia coli enhanced the accumulation of FMO NPs at the tumor tissue through neutrophil-mediated targeted delivery. The FMO NPs underwent reactive decomposition in the tumor site, generating manganese and iron ions that induced ICD and O2 that regulated the tumor hypoxia environment. Moreover, OMVs are rich in pathogen-associated pattern molecules that can overcome the tumor immunosuppressive microenvironment and effectively activate immune cells, thereby enhancing specific immune responses. Photothermal therapy (PTT) caused by MnO2 and Fe3O4 can not only indirectly stimulate systemic immunity by directly destroying tumor cells but also promote the enrichment of neutrophil-equipped nanoparticles by enhancing the inflammatory response at the tumor site. Finally, the proposed multi-modal treatment system with targeted delivery capability realized effective tumor immunotherapy to prevent tumor growth and recurrence.
Collapse
Affiliation(s)
- Xin Zheng Liu
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
- Shandong Laboratory of Advanced Materials and Green Manufacturing, Yantai264000, People's Republic of China
| | - Zhi Juan Wen
- Binzhou Medical University Hospital, Binzhou256603PR China
| | - Yun Meng Li
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
| | - Wan Ru Sun
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
| | - Xiao Qian Hu
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
| | - Jia Zhi Zhu
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
| | - Xin Yu Li
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
| | - Ping Yu Wang
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai264003, People's Republic of China
| | - José Luis Pedraz
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006Vitoria-Gasteiz, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, 28029Madrid, Spain
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan31116, Republic of Korea
- Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan31116, Republic of Korea
- Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan31116, Republic of Korea
| | - Murugan Ramalingam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan31116, Republic of Korea
- Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan31116, Republic of Korea
- School of Basic Medical Sciences, Chengdu University, Chengdu610106, People's Republic of China
- Department of Metallurgical and Materials Engineering, Atilim University, Ankara06830, Turkey
| | - Shuyang Xie
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai264003, People's Republic of China
| | - Ranran Wang
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
- Shandong Laboratory of Advanced Materials and Green Manufacturing, Yantai264000, People's Republic of China
| |
Collapse
|
140
|
Li K, Huo Q, Li BY, Yokota H. The Double-Edged Proteins in Cancer Proteomes and the Generation of Induced Tumor-Suppressing Cells (iTSCs). Proteomes 2023; 11:5. [PMID: 36810561 PMCID: PMC9944087 DOI: 10.3390/proteomes11010005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Unlike a prevalent expectation that tumor cells secrete tumor-promoting proteins and stimulate the progression of neighboring tumor cells, accumulating evidence indicates that the role of tumor-secreted proteins is double-edged and context-dependent. Some of the oncogenic proteins in the cytoplasm and cell membranes, which are considered to promote the proliferation and migration of tumor cells, may inversely act as tumor-suppressing proteins in the extracellular domain. Furthermore, the action of tumor-secreted proteins by aggressive "super-fit" tumor cells can be different from those derived from "less-fit" tumor cells. Tumor cells that are exposed to chemotherapeutic agents could alter their secretory proteomes. Super-fit tumor cells tend to secrete tumor-suppressing proteins, while less-fit or chemotherapeutic agent-treated tumor cells may secrete tumor-promotive proteomes. Interestingly, proteomes derived from nontumor cells such as mesenchymal stem cells and peripheral blood mononuclear cells mostly share common features with tumor cell-derived proteomes in response to certain signals. This review introduces the double-sided functions of tumor-secreted proteins and describes the proposed underlying mechanism, which would possibly be based on cell competition.
Collapse
Affiliation(s)
- Kexin Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Qingji Huo
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Bai-Yan Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| |
Collapse
|
141
|
Mosale Seetharam S, Liu Y, Wu J, Fechter L, Murugesan K, Maecker H, Gotlib J, Zehnder J, Paulmurugan R, Krishnan A. Enkurin: A novel marker for myeloproliferative neoplasms from platelet, megakaryocyte, and whole blood specimens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.07.523111. [PMID: 36712071 PMCID: PMC9881897 DOI: 10.1101/2023.01.07.523111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Impaired protein homeostasis, though well established in age-related disorders, has been linked in recent research with the pathogenesis of myeloproliferative neoplasms (MPNs). As yet, however, little is known about MPN-specific modulators of proteostasis, thus impeding our ability for increased mechanistic understanding and discovery of additional therapeutic targets. Loss of proteostasis, in itself, is traced to dysregulated mechanisms in protein folding and intracellular calcium signaling at the endoplasmic reticulum (ER). Here, using ex vivo and in vitro systems (including CD34 + cultures from patient bone marrow, and healthy cord/peripheral blood specimens), we extend our prior data from MPN patient platelet RNA sequencing, and discover select proteostasis-associated markers at RNA and/or protein levels in each of platelets, parent megakaryocytes, and whole blood specimens. Importantly, we identify a novel role in MPNs for enkurin ( ENKUR ), a calcium mediator protein, implicated originally only in spermatogenesis. Our data reveal consistent ENKUR downregulation at both RNA and protein levels across MPN patient specimens and experimental models, with a concomitant upregulation of a cell cycle marker, CDC20 . Silencing of ENKUR by shRNA in CD34 + derived megakaryocytes further confirm this association with CDC20 at both RNA and protein levels; and indicate a likely role for the PI3K/Akt pathway. The inverse association of ENKUR and CDC20 expression was further confirmed upon treatment with thapsigargin (an agent that causes protein misfolding in the ER by selective loss of calcium) in both megakaryocyte and platelet fractions at RNA and protein levels. Together, our work sheds light on enkurin as a novel marker of MPN pathogenesis beyond the genetic alterations; and indicates further mechanistic investigation into a role for dysregulated calcium homeostasis, and ER and protein folding stress in MPN transformation. VISUAL ABSTRACT Key Points Enkurin, a calcium adaptor protein, is identified as a novel marker of pathogenesis in MPNs.MPN megakaryocyte and platelet expression of enkurin at RNA and protein levels is inversely associated with a cell differentiation cycle gene, CDC20.Likely role for dysregulated calcium homeostasis, and ER and protein folding stress in MPN transformation.
Collapse
Affiliation(s)
| | - Yi Liu
- Department of Radiology, Stanford University, Stanford, CA
| | - Jason Wu
- High-Throughput Bioscience Center (HTBC), Stanford University School of Medicine, Stanford, CA
| | - Lenn Fechter
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | | | - Holden Maecker
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA
| | - Jason Gotlib
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - James Zehnder
- Department of Pathology, Stanford University, Stanford, CA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Anandi Krishnan
- Department of Pathology, Stanford University, Stanford, CA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
142
|
Xian S, Chen L, Yan Y, Chen J, Yu G, Shao Y, Zhan B, Wang Y, Zhao L. Echinococcus multilocularis Calreticulin Interferes with C1q-Mediated Complement Activation. Trop Med Infect Dis 2023; 8:tropicalmed8010047. [PMID: 36668954 PMCID: PMC9864966 DOI: 10.3390/tropicalmed8010047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
As a zoonotic disease caused by Echinococcus multilocularis larvae, alveolar echinococcosis (AE) is one of the most severe forms of parasitic infection. Over a long evolutional process E. multilocularis has developed complex strategies to escape host immune attack and survive within a host. However, the mechanisms underlying immune evasion remain unclear. Here we investigated the binding activity of E. multilocularis calreticulin (EmCRT), a highly conserved Ca2+-binding protein, to human complement C1q and its ability to inhibit classical complement activation. ELISA, Far Western blotting and immunoprecipitation results demonstrated that both recombinant and natural EmCRTs bound to human C1q, and the interaction of recombinant EmCRT (rEmCRT) inhibited C1q binding to IgM. Consequently, rEmCRT inhibited classical complement activation manifested as decreasing C4/C3 depositions and antibody-sensitized cell lysis. Moreover, rEmCRT binding to C1q suppressed C1q binding to human mast cell, HMC-1, resulting in reduced C1q-induced mast cell chemotaxis. According to these results, E. multilocularis expresses EmCRT to interfere with C1q-mediated complement activation and C1q-dependent non-complement activation of immune cells, possibly as an immune evasion strategy of the parasite in the host.
Collapse
Affiliation(s)
- Siqi Xian
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, Baotou Medical College, Baotou 014040, China
| | - Lujuan Chen
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, Baotou Medical College, Baotou 014040, China
| | - Yan Yan
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, Baotou Medical College, Baotou 014040, China
| | - Jianfang Chen
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, Baotou Medical College, Baotou 014040, China
| | - Guixia Yu
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, Baotou Medical College, Baotou 014040, China
| | - Yuxiao Shao
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, Baotou Medical College, Baotou 014040, China
| | - Bin Zhan
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yanhai Wang
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- Correspondence: (Y.W.); (L.Z.)
| | - Limei Zhao
- Department of Pathogenic Biology, School of Basic Medical Sciences and Forensic Medicine, Baotou Medical College, Baotou 014040, China
- Correspondence: (Y.W.); (L.Z.)
| |
Collapse
|
143
|
Kuang Y, Jiang B, Zhu H, Zhou Y, Huang H, Li C, Zhang W, Li X, Cao Y. Classification related to immunogenic cell death predicts prognosis, immune microenvironment characteristics, and response to immunotherapy in lower-grade gliomas. Front Immunol 2023; 14:1102094. [PMID: 37153540 PMCID: PMC10154552 DOI: 10.3389/fimmu.2023.1102094] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/04/2023] [Indexed: 05/09/2023] Open
Abstract
Background Immunogenic cell death (ICD) is a form of cell death that elicits immune responses against the antigens found in dead or dying tumor cells. Growing evidence implies that ICD plays a significant role in triggering antitumor immunity. The prognosis for glioma remains poor despite many biomarkers being reported, and identifying ICD-related biomarkers is imminent for better-personalized management in patients with lower-grade glioma (LGG). Materials and methods We identified ICD-related differentially expressed genes (DEGs) by comparing gene expression profiles obtained across Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) cohorts. On the foundation of ICD-related DEGs, two ICD-related clusters were identified through consensus clustering. Then, survival analysis, functional enrichment analysis, somatic mutation analysis, and immune characteristics analysis were performed in the two ICD-related subtypes. Additionally, we developed and validated a risk assessment signature for LGG patients. Finally, we selected one gene (EIF2AK3) from the above risk model for experimental validation. Results 32 ICD-related DEGs were screened, dividing the LGG samples from the TCGA database into two distinct subtypes. The ICD-high subgroup showed worse overall survival (OS), greater immune infiltration, more active immune response process, and higher expression levels of HLA genes than the ICD-low subgroup. Additionally, nine ICD-related DEGs were identified to build the prognostic signature, which was highly correlated with the tumor-immune microenvironment and could unambiguously be taken as an independent prognostic factor and further verified in an external dataset. The experimental results indicated that EIF2AK3 expression was higher in tumors than paracancerous tissues, and high-expression EIF2AK3 was enriched in WHO III and IV gliomas by qPCR and IHC, and Knockdown of EIF2AK3 suppressed cell viability and mobility in glioma cells. Conclusion We established novel ICD-related subtypes and risk signature for LGG, which may be beneficial to improving clinical outcome prediction and guiding individualized immunotherapy.
Collapse
Affiliation(s)
- Yirui Kuang
- Department of Neurosurgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bincan Jiang
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hecheng Zhu
- Department of Oncology Radiology, Changsha Kexin Cancer Hospital, Changsha, Hunan, China
| | - Yi Zhou
- Department of Neurosurgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haoxuan Huang
- Department of Neurosurgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Can Li
- Department of Neurosurgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenlong Zhang
- Department of Neurosurgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuewen Li
- Department of Oncology Radiology, Changsha Kexin Cancer Hospital, Changsha, Hunan, China
| | - Yudong Cao
- Department of Neurosurgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Yudong Cao,
| |
Collapse
|
144
|
Zhai J, Gu X, Liu Y, Hu Y, Jiang Y, Zhang Z. Chemotherapeutic and targeted drugs-induced immunogenic cell death in cancer models and antitumor therapy: An update review. Front Pharmacol 2023; 14:1152934. [PMID: 37153795 PMCID: PMC10160433 DOI: 10.3389/fphar.2023.1152934] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/04/2023] [Indexed: 05/10/2023] Open
Abstract
As traditional strategies for cancer treatment, some chemotherapy agents, such as doxorubicin, oxaliplatin, cyclophosphamide, bortezomib, and paclitaxel exert their anti-tumor effects by inducing immunogenic cell death (ICD) of tumor cells. ICD induces anti-tumor immunity through release of, or exposure to, damage-related molecular patterns (DAMPs), including high mobility group box 1 (HMGB1), calreticulin, adenosine triphosphate, and heat shock proteins. This leads to activation of tumor-specific immune responses, which can act in combination with the direct killing functions of chemotherapy drugs on cancer cells to further improve their curative effects. In this review, we highlight the molecular mechanisms underlying ICD, including those of several chemotherapeutic drugs in inducing DAMPs exposed during ICD to activate the immune system, as well as discussing the prospects for application and potential role of ICD in cancer immunotherapy, with the aim of providing valuable inspiration for future development of chemoimmunotherapy.
Collapse
|
145
|
Yang Y, Huang J, Liu M, Qiu Y, Chen Q, Zhao T, Xiao Z, Yang Y, Jiang Y, Huang Q, Ai K. Emerging Sonodynamic Therapy-Based Nanomedicines for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204365. [PMID: 36437106 PMCID: PMC9839863 DOI: 10.1002/advs.202204365] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/25/2022] [Indexed: 05/08/2023]
Abstract
Cancer immunotherapy effect can be greatly enhanced by other methods to induce immunogenic cell death (ICD), which has profoundly affected immunotherapy as a highly efficient paradigm. However, these treatments have significant limitations, either by causing damage of the immune system or limited to superficial tumors. Sonodynamic therapy (SDT) can induce ICD to promote immunotherapy without affecting the immune system because of its excellent spatiotemporal selectivity and low side effects. Nevertheless, SDT is still limited by low reactive oxygen species yield and the complex tumor microenvironment. Recently, some emerging SDT-based nanomedicines have made numerous attractive and encouraging achievements in the field of cancer immunotherapy due to high immunotherapeutic efficiency. However, this cross-cutting field of research is still far from being widely explored due to huge professional barriers. Herein, the characteristics of the tumor immune microenvironment and the mechanisms of ICD are firstly systematically summarized. Subsequently, the therapeutic mechanism of SDT is fully summarized, and the advantages and limitations of SDT are discussed. The representative advances of SDT-based nanomedicines for cancer immunotherapy are further highlighted. Finally, the application prospects and challenges of SDT-based immunotherapy in future clinical translation are discussed.
Collapse
Affiliation(s)
- Yunrong Yang
- Department of PharmacyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jia Huang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Min Liu
- Department of PharmacyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Yige Qiu
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Tianjiao Zhao
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Yuqi Yang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Yitian Jiang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| | - Qiong Huang
- Department of PharmacyXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Kelong Ai
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410078P. R. China
| |
Collapse
|
146
|
Aydın EB, Aydın M, Sezgintürk MK. A Simple and Low-Cost Electrochemical Immunosensor for Ultrasensitive Determination of Calreticulin Biomarker in Human Serum. Macromol Biosci 2023; 23:e2200390. [PMID: 36419333 DOI: 10.1002/mabi.202200390] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/13/2022] [Indexed: 11/27/2022]
Abstract
An early on time detection of breast cancer significantly affects the treatment process and outcome. Herein, a new label-free impedimetric biosensor is developed to determine the lowest change in the level of calreticulin (CALR), which is a new biomarker of breast carcinoma. The proposed immunosensor is fabricated by using reduced graphene oxide/amino substituted polypyrrole polymer (rGO-PPyNH2 ) nanocomposite modified disposable electrode. The anti-CALR antibodies are first attached on the rGO-PPyNH2 nanocomposite coated electrode through glutaraldehyde crosslinking; the CALR antigens are then immobilized with the addition of CALR antigens to form an immunocomplex on the sensing surface. This immunocomplex induces considerably larger interfacial electron transport resistance (Rct ). The variation in the Rct has a linear relationship with CALR level in the detection range of 0.025 to 75 pg mL-1 , with a detection limit of 10.4 fg mL-1 . The suggested biosensor shows high selectivity to CALR, good storage stability (at least 5 weeks) and suitable reproducibility results as shown in quality control chart. The designed immunosensor is utilized to analyze CALR levels in human sera with satisfying results. This immunosensor provides a novel way for the clinical determination of CALR and other cancer biological markers.
Collapse
Affiliation(s)
- Elif Burcu Aydın
- Tekirdağ Namık Kemal University, Scientific and Technological Research Center, Tekirdağ, 59000, Turkey
| | - Muhammet Aydın
- Tekirdağ Namık Kemal University, Scientific and Technological Research Center, Tekirdağ, 59000, Turkey
| | - Mustafa Kemal Sezgintürk
- Çanakkale Onsekiz Mart University, Faculty of Engineering, Bioengineering Department, Çanakkale, 17000, Turkey
| |
Collapse
|
147
|
Liu Z, Wan R, Bai H, Wang J. Damage-associated molecular patterns and sensing receptors based molecular subtypes in malignant pleural mesothelioma and implications for immunotherapy. Front Immunol 2023; 14:1104560. [PMID: 37033966 PMCID: PMC10079989 DOI: 10.3389/fimmu.2023.1104560] [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: 11/21/2022] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Objectives Malignant pleural mesothelioma (MPM) is characterized as an incredibly aggressive form of cancer with a dismal diagnosis and a dearth of specific biomarkers and therapeutic options. For MPM patients, the effectiveness of immunotherapy may be influenced by damage-associated molecular pattern (DAMP)-induced immunogenic cell death (ICD).The objective of this work is to create a molecular profile associated with DAMPs to categorize MPM patients and predict their prognosis and response to immunotherapy. Methods The RNA-seq of 397 patients (263 patients with clinical data, 57.2% male, 73.0% over 60 yrs.) were gathered from eight public datasets as a training cohort to identify the DAMPs-associated subgroups of MPMs using K-means analysis. Three validation cohorts of patients or murine were established from TCGA and GEO databases. Comparisons were made across each subtype's immune status, gene mutations, survival prognosis, and predicted response to therapy. Results Based on the DAMPs gene expression, MPMs were categorized into two subtypes: the nuclear DAMPs subtype, which is classified by the upregulation of immune-suppressed pathways, and the inflammatory DAMPs subtype, which is distinguished by the enrichment of proinflammatory cytokine signaling. The inflammatory DAMPs subgroup had a better prognosis, while the nuclear DAMPs subgroup exhibited a worse outcome. In validation cohorts, the subtyping system was effectively verified. We further identified the genetic differences between the two DAMPs subtypes. It was projected that the inflammatory DAMPs subtype will respond to immunotherapy more favorably, suggesting that the developed clustering method may be implemented to predict the effectiveness of immunotherapy. Conclusion We constructed a subtyping model based on ICD-associated DAMPs in MPM, which might serve as a signature to gauge the outcomes of immune checkpoint blockades. Our research may aid in the development of innovative immunomodulators as well as the advancement of precision immunotherapy for MPM.
Collapse
Affiliation(s)
- Zheng Liu
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui Wan
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hua Bai
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
148
|
Huang S, Zeng L. Flow Cytometric Analysis of Regulated Cell Death. Methods Mol Biol 2023; 2712:165-178. [PMID: 37578705 DOI: 10.1007/978-1-0716-3433-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Cell death is a crucial and fundamental process in the biology of all living organisms and plays an essential role in developmental, cellular, and molecular biology. With a deeper understanding of the mechanisms of different types of cell death, quantitative experiments are becoming increasingly necessary to analyze the dynamic changes and coordinate physiological processes. Flow cytometry is the most widely used method for detecting and quantifying cell processes in mammalian cells, providing a comprehensive and high-throughput approach, even at the single-cell level. This chapter provides a brief overview of guidelines for performing flow cytometry in the detection of regulated cell deaths, including apoptosis, autophagy, ferroptosis, pyroptosis, and immunogenic cell death.
Collapse
Affiliation(s)
- Siyuan Huang
- Department of Trauma Medical Center, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China
| | - Ling Zeng
- Department of Trauma Medical Center, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
149
|
Yang R, Jia L, Lv Z, Cui J. Case report: Hilar metastasis of breast cancer: A single-center retrospective case-control study. Front Surg 2023; 10:1025287. [PMID: 36896259 PMCID: PMC9989156 DOI: 10.3389/fsurg.2023.1025287] [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: 08/29/2022] [Accepted: 01/24/2023] [Indexed: 02/25/2023] Open
Abstract
Purpose The lungs are a common metastatic organ in breast cancer, mainly due to blood metastasis. On imaging, most metastatic lesions show a peripheral round mass in the lung, occasionally with a hilar mass as the primary manifestation, showing burr and lobulation signs. This study aimed to investigate breast cancer patient's clinical characteristics and prognosis with two different metastatic sites in the lung. Methods We retrospectively analyzed patients admitted to the First Hospital of Jilin University between 2016 and 2021 diagnosed with breast cancer lung metastases. Forty breast cancer patients with hilar metastases (HM) and 40 patients with peripheral lung metastases (PLM) were matched 1:1 using a pairing method. To analyze the patient's prognosis, the clinical characteristics of patients with two different metastatic sites were compared using the chi-square test, Kaplan-Meier curve, and Cox proportional hazards model. Results The median follow-up time was 38 months (2-91 months). The median age of patients with HM was 56 years (25-75 years), and that of patients with PLM was 59 years (44-82 years). The median overall survival (mOS) was 27 months in the HM group and 42 months in the PLM group (p = 0.001). The results of the Cox proportional hazards model showed that the histological grade (hazard ratio = 2.741, 95% confidence interval 1.442-5.208, p = 0.002) was a prognostic factor in the HM group. Conclusion The number of young patients in the HM group was higher than that in the PLM group, with higher Ki-67 indexes and histological grades. Most patients had mediastinal lymph node metastasis, with shorter DFI and OS and poor prognosis.
Collapse
Affiliation(s)
- Ruohan Yang
- Cancer Center, the First Hospital of Jilin University, Changchun, China
| | - Lin Jia
- Cancer Center, the First Hospital of Jilin University, Changchun, China
| | - Zheng Lv
- Cancer Center, the First Hospital of Jilin University, Changchun, China
| | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
150
|
Zhou Q, Wu W, Jia K, Qi G, Sun XS, Li P. Design and characterization of PROTAC degraders specific to protein N-terminal methyltransferase 1. Eur J Med Chem 2022; 244:114830. [PMID: 36228414 PMCID: PMC10520980 DOI: 10.1016/j.ejmech.2022.114830] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 11/24/2022]
Abstract
Protein N-terminal methylation catalyzed by N-terminal methyltransferase 1 (NTMT1) is an emerging methylation present in eukaryotes, playing important regulatory roles in various biological and cellular processes. Although dysregulation of NTMT1 has been linked to many diseases such as colorectal cancer, their molecular and cellular mechanisms remain elusive due to inaccessibility to an effective cellular probe. Here we report the design, synthesis, and characterization of the first-in-class NTMT1 degraders based on proteolysis-targeting chimera (PROTAC) strategy. Through a brief structure-activity relationship (SAR) study of linker length, a cell permeable degrader 1 involving a von Hippel-Lindau (VHL) E3 ligase ligand was developed and demonstrated to reduce NTMT1 protein levels effectively and selectively in time- and dose-dependent manners in colorectal carcinoma cell lines HCT116 and HT29. Degrader 1 displayed DC50 = 7.53 μM and Dmax > 90% in HCT116 (cellular IC50 > 100 μM for its parent inhibitor DC541). While degrader 1 had marginal cytotoxicity, it displayed anti-proliferative activity in 2D and 3D culture environment, resulting from cell cycle arrested at G0/G1 phase in HCT116. Label-free global proteomic analysis revealed that degrader 1 induced overexpression of calreticulin (CALR), an immunogenic cell death (ICD) signal protein that is known to elicit antitumor immune response and clinically linked to a high survival rate of patients with colorectal cancer upon its upregulation. Collectively, degrader 1 offers the first selective cellular probe for NTMT1 exploration and a new drug discovery modality for NTMT1-related oncology and diseases.
Collapse
Affiliation(s)
- Qilong Zhou
- Department of Chemistry, Kansas State University, Manhattan, KS, 66506, USA; Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine
| | - Wei Wu
- Department of Chemistry, Kansas State University, Manhattan, KS, 66506, USA
| | - Kaimin Jia
- Department of Chemistry, Kansas State University, Manhattan, KS, 66506, USA
| | - Guangyan Qi
- Department of Grain Science and Industry, Kansas State University, Manhattan, KS, 66506, USA
| | - Xiuzhi Susan Sun
- Department of Grain Science and Industry, Kansas State University, Manhattan, KS, 66506, USA; Department of Biological and Agricultural Engineering, Kansas State University, Manhattan, KS, 66506, USA
| | - Ping Li
- Department of Chemistry, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|