1
|
Qi R, Cheng X, Chen S, Fan J. Extracellular HSP70 facilitated β-glucan induced trained immunity in macrophages to suppress sepsis via TLR2-NF-κB axis. Cytokine 2025; 187:156861. [PMID: 39823994 DOI: 10.1016/j.cyto.2025.156861] [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: 12/01/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/20/2025]
Abstract
Sepsis is a common systemic infectious disease followed by extremely high incidence and mortality with no effective treatment and clinical drugs. As a key mediator involved in infection and immunity, it has been reported that sepsis patients are accompanied by increased heat shock protein 70 (HSP70). Trained immunity is a novel innate immunity approach that can be activated by β-glucan to fight against sepsis. The mechanism of HSP70 activating trained macrophages against sepsis needs further elucidation. Trained immunity and sepsis models were established by β-glucan and LPS individually both in vivo and in vitro. We demonstrated that HSP70 was significantly upregulated in septic mice serum, and HSP70 could protect mice from sepsis by activating β-glucan-trained macrophages as an ideal secondary inducer via TLR2-NF-κB pathway. Additionally, the sepsis resistant effects of HSP70 could be blocked by its antibody. In summary, more than a molecular chaperone to maintain homeostasis, HSP70 could be an important trained immunity inducer to help the body fighting against sepsis, which provided new stimuli for trained immunity and novel therapeutic solutions for sepsis.
Collapse
Affiliation(s)
- Ran Qi
- Department of Clinical Laboratory, The Second Children & Women's Healthcare of Jinan City, Jinan, Shandong, China
| | - Xin Cheng
- Department of Clinical Laboratory, Jinan City People's Hospital, Jinan, Shandong, China
| | - Shan Chen
- Department of Clinical Laboratory, The Second Children & Women's Healthcare of Jinan City, Jinan, Shandong, China
| | - Jinjun Fan
- Department of Clinical Laboratory, The Second Children & Women's Healthcare of Jinan City, Jinan, Shandong, China.
| |
Collapse
|
2
|
Xu F, Ni Q, Gong N, Xia B, Zhang J, Guo W, Hu Z, Li J, Liang XJ. Delivery Systems Developed for Treatment Combinations to Improve Adoptive Cell Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407525. [PMID: 39165065 DOI: 10.1002/adma.202407525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/26/2024] [Indexed: 08/22/2024]
Abstract
Adoptive cell therapy (ACT) has shown great success in the clinic for treating hematologic malignancies. However, solid tumor treatment with ACT monotherapy is still challenging, owing to insufficient expansion and rapid exhaustion of adoptive cells, tumor antigen downregulation/loss, and dense tumor extracellular matrix. Delivery strategies for combination cell therapy have great potential to overcome these hurdles. The delivery of vaccines, immune checkpoint inhibitors, cytokines, chemotherapeutics, and photothermal reagents in combination with adoptive cells, have been shown to improve the expansion/activation, decrease exhaustion, and promote the penetration of adoptive cells in solid tumors. Moreover, the delivery of nucleic acids to engineer immune cells directly in vivo holds promise to overcome many of the hurdles associated with the complex ex vivo cell engineering strategies. Here, these research advance, as well as the opportunities and challenges for integrating delivery technologies into cell therapy s are discussed, and the outlook for these emerging areas are criticlly analyzed.
Collapse
Affiliation(s)
- Fengfei Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Qiankun Ni
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, New Cornerstone Science Institute, Tsinghua University, Beijing, China
| | - Ningqiang Gong
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinchao Zhang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Weisheng Guo
- College of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
| | - Zhongbo Hu
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinghong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, New Cornerstone Science Institute, Tsinghua University, Beijing, China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
3
|
Li M, Li S, Guo Y, Hu P, Shi J. Magnetothermal-activated gene editing strategy for enhanced tumor cell apoptosis. J Nanobiotechnology 2024; 22:450. [PMID: 39080645 PMCID: PMC11287911 DOI: 10.1186/s12951-024-02734-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024] Open
Abstract
Precise and effective initiation of the apoptotic mechanism in tumor cells is one of the most promising approaches for the treatment of solid tumors. However, current techniques such as high-temperature ablation or gene editing suffer from the risk of damage to adjacent normal tissues. This study proposes a magnetothermal-induced CRISPR-Cas9 gene editing system for the targeted knockout of HSP70 and BCL2 genes, thereby enhancing tumor cell apoptosis. The magnetothermal nanoparticulate platform is composed of superparamagnetic ZnCoFe2O4@ZnMnFe2O4 nanoparticles and the modified polyethyleneimine (PEI) and hyaluronic acid (HA) on the surface, on which plasmid DNA can be effectively loaded. Under the induction of a controllable alternating magnetic field, the mild magnetothermal effect (42℃) not only triggers dual-genome editing to disrupt the apoptosis resistance mechanism of tumor cells but also sensitizes tumor cells to apoptosis through the heat effect itself, achieving a synergistic therapeutic effect. This strategy can precisely regulate the activation of the CRISPR-Cas9 system for tumor cell apoptosis without inducing significant damage to healthy tissues, thus providing a new avenue for cancer treatment.
Collapse
Affiliation(s)
- Mingyuan Li
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Siqian Li
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - YueDong Guo
- Shanghai Tenth People's Hospital, Medical School of Tongji University, 38 Yun-xin Road, Shanghai, 200435, P.R. China
| | - Ping Hu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China.
- Shanghai Tenth People's Hospital, Medical School of Tongji University, 38 Yun-xin Road, Shanghai, 200435, P.R. China.
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
4
|
Chen J, Liu J, Liu X, Wang J, Wang X, Ye X, Xie Q, Liang J, Li Y. Shikonin improves the effectiveness of PD-1 blockade in colorectal cancer by enhancing immunogenicity via Hsp70 upregulation. Mol Biol Rep 2024; 51:86. [PMID: 38183539 PMCID: PMC10771352 DOI: 10.1007/s11033-023-09056-2] [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: 05/16/2023] [Accepted: 10/23/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND PD-1 blockade has shown impressive clinical outcomes in colorectal cancers patients with high microsatellite instability (MSI-H). However, the majority of patients with colorectal cancer who present low microsatellite instability (MSI-L) or stable microsatellites (MSS) show little response to PD-1 blockade therapy. Here, we have demonstrated that Shikonin (SK) could induce cell death of CT26 cells via classically programmed and immunogenic pathways. METHODS AND RESULTS SK promoted the membrane exposure of calreticulin and upregulated the expression of heat shock protein 70 (Hsp70). The upregulation of Hsp70 was dependent on ROS induced by SK and silencing of PKM2 in CT26 cells reverts ROS upregulation. Besides, SK synergizes with PD-1 blockade in CT26 tumor mice model, with the increase of intramural DC cells and CD8+ T cells. The expression of Hsp70 in tumor tissue was also increased in combinational SK plus αPD-1 therapy group. CONCLUSIONS Our study elucidated the potential role of 'Shikonin-PKM2-ROS-Hsp70' axis in the promotion of efficacy of PD-1 blockade in CRC treatments, providing a potential strategy and targets for improving the efficacy of PD-1 blockade in colorectal cancer.
Collapse
Affiliation(s)
- Jinghua Chen
- Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jie Liu
- Department of Pediatric Intensive Care Unit, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, China
| | - Xiaolin Liu
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jun Wang
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiumei Wang
- Department of Oncology, The Yuncheng Chenxin Hospital, Heze, China
| | - Xin Ye
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Qi Xie
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.
| | - Jing Liang
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.
| | - Yan Li
- Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China.
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.
| |
Collapse
|
5
|
Bie N, Yong T, Wei Z, Liang Q, Zhang X, Li S, Li X, Li J, Gan L, Yang X. Tumor-repopulating cell-derived microparticles elicit cascade amplification of chemotherapy-induced antitumor immunity to boost anti-PD-1 therapy. Signal Transduct Target Ther 2023; 8:408. [PMID: 37875473 PMCID: PMC10598206 DOI: 10.1038/s41392-023-01658-3] [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: 05/07/2023] [Revised: 08/30/2023] [Accepted: 09/15/2023] [Indexed: 10/26/2023] Open
Abstract
Immune checkpoint blockade (ICB) therapy, particularly antibodies targeting the programmed death receptor 1 (PD-1) and its ligand (PD-L1), has revolutionized cancer treatment. However, its efficacy as a standalone therapy remains limited. Although ICB therapy in combination with chemotherapy shows promising therapeutic responses, the challenge lies in amplifying chemotherapy-induced antitumor immunity effectively. This relies on efficient drug delivery to tumor cells and robust antigen presentation by dendritic cells (DCs). Here, we developed tumor-repopulating cell (TRC)-derived microparticles with exceptional tumor targeting to deliver doxorubicin (DOX@3D-MPs) for improve anti-PD-1 therapy. DOX@3D-MPs effectively elicit immunogenic tumor cell death to release sufficient tumor antigens. Heat shock protein 70 (HSP70) overexpressed in DOX@3D-MPs contributes to capturing tumor antigens, promoting their phagocytosis by DCs, and facilitating DCs maturation, leading to the activation of CD8+ T cells. DOX@3D-MPs significantly enhance the curative response of anti-PD-1 treatment in large subcutaneous H22 hepatoma, orthotopic 4T1 breast tumor and Panc02 pancreatic tumor models. These results demonstrate that DOX@3D-MPs hold promise as agents to improve the response rate to ICB therapy and generate long-lasting immune memory to prevent tumor relapse.
Collapse
Affiliation(s)
- Nana Bie
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Zhaohan Wei
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Qingle Liang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Xiaoqiong Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Shiyu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Xin Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Jianye Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China.
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China.
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, 430074, Wuhan, China.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China.
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China.
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, 430074, Wuhan, China.
| |
Collapse
|
6
|
Xue S, Jiao J, Miao S, Wang L, Liu Y, Zhang Q, Wang Q, Xi Y, Zhang Y. Lipid-coated bismuth nanoflower as the thermos-radio sensiti for therapy of lung metastatic breast cancer: Preparation, optimisation, and characterisation. IET Nanobiotechnol 2022; 16:305-315. [PMID: 36036543 DOI: 10.1049/nbt2.12097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/19/2022] Open
Abstract
Lung metastatic breast cancer (LMBC) leads to a large number of deaths in women with breast cancer, and radiotherapy has been considered the common assay for tumour therapy except for surgery. However, radiotherapy still faces problems of low efficiency due to resistance and easily induced side effects. Here, the authors designed lipid-decorated bismuth-based nanoflowers (DP-BNFs) as both a radiosensitiser and a photothermal therapy agent for LMBC treatment. The BNFs were prepared by oxidation of bismuth nitrate and subsequent reduction using sodium borohydride. The preparation parameters and formulation of DP-BNFs were optimised via a single-factor experiment, with the factors including reaction temperature, a molar ratio of reducing agents, and the types and amount of decorated lipid materials. The result indicated that the BNFs prepared at 170°C with the Bi/NaBH4 ratio of 1:0.7 exhibited the best yield and particle size around 160 nm. After being spray dried with lactose to prepare dry powder inhalation (DP-BNF@Lat-MPs), their effects on improving therapeutic efficiency of the radiotherapy and photothermal therapy combination were measured using the western blot assay to determine the tumour apoptosis. In a word, DP-BNF@Lat-MPs could be a novel inhalable integrated microsphere that provides a new possibility for thermoradiotherapy of LMBC.
Collapse
Affiliation(s)
- Shushu Xue
- Department of Radiotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Junrong Jiao
- Department of Radiotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Si Miao
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, Nanjing, China
| | - Lijun Wang
- Department of Radiotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Liu
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, Nanjing, China
| | - Qingjie Zhang
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, Nanjing, China
| | - Qiyue Wang
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, China
| | - Yu Xi
- Department of Pharmacy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yuanyuan Zhang
- Department of Pharmacy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Zhao L, Zhou Y, Bai Z, Zhang F, Yang X. The underlying molecular mechanism of intratumoral radiofrequency hyperthermia-enhanced chemotherapy of pancreatic cancer. J Interv Med 2022; 5:57-63. [PMID: 35936663 PMCID: PMC9349012 DOI: 10.1016/j.jimed.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 02/22/2022] [Accepted: 02/28/2022] [Indexed: 02/07/2023] Open
Abstract
Background To investigate the underlying molecular mechanisms of radiofrequency hyperthermia (RFH)-enhanced direct chemotherapy of pancreatic cancers. Method Rat ductal PaCa cell line DSL-6A/C1 and orthotopic pancreatic cancers of Lewis rats were divided into four study groups with various treatments: i) phosphate-buffered saline (PBS) as a control; ii) RFH alone; iii) intratumoral chemotherapy alone (gemcitabine); and (iv) combination therapy of gemcitabine plus intratumoral RFH at 42 °C for 30 min. In the in-vitro confirmation experiments, the viability and apoptosis of DSL-6A/C1 cells in each treatment group were evaluated using cell live/dead staining, flow cytometry, and Western blot. In the in vivo validation experiments, related proteins were evaluated by immunohistochemistry (IHC) staining of tumors. Results Of the in-vitro experiments, the lowest cell viability and more apoptotic cells were shown in the group with combination therapy compared to other treatments. Western blot data showed elevated Bax/Bcl-2, Caspase-3, and HSP70 expressions in DSL cells with combination therapy, compared to other treatments. Of the in vivo experiments, IHC staining detected the significantly increased expressions of HSP70, IL-1β, TNF-ɑ, Bax, and Caspase-3 in pancreatic cancer tissues of the animal group treated by combination therapy of gemcitabine with RFH. Conclusion Molecular imaging-guided interventional RFH can significantly enhance the chemotherapeutic effect on pancreatic cancers via potential molecular mechanisms of up-regulating Bax/caspase-3-dependent apoptosis pathways.
Collapse
|
8
|
El Meskini R, Atkinson D, Kulaga A, Abdelmaksoud A, Gumprecht M, Pate N, Hayes S, Oberst M, Kaplan IM, Raber P, Van Dyke T, Sharan SK, Hollingsworth R, Day CP, Merlino G, Weaver Ohler Z. Distinct Biomarker Profiles and TCR Sequence Diversity Characterize the Response to PD-L1 Blockade in a Mouse Melanoma Model. Mol Cancer Res 2021; 19:1422-1436. [PMID: 33888600 DOI: 10.1158/1541-7786.mcr-20-0881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 03/09/2021] [Accepted: 04/19/2021] [Indexed: 11/16/2022]
Abstract
Only a subset of patients responds to immune checkpoint blockade (ICB) in melanoma. A preclinical model recapitulating the clinical activity of ICB would provide a valuable platform for mechanistic studies. We used melanoma tumors arising from an Hgftg;Cdk4R24C/R24C genetically engineered mouse (GEM) model to evaluate the efficacy of an anti-mouse PD-L1 antibody similar to the anti-human PD-L1 antibodies durvalumab and atezolizumab. Consistent with clinical observations for ICB in melanoma, anti-PD-L1 treatment elicited complete and durable response in a subset of melanoma-bearing mice. We also observed tumor growth delay or regression followed by recurrence. For early treatment assessment, we analyzed gene expression profiles, T-cell infiltration, and T-cell receptor (TCR) signatures in regressing tumors compared with tumors exhibiting no response to anti-PD-L1 treatment. We found that CD8+ T-cell tumor infiltration corresponded to response to treatment, and that anti-PD-L1 gene signature response indicated an increase in antigen processing and presentation, cytokine-cytokine receptor interaction, and natural killer cell-mediated cytotoxicity. TCR sequence data suggest that an anti-PD-L1-mediated melanoma regression response requires not only an expansion of the TCR repertoire that is unique to individual mice, but also tumor access to the appropriate TCRs. Thus, this melanoma model recapitulated the variable response to ICB observed in patients and exhibited biomarkers that differentiate between early response and resistance to treatment, providing a valuable platform for prediction of successful immunotherapy. IMPLICATIONS: Our melanoma model recapitulates the variable response to anti-PD-L1 observed in patients and exhibits biomarkers that characterize early antibody response, including expansion of the TCR repertoire.
Collapse
Affiliation(s)
- Rajaa El Meskini
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland.
| | - Devon Atkinson
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Alan Kulaga
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Abdalla Abdelmaksoud
- Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research (CCR), National Cancer Institute, Bethesda, Maryland.,Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Michelle Gumprecht
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Nathan Pate
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | | | | | | | | | - Terry Van Dyke
- Mouse Cancer Genetics Program, CCR, NCI/NIH, Frederick, Maryland
| | - Shyam K Sharan
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland.,Mouse Cancer Genetics Program, CCR, NCI/NIH, Frederick, Maryland
| | | | - Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, CCR, NCI/NIH, Bethesda, Maryland
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, CCR, NCI/NIH, Bethesda, Maryland
| | - Zoë Weaver Ohler
- Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland.
| |
Collapse
|
9
|
Sheikh-Hosseini M, Larijani B, Gholipoor Kakroodi Z, Shokoohi M, Moarefzadeh M, Sayahpour FA, Goodarzi P, Arjmand B. Gene Therapy as an Emerging Therapeutic Approach to Breast Cancer: New Developments and Challenges. Hum Gene Ther 2021; 32:1330-1345. [PMID: 33307949 DOI: 10.1089/hum.2020.199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Breast cancer is a heterogeneous disease, which is the consequence of several genetic and environmental factors. Also, it is one of the most common causes of cancer death and second leading cancer among women all around the world. Therefore, it is necessary to develop novel therapeutic approaches useful for the successful treatment of breast cancer. As conventional treatments had limited success, alternative approaches for the treatment of breast cancer have been applied in recent years. Hence, the molecular basis of breast cancer has provided the opportunity of using genetic materials for therapeutic uses. In this regard, gene therapy as one of the potentially efficient and beneficial treatments among various techniques became a popular treatment for different cancers, especially breast cancer. Accordingly, there are plenty of targets available for gene therapy of breast cancer. Gene therapy strategies have the potential to correct molecular defects that contributed to the cancer progression. These techniques should selectively target tumor cells without affecting normal cells. Moreover, data of clinical trials in gene therapy for breast cancer indicated that this approach has little toxicity compared to other therapeutic approaches. In this study, different aspects of breast neoplasm, gene therapy techniques, challenges, and recent developments will be mentioned.
Collapse
Affiliation(s)
- Motahareh Sheikh-Hosseini
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Gholipoor Kakroodi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahshid Shokoohi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Moarefzadeh
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Forough Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Khan M, Zhao Z, Arooj S, Fu Y, Liao G. Soluble PD-1: Predictive, Prognostic, and Therapeutic Value for Cancer Immunotherapy. Front Immunol 2020; 11:587460. [PMID: 33329567 PMCID: PMC7710690 DOI: 10.3389/fimmu.2020.587460] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022] Open
Abstract
Programmed death protein 1 (PD-1) interaction with PD-L1 deliver immunosuppressive environment for tumor growth, and its blockade with directed monoclonal antibodies (anti-PD-1/anti-PD-L1) has shown remarkable clinical outcome. Lately, their soluble counterparts, sPD-1 and sPD-L1, have been detected in plasma, and elevated levels have been associated with advanced disease, clinical stages, and worst prognosis for cancer patients. Elevated plasma levels of sPD-L1 have been correlated with worst prognosis in several studies and has displayed a persistent outlook. On the other hand, sPD-1 levels have been inconsistent in their predictive and prognostic ability. Pretherapeutic higher sPD-1 plasma levels have shown to predict advanced disease state and to a lesser extent worst prognosis. Any increase in sPD-1 plasma level post therapeutically have been correlated with improved survival for various cancers. In vitro and in vivo studies have shown sPD-1 ability to bind PD-L1 and PD-L2 and block PD-1/PD-L1 interaction. Local delivery of sPD-1 in cancer tumor microenvironment through local gene therapy have demonstrated an increase in tumor specific CD8+ T cell immunity and tumor growth reduction. It had also exhibited enhancement of T cell immunity induced by vaccination and other gene therapeutic agents. Furthermore, it may also lessen the inhibitory effect of circulating sPD-L1 and enhance the effects of mAb-based immunotherapy. In this review, we highlight various aspects of sPD-1 role in cancer prediction, prognosis, and anti-cancer immunity, as well as, its therapeutic value for local gene therapy or systemic immunotherapy in blocking the PD-1 and PD-L1 checkpoint interactions.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China.,Department of Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhihong Zhao
- Department of Nephrology, Shenzhen People's Hospital, Second Clinical Medicine Centre, Jinan University, Shenzhen, China
| | - Sumbal Arooj
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China.,Department of Biochemistry, University of Sialkot, Sialkot, Pakistan
| | - Yuxiang Fu
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Guixiang Liao
- Department of Radiation Oncology, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
11
|
Molecular Chaperones: Molecular Assembly Line Brings Metabolism and Immunity in Shape. Metabolites 2020; 10:metabo10100394. [PMID: 33023034 PMCID: PMC7600384 DOI: 10.3390/metabo10100394] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Molecular chaperones are a set of conserved proteins that have evolved to assist the folding of many newly synthesized proteins by preventing their misfolding under conditions such as elevated temperatures, hypoxia, acidosis and nutrient deprivation. Molecular chaperones belong to the heat shock protein (HSP) family. They have been identified as important participants in immune functions including antigen presentation, immunostimulation and immunomodulation, and play crucial roles in metabolic rewiring and epigenetic circuits. Growing evidence has accumulated to indicate that metabolic pathways and their metabolites influence the function of immune cells and can alter transcriptional activity through epigenetic modification of (de)methylation and (de)acetylation. However, whether molecular chaperones can regulate metabolic programs to influence immune activity is still largely unclear. In this review, we discuss the available data on the biological function of molecular chaperones to immune responses during inflammation, with a specific focus on the interplay between molecular chaperones and metabolic pathways that drive immune cell fate and function.
Collapse
|
12
|
Guo D, Zhang A, Huang J, Suo M, Zhong Y, Liang Y. Suppression of HSP70 inhibits the development of acute lymphoblastic leukemia via TAK1/Egr-1. Biomed Pharmacother 2019; 119:109399. [PMID: 31521893 DOI: 10.1016/j.biopha.2019.109399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 08/21/2019] [Accepted: 08/28/2019] [Indexed: 02/04/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL), usually treated with chemotherapy, has limited therapeutic effects and high toxicity. Upregulation of HSP70 induces tumor development, however, the molecular mechanism of HSP70 in ALL remains unclear. In our research, we aimed to investigate the role of HSP70 in ALL, specifically the molecular mechanisms underlying cell apoptosis and proliferation. We found that HSP70 expression in leukomonocytes from ALL patients was increased compared with the control group. HSP70 expression in NALM-6 and BE-13 was also up-regulated contrast with AHH-1. Inhibition of HSP70 significantly promoted cell apoptosis and suppressed cell proliferation in ALL cell lines. Suppression of HSP70 decreased TAK1 and increased Egr-1 protein expression. Further experiments indicated that overexpression of TAK1 ameliorated the effect of HSP70 inhibition on Egr-1 protein expression, cell apoptosis and proliferation. In order to determine whether the effect of HSP70 inhibition on apoptosis and proliferation of ALL cell lines could be achieved via regulation of Egr-1, we performed a loss-of-function experiment for Egr-1. Egr-1 suppression was found to reverse the effect of HSP70 inhibition on cell apoptosis and proliferation in ALL. Taken together, our results suggest that HSP70 inhibition upregulates Egr-1 via TAK1, inducing apoptosis and restricting proliferation in ALL cells.
Collapse
Affiliation(s)
- Dongfang Guo
- Department of Clinical Laboratory, Zhumadian Central Hospital, Zhumadian, 463100, China.
| | - Airong Zhang
- Department of Clinical Laboratory, Zhumadian Central Hospital, Zhumadian, 463100, China
| | - Jing Huang
- Department of Clinical Laboratory, Zhumadian Central Hospital, Zhumadian, 463100, China
| | - Meifang Suo
- Department of Clinical Laboratory, Zhumadian Central Hospital, Zhumadian, 463100, China
| | - Yaping Zhong
- Deparment of Hematopathology, Zhumadian Central Hospital, Zhumadian, 463100, China
| | - Yile Liang
- Deparment of Infectious Diseases, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471000, China
| |
Collapse
|
13
|
Lorenzo-Herrero S, Sordo-Bahamonde C, González S, López-Soto A. Immunosurveillance of cancer cell stress. Cell Stress 2019; 3:295-309. [PMID: 31535086 PMCID: PMC6732214 DOI: 10.15698/cst2019.09.198] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 12/11/2022] Open
Abstract
Cancer development is tightly controlled by effector immune responses that recognize and eliminate malignantly transformed cells. Nonetheless, certain immune subsets, such as tumor-associated macrophages, have been described to promote tumor growth, unraveling a double-edge role of the immune system in cancer. Cell stress can modulate the crosstalk between immune cells and tumor cells, reshaping tumor immunogenicity and/or immune function and phenotype. Infiltrating immune cells are exposed to the challenging conditions typically present in the tumor microenvironment. In return, the myriad of signaling pathways activated in response to stress conditions may tip the balance toward stimulation of antitumor responses or immune-mediated tumor progression. Here, we explore how distinct situations of cellular stress influence innate and adaptive immunity and the consequent impact on cancer establishment and progression.
Collapse
Affiliation(s)
- Seila Lorenzo-Herrero
- Departamento de Biología Funcional, Inmunología, Universidad de Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA) Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Christian Sordo-Bahamonde
- Departamento de Biología Funcional, Inmunología, Universidad de Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA) Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Segundo González
- Departamento de Biología Funcional, Inmunología, Universidad de Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA) Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Alejandro López-Soto
- Departamento de Biología Funcional, Inmunología, Universidad de Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA) Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
14
|
Lei YMM, Jianquan ZMD, Jianguo SMM, Hang ZMM, Zongping DMM, Jianming ZP. The Significance of Heat Shock Protein 70 Expression in Benign Thyroid Nodules During Thermal Ablation. ADVANCED ULTRASOUND IN DIAGNOSIS AND THERAPY 2019. [DOI: 10.37015/audt.2019.190817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
15
|
Class A1 scavenger receptor modulates glioma progression by regulating M2-like tumor-associated macrophage polarization. Oncotarget 2018; 7:50099-50116. [PMID: 27367025 PMCID: PMC5226571 DOI: 10.18632/oncotarget.10318] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 06/12/2016] [Indexed: 01/10/2023] Open
Abstract
Macrophages enhance glioma development and progression by shaping the tumor microenvironment. Class A1 scavenger receptor (SR-A1), a pattern recognition receptor primarily expressed in macrophages, is up-regulated in many human solid tumors. We found that SR-A1 expression in 136 human gliomas was positively correlated with tumor grade (P<0.01), but not prognosis or tumor recurrence. SR-A1-expressing macrophages originated primarily from circulating monocytes attracted to tumor tissue, and were almost twice as numerous as resident microglia in glioma tissues (P<0.001). The effects of SR-A1 on glioma proliferation and invasion were assessed in vivo using an SR-A1-deficient murine orthotopic glioma model. SR-A1 deletion promoted M2-like tumor-associated macrophage (TAM) polarization in mice by activating STAT3 and STAT6, which resulted in robust orthotopic glioma proliferation and angiogenesis. Finally, we found that HSP70 might be an endogenous ligand that activates SR-A1-dependent anti-tumorigenic pathways in gliomas, although its expression does not appear informative for diagnostic purposes. Our findings demonstrate a relationship between TAMs, SR-A1 expression and glioma growth and provide new insights into the pathogenic role of TAMs in glioma.
Collapse
|
16
|
Yang X, Guo Y, Guo Z, Si T, Xing W, Yu W, Wang Y. Cryoablation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Oncotarget 2018; 10:4180-4191. [PMID: 31289616 PMCID: PMC6609244 DOI: 10.18632/oncotarget.24105] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/03/2018] [Indexed: 11/25/2022] Open
Abstract
Cryoablation is moderately effective against prostate cancer. Of note, the off-target or enlarged therapeutic effect after cryoablation is reported in routine clinical practice. To uncover it, we constructed a bilateral inguinal transplantation model of prostate cancer. All the mice were randomly subdivided into three groups: Group A (Control group), Group B (Surgery group), and Group C (Cryoablation group). All the procedures in three groups were conducted only for tumors in the target region (right groin). The tumors in untargeted region (left groin) received no treatment. We measured the growth of untargeted tumors and lung metastasis rate, and then explored the changes in a series of immune cells and danger signals. First, our results revealed the protective effect of cryoablation treatment against the abscopal tumor. The possible mechanism was mediated by an increase in the number of CD4+ T cells, CD8+ T cells, ratio T helper 1 (Th1)/Th2, the killing activity of cytotoxic T lymphocytes and NK cells. Hsp70 may be involved in the modulation of the immune response. The combination of weakened Ki67 activity and activated immune response delayed spectator tumor growth, decreased the pulmonary metastasis rate, and prolonged animal survival, with an inducible abscopal effect.
Collapse
Affiliation(s)
- Xueling Yang
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,National Clinical Research Center of Cancer, Tianjin 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Yongfei Guo
- Department of Interventional Radiology, Tianjin Hospital of ITCWM Nankai Hospital, Tianjin 100020, China
| | - Zhi Guo
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,National Clinical Research Center of Cancer, Tianjin 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Tongguo Si
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,National Clinical Research Center of Cancer, Tianjin 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Wenge Xing
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,National Clinical Research Center of Cancer, Tianjin 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Wenwen Yu
- National Clinical Research Center of Cancer, Tianjin 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.,Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Yan Wang
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,National Clinical Research Center of Cancer, Tianjin 300060, China.,Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| |
Collapse
|
17
|
Qi YX, Liu K, Yin J, Li L. Evaluation of short- and long-term efficacy of chemoradiotherapy for advanced cervical cancer using HSP70 protein combined with multimodal MRI. J Cell Biochem 2017; 119:3017-3029. [PMID: 29023986 DOI: 10.1002/jcb.26424] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 10/10/2017] [Indexed: 01/16/2023]
Abstract
This study aims to evaluate the efficacy of chemoradiotherapy for advanced cervical cancer using heat shock protein 70 (HSP70) combined with multimodal MRI. The protein expressions of HSP70 in biopsy specimens obtained from 101 patients with cervical cancer were detected by immunohistochemistry. Plain MRI scan, DWI, DCE-MR, and MRS were performed before and after a period of 6 months of chemoradiotherapy. All patients were assigned into the complete response (CR) and partial response (PR) groups. HSP70 protein expression, tumor diameter, and tumor volume were lower in the CR group than in the PR group. The rate of tumor shrinkage, relative positive enhancement integral (rPEI), relative maximum slope of decrease (rMSD), relative signal enhancement ratio at 60 s (rSER60 ) and maximum SER (rSERmax ), mean apparent diffusion coefficient (ADCmean ) and minimum ADC value (ADCmin ) values in the CR group were higher than those in the PR group. Tumor diameter and volume, rSER15 and rSER30 were reduced after chemoradiotherapy, while rMSD, rSERmax , time to peak (TTP), ADCmean and ADCmin were higher after the treatment. The receiver operating characteristic (ROC) curves indicated that HSP70 expression, tumor diameter, rPEI, ADCmean and Cho peak showed the better chemoradiotherapy efficacy. Our data demonstrates that HSP70 protein combined with multimodal MRI may accurately evaluate the chemoradiotherapy efficacy of patients with advanced cervical cancer. The recurrence of cervical cancer significantly decreased in patients with negative expression of HSP70 and HSP70 protein detection provides potential therapy for the prevention, diagnosis, and prognosis of cervical cancer.
Collapse
Affiliation(s)
- Yun-Xiang Qi
- Sichuan Cancer Hospital and Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Kun Liu
- Department of Radiology, Daqing Longnan Hospital, Daqing, P.R. China
| | - Jun Yin
- Sichuan Cancer Hospital and Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Lu Li
- Sichuan Cancer Hospital and Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, P.R. China
| |
Collapse
|
18
|
Zhang D, Zhang Y, Zhu B, Zhang H, Sun Y, Sun C. Resveratrol may reverse the effects of long-term occupational exposure to electromagnetic fields on workers of a power plant. Oncotarget 2017; 8:47497-47506. [PMID: 28537898 PMCID: PMC5564581 DOI: 10.18632/oncotarget.17668] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/21/2017] [Indexed: 11/25/2022] Open
Abstract
High-voltage electricity lines are known to generate extremely low-frequency electromagnetic fields (ELF-EMFs). With the process of urbanization, increasing concerns has been focused on the potentially hazardous impacts of ELF-EMF on human health, and the conclusions are controversial. Little is known about the method of prevention against ELF-EMF induced healthy problems. A total of 186 male workers with occupational exposure to high-voltage electricity lines, and 154 male subjects with insignificant exposure as reference control were enrolled in this study. Resveratrol or placebo was given as dietary supplements (500 mg twice daily), and several inflammatory biomarkers and biomarkers of oxidative stress were assessed. Workers who had long-term exposure to high-voltage electricity lines exhibited elevated urinary levels of 8-hydroxy-2-deoxy-guanosine (8-OHdG) and F2-isoprostane, compared to the reference group. Lower plasma nuclear factor kappa B (NF-κB) and interleukin (IL)-6 were observed in exposed workers compared to the reference group. Resveratrol significantly reversed the adverse impacts of ELF-EMF. Stimulated cytokine production by resveratrol was found in exposed workers but not in the reference group. This study supported that occupational and long-term exposure to high-voltage electricity lines has an adverse effect on homeostasis of human body, and resveratrol supplement could be an effective protection strategy against the adverse effects induced by ELF-EMFs.
Collapse
Affiliation(s)
- Dan Zhang
- Electrical Power Research Institute, Jilin Electrical Power Company Limited, Changchun 130021, Jilin, China
| | - Yang Zhang
- Electrical Power Research Institute, Jilin Electrical Power Company Limited, Changchun 130021, Jilin, China
| | - Baoyu Zhu
- Electrical Power Research Institute, Jilin Electrical Power Company Limited, Changchun 130021, Jilin, China
| | - He Zhang
- Electrical Power Research Institute, Jilin Electrical Power Company Limited, Changchun 130021, Jilin, China
| | - Ye Sun
- Electrical Power Research Institute, Jilin Electrical Power Company Limited, Changchun 130021, Jilin, China
| | - Chengxun Sun
- Electrical Power Research Institute, Jilin Electrical Power Company Limited, Changchun 130021, Jilin, China
| |
Collapse
|
19
|
Zhang W, Tao H, Zeng J, Fang G, Liang B, Zhou L, Luo X, Shi J, Niu L. Laparotomy Cryoablation in Rabbit VX2 Pancreatic Carcinoma. Pancreas 2017; 46:288-295. [PMID: 28129233 DOI: 10.1097/mpa.0000000000000798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE The aim of this study was to establish a suitable rabbit model and select the optimal protocol for laparotomy cryoablation of pancreatic carcinoma. METHODS VX2 tumor tissues were inoculated into rabbit pancreases to build the pancreatic carcinoma model; then, the tumor-bearing rabbits were randomly divided into 4 groups: control, treatment A (the cryoablated-iceball diameter was bigger than the tumor), treatment B (iceball was as big as the tumor), and treatment C (iceball was smaller than the tumor). Related laboratory tests were conducted, and survival time was recorded. RESULTS The VX2 pancreatic carcinoma model was successfully established, and serum neuron-specific enolase levels increased continuously after inoculation. Compared with controls, rabbits in treatments A and C groups had no significant survival benefit (P > 0.05), but treatment B significantly prolonged the survival time (P < 0.01). CONCLUSIONS VX2 pancreatic cancer model was successfully established with neuron-specific enolase as biomarker. Treatment B may be the optimal protocol for pancreatic carcinoma and a new treatment option for patients with unresectable pancreatic cancer.
Collapse
Affiliation(s)
- Wenlong Zhang
- From the *Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun; and †Fuda Cancer Hospital, Jinan University School of Medicine (Guangzhou Fuda Cancer Hospital), and ‡Guangzhou Fuda Cancer Institute, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Gabai VL, Yaglom JA, Wang Y, Meng L, Shao H, Kim G, Colvin T, Gestwicki J, Sherman MY. Anticancer Effects of Targeting Hsp70 in Tumor Stromal Cells. Cancer Res 2016; 76:5926-5932. [PMID: 27503927 DOI: 10.1158/0008-5472.can-16-0800] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/12/2016] [Indexed: 12/27/2022]
Abstract
The stress-induced chaperone protein Hsp70 enables the initiation and progression of many cancers, making it an appealing therapeutic target for development. Here, we show that cancer cells resistant to Hsp70 inhibitors in vitro remain sensitive to them in vivo, revealing the pathogenic significance of Hsp70 in tumor stromal cells rather than tumor cells as widely presumed. Using transgenic mouse models of cancer, we found that expression of Hsp70 in host stromal cells was essential to support tumor growth. Furthermore, genetic ablation or pharmacologic inhibition of Hsp70 suppressed tumor infiltration by macrophages needed to enable tumor growth. Overall, our results illustrate how Hsp70 inhibitors mediate the anticancer effects by targeting both tumor cells and tumor stromal cells, with implications for the broad use of these inhibitors as tools to ablate tumor-associated macrophages that enable malignant progression. Cancer Res; 76(20); 5926-32. ©2016 AACR.
Collapse
Affiliation(s)
- Vladimir L Gabai
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts. CureLab Oncology, Needham, Massachusetts
| | - Julia A Yaglom
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Yongmei Wang
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Le Meng
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts. Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, Massachusetts
| | - Hao Shao
- Department of Pharmaceutical Chemistry and Institute for Neurodegenerative Disease, University of California at San Francisco, San Francisco, California
| | - Geunwon Kim
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Teresa Colvin
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Jason Gestwicki
- Department of Pharmaceutical Chemistry and Institute for Neurodegenerative Disease, University of California at San Francisco, San Francisco, California
| | - Michael Y Sherman
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
21
|
Abstract
Heat shock protein 70, (Hsp70) constitutes a powerful system of cytoprotection in all organisms studied to date. Exerting such activity, Hsp70 rescues cancer cells from antitumor therapy, posing a great challenge for oncologists. In contrast to its protective action, Hsp70 was found to be released from cancer cells, prompting cytotoxic lymphocytes to target and kill the tumor. A great number of vaccines have been developed on the basis of the ability of Hsp70 to present tumor antigen or to elevate the sensitivity of cancer cells to cytotoxic lymphocytes. In this commentary, we consider novel data on the employment of pure Hsp70 in the therapy of glioma and melanoma malignancies. We show that intratumorally delivered Hsp70 penetrates cancer cells and pulls its intracellular analog outside of the cell. This displacement may activate cells, constituting both innate and adaptive immunity. In vivo delivery of Hsp70 was found to inhibit tumor growth and to extend survival. The technology of intratumoral injection of pure Hsp70 passed through preclinical trials and was investigated in clinics for children with brain cancer; the results show the safety and feasibility of a new approach.
Collapse
Affiliation(s)
- Irina V Guzhova
- a Institute of Cytology of Russian Academy of Sciences , St. Petersburg , Russia
| | - Boris A Margulis
- a Institute of Cytology of Russian Academy of Sciences , St. Petersburg , Russia
| |
Collapse
|
22
|
Mining for Candidate Genes Related to Pancreatic Cancer Using Protein-Protein Interactions and a Shortest Path Approach. BIOMED RESEARCH INTERNATIONAL 2015; 2015:623121. [PMID: 26613085 PMCID: PMC4647023 DOI: 10.1155/2015/623121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 10/15/2015] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer (PC) is a highly malignant tumor derived from pancreas tissue and is one of the leading causes of death from cancer. Its molecular mechanism has been partially revealed by validating its oncogenes and tumor suppressor genes; however, the available data remain insufficient for medical workers to design effective treatments. Large-scale identification of PC-related genes can promote studies on PC. In this study, we propose a computational method for mining new candidate PC-related genes. A large network was constructed using protein-protein interaction information, and a shortest path approach was applied to mine new candidate genes based on validated PC-related genes. In addition, a permutation test was adopted to further select key candidate genes. Finally, for all discovered candidate genes, the likelihood that the genes are novel PC-related genes is discussed based on their currently known functions.
Collapse
|
23
|
Sherman MY, Gabai VL. Hsp70 in cancer: back to the future. Oncogene 2015; 34:4153-61. [PMID: 25347739 PMCID: PMC4411196 DOI: 10.1038/onc.2014.349] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/08/2014] [Accepted: 09/22/2014] [Indexed: 12/24/2022]
Abstract
Mechanistic studies from cell culture and animal models have revealed critical roles for the heat shock protein Hsp70 in cancer initiation and progression. Surprisingly, many effects of Hsp70 on cancer have not been related to its chaperone activity, but rather to its role(s) in regulating cell signaling. A major factor that directs Hsp70 signaling activity appears to be the co-chaperone Bag3. Here, we review these recent breakthroughs, and how these discoveries drive drug development efforts.
Collapse
Affiliation(s)
- Michael Y. Sherman
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, Tel 617-638-5971, Fax 617-638-5339
| | - Vladimir L. Gabai
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, Tel 617-638-5971, Fax 617-638-5339
| |
Collapse
|
24
|
Proia DA, Kaufmann GF. Targeting Heat-Shock Protein 90 (HSP90) as a Complementary Strategy to Immune Checkpoint Blockade for Cancer Therapy. Cancer Immunol Res 2015; 3:583-9. [PMID: 25948551 DOI: 10.1158/2326-6066.cir-15-0057] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 03/16/2015] [Indexed: 11/16/2022]
Abstract
The demonstration that immune checkpoint blockade can meaningfully improve outcomes for cancer patients has revolutionized the field of immuno-oncology. New biologic agents targeting specific checkpoints have shown remarkable durability in terms of patient response and, importantly, exhibit clinical activity across a range of human malignancies, including many that have traditionally proven refractory to other immunotherapies. In this rapidly evolving area, a key consideration relates to the identification of novel combinatorial strategies that exploit existing or investigational cancer therapies in order to optimize patient outcomes and the proportion of individuals able to derive benefit from this approach. In this regard, heat-shock protein 90 (HSP90) represents an important emerging target for cancer therapy because its inactivation results in the simultaneous blockade of multiple signaling pathways and can sensitize tumor cells to other anticancer agents. Within the context of immunology, HSP90 plays a dual regulatory role, with its functional inhibition resulting in both immunosuppressive and immunostimulatory effects. In this Cancer Immunology at the Crossroads overview, the anticancer activity profile of targeted HSP90 inhibitors is discussed along with their paradoxical roles in immunology. Overall, we explore the rationale for combining the modalities of HSP90 inhibition and immune checkpoint blockade in order to augment the antitumor immune response in cancer.
Collapse
Affiliation(s)
- David A Proia
- Synta Pharmaceuticals Corporation, Lexington, Massachusetts.
| | | |
Collapse
|
25
|
Land WG. The Role of Damage-Associated Molecular Patterns (DAMPs) in Human Diseases: Part II: DAMPs as diagnostics, prognostics and therapeutics in clinical medicine. Sultan Qaboos Univ Med J 2015; 15:e157-e170. [PMID: 26052447 PMCID: PMC4450777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/05/2014] [Accepted: 10/30/2014] [Indexed: 06/04/2023] Open
Abstract
This article is the second part of a review that addresses the role of damage-associated molecular patterns (DAMPs) in human diseases by presenting examples of traumatic (systemic inflammatory response syndrome), cardiovascular (myocardial infarction), metabolic (type 2 diabetes mellitus), neurodegenerative (Alzheimer's disease), malignant and infectious diseases. Various DAMPs are involved in the pathogenesis of all these diseases as they activate innate immune machineries including the unfolded protein response and inflammasomes. These subsequently promote sterile autoinflammation accompanied, at least in part, by subsequent adaptive autoimmune processes. This review article discusses the future role of DAMPs in routine practical medicine by highlighting the possibility of harnessing and deploying DAMPs either as biomarkers for the appropriate diagnosis and prognosis of diseases, as therapeutics in the treatment of tumours or as vaccine adjuncts for the prophylaxis of infections. In addition, this article examines the potential for developing strategies aimed at mitigating DAMPs-mediated hyperinflammatory responses, such as those seen in systemic inflammatory response syndrome associated with multiple organ failure.
Collapse
Affiliation(s)
- Walter G Land
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, LabEx Transplantex, Centre de Recherche d'Immunologie et d'Hématologie, Université de Strasbourg, Strasbourg, France, E-mail:
| |
Collapse
|