1
|
Hoover AR, Liu K, Furrer C, Lam SSK, Anderson DW, Zhou Z, Yang J, Wong CF, Medcalf AD, Sun XH, Hode T, Alleruzzo L, Delawder A, Raker J, Abousleiman G, Valerio TI, Sun Y, Papin JF, Li M, Chen WR. N-Dihydrogalactochitosan Drives Conventional and Alternative Activations of STING to Synergize Type I IFN and IL-1β Productions for Antitumor Immunity. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2410079. [PMID: 39896882 PMCID: PMC11784597 DOI: 10.1002/adfm.202410079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Indexed: 02/04/2025]
Abstract
N-dihydrogalactochitosan (GC) is an immune stimulant/adjuvant. Synthesized from chitosan and galactose, GC is a new chemical entity that significantly enhances the immune-stimulating properties of its parental material, chitosan, making it a promising therapeutic agent. When used in combination with antigenic material, GC stimulates innate and adaptive antitumor and antiviral immunities. However, its mechanism has not been fully investigated. Herein we demonstrate that GC drives type I IFN activation in antigen-presenting cells (APCs). More importantly, GC drives alternative STING pathways, leading to inflammatory cell death that enhances dendritic cell (DC) activation. GC-activated DCs trigger a variety of nucleic acid sensing pattern recognition receptors (PRRs) pathways and IL-1β production via the activation of the inflammasome. In vivo, GC induces a potent response of type I IFNs and upregulates genes associated with STING signaling within the tumor microenvironment (TME). Moreover, intratumoral delivery of GC reduces the numbers of M2-like macrophages and increases M1-like macrophages residing within the TME, while subsequently increasing the number of activated DCs. Our findings demonstrate that GC acts as a multimodal immune stimulant via STING to generate a broad type I IFN response. This uniquely broad response holds therapeutic promise in generating enhanced antitumor and antiviral immunities.
Collapse
Affiliation(s)
- Ashley R. Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Coline Furrer
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | | | | | - Zhijun Zhou
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jingxuan Yang
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | - Alexandra D. Medcalf
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Xiao-Hong Sun
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Tomas Hode
- Immunophotonics, Inc., Saint Louis, MO, USA
| | | | | | | | - Ghainaa Abousleiman
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Trisha I. Valerio
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Yuanhong Sun
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - James F. Papin
- Department Pathology and Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Min Li
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Wei R. Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
2
|
Xu S, Hu E, Cai Y, Xie Z, Luo X, Zhan L, Tang W, Wang Q, Liu B, Wang R, Xie W, Wu T, Xie L, Yu G. Using clusterProfiler to characterize multiomics data. Nat Protoc 2024; 19:3292-3320. [PMID: 39019974 DOI: 10.1038/s41596-024-01020-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 05/13/2024] [Indexed: 07/19/2024]
Abstract
With the advent of multiomics, software capable of multidimensional enrichment analysis has become increasingly crucial for uncovering gene set variations in biological processes and disease pathways. This is essential for elucidating disease mechanisms and identifying potential therapeutic targets. clusterProfiler stands out for its comprehensive utilization of databases and advanced visualization features. Importantly, clusterProfiler supports various biological knowledge, including Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, through performing over-representation and gene set enrichment analyses. A key feature is that clusterProfiler allows users to choose from various graphical outputs to visualize results, enhancing interpretability. This protocol describes innovative ways in which clusterProfiler has been used for integrating metabolomics and metagenomics analyses, identifying and characterizing transcription factors under stress conditions, and annotating cells in single-cell studies. In all cases, the computational steps can be completed within ~2 min. clusterProfiler is released through the Bioconductor project and can be accessed via https://bioconductor.org/packages/clusterProfiler/ .
Collapse
Affiliation(s)
- Shuangbin Xu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Division of Laboratory Medicine, Microbiome Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Erqiang Hu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yantong Cai
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Zijing Xie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao Luo
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Li Zhan
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wenli Tang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qianwen Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bingdong Liu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Rui Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wenqin Xie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tianzhi Wu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Guangchuang Yu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
- Division of Laboratory Medicine, Microbiome Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
- Dermatology Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Li Y, Lam SSK, Wong CF, Hode T, Anderson D, Martin RCG. Thermal ablation enhances immunotherapeutic effect of IP-001 on orthotopic liver cancer in a rat model. Int J Hyperthermia 2024; 41:2413591. [PMID: 39389594 DOI: 10.1080/02656736.2024.2413591] [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: 06/10/2024] [Revised: 09/18/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Thermal ablation is reported to increase immunogenicity in tumor cells via expressing tumor antigens. IP-001, a synthesized molecule, is created by attaching galactose molecules to the free amino groups of partially deacetylated glucosamine polymers. As a member of a new class of polycationic immunoadjuvants that activate multiple immune response pathways, IP-001 can both sequester ablation-released tumor antigens in situ and independently recruit and stimulate antigen presenting cells (APCs) to induce a potent tumor-specific Th1 type T cell response. METHODS An orthotopic HCC rat model is established by implantation of 5 × 106 N1-S1 cells into the left lobe of liver. When tumor size reached 1.0-1.5 cm3, the animals were divided randomly into 4 groups, (1) MWA+IP-001; (2) MWA+saline; (3) sham MWA+IP-001 and (4) sham MWA+saline (n = 5 each group). RESULTS IP001 + MWA treatment significantly suppressed tumor growth in comparison to the other 3 groups. Significantly increased infiltration of inflammatory/immune cells were found in the tumor adjacent tissues of MWA+IP-001 mice, compared to the other 3 groups. Flow cytometry results indicated that there were significant increases of cytotoxic T cells, macrophages, dendritic cells and NK cell in the combination of MWA and IP001 treated mice, compared to other 3 groups (p < 0.01). Significantly decreased number of Treg cells were found in all the treatment arms compared to untreated control (p < 0.01). CONCLUSION Combination of MWA and IP001 enhances tumor suppression in an orthotopic HCC rat model. The tumor suppression is associated to the enhanced immune responses in terms of recruiting the important cell subpopulations such as CD8 + T-cells and NK cells into tumor microenvironment and abolishing immune suppressor such as Treg cells.
Collapse
Affiliation(s)
- Yan Li
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY, USA
| | | | | | | | | | - Robert C G Martin
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
4
|
Shi L, Zhang F, Yan J, Luo M, Liu K, Liu P, Yan G, Li C, Yang Y, Zeng Q, Zhang G, Chen WR, Wang X. Photothermal therapy improves the efficacy of topical immunotherapy against melanoma. Photodiagnosis Photodyn Ther 2024; 49:104290. [PMID: 39067671 DOI: 10.1016/j.pdpdt.2024.104290] [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: 05/08/2024] [Revised: 07/03/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Melanoma is an aggressive cancer with poor response to traditional therapies. A combination of photothermal therapy and topical immunotherapy may enhance elimination of melanoma.. MATERIALS AND METHODS C57BL/6 mice with early stage and metastatic melanoma were treated with laser immunotherapy (LIT), combining near-infrared laser-based photothermal therapy (PTT) and topical imiquimod (IMQ)-based immunotherapy. The volume of primary and abscopal melanoma, animal survival, tissue temperature, transcriptome, and immune cell response were investigated to evaluate the effect of LIT. RESULTS LIT could eliminate primary tumors, inhibite abscopal tumors, and prolong animal survival. The tumor tissues were selectively destroyed under a photothermal gradient between 38.2 ± 3.7 °C and 73.0 ± 2.3 °C. Gene expression analysis showed a significant increase in the expression of damage associated molecular patterns. Additionally, the population of mature dendritic cells, CD4+ T cells, and CD8+ T cells were increased, while myeloid-derived suppressor cells were downregulated after LIT. CONCLUSION The study showed that LIT inhibited the growth of both primary and abscopal melanoma by activating systemic antitumor immune responses and reversing the immunosuppressive tumor microenvironment, making LIT a potential method for advanced melanoma treatment.
Collapse
Affiliation(s)
- Lei Shi
- Department of Dermatology, Huadong Hospital, Fudan University, Shanghai 200040, PR China
| | - Fuhe Zhang
- Institute of Photomedicine, School of Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200092, PR China
| | - Jia Yan
- Institute of Photomedicine, School of Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200092, PR China
| | - Min Luo
- Institute of Photomedicine, School of Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200092, PR China
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Pei Liu
- Institute of Photomedicine, School of Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200092, PR China
| | - Guorong Yan
- Institute of Photomedicine, School of Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200092, PR China
| | - Chunxiao Li
- Institute of Photomedicine, School of Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200092, PR China
| | - Yutong Yang
- Institute of Photomedicine, School of Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200092, PR China
| | - Qingyu Zeng
- Institute of Photomedicine, School of Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200092, PR China
| | - Guolong Zhang
- Institute of Photomedicine, School of Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200092, PR China
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA.
| | - Xiuli Wang
- Institute of Photomedicine, School of Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai 200092, PR China.
| |
Collapse
|
5
|
Chen Y, Shen J, Ling C, Liang Z, Huang S, Lin W, Qin Y, Meng L, Luo Y. Exploring the role of CD8 + T cells in clear renal cell carcinoma metastasis. FEBS Open Bio 2024; 14:1205-1217. [PMID: 38872260 PMCID: PMC11216920 DOI: 10.1002/2211-5463.13819] [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: 08/23/2023] [Revised: 04/04/2024] [Accepted: 05/03/2024] [Indexed: 06/15/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) accounts for approximately 75-80% of all patients with renal cell carcinoma. Despite its prevalence, little is known regarding the key components involved in ccRCC metastasis. In this study, scRNA-seq analysis was employed to classify CD8+ T cells into four sub-clusters based on their genetic profiles and immunofluorescence experiments were used to validate two key clusters. Through gene set enrichment analysis, these newly identified sub-clusters were found to exhibit distinct biological characteristics. Notably, TYMP, TOP2A, CHI3L2, CDKN3, CENPM, and RZH2 were highly expressed in these sub-clusters, indicating a correlation with poor prognosis. Among these sub-clusters, CD8+ T cells (MT-ND4) were identified as potentially playing a critical role in mediating ccRCC metastasis. These results contribute to our understanding of CD8+ T cell heterogeneity in ccRCC and shed light on the mechanisms underlying the loss of immune response against cancer.
Collapse
Affiliation(s)
- Yuanhong Chen
- Center for Systemic Inflammation Research (CSIR), School of Preclinical MedicineYoujiang Medical University for NationalitiesBaiseChina
- Department of Pathogenic Biology and ImmunologyYoujiang Medical University for NationalitiesBaiseChina
| | - Jiajia Shen
- Center for Systemic Inflammation Research (CSIR), School of Preclinical MedicineYoujiang Medical University for NationalitiesBaiseChina
| | - Caixia Ling
- Modern Industrial College of Biomedicine and Great HealthYoujiang Medical University for NationalitiesBaiseChina
| | - Zhengfang Liang
- Department of Urinary SurgeryThe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseChina
| | - Shaoang Huang
- Center for Systemic Inflammation Research (CSIR), School of Preclinical MedicineYoujiang Medical University for NationalitiesBaiseChina
| | - Wenxian Lin
- Center for Systemic Inflammation Research (CSIR), School of Preclinical MedicineYoujiang Medical University for NationalitiesBaiseChina
- Department of Interventional OncologyAffiliated Hospital of Youjiang Medical College for NationalitiesBaiseChina
| | - Yujuan Qin
- Center for Systemic Inflammation Research (CSIR), School of Preclinical MedicineYoujiang Medical University for NationalitiesBaiseChina
| | - Lingzhang Meng
- Center for Systemic Inflammation Research (CSIR), School of Preclinical MedicineYoujiang Medical University for NationalitiesBaiseChina
- Institute of Cardiovascular SciencesGuangxi Academy of Medical SciencesNanningChina
| | - Yanhong Luo
- Center for Systemic Inflammation Research (CSIR), School of Preclinical MedicineYoujiang Medical University for NationalitiesBaiseChina
- Modern Industrial College of Biomedicine and Great HealthYoujiang Medical University for NationalitiesBaiseChina
| |
Collapse
|
6
|
Liu P, Wei Z, Ye X. Immunostimulatory effects of thermal ablation: Challenges and future prospects. J Cancer Res Ther 2024; 20:531-539. [PMID: 38687922 DOI: 10.4103/jcrt.jcrt_2484_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 12/13/2023] [Indexed: 05/02/2024]
Abstract
ABSTRACT This literature explores the immunostimulatory effects of thermal ablation in the tumor microenvironment, elucidating the mechanisms such as immunogenic cell death, tumor-specific antigens, and damage-associated molecular patterns. Furthermore, it outlines critical issues associated with thermal ablation-induced immunostimulatory challenges and offers insights into future research avenues and potential therapeutic strategies.
Collapse
Affiliation(s)
- Peng Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, No. 16766 Jingshi Road, Jinan, Shandong Province, China
| | | | | |
Collapse
|
7
|
Hoover AR, More S, Liu K, West CL, Valerio TI, Furrer CL, Adams JP, Yu N, Villalva C, Kumar A, Alleruzzo L, Lam SSK, Hode T, Papin JF, Chen WR. N-dihydrogalactochitosan serves as an effective mucosal adjuvant for intranasal vaccine in combination with recombinant viral proteins against respiratory infection. Acta Biomater 2024; 175:279-292. [PMID: 38160856 DOI: 10.1016/j.actbio.2023.12.039] [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: 04/18/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
Mucosal vaccinations for respiratory pathogens provide effective protection as they stimulate localized cellular and humoral immunities at the site of infection. Currently, the major limitation of intranasal vaccination is using effective adjuvants capable of withstanding the harsh environment imposed by the mucosa. Herein, we describe the efficacy of using a unique biopolymer, N-dihydrogalactochitosan (GC), as a nasal mucosal vaccine adjuvant against respiratory infections. Specifically, we mixed GC with recombinant SARS-CoV-2 trimeric spike (S) and nucleocapsid (NC) proteins to intranasally vaccinate K18-hACE2 transgenic mice, in comparison with Addavax (AV), an MF-59 equivalent. In contrast to AV, intranasal application of GC induces a robust, systemic antigen-specific antibody response and increases the number of T cells in the cervical lymph nodes. Moreover, GC+S+NC-vaccinated animals were largely resistant to the lethal SARS-CoV-2 challenge and experienced drastically reduced morbidity and mortality, with animal weights and behavior returning to normal 22 days post-infection. In contrast, animals intranasally vaccinated with AV+S+NC experienced severe weight loss, mortality, and respiratory distress, with none surviving beyond 6 days post-infection. Our findings demonstrate that GC can serve as a potent mucosal vaccine adjuvant against SARS-CoV-2 and potentially other respiratory viruses. STATEMENT OF SIGNIFICANCE: We demonstrated that a unique biopolymer, N-dihydrogalactochitosan (GC), was an effective nasal mucosal vaccine adjuvant against respiratory infections. Specifically, we mixed GC with recombinant SARS-CoV-2 trimeric spike (S) and nucleocapsid (NC) proteins to intranasally vaccinate K18-hACE2 transgenic mice, in comparison with Addavax (AV). In contrast to AV, GC induces a robust, systemic antigen-specific antibody response and increases the number of T cells in the cervical lymph nodes. About 90 % of the GC+S+NC-vaccinated animals survived the lethal SARS-CoV-2 challenge and remained healthy 22 days post-infection, while the AV+S+NC-vaccinated animals experienced severe weight loss and respiratory distress, and all died within 6 days post-infection. Our findings demonstrate that GC is a potent mucosal vaccine adjuvant against SARS-CoV-2 and potentially other respiratory viruses.
Collapse
Affiliation(s)
- Ashley R Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA; Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sunil More
- Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK USA
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Connor L West
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Trisha I Valerio
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Coline L Furrer
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Jacob P Adams
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Ningli Yu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Crystal Villalva
- Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK USA
| | - Amit Kumar
- Biogen Inc., 225 Bnney Street, Cambridge, MA, USA
| | - Lu Alleruzzo
- Immunophotonics, Inc., 4340 Duncan Avenue, Suite 212, Saint Louis, MO, USA
| | - Samuel S K Lam
- Immunophotonics, Inc., 4340 Duncan Avenue, Suite 212, Saint Louis, MO, USA
| | - Tomas Hode
- Immunophotonics, Inc., 4340 Duncan Avenue, Suite 212, Saint Louis, MO, USA
| | - James F Papin
- Department Pathology and Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA.
| |
Collapse
|
8
|
Xia H, Zhu J, Zheng Z, Xiao P, Yu X, Wu M, Xue L, Xu X, Wang X, Guo Y, Zheng C, Ding S, Wang Y, Peng X, Fu S, Li J, Deng X. Amino acids and their roles in tumor immunotherapy of breast cancer. J Gene Med 2024; 26:e3647. [PMID: 38084655 DOI: 10.1002/jgm.3647] [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: 06/11/2023] [Revised: 10/20/2023] [Accepted: 11/13/2023] [Indexed: 01/30/2024] Open
Abstract
Breast cancer is the most commonly diagnosed cancer among women. The primary treatment options include surgery, radiotherapy, chemotherapy, targeted therapy and hormone therapy. The effectiveness of breast cancer therapy varies depending on the stage and aggressiveness of the cancer, as well as individual factors. Advances in early detection and improved treatments have significantly increased survival rates for breast cancer patients. Nevertheless, specific subtypes of breast cancer, particularly triple-negative breast cancer, still lack effective treatment strategies. Thus, novel and effective therapeutic targets for breast cancer need to be explored. As substrates of protein synthesis, amino acids are important sources of energy and nutrition, only secondly to glucose. The rich supply of amino acids enables the tumor to maintain its proliferative competence through participation in energy generation, nucleoside synthesis and maintenance of cellular redox balance. Amino acids also play an important role in immune-suppressive microenvironment formation. Thus, the biological effects of amino acids may change unexpectedly in tumor-specific or oncogene-dependent manners. In recent years, there has been significant progress in the study of amino acid metabolism, particularly in their potential application as therapeutic targets in breast cancer. In this review, we provide an update on amino acid metabolism and discuss the therapeutic implications of amino acids in breast cancer.
Collapse
Affiliation(s)
- Hongzhuo Xia
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
| | - Jianyu Zhu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
- Department of Pathophysiology, Jishou University, Jishou, Hunan, China
| | - Zhuomeng Zheng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
| | - Peiyao Xiao
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
| | - Xiaohui Yu
- Department of Pathology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Muyao Wu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
| | - Lian Xue
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
| | - Xi Xu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
| | - Xinyu Wang
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
| | - Yuxuan Guo
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
| | - Chanjuan Zheng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
| | - Siyu Ding
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
| | - Yian Wang
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
| | - Xiaoning Peng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
- Department of Pathophysiology, Jishou University, Jishou, Hunan, China
| | - Shujun Fu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
| | - Junjun Li
- Department of Pathology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiyun Deng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Pathophysiology, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
9
|
Liu K, Sadeghipour N, Hoover AR, Valero TI, Furrer C, Adams J, Naqash AR, Zhao M, Papin JF, Chen WR. Single-cell transcriptomics reveals that tumor-infiltrating natural killer cells are activated by localized ablative immunotherapy and share anti-tumor signatures induced by immune checkpoint inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539163. [PMID: 37205468 PMCID: PMC10187236 DOI: 10.1101/2023.05.02.539163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Rationale Natural killer (NK) cells provide protective anti-cancer immunity. However, the cancer therapy induced activation gene signatures and pathways in NK cells remain unclear. Methods We applied a novel localized ablative immunotherapy (LAIT) by synergizing photothermal therapy (PTT) with intra-tumor delivering of the immunostimulant N-dihydrogalactochitosan (GC), to treat breast cancer using a mammary tumor virus-polyoma middle tumor-antigen (MMTV-PyMT) mouse model. We performed single-cell RNA sequencing (scRNAseq) analysis to unveil the cellular heterogeneity and compare the transcriptional alterations induced by PTT, GC, and LAIT in NK cells within the tumor microenvironment (TME). Results ScRNAseq showed that NK subtypes, including cycling, activated, interferon-stimulated, and cytotoxic NK cells. Trajectory analysis revealed a route toward activation and cytotoxicity following pseudotime progression. Both GC and LAIT elevated gene expression associated with NK cell activation, cytolytic effectors, activating receptors, IFN pathway components, and cytokines/chemokines in NK subtypes. Single-cell transcriptomics analysis using immune checkpoint inhibitor (ICI)-treated animal and human samples revealed that ICI-induced NK activation and cytotoxicity across several cancer types. Furthermore, ICI-induced NK gene signatures were also induced by LAIT treatment. We also discovered that several types of cancer patients had significantly longer overall survival when they had higher expression of genes in NK cells that were also specifically upregulated by LAIT. Conclusion Our findings show for the first time that LAIT activates cytotoxicity in NK cells and the upregulated genes positively correlate with beneficial clinical outcomes for cancer patients. More importantly, our results further establish the correlation between the effects of LAIT and ICI on NK cells, hence expanding our understanding of mechanism of LAIT in remodeling TME and shedding light on the potentials of NK cell activation and anti-tumor cytotoxic functions in clinical applications.
Collapse
|
10
|
Valerio TI, Furrer CL, Sadeghipour N, Patrock SJX, Tillery SA, Hoover AR, Liu K, Chen WR. Immune modulations of the tumor microenvironment in response to phototherapy. JOURNAL OF INNOVATIVE OPTICAL HEALTH SCIENCES 2023; 16:2330007. [PMID: 38550850 PMCID: PMC10976517 DOI: 10.1142/s1793545823300070] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/01/2024]
Abstract
The tumor microenvironment (TME) promotes pro-tumor and anti-inflammatory metabolisms and suppresses the host immune system. It prevents immune cells from fighting against cancer effectively, resulting in limited efficacy of many current cancer treatment modalities. Different therapies aim to overcome the immunosuppressive TME by combining various approaches to synergize their effects for enhanced anti-tumor activity and augmented stimulation of the immune system. Immunotherapy has become a major therapeutic strategy because it unleashes the power of the immune system by activating, enhancing, and directing immune responses to prevent, control, and eliminate cancer. Phototherapy uses light irradiation to induce tumor cell death through photothermal, photochemical, and photo-immunological interactions. Phototherapy induces tumor immunogenic cell death, which is a precursor and enhancer for anti-tumor immunity. However, phototherapy alone has limited effects on long-term and systemic anti-tumor immune responses. Phototherapy can be combined with immunotherapy to improve the tumoricidal effect by killing target tumor cells, enhancing immune cell infiltration in tumors, and rewiring pathways in the TME from anti-inflammatory to pro-inflammatory. Phototherapy-enhanced immunotherapy triggers effective cooperation between innate and adaptive immunities, specifically targeting the tumor cells, whether they are localized or distant. Herein, the successes and limitations of phototherapy combined with other cancer treatment modalities will be discussed. Specifically, we will review the synergistic effects of phototherapy combined with different cancer therapies on tumor elimination and remodeling of the immunosuppressive TME. Overall, phototherapy, in combination with other therapeutic modalities, can establish anti-tumor pro-inflammatory phenotypes in activated tumor-infiltrating T cells and B cells and activate systemic anti-tumor immune responses.
Collapse
Affiliation(s)
- Trisha I. Valerio
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Coline L. Furrer
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Negar Sadeghipour
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
- School of Electrical and Computer Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Sophia-Joy X. Patrock
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Sayre A. Tillery
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Ashley R. Hoover
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Kaili Liu
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Wei R. Chen
- Stephenson School of Biomedical Engineering University of Oklahoma, Norman, Oklahoma 73019, USA
| |
Collapse
|
11
|
Wang S, Sun ST, Zhang XY, Ding HR, Yuan Y, He JJ, Wang MS, Yang B, Li YB. The Evolution of Single-Cell RNA Sequencing Technology and Application: Progress and Perspectives. Int J Mol Sci 2023; 24:ijms24032943. [PMID: 36769267 PMCID: PMC9918030 DOI: 10.3390/ijms24032943] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/01/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
As an emerging sequencing technology, single-cell RNA sequencing (scRNA-Seq) has become a powerful tool for describing cell subpopulation classification and cell heterogeneity by achieving high-throughput and multidimensional analysis of individual cells and circumventing the shortcomings of traditional sequencing for detecting the average transcript level of cell populations. It has been applied to life science and medicine research fields such as tracking dynamic cell differentiation, revealing sensitive effector cells, and key molecular events of diseases. This review focuses on the recent technological innovations in scRNA-Seq, highlighting the latest research results with scRNA-Seq as the core technology in frontier research areas such as embryology, histology, oncology, and immunology. In addition, this review outlines the prospects for its innovative application in traditional Chinese medicine (TCM) research and discusses the key issues currently being addressed by scRNA-Seq and its great potential for exploring disease diagnostic targets and uncovering drug therapeutic targets in combination with multiomics technologies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Bin Yang
- Correspondence: (B.Y.); (Y.-B.L.)
| | - Yu-Bo Li
- Correspondence: (B.Y.); (Y.-B.L.)
| |
Collapse
|
12
|
Hoover AR, Kaabinejadian S, Krawic JR, Sun XH, Naqash AR, Yin Q, Yang X, Christopher Garcia K, Davis MM, Hildebrand WH, Chen WR. Localized ablative immunotherapy drives de novo CD8 + T-cell responses to poorly immunogenic tumors. J Immunother Cancer 2022; 10:e004973. [PMID: 36253002 PMCID: PMC9577935 DOI: 10.1136/jitc-2022-004973] [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] [Accepted: 09/17/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Localized ablative immunotherapies hold great promise in stimulating antitumor immunity to treat metastatic and poorly immunogenic tumors. Tumor ablation is well known to release tumor antigens and danger-associated molecular patterns to stimulate T-cell immunity, but its immune stimulating effect is limited, particularly against metastatic tumors. METHODS In this study, we combined photothermal therapy with a potent immune stimulant, N-dihydrogalactochitosan, to create a local ablative immunotherapy which we refer to as laser immunotherapy (LIT). Mice bearing B16-F10 tumors were treated with LIT when the tumors reached 0.5 cm3 and were monitored for survival, T-cell activation, and the ability to resist tumor rechallenge. RESULTS We found that LIT stimulated a stronger and more consistent antitumor T-cell response to the immunologically 'cold' B16-F10 melanoma tumors and conferred a long-term antitumor memory on tumor rechallenge. Furthermore, we discovered that LIT generated de novo CD8+ T-cell responses that strongly correlated with animal survival and tumor rejection. CONCLUSION In summary, our findings demonstrate that LIT enhances the activation of T cells and drives de novo antitumor T-cell responses. The data presented herein suggests that localized ablative immunotherapies have great potential to synergize with immune checkpoint therapies to enhance its efficacy, resulting in improved antitumor immunity.
Collapse
Affiliation(s)
- Ashley R Hoover
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
| | - Saghar Kaabinejadian
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Jason R Krawic
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Xiao-Hong Sun
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Abdul Rafeh Naqash
- Medical Oncology/TSET Phase 1 Program, The University of Oklahoma Stephenson Cancer Center, Oklahoma City, Oklahoma, USA
| | - Qian Yin
- Institute for Immunity, Stanford University School of Medicine, Stanford, California, USA
| | - Xinbo Yang
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, California, USA
| | - K Christopher Garcia
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - William H Hildebrand
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|