1
|
Jiang KC, Zhu YH, Jiang ZL, Liu Y, Hussain W, Luo HY, Sun WH, Ji XY, Li DX. Regulation of PEST-containing nuclear proteins in cancer cells: implications for cancer biology and therapy. Front Oncol 2025; 15:1548886. [PMID: 40330830 PMCID: PMC12052563 DOI: 10.3389/fonc.2025.1548886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
The PEST-containing nuclear protein (PCNP) is a nuclear protein involved in the regulation of cell cycle progression, protein degradation, and tumorigenesis. PCNP contains a PEST sequence, a polypeptide structural motif rich in proline (P), glutamic acid (E), serine (S), and threonine (T), which serves as a proteolytic recognition signal. The degradation of specific proteins via the PEST sequence plays a crucial role in modulating signaling pathways that control cell growth, differentiation, apoptosis, and stress responses. PCNP is primarily degraded through the ubiquitin-proteasome system (UPS) and the calpain pathway, with phosphorylation of threonine and serine residues further accelerating its degradation. The ubiquitination of PCNP by the ring finger protein NIRF in an E3 ligase-dependent manner is well documented, along with its involvement in the MAPK and PI3K/AKT/mTOR signaling pathways. Additionally, PCNP is implicated in p53-mediated cell cycle arrest and apoptosis, which are essential for inhibiting tumor growth. To explore the role of PCNP in cancer, this review examines its effects on cell growth, differentiation, proliferation, and apoptosis in lung adenocarcinoma, thyroid cancer, ovarian cancer, and other malignancies derived from glandular epithelial cells. By focusing on PCNP and its regulatory mechanisms, this study provides a scientific basis for further research on the biological functions of the PEST sequence in tumor development and cancer progression.
Collapse
Affiliation(s)
- Kai-Chun Jiang
- Department of Traditional Chinese Medicine, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, China
| | - Yong-Hao Zhu
- School of Stomatology, Henan University, Kaifeng, Henan, China
| | - Zhi-Liang Jiang
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Urology, Institute of Urology, Sichuan University, Chengdu, China
| | - Yi Liu
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Wahab Hussain
- School of Stomatology, Henan University, Kaifeng, Henan, China
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
| | - Huang-Yin Luo
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Urology, Institute of Urology, Sichuan University, Chengdu, China
| | - Wei-Hang Sun
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Urology, Institute of Urology, Sichuan University, Chengdu, China
| | - Xin-Ying Ji
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Oncology, Huaxian County Hospital, Anyang, Henan, China
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, China
| | - Ding-Xi Li
- The Affiliated Cancer Hospital, Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
2
|
Luanpitpong S, Janan M, Poohadsuan J, Rodboon N, Samart P, Rungarunlert S, Issaragrisil S. A High-Throughput, Three-Dimensional Multiple Myeloma Model Recapitulating Tumor-Stroma Interactions for CAR-Immune Cell-Mediated Cytotoxicity Assay. Immunotargets Ther 2025; 14:321-338. [PMID: 40182067 PMCID: PMC11967349 DOI: 10.2147/itt.s503984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
Background Multiple myeloma (MM) is characterized by an excessive proliferation of clonal plasma cells in the bone marrow (BM). Components in BM niche contribute to the immunosuppressive tumor microenvironment (TME), but three-dimensional (3D) MM models that recreate the complex TME and enable high-throughput cytotoxicity assay of chimeric antigen receptor (CAR)-engineered immune cells are still lacking. Methods Stable, luciferase (Luc)-labeled target MM cells were generated using Luc/RFP dual reporter system to track MM growth. 3D spheroids were formed in a 96-well plate in the presence or absence of cancer-associated fibroblast (CAF)-like stromal cells activated by MM-derived conditioned medium and the cytotoxicity of CAR-immune cells, which were represented by third-generation anti-CD138 CAR-NK-92 cells, was evaluated by luciferase assay using a multimode microplate reader. Immune cell infiltration was visualized under a fluorescence microscope by using multiple fluorescent dyes. Results We first showed that luciferase assay provides a relatively simple and robust means to specifically monitor Luc-labeled tumor cell growth in a coculture system, allowing the high-throughput assessment of CAR-immune cytotoxicity. Through this assay, we demonstrated that CAF-like stromal cells impaired NK cell effector function in 2D culture and 3D spheroids, likely via paracrine signaling and physical barrier function. Importantly, we showed that 3D spheroids consisting of MM cells and CAF-like stromal cells provide a more comprehensive, physiologically relevant immuno-oncology model. Our established model could also be used to investigate the trafficking and infiltration of immune cells into the core of spheroids. Herein, we showed that CAR incorporation did improve the ability of NK cells to infiltrate 3D spheroids. Conclusion Our established 3D spheroid model, which partially recapitulates the complex TME with immunosuppressive environment, is suitable for high-throughput screening of CAR-immune cytotoxicity and could be important in accelerating immuno-oncology drug discovery for MM since there is a pressing need to establish innovative CAR-immune cells.
Collapse
Affiliation(s)
- Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Cell Factory for Cancer Immunotherapy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Montira Janan
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Cell Factory for Cancer Immunotherapy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jirarat Poohadsuan
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Napachai Rodboon
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Parinya Samart
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sasitorn Rungarunlert
- Department of Preclinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- BDMS Center of Excellence for Hematology, Wattanosoth Cancer Hospital, Bangkok, Thailand
| |
Collapse
|
3
|
Wang B, Yin Y, Wang A, Liu W, Chen J, Li T. SMR-guided molecular subtyping and machine learning model reveals novel prognostic biomarkers and therapeutic targets in non-small cell lung adenocarcinoma. Sci Rep 2025; 15:1640. [PMID: 39794414 PMCID: PMC11723915 DOI: 10.1038/s41598-025-85471-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Non-small cell lung adenocarcinoma (LUAD) is a markedly heterogeneous disease, with its underlying molecular mechanisms and prognosis prediction presenting ongoing challenges. In this study, we integrated data from multiple public datasets, including TCGA, GSE31210, and GSE13213, encompassing a total of 867 tumor samples. By employing Mendelian randomization (MR) analysis, machine learning techniques, and comprehensive bioinformatics approaches, we conducted an in-depth investigation into the molecular characteristics, prognostic markers, and potential therapeutic targets of LUAD. Our analysis identified 321 genes significantly associated with LUAD, with CENP-A, MCM7, and DLGAP5 emerging as highly connected nodes in network analyses. By performing correlation analysis and Cox regression analysis, we identified 26 prognostic genes and classified LUAD samples into two molecular subtypes with significantly distinct survival outcomes. The Random Survival Forest (RSF) model exhibited robust prognostic predictive capabilities across multiple independent cohorts (AUC > 0.75). Beyond merely predicting patient outcomes, this model also captures key features of the tumor immune microenvironment and potential therapeutic responses. Functional enrichment analysis revealed the complex interplay of cell cycle regulation, DNA repair, immune response, and metabolic reprogramming in the progression of LUAD. Furthermore, we observed a strong correlation between risk scores and the expression of specific cytokines, such as CCL17, CCR2, and CCL20, suggesting novel avenues for developing cytokine network-based therapeutic strategies. This study offers fresh insights into the molecular subtyping, prognostic prediction, and personalized therapeutic decision-making in LUAD, laying a critical foundation for future clinical applications and targeted therapy research.
Collapse
Affiliation(s)
- Baozhen Wang
- School of Clinical Medicine, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- Department of Surgical Oncology II, The General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia, 750004, China
| | - Yichen Yin
- School of Clinical Medicine, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- Department of Surgical Oncology II, The General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia, 750004, China
| | - Anqi Wang
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
| | - Weidi Liu
- School of Clinical Medicine, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China
- Department of Surgical Oncology II, The General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia, 750004, China
| | - Jing Chen
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China.
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia, 750004, China.
| | - Tao Li
- Department of Surgical Oncology II, The General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan, Ningxia, 750004, China.
| |
Collapse
|
4
|
Zheng S, Feng J, Chen Z, Wei C, Pan Y, Liu J. The Impact of Socioeconomic Status on the Incidence and Stage of Melanoma in China: A Single-Center Observational Study. Ann Plast Surg 2024; 93:S59-S63. [PMID: 38718342 DOI: 10.1097/sap.0000000000003925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
BACKGROUND The role of high socioeconomic status (SES) as an established risk factor for melanoma has been well documented in Western countries and regions. However, research on the association between melanoma and SES in China remains limited. This study aimed to investigate the association between SES and melanoma incidence and stage in China. METHODS Five measures of SES were accessed, including education level, ethnic background, per capita household income, occupation, and medical insurance coverage. A scoring system based on the Kuppuswamy Socio-Economic Scale was used to create a quantitative assessment of SES. To improve clarity and precision, we refined the language in the original text. Clinical stage at diagnosis was classified according to the Chinese Society Oncology Melanoma Guidelines. RESULTS A total of 122 patients with pathologic melanoma were enrolled in this study from January 2013 to December 2017. Of these patients, 58 (48%) were male and 64 (52%) were female, with a mean age of 59.23 ± 9.91 years. Patients in the age groups of 45-59 and 60-73 had a higher incidence of melanoma compared to other age groups. Acral lentiginous melanoma was the most commonly observed subtype, accounting for 48% of cases. Patients with a low level of education (middle school and below) and a low level of monthly household income (<3000 CNY) had a higher risk of developing melanoma, as did those who were unemployed. Interestingly, a higher proportion of melanoma diagnoses were made in patients with medical insurance than those without. However, no significant differences in melanoma staging were found based on education level ( P = 0.153), monthly household income ( P = 0.507), occupation ( P = 0.687), or insurance status ( P = 0.537). According to the Kuppuswamy Socio-Economic Scale, there were 0 in upper class, 50 in upper middle class, 44 in lower middle class, 28 in upper lower class, 0 in lower class. The mean K-score was 13.85. No statistically significant interaction was observed between K-score and tumor stage. CONCLUSIONS Patients with lower SES have a higher risk of developing melanoma. However, no significant differences were found in melanoma staging based on SES.
Collapse
Affiliation(s)
- Shaoluan Zheng
- From the Plastic and Reconstructive Surgery, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen
| | - Jia Feng
- Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai
| | - Zhiwei Chen
- Artificial Intelligence Center for Plastic Surgery and Cutaneous Soft Tissue Cancers, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuanyuan Wei
- Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai
| | - Yuyan Pan
- Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai
| | | |
Collapse
|
5
|
Yuan P, Long Y, Wei N, Wang Y, Zhu Z, Han J, Jiang D, Lan X, Gai Y. Peptide-based PET tracer targeting LAG-3 for evaluating the efficacy of immunotherapy in melanoma. J Immunother Cancer 2024; 12:e009010. [PMID: 39043603 DOI: 10.1136/jitc-2024-009010] [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] [Accepted: 07/12/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Lymphocyte activation gene 3 (LAG-3) is expressed on activated immune cells and has emerged as a promising target for immune checkpoints blockade. However, conflicting findings have been reported regarding the association between LAG-3 expression in tumors and patient prognosis, indicating the need for further investigation into the significance of LAG-3 expression levels in tumor therapies. In this study, 68Ga-NOTA-XH05, a novel peptide-based positron emission tomography (PET) tracer targeting LAG-3, was constructed to non-invasively detect LAG-3 expression in melanoma after CpG oligonucleotide (CpG) treatment and explore the relationship between LAG-3 expression and therapeutic effect. METHODS The tracer 68Ga-NOTA-XH05 was identified by high-performance liquid chromatography after being prepared and purified. Cell uptake and blocking essays were performed to verify the specificity of the tracer in vitro. The expression of LAG-3 in B16-F10 subcutaneous tumors was monitored by flow cytometry, and its correlation with the tracer uptake was analyzed to evaluate the tracer specificity. PET imaging and biodistribution studies were conducted after CpG treatment of unilateral or bilateral B16-F10 subcutaneous tumor models to assess the ability of 68Ga-NOTA-XH05 in monitoring immunotherapy efficacy and the abscopal effect of CpG. RESULTS Following purification, 68Ga-NOTA-XH05 exhibited high radiochemical purity and specificity. Flow cytometry analysis revealed a positive correlation between LAG-3 expression in tumors and the uptake of 68Ga-NOTA-XH05. In B16-F10 bearing mice treated with CpG, PET imaging using 68Ga-NOTA-XH05 demonstrated a higher tumor to blood ratio (TBR) compared with the control group. Furthermore, TBR values obtained from CpG-treated mice allowed for differentiation between responders and non-responders. In a bilateral subcutaneous tumor model where only right-sided tumors were treated with intratumoral injection of CpG, TBR values of left-sided tumors were significantly higher than those in the control group, indicating that 68Ga-NOTA-XH05 could effectively monitor the systemic effect of local CpG injection. CONCLUSION Our findings highlight the detection capability of 68Ga-NOTA-XH05 in assessing LAG-3 expression levels within tumors and evaluating response to immunotherapy, thereby suggesting promising clinical translational prospects.
Collapse
Affiliation(s)
- Peizhe Yuan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| | - Yu Long
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| | - Nannan Wei
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| | - Yan Wang
- Department of Oncology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyang Zhu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| | | | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, People's Republic of China
| |
Collapse
|
6
|
Tang M, Wang Y, Li P, Han R, Wang R. Assessing the benefits and safety profile of incorporating poly ADP-ribose polymerase (PARP) inhibitors in the treatment of advanced lung cancer: a thorough systematic review and meta-analysis. Front Pharmacol 2024; 15:1338442. [PMID: 38989152 PMCID: PMC11234112 DOI: 10.3389/fphar.2024.1338442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/29/2024] [Indexed: 07/12/2024] Open
Abstract
Background Poly (ADP-Ribose) Polymerase (PARP) inhibitors represent a novel class of drugs that hinder DNA repair mechanisms in tumor cells, leading to cell death. This systematic review aims to evaluate the effectiveness, safety, and potential adverse effects of PARP inhibitors (PARPi) in the management of patients with advanced lung cancer. Materials and Methods We conducted a comprehensive search for relevant studies in PubMed, Embase, Cochrane, and ClinicalTrials.gov. We extracted primary and secondary outcome measures, including progression-free survival (PFS), overall survival (OS), and adverse events (AEs), from the identified literature for subsequent meta-analysis and systematic review. Results This study encompassed twelve randomized controlled trials, involving 3,132 patients with advanced lung cancer. In comparison to non-PARPi treatments, the administration of PARPi significantly extended OS (hazard ratio (HR) = 0.90, 95% CI = 0.83-0.97, p = 0.006). However, the difference in PFS did not reach statistical significance. Conclusion In summary, therapies incorporating PARPi provide a degree of benefit by extending OS in patients with advanced lung cancer. Nonetheless, further trials are necessary to furnish additional evidence regarding the efficacy and safety of PARPi in the treatment of lung cancer. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO/, identifier number: CRD42023424673.
Collapse
Affiliation(s)
- Min Tang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yue Wang
- Department of Infectious Disease, Hefei Second People's Hospital, Hefei, China
| | - Pulin Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Rui Han
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ran Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
7
|
Wang J, Wang S, Zhang Y, Zhang W. Bibliometric analysis of evolutionary trajectory and prospective directions of LAG-3 in cancer. Front Immunol 2024; 15:1329775. [PMID: 38390331 PMCID: PMC10881671 DOI: 10.3389/fimmu.2024.1329775] [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: 10/29/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Objectives Perform a bibliometric analysis on the role of LAG-3 in the domain of cancer, elucidate the prevailing areas of research, and visually depict the evolutionary trajectory and prospective directions of LAG-3 research over the past twenty-three decades. Materials and methods Between 2000 and 2023, a comprehensive review of scholarly articles pertaining to LAG-3 research in the context of cancer was carried out using the Web of Science Core Collection (WoSCC) database. Bibliometric analysis can be conducted by taking advantage of VOSviewer (version 1.6.16) and CiteSpace (version 6.2.R4). Create a network diagram to visually represent various authors, countries, and organizations while assessing the publishing years, journals, references, and keywords. Results In conclusion, 1841 records were identified and published in 587 publications. These records were authored by 12,849 individuals affiliated with 2491 institutes across 74 countries. There has been a substantial surge in publications subsequent to 2013. The USA, China, and Germany gave the majority of records, amounting to 69.69%. American institutions actively engage in collaboration with institutions located in other countries. Triebel, F., Vignali, Dario A. A., Workman, Creg J. Drake, Charles G., and Elkord, Eyad are highly regarded authors in their respective fields. However, it is worth noting that Triebel exhibits limited collaboration with other writers. The examination of the role of LAG-3 in cancer and its potential for use in clinical settings is a discernible trend, as seen by keyword analysis. Conclusion The scientific interest in and attention towards LAG-3 has experienced a significant rise since 2013. The United States is leading the way, with China following closely behind. Promoting collaboration among writers, nations, and institutions with varied backgrounds is imperative. The discipline of immunotherapy is currently seeing ongoing progress. A thorough investigation of the distinctive cis ligand TCR-CD3 complex of LAG-3 and its signal transduction mechanism is necessary. Additionally, it is worthwhile to explore novel combinations of LAG-3 therapy.
Collapse
Affiliation(s)
| | | | | | - Wei Zhang
- Department of Breast Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
8
|
Patwekar M, Sehar N, Patwekar F, Medikeri A, Ali S, Aldossri RM, Rehman MU. Novel immune checkpoint targets: A promising therapy for cancer treatments. Int Immunopharmacol 2024; 126:111186. [PMID: 37979454 DOI: 10.1016/j.intimp.2023.111186] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/20/2023]
Abstract
The immune system frequently comprises immunological checkpoints. They serve as a barrier to keep the immune system from overreacting and damaging cells that are robust. Immune checkpoint inhibitors (ICIs) are utilized in immunotherapy to prevent the synergy of partner proteins of checkpoint proteins with auxiliary proteins. Moreover, the T cells may target malignant cells since the "off" signal cannot be conveyed. ICIs, which are mostly composed of monoclonal antibodies (mAbs) against cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and anti- programmed death-1/programmed ligand 1 (anti-PD-1/PD-L1), might transform the context of cancer therapy. Further, more patients continued to exhibit adaptive resistance, even though several ICIs demonstrated convincing therapeutic benefits in selective tumor types. Immune checkpoint therapy's overall effectiveness is still lacking at this time. A popular area of study involves investigating additional immune checkpoint molecules. Recent research has found a number of fresh immune checkpoint targets, including NKG2A ligands, TIGIT, B7-H6 ligands, Galectin 3, TIM3, and so on. These targets have been focus of the study, and recent investigational approaches have shown encouraging outcomes. In this review article, we covered the development and present level understanding of these recently identified immune checkpoint molecules, its effectiveness and limitations.
Collapse
Affiliation(s)
| | - Nouroz Sehar
- Centre for Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard University, New Delhi, 110062, India
| | - Faheem Patwekar
- Luqman College of Pharmacy, Gulbarga, 585102, Karnataka, India
| | | | - Shafat Ali
- Cytogenetics and Molecular Biology Laboratory, Centre of Research for Development, University of Kashmir, Srinagar, 190006, Jammu and Kashmir, India.
| | - Rana M Aldossri
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Muneeb U Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
9
|
Wusiman D, Guo L, Li L, Zhang X, Zhao X, An Z, Huang Z, Zhang Y, Li Z, Ying J, Wei M, Li W, An C. Clinicopathological and prognostic significance of PD-L1 and TIM-3 expression in medullary thyroid carcinoma: a retrospective immunohistochemistry study. J Endocrinol Invest 2024; 47:91-100. [PMID: 37464189 PMCID: PMC10776706 DOI: 10.1007/s40618-023-02126-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/30/2023] [Indexed: 07/20/2023]
Abstract
PURPOSE Expression of the programmed death-ligand 1 (PD-L1) and T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) in medullary thyroid carcinoma (MTC) has been controversial and rarely reported. METHODS Surgical specimens of 190 MTC patients who had initial curative-intent surgery were collected. Immunohistochemistry of PD-L1 and TIM-3 was performed using 22C3 pharmDx (Dako, Carpinteria, CA) and anti-TIM-3 (1:500, ab241332, Abcam). Stained slides were scored using a combined positive score (CPS) with a cutoff of ≥ 1. We established correlations between PD-L1 expression, TIM-3 expression, clinicopathological, and survival data. RESULTS 13 cases (13/190, 6.84%) were positive for PD-L1 expression, and 42 cases (42/154, 27.27%) for TIM-3 expression. PD-L1 expression was correlated to TIM-3 expression (P = 0.002), but was not related to overall survival (OS) or progression-free survival (PFS). TIM-3 expression was correlated to perineural invasion (P = 0.040). Multivariate Cox analysis showed that lymphovascular invasion (LVI) was independently associated with OS. And tumor size, LVI, and lymph node metastases were significantly associated with PFS. Furthermore, the multivariate logistic analysis showed multifocal status, LVI, pathological T stage and lymph node metastasis were independent risk factors for biochemical recurrence/persistent disease. CONCLUSIONS We demonstrated that PD-L1 and TIM-3 expression were not frequent in MTC and were not associated with survival prognosis. Our results should be considered when clinical trials of PD-L1 or TIM-3 blockades are implemented.
Collapse
Affiliation(s)
- D Wusiman
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - L Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - L Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - X Zhang
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - X Zhao
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Z An
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Z Huang
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Y Zhang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Z Li
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - J Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - M Wei
- Department of Head and Neck Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 518116, Shenzhen, China.
| | - W Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| | - C An
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| |
Collapse
|
10
|
Youssef R, Maniar R, Khan J, Mesa H. Metabolic Interplay in the Tumor Microenvironment: Implications for Immune Function and Anticancer Response. Curr Issues Mol Biol 2023; 45:9753-9767. [PMID: 38132455 PMCID: PMC10742411 DOI: 10.3390/cimb45120609] [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: 10/31/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023] Open
Abstract
Malignant tumors exhibit rapid growth and high metabolic rates, similar to embryonic stem cells, and depend on aerobic glycolysis, known as the "Warburg effect". This understanding has enabled the use of radiolabeled glucose analogs in tumor staging and therapeutic response assessment via PET scans. Traditional treatments like chemotherapy and radiotherapy target rapidly dividing cells, causing significant toxicity. Despite immunotherapy's impact on solid tumor treatment, gaps remain, leading to research on cancer cell evasion of immune response and immune tolerance induction via interactions with the tumor microenvironment (TME). The TME, consisting of immune cells, fibroblasts, vessels, and the extracellular matrix, regulates tumor progression and therapy responses. TME-targeted therapies aim to transform this environment from supporting tumor growth to impeding it and fostering an effective immune response. This review examines the metabolic disparities between immune cells and cancer cells, their impact on immune function and therapeutic targeting, the TME components, and the complex interplay between cancer cells and nontumoral cells. The success of TME-targeted therapies highlights their potential to achieve better cancer control or even a cure.
Collapse
Affiliation(s)
- Reem Youssef
- Department of Laboratory Medicine and Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Rohan Maniar
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jaffar Khan
- Department of Laboratory Medicine and Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hector Mesa
- Department of Laboratory Medicine and Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
11
|
Patel M, Hudson O, Han J, Kondapalli L, Arora G, Hawi R, Andrikopoulou E, Estes C, Johnson AM, Lenneman C. Update on Immunotherapy Cardiotoxicity: Checkpoint Inhibitors, CAR T, and Beyond. Curr Treat Options Oncol 2023; 24:1489-1503. [PMID: 37624557 DOI: 10.1007/s11864-023-01130-y] [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] [Accepted: 07/18/2023] [Indexed: 08/26/2023]
Abstract
OPINION STATEMENT Immunotherapy is an innovative approach to cancer treatment that involves using the body's immune system to fight cancer. The landscape of immunotherapy is constantly evolving, as new therapies are developed and refined. Some of the most promising approaches in immunotherapy include immune checkpoint inhibitors (ICIs): these drugs target proteins on the surface of T-cells that inhibit their ability to attack cancer cells. By blocking these proteins, checkpoint inhibitors allow T-cells to recognize and destroy cancer cells more effectively. CAR T-cell therapy: this therapy involves genetically modifying a patient's own T-cells to recognize and attack cancer cells. CAR T-cell therapy exhibits favorable response in many patients with refractory hematological cancers with growing clinical trials in solid tumors. Immune system modulators: these drugs enhance the immune system's ability to fight cancer by stimulating the production of immune cells or inhibiting the activity of immune-suppressing cells. While immunotherapy has shown great promise in the treatment of cancer, it can also pose significant cardiac side effects. Some immunotherapy drugs like ICIs can cause myocarditis, which can lead to chest pain, shortness of breath, and heart failure. Other cardiac side effects of ICIs include arrhythmias, pericarditis, vasculitis, and accelerated atherosclerosis. It is important for patients receiving immunotherapy to be monitored closely for these side effects, as prompt treatment can help prevent serious complications. Patients should also report any symptoms to their healthcare providers right away, so that appropriate action can be taken. CAR T-cell therapy can also illicit an exaggerated immune response creating cytokine release syndrome (CRS) that may precipitate cardiovascular events: arrhythmias, myocardial infarction, and heart failure. Overall, while immune modulating therapy is a promising and expanding approach to cancer treatment, it is important to weigh the potential benefits against the risks and side effects, especially in patients with high risk for cardiovascular complications.
Collapse
Affiliation(s)
- Murti Patel
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Olivia Hudson
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Jingnan Han
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Lavanya Kondapalli
- University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Garima Arora
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Riem Hawi
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | | | - Courtney Estes
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Abigail M Johnson
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Carrie Lenneman
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA.
| |
Collapse
|
12
|
Klaihmon P, Kang X, Issaragrisil S, Luanpitpong S. Generation and Functional Characterization of Anti-CD19 Chimeric Antigen Receptor-Natural Killer Cells from Human Induced Pluripotent Stem Cells. Int J Mol Sci 2023; 24:10508. [PMID: 37445684 DOI: 10.3390/ijms241310508] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Natural killer (NK) cells are a part of innate immunity that can be activated rapidly in response to malignant transformed cells without prior sensitization. Engineering NK cells to express chimeric antigen receptors (CARs) allows them to be directed against corresponding target tumor antigens. CAR-NK cells are regarded as a promising candidate for cellular immunotherapy alternatives to conventional CAR-T cells, due to the relatively low risk of graft-versus-host disease and safer clinical profile. Human induced pluripotent stem cells (iPSCs) are a promising renewable cell source of clinical NK cells. In the present study, we successfully introduced a third-generation CAR targeting CD19, which was validated to have effective signaling domains suitable for NK cells, into umbilical cord blood NK-derived iPSCs, followed by a single-cell clone selection and thorough iPSC characterization. The established single-cell clone of CAR19-NK/iPSCs, which is highly desirable for clinical application, can be differentiated using serum- and feeder-free protocols into functional CAR19-iNK-like cells with improved anti-tumor activity against CD19-positive hematologic cancer cells when compared with wild-type (WT)-iNK-like cells. With the feasibility of being an alternative source for off-the-shelf CAR-NK cells, a library of single-cell clones of CAR-engineered NK/iPSCs targeting different tumor antigens may be created for future clinical application.
Collapse
Affiliation(s)
- Phatchanat Klaihmon
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Xing Kang
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- BDMS Center of Excellence for Hematology, Wattanosoth Cancer Hospital, Bangkok 10310, Thailand
| | - Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
13
|
Lei Q, Wang Y, Li J, Wang S, Hu Y, Duan L, Huo Y, Wu Y, Liu H. Increased SEMA6B expression as a potential prognostic and immune cell infiltration biomarker in thyroid cancer patients. Aging (Albany NY) 2023; 15:3572-3585. [PMID: 37155149 PMCID: PMC10449300 DOI: 10.18632/aging.204691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND Even today, thyroid cancer (THCA) remains an important threat to global health. For THCA patients, differentiated thyroid cancer is the most commonly identified pathological subtype, and those diagnosed with papillary thyroid cancer generally have good overall prognosis. For poorly differentiated subtype THCA, patients have aggressive disease course, higher risk of distant organ metastasis and inferior overall prognosis. METHODS RNA-seq data from TCGA and GTEx databases are collected and analyzed via R. The correlation between SEMA6B expression level and pathological as well as clinical parameters of THCA patients was respectively investigated. Gene expression profiling and subsequent functional clustering analysis was the performed utilizing GSEA. The receiver operating characteristic (ROC) curve was utilized to evaluate the diagnostic value of SEMA6B expression. RESULTS Increased SEMA6B expression was characteristic in THCA tumor samples and was associated with specific pathologic and clinical features for TCHA patients. Univariate and multivariate analysis indicated that SEMA6B was independent predictive marker for THCA patients' prognosis. Gene expression profiling and functional clustering analysis suggested that SEMA6B high-expression was related with increased expression of multiple signal pathways and signatures of multiple immune cell infiltration. CONCLUSIONS In this study, through bioinformatic analysis and clinical data investigation, we demonstrated the potential value of SEMA6B as diagnostic and prognostic marker in THCA patient treatment.
Collapse
Affiliation(s)
- Qinghua Lei
- Physical Examination Center, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Yunha Wang
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing 100853, China
| | - Junhua Li
- Physical Examination Center, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Shanshan Wang
- Physical Examination Center, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Yanyan Hu
- Physical Examination Center, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Lihua Duan
- Department of Ultrasound Medicine, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Yanfei Huo
- Physical Examination Center, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Yiping Wu
- Department of Neurology, Handan Central Hospital, Congtai, Handan 056008, Hebei Province, China
| | - Hongzhou Liu
- Department of Endocrinology, First Hospital of Handan City, Congtai, Handan 056002, Hebei Province, China
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing 100853, China
| |
Collapse
|
14
|
Wang C, Zhu H, Huang H, Sun Z, Teng Y, Li Y. Immune-induced pneumonia in patients with advanced solid tumors treated with immunotherapy: a real-world assessment. Future Oncol 2023; 19:259-270. [PMID: 36891950 DOI: 10.2217/fon-2022-0826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023] Open
Abstract
Aim: To investigate the computed tomography (CT) and clinical characteristics of immunotherapy-induced pneumonitis (IIP) in patients with advanced solid tumors. Patients & methods: CT and clinical data of 254 patients with advanced solid tumors treated with immune checkpoint inhibitors in our hospital were collected retrospectively. Results: The incidences of IIP in patients with non-small-cell lung cancer, lymphoma and gastrointestinal tumors were 19% (19/100), 9.8% (6/61) and 6.2% (4/65), respectively. The median onset time for all 31 IIP patients was 44 days (interquartile range: 24-65). Most IIP patients (21/31) had grade 1-2 disease. Multifocal ground-glass opacities (seen in 21/31 patients) were the main CT findings of IIP. Conclusion: Patients should be alerted to the risk of IIP, an adverse reaction that has a relatively low incidence but which is sometimes life-threatening.
Collapse
Affiliation(s)
- Chuhan Wang
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, 215000, China
| | - Hong Zhu
- Department of Medical Oncology, the First Affiliated Hospital of Soochow University, Suzhou City Jiangsu Province, 215006, China
| | - Haiwen Huang
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, 215000, China.,Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, 215006, China
| | - Zongqiong Sun
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi City, Jiangsu Province, 214000, China
| | - Yue Teng
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, 215000, China
| | - Yonggang Li
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, 215000, China.,National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, 215000, China.,Institute of Medical Imaging, Soochow University, Suzhou City, Jiangsu Province, 215000, China
| |
Collapse
|
15
|
Supramolecular assembly of a trivalent peptide hydrogel vaccine for cancer immunotherapy. Acta Biomater 2023; 158:535-546. [PMID: 36632876 DOI: 10.1016/j.actbio.2022.12.070] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/10/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023]
Abstract
Vaccination shows great promise in cancer immunotherapy. However, the induction of robust and broad therapeutic CD8 T cell immunity against tumors is challenging due to the essential heterogenicity of tumor antigen expression. Recently, bioinspired materials have reshaped the field of cancer nanomedicine. Herein, a bioinspired nanofibrous trivalent peptide hydrogel vaccine was constructed using the spontaneous supramolecular co-assembly of three antigenic epitope-conjugated peptides, which could mimic the fibrillar structure and biological function of the extracellular matrix and naturally occurring protein assembly. The hydrogel vaccine could be accurately and flexibly adjusted to load each antigenic peptide at a defined ratio, which facilitated the antigen presentation of dendritic cells and significantly improved the initiation of CD8 T cell response and the secretion of interferon-γ (IFN-γ). C57BL/6 mice were immunized with the trivalent peptide hydrogel vaccine, where it elicited a high broad-spectrum antitumor CD8 T cell response that significantly inhibited the growth of B16 tumors in the absence of additional immunoadjuvants or delivery systems. In summary, the supramolecular assembly of triple antigenic epitope-conjugated peptides offers a simple, customizable, and versatile approach for the development of cancer vaccines with remarkable therapeutic efficacy, thereby providing a highly versatile platform for the application of personalized multivalent tumor vaccines. STATEMENT OF SIGNIFICANCE: (1) We report a feasible, versatile and bioinspired approach to manufacture a multivalent peptide-based hydrogel cancer vaccine in the absence of additional adjuvants, which closely mimics immune niches, co-delivers antigen epitopes, greatly promotes antigen presentation to DCs and their subsequent homing to dLNs and elicits a broad-spectrum antitumor CD8 T cell response, resulting in significant inhibition of B16 tumor growth. (2) This feasible and efficient co-assembly strategy provides an attractive platform for engineering a range of multivalent vaccines at defined ratios to further enhance antigen-specific T cell responses. This approach may also be used for personalized immunotherapy with neo-epitopes.
Collapse
|
16
|
Ma W, Xue R, Zhu Z, Farrukh H, Song W, Li T, Zheng L, Pan CX. Increasing cure rates of solid tumors by immune checkpoint inhibitors. Exp Hematol Oncol 2023; 12:10. [PMID: 36647169 PMCID: PMC9843946 DOI: 10.1186/s40164-023-00372-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 01/04/2023] [Indexed: 01/18/2023] Open
Abstract
Immunotherapy has become the central pillar of cancer therapy. Immune checkpoint inhibitors (ICIs), a major category of tumor immunotherapy, reactivate preexisting anticancer immunity. Initially, ICIs were approved only for advanced and metastatic cancers in the salvage setting after or concurrent with chemotherapy at a response rate of around 20-30% with a few exceptions. With significant progress over the decade, advances in immunotherapy have led to numerous clinical trials investigating ICIs as neoadjuvant and/or adjuvant therapies for resectable solid tumors. The promising results of these trials have led to the United States Food and Drug Administration (FDA) approvals of ICIs as neoadjuvant or adjuvant therapies for non-small cell lung cancer, melanoma, triple-negative breast cancer, and bladder cancer, and the list continues to grow. This therapy represents a paradigm shift in cancer treatment, as many early-stage cancer patients could be cured with the introduction of immunotherapy in the early stages of cancer. Therefore, this topic became one of the main themes at the 2021 China Cancer Immunotherapy Workshop co-organized by the Chinese American Hematologist and Oncologist Network, the China National Medical Products Administration and the Tsinghua University School of Medicine. This review article summarizes the current landscape of ICI-based immunotherapy, emphasizing the new clinical developments of ICIs as curative neoadjuvant and adjuvant therapies for early-stage disease.
Collapse
Affiliation(s)
- Weijie Ma
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Ruobing Xue
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Ellis Fischel Cancer Center, University of Missouri, 1 Hospital Dr, Columbia, MO, 65201, USA
| | - Zheng Zhu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Hizra Farrukh
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Wenru Song
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Kira Pharmaceuticals, Cambridge, MA, USA
| | - Tianhong Li
- Chinese American Hematologist and Oncologist Network, New York, NY, USA. .,Department of Medicine, Division of Hematology & Oncology, University of California Davis, Sacramento, CA, 95817, USA. .,Department of Medicine, VA Northern California Health Care System, Mather, CA, USA.
| | - Lei Zheng
- Chinese American Hematologist and Oncologist Network, New York, NY, USA. .,The Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Chong-Xian Pan
- Chinese American Hematologist and Oncologist Network, New York, NY, USA. .,VA Boston Healthcare System, Boston, MA, 02132, USA. .,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Akter Z, Salamat N, Ali MY, Zhang L. The promise of targeting heme and mitochondrial respiration in normalizing tumor microenvironment and potentiating immunotherapy. Front Oncol 2023; 12:1072739. [PMID: 36686754 PMCID: PMC9851275 DOI: 10.3389/fonc.2022.1072739] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
Cancer immunotherapy shows durable treatment responses and therapeutic benefits compared to other cancer treatment modalities, but many cancer patients display primary and acquired resistance to immunotherapeutics. Immunosuppressive tumor microenvironment (TME) is a major barrier to cancer immunotherapy. Notably, cancer cells depend on high mitochondrial bioenergetics accompanied with the supply of heme for their growth, proliferation, progression, and metastasis. This excessive mitochondrial respiration increases tumor cells oxygen consumption, which triggers hypoxia and irregular blood vessels formation in various regions of TME, resulting in an immunosuppressive TME, evasion of anti-tumor immunity, and resistance to immunotherapeutic agents. In this review, we discuss the role of heme, heme catabolism, and mitochondrial respiration on mediating immunosuppressive TME by promoting hypoxia, angiogenesis, and leaky tumor vasculature. Moreover, we discuss the therapeutic prospects of targeting heme and mitochondrial respiration in alleviating tumor hypoxia, normalizing tumor vasculature, and TME to restore anti-tumor immunity and resensitize cancer cells to immunotherapy.
Collapse
|
18
|
Jlassi A, Manai M, Morjen M, Sahraoui G, Elasmi Allal M, ELBini-Dhouib I, Naija L, Charfi L, Rejaibi R, Ben Ahmed M, Marrakchi N, Srairi-Abid N, Mezlini A, Manai M, Mrad K, Doghri R. VISTA+/CD8+ status correlates with favorable prognosis in Epithelial ovarian cancer. PLoS One 2023; 18:e0278849. [PMID: 36952478 PMCID: PMC10035885 DOI: 10.1371/journal.pone.0278849] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/23/2022] [Indexed: 03/25/2023] Open
Abstract
Immunotherapy by blocking immune checkpoint regulators has emerged as a new targeted therapy for some cancers. Among them V-domain Ig suppressor of Tcell activation (VISTA) which is identified as a novel checkpoint regulator in ovarian cancer. This study aimed to investigate the VISTA role in Epithelial ovarian cancer (EOC), and its relationship with tumor-infiltrating lymphocytes (TILs) markers and its prognostic value. The expression of VISTA, CD3, CD8, CD4, FOXP3, and CD56 was assessed in 168 EOC tissue microarrays (TMA) by immunohistochemistry (IHC). In addition, associations between VISTA, TILs, clinicopathological variables, and overall survival (OS) were analyzed. VISTA expression in IGRov1 cells, as well as in PBMC of EOC patient, was evaluated by western blot. VISTA expression was detected in 64,28% of tissues, among which 42.3% were positive for tumor cells (TCs), and 47,9% were positive for immune cells (ICs). In univariate analysis, VISTA expression was significantly associated with a high density of TILs:CD3+ (p = 0,001), CD4+ (p = 0,002) and CD8+ (p≤0,001), in ICs but not in TCs. In terms of OS, multivariate analysis showed a significant association between the high density of CD8+ TILs and VISTA positive staining in ICs (p = 0,044), but not in TCs (p = 0,108). Kaplan-Meier curves demonstrated no correlation between VISTA expression and prolonged OS in both ICs (p = 0,841) and TCs (p = 0,090). Classification of EOC tumor microenvironment based on VISTA and CD8+TILs expression, demonstrated four immune subtypes: VISTA+/CD8+, VISTA+/CD8-, VISTA-/CD8+ and VISTA-/CD8-. The dual positive VISTA+/CD8+ subtype was significantly associated with prolonged OS in both TCs and ICs (p = 0,012 and p≤0,01, respectively), whereas patients with VISTA+/CD8- had the worst OS. Our results showed that VISTA is highly expressed in the IGRov1 cell line and LT-CD8 from a patient with EOC. Our results highlighted the association of VISTA expression and CD8+ TILs in EOC, with prolonged OS in patients with VISTA+/CD8+ and proposed VISTA as a potential immunotherapeutic target in EOC.
Collapse
Affiliation(s)
- Aida Jlassi
- Department of Biology, Mycology, Pathologies and Biomarkers Laboratory (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
| | - Maroua Manai
- Department of Biology, Mycology, Pathologies and Biomarkers Laboratory (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia
- Human Genetics Laboratory (LR99ES10), Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Department of Medicine, Division of Hematology-oncology, New York, New York, United States of America
| | - Maram Morjen
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Ghada Sahraoui
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | | | - Ines ELBini-Dhouib
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Lamia Naija
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Department of Surgical Oncology, Salah Aziz Institute, Tunis, Tunisia
| | - Lamia Charfi
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Rim Rejaibi
- Department of Biology, Mycology, Pathologies and Biomarkers Laboratory (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
| | - Melika Ben Ahmed
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infections - LR16IPT02, Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Naziha Marrakchi
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Najet Srairi-Abid
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Amel Mezlini
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Department of Medical Oncology, Salah Aziz Institute, Tunis, Tunisia
| | - Mohamed Manai
- Department of Biology, Mycology, Pathologies and Biomarkers Laboratory (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia
| | - Karima Mrad
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Raoudha Doghri
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| |
Collapse
|
19
|
Lv J, Qin L, Zhao R, Wu D, Wu Z, Zheng D, Li S, Luo M, Wu Q, Long Y, Tang Z, Tang YL, Luo X, Yao Y, Yang LH, Li P. Disruption of CISH promotes the antitumor activity of human T cells and decreases PD-1 expression levels. Mol Ther Oncolytics 2022; 28:46-58. [PMID: 36654786 PMCID: PMC9827364 DOI: 10.1016/j.omto.2022.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Tumor cells and the immunosuppressive tumor microenvironment suppress the antitumor activity of T cells through immune checkpoints, including the PD-L1/PD-1 axis. Cytokine-inducible SH2-containing protein (CISH), a member of the suppressor of cytokine signaling (SOCS) family, inhibits JAK-STAT and T cell receptor (TCR) signaling in T and natural killer (NK) cells. However, its role in the regulation of immune checkpoints in T cells remains unclear. In this study, we ablated CISH in T cells with CRISPR-Cas9 and found that the sensitivity of T cells to TCR and cytokine stimulation was increased. In addition, chimeric antigen receptor T cells with CISH deficiency exhibited longer survival and higher cytokine secretion and antitumor activity. Notably, PD-1 expression was decreased in activated CISH-deficient T cells in vitro and in vivo. The level of FBXO38, a ubiquitination-regulating protein that reduces PD-1 expression, was elevated in activated T cells after CISH ablation. Hence, this study reveals a mechanism by which CISH promotes PD-1 expression by suppressing the expression of FBXO38 and proposes a new strategy for augmenting the therapeutic effect of CAR-T cells by inhibiting CISH.
Collapse
Affiliation(s)
- Jiang Lv
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Le Qin
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Ruocong Zhao
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR 999077, China
| | - Di Wu
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Zhiping Wu
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Diwei Zheng
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Siyu Li
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China
| | - Mintao Luo
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiting Wu
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Youguo Long
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Zhaoyang Tang
- Guangdong Zhaotai InVivo Biomedicine Co., Ltd., Guangzhou 510700, China
| | - Yan-Lai Tang
- Department of Paediatrics, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Xuequn Luo
- Department of Paediatrics, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yao Yao
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Li-Hua Yang
- Department of Paediatrics, Zhujiang Hospital, Southern China Medical University, Guangzhou, Guangdong 510280, China,Corresponding author Li-Hua Yang, Department of Paediatrics, Zhujiang Hospital, Southern China Medical University, Guangzhou, Guangdong 510280, China.
| | - Peng Li
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China,University of Chinese Academy of Sciences, Beijing 100049, China,Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR 999077, China,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Corresponding author Peng Li, China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
| |
Collapse
|
20
|
Guven DC, Kavgaci G, Aktepe OH, Yildirim HC, Sahin TK, Aksoy S, Erman M, Kilickap S, Yalcin S. The burden of polypharmacy and drug-drug interactions in older cancer patients treated with immunotherapy. J Oncol Pharm Pract 2022; 28:785-793. [PMID: 33878976 DOI: 10.1177/10781552211012038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Polypharmacy is a common problem in older cancer patients, although the data about polypharmacy and potentially inappropriate prescription practices is limited in patients treated with immune checkpoint inhibitors (ICIs). Therefore, we aimed to evaluate the polypharmacy frequency and drug-drug interactions in older cancer patients (≥65 years) treated with ICIs. METHODS A total of 70 geriatric patients with advanced cancer were included. The polypharmacy was defined as regular use of 5 or more drugs. The START/STOPP Criteria Version 2 was used for the potentially inappropriate medications (PIM) and potential prescription omissions (PPO). The Medscape Drug Interaction Checker was used for potential drug-drug interactions. RESULTS The patients had a median of 6 regular drugs, and polypharmacy was present in 77.1%. The polypharmacy risk was significantly increased in patients over 75 years of age (p = 0.028) and with opioid use (p = 0.048). The 50% of patients had category D or X interactions. Patients with higher Charlson Comorbidity Index had significantly increased risk for drug interactions (CCI ≤10 vs. >10, p = 0.017). The PIMs were present in 44.3% and the PPOs in 68.6% of the patients. While the overall survival and immune related adverse events were similar according to polypharmacy, in patients using seven or more drugs, the acute kidney injury risk was increased (HR: 4.667, p = 0.038). CONCLUSION In this study, we observed a high rate of polypharmacy and inappropriate prescription practices in ICI-treated patients. These issues pointed out the need for improved general medical care and attention for better comedication management in ICI-treated patients.
Collapse
Affiliation(s)
- Deniz C Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Gozde Kavgaci
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Oktay H Aktepe
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Hasan C Yildirim
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Taha K Sahin
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Sercan Aksoy
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Mustafa Erman
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Saadettin Kilickap
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Suayib Yalcin
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
21
|
CheshmehSohrabi M, Shabani R, Shirdavani S. Tops and Trends in Iranian Cancer Research: A Bibliometric Analysis. ARCHIVES OF IRANIAN MEDICINE 2022; 25:224-234. [PMID: 35942994 PMCID: PMC11897874 DOI: 10.34172/aim.2022.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 04/21/2021] [Indexed: 06/15/2023]
Abstract
BACKGROUND Detecting the main actors and important topics of Iranian cancer research is essential for Iranian policymakers, clinicians, and researchers. This study was conducted to demonstrate the trends and tops in Iranian cancer research from 1960 to 2018. METHODS A total of 22,370 Iranian cancer articles in Web of Science (WoS), PubMed, and Scopus, from 1960 through 2018, were extracted and preprocessed based on data mining techniques and analyzed using the collaboration network analysis, keywords analysis, and bibliometrics methods. RESULTS The results reveal that, Tehran University of Medical Sciences (11.46%) among organizations, Asian Pacific Journal of Cancer Prevention (5%) among journals, Malekzadeh R (1.09%) among authors, and Breast cancer (10.37%) among topics ranked the first. The trend of Iranian cancer research represents three periods: 1) germinating period, from 1970 to 2000, 2) developing period, from 2002 to 2014, and 3) flourishing period, from 2014 to 2018. It is expected that this trend will continue. The results indicate an average 12.8% increase in the logarithm of the count of articles published by Iranian cancer researchers each year. The findings are contextualized with Price's publications trends principal for determining global and Iranian cancer research publication trends. CONCLUSION The number of research papers published by Iranian researchers on cancer is increasing. In order to maintain the publication growth in this field, greater participation by other Iranian institutions is suggested. Although the quantity and quality of papers are increasing in some topics, certain topics and types of cancers should be still further studied and the Iranian policymakers should be encouraged to invest more in these topics.
Collapse
Affiliation(s)
- Mozaffar CheshmehSohrabi
- Department of knowledge and Information Science, Faculty of Education and Psychology, University of Isfahan, Isfahan, Iran
| | - Rasoul Shabani
- Department of knowledge and Information Science, Faculty of Education and Psychology, University of Isfahan, Isfahan, Iran
| | - Shiva Shirdavani
- Department of knowledge and Information Science, Faculty of Education and Psychology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
22
|
Yadav D, Kwak M, Chauhan PS, Puranik N, Lee PCW, Jin JO. Cancer immunotherapy by immune checkpoint blockade and its advanced application using bio-nanomaterials. Semin Cancer Biol 2022; 86:909-922. [PMID: 35181474 DOI: 10.1016/j.semcancer.2022.02.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/09/2022] [Accepted: 02/13/2022] [Indexed: 02/07/2023]
Abstract
Cancer is the second leading cause of death worldwide. Traditional approaches, such as surgery, chemotherapy, and radiotherapy have been the main cancer therapeutic modalities in recent years. Cancer immunotherapy is a novel therapeutic modality that potentiates the immune responses of patients against malignancy. Immune checkpoint proteins expressed on T cells or tumor cells serve as a target for inhibiting T cell overactivation, maintaining the balance between self-reactivity and autoimmunity. Tumors essentially hijack the immune checkpoint pathway in order to survive and spread. Immune checkpoint inhibitors (ICIs) are being developed as a result to reactivate the anti-tumor immune response. Recent advances in nanotechnology have contributed to the development of successful, safe, and efficient anticancer drug systems based on nanoparticles. Nanoparticle-based cancer immunotherapy overcomes numerous challenges and offers novel strategies for improving conventional immunotherapies. The fundamental and physiochemical properties of nanoparticles depend on various cancer therapeutic strategies, such as chemotherapeutics, nucleic acid-based treatments, photothermal therapy, and photodynamic agents. The review discusses the use of nanoparticles as carriers for delivering immune checkpoint inhibitors and their efficacy in cancer combination therapy.
Collapse
Affiliation(s)
- Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Minseok Kwak
- Department of Chemistry and Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, South Korea
| | | | - Nidhi Puranik
- Biological Sciences Department, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Peter C W Lee
- Department of Biomedical Sciences, University of Ulsan College of Medicine, ASAN Medical Center, Seoul, South Korea.
| | - Jun-O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea.
| |
Collapse
|
23
|
Zhang L, Jia H, Liu X, Zou Y, Sun J, Liu M, Jia S, Liu N, Li Y, Wang Q. Heptamethine Cyanine–Based Application for Cancer Theranostics. Front Pharmacol 2022; 12:764654. [PMID: 35222006 PMCID: PMC8874131 DOI: 10.3389/fphar.2021.764654] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 12/09/2021] [Indexed: 01/31/2023] Open
Abstract
Cancer is the most common life-threatening malignant disease. The future of personalized cancer treatments relies on the development of functional agents that have tumor-targeted anticancer activities and can be detected in tumors through imaging. Cyanines, especially heptamethine cyanine (Cy7), have prospective application because of their excellent tumor-targeting capacity, high quantum yield, low tissue autofluorescence, long absorption wavelength, and low background interference. In this review, the application of Cy7 and its derivatives in tumors is comprehensively explored. Cy7 is enormously acknowledged in the field of non-invasive therapy that can “detect” and “kill” tumor cells via near-infrared fluorescence (NIRF) imaging, photothermal therapy (PTT), and photodynamic therapy (PDT). Furthermore, Cy7 is more available and has excellent properties in cancer theranostics by the presence of multifunctional nanoparticles via fulfilling multimodal imaging and combination therapy simultaneously. This review provides a comprehensive scope of Cy7’s application for cancer NIRF imaging, phototherapy, nanoprobe-based combination therapy in recent years. A deeper understanding of the application of imaging and treatment underlying Cy7 in cancer may provide new strategies for drug development based on cyanine. Thus, the review will lead the way to new types with optical properties and practical transformation to clinical practice.
Collapse
Affiliation(s)
- Lei Zhang
- School of Basic Medical Sciences, Laboratory for Nanomedicine, Henan University, Kaifeng, China
| | - Hang Jia
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Xuqian Liu
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Yaxin Zou
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Jiayi Sun
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Mengyu Liu
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Shuangshuang Jia
- School of Basic Medical Sciences, Laboratory for Nanomedicine, Henan University, Kaifeng, China
| | - Nan Liu
- Obstetrics Department, Kaifeng Maternity Hospital, Kaifeng, China
| | - Yanzhang Li
- School of Basic Medical Sciences, Laboratory for Nanomedicine, Henan University, Kaifeng, China
- *Correspondence: Qun Wang, ; Yanzhang Li,
| | - Qun Wang
- School of Basic Medical Sciences, Laboratory for Nanomedicine, Henan University, Kaifeng, China
- *Correspondence: Qun Wang, ; Yanzhang Li,
| |
Collapse
|
24
|
Abu N, Rus Bakarurraini NAA. The interweaving relationship between extracellular vesicles and T cells in cancer. Cancer Lett 2021; 530:1-7. [PMID: 34906625 DOI: 10.1016/j.canlet.2021.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/26/2021] [Accepted: 12/07/2021] [Indexed: 12/21/2022]
Abstract
The interdependency between cancer cells and immune cells is an important link in understanding cancer pathogenesis. T cells are important immune cells that are able to either impede or promote tumor growth. Extracellular vesicles or EVs are membrane-encapsulated vesicles that are released by both cancer and immune cells that can act as communicators. Studies have shown that tumor-derived EVs can interact with immune cells, particularly T cells. Vice versa, T cells-derived EVs have also been shown to possess immunomodulatory roles. Therefore, the purpose of this mini-review is to understand the role of tumor-derived EVs and T-cells derived EVs on cancer immunosuppression especially the interweaving role of different types of EVs and how it affects tumor immunity. We also discuss the role of EVs in different types of T cells namely CD8+, CD4+ Th17 and Treg cells. More importantly, we include the limitations and future directions involving this type of research. This will further elucidate our understanding of the important functions of these tiny mediators.
Collapse
Affiliation(s)
- Nadiah Abu
- UKM Medical Molecular Biology Institute (UMBI), UKM Medical Center, 56000, Kuala Lumpur, Malaysia.
| | | |
Collapse
|
25
|
Li L, Shi W, Zhou J. Effect of CMNa combined with radiotherapy on the tumor immune microenvironment of mouse cervical cancer cell transplantation tumor model. Bioengineered 2021; 12:1066-1077. [PMID: 33784955 PMCID: PMC8806344 DOI: 10.1080/21655979.2021.1899532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 03/02/2021] [Indexed: 01/30/2023] Open
Abstract
In this study, we construct a subcutaneous tumor mice model of U14 cells, observe the tumor growth, and detect the expression of Foxp3 and VISTA in cervical cancer tissues and adjacent tissues during CMNa-enhancing radiotherapy.From the 15th day, compared with the control group, the tumor volume changes in each treatment group were significant (P < 0.01). CMNa combined with radiotherapy had an interactive effect and a positive effect in inhibiting tumor volume growth. There was no significant difference in the expression of Foxp3 and VISTA in mouse cervical cancer tissues and adjacent tissues in each group. The Foxp3 level in the RT group was the highest, and the CMNa group was the lowest. The VISTA level of the CMNa+RT group was the highest, the RT group is followed by, and the Control group is the lowest. The Foxp3 level of the CMNa group did not change much at each different point. The Foxp3 level in RT and CMNa+RT group gradually decreased after a transient increase, and the VISTA level in the CMNa+RT group increased more.Our results show that CMNa can enhance the efficacy of radiotherapy, and at the same time can reduce the compensatory increase in regulatory T cell Foxp3 levels caused by radiotherapy, and reduce the radiotherapy response. However, in the course of the treatment of the two, there may be a substantial increase in the level of VISTA, and the combined application of VISTA inhibitors may increase the anti-tumor response.
Collapse
Affiliation(s)
- Li Li
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weiqiang Shi
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Juying Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
26
|
Guven DC, Acar R, Yekeduz E, Bilgetekin I, Baytemur NK, Erol C, Ceylan F, Sendur MA, Demirci U, Urun Y, Karadurmus N, Erman M, Kilickap S. The association between antibiotic use and survival in renal cell carcinoma patients treated with immunotherapy: a multi-center study. Curr Probl Cancer 2021; 45:100760. [PMID: 34130864 DOI: 10.1016/j.currproblcancer.2021.100760] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 05/02/2021] [Accepted: 05/07/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Immunotherapy improves overall survival (OS) in the second and later lines of renal cell carcinoma (RCC) treatment. Recent studies have suggested that antibiotic (ATB) use either shortly before or after the start of immunotherapy could lead to decreased OS. Herein, we evaluate the impact of ATB use on OS in RCC patients treated with nivolumab in a multi-center cohort from Turkey. METHODS The data of 93 metastatic RCC patients treated with nivolumab in the second line or later were retrospectively collected from 6 oncology centers. Previous treatments, sites of metastases, International Metastatic RCC Database Consortium risk classification, and ATB use in the three months before (-3) or three months after (+3) the start of immunotherapy were recorded together with survival data. The association of clinical factors with OS and progression-free survival (PFS) was analyzed with univariate and multivariable analyses. RESULTS The median age was 61 (interquartile range 54-67), and 76.3% of the patients were male. The median OS of the cohort was 23.75 ± 4.41, and the PFS was 8.44 ± 1.61 months. Thirty-one (33.3%) patients used ATBs in the 3 months before (-3) or 3 months after (+3) nivolumab initiation. In the multivariable analyses, ATB exposure (HR: 2.306, 95% confidence interval [CI]: 1.155-4.601, P = 0.018) and the presence of brain metastases at the baseline (HR: 2.608, 95% CI: 1.200-5.666, P = 0.015) had a statistically significant association with OS, while ATB exposure was the only statistically significant parameter associated with PFS (HR: 2.238, 95% CI: 1.284-3.900, P = 0.004). CONCLUSION In our study, patients with ATB exposure in the 3 months before or 3 months after the start of immunotherapy had shorter OS. Our findings further support meticulous risk-benefit assessments of prescribing ATBs for patients who are either receiving or are expected to receive immunotherapy.
Collapse
Affiliation(s)
- Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Oncology Hospital, Ankara, Turkey.
| | - Ramazan Acar
- Department of Medical Oncology, Health Sciences University, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Emre Yekeduz
- Department of Medical Oncology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Irem Bilgetekin
- Department of Medical Oncology, Dr. Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, Ankara, Turkey
| | | | - Cihan Erol
- Department of Medical Oncology, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Furkan Ceylan
- Department of Medical Oncology, Hacettepe University Oncology Hospital, Ankara, Turkey
| | - Mehmet Ali Sendur
- Department of Medical Oncology, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Umut Demirci
- Department of Medical Oncology, Memorial Ankara Hospital, Ankara, Turkey
| | - Yuksel Urun
- Department of Medical Oncology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Nuri Karadurmus
- Department of Medical Oncology, Health Sciences University, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Mustafa Erman
- Department of Medical Oncology, Hacettepe University Oncology Hospital, Ankara, Turkey
| | - Saadettin Kilickap
- Department of Medical Oncology, Hacettepe University Oncology Hospital, Ankara, Turkey; Department of Medical Oncology, Istinye University Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
27
|
He W, Li Q, Lu Y, Ju D, Gu Y, Zhao K, Dong C. Cancer treatment evolution from traditional methods to stem cells and gene therapy. Curr Gene Ther 2021; 22:368-385. [PMID: 34802404 DOI: 10.2174/1566523221666211119110755] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 06/25/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cancer, a malignant tumor, is caused by the failure of the mechanism that controls cell growth and proliferation. Late clinical symptoms often manifest as lumps, pain, ulcers, and bleeding. Systemic symptoms include weight loss, fatigue, and loss of appetite. It is a major disease that threatens human life and health. How to treat cancer is a long-standing problem that needs to be overcome in the history of medicine. METHOD Traditional tumor treatment methods are poorly targeted, and the side effects of treatment seriously damage the physical and mental health of patients. In recent years, with the advancement of medical science and technology, the research on gene combined with mesenchymal stem cells to treat tumors has been intensified. Mesenchymal stem cells carry genes to target cancer cells, which can achieve better therapeutic effects. DISCUSSION In the text, we systematically review the cancer treatment evolution from traditional methods to novel approaches that include immunotherapy, nanotherapy, stem cell theapy, and gene therapy. We provide the latest review of the application status, clinical trials and development prospects of mesenchymal stem cells and gene therapy for cancer, as well as their integration in cancer treatment. Mesenchymal stem cells are effective carriers carrying genes and provide new clinical ideas for tumor treatment. CONCLUSION This review focuses on the current status, application prospects and challenges of mesenchymal stem cell combined gene therapy for cancer, and provides new ideas for clinical research.
Collapse
Affiliation(s)
- Wenhua He
- Department of Anatomy, Medical College of Nantong University, Nantong 226001. China
| | - Qingxuan Li
- Department of Anatomy, Medical College of Nantong University, Nantong 226001. China
| | - Yan Lu
- Department of Anatomy, Medical College of Nantong University, Nantong 226001. China
| | - Dingyue Ju
- Department of Anatomy, Medical College of Nantong University, Nantong 226001. China
| | - Yu Gu
- Department of Anatomy, Medical College of Nantong University, Nantong 226001. China
| | - Kai Zhao
- Department of Anatomy, Medical College of Nantong University, Nantong 226001. China
| | - Chuanming Dong
- Department of Anatomy, Medical College of Nantong University, Nantong 226001. China
| |
Collapse
|
28
|
Xing C, Li H, Li RJ, Yin L, Zhang HF, Huang ZN, Cheng Z, Li J, Wang ZH, Peng HL. The roles of exosomal immune checkpoint proteins in tumors. Mil Med Res 2021; 8:56. [PMID: 34743730 PMCID: PMC8573946 DOI: 10.1186/s40779-021-00350-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/19/2021] [Indexed: 02/08/2023] Open
Abstract
Targeting immune checkpoints has achieved great therapeutic effects in the treatment of early-stage tumors. However, most patients develop adaptive resistance to this therapy. The latest evidence demonstrates that tumor-derived exosomes may play a key role in systemic immune suppression and tumor progression. In this article, we highlight the role of exosomal immune checkpoint proteins in tumor immunity, with an emphasis on programmed death ligand 1 (PD-L1) and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), as well as emerging evidence on roles of T cell immunoglobulin-3 (TIM-3), arginase 1 (ARG1), and estrogen receptor binding fragment-associated antigen 9 (EBAG9) expressed by exosomes.
Collapse
Affiliation(s)
- Cheng Xing
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011 China
- Institute of Molecular Hematology, Central South University, Changsha, 410011 China
| | - Heng Li
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011 China
- Institute of Molecular Hematology, Central South University, Changsha, 410011 China
| | - Rui-Juan Li
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011 China
- Institute of Molecular Hematology, Central South University, Changsha, 410011 China
| | - Le Yin
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011 China
- Institute of Molecular Hematology, Central South University, Changsha, 410011 China
| | - Hui-Fang Zhang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011 China
- Institute of Molecular Hematology, Central South University, Changsha, 410011 China
| | - Zi-Neng Huang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011 China
- Institute of Molecular Hematology, Central South University, Changsha, 410011 China
| | - Zhao Cheng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011 China
- Institute of Molecular Hematology, Central South University, Changsha, 410011 China
| | - Ji Li
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011 China
- Institute of Molecular Hematology, Central South University, Changsha, 410011 China
| | - Zhi-Hua Wang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011 China
- Institute of Molecular Hematology, Central South University, Changsha, 410011 China
| | - Hong-Ling Peng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011 China
- Institute of Molecular Hematology, Central South University, Changsha, 410011 China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Changsha, 410011 China
| |
Collapse
|
29
|
Lecocq Q, Debie P, Puttemans J, Awad RM, De Beck L, Ertveldt T, De Vlaeminck Y, Goyvaerts C, Raes G, Keyaerts M, Breckpot K, Devoogdt N. Evaluation of single domain antibodies as nuclear tracers for imaging of the immune checkpoint receptor human lymphocyte activation gene-3 in cancer. EJNMMI Res 2021; 11:115. [PMID: 34727262 PMCID: PMC8563901 DOI: 10.1186/s13550-021-00857-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/22/2021] [Indexed: 12/29/2022] Open
Abstract
Recent advancements in the field of immune-oncology have led to a significant increase in life expectancy of patients with diverse forms of cancer, such as hematologic malignancies, melanoma and lung cancer. Unfortunately, these encouraging results are not observed in the majority of patients, who remain unresponsive and/or encounter adverse events. Currently, researchers are collecting more insight into the cellular and molecular mechanisms that underlie these variable responses. As an example, the human lymphocyte activation gene-3 (huLAG-3), an inhibitory immune checkpoint receptor, is increasingly studied as a therapeutic target in immune-oncology. Noninvasive molecular imaging of the immune checkpoint programmed death protein-1 (PD-1) or its ligand PD-L1 has shown its value as a strategy to guide and monitor PD-1/PD-L1-targeted immune checkpoint therapy. Yet, radiotracers that allow dynamic, whole body imaging of huLAG-3 expression are not yet described. We here developed single-domain antibodies (sdAbs) that bind huLAG-3 and showed that these sdAbs can image huLAG-3 in tumors, therefore representing promising tools for further development into clinically applicable radiotracers.
Collapse
Affiliation(s)
- Q Lecocq
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - P Debie
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K, 1090, Brussels, Belgium
| | - J Puttemans
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K, 1090, Brussels, Belgium
| | - R M Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - L De Beck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - T Ertveldt
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Y De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - C Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - G Raes
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium.,Cellular and Molecular Immunology Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - M Keyaerts
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K, 1090, Brussels, Belgium.,Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - K Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium.
| | - N Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K, 1090, Brussels, Belgium.
| |
Collapse
|
30
|
Lecocq Q, Awad RM, De Vlaeminck Y, de Mey W, Ertveldt T, Goyvaerts C, Raes G, Thielemans K, Keyaerts M, Devoogdt N, Breckpot K. Single-Domain Antibody Nuclear Imaging Allows Noninvasive Quantification of LAG-3 Expression by Tumor-Infiltrating Leukocytes and Predicts Response of Immune Checkpoint Blockade. J Nucl Med 2021; 62:1638-1644. [PMID: 33712537 PMCID: PMC8612328 DOI: 10.2967/jnumed.120.258871] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/03/2021] [Indexed: 11/29/2022] Open
Abstract
Recent advances in the field of immune-oncology led to the discovery of next-generation immune checkpoints (ICPs). Lymphocyte activation gene-3 (LAG-3), being the most widely studied among them, is being explored as a target for the treatment of cancer patients. Several antagonistic anti-LAG-3 antibodies are being developed and are prime candidates for clinical application. Furthermore, validated therapies targeting cytotoxic T-lymphocyte-associated protein-4, programmed cell-death protein-1, or programmed cell-death ligand-1 showed that only subsets of patients respond. This finding highlights the need for better tools for patient selection and monitoring. The potential of molecular imaging to detect ICPs noninvasively in cancer is supported by several preclinical and clinical studies. Here, we report on a single-domain antibody to evaluate whole-body LAG-3 expression in various syngeneic mouse cancer models using nuclear imaging. Methods: SPECT/CT scans of tumor-bearing mice were performed 1 h after injection with radiolabeled single-domain antibody. Organs and tumors of mice were isolated and evaluated for the presence of the radiolabeled tracer and LAG-3-expressing immune cells using a γ-counter and flow cytometry respectively. PD-1/LAG-3-blocking antibodies were injected in MC38-bearing mice. Results: The radiolabeled single-domain antibody detected LAG-3 expression on tumor-infiltrating lymphocytes (TILs) as soon as 1 h after injection in MC38, MO4, and TC-1 cancer models. The single-domain antibody tracer visualized a compensatory upregulation of LAG-3 on TILs in MC38 tumors of mice treated with PD-1-blocking antibodies. When PD-1 blockade was combined with LAG-3 blockade, a synergistic effect on tumor growth delay was observed. Conclusion: These findings consolidate LAG-3 as a next-generation ICP and support the use of single-domain antibodies as tools to noninvasively monitor the dynamic evolution of LAG-3 expression by TILs, which could be exploited to predict therapy outcome.
Collapse
Affiliation(s)
- Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Wout de Mey
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Geert Raes
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
- Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kris Thielemans
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marleen Keyaerts
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium; and
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium; and
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
31
|
Liu M, Gao Y, Yuan Y, Shi S, Wu J, Tian J, Zhang J. An evidence mapping and scientometric analysis of the top-100 most cited clinical trials of anti-PD-1/PD-L1 drugs to treat cancers. Biomed Pharmacother 2021; 143:112238. [PMID: 34649362 DOI: 10.1016/j.biopha.2021.112238] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE To gain a deeper understanding of the hot topics and future prospects of programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) inhibitors treatment of cancer through scientometric analysis of the top-100 most cited clinical trials. MATERIALS AND METHODS We searched the Web of Science Core Collection database from 1980 to June 2019. Two reviewers independently screened the top-100 most cited clinical trials that defined by the National Institutes of Health starting from the most cited article. Title, year of publication, citations, type of cancer, and focused aspects of outcomes were extracted from included clinical trials. VOSviewer software (version 1.6.9) and Excel 2016 were used to do statistical analysis. The evidence mapping was used to present the relationship between cancers, drugs, citations, and outcomes, etc. RESULTS: The top-100 most cited clinical trials published from 2010 to 2018 in nine journals with high impact factor (IF) (IF2018:6.68-70.67), and Lancet Oncology (USA) published the most clinical trials (n = 29, IF2018 = 35.3856). The total number of citations of the top-100 most cited clinical trials was from 59 to 5606. 920 authors from 34 countries and 458 organizations participated in publishing the top-100 most cited clinical trials. The USA (n = 95) and Memorial Sloan-Kettering Cancer Center (n = 31) contributed the most publications. Based on the evidence mapping, there are 25 different types of cancers (e.g. lung cancer, melanoma, and renal cell cancer) and five focused aspects of outcomes (e.g. safety and efficacy). CONCLUSION The USA was the dominant country. Anti-PD-1/PD-L1 drugs were widely used to treat lung cancer, melanoma, renal cell cancer, and Hodgkin lymphoma. More exploration should be done to explore the use of anti-PD-1/PD-L1 drugs to treat more type of cancers in future research.
Collapse
Affiliation(s)
- Ming Liu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou 730000, China
| | - Ya Gao
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou 730000, China
| | - Yuan Yuan
- Gansu Provincial Maternity and Child-care Hospital, Lanzhou 730000, China
| | - Shuzhen Shi
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou 730000, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100000, China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou 730000, China.
| | - Junhua Zhang
- Evidence-Based Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| |
Collapse
|
32
|
Ma P, Jin X, Fan Z, Wang Z, Yue S, Wu C, Chen S, Wu Y, Chen M, Gu D, Zhang S, Mao R, Fan Y. Super-enhancer receives signals from the extracellular matrix to induce PD-L1-mediated immune evasion via integrin/BRAF/TAK1/ERK/ETV4 signaling. Cancer Biol Med 2021; 19:j.issn.2095-3941.2021.0137. [PMID: 34623791 PMCID: PMC9196059 DOI: 10.20892/j.issn.2095-3941.2021.0137] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE PD-L1 and PD-L2 expression levels determine immune evasion and the therapeutic efficacy of immune checkpoint blockade. The factors that drive inducible PD-L1 expression have been extensively studied, but mechanisms that result in constitutive PD-L1 expression in cancer cells are largely unknown. METHODS DNA elements were deleted in cells by CRISPR/Cas9-mediated knockout. Protein function was inhibited by chemical inhibitors. Protein levels were examined by Western blot, mRNA levels were examined by real-time RT-PCR, and surface protein expression was determined by cellular immunofluorescence and flow cytometry. Immune evasion was examined by in vitro T cell-mediated killing. RESULTS We determined the core regions (chr9: 5, 496, 378-5, 499, 663) of a previously identified PD-L1L2-super-enhancer (SE). Through systematic analysis, we found that the E26 transformation-specific (ETS) variant transcription factor (ETV4) bound to this core DNA region but not to DNA surrounding PD-L1L2SE. Genetic knockout of ETV4 dramatically reduced the expressions of both PD-L1 and PD-L2. ETV4 transcription was dependent on ERK activation, and BRAF/TAK1-induced ERK activation was dependent on extracellular signaling from αvβ3 integrin, which profoundly affected ETV4 transcription and PD-L1/L2 expression. Genetic silencing or pharmacological inhibition of components of the PD-L1L2-SE-associated pathway rendered cancer cells susceptible to T cell-mediated killing. CONCLUSIONS We identified a pathway originating from the extracellular matrix that signaled via integrin/BRAF/TAK1/ERK/ETV4 to PD-L1L2-SE to induce PD-L1-mediated immune evasion. These results provided new insights into PD-L1L2-SE activation and pathways associated with immune checkpoint regulation in cancer.
Collapse
Affiliation(s)
- Panpan Ma
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong 226001, China
- Department of Clinical Laboratory, Yancheng No. 1 People's Hospital, Yancheng 224005, China
| | - Xinxin Jin
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Zhiwei Fan
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Zhou Wang
- School of Life Sciences, Nantong University, Nantong 226001, China
| | - Suhui Yue
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Changyue Wu
- Department of Dermatology, Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| | - Shiyin Chen
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Yuanyuan Wu
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Miaomiao Chen
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
| | - Donghua Gu
- The Department of Urology, the Second Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| | - Siliang Zhang
- The Department of Radiotherapy Oncology, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong 226001, China
| | - Yihui Fan
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong 226001, China
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong 226001, China
| |
Collapse
|
33
|
Pham MM, Ngoi NYL, Peng G, Tan DSP, Yap TA. Development of poly(ADP-ribose) polymerase inhibitor and immunotherapy combinations: progress, pitfalls, and promises. Trends Cancer 2021; 7:958-970. [PMID: 34158277 PMCID: PMC8458234 DOI: 10.1016/j.trecan.2021.05.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/21/2022]
Abstract
The efficacy of poly(ADP-ribose) polymerase inhibitors (PARPi) is restricted by inevitable drug resistance, while their use in combination with chemotherapy and targeted agents is commonly associated with dose-limiting toxicities. Immune checkpoint blockade (ICB) has demonstrated durable responses in different solid tumors and is well-established across multiple cancers. Despite this, single agent activity is limited to a minority of patients and drug resistance remains an issue. Building on the monotherapy success of both drug classes, combining PARPi with ICB may be a safe and well-tolerated strategy with the potential to improve survival outcomes. In this review, we present the preclinical, translational, and clinical data supporting the combination of DNA damage response (DDR) and ICB as well as consider important questions to be addressed with future research.
Collapse
Affiliation(s)
- Melissa M Pham
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalie Y L Ngoi
- Department of Hematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Guang Peng
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S P Tan
- Department of Hematology-Oncology, National University Cancer Institute, National University Health System, Singapore; Cancer Science Institute, National University of Singapore, Singapore
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
34
|
Zhu S, Zhang T, Zheng L, Liu H, Song W, Liu D, Li Z, Pan CX. Combination strategies to maximize the benefits of cancer immunotherapy. J Hematol Oncol 2021; 14:156. [PMID: 34579759 PMCID: PMC8475356 DOI: 10.1186/s13045-021-01164-5] [Citation(s) in RCA: 344] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/07/2021] [Indexed: 12/15/2022] Open
Abstract
Immunotherapies such as immune checkpoint blockade (ICB) and adoptive cell therapy (ACT) have revolutionized cancer treatment, especially in patients whose disease was otherwise considered incurable. However, primary and secondary resistance to single agent immunotherapy often results in treatment failure, and only a minority of patients experience long-term benefits. This review article will discuss the relationship between cancer immune response and mechanisms of resistance to immunotherapy. It will also provide a comprehensive review on the latest clinical status of combination therapies (e.g., immunotherapy with chemotherapy, radiation therapy and targeted therapy), and discuss combination therapies approved by the US Food and Drug Administration. It will provide an overview of therapies targeting cytokines and other soluble immunoregulatory factors, ACT, virotherapy, innate immune modifiers and cancer vaccines, as well as combination therapies that exploit alternative immune targets and other therapeutic modalities. Finally, this review will include the stimulating insights from the 2020 China Immuno-Oncology Workshop co-organized by the Chinese American Hematologist and Oncologist Network (CAHON), the China National Medical Product Administration (NMPA) and Tsinghua University School of Medicine.
Collapse
Affiliation(s)
- Shaoming Zhu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Tian Zhang
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, DUMC 103861, Durham, NC, 27710, USA
| | - Lei Zheng
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,The Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Hongtao Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,University of Chicago, Chicago, IL, USA
| | - Wenru Song
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Kira Pharmaceuticals, Cambridge, MA, USA
| | - Delong Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,New York Medical College, Valhalla, NY, USA
| | - Zihai Li
- Chinese American Hematologist and Oncologist Network, New York, NY, USA. .,Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, USA.
| | - Chong-Xian Pan
- Chinese American Hematologist and Oncologist Network, New York, NY, USA. .,Harvard Medical School, West Roxbury, MA, 02132, USA.
| |
Collapse
|
35
|
Li L, Xu XT, Wang LL, Qin SB, Zhou JY. Expression and clinicopathological significance of Foxp3 and VISTA in cervical cancer. Am J Transl Res 2021; 13:10428-10438. [PMID: 34650712 PMCID: PMC8507058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 01/27/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE To detect the expression differences of Foxp3 and VISTA in chronic cervical inflammation, cervical intraepithelial neoplasia, and cervical cancer, and to explore the role of Foxp3 and VISTA in the development of cervical cancer and the effect of Foxp3 and VISTA on the prognosis of cervical cancer, to provide a theoretical basis for clinical immunotherapy of cervical cancer. METHODS We collected 130 paraffin specimens of cervical tissue, which included 70 cases of cervical cancer tissue, 40 cases of cervical intraepithelial neoplasia tissues and 20 cases of chronic cervicitis. The expression of Foxp3 and VISTA in each group was detected, and the study was conducted based on the clinicopathological characteristics of the patients. The patients were followed up and the prognosis was statistically analyzed. RESULT 1. The expression of Foxp3 and VISTA was statistically different between the cervical cancer group and other groups. 2. Expressions of Foxp3 and VISTA were significantly correlated. 3. In 70 cases of cervical cancer, the expression of Foxp3 and VISTA was related to the clinical stage. 4. The 3-year survival rate of 70 patients with cervical cancer was 72.9%, and there were no factors affecting 3-year OS found. The expression of Foxp3 and VISTA was significantly correlated with the prognosis of cervical cancer. Foxp3 and VISTA double positive expression group had the worst prognosis. CONCLUSION 1. In cervical cancer, the expression of Foxp3 and VISTA was significantly higher than that of cervical intraepithelial neoplasia and chronic cervicitis, which suggested that they were closely related to the occurrence and growth of cervical cancer. 2. The expression of Foxp3 and VISTA was significantly related. 3. The positive expression of Foxp3 and VISTA could be used as independent prognostic factors for cervical cancer prognosis providing a strong basis for cervical cancer immunotherapy.
Collapse
Affiliation(s)
- Li Li
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University Suzhou 215006, China
| | - Xiao-Ting Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University Suzhou 215006, China
| | - Li-Li Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University Suzhou 215006, China
| | - Song-Bing Qin
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University Suzhou 215006, China
| | - Ju-Ying Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University Suzhou 215006, China
| |
Collapse
|
36
|
Expression of Four Autophagy-Related Genes Accurately Predicts the Prognosis of Gastrointestinal Cancer in Asian Patients. DISEASE MARKERS 2021; 2021:7253633. [PMID: 34484469 PMCID: PMC8413069 DOI: 10.1155/2021/7253633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/07/2021] [Accepted: 08/07/2021] [Indexed: 12/11/2022]
Abstract
Gastrointestinal (GI) cancers are among the most fatal diseases in the world. Numerous studies have demonstrated the relationship between autophagy and development of gastrointestinal cancers. However, whether autophagy-related genes can predict prognosis of GI cancers in individuals of Asian ancestry has not been defined. This study, evaluated the prognostic value of autophagy-related genes in gastrointestinal cancer. Expression profile of autophagy-related genes for 296 gastrointestinal cancer patients of Asian ancestry was downloaded from the TCGA database (TCGA-LIHC, TCGA-STAD, TCGA-ESCA, TCGA-PAAD, TCGA-COAD, TCGA-CHOL, and TCGA-READ). The prognostic value of the autophagy-related genes was evaluated using univariate Cox, LASSO, and multivariate Cox regression analyses. The risk score of the autophagy-related gene signature was calculated to assess its predictive prognostic value for GI cancers. Forty-seven differentially expressed autophagy-related genes, in Asian patients with gastrointestinal cancers, were identified. Of the 47 genes, 4 were associated with prognosis of GI cancer (SQSTM1, BIRC5, NRG3, and CXCR4). A prognostic model for GI cancer, based on the expression of the above 4 genes in the training set, showed that cancer patients were stratified into high-risk and low-risk groups (P < 0.05). The utility of the model for overall survival (OS) of GI cancer patients was consistent across the entire set, training set, and test set (entire set: P = 4.568 × 10−4; train set: P = 5.718 × 10−3; test set: P = 3.516 × 10−2). The sensitivity and specificity of the ROC curve of the above prognostic model in predicting the 5-year prognosis of GI cancer was satisfactory (entire set: 0.728; train set: 0.727; test set: 0.733). Analysis of clinical samples validated the overexpression of the 4 genes (SQSTM1, BIRC5, NRG3, and CXCR4) in tumor tissues relative to paired normal tissues, consistent with bioinformatic findings. Expression of the 4 autophagy-related genes (SQSTM1, BIRC5, NRG3, and CXCR4) can accurately predict the prognosis of gastrointestinal tumors in Asian patients.
Collapse
|
37
|
Selective Cytotoxicity of Single and Dual Anti-CD19 and Anti-CD138 Chimeric Antigen Receptor-Natural Killer Cells against Hematologic Malignancies. J Immunol Res 2021; 2021:5562630. [PMID: 34337077 PMCID: PMC8289607 DOI: 10.1155/2021/5562630] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 06/25/2021] [Indexed: 12/03/2022] Open
Abstract
Natural killer (NK) cells are part of the first line of defense that rapidly respond to malignant transformed cells. Chimeric antigen receptor- (CAR-) engineered NK cells, although are still at the preliminary stage, have been shown to be alternative to CAR-T cells, mainly due to the absence of graft-versus-host disease and safer clinical profile. Allogeneic human NK cell line NK-92 cells, equipped by CAR, are being developed for clinical applications. Herein, we designed third-generation CARs, optimized the production protocol, and generated CAR-NK-92 cells, targeting CD19 and/or CD138 antigens that employ CD28, 4-1BB, and CD3ζ signaling, with >80% CAR expression, designated as CD19-NK-92, CD138-NK-92, and dual-NK-92 cells. The generated CAR-NK-92 cells displayed high and selective cytotoxicity toward various corresponding leukemia, lymphoma, and multiple myeloma cell lines in vitro. Multitargeting approach using a mixture of CD19-NK-92 and CD138-NK-92 cells was also evaluated at various ratios to test the idea of personalized formulation to match the patients' antigen expression profile. Our data indicate that increasing the ratio of CD19-NK-92 to CD138-NK-92 could improve NK cytotoxicity in leukemia cells with a relatively higher expression of CD19 over CD138, supporting the personalized proof of concept. This information represents the basis for further in vivo studies and future progress to clinical trials.
Collapse
|
38
|
Peña-Cardelles JF, Salgado-Peralvo AO, Garrido-Martínez P, Cebrián-Carretero JL, Pozo-Kreilinger JJ, Moro-Rodríguez JE. Oral mucositis. Is it present in the immunotherapy of the immune checkpoint pd1/pd-l1 against oral cancer? A systematic review. Med Oral Patol Oral Cir Bucal 2021; 26:e494-e501. [PMID: 33772569 PMCID: PMC8254888 DOI: 10.4317/medoral.24353] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 03/01/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Oral mucositis (OM) is a painful lesion that takes place in the mucosa of the oral cavity, usually its etiology is associated with drug therapies in cancer patients. It is presented as well-defined ulcers whose painful symptomatology sometimes implies the suspension of oncological treatment or parenteral feeding, being therefore an important adverse effect, marking the evolution of these types of therapies against cancer. The present work aim is to know the prevalence of oral mucositis in oral cancer immunotherapy compared to its prevalence in standard therapy. MATERIAL AND METHODS A protocol was developed for a systematic review following PRISMA® guidelines and a focused question (PICO) was constructed. A comprehensive literature search was conducted on electronic databases including PubMed, the SCOPUS database, the Cochrane library and the Web of Science (WOS). RESULTS Six clinical trials were included that met the different inclusion criteria. In these articles, a discrepancy between the prevalence of OM in patients treated with chemotherapy and patients treated with immunotherapy related to the immune checkpoint PD-1/PD-L1 (Nivolumab and Pembrolizumab) was observed. CONCLUSIONS The prevalence of oral mucositis is lower in new immunotherapy with monoclonal antibodies against oral cancer than drugs used so far (chemotherapy drugs [methotrexate, cisplatin] as well as cetuximab). However, more studies should be carried out to confirm these data.
Collapse
Affiliation(s)
- J-F Peña-Cardelles
- Universidad Rey Juan Carlos Av. de Atenas, S/N 28922, Alcorcón, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
39
|
Chiang AWT, Baghdassarian HM, Kellman BP, Bao B, Sorrentino JT, Liang C, Kuo CC, Masson HO, Lewis NE. Systems glycobiology for discovering drug targets, biomarkers, and rational designs for glyco-immunotherapy. J Biomed Sci 2021; 28:50. [PMID: 34158025 PMCID: PMC8218521 DOI: 10.1186/s12929-021-00746-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer immunotherapy has revolutionized treatment and led to an unprecedented wave of immuno-oncology research during the past two decades. In 2018, two pioneer immunotherapy innovators, Tasuku Honjo and James P. Allison, were awarded the Nobel Prize for their landmark cancer immunotherapy work regarding “cancer therapy by inhibition of negative immune regulation” –CTLA4 and PD-1 immune checkpoints. However, the challenge in the coming decade is to develop cancer immunotherapies that can more consistently treat various patients and cancer types. Overcoming this challenge requires a systemic understanding of the underlying interactions between immune cells, tumor cells, and immunotherapeutics. The role of aberrant glycosylation in this process, and how it influences tumor immunity and immunotherapy is beginning to emerge. Herein, we review current knowledge of miRNA-mediated regulatory mechanisms of glycosylation machinery, and how these carbohydrate moieties impact immune cell and tumor cell interactions. We discuss these insights in the context of clinical findings and provide an outlook on modulating the regulation of glycosylation to offer new therapeutic opportunities. Finally, in the coming age of systems glycobiology, we highlight how emerging technologies in systems glycobiology are enabling deeper insights into cancer immuno-oncology, helping identify novel drug targets and key biomarkers of cancer, and facilitating the rational design of glyco-immunotherapies. These hold great promise clinically in the immuno-oncology field.
Collapse
Affiliation(s)
- Austin W T Chiang
- Department of Pediatrics, University of California, 9500 Gilman Drive MC 0760, La Jolla, San Diego, CA, 92093, USA. .,The Novo Nordisk Foundation Center for Biosustainability at the University of California, La Jolla, San Diego, CA, 92093, USA.
| | - Hratch M Baghdassarian
- Department of Pediatrics, University of California, 9500 Gilman Drive MC 0760, La Jolla, San Diego, CA, 92093, USA.,The Novo Nordisk Foundation Center for Biosustainability at the University of California, La Jolla, San Diego, CA, 92093, USA.,Bioinformatics and Systems Biology Graduate Program, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Benjamin P Kellman
- Department of Pediatrics, University of California, 9500 Gilman Drive MC 0760, La Jolla, San Diego, CA, 92093, USA.,The Novo Nordisk Foundation Center for Biosustainability at the University of California, La Jolla, San Diego, CA, 92093, USA.,Bioinformatics and Systems Biology Graduate Program, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Bokan Bao
- Department of Pediatrics, University of California, 9500 Gilman Drive MC 0760, La Jolla, San Diego, CA, 92093, USA.,The Novo Nordisk Foundation Center for Biosustainability at the University of California, La Jolla, San Diego, CA, 92093, USA.,Bioinformatics and Systems Biology Graduate Program, University of California, La Jolla, San Diego, CA, 92093, USA
| | - James T Sorrentino
- Department of Pediatrics, University of California, 9500 Gilman Drive MC 0760, La Jolla, San Diego, CA, 92093, USA.,The Novo Nordisk Foundation Center for Biosustainability at the University of California, La Jolla, San Diego, CA, 92093, USA.,Bioinformatics and Systems Biology Graduate Program, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Chenguang Liang
- Department of Pediatrics, University of California, 9500 Gilman Drive MC 0760, La Jolla, San Diego, CA, 92093, USA.,Department of Bioengineering, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Chih-Chung Kuo
- Department of Pediatrics, University of California, 9500 Gilman Drive MC 0760, La Jolla, San Diego, CA, 92093, USA.,The Novo Nordisk Foundation Center for Biosustainability at the University of California, La Jolla, San Diego, CA, 92093, USA.,Department of Bioengineering, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Helen O Masson
- Department of Pediatrics, University of California, 9500 Gilman Drive MC 0760, La Jolla, San Diego, CA, 92093, USA.,Department of Bioengineering, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, 9500 Gilman Drive MC 0760, La Jolla, San Diego, CA, 92093, USA.,The Novo Nordisk Foundation Center for Biosustainability at the University of California, La Jolla, San Diego, CA, 92093, USA.,Department of Bioengineering, University of California, La Jolla, San Diego, CA, 92093, USA.,The National Biologics Facility, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
40
|
Zhao G, Liu C, Wen X, Luan G, Xie L, Guo X. The translational values of TRIM family in pan-cancers: From functions and mechanisms to clinics. Pharmacol Ther 2021; 227:107881. [PMID: 33930453 DOI: 10.1016/j.pharmthera.2021.107881] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/08/2023]
Abstract
Cancer is the second leading cause of human death across the world. Tripartite motif (TRIM) family, with E3 ubiquitin ligase activities in majority of its members, is reported to be involved in multiple cellular processes and signaling pathways. TRIM proteins have critical effects in the regulation of biological behaviors of cancer cells. Here, we discussed the current understanding of the molecular mechanism of TRIM proteins regulation of cancer cells. We also comprehensively reviewed published studies on TRIM family members as oncogenes or tumor suppressors in the oncogenesis, development, and progression of a variety of types of human cancers. Finally, we highlighted that certain TRIM family members are potential molecular biomarkers for cancer diagnosis and prognosis, and potential therapeutic targets.
Collapse
Affiliation(s)
- Guo Zhao
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Chuan Liu
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Xin Wen
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Gan Luan
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Longxiang Xie
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Xiangqian Guo
- Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| |
Collapse
|
41
|
Zhang L, Zhao Y, Tu Q, Xue X, Zhu X, Zhao KN. The Roles of Programmed Cell Death Ligand-1/ Programmed Cell Death-1 (PD-L1/PD-1) in HPV-induced Cervical Cancer and Potential for their Use in Blockade Therapy. Curr Med Chem 2021; 28:893-909. [PMID: 32003657 DOI: 10.2174/0929867327666200128105459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/13/2019] [Accepted: 12/14/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cervical cancer induced by infection with human papillomavirus (HPV) remains a leading cause of mortality for women worldwide although preventive vaccines and early diagnosis have reduced morbidity and mortality. Advanced cervical cancer can only be treated with either chemotherapy or radiotherapy but the outcomes are poor. The median survival for advanced cervical cancer patients is only 16.8 months. METHODS We undertook a structural search of peer-reviewed published studies based on 1). Characteristics of programmed cell death ligand-1/programmed cell death-1(PD-L1/PD-1) expression in cervical cancer and upstream regulatory signals of PD-L1/PD-1 expression, 2). The role of the PD-L1/PD-1 axis in cervical carcinogenesis induced by HPV infection and 3). Whether the PD-L1/PD-1 axis has emerged as a potential target for cervical cancer therapies. RESULTS One hundred and twenty-six published papers were included in the review, demonstrating that expression of PD-L1/PD-1 is associated with HPV-caused cancer, especially with HPV 16 and 18 which account for approximately 70% of cervical cancer cases. HPV E5/E6/E7 oncogenes activate multiple signalling pathways including PI3K/AKT, MAPK, hypoxia-inducible factor 1α, STAT3/NF-kB and microRNA, which regulate PD-L1/PD-1 axis to promote HPV-induced cervical carcinogenesis. The PD-L1/PD-1 axis plays a crucial role in the immune escape of cervical cancer through inhibition of host immune response. Creating an "immune-privileged" site for initial viral infection and subsequent adaptive immune resistance, which provides a rationale for the therapeutic blockade of this axis in HPV-positive cancers. Currently, Phase I/II clinical trials evaluating the effects of PDL1/ PD-1 targeted therapies are in progress for cervical carcinoma, which provide an important opportunity for the application of anti-PD-L1/anti-PD-1 antibodies in cervical cancer treatment. CONCLUSION Recent research developments have led to an entirely new class of drugs using antibodies against the PD-L1/PD-1 thus promoting the body's immune system to fight cancer. The expression and roles of the PD-L1/ PD-1 axis in the progression of cervical cancer provide great potential for using PD-L1/PD-1 antibodies as a targeted cancer therapy.
Collapse
Affiliation(s)
- Lifang Zhang
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325035 Zhejiang, China
| | - Yu Zhao
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Quanmei Tu
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Xiangyang Xue
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325035 Zhejiang, China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Kong-Nan Zhao
- School of Basic Medical Science, Wenzhou Medical University, Wenzhou, 325035 Zhejiang, China
| |
Collapse
|
42
|
Fu D, Wang Z, Tu Y, Peng F. Interactions between Biomedical Micro-/Nano-Motors and the Immune Molecules, Immune Cells, and the Immune System: Challenges and Opportunities. Adv Healthc Mater 2021; 10:e2001788. [PMID: 33506650 DOI: 10.1002/adhm.202001788] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/21/2020] [Indexed: 12/16/2022]
Abstract
Mobile micro- and nano-motors (MNMs) emerge as revolutionary platforms for biomedical applications, including drug delivery, biosensing, non-invasive surgery, and cancer therapy. While for applications in biomedical fields and practical clinical translation, the interactions of these untethered tiny machines with the immune system is an essential issue to be considered. This review highlights the recent approaches of surpassing immune barriers to prevent foreign motors from triggering immune responses. In addition to trials focusing on the function preservation of MNMs, examples of versatile MNMs working with the immune components (immune molecules, immune cells and the whole system) to achieve cancer immunotherapy, immunoassay, and detoxification are outlined. The immune interference part provides researchers an idea about what is the limit presented by the immune components. The coworking part suggests ways to bypass or even utilize the limit. With interdisciplinary cooperation of nanoengineering, materials science, and immunology field, the rationally designed functional MNMs are expected to provide novel opportunities for the biomedical field.
Collapse
Affiliation(s)
- Dongmei Fu
- School of Materials Science and Engineering Sun‐Yat‐sen University Guangzhou 510275 China
| | - Zhen Wang
- School of Materials Science and Engineering Sun‐Yat‐sen University Guangzhou 510275 China
| | - Yingfeng Tu
- School of Pharmaceutical Science Southern Medical University Guangzhou 510515 China
| | - Fei Peng
- School of Materials Science and Engineering Sun‐Yat‐sen University Guangzhou 510275 China
| |
Collapse
|
43
|
Guven DC, Sahin TK, Aksun MS, Taban H, Aktepe OH, Aksu NM, Akkaş M, Erman M, Kilickap S, Dizdar O, Aksoy S. Evaluation of emergency departments visits in patients treated with immune checkpoint inhibitors. Support Care Cancer 2021; 29:2029-2035. [PMID: 32851486 DOI: 10.1007/s00520-020-05702-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/19/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND The emergency department (ED) is a crucial encounter point in cancer care. Yet, data on the causes of ED visits are limited in patients treated with immune checkpoint inhibitors (ICI). Therefore, we evaluated ED visits in patients treated with ICIs in attempt to determine the predisposing factors. METHODS We performed a retrospective chart review on adult cancer patients treated with ICIs for any type of cancer in the Hacettepe University Cancer Center. The data on ED visits after the first dose of ICIs to 6 months after the last cycle of ICIs were collected. RESULTS A total of 221 patients were included in the study. The mean age was 58.46 ± 13.87 years, and 65.6% of patients were males. Melanoma was the most common diagnosis (27.6%), followed by kidney and lung cancers. Eighty-three of these patients (37.6%) had at least one emergency department (ED) visit. Most of the ED visits were related to symptoms attributable to the disease burden itself, while immune-related adverse events comprised less than 10% of these visits. While baseline Eastern Cooperative Oncology Group performance status, age, polypharmacy, concomitant chemotherapy, eosinophilia, and lactate dehydrogenase levels did not significantly increase the risk, patients with regular opioid use and baseline neutrophilia (> 8000/mm3) had a statistically significant increased risk of visiting the ED (p = 0.001 and 0.19, respectively). These two factors remained significant in the multivariate analyses. CONCLUSION In this study, almost 40% of ICI-treated patients had ED visits. Collaboration with other specialties like emergency medicine is vital for improving the care of patients receiving immunotherapy.
Collapse
Affiliation(s)
- Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey.
| | - Taha Koray Sahin
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Melek Seren Aksun
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Hakan Taban
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Oktay Halit Aktepe
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Nalan Metin Aksu
- Deparment of Emergency Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Meltem Akkaş
- Deparment of Emergency Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Mustafa Erman
- Department of Preventive Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Saadettin Kilickap
- Department of Preventive Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Omer Dizdar
- Department of Preventive Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Sercan Aksoy
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
44
|
Immune Checkpoint Inhibitor-Induced Diabetes Mellitus: Potential Role of T Cells in the Underlying Mechanism. Int J Mol Sci 2021; 22:ijms22042093. [PMID: 33672515 PMCID: PMC7923776 DOI: 10.3390/ijms22042093] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 01/30/2021] [Accepted: 02/02/2021] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy is now a recognized treatment option for several types of cancer. However, some cancer patients treated with immune checkpoint inhibitors (ICIs) are subject to immune-related adverse events, including induced diabetes mellitus. The exact role and molecular/genetic action of ICIs in diabetes are still not well understood. Elucidating the underlying mechanisms in a proper fashion would allow better refining of biomarkers that would help diagnose patients at risk of altered immune system homeostasis, but would also hold the potential of new therapeutic options for diabetes. In the present narrative review, we propose to discuss the case of autoimmune diabetes following treatment with ICIs and the role of ICIs in the pathophysiology of diabetes. We also present some scarce available data on interesting potential immune therapies for diabetes.
Collapse
|
45
|
Li T, Yao F, An Y, Li X, Duan J, Yang XD. Novel Complex of PD-L1 Aptamer and Holliday Junction Enhances Antitumor Efficacy in Vivo. Molecules 2021; 26:1067. [PMID: 33670583 PMCID: PMC7921949 DOI: 10.3390/molecules26041067] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 12/11/2022] Open
Abstract
Blocking the PD-1/PD-L1 pathway can diminish immunosuppression and enhance anticancer immunity. PD-1/PD-L1 blockade can be realized by aptamers, which have good biocompatibility and can be synthesized in quantity economically. For in vivo applications, aptamers need to evade renal clearance and nuclease digestion. Here we investigated whether DNA nanostructures could be used to enhance the function of PD-L1 aptamers. Four PD-L1 aptamers (Apt) were built into a Holliday Junction (HJ) to form a tetravalent DNA nanostructure (Apt-HJ). The average size of Apt-HJ was 13.22 nm, which was above the threshold for renal clearance. Apt-HJ also underwent partial phosphorothioate modification and had improved nuclease resistance. Compared with the monovalent PD-L1 aptamer, the tetravalent Apt-HJ had stronger affinity to CT26 colon cancer cells. Moreover, Apt-HJ markedly boosted the antitumor efficacy in vivo vs. free PD-L1 aptamers without raising systemic toxicity. The results indicate that multiple aptamers attached to a DNA nanostructure may significantly improve the function of PD-L1 aptamers in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | - Xian-Da Yang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; (T.L.); (F.Y.); (Y.A.); (X.L.); (J.D.)
| |
Collapse
|
46
|
Kazemi M, Jafarzadeh A, Nemati M, Taghipour F, Oladpour O, Rezayati MT, Khorramdelazad H, Hassan ZM. Zingerone improves the immune responses in an animal model of breast cancer. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2021; 18:303-310. [PMID: 33544516 DOI: 10.1515/jcim-2019-0135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/28/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The potent anti-tumorigenic effects were attributed to ginger and there are some reports regarding the anti-cancer and immunomodulatory properties ginger-derived components. This study aimed to investigate the effects of zingerone on some immune-related parameters in an animal model of breast cancer. METHODS The breast cancer was established in female BALB/c mice using a carcinogenic 4T1 cell line. At day 10 after cancer induction, tumor-bearing mice were divided into five groups and treated intraperitoneal (daily from days 11-30) with saline or zingerone (at doses 10, 20, 50 and 100 mg/kg/day). The mice were sacrificed on day 31 and the number of splenic Th1- and Treg cells, the expression of IFN-γ and TGF-β in the blood mononuclear cells, the antibody production against sheep red blood cell (SRBC) were determined using flow cytometry, real time-PCR and a standard hemagglutination assay, respectively. RESULTS Zingerone at doses 50 and 100 mg/kg enhanced the number of splenic Th1 cells (p<0.03 and 0.007, respectively); at doses 10, 20, 50 and 100 mg/kg reduced the number of splenic Treg cells (p<0.02, 0.01, and 0.01, respectively), at doses 50 and 100 mg/kg enhanced the expression of IFN-γ (p<0.03), at doses 50 and 100 mg/kg reduced the expression of TGF-β, at doses 50 mg/kg reduced the titer of anti-SRBC antibody (p<0.05). CONCLUSIONS Zingerone improve the T cell-mediated and antibody responses in a mouse model of breast cancer. The immunotherapeutic potentials of zingerone in cancers need more considerations.
Collapse
Affiliation(s)
- Modje Kazemi
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Islamic Republic of Iran
| | - Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Islamic Republic of Iran
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Islamic Republic of Iran
| | - Maryam Nemati
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Islamic Republic of Iran
- Department of Laboratory Sciences, Para-Medical School, Kerman University of Medical Sciences, Kerman, Islamic Republic of Iran
| | - Fereshteh Taghipour
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Islamic Republic of Iran
| | - Omolbanin Oladpour
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Islamic Republic of Iran
| | - Mohammad Taghi Rezayati
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Islamic Republic of Iran
| | - Hossain Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Islamic Republic of Iran
| | - Zuhair Mohammad Hassan
- Department of Immunology, School of Medicine, Tarbiat Modarres University, Tehran, Islamic Republic of Iran
| |
Collapse
|
47
|
Zhou Y, Wang Y, Chen H, Xu Y, Luo Y, Deng Y, Zhang J, Shao A. Immuno-oncology: are TAM receptors in glioblastoma friends or foes? Cell Commun Signal 2021; 19:11. [PMID: 33509214 PMCID: PMC7841914 DOI: 10.1186/s12964-020-00694-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/01/2020] [Indexed: 12/21/2022] Open
Abstract
Tyro3, Axl, and Mertk (TAM) receptors are a subfamily of receptor tyrosine kinases. TAM receptors have been implicated in mediating efferocytosis, regulation of immune cells, secretion of inflammatory factors, and epithelial-to-mesenchymal transition in the tumor microenvironment, thereby serving as a critical player in tumor development and progression. The pro-carcinogenic role of TAM receptors has been widely confirmed, overexpression of TAM receptors is tied to tumor cells growth, metastasis, invasion and treatment resistance. Nonetheless, it is surprising to detect that inhibiting TAM signaling is not all beneficial in the tumor immune microenvironment. The absence of TAM receptors also affects anti-tumor immunity under certain conditions by modulating different immune cells, as the functional diversification of TAM signaling is closely related to tumor immunotherapy. Glioblastoma is the most prevalent and lethal primary brain tumor in adults. Although research regarding the crosstalk between TAM receptors and glioblastoma remains scarce, it appears likely that TAM receptors possess potential anti-tumor effects rather than portraying a total cancer-driving role in the context of glioblastoma. Accordingly, we doubt whether TAM receptors play a double-sided role in glioblastoma, and propose the Janus-faced TAM Hypothesis as a conceptual framework for comprehending the precise underlying mechanisms of TAMs. In this study, we aim to cast a spotlight on the potential multidirectional effects of TAM receptors in glioblastoma and provide a better understanding for TAM receptor-related targeted intervention. Video Abstract
Collapse
Affiliation(s)
- Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yali Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hailong Chen
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yanyan Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, 211126, Jiangsu, China
| | - Yi Luo
- The Second Affiliated Hospital of Zhejiang University School of Medicine (Changxing Branch), Changxing, Huzhou, 313100, Zhejiang, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
48
|
Fernandez-Rozadilla C, Simões AR, Lleonart ME, Carnero A, Carracedo Á. Tumor Profiling at the Service of Cancer Therapy. Front Oncol 2021; 10:595613. [PMID: 33505911 PMCID: PMC7832432 DOI: 10.3389/fonc.2020.595613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer treatment options have evolved significantly in the past few years. From the initial surgical procedures, to the latest next-generation technologies, we are now in the position to analyze and understand tumors in a one-by-one basis and use that to our advantage to provide with individualized treatment options that may increase patient survival. In this review, we will focus on how tumor profiling has evolved over the past decades to deliver more efficient and personalized treatment options, and how novel technologies can help us envisage the future of precision oncology toward a better management and, ultimately, increased survival.
Collapse
Affiliation(s)
- Ceres Fernandez-Rozadilla
- Grupo de Medicina Xenómica (USC), Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Ana Rita Simões
- Grupo de Medicina Xenómica (USC), Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Matilde E Lleonart
- Biomedical Research in Cancer Stem Cells, Vall d´Hebron Research Institute (VHIR), Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology, CIBERONC, Madrid, Spain
| | - Amancio Carnero
- Spanish Biomedical Research Network Centre in Oncology, CIBERONC, Madrid, Spain.,Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
| | - Ángel Carracedo
- Grupo de Medicina Xenómica (USC), Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain.,Grupo de Medicina Xenómica (USC), Fundación Pública Galega de Medicina Xenómica, Santiago de Compostela, Spain
| |
Collapse
|
49
|
Hong M, Tao S, Zhang L, Diao LT, Huang X, Huang S, Xie SJ, Xiao ZD, Zhang H. RNA sequencing: new technologies and applications in cancer research. J Hematol Oncol 2020; 13:166. [PMID: 33276803 PMCID: PMC7716291 DOI: 10.1186/s13045-020-01005-x] [Citation(s) in RCA: 300] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023] Open
Abstract
Over the past few decades, RNA sequencing has significantly progressed, becoming a paramount approach for transcriptome profiling. The revolution from bulk RNA sequencing to single-molecular, single-cell and spatial transcriptome approaches has enabled increasingly accurate, individual cell resolution incorporated with spatial information. Cancer, a major malignant and heterogeneous lethal disease, remains an enormous challenge in medical research and clinical treatment. As a vital tool, RNA sequencing has been utilized in many aspects of cancer research and therapy, including biomarker discovery and characterization of cancer heterogeneity and evolution, drug resistance, cancer immune microenvironment and immunotherapy, cancer neoantigens and so on. In this review, the latest studies on RNA sequencing technology and their applications in cancer are summarized, and future challenges and opportunities for RNA sequencing technology in cancer applications are discussed.
Collapse
Affiliation(s)
- Mingye Hong
- Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Shuang Tao
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Ling Zhang
- Health Science Center, The University of Texas, Houston, 77030, USA
| | - Li-Ting Diao
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Xuanmei Huang
- Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Shaohui Huang
- Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Shu-Juan Xie
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Zhen-Dong Xiao
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| | - Hua Zhang
- Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
50
|
Small molecules targeting the innate immune cGAS‒STING‒TBK1 signaling pathway. Acta Pharm Sin B 2020; 10:2272-2298. [PMID: 33354501 PMCID: PMC7745059 DOI: 10.1016/j.apsb.2020.03.001] [Citation(s) in RCA: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/20/2020] [Accepted: 02/28/2020] [Indexed: 12/19/2022] Open
Abstract
Multiple cancer immunotherapies including chimeric antigen receptor T cell and immune checkpoint inhibitors (ICIs) have been successfully developed to treat various cancers by motivating the adaptive anti-tumor immunity. Particularly, the checkpoint blockade approach has achieved great clinic success as evidenced by several U.S. Food and Drug Administration (FDA)-approved anti-programmed death receptor 1/ligand 1 or anti-cytotoxic T lymphocyte associated protein 4 antibodies. However, the majority of cancers have low clinical response rates to these ICIs due to poor tumor immunogenicity. Indeed, the cyclic guanosine monophosphate-adenosine monophosphate synthase‒stimulator of interferon genes‒TANK-binding kinase 1 (cGAS‒STING‒TBK1) axis is now appreciated as the major signaling pathway in innate immune response across different species. Aberrant signaling of this pathway has been closely linked to multiple diseases, including auto-inflammation, virus infection and cancers. In this perspective, we provide an updated review on the latest progress on the development of small molecule modulators targeting the cGAS‒STING‒TBK1 signaling pathway and their preclinical and clinical use as a new immune stimulatory therapy. Meanwhile, highlights on the clinical candidates, limitations and challenges, as well as future directions in this field are also discussed. Further, small molecule inhibitors targeting this signaling axis and their potential therapeutic use for various indications are discussed as well.
Collapse
Key Words
- ABZI, amidobenzimidazole
- ACMA, 9-amino-6-chloro-2-methoxyacridine
- AMP, adenosine monophosphate
- ATP, adenosine triphosphate
- Anti-tumor
- BNBC, 6-bromo-N-(naphthalen-1-yl)benzo[d][1,3]dioxole-5-carboxamide
- CBD, cyclic dinucleotide-binding domain
- CDA, cyclic diadenosine monophosphate (c-di-AMP)
- CDG, cyclic diguanosine monophosphate (c-di-GMP)
- CDN, cyclic dinucleotide
- CMA, 10-carboxymethyl-9-acridanone
- CTD, C-terminal domain
- CTLA-4, cytotoxic T lymphocyte associated protein 4
- CTT, C-terminal tail
- CXCL, chemokine (C-X-C motif) ligand
- DC50, concentration for 50% degradation
- DCs, dendritic cells
- DMXAA, 5,6-dimethylxanthenone-4-acetic acid
- DSDP, dispiro diketopiperzine
- EM, cryo-electron microscopy
- ENPP1, ecto-nucleotide pyrophosphatase/phosphodiesterase
- ER, endoplasmic reticulum
- FAA, flavone-8-acetic acid
- FDA, U.S. Food and Drug Administration
- FP, fluorescence polarization
- GMP, guanosine monophosphate
- GTP, guanosine triphosphate
- HCQ, hydrochloroquine
- HTS, high throughput screening
- ICI, immune checkpoint inhibitor
- IKK, IκB kinase
- IO, immune-oncology
- IRF3, interferon regulatory factor 3
- ISG, interferon stimulated gene
- ITC, isothermal titration calorimetry
- Immunotherapy
- KD, kinase domain
- LBD, ligand-binding domain
- MDCK, Madin–Darby canine kidney
- MG, Mangostin
- MI, maximum induction
- MLK, mixed lineage kinase
- MinEC5×, minimum effective concentration for inducing 5-fold luciferase activity
- NF-κB, nuclear factor-κB
- Ntase, nucleotidyl transferase
- PBMCs, peripheral-blood mononuclear cells
- PD-1, programmed death receptor 1
- PD-L1, programmed death ligand 1
- PDE, phosphodiesterases
- PDK1, 3-phosphoinositide-dependent protein kinase 1
- PPi, pyrophosphoric acid
- PROTACs, proteolysis targeting chimeras
- PRRs, pattern recognition receptors
- QC, quinacrine
- SAR, structure–activity relationship
- SDD, scaffold and dimerization domain
- STAT, signal transducer and activator of transcription
- STING
- STING, stimulator of interferon genes
- Small molecule modulators
- TBK1
- TBK1, TANK-binding kinase 1
- THIQCs, tetrahydroisoquinolone acetic acids
- TNFRSF, tumor necrosis factor receptor superfamily
- ULD, ubiquitin-like domain
- VHL, von Hippel–Lindau
- cAIMP, cyclic adenosine-inosine monophosphate
- cGAMP, cyclic guanosine monophosphate-adenosine monophosphate
- cGAS
- cGAS, cyclic guanosine monophosphate-adenosine monophosphate synthase
- dsDNA, double-stranded DNA
- i.t., intratumoral
Collapse
|