1
|
Liu X, Zhang W, Wei S, Liang X, Luo B. Targeting cuproptosis with nano material: new way to enhancing the efficacy of immunotherapy in colorectal cancer. Front Pharmacol 2024; 15:1451067. [PMID: 39691393 PMCID: PMC11649426 DOI: 10.3389/fphar.2024.1451067] [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: 06/18/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Colorectal cancer has emerged as one of the predominant malignant tumors globally. Immunotherapy, as a novel therapeutic methodology, has opened up new possibilities for colorectal cancer patients. However, its actual clinical efficacy requires further enhancement. Copper, as an exceptionally crucial trace element, can influence various signaling pathways, gene expression, and biological metabolic processes in cells, thus playing a critical role in the pathogenesis of colorectal cancer. Recent studies have revealed that cuproptosis, a novel mode of cell death, holds promise to become a potential target to overcome resistance to colorectal cancer immunotherapy. This shows substantial potential in the combination treatment of colorectal cancer. Conveying copper into tumor cells via a nano-drug delivery system to induce cuproptosis of colorectal cancer cells could offer a potential strategy for eliminating drug-resistant colorectal cancer cells and vastly improving the efficacy of immunotherapy while ultimately destroy colorectal tumors. Moreover, combining the cuproptosis induction strategy with other anti-tumor approaches such as photothermal therapy, photodynamic therapy, and chemodynamic therapy could further enhance its therapeutic effect. This review aims to illuminate the practical significance of cuproptosis and cuproptosis-inducing nano-drugs in colorectal cancer immunotherapy, and scrutinize the current challenges and limitations of this methodology, thereby providing innovative thoughts and references for the advancement of cuproptosis-based colorectal cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Xiangdong Liu
- Department of Radiotherapy Center, Hubei Cancer Hospital, The Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
| | - Wanqiu Zhang
- Department of Radiotherapy Center, Hubei Cancer Hospital, The Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
| | - Shaozhong Wei
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
- Department of Gastrointestinal Oncology Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinjun Liang
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
- Department of Abdominal Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Luo
- Department of Radiotherapy Center, Hubei Cancer Hospital, The Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
| |
Collapse
|
2
|
Kos M, Bojarski K, Mertowska P, Mertowski S, Tomaka P, Zaborek-Łyczba M, Łyczba J, Dziki Ł, Grywalska E. Can the Analysis of Toll-like Receptors (TLR) on NK and NKT-like Cells Improve Gastric Cancer Diagnostics and Treatment? Cancers (Basel) 2024; 16:3854. [PMID: 39594809 PMCID: PMC11592653 DOI: 10.3390/cancers16223854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: The aim of this study was to determine the assessment of the percentage of NK and NKT-like cells expressing Toll-like receptors (TLR-2, TLR-3, TLR-4, and TLR-9) in patients with gastric cancer (GC) compared with healthy volunteers (HV) and to investigate differences according to cancer subtype. We also assessed TLR gene expression by RT-qPCR to assess whether TLRs could be diagnostic and prognostic biomarkers. Methods: The study included 86 patients with histologically confirmed gastric cancer and 30 healthy volunteers. Peripheral blood samples were collected from the participants, and TLR expression on NK and NKT-like cells was assessed by flow cytometry and RT-qPCR. The expression of TLR2, TLR3, TLR4, and TLR9 genes was assessed using genetic material derived from NK and NKT-like cells sourced from PBMC. The obtained results were statistically analyzed using Mann-Whitney U and Kruskal-Wallis tests, and the predictive ability of variables was assessed using ROC curve analysis. Results: A significantly higher expression of TLR receptors (TLR-2, TLR-3, TLR-4, and TLR-9) was found in patients with gastric cancer compared to healthy volunteers (p < 0.05). TLR expression also differed depending on the cancer subtype, and higher expression was observed in more advanced GC subtypes. RT-qPCR analysis showed significantly increased expression of TLR genes in the group of GC patients. ROC curves indicate a high ability of TLRs to differentiate between GC patients and healthy individuals. Conclusions: The expression of TLRs on NK and NKT-like cells is clearly increased in patients with gastric cancer, especially in more advanced subtypes of the tumor. The results suggest that TLRs could potentially be used as diagnostic and prognostic biomarkers and represent potential targets for immune therapies in GC. However, further studies are needed to determine the functional role of TLRs in disease progression and the possibility of their use in personalized treatment.
Collapse
Affiliation(s)
- Marek Kos
- Department of Public Health, Medical University of Lublin, 1 Chodźki Street, 20-093 Lublin, Poland
| | - Krzysztof Bojarski
- General Surgery Department, Independent Public Health Care Center in Łęczna (SP ZOZ in Leczna), 52 Krasnystawska Street, 21-010 Leczna, Poland
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodźki Street, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodźki Street, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Piotr Tomaka
- Department of Anesthesiology and Intensive Care, Independent Public Health Care Center in Łęczna (SP ZOZ in Leczna), 52 Krasnystawska Street, 21-010 Leczna, Poland
| | - Monika Zaborek-Łyczba
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodźki Street, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Jakub Łyczba
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodźki Street, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Łukasz Dziki
- Department of General and Oncological Surgery, Faculty of Medicine, Medical University of Lodz, 251 Street, 92-213 Lodz, Poland
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodźki Street, 20-093 Lublin, Poland; (S.M.); (E.G.)
| |
Collapse
|
3
|
Li J, Zhang W, Chen L, Wang X, Liu J, Huang Y, Qi H, Chen L, Wang T, Li Q. Targeting extracellular matrix interaction in gastrointestinal cancer: Immune modulation, metabolic reprogramming, and therapeutic strategies. Biochim Biophys Acta Rev Cancer 2024; 1879:189225. [PMID: 39603565 DOI: 10.1016/j.bbcan.2024.189225] [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: 09/20/2024] [Revised: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
The extracellular matrix (ECM) is a major constituent of the tumor microenvironment, acting as a mediator that supports the progression of gastrointestinal (GI) cancers, particularly in mesenchymal subtypes. Beyond providing structural support, the ECM actively shapes the tumor microenvironment (TME) through complex biochemical and biomechanical remodeling. Dysregulation of ECM composition and signaling is closely linked to increased cancer aggressiveness, poor prognosis, and resistance to therapy. ECM components, such as collagen, fibronectin, laminin, and periostin, influence tumor growth, metastasis, immune modulation, and metabolic reprogramming by interacting with tumor cells, immune cells, and cancer-associated fibroblasts. In this review, we highlight the heterogeneous nature of the ECM and the dualistic roles of its components across GI cancers, with a focus on their contributions to immune evasion and metabolic remodeling via intercellular interactions. Additionally, we explore therapeutic strategies targeting ECM remodeling and ECM-centered interactions, emphasizing their potential in enhancing existing anti-tumor therapies.
Collapse
Affiliation(s)
- Jiyifan Li
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenxin Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Lu Chen
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinhai Wang
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiafeng Liu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuxin Huang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Huijie Qi
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Li Chen
- Department of Pharmacy, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China
| | - Tianxiao Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China.
| | - Qunyi Li
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Tu Y, Wang S, Wang H, Zhang P, Wang M, Liu C, Yang C, Jiang R. The role of perioperative factors in the prognosis of cancer patients: A coin has two sides. J Biomed Res 2024; 38:1-12. [PMID: 39314042 DOI: 10.7555/jbr.38.20240164] [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: 09/25/2024] Open
Abstract
Cancer, the second leading cause of mortality globally, poses a significant health challenge. The conventional treatment for solid tumors involves surgical intervention, followed by chemo- and radio-therapies as well as target therapies, but the recurrence and metastasis of cancers remain a major issue. Anesthesia is essential for ensuring patient comfort and safety during surgical procedures. Despite its crucial role during the surgery, the precise effect of anesthesia on cancer patient outcomes is not clearly understood. This comprehensive review aims to elucidate the various anesthesia strategies used in the perioperative care of cancer patients and their potential effects on patients' prognosis, but understanding the complex relationship between anesthesia and cancer outcomes is crucial, given the complexity in cancer treaments. Examining potential implications of anesthesia strategies on cancer patient prognosis may help better understand treatment efficacy and risk factors of cancer recurrence and metastasis. Through a detailed analysis of anesthesia practices in cancer surgery, this review aims to provide insights that may lead to improving the existing anesthesia protocols and ultimately reduce risk factors for patient outcomes in the field of oncology.
Collapse
Affiliation(s)
- Yingzhou Tu
- Department of Anesthesiology and Perioperative Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Sen Wang
- Department of Anesthesiology and Perioperative Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Haoran Wang
- Department of Anesthesiology and Perioperative Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Peiyao Zhang
- Department of Anesthesiology and Perioperative Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mengyu Wang
- Department of Anesthesiology and Perioperative Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Department of Radiation Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Riyue Jiang
- Department of Radiation Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
5
|
Bakhtiyaridovvombaygi M, Yazdanparast S, Kheyrandish S, Safdari SM, Amiri Samani F, Sohani M, Jaafarian AS, Damirchiloo F, Izadpanah A, Parkhideh S, Mikanik F, Roshandel E, Hajifathali A, Gharehbaghian A. Harnessing natural killer cells for refractory/relapsed non-Hodgkin lymphoma: biological roles, clinical trials, and future prospective. Biomark Res 2024; 12:66. [PMID: 39020411 PMCID: PMC11253502 DOI: 10.1186/s40364-024-00610-z] [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: 04/11/2024] [Accepted: 06/28/2024] [Indexed: 07/19/2024] Open
Abstract
Non-Hodgkin lymphomas (NHLs) are heterogeneous and are among the most common hematological malignancies worldwide. Despite the advances in the treatment of patients with NHLs, relapse or resistance to treatment is anticipated in several patients. Therefore, novel therapeutic approaches are needed. Recently, natural killer (NK) cell-based immunotherapy alone or in combination with monoclonal antibodies, chimeric antigen receptors, or bispecific killer engagers have been applied in many investigations for NHL treatment. The functional defects of NK cells and the ability of cancerous cells to escape NK cell-mediated cytotoxicity within the tumor microenvironment of NHLs, as well as the beneficial results from previous studies in the context of NK cell-based immunotherapy in NHLs, direct our attention to this therapeutic strategy. This review aims to summarize clinical studies focusing on the applications of NK cells in the immunotherapy of patients with NHL.
Collapse
Affiliation(s)
- Mehdi Bakhtiyaridovvombaygi
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Yazdanparast
- Department of Hematology and Blood Banking, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Setare Kheyrandish
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mehrab Safdari
- Departments of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fateme Amiri Samani
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Mahsa Sohani
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Akram Sadat Jaafarian
- Departments of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fateme Damirchiloo
- Departments of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Izadpanah
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Parkhideh
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mikanik
- Laboratory Hematology and Blood Bank Department, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Roshandel
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ahmad Gharehbaghian
- Laboratory Hematology and Blood Bank Department, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Mangiola S, Milton M, Ranathunga N, Li-Wai-Suen C, Odainic A, Yang E, Hutchison W, Garnham A, Iskander J, Pal B, Yadav V, Rossello J, Carey VJ, Morgan M, Bedoui S, Kallies A, Papenfuss AT. A multi-organ map of the human immune system across age, sex and ethnicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.08.542671. [PMID: 38746418 PMCID: PMC11092463 DOI: 10.1101/2023.06.08.542671] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Understanding tissue biology's heterogeneity is crucial for advancing precision medicine. Despite the centrality of the immune system in tissue homeostasis, a detailed and comprehensive map of immune cell distribution and interactions across human tissues and demographics remains elusive. To fill this gap, we harmonised data from 12,981 single-cell RNA sequencing samples and curated 29 million cells from 45 anatomical sites to create a comprehensive compositional and transcriptional healthy map of the healthy immune system. We used this resource and a novel multilevel modelling approach to track immune ageing and test differences across sex and ethnicity. We uncovered conserved and tissue-specific immune-ageing programs, resolved sex-dependent differential ageing and identified ethnic diversity in clinically critical immune checkpoints. This study provides a quantitative baseline of the immune system, facilitating advances in precision medicine. By sharing our immune map, we hope to catalyse further breakthroughs in cancer, infectious disease, immunology and precision medicine.
Collapse
Affiliation(s)
- S Mangiola
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - M Milton
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - N Ranathunga
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Csn Li-Wai-Suen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - A Odainic
- The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - E Yang
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - W Hutchison
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - A Garnham
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - J Iskander
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - B Pal
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
| | - V Yadav
- Systems Biology of Aging Laboratory, Columbia University; New York, USA
| | - Jfj Rossello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia
- Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Victoria, Australia
| | - V J Carey
- Channing Division of Network Medicine, Mass General Brigham, Harvard Medical School, Harvard University, Boston, USA
| | - M Morgan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, NY, USA
| | - S Bedoui
- The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - A Kallies
- The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - A T Papenfuss
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
7
|
Wu H, Geng Q, Shi W, Qiu C. Comprehensive pan-cancer analysis reveals CCDC58 as a carcinogenic factor related to immune infiltration. Apoptosis 2024; 29:536-555. [PMID: 38066393 DOI: 10.1007/s10495-023-01919-0] [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: 11/06/2023] [Indexed: 02/18/2024]
Abstract
CCDC58, a member of the CCDC protein family, has been primarily associated with the malignant progression of hepatocellular carcinoma (HCC) and breast cancer, with limited research conducted on its involvement in other tumor types. We aimed to assess the significance of CCDC58 in pan-cancer. We utilized the TCGA, GTEx, and UALCAN databases to perform the differential expression of CCDC58 at both mRNA and protein levels. Prognostic value was evaluated through univariate Cox regression and Kaplan-Meier methods. Mutation and methylation analyses were conducted using the cBioPortal and SMART databases. We identified genes interacting with and correlated to CCDC58 through STRING and GEPIA2, respectively. Subsequently, we performed GO and KEGG enrichment analyses. To gain insights into the functional status of CCDC58 at the single-cell level, we utilized CancerSEA. We explored the correlation between CCDC58 and immune infiltration as well as immunotherapy using the ESTIMATE package, TIMER2.0, TISIDB, TIDE, TIMSO, and TCIA. We examined the relationship between CCDC58 and tumor heterogeneity, stemness, DNA methyltransferases, and MMR genes. Lastly, we constructed a nomogram based on CCDC58 in HCC and investigated its association with drug sensitivity. CCDC58 expression was significantly upregulated and correlated with poor prognosis across various tumor types. The mutation frequency of CCDC58 was found to be increased in 25 tumors. We observed a negative correlation between CCDC58 expression and the methylation sites in the majority of tumors. CCDC58 showed negative correlations with immune and stromal scores, as well as with NK T cells, Tregs, CAFs, endothelial cells, and immunomodulators. Its value in immunotherapy was comparable to that of tumor mutational burden. CCDC58 exhibited positive correlations with tumor heterogeneity, stemness, DNA methyltransferase genes, and MMR genes. In HCC, CCDC58 was identified as an independent risk factor and demonstrated potential associations with multiple drugs. CCDC58 demonstrates significant clinical value as a prognostic marker and indicator of immune response across various tumor types. Its comprehensive analysis provides insights into its potential implications in pan-cancer research.
Collapse
Affiliation(s)
- Huili Wu
- Department of Endodontics, Zhonglou Hospital, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China
| | - Qing Geng
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Wenxiang Shi
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chenjie Qiu
- Department of General Surgery, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China.
| |
Collapse
|
8
|
Wu CY, Kuo TC, Lin HW, Yang JT, Chen WH, Cheng WF, Tien YW, Chan KC. Immunocyte profiling changes in patients received epidural versus intravenous analgesia after pancreatectomy: A randomized controlled trial. J Formos Med Assoc 2024:S0929-6646(24)00148-7. [PMID: 38494360 DOI: 10.1016/j.jfma.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND Perioperative immunosuppressants, such as surgical stress and opioid use may downregulate anti-cancer immunocytes for patients undergoing pancreatectomy. Thoracic epidural analgesia (TEA) may attenuate these negative effects and provide better anti-cancer immunocyte profile change than intravenous analgesia using opioid. METHODS We randomly assigned 108 adult patients undergoing pancreatectomy to receive one of two 72-h postoperative analgesia protocols: one was TEA, and the other was intravenous patient-controlled analgesia (IV-PCA). The perioperative proportional changes of immunocytes relevant to anticancer immunity-namely natural killer (NK) cells, cytotoxic T cells, helper T cells, mature dendritic cells, and regulatory T (Treg) cells were determined at 1 day before surgery, at the end of surgery and on postoperative day 1,4 and 7 using flow cytometry. In addition, the progression-free survival and overall survival between the two groups were compared. RESULTS After surgery, the proportions of NK cells and cytotoxic T cells were significantly decreased; the proportion of B cells and mature dendritic cells and Treg cells were significantly increased. However, the proportions of helper T cells exhibited no significant change. These results were comparable between the two groups. Furthermore, there were no significant differences in progression-free survival (52.75 [39.96] and 57.48 [43.66] months for patients in the TEA and IV-PCA groups, respectively; p = 0.5600) and overall survival (62.71 [35.48] and 75.11 [33.10] months for patients in the TEA and IV-PCA groups, respectively; p = 0.0644). CONCLUSIONS TEA was neither associated with favorable anticancer immunity nor favorable oncological outcomes for patients undergoing pancreatectomy.
Collapse
Affiliation(s)
- Chun-Yu Wu
- Department of Anesthesiology, National Taiwan University Hospital, Hsinchu branch, Hsinchu, Taiwan
| | - Ting-Chun Kuo
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Han-Wei Lin
- Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Jen-Ting Yang
- Department of Health Services, University of Washington, Seattle, United States
| | - Wen-Hsiu Chen
- Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Fang Cheng
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Wen Tien
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuang-Cheng Chan
- Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
9
|
Samareh Salavatipour M, Poursalehi Z, Hosseini Rouzbahani N, Mohammadyar S, Vasei M. CRISPR-Cas9 in basic and translational aspects of cancer therapy. BIOIMPACTS : BI 2024; 14:30087. [PMID: 39493894 PMCID: PMC11530967 DOI: 10.34172/bi.2024.30087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 11/05/2024]
Abstract
Introduction The discovery of gene editing techniques has opened a new era within the field of biology and enabled scientists to manipulate nucleic acid molecules. CRISPR-Cas9 genome engineering has revolutionized this achievement by successful targeting the DNA molecule and editing its sequence. Since genomic changes are the basis of the birth and growth of many tumors, CRISPR-Cas9 method has been successfully applied to identify and manipulate the genes which are involved in initiating and driving some neoplastic processes. Methods By review of the existing literature on application of CRISPR-Cas9 in cancer, different databases, such as PubMed and Google Scholar, we started data collection for "CRISPR-Cas9", "Genome Editing", "Cancer", "Solid tumors", "Hematologic malignancy" "Immunotherapy", "Diagnosis", "Drug resistance" phrases. Clinicaltrials.gov, a resource that provides access to information on clinical trials, was also searched in this review. Results We have defined the basics of this technology and then mentioned some clinical and preclinical studies using this technology in the treatment of a variety of solid tumors as well as hematologic neoplasms. Finally, we described the progress made by this technology in boosting immune-mediated cell therapy in oncology, such as CAR-T cells, CAR-NK cells, and CAR-M cells. Conclusion CRISPR-Cas9 system revolutionized the therapeutic strategies in some solid malignant tumors and leukemia through targeting the key genes involved in the pathogenesis of these cancers.
Collapse
Affiliation(s)
- Maryam Samareh Salavatipour
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Poursalehi
- Department of Applied Cell Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Negin Hosseini Rouzbahani
- Department of Medical Immunology, Faculty of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Sohaib Mohammadyar
- Department of Hematology and Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Vasei
- Gene Therapy Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Maia A, Tarannum M, Lérias JR, Piccinelli S, Borrego LM, Maeurer M, Romee R, Castillo-Martin M. Building a Better Defense: Expanding and Improving Natural Killer Cells for Adoptive Cell Therapy. Cells 2024; 13:451. [PMID: 38474415 DOI: 10.3390/cells13050451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/01/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Natural killer (NK) cells have gained attention as a promising adoptive cell therapy platform for their potential to improve cancer treatments. NK cells offer distinct advantages over T-cells, including major histocompatibility complex class I (MHC-I)-independent tumor recognition and low risk of toxicity, even in an allogeneic setting. Despite this tremendous potential, challenges persist, such as limited in vivo persistence, reduced tumor infiltration, and low absolute NK cell numbers. This review outlines several strategies aiming to overcome these challenges. The developed strategies include optimizing NK cell expansion methods and improving NK cell antitumor responses by cytokine stimulation and genetic manipulations. Using K562 cells expressing membrane IL-15 or IL-21 with or without additional activating ligands like 4-1BBL allows "massive" NK cell expansion and makes multiple cell dosing and "off-the-shelf" efforts feasible. Further improvements in NK cell function can be reached by inducing memory-like NK cells, developing chimeric antigen receptor (CAR)-NK cells, or isolating NK-cell-based tumor-infiltrating lymphocytes (TILs). Memory-like NK cells demonstrate higher in vivo persistence and cytotoxicity, with early clinical trials demonstrating safety and promising efficacy. Recent trials using CAR-NK cells have also demonstrated a lack of any major toxicity, including cytokine release syndrome, and, yet, promising clinical activity. Recent data support that the presence of TIL-NK cells is associated with improved overall patient survival in different types of solid tumors such as head and neck, colorectal, breast, and gastric carcinomas, among the most significant. In conclusion, this review presents insights into the diverse strategies available for NK cell expansion, including the roles played by various cytokines, feeder cells, and culture material in influencing the activation phenotype, telomere length, and cytotoxic potential of expanded NK cells. Notably, genetically modified K562 cells have demonstrated significant efficacy in promoting NK cell expansion. Furthermore, culturing NK cells with IL-2 and IL-15 has been shown to improve expansion rates, while the presence of IL-12 and IL-21 has been linked to enhanced cytotoxic function. Overall, this review provides an overview of NK cell expansion methodologies, highlighting the current landscape of clinical trials and the key advancements to enhance NK-cell-based adoptive cell therapy.
Collapse
Affiliation(s)
- Andreia Maia
- Molecular and Experimental Pathology Laboratory, Champalimaud Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
- NK Cell Gene Manipulation and Therapy Laboratory, Division of Cellular Therapy and Stem Cell Transplant, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- NOVA Medical School, NOVA University of Lisbon, 1099-085 Lisbon, Portugal
| | - Mubin Tarannum
- NK Cell Gene Manipulation and Therapy Laboratory, Division of Cellular Therapy and Stem Cell Transplant, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Joana R Lérias
- ImmunoTherapy/ImmunoSurgery, Champalimaud Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Sara Piccinelli
- NK Cell Gene Manipulation and Therapy Laboratory, Division of Cellular Therapy and Stem Cell Transplant, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Luis Miguel Borrego
- Comprehensive Health Research Centre (CHRC), NOVA Medical School, Faculdade de Ciências Médicas (FCM), NOVA University of Lisbon, 1099-085 Lisbon, Portugal
- Immunoallergy Department, Hospital da Luz, 1600-209 Lisbon, Portugal
| | - Markus Maeurer
- ImmunoTherapy/ImmunoSurgery, Champalimaud Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
- I Medical Clinic, University of Mainz, 55131 Mainz, Germany
| | - Rizwan Romee
- NK Cell Gene Manipulation and Therapy Laboratory, Division of Cellular Therapy and Stem Cell Transplant, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Mireia Castillo-Martin
- Molecular and Experimental Pathology Laboratory, Champalimaud Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Pathology Service, Champalimaud Clinical Center, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| |
Collapse
|
11
|
Taheri M, Tehrani HA, Daliri F, Alibolandi M, Soleimani M, Shoari A, Arefian E, Ramezani M. Bioengineering strategies to enhance the interleukin-18 bioactivity in the modern toolbox of cancer immunotherapy. Cytokine Growth Factor Rev 2024; 75:65-80. [PMID: 37813764 DOI: 10.1016/j.cytogfr.2023.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023]
Abstract
Cytokines are the first modern immunotherapeutic agents used for activation immunotherapy. Interleukin-18 (IL-18) has emerged as a potent anticancer immunostimulatory cytokine over the past three decades. IL-18, structurally is a stable protein with very low toxicity at biological doses. IL-18 promotes the process of antigen presentation and also enhances innate and acquired immune responses. It can induce the production of proinflammatory cytokines and increase tumor infiltration of effector immune cells to revert the immunosuppressive milieu of tumors. Furthermore, IL-18 can reduce tumorigenesis, suppress tumor angiogenesis, and induce tumor cell apoptosis. These characteristics present IL-18 as a promising option for cancer immunotherapy. Although several preclinical studies have reported the immunotherapeutic potential of IL-18, clinical trials using it as a monotherapy agent have reported disappointing results. These results may be due to some biological characteristics of IL-18. Several bioengineering approaches have been successfully used to correct its defects as a bioadjuvant. Currently, the challenge with this anticancer immunotherapeutic agent is mainly how to use its capabilities in a rational combinatorial therapy for clinical applications. The present study discussed the strengths and weaknesses of IL-18 as an immunotherapeutic agent, followed by comprehensive review of various promising bioengineering approaches that have been used to overcome its disadvantages. Finally, this study highlights the promising application of IL-18 in modern combinatorial therapies, such as chemotherapy, immune checkpoint blockade therapy, cell-based immunotherapy and cancer vaccines to guide future studies, circumventing the barriers to administration of IL-18 for clinical applications, and bring it to fruition as a potent immunotherapy agent in cancer treatment.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | | | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoud Soleimani
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Iran
| | - Alireza Shoari
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Li Y, Sui S, Goel A. Extracellular vesicles associated microRNAs: Their biology and clinical significance as biomarkers in gastrointestinal cancers. Semin Cancer Biol 2024; 99:5-23. [PMID: 38341121 DOI: 10.1016/j.semcancer.2024.02.001] [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: 10/13/2023] [Revised: 01/26/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024]
Abstract
Gastrointestinal (GI) cancers, including colorectal, gastric, esophageal, pancreatic, and liver, are associated with high mortality and morbidity rates worldwide. One of the underlying reasons for the poor survival outcomes in patients with these malignancies is late disease detection, typically when the tumor has already advanced and potentially spread to distant organs. Increasing evidence indicates that earlier detection of these cancers is associated with improved survival outcomes and, in some cases, allows curative treatments. Consequently, there is a growing interest in the development of molecular biomarkers that offer promise for screening, diagnosis, treatment selection, response assessment, and predicting the prognosis of these cancers. Extracellular vesicles (EVs) are membranous vesicles released from cells containing a repertoire of biological molecules, including nucleic acids, proteins, lipids, and carbohydrates. MicroRNAs (miRNAs) are the most extensively studied non-coding RNAs, and the deregulation of miRNA levels is a feature of cancer cells. EVs miRNAs can serve as messengers for facilitating interactions between tumor cells and the cellular milieu, including immune cells, endothelial cells, and other tumor cells. Furthermore, recent years have witnessed considerable technological advances that have permitted in-depth sequence profiling of these small non-coding RNAs within EVs for their development as promising cancer biomarkers -particularly non-invasive, liquid biopsy markers in various cancers, including GI cancers. Herein, we summarize and discuss the roles of EV-associated miRNAs as they play a seminal role in GI cancer progression, as well as their promising translational and clinical potential as cancer biomarkers as we usher into the area of precision oncology.
Collapse
Affiliation(s)
- Yuan Li
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA, USA; Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Silei Sui
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA, USA; Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA, USA.
| |
Collapse
|
13
|
Ahluwalia P, Ballur K, Leeman T, Vashisht A, Singh H, Omar N, Mondal AK, Vaibhav K, Baban B, Kolhe R. Incorporating Novel Technologies in Precision Oncology for Colorectal Cancer: Advancing Personalized Medicine. Cancers (Basel) 2024; 16:480. [PMID: 38339232 PMCID: PMC10854941 DOI: 10.3390/cancers16030480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 02/12/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most heterogeneous and deadly diseases, with a global incidence of 1.5 million cases per year. Genomics has revolutionized the clinical management of CRC by enabling comprehensive molecular profiling of cancer. However, a deeper understanding of the molecular factors is needed to identify new prognostic and predictive markers that can assist in designing more effective therapeutic regimens for the improved management of CRC. Recent breakthroughs in single-cell analysis have identified new cell subtypes that play a critical role in tumor progression and could serve as potential therapeutic targets. Spatial analysis of the transcriptome and proteome holds the key to unlocking pathogenic cellular interactions, while liquid biopsy profiling of molecular variables from serum holds great potential for monitoring therapy resistance. Furthermore, gene expression signatures from various pathways have emerged as promising prognostic indicators in colorectal cancer and have the potential to enhance the development of equitable medicine. The advancement of these technologies for identifying new markers, particularly in the domain of predictive and personalized medicine, has the potential to improve the management of patients with CRC. Further investigations utilizing similar methods could uncover molecular subtypes specific to emerging therapies, potentially strengthening the development of personalized medicine for CRC patients.
Collapse
Affiliation(s)
- Pankaj Ahluwalia
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Kalyani Ballur
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Tiffanie Leeman
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Ashutosh Vashisht
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Harmanpreet Singh
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Nivin Omar
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Ashis K. Mondal
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Kumar Vaibhav
- Department of Neurosurgery, Augusta University, Augusta, GA 30912, USA;
| | - Babak Baban
- Departments of Neurology and Surgery, Augusta University, Augusta, GA 30912, USA;
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| |
Collapse
|
14
|
Xie Y, Jiang H. The exploration of mitochondrial-related features helps to reveal the prognosis and immunotherapy methods of colorectal cancer. Cancer Rep (Hoboken) 2024; 7:e1914. [PMID: 37903487 PMCID: PMC10809275 DOI: 10.1002/cnr2.1914] [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: 07/06/2023] [Revised: 09/01/2023] [Accepted: 09/29/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Cancer cell survival, proliferation, and metabolism are all intertwined with mitochondria. However, a complete description of how the features of mitochondria relate to the tumor microenvironment (TME) and immunological landscape of colorectal cancer (CRC) has yet to be made. We performed subgroup analysis on CRC patient data obtained from the databases using non-negative matrix factorization (NMF) clustering. Construct a prognostic model using the mitochondrial-related gene (MRG) risk score, and then compare it to other models for accuracy. Comprehensive analyses of the risk score, in conjunction with the TME and immune landscape, were performed, and the relationship between the model and different types of cell death, radiation and chemotherapy, and drug resistance was investigated. Results from immunohistochemistry and single-cell sequencing were utilized to verify the model genes, and a drug sensitivity analysis was conducted to evaluate possible therapeutic medicines. The pan-cancer analysis is utilized to further investigate the role of genes in a wider range of malignancies. METHODS AND RESULTS We found that CRC patients based on MRG were divided into two groups with significant differences in survival outcomes and TME between groups. The predictive power of the risk score was further shown by building a prognostic model and testing it extensively in both internal and external cohorts. Multiple immune therapeutic responses and the expression of immunological checkpoints demonstrate that the risk score is connected to immunotherapy success. The correlation analysis of the risk score provide more ideas and guidance for prognostic models in clinical treatment. CONCLUSION The TME, immune cell infiltration, and responsiveness to immunotherapy in CRC were all thoroughly evaluated on the basis of MRG features. The comparative validation of multiple queues and models combined with clinical data ensures the effectiveness and clinical practicality of MRG features. Our studies help clinicians create individualized treatment programs for individuals with cancer.
Collapse
Affiliation(s)
- Yun‐hui Xie
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of ChengduAffiliated Hospital of Southwest Jiaotong UniversityChengduChina
| | - Hui‐zhong Jiang
- College of GraduateGuizhou University of Traditional Chinese MedicineGuiyangChina
| |
Collapse
|
15
|
龚 英, 艾丽飞热·艾麦提, 何 宗. [ARL67156, a small-molecule CD39 inhibitor, enhances natural killer cell cytotoxicity against gastric cancer cells in vitro and in nude mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:2006-2014. [PMID: 38189385 PMCID: PMC10774095 DOI: 10.12122/j.issn.1673-4254.2023.12.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Indexed: 01/09/2024]
Abstract
OBJECTIVE To investigate the effect of ARL67156, a small-molecule inhibitor of CD39, on cytotoxicity of natural killer (NK) cells against gastric cancer cells. METHODS Human peripheral blood-derived primary NK cells isolated and purified using a magnetic bead antibody method were treated with 100 μmol/L ARL67156 for 24 h, and the signaling pathway of NK cell activation was detected by Western blotting. The level of interferon-γ (IFN-γ) in the supernatant of NK cells co-cultured with gastric cancer cells was detected using ELISA, and NK cell CD107a degranulation was measured with flow cytometry. The cytotoxicity of NK cells against co-cultured gastric cancer cells was evaluated using flow cytometry. In a nude mouse model bearing subcutaneous gastric cancer xenografts, the therapeutic effect of intravenous transfusion of NK cells and intraperitoneal injection of ARL67156 was assessed by measuring the changes in tumor volume. RESULTS (25.97 ± 5.69) % of peripheral blood NK cells from healthy individuals positive for CD39 expression. Treatment with ARL67156 significantly upregulated the activation molecules including NKG2D, DAP10, CD57, and CD16 and reduced the expressions of the inhibitory receptors TIGIT and KIR, thereby promoting the secretion of IFN-γ and CD107a degranulation in NK cells (P < 0.05). In both the in vitro and in vivo experiments, ARL67156 significantly enhanced the cytotoxicity of NK cells against gastric cancer cells (P < 0.05). CONCLUSION ARL67156 activates NK cells through the vav1-Syk signaling pathway to enhance their cytotoxicity against gastric cancer cells, which may serve as a new strategy for NK cell immunotherapy for gastric cancer.
Collapse
Affiliation(s)
- 英 龚
- 南方医科大学南方医院检验医学科,广东 广州 510515Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 艾丽飞热·艾麦提
- 南方医科大学南方医院检验医学科,广东 广州 510515Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 宗忠 何
- 南部战区总医院输血医学科,广东 广州 510010Department of Transfusion Medicine, Southern Theater Command General Hospital, Guangzhou 510010, China
| |
Collapse
|
16
|
Hojjatipour T, Sharifzadeh Z, Maali A, Azad M. Chimeric antigen receptor-natural killer cells: a promising sword against insidious tumor cells. Hum Cell 2023; 36:1843-1864. [PMID: 37477869 DOI: 10.1007/s13577-023-00948-w] [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: 04/01/2023] [Accepted: 06/23/2023] [Indexed: 07/22/2023]
Abstract
Natural killer (NK) cells are a critical component of innate immunity, particularly in initial cancer recognition and inhibition of additional tumor growth or metastasis propagation. NK cells recognize transformed cells without prior sensitization via stimulatory receptors and rapidly eradicate them. However, the protective tumor microenvironment facilitates tumor escaping via induction of an exhaustion state in immune cells, including NK cells. Hence, genetic manipulation of NK cells for specific identification of tumor-associated antigens or a more robust response against tumor cells is a promising strategy for NK cells' tumoricidal augmentation. Regarding the remarkable achievement of engineered CAR-T cells in treating hematologic malignancies, there is evolving interest in CAR-NK cell recruitment in cancer immunotherapy. Innate functionality of NK cells, higher safety, superior in vivo maintenance, and the off-the-shelf potential move CAR-NK-based therapy superior to CAR-T cells treatment. In this review, we have comprehensively discussed the recent genetic manipulations of CAR-NK cell manufacturing regarding different domains of CAR constructs and their following delivery systems into diverse sources of NK cells. Then highlight the preclinical and clinical investigations of CAR-NK cells and examine the current challenges and prospects as an optimistic remedy in cancer immunotherapy.
Collapse
Affiliation(s)
- Tahereh Hojjatipour
- Department of Hematology and Blood Transfusion, Students Research Center, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciecnes, Qazvin, Iran
| | - Mehdi Azad
- Department of Medical Laboratory Sciences, School of Paramedicine, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, 3419759811, Iran.
| |
Collapse
|
17
|
Ren X, Tang X, Huang T, Hu Z, Wang Y, Zhou Y. FTO plays a crucial role in gastrointestinal cancer and may be a target for immunotherapy: an updated review. Front Oncol 2023; 13:1241357. [PMID: 37916161 PMCID: PMC10616962 DOI: 10.3389/fonc.2023.1241357] [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: 06/16/2023] [Accepted: 09/22/2023] [Indexed: 11/03/2023] Open
Abstract
Gastrointestinal cancer is a common malignancy with high mortality and poor prognosis. Therefore, developing novel effective markers and therapeutic targets for gastrointestinal cancer is currently a challenging and popular topic in oncology research. Accumulating studies have reported that N6-methyladenosine is the most abundant epigenetic modification in eukaryotes. N6-methyladenosine plays an essential role in regulating RNA expression and metabolism, including splicing, translation, stability, decay, and transport. FTO, the earliest demethylase discovered to maintain the balance of N6-adenosine methylation, is abnormally expressed in many tumors. In this review, we discuss the molecular structure and substrate selectivity of FTO. we focus on the role of FTO in gastrointestinal tumor proliferation, migration, invasion, apoptosis, autophagy, immune microenvironment, and its molecular mechanisms. We also discuss its potential in the treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Xiangqing Ren
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaolong Tang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Tian Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zenan Hu
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yongning Zhou
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
18
|
Konstantis G, Tsaousi G, Kitsikidou E, Zacharoulis D, Pourzitaki C. The Immunomodulatory Effect of Various Anaesthetic Practices in Patients Undergoing Gastric or Colon Cancer Surgery: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. J Clin Med 2023; 12:6027. [PMID: 37762967 PMCID: PMC10531584 DOI: 10.3390/jcm12186027] [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: 07/19/2023] [Revised: 08/26/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Gastric and colorectal carcinomas are associated with increased mortality and an increasing incidence worldwide, while surgical resection remains the primary approach for managing these conditions. Emerging evidence suggests that the immunosuppression induced by the chosen anaesthesia approach, during the perioperative period, can have a significant impact on the immune system and consequently the prognosis of these patients. AIM This systematic review aims to comprehensively summarize the existing literature on the effects of different anaesthesia techniques on immune system responses, focusing on cellular immunity in patients undergoing the surgical removal of gastric or colorectal carcinomas. There is no meta-analysis investigating anaesthesia's impact on immune responses in gastric and colorectal cancer surgery. Anaesthesia is a key perioperative factor, yet its significance in this area has not been thoroughly investigated. The clinical question of how the anaesthetic technique choice affects the immune system and prognosis remains unresolved. METHODS Major electronic databases were searched up to February 2023 to May 2023 for relevant randomized controlled trials (RCTs). The study protocol has been registered with Prospero (CRD42023441383). RESULTS Six RCTs met the selection criteria. Among these, three RCTs investigated the effects of volatile-based anaesthesia versus total intravenous anaesthesia (TIVA), while the other three RCTs compared general anaesthesia alone to the combination of general anaesthesia with epidural anaesthesia. According to our analysis, there were no significant differences between TIVA and volatile-based anaesthesia, in terms of primary and secondary endpoints. The combination of general anaesthesia with epidural analgesia had a positive impact on NK cell counts (SMD 0.61, 95% CI 0.28 to 0.94, I2 0.0% at 24 and 72 h after the operation), as well as on CD4+ cells (SMD 0.59, CI 95% 0.26 to 0.93, I2 0.0%). However, the CD3+ cell count, CD4+/CD8+ ratio, neutrophil-to-lymphocyte ratio (NLR), IL-6 and TNF-α levels remained unaffected. CONCLUSIONS The combination of epidural analgesia and general anaesthesia can potentially improve, postoperatively, the NK cell count and CD4+ cell levels in gastric or colon surgery patients. However, the specific impact of TIVA or volatile-based anaesthesia remains uncertain. To gain a better understanding of the immunomodulatory effects of anaesthesia, in this particular group of cancer patients, further well-designed trials are required.
Collapse
Affiliation(s)
- Georgios Konstantis
- Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany
| | - Georgia Tsaousi
- Department of Anesthesiology and ICU, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Elisavet Kitsikidou
- Department of Internal Medicine, Evangelical Hospital Dusseldorf, 40217 Dusseldorf, Germany;
| | | | - Chryssa Pourzitaki
- Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| |
Collapse
|
19
|
Durham J, Tessmann JW, Deng P, Hennig B, Zaytseva YY. The role of perfluorooctane sulfonic acid (PFOS) exposure in inflammation of intestinal tissues and intestinal carcinogenesis. FRONTIERS IN TOXICOLOGY 2023; 5:1244457. [PMID: 37662676 PMCID: PMC10469509 DOI: 10.3389/ftox.2023.1244457] [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: 06/22/2023] [Accepted: 08/03/2023] [Indexed: 09/05/2023] Open
Abstract
PFAS (per- and polyfluoroalkyl substances) are organofluorine substances that are used commercially in products like non-stick cookware, food packaging, personal care products, fire-fighting foam, etc. These chemicals have several different subtypes made of varying numbers of carbon and fluorine atoms. PFAS substances that have longer carbon chains, such as PFOS (perfluorooctane sulfonic acid), can potentially pose a significant public health risk due to their ability to bioaccumulate and persist for long periods of time in the body and the environment. The National Academies Report suggests there is some evidence of PFOS exposure and gastrointestinal (GI) inflammation contributing to ulcerative colitis. Inflammatory bowel diseases such as ulcerative colitis are precursors to colorectal cancer. However, evidence about the association between PFOS and colorectal cancer is limited and has shown contradictory findings. This review provides an overview of population and preclinical studies on PFOS exposure and GI inflammation, metabolism, immune responses, and carcinogenesis. It also highlights some mitigation approaches to reduce the harmful effects of PFOS on GI tract and discusses the dietary strategies, such as an increase in soluble fiber intake, to reduce PFOS-induced alterations in cellular lipid metabolism. More importantly, this review demonstrates the urgent need to better understand the relationship between PFOS and GI pathology and carcinogenesis, which will enable development of better approaches for interventions in populations exposed to high levels of PFAS, and in particular to PFOS.
Collapse
Affiliation(s)
- Jerika Durham
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, United States
| | - Josiane Weber Tessmann
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, United States
| | - Pan Deng
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Bernhard Hennig
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY, United States
| | - Yekaterina Y. Zaytseva
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
20
|
Hatami Z, Hashemi ZS, Eftekhary M, Amiri A, Karpisheh V, Nasrollahi K, Jafari R. Natural killer cell-derived exosomes for cancer immunotherapy: innovative therapeutics art. Cancer Cell Int 2023; 23:157. [PMID: 37543612 PMCID: PMC10403883 DOI: 10.1186/s12935-023-02996-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 07/19/2023] [Indexed: 08/07/2023] Open
Abstract
Chimeric antigen receptor natural killer cells (CAR-NK) promote off-the-shelf cellular therapy for solid tumors and malignancy.However,, the development of CAR-NK is due to their immune surveillance uncertainty and cytotoxicity challenge was restricted. Natural killer cell-derived exosome (NK-Exo) combine crucial targeted cellular therapies of NK cell therapies with unique non-toxic Exo as a self-origin shuttle against cancer immunotherapy. This review study covers cytokines, adoptive (autologous and allogenic) NK immunotherapy, stimulatory and regulatory functions, and cell-free derivatives from NK cells. The future path of NK-Exo cytotoxicity and anti-tumor activity with considering non-caspase-independent/dependent apoptosis and Fas/FasL pathway in cancer immunotherapy. Finally, the significance and implication of NK-Exo therapeutics through combination therapy and the development of emerging approaches for the purification and delivery NK-Exo to severe immune and tumor cells and tissues were discussed in detail.
Collapse
Affiliation(s)
- Zahra Hatami
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Sadat Hashemi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| | - Mohamad Eftekhary
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ala Amiri
- Department of Biotechnology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Vahid Karpisheh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kaveh Nasrollahi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
21
|
Khawar MB, Gao G, Rafiq M, Shehzadi A, Afzal A, Abbasi MH, Sheikh N, Afzal N, Ashraf MA, Hamid SE, Shahzaman S, Kawish N, Sun H. Breaking down barriers: The potential of smarter CAR-engineered NK cells against solid tumors. J Cell Biochem 2023; 124:1082-1104. [PMID: 37566723 DOI: 10.1002/jcb.30460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/04/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023]
Abstract
Natural killer (NK) cells are considered to be the foremost fighters of our innate immune system against foreign invaders and thus tend to promptly latch onto the virus-infected and tumor/cancerous cells, killing them through phagocytosis. At present, the application of genetically engineered Chimeric antigen receptor (CAR) receptors ensures a guaranteed optimistic response with NK cells and would not allow the affected cells to dodge or escape unchecked. Hence the specificity and uniqueness of CAR-NK cells over CAR-T therapy make them a better immunotherapeutic choice to reduce the load of trafficking of numerous tumor cells near the healthy cell populations in a more intact way than offered by CAR-T immunotherapy. Our review mainly focuses on the preclinical, clinical, and recent advances in clinical research trials and further strategies to achieve an augmented and efficient cure against solid tumors.
Collapse
Affiliation(s)
- Muhammad B Khawar
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental and, Translational Noncoding RNA Research, Yangzhou, China
- Department of Zoology, Applied Molecular Biology and Biomedicine Lab, University of Narowal, Narowal, Pakistan
| | - Guangzhong Gao
- Department of Physiatry, Haian Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nantong, Jiangsu, China
| | - Mussarat Rafiq
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Anila Shehzadi
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | - Ali Afzal
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | | | - Nadeem Sheikh
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Nimra Afzal
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | | | - Syeda E Hamid
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | - Sara Shahzaman
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | - Naseer Kawish
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Haibo Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental and, Translational Noncoding RNA Research, Yangzhou, China
| |
Collapse
|
22
|
Wang J, Ge H, Tian Z. Immunotherapy Plus Radiotherapy for the Treatment of Sarcomas: Is There a Potential for Synergism? Onco Targets Ther 2023; 16:385-397. [PMID: 37313391 PMCID: PMC10258041 DOI: 10.2147/ott.s410693] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
Soft tissue sarcoma (STS) is a highly heterogeneous malignant tumor derived from mesenchymal tissue. Advanced STS has a poor response to the current anti-cancer therapeutic options, with a median overall survival of less than two years. Thus, new and more effective treatment methods for STS are needed. Increasing evidence has shown that immunotherapy and radiotherapy have synergistic therapeutic effects against malignant tumors. In addition, immunoradiotherapy has yielded positive results in clinical trials for various cancers. In this review, we discuss the synergistic mechanism of immunoradiotherapy in cancer treatment and the application of this combined regimen for the treatment of several cancers. In addition, we summarize the existing evidence on the use of immunoradiotherapy for the treatment of STS and the relevant clinical trials that are currently ongoing. Furthermore, we identify challenges in the use of immunoradiotherapy for the treatment of sarcomas and propose methods and precautions for overcoming these challenges. Lastly, we propose clinical research strategies and future research directions to help in the research and treatment of STS.
Collapse
Affiliation(s)
- Jiaqiang Wang
- Department of Bone and Soft Tissue, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan Province, 450008, People’s Republic of China
| | - Hong Ge
- Department of Radiotherapy, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan Province, 450008, People’s Republic of China
| | - Zhichao Tian
- Department of Bone and Soft Tissue, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan Province, 450008, People’s Republic of China
| |
Collapse
|
23
|
Pimenta BV, Madrid RRM, Mathews PD, Riske KA, Loh W, Angelov B, Angelova A, Mertins O. Interaction of polyelectrolyte-shell cubosomes with serum albumin for triggering drug release in gastrointestinal cancer. J Mater Chem B 2023; 11:2490-2503. [PMID: 36852541 DOI: 10.1039/d2tb02670h] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Nano-structured and functionalized materials for encapsulation, transport, targeting and controlled release of drugs are of high interest to overcome low bioavailability in oral administration. We develop lipid-based cubosomes, which are surface-functionalized with biocompatible chitosan-N-arginine and alginate, displaying internal liquid crystalline structures. Polyelectrolyte-shell (PS) cubosomes have pH-responsive characteristics profitable for oral delivery. The obtained PScubosomes can strongly interact with serum albumin, a protein which is released in the stomach under gastric cancer conditions. An effective thermodynamic PScubosome-protein interaction was characterized at pH 2.0 and 7.4 by isothermal titration calorimetry at 37 °C. A high increment of the albumin conformation transition temperature was evidenced by differential scanning calorimetry upon incubation with PScubosomes. The performed structural studies by synchrotron small-angle X-ray scattering (SAXS) revealed essential alterations in the internal liquid crystalline topology of the nanocarriers including an Im3m to Pn3m transition and a reduction of the cubic lattice parameters. The PScubosome nanoparticle interaction with serum albumin, leading to inner structural changes in a range of temperatures, promoted the release of water from the cubosomal nanochannels. Altogether, the results revealed effective interactions of the PScubosomes with albumin under simulated gastrointestinal pH conditions and suggested promising nanocarrier characteristics for triggered oral drug release.
Collapse
Affiliation(s)
- Barbara V Pimenta
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil.
| | - Rafael R M Madrid
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil.
| | - Patrick D Mathews
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil.
| | - Karin A Riske
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil.
| | - Watson Loh
- Institute of Chemistry, State University of Campinas (UNICAMP), 13083-970 Campinas, Brazil
| | - Borislav Angelov
- Institute of Physics, ELI Beamlines, Academy of Sciences of the Czech Republic, CZ-18221 Prague, Czech Republic
| | - Angelina Angelova
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France.
| | - Omar Mertins
- Laboratory of Nano Bio Materials (LNBM), Department of Biophysics, Paulista Medical School, Federal University of Sao Paulo (UNIFESP), 04023-062 Sao Paulo, Brazil. .,Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France.
| |
Collapse
|
24
|
Minaei N, Ramezankhani R, Tamimi A, Piryaei A, Zarrabi A, Aref AR, Mostafavi E, Vosough M. Immunotherapeutic approaches in Hepatocellular carcinoma: Building blocks of hope in near future. Eur J Cell Biol 2023; 102:151284. [PMID: 36584598 DOI: 10.1016/j.ejcb.2022.151284] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/30/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary hepatic cancer and is among the major causes of mortality due to cancer. Due to the lack of efficient conventional therapeutic options for this cancer, particularly in advanced cases, novel treatments including immunotherapy have been considered. However, despite the encouraging clinical outcomes after implementing these innovative approaches, such as oncolytic viruses (OVs), adoptive cell therapies (ACT), immune checkpoint blockades (ICBs), and cancer vaccines, several factors have restricted their therapeutic effect. The main concern is the existence of an immunosuppressive tumor microenvironment (TME). Combination of different ICBs or ICBs plus tyrosine kinase inhibitors have shown promising results in overcoming these limiting factors to some extent. Combination of programmed cell death ligand-1 (PD-L1) antibody Atezolizumab and vascular endothelial growth factor (VEGF) antibody Bevacizumab has become the standard of care in the first-line therapy for untestable HCC, approved by regulatory agencies. This paper highlighted a wide overview of the direct and indirect immunotherapeutic strategies proposed for the treatment of HCC patients and the common challenges that have hindered their further clinical applications.
Collapse
Affiliation(s)
- Neda Minaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Roya Ramezankhani
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran; Department of Development and Regeneration, KU Leuven Stem Cell Institute, Leuven, Belgium
| | - Atena Tamimi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital-Huddinge, Sweden.
| |
Collapse
|
25
|
Zeng Z, Liu Z, Li J, Sun J, Ma M, Ye X, Yu J, Kang W. Baseline splenic volume as a biomarker for clinical outcome and circulating lymphocyte count in gastric cancer. Front Oncol 2023; 12:1065716. [PMID: 36793344 PMCID: PMC9923954 DOI: 10.3389/fonc.2022.1065716] [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: 10/21/2022] [Accepted: 12/28/2022] [Indexed: 01/31/2023] Open
Abstract
Background The spleen is the largest peripheral lymphoid organ in the body. Studies have implicated the spleen in the development of cancer. However, it is unknown whether splenic volume (SV) is associated with the clinical outcome of gastric cancer. Methods Data of gastric cancer patients treated with surgical resection were retrospectively analyzed. Patients were divided into three groups: underweight, normal-weight and overweight. Overall survival was compared in patients with high and low splenic volume. The correlation between splenic volume and peripheral immune cells were analyzed. Results Of 541 patients, 71.2% were male and the median age was 60. Underweight, normal-weight and overweight patients accounted for 5.4%, 62.3% and 32.3%, respectively. High splenic volume was associated with unfavorable prognosis across the three groups. In addition, the increase of splenic volume during neoadjuvant chemotherapy was not associated with prognosis. The baseline splenic volume was negatively correlated with lymphocytes (r=-0.21, p<0.001) and positively correlated with NLR (neutrophil-to-lymphocyte ratio) (r=0.24, p<0.001). In a group of patients (n=56), splenic volume was found to have negative correlation with CD4+T cells (r=-0.27, p=0.041) and NK cells (r=-0.30, p=0.025). Conclusions The presence of high splenic volume is a biomarker of unfavorable prognosis and reduced circulating lymphocytes in gastric cancer.
Collapse
|
26
|
Prakash A, Gates T, Zhao X, Wangmo D, Subramanian S. Tumor-derived extracellular vesicles in the colorectal cancer immune environment and immunotherapy. Pharmacol Ther 2023; 241:108332. [PMID: 36526013 DOI: 10.1016/j.pharmthera.2022.108332] [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: 09/07/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Despite significant advances in the screening, diagnosis, and treatment of colorectal cancer (CRC) immune checkpoint inhibitors (ICIs) continue to have limited utility outside of microsatellite-high disease. Given the durable response to immunotherapy seen across malignancies, increasing CRC response rates to ICI therapy is an active area of clinical research. An increasing body of work has demonstrated that tumor-derived extracellular vesicles (TEVs) are key modulators in tumor signaling and the determinants of the tumor microenvironment. Pre-clinical models have shown that TEVs are directly involved in antigen presentation and are involved in radiation-induced DNA damage signaling. Both direct and indirect modifications of these TEVs can alter CRC immunogenicity and ICI treatment response, making them attractive targets for potential therapeutic development. In addition, modified TEVs can be developed using several different mechanisms, with varied cargo including micro-RNAs and small peptide molecules. Recent work has shown strong pre-clinical evidence of injected modified TEV-induced ICI activity, with knockdown of the micro-RNA miR-424 in TEVs improving CRC immunogenicity and increasing anti-PD-1 activity in mouse models. Clinical trials are ongoing in the evaluation of modified TEVs in cancer therapy, but they appear to be a promising therapeutic target in CRC.
Collapse
Affiliation(s)
- Ajay Prakash
- Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN, United States of America.
| | - Travis Gates
- Department of Surgery, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Xianda Zhao
- Department of Surgery, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Dechen Wangmo
- Department of Surgery, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Subbaya Subramanian
- Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN, United States of America; Department of Surgery, University of Minnesota Medical School, Minneapolis, MN, United States of America; Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| |
Collapse
|
27
|
Gut Microbiota Host-Gene Interaction. Int J Mol Sci 2022; 23:ijms232213717. [PMID: 36430197 PMCID: PMC9698405 DOI: 10.3390/ijms232213717] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/28/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022] Open
Abstract
Studies carried out in the last ten years have shown that the metabolites made up from the gut microbiota are essential for multiple functions, such as the correct development of the immune system of newborns, interception of pathogens, and nutritional enrichment of the diet. Therefore, it is not surprising that alteration of the gut microbiota is the starting point of gastrointestinal infection, obesity, type 2 diabetes, inflammatory bowel disease, colorectal cancer, and lung cancer. Diet changes and antibiotics are the major factors damaging the gut microbiota. Early exposure of the newborns to antibiotics may prevent their correct development of the immune system, exposing them to pathogen infections, allergies, and chronic inflammatory diseases. We already know much on how host genes, microbiota, and the environment interact, owing to experiments in several model animals, especially in mice; advances in molecular technology; microbiota transplantation; and comparative metagenomic analysis. However, much more remains to be known. Longitudinal studies on patients undergoing to therapy, along with the identification of bacteria prevalent in responding patients may provide valuable data for improving therapies.
Collapse
|
28
|
Wang D, Ye Q, Gu H, Chen Z. The role of lipid metabolism in tumor immune microenvironment and potential therapeutic strategies. Front Oncol 2022; 12:984560. [PMID: 36172157 PMCID: PMC9510836 DOI: 10.3389/fonc.2022.984560] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/01/2022] [Indexed: 11/20/2022] Open
Abstract
Aberrant lipid metabolism is nonnegligible for tumor cells to adapt to the tumor microenvironment (TME). It plays a significant role in the amount and function of immune cells, including tumor-associated macrophages, T cells, dendritic cells and marrow-derived suppressor cells. It is well-known that the immune response in TME is suppressed and lipid metabolism is closely involved in this process. Immunotherapy, containing anti-PD1/PDL1 therapy and adoptive T cell therapy, is a crucial clinical cancer therapeutic strategy nowadays, but they display a low-sensibility in certain cancers. In this review, we mainly discussed the importance of lipid metabolism in the formation of immunosuppressive TME, and explored the effectiveness and sensitivity of immunotherapy treatment by regulating the lipid metabolism.
Collapse
Affiliation(s)
- Danting Wang
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qizhen Ye
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haochen Gu
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhigang Chen
- Department of Breast Surgery (Surgical Oncology), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University, Hangzhou, China
- Cancer Centre, Zhejiang University, Hangzhou, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Zhigang Chen,
| |
Collapse
|
29
|
Kim S, Kim K. Lipid-mediated ex vivo cell surface engineering for augmented cellular functionalities. BIOMATERIALS ADVANCES 2022; 140:213059. [PMID: 35961186 DOI: 10.1016/j.bioadv.2022.213059] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/23/2022] [Accepted: 07/31/2022] [Indexed: 06/15/2023]
Abstract
Once administrated, intercellular adhesion to recognize and/or arrest target cells is essential for specific treatments, especially for cancer or tumor. However, immune cells administrated into the tumor-microenvironment could lose their intrinsic functionalities such as target recognition ability, resulting in an ineffective cancer immunotherapy. Various manipulation techniques for decorating functional moieties onto cell surface and enhancing target recognition have been developed. A hydrophobic interaction-mediated ex-vivo cell surface engineering using lipid-based biomaterials could be a state-of-the-art engineering technique that could achieve high-efficiency cell surface modification by a single method without disturbance of intrinsic characteristics of cells. In this regard, this review provides design principles for the development of lipid-based biomaterials with a linear structure of lipid, polyethylene glycol, and functional group, strategies for the synthesis process, and their practical applications in biomedical engineering. Especially, we provide new insights into the development of a novel surface coating techniques for natural killer (NK) cells with engineering decoration of cancer targeting moieties on their cell surfaces. Among immune cells, NK cells are interesting cell population for substituting T cells because of their excellent safety and independent anticancer efficacy. Thus, optimal strategies to select cancer-type-specific targeting moieties and present them onto the surface of immune cells (especially, NK cells) using lipid-based biomaterials could provide additional tools to capture cancer cells for developing novel immune cell therapy products. Enhanced anticancer efficacies by surface-engineered NK cells have been demonstrated both in vitro and in vivo. Therefore, it could be speculated that recent progresses in cell surface modification technology via lipid-based biomaterials could strengthen immune surveillance and immune synapses for utilization in a next-generation cancer immunotherapy, beyond currently available genetic engineering tool such as chimeric antigen receptor-mediated immune cell modulation.
Collapse
Affiliation(s)
- Sungjun Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, Republic of Korea
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Skorupan N, Palestino Dominguez M, Ricci SL, Alewine C. Clinical Strategies Targeting the Tumor Microenvironment of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:4209. [PMID: 36077755 PMCID: PMC9454553 DOI: 10.3390/cancers14174209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 12/04/2022] Open
Abstract
Pancreatic cancer has a complex tumor microenvironment which engages in extensive crosstalk between cancer cells, cancer-associated fibroblasts, and immune cells. Many of these interactions contribute to tumor resistance to anti-cancer therapies. Here, new therapeutic strategies designed to modulate the cancer-associated fibroblast and immune compartments of pancreatic ductal adenocarcinomas are described and clinical trials of novel therapeutics are discussed. Continued advances in our understanding of the pancreatic cancer tumor microenvironment are generating stromal and immune-modulating therapeutics that may improve patient responses to anti-tumor treatment.
Collapse
Affiliation(s)
- Nebojsa Skorupan
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Medical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mayrel Palestino Dominguez
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Samuel L. Ricci
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christine Alewine
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
31
|
Tian Z, Yao W. PD-1/L1 inhibitor plus chemotherapy in the treatment of sarcomas. Front Immunol 2022; 13:898255. [PMID: 36072581 PMCID: PMC9441887 DOI: 10.3389/fimmu.2022.898255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
There is an urgent clinical need for new therapeutic regimens for the effective treatment of advanced sarcomas. Accumulating evidence suggests that programmed death receptor-1/programmed death protein ligand-1 (PD-1/L1) inhibitors have synergistic effects with chemotherapy and have been approved for treatment of lung cancer, gastroesophageal cancer, and breast cancer. In this review, we reviewed the synergistic mechanism of PD-1/L1 inhibitors plus chemotherapy in the treatment of cancers, and the application of this combined regimen in several cancers, followed by a summary of the current evidence on the application of this combined regimen in the treatment of sarcomas as well as the main clinical trials currently underway. Based on the findings of this review, we believe that this combined approach will play an important role in the treatment of some subtypes of sarcomas in the future.
Collapse
|
32
|
Fakharian F, Asgari B, Nabavi-Rad A, Sadeghi A, Soleimani N, Yadegar A, Zali MR. The interplay between Helicobacter pylori and the gut microbiota: An emerging driver influencing the immune system homeostasis and gastric carcinogenesis. Front Cell Infect Microbiol 2022; 12:953718. [PMID: 36046747 PMCID: PMC9423097 DOI: 10.3389/fcimb.2022.953718] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/25/2022] [Indexed: 01/06/2023] Open
Abstract
The human gut microbiota are critical for preserving the health status because they are required for digestion and nutrient acquisition, the development of the immune system, and energy metabolism. The gut microbial composition is greatly influenced by the colonization of the recalcitrant pathogen Helicobacter pylori (H. pylori) and the conventional antibiotic regimens that follow. H. pylori is considered to be the main microorganism in gastric carcinogenesis, and it appears to be required for the early stages of the process. However, a non-H. pylori microbiota profile is also suggested, primarily in the later stages of tumorigenesis. On the other hand, specific groups of gut microbes may produce beneficial byproducts such as short-chain fatty acids (acetate, butyrate, and propionate) that can modulate inflammation and tumorigenesis pathways. In this review, we aim to present how H. pylori influences the population of the gut microbiota to modify the host immunity and trigger the development of gastric carcinogenesis. We will also highlight the effect of the gut microbiota on immunotherapeutic approaches such as immune checkpoint blockade in cancer treatment to present a perspective for further development of innovative therapeutic paradigms to prevent the progression of H. pylori-induced stomach cancer.
Collapse
Affiliation(s)
- Farzaneh Fakharian
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnoush Asgari
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Soleimani
- Department of Microbiology, Faculty of Biological Sciences and Technology, Shahid Beheshti University, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Abbas Yadegar, ;
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Behfar M, Muhammadnejad S, Abdolahi S, Mohseni R, Shoae-Hassani A, Monzavi SM, Hamidieh AA. Adoptive NK-cell transfer as a potential treatment paradigm for Wilms tumor: A preclinical study. Pediatr Blood Cancer 2022; 69:e29676. [PMID: 35441789 DOI: 10.1002/pbc.29676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 02/15/2022] [Accepted: 03/05/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Natural killer (NK) cell therapy has been shown to be effective in the treatment of some cancers. However, the effects of this adoptive immunotherapy have not been investigated for Wilms tumor (WT). In this study, the effects of adoptive NK-cell transfer on a patient-derived xenograft (PDX) model of anaplastic WT were evaluated, and the impacts of cell source and ex vivo activation strategy on the therapeutic efficacy of NK-cell product were appraised. METHODS NK cells were isolated from human peripheral blood mononuclear cells (NKPB ) and human cord blood (NKCB ), and were expanded and activated using a cytokine cocktail. Another group of NK cells (NKET ) was produced through activation with the exosomes extracted from previously challenged NKPB cells with WT. PDX-bearing mice were treated with clinically relevant doses of NKPB , NKCB , NKET , standard chemotherapy, and placebo (phosphate-buffered saline). RESULTS PDX models treated with NKCB showed a better survival rate, though the difference among the study groups was not significant. Compared with the placebo control group, NKCB significantly improved the histopathologic response, NKPB significantly inhibited the proliferation of neoplastic cells, and NKET led to a significant decrease in the metastasis score (all p-values <.05). Standard chemotherapy provided the greatest tumor growth inhibition and the lowest mitotic count, though it did not show any significant advantage over NK-cell therapies in any of the outcome parameters in two-by-two comparisons. CONCLUSIONS This study spotlights the efficacy of adoptive NK-cell transfer as a potential treatment candidate for high-risk WT.
Collapse
Affiliation(s)
- Maryam Behfar
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Samad Muhammadnejad
- Gene Therapy Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahrokh Abdolahi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rashin Mohseni
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Shoae-Hassani
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mostafa Monzavi
- Gene Therapy Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Liu S, Tang W, Cao J, Shang M, Sun H, Gong J, Hu B. A Comprehensive Analysis of HAVCR1 as a Prognostic and Diagnostic Marker for Pan-Cancer. Front Genet 2022; 13:904114. [PMID: 35754803 PMCID: PMC9213751 DOI: 10.3389/fgene.2022.904114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Hepatitis A virus cellular receptor (HAVCR1) is a type-1 integral membrane glycoprotein that plays a key role in immunity and renal regeneration and is abnormally expressed in various tumor types. Nonetheless, the function of HAVCR1 in pan-cancer remains unknown. In this study, we comprehensively analyzed the expression and promoter methylation level of HAVCR1 and assessed the immune cell infiltration, correlation between stromal and immune cell admixture, CD (Cluster of Differentiation) and HAVCR1 expression and prognostic value of HAVCR1 mRNA expression in Liver hepatocellular carcinoma (LIHC) and Pancreatic adenocarcinoma (PAAD). Our results showed that HAVCR1 was overexpressed while the promoter methylation of HAVCR1 was decreased in Liver hepatocellular carcinoma and Pancreatic adenocarcinoma. HAVCR1 was associated with increased infiltration of B cells, CD8 cells, macrophages, neutrophils and Dendritic cells in Liver hepatocellular carcinoma and Pancreatic adenocarcinoma. HAVCR1 expression was positively correlated with the immune, stromal and estimate scores of Pancreatic adenocarcinoma and the stromal and estimate scores of Liver hepatocellular carcinoma. Furthermore, HAVCR1 expression was correlated with other immune molecules such as HHLA2 (Human endogenous retrovirus-H long terminal repeat-associating protein 2), CD44 and TNFRSF4 (TNF Receptor Superfamily Member 4) in Liver hepatocellular carcinoma and Pancreatic adenocarcinoma. During Kaplan-Meier analysis, high HAVCR1 expression in Liver hepatocellular carcinoma and Pancreatic adenocarcinoma correlated with poor survival. A marginally significant p-value (p = 0.051) was obtained when the relationship between HAVCR1 expression in Liver hepatocellular carcinoma and prognosis was analyzed, attributed to the small sample size. Overall, we provided compelling evidence that HAVCR1 could be a prognostic and diagnostic marker for Liver hepatocellular carcinoma and Pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Sheng Liu
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenting Tang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen Cancer Center, Guangzhou, China
- Department of Molecular Diagnostics, Sun Yat-sen Cancer Center, Guangzhou, China
| | - Jing Cao
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Mei Shang
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hengchang Sun
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiao Gong
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bo Hu
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
35
|
Maddineni S, Silberstein JL, Sunwoo JB. Emerging NK cell therapies for cancer and the promise of next generation engineering of iPSC-derived NK cells. J Immunother Cancer 2022; 10:jitc-2022-004693. [PMID: 35580928 PMCID: PMC9115029 DOI: 10.1136/jitc-2022-004693] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2022] [Indexed: 12/11/2022] Open
Abstract
Adoptive cell therapy is a rapidly advancing approach to cancer immunotherapy that seeks to facilitate antitumor responses by introducing potent effector cells into the tumor microenvironment. Expanded autologous T cells, particularly T cells with engineered T cell receptors (TCR) and chimeric antigen receptor-T cells have had success in various hematologic malignancies but have faced challenges when applied to solid tumors. As a result, other immune subpopulations may provide valuable and orthogonal options for treatment. Natural killer (NK) cells offer the possibility of significant tumor clearance and recruitment of additional immune subpopulations without the need for prior antigen presentation like in T or B cells that could require removal of endogenous antigen specificity mediated via the T cell receptor (TCR and/or the B ecll receptor (BCR). In recent years, NK cells have been demonstrated to be increasingly important players in the immune response against cancer. Here, we review multiple avenues for allogeneic NK cell therapy, including derivation of NK cells from peripheral blood or umbilical cord blood, the NK-92 immortalized cell line, and induced pluripotent stem cells (iPSCs). We also describe the potential of engineering iPSC-derived NK cells and the utility of this platform. Finally, we consider the benefits and drawbacks of each approach and discuss recent developments in the manufacturing and genetic or metabolic engineering of NK cells to have robust and prolonged antitumor responses in preclinical and clinical settings.
Collapse
Affiliation(s)
- Sainiteesh Maddineni
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - John L Silberstein
- Program in Immunology, Stanford University School of Medicine, Palo Alto, California, USA.,Department of Bioengineering, Stanford University, Palo Alto, California, USA
| | - John B Sunwoo
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| |
Collapse
|
36
|
Hossian AKMN, Hackett CS, Brentjens RJ, Rafiq S. Multipurposing CARs: Same engine, different vehicles. Mol Ther 2022; 30:1381-1395. [PMID: 35151842 PMCID: PMC9077369 DOI: 10.1016/j.ymthe.2022.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/16/2021] [Accepted: 02/08/2022] [Indexed: 11/22/2022] Open
Abstract
T cells genetically engineered to recognize and eliminate tumor cells through synthetic chimeric antigen receptors (CARs) have demonstrated remarkable clinical efficacy against B cell leukemia over the past decade. This therapy is a form of highly personalized medicine that involves genetically modifying a patient's T cells to recognize and kill cancer cells. With the FDA approval of 5 CAR T cell products, this approach has been validated as a powerful new drug in the therapeutic armamentarium against cancer. Researchers are now studying how to expand this technology beyond its use in conventional polyclonal αβ T cells to address limitations to the current therapy in cancer and applications beyond it. Considering the specific characteristics of immune cell from diverse lineages, several preclinical and clinical studies are under way to assess the advantages of CAR-redirected function in these cells and apply the lessons learned from CAR T cell therapy in cancer to other diseases.
Collapse
Affiliation(s)
- A K M Nawshad Hossian
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
| | - Christopher S Hackett
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Renier J Brentjens
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA.
| | - Sarwish Rafiq
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
37
|
Zhu M, Zhang H, Pedersen KS, Foster NR, Jaszewski BL, Liu X, Hirdler JB, An Z, Bekaii-Saab TS, Halfdanarson TR, Boland PM, Yan Y, Hubbard JH, Ma WW, Yoon HH, Revzin A, Fernandez-Zapico ME, Overman MJ, McWilliams RR, Dong H. Understanding Suboptimal Response to Immune Checkpoint Inhibitors. Adv Biol (Weinh) 2022; 7:e2101319. [PMID: 35343107 DOI: 10.1002/adbi.202101319] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/28/2022] [Indexed: 12/31/2022]
Abstract
Immune checkpoint inhibitors (ICIs), as a novel class of anticancer therapy, can be more efficacious and less toxic than chemotherapy, but their clinical success is confined to certain tumor types. Elucidating their targets, mechanisms and scope of action, and potential synergism with chemotherapy and/or targeted therapies are critical to widen their clinical indications. Treatment response to an ICI targeting programmed death-1 (anti-PD-1) is sought to be understood here by conducting a preplanned correlative analysis of a phase II clinical trial in patients with small bowel adenocarcinoma (SBA). The cytolytic capacity of circulating immune cells in cancer patients using a novel ex vivo cytotoxicity assay is evaluated, and the utility of circulating biomarkers is investigated to predict and monitor the treatment effect of anti-PD-1. Baseline expression of Bim and NKG7 and upregulation of CX3CR1 in circulating T cells are associated with the clinical benefit of anti-PD-1 in patients with SBA. Overall, these findings suggest that the frequency and cytolytic capacity of circulating, effector immune cells may differentiate clinical response to ICIs, providing a strong rationale to support immune monitoring using patient peripheral blood.
Collapse
Affiliation(s)
- Mojun Zhu
- Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Henan Zhang
- Urology and Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Nathan R Foster
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Brandy L Jaszewski
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xin Liu
- Urology and Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jacob B Hirdler
- Urology and Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Zesheng An
- Urology and Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | | | - Patrick M Boland
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA
| | - Yiyi Yan
- Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Wen Wee Ma
- Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Harry H Yoon
- Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Alexander Revzin
- Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | | | | | | | - Haidong Dong
- Urology and Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
38
|
Chang M, Tang X, Nelson L, Nyberg G, Du Z. Differential effects on natural killer cell production by membrane-bound cytokine stimulations. Biotechnol Bioeng 2022; 119:1820-1838. [PMID: 35297033 DOI: 10.1002/bit.28086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 11/07/2022]
Abstract
Robust manufacturing production of natural killer (NK) cells has been challenging in allogeneic NK cell-based therapy. Here, we compared the impact of cytokines on NK cell expansion by developing recombinant K562 feeder cell lines expressing membrane-bound cytokines, mIL15, mIL21, and 41BBL, individually or in combination. We found that 41BBL played a dominant role in promoting up to 500,000-fold of NK cell expansion after a 21-day culture process without inducing exhaustion. However, 41BBL stimulation reduced the overall cytotoxic activity of NK cells when combined with mIL15 and mIL21. Additionally, long-term stimulation with mIL15 and mIL21, but not 41BBL, increased CD56 expression and CD56bright population, which is unexpectedly correlated with the NK cell cytotoxicity. By conducting single-cell sequencing, we identified distinct subpopulations of NK cells induced by different cytokines, including an adaptive-like CD56brightCD16-CD49a+ subset induced by mIL15. Through gene expression analysis, we found that cytokines modulated signaling pathways and target genes involved in cell cycle, senescence, self-renewal, migration, and maturation, in a different manner. Together, our study demonstrated cytokine signal pathways play different roles in NK cell expansion and differentiation, which shed light on NK cell process design to improve productivity and product quality. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Meiping Chang
- Process Cell Sciences, Biologics Process R&D, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Xiaoyan Tang
- Process Cell Sciences, Biologics Process R&D, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Luke Nelson
- Process Cell Sciences, Biologics Process R&D, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Gregg Nyberg
- Process Cell Sciences, Biologics Process R&D, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Zhimei Du
- Process Cell Sciences, Biologics Process R&D, Merck & Co., Inc., Kenilworth, NJ, USA
| |
Collapse
|
39
|
Faghfuri E, Shadbad MA, Faghfouri AH, Soozangar N. Cellular immunotherapy in gastric cancer: adoptive cell therapy and dendritic cell-based vaccination. Immunotherapy 2022; 14:475-488. [PMID: 35232264 DOI: 10.2217/imt-2021-0285] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is one of the most frequently diagnosed malignancies. Recent studies have highlighted cellular immunotherapy (CI) as a promising approach for treating this disease. Among the CI-based approaches, adoptive cell therapy and dendritic cell-based vaccination are commonly studied in preclinical and clinical trials. Here we review the current evidence on the potentiality of CI in treating GC, the targets for adoptive cell therapy, ongoing clinical trials, constraints and the future outlook. The results suggest that there is a need to identify novel biomarkers that predict which GC patients will most likely respond to these approaches. Also, CI plus chemotherapy or immune checkpoint inhibitors can improve the survival of patients with late-stage GC. Therefore, this approach can be promising for treating these patients.
Collapse
Affiliation(s)
- Elnaz Faghfuri
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | | | - Narges Soozangar
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
40
|
El-Deeb NM, Ibrahim OM, Mohamed MA, Farag MMS, Farrag AA, El-Aassar MR. Alginate/κ-carrageenan oral microcapsules loaded with Agaricus bisporus polysaccharides MH751906 for natural killer cells mediated colon cancer immunotherapy. Int J Biol Macromol 2022; 205:385-395. [PMID: 35183600 DOI: 10.1016/j.ijbiomac.2022.02.058] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/28/2022] [Accepted: 02/11/2022] [Indexed: 12/24/2022]
Abstract
The current study explores the effect of the extracted novel Mushroom polysaccharides and its formulation onto Alginate (Alg.)/kappa carrageenan microcapsules to exert immunotherapeutic effect upon activating gut resident natural killer cells (NK) against colon cancer. The extracted polysaccharides of Agaricus bisporus MH751906 was microcapsulated in Alg/κ-carrageenan microcapsules as an oral delivery system for colon cancer. The microcapsule is characterized by SEM, FTIR, Raman and TGA; and showed a superior acidic stability, controlled release, and thermal stability at high temperature with higher hydrogel swelling rate in colon-mimicking pH. Upon activation of human NK cells with microcapsules (ANK cells), a significant increase in CD16+CD56+ NK cell populations were recorded. These activated NK cells showed 74.09% cytotoxic effects against human colon cancer Caco-2 cells where majority of cancer cell populations arrested at G0/G1 phase leading to apoptosis. The apoptotic molecular mechanism induced by ANK cells on Caco-2 treated cells is through down regulations of both BCL2 and TGF surviving genes and up regulation in IkappaB-α gene expression. Therefore, this novel polysaccharides-alginate/κ-carrageenan microcapsules can be used as an oral targeted delivery system for colon cancer immunotherapy.
Collapse
Affiliation(s)
- Nehal M El-Deeb
- Biopharmaceutical Products Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City 21934, Alexandria, Egypt; Pharmaceutical and Fermentation Industries Development Center, City of Scientific Research and Technological Applications (SRTA city), New Borg El-Arab City 21934, Alexandria, Egypt
| | - Omar M Ibrahim
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, United States
| | - Mahmoud A Mohamed
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, 11884 Nasr City, Cairo, Egypt
| | - Mohamed M S Farag
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, 11884 Nasr City, Cairo, Egypt
| | - Ayman A Farrag
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, 11884 Nasr City, Cairo, Egypt; Fermentation Biotechnology and Applied Microbiology Center, Al-Azhar University, 11884 Nasr City, Cairo, Egypt
| | - M R El-Aassar
- Department of Chemistry, College of Science, Jouf University, Sakaka 2014, Saudi Arabia.
| |
Collapse
|
41
|
Mortezaee K, Majidpoor J. (Im)maturity in Tumor Ecosystem. Front Oncol 2022; 11:813897. [PMID: 35145911 PMCID: PMC8821092 DOI: 10.3389/fonc.2021.813897] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/27/2021] [Indexed: 01/10/2023] Open
Abstract
Tumors have special features that make them distinct from their normal counterparts. Immature cells in a tumor mass and their critical contributions to the tumorigenesis will open new windows toward cancer therapy. Incomplete cellular development brings versatile and unique functionality in the cellular tumor ecosystem, such as what is seen for highly potential embryonic cells. There is evidence that maturation of certain types of cells in this ecosystem can recover the sensitivity of the tumor. Therefore, understanding more about the mechanisms that contributed to this immaturity will render new therapeutic approaches in cancer therapy. Targeting such mechanisms can be exploited as a supplementary to the current immunotherapeutic treatment schedules, such as immune checkpoint inhibitor (ICI) therapy. The key focus of this review is to discuss the impact of (im)maturity in cellular tumor ecosystems on cancer progression, focusing mainly on immaturity in the immune cell compartment of the tumor, as well as on the stemness of tumor cells.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
42
|
Expression Profiles of Immune Cells after Propofol or Sevoflurane Anesthesia for Colorectal Cancer Surgery: A Prospective Double-blind Randomized Trial. Anesthesiology 2022; 136:448-458. [PMID: 35051263 DOI: 10.1097/aln.0000000000004119] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The antitumor effects of natural killer cells, helper T cells, and cytotoxic T cells after cancer surgery were reported previously. This study hypothesized that propofol-based anesthesia would have fewer harmful effects on immune cells than volatile anesthetics-based anesthesia during colorectal cancer surgery. METHODS In total, 153 patients undergoing colorectal cancer surgery were randomized and included in the analysis. The primary outcome was the fraction of circulating natural killer cells over time in the propofol and sevoflurane groups. The fractions of circulating natural killer, type 1, type 17 helper T cells, and cytotoxic T cells were investigated. The fractions of CD39 and CD73 expressions on circulating regulatory T cells were investigated, along with the proportions of circulating neutrophils, lymphocytes, and monocytes. RESULTS The fraction of circulating natural killer cells was not significantly different between the propofol and sevoflurane groups until 24 h postoperatively (20.4 ± 13.4% vs. 20.8 ± 11.3%, 17.9 ± 12.7% vs. 20.7 ± 11.9%, and 18.6 ± 11.6% vs. 21.3 ± 10.8% before anesthesia and after 1 and 24 h after anesthesia, respectively; difference [95% CI], -0.3 [-4.3 to 3.6], -2.8 [-6.8 to 1.1], and -2.6 [-6.2 to 1.0]; P = 0.863, P = 0.136, and P = 0.151 before anesthesia and after 1 and 24 h, respectively). The fractions of circulating type 1 and type 17 helper T cells, cytotoxic T cells, and CD39+ and CD73+ circulating regulatory T cells were not significantly different between the two groups. The neutrophil to lymphocyte ratio in both groups remained within the normal range and was not different between the groups. CONCLUSIONS Propofol-based anesthesia was not superior to sevoflurane-based anesthesia in terms of alleviating suppression of immune cells including natural killer cells and T lymphocytes during colorectal cancer surgery. EDITOR’S PERSPECTIVE
Collapse
|
43
|
Väyrynen JP, Haruki K, Lau MC, Väyrynen SA, Ugai T, Akimoto N, Zhong R, Zhao M, Dias Costa A, Borowsky J, Bell P, Takashima Y, Fujiyoshi K, Arima K, Kishikawa J, Shi SS, Twombly TS, Song M, Wu K, Chan AT, Zhang X, Fuchs CS, Meyerhardt JA, Giannakis M, Ogino S, Nowak JA. Spatial organization and prognostic significance of NK and NKT-like cells via multimarker analysis of the colorectal cancer microenvironment. Cancer Immunol Res 2021; 10:215-227. [DOI: 10.1158/2326-6066.cir-21-0772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/24/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022]
|
44
|
Zhong F, Lin Y, Jing X, Ye Y, Wang S, Shen Z. Innate tumor killers in colorectal cancer. Cancer Lett 2021; 527:115-126. [PMID: 34952144 DOI: 10.1016/j.canlet.2021.12.022] [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: 11/15/2021] [Revised: 12/11/2021] [Accepted: 12/18/2021] [Indexed: 12/12/2022]
Abstract
Standard treatment of colorectal cancer (CRC) improves the prognosis of CRC patients, but it is still intractable to control the progression of metastatic CRC. Immune microenvironment and immunotherapies of CRC have received extensive attention in recent years, but present immunotherapies of CRC have mainly focused on T cells and therapeutic response is only observed in a small proportion of patients. Innate immune cells are the first-line of defense in the development of malignancies. Natural killer (NK) cells, NKT cells and γδT cells are three types of innate cells of lymphoid origin and show cytotoxicity against various tumor cells including CRC. Besides, in the development of CRC, they can also be inhibited or express regulatory type, promoting tumor progression. Researches about anti-tumorigenic and pro-tumorigenic mechanisms of these cells are ongoing and regulation of these cells is also being unearthed. Meanwhile, immunotherapies using these cells more or less have shown efficacy in animal models and some of them are under exploration in clinical trials. This review provides an overview of intrinsic properties of NK cell, NKT cell and γδT cell, and summarizes current related promising treatment strategies.
Collapse
Affiliation(s)
- Fengyun Zhong
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China; Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, 100044, PR China.
| | - Yilin Lin
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China; Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, 100044, PR China.
| | - Xiangxiang Jing
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China; Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, 100044, PR China.
| | - Yingjiang Ye
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China; Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, 100044, PR China.
| | - Shan Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China; Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, 100044, PR China.
| | - Zhanlong Shen
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, PR China; Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, 100044, PR China.
| |
Collapse
|
45
|
Zhao LX, Zhang K, Shen BB, Li JN. Mesenchymal stem cell-derived exosomes for gastrointestinal cancer. World J Gastrointest Oncol 2021; 13:1981-1996. [PMID: 35070036 PMCID: PMC8713327 DOI: 10.4251/wjgo.v13.i12.1981] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/15/2021] [Accepted: 09/08/2021] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal (GI) malignancies, a series of malignant conditions originating from the digestive system, include gastric cancer, hepatocellular carcinoma, pancreatic cancer, and colorectal cancer. GI cancers have been regarded as the leading cancer-related cause of death in recent years. Therefore, it is essential to develop effective treatment strategies for GI malignancies. Mesenchymal stem cells (MSCs), a type of distinct non-hematopoietic stem cells and an important component of the tumor microenvironment, play important roles in regulating GI cancer development and progression through multiple mechanisms, such as secreting cytokines and direct interactions. Currently, studies are focusing on the anti-cancer effect of MSCs on GI malignancies. However, the effects and functional mechanisms of MSC-derived exosomes on GI cancer are less studied. MSC-derived exosomes can regulate GI tumor growth, drug response, metastasis, and invasion through transplanting proteins and miRNA to tumor cells to activate the specific signal pathway. Besides, the MSC-derived exosomes are also seen as an important drug delivery system and have shown potential in anti-cancer treatment. This study aims to summarize the effect and biological functions of MSC-derived exosomes on the development of GI cancers and discuss their possible clinical applications for the treatment of GI malignancies.
Collapse
Affiliation(s)
- Lin-Xian Zhao
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, Jilin Province, China
| | - Kai Zhang
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, Jilin Province, China
| | - Bing-Bing Shen
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Jian-Nan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, Jilin Province, China
| |
Collapse
|
46
|
Nguyen OTP, Misun PM, Lohasz C, Lee J, Wang W, Schroeder T, Hierlemann A. An Immunocompetent Microphysiological System to Simultaneously Investigate Effects of Anti-Tumor Natural Killer Cells on Tumor and Cardiac Microtissues. Front Immunol 2021; 12:781337. [PMID: 34925361 PMCID: PMC8675866 DOI: 10.3389/fimmu.2021.781337] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/16/2021] [Indexed: 12/26/2022] Open
Abstract
Existing first-line cancer therapies often fail to cope with the heterogeneity and complexity of cancers, so that new therapeutic approaches are urgently needed. Among novel alternative therapies, adoptive cell therapy (ACT) has emerged as a promising cancer treatment in recent years. The limited clinical applications of ACT, despite its advantages over standard-of-care therapies, can be attributed to (i) time-consuming and cost-intensive procedures to screen for potent anti-tumor immune cells and the corresponding targets, (ii) difficulties to translate in-vitro and animal-derived in-vivo efficacies to clinical efficacy in humans, and (iii) the lack of systemic methods for the safety assessment of ACT. Suitable experimental models and testing platforms have the potential to accelerate the development of ACT. Immunocompetent microphysiological systems (iMPS) are microfluidic platforms that enable complex interactions of advanced tissue models with different immune cell types, bridging the gap between in-vitro and in-vivo studies. Here, we present a proof-of-concept iMPS that supports a triple culture of three-dimensional (3D) colorectal tumor microtissues, 3D cardiac microtissues, and human-derived natural killer (NK) cells in the same microfluidic network. Different aspects of tumor-NK cell interactions were characterized using this iMPS including: (i) direct interaction and NK cell-mediated tumor killing, (ii) the development of an inflammatory milieu through enrichment of soluble pro-inflammatory chemokines and cytokines, and (iii) secondary effects on healthy cardiac microtissues. We found a specific NK cell-mediated tumor-killing activity and elevated levels of tumor- and NK cell-derived chemokines and cytokines, indicating crosstalk and development of an inflammatory milieu. While viability and morphological integrity of cardiac microtissues remained mostly unaffected, we were able to detect alterations in their beating behavior, which shows the potential of iMPS for both, efficacy and early safety testing of new candidate ACTs.
Collapse
Affiliation(s)
- Oanh T. P. Nguyen
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Patrick M. Misun
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Christian Lohasz
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Jihyun Lee
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Weijia Wang
- Cell Systems Dynamics Group, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Timm Schroeder
- Cell Systems Dynamics Group, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Andreas Hierlemann
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| |
Collapse
|
47
|
Cascio S, Chandler C, Zhang L, Sinno S, Gao B, Onkar S, Bruno TC, Vignali DAA, Mahdi H, Osmanbeyoglu HU, Vlad AM, Coffman LG, Buckanovich RJ. Cancer-associated MSC drive tumor immune exclusion and resistance to immunotherapy, which can be overcome by Hedgehog inhibition. SCIENCE ADVANCES 2021; 7:eabi5790. [PMID: 34767446 PMCID: PMC8589308 DOI: 10.1126/sciadv.abi5790] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/23/2021] [Indexed: 05/10/2023]
Abstract
We investigated the impact of cancer-associated mesenchymal stem cells (CA-MSCs) on ovarian tumor immunity. In patient samples, CA-MSC presence inversely correlates with the presence of intratumoral CD8+ T cells. Using an immune “hot” mouse ovarian cancer model, we found that CA-MSCs drive CD8+ T cell tumor immune exclusion and reduce response to anti–PD-L1 immune checkpoint inhibitor (ICI) via secretion of numerous chemokines (Ccl2, Cx3cl1, and Tgf-β1), which recruit immune-suppressive CD14+Ly6C+Cx3cr1+ monocytic cells and polarize macrophages to an immune suppressive Ccr2hiF4/80+Cx3cr1+CD206+ phenotype. Both monocytes and macrophages express high levels of transforming growth factor β–induced (Tgfbi) protein, which suppresses NK cell activity. Hedgehog inhibitor (HHi) therapy reversed CA-MSC effects, reducing myeloid cell presence and expression of Tgfbi, increasing intratumoral NK cell numbers, and restoring response to ICI therapy. Thus, CA-MSCs regulate antitumor immunity, and CA-MSC hedgehog signaling is an important target for cancer immunotherapy.
Collapse
Affiliation(s)
- Sandra Cascio
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Chelsea Chandler
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Linan Zhang
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Sarah Sinno
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Bingsi Gao
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sayali Onkar
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Tullia C. Bruno
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Dario A. A. Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Haider Mahdi
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Hatice U. Osmanbeyoglu
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15213 USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Anda M. Vlad
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lan G. Coffman
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15232, USA
| | - Ronald J. Buckanovich
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15232, USA
| |
Collapse
|
48
|
Songjang W, Nensat C, Pongcharoen S, Jiraviriyakul A. The role of immunogenic cell death in gastrointestinal cancer immunotherapy (Review). Biomed Rep 2021; 15:86. [PMID: 34512974 PMCID: PMC8411483 DOI: 10.3892/br.2021.1462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/30/2021] [Indexed: 12/24/2022] Open
Abstract
Modern cancer immunotherapy techniques are aimed at enhancing the responses of the patients' immune systems to fight against the cancer. The main promising strategies include active vaccination of tumor antigens, passive vaccination with antibodies specific to cancer antigens, adoptive transfer of cancer-specific T cells and manipulation of the patient's immune response by inhibiting immune checkpoints. The application of immunogenic cell death (ICD) inducers has been proven to enhance the immunity of patients undergoing various types of immunotherapy. The dying, stressed or injured cells release or present molecules on the cell surface, which function as either adjuvants or danger signals for detection by the innate immune system. These molecules are now termed 'damage-associated molecular patterns'. The term 'ICD' indicates a type of cell death that triggers an immune response against dead-cell antigens, particularly those derived from cancer cells, and it was initially proposed with regards to the effects of anticancer chemotherapy with conventional cytotoxic drugs. The aim of the present study was to review and discuss the role and mechanisms of ICD as a promising combined immunotherapy for gastrointestinal tumors.
Collapse
Affiliation(s)
- Worawat Songjang
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Chatchai Nensat
- Biomedical Sciences, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Sutatip Pongcharoen
- Division of Immunology, Department of Medicine, Faculty of Medicine, Naresuan University, Phitsanulok 65000, Thailand
| | - Arunya Jiraviriyakul
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| |
Collapse
|
49
|
Kumar S, Sarthi P, Mani I, Ashraf MU, Kang MH, Kumar V, Bae YS. Epitranscriptomic Approach: To Improve the Efficacy of ICB Therapy by Co-Targeting Intracellular Checkpoint CISH. Cells 2021; 10:2250. [PMID: 34571899 PMCID: PMC8466810 DOI: 10.3390/cells10092250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 02/07/2023] Open
Abstract
Cellular immunotherapy has recently emerged as a fourth pillar in cancer treatment co-joining surgery, chemotherapy and radiotherapy. Where, the discovery of immune checkpoint blockage or inhibition (ICB/ICI), anti-PD-1/PD-L1 and anti-CTLA4-based, therapy has revolutionized the class of cancer treatment at a different level. However, some cancer patients escape this immune surveillance mechanism and become resistant to ICB-therapy. Therefore, a more advanced or an alternative treatment is required urgently. Despite the functional importance of epitranscriptomics in diverse clinico-biological practices, its role in improving the efficacy of ICB therapeutics has been limited. Consequently, our study encapsulates the evidence, as a possible strategy, to improve the efficacy of ICB-therapy by co-targeting molecular checkpoints especially N6A-modification machineries which can be reformed into RNA modifying drugs (RMD). Here, we have explained the mechanism of individual RNA-modifiers (editor/writer, eraser/remover, and effector/reader) in overcoming the issues associated with high-dose antibody toxicities and drug-resistance. Moreover, we have shed light on the importance of suppressor of cytokine signaling (SOCS/CISH) and microRNAs in improving the efficacy of ICB-therapy, with brief insight on the current monoclonal antibodies undergoing clinical trials or already approved against several solid tumor and metastatic cancers. We anticipate our investigation will encourage researchers and clinicians to further strengthen the efficacy of ICB-therapeutics by considering the importance of epitranscriptomics as a personalized medicine.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Biological Sciences, Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (M.U.A.); (M.-H.K.)
- Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea
| | - Parth Sarthi
- University Department of Botany, M.Sc. Biotechnology, Ranchi University, Ranchi 834008, India;
| | - Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi 110049, India;
| | - Muhammad Umer Ashraf
- Department of Biological Sciences, Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (M.U.A.); (M.-H.K.)
- Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea
| | - Myeong-Ho Kang
- Department of Biological Sciences, Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (M.U.A.); (M.-H.K.)
- Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea
| | - Vishal Kumar
- Department of Pharmaceutical Science, Dayananda Sagar University, Bengaluru 560078, India;
| | - Yong-Soo Bae
- Department of Biological Sciences, Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea; (M.U.A.); (M.-H.K.)
- Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea
| |
Collapse
|
50
|
Zhu H, Liu X. Advances of Tumorigenesis, Diagnosis at Early Stage, and Cellular Immunotherapy in Gastrointestinal Malignancies. Front Oncol 2021; 11:666340. [PMID: 34434889 PMCID: PMC8381364 DOI: 10.3389/fonc.2021.666340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/19/2021] [Indexed: 01/10/2023] Open
Abstract
Globally, in 2018, 4.8 million new patients have a diagnosis of gastrointestinal (GI) cancers, while 3.4 million people died of such disorders. GI malignancies are tightly relevant to 26% of the world-wide cancer incidence and occupies 35% of all cancer-associated deaths. In this article, we principally investigated molecular and cellular mechanisms of tumorigenesis in five major GI cancers occurring at esophagus, stomach, liver, pancreas, and colorectal region that illustrate high morbidity in Eastern and Western countries. Moreover, through this investigation, we not only emphasize importance of the tumor microenvironment in development and treatment of malignant tumors but also identify significance of M2PK, miRNAs, ctDNAs, circRNAs, and CTCs in early detection of GI cancers, as well as systematically evaluate contribution of personalized precision medicine including cellular immunotherapy, new antigen and vaccine therapy, and oncolytic virotherapy in treatment of GI cancers.
Collapse
Affiliation(s)
- Haipeng Zhu
- Precision and Personalized Cancer Treatment Center, Division of Cancer Diagnosis & Therapy, Ciming Boao International Hospital, Boao Lecheng International Medical Tourism Pilot Zone, Qionghai, China.,Stem Cell and Biotherapy Technology Research Center, Xinxiang Medical College, Xinxiang, China
| | - Xiaojun Liu
- Division of Cellular & Biomedical Science, Ciming Boao International Hospital, Boao Lecheng International Medical Tourism Pilot Zone, Qionghai, China
| |
Collapse
|