1
|
Hosseini SA, Nasab NK, Kargozar S, Wang AZ. Advanced biomaterials and scaffolds for cancer immunotherapy. BIOMATERIALS FOR PRECISION CANCER MEDICINE 2025:377-424. [DOI: 10.1016/b978-0-323-85661-4.00016-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
2
|
Gharatape A, Amanzadi B, Mohamadi F, Rafieian M, Faridi-Majidi R. Recent advances in polymeric and lipid stimuli-responsive nanocarriers for cell-based cancer immunotherapy. Nanomedicine (Lond) 2024; 19:2655-2678. [PMID: 39540464 DOI: 10.1080/17435889.2024.2416377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Conventional cancer therapy has major limitations, including non-specificity, unavoidable side effects, low specific tumor accumulation and systemic toxicity. In recent years, more effective and precise treatment methods have been developed, including cell-based immunotherapy. Carriers that can accurately and specifically target cells and equip them to combat cancer cells are particularly important for developing this therapy. As a result, attention has been drawn to smart nanocarriers that can react to specific stimuli. Thus, stimuli-responsive nanocarriers have attracted increasing attention because they can change their physicochemical properties in response to stimulus conditions, such as pH, enzymes, redox agents, hypoxia, light and temperature. This review highlights recent advances in various stimuli-responsive nanocarriers, discussing loading, targeted delivery, cellular uptake, biocompatibility and immunomodulation in cell-based immunotherapy. Finally, future challenges and perspectives regarding the possible clinical translation of nanocarriers are discussed.
Collapse
Affiliation(s)
- Alireza Gharatape
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Bentolhoda Amanzadi
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Faranak Mohamadi
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Mahdieh Rafieian
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Reza Faridi-Majidi
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
- Pharmaceutical Nanotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Chatterjee D, Bhattacharya S, Kumari L, Datta A. Aptamers: ushering in new hopes in targeted glioblastoma therapy. J Drug Target 2024; 32:1005-1028. [PMID: 38923419 DOI: 10.1080/1061186x.2024.2373306] [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: 04/16/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024]
Abstract
Glioblastoma, a formidable brain cancer, has remained a therapeutic challenge due to its aggressive nature and resistance to conventional treatments. Recent data indicate that aptamers, short synthetic DNA or RNA molecules can be used in anti-cancer therapy due to their better tumour penetration, specific binding affinity, longer retention in tumour sites and their ability to cross the blood-brain barrier. With the ability to modify these oligonucleotides through the selection process, and using rational design to modify them, post-SELEX aptamers offer several advantages in glioblastoma treatment, including precise targeting of cancer cells while sparing healthy tissue. This review discusses the pivotal role of aptamers in glioblastoma therapy and diagnosis, emphasising their potential to enhance treatment efficacy and also highlights recent advancements in aptamer-based therapies which can transform the landscape of glioblastoma treatment, offering renewed hope to patients and clinicians alike.
Collapse
Affiliation(s)
- Debarpan Chatterjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Srijan Bhattacharya
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Leena Kumari
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Aparna Datta
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| |
Collapse
|
4
|
Hoch CC, Hachani K, Han Y, Schmidl B, Wirth M, Multhoff G, Bashiri Dezfouli A, Wollenberg B. The future of interleukin gene therapy in head and neck cancers. Expert Opin Biol Ther 2024; 24:1057-1073. [PMID: 39291462 DOI: 10.1080/14712598.2024.2405568] [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: 06/26/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
INTRODUCTION Head and neck cancer (HNC), primarily head and neck squamous cell carcinomas, originates from the squamous epithelium in areas like the oral cavity, lip, larynx, and oropharynx. With high morbidity impacting critical functions, combined treatments like surgery, radiation, and chemotherapy often fall short in advanced stages, highlighting the need for innovative therapies. AREAS COVERED This review critically evaluates interleukin (IL) gene therapy for treating HNC. The discussion extends to key ILs in HNC, various gene therapy techniques and delivery methods. We particularly focus on the application of IL-2, IL-12, and IL-24 gene therapies, examining their mechanisms and outcomes in preclinical studies and clinical trials. The final sections address IL gene therapy challenges in HNC, exploring solutions and critically assessing future therapeutic directions. EXPERT OPINION Despite advancements in genomic and immunotherapy, significant challenges in HNC treatment persist, primarily due to the immunosuppressive nature of the tumor microenvironment and the adverse effects of current therapies. The therapeutic efficacy of IL gene therapy hinges on overcoming these hurdles through refined delivery methods that ensure targeted, tumor-specific gene expression. Future strategies should focus on refining gene delivery methods and combining IL gene therapy with other treatments to optimize efficacy and minimize toxicity.
Collapse
Affiliation(s)
- Cosima C Hoch
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Khouloud Hachani
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Yu Han
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Benedikt Schmidl
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Markus Wirth
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Gabriele Multhoff
- Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Munich, Germany
- Department of Radiation Oncology, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Ali Bashiri Dezfouli
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
- Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Munich, Germany
- Department of Radiation Oncology, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| |
Collapse
|
5
|
Zhang Y, Chen X, Hu B, Zou B, Xu Y. Advancements in nanomedicine delivery systems: unraveling immune regulation strategies for tumor immunotherapy. Nanomedicine (Lond) 2024; 19:1821-1840. [PMID: 39011582 PMCID: PMC11418288 DOI: 10.1080/17435889.2024.2374230] [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: 03/27/2024] [Accepted: 06/26/2024] [Indexed: 07/17/2024] Open
Abstract
This review highlights the significant role of nanodrug delivery systems (NDDS) in enhancing the efficacy of tumor immunotherapy. Focusing on the integration of NDDS with immune regulation strategies, it explores their transformative impacts on the tumor microenvironment and immune response dynamics. Key advancements include the optimization of drug delivery through NDDS, targeting mechanisms like immune checkpoint blockade and modulating the immunosuppressive tumor environment. Despite the progress, challenges such as limited clinical efficacy and complex manufacturing processes persist. The review emphasizes the need for further research to optimize these systems, potentially revolutionizing cancer treatment by improving delivery efficiency, reducing toxicity and overcoming immune resistance.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Xi Chen
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Binbin Hu
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Bingwen Zou
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Yong Xu
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| |
Collapse
|
6
|
Narayan VM, Meeks JJ, Jakobsen JS, Shore ND, Sant GR, Konety BR. Mechanism of action of nadofaragene firadenovec-vncg. Front Oncol 2024; 14:1359725. [PMID: 38559556 PMCID: PMC10979480 DOI: 10.3389/fonc.2024.1359725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/14/2024] [Indexed: 04/04/2024] Open
Abstract
Effective bladder-preserving therapeutic options are needed for patients with bacillus Calmette-Guérin unresponsive non-muscle-invasive bladder cancer. Nadofaragene firadenovec-vncg (Adstiladrin®) was approved by the US Food and Drug Administration as the first gene therapy in urology and the first intravesical gene therapy indicated for the treatment of adult patients with high-risk bacillus Calmette-Guérin-unresponsive non-muscle-invasive bladder cancer with carcinoma in situ with or without papillary tumors. The proposed mechanism of action underlying nadofaragene firadenovec efficacy is likely due to the pleiotropic nature of interferon-α and its direct and indirect antitumor activities. Direct activities include cell death and the mediation of an antiangiogenic effect, and indirect activities are those initiated through immunomodulation of the innate and adaptive immune responses. The sustained expression of interferon-α that results from this treatment modality contributes to a durable response. This review provides insight into potential mechanisms of action underlying nadofaragene firadenovec efficacy.
Collapse
Affiliation(s)
| | - Joshua J. Meeks
- Department of Urology, Northwestern University, Chicago, IL, United States
| | - Jørn S. Jakobsen
- Ferring Pharmaceuticals, International PharmaScience Center, Copenhagen, Denmark
| | - Neal D. Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, United States
| | - Grannum R. Sant
- Department of Urology, Tufts University School of Medicine, Boston, MA, United States
| | | |
Collapse
|
7
|
Zhang J, Xiao Y, Zhang J, Yang Y, Zhang L, Liang F. Recent advances of engineered oncolytic viruses-based combination therapy for liver cancer. J Transl Med 2024; 22:3. [PMID: 38167076 PMCID: PMC10763442 DOI: 10.1186/s12967-023-04817-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
Liver cancer is a major malignant tumor, which seriously threatens human health and increases the economic burden on patients. At present, gene therapy has been comprehensively studied as an excellent therapeutic measure in liver cancer treatment. Oncolytic virus (OV) is a kind of virus that can specifically infect and kill tumor cells. After being modified by genetic engineering, the specificity of OV infection to tumor cells is increased, and its influence on normal cells is reduced. To date, OV has shown its effectiveness and safety in experimental and clinical studies on a variety of tumors. Thus, this review primarily introduces the current status of different genetically engineered OVs used in gene therapy for liver cancer, focuses on the application of OVs and different target genes for current liver cancer therapy, and identifies the problems encountered in OVs-based combination therapy and the corresponding solutions, which will provide new insights into the treatment of liver cancer.
Collapse
Affiliation(s)
- Junhe Zhang
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China.
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China.
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Yunxi Xiao
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
| | - Jie Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yun Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Liao Zhang
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
| | - Fan Liang
- Institutes of Health Central Plains, Xinxiang Medical University, No. 601 Jinsui Road, Xinxiang, 453003, Henan Province, China
| |
Collapse
|
8
|
Wang C, Shen Y, Ma Y. Bifidobacterium infantis-Mediated Herpes Simplex Virus-TK/Ganciclovir Treatment Inhibits Cancer Metastasis in Mouse Model. Int J Mol Sci 2023; 24:11721. [PMID: 37511481 PMCID: PMC10380465 DOI: 10.3390/ijms241411721] [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: 06/13/2023] [Revised: 07/03/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Previous studies have found that Bifidobacterium infantis-mediated herpes simplex virus-TK/ganciclovir (BF-TK/GCV) reduces the expression of VEGF and CD146, implying tumor metastasis inhibition. However, the mechanism by which BF-TK/GCV inhibits tumor metastasis is not fully studied. Here, we comprehensively identified and quantified protein expression profiling for the first time in gastric cancer (GC) cells MKN-45 upon BF-TK/GCV treatment using quantitative proteomics. A total of 159 and 72 differential expression proteins (DEPs) were significantly changed in the BF-TK/GCV/BF-TK and BF-TK/GCV/BF/GCV comparative analysis. Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis enriched some metastasis-related pathways such as gap junction and cell adhesion molecules pathways. Moreover, the transwell assay proved that BF-TK/GCV inhibited the invasion and migration of tumor cells. Furthermore, immunohistochemistry (IHC) demonstrated that BF-TK/GCV reduced the expression of HIF-1α, mTOR, NF-κB1-p105, VCAM1, MMP13, CXCL12, ATG16, and CEBPB, which were associated with tumor metastasis. In summary, BF-TK/GCV inhibited tumor metastasis, which deepened and expanded the understanding of the antitumor mechanism of BF-TK/GCV.
Collapse
Affiliation(s)
- Changdong Wang
- Department of Biochemistry & Molecular Biology, Molecular Medicine & Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yanxi Shen
- Department of Biochemistry & Molecular Biology, Molecular Medicine & Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yongping Ma
- Department of Biochemistry & Molecular Biology, Molecular Medicine & Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
9
|
Zhu M, Tang X, Zhu Z, Gong Z, Tang W, Hu Y, Cheng C, Wang H, Sarwar A, Chen Y, Liu F, Huo J, Wang X, Zhang Y. STING activation in macrophages by vanillic acid exhibits antineoplastic potential. Biochem Pharmacol 2023; 213:115618. [PMID: 37211172 DOI: 10.1016/j.bcp.2023.115618] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
The host stimulator of interferon genes (STING) signaling pathway is a major innate immune sensing pathway, and the stimulation of this pathway within antigen-presenting cells shows promise in targeting immune-suppressed tumors. Macrophages resident in tumors exhibit anti-inflammatory properties and enhance tumor growth and development. Polarizing such macrophages towards a pro-inflammatory phenotype is an effective strategy for tumor suppression. In the present study, we observed that the STING pathway was inactivated in breast and lung carcinomas, and a positive correlation existed between STING and macrophage markers in these tumors. We found that vanillic acid (VA) could stimulate the STING/TBK1/IRF3 pathway. VA mediated the production of type I IFN and promoted macrophage polarization into the M1 phenotype; this activity was dependent on STING activation. A direct-contact co-culture model and a transwell co-culture model revealed that macrophages with VA-induced STING activation exhibited anti-proliferative effects on SKBR3 and H1299 cells, although a STING antagonist and M2 macrophage-related cytokines alleviated this anti-proliferative effect. Further investigation indicated that phagocytosis and apoptosis-inducing effects were the major mediators of the anti-tumor effect of VA-treated macrophages. Mechanistically, VA promoted the polarization of macrophages to a M1 phenotype via IL-6R/JAK signaling, resulting in enhanced phagocytosis and apoptosis-induction effects. Additionally, STING activation-induced IFNβ production also participated in the apoptosis mediated by VA-treated macrophage in SKBR3 and H1299 cells. Mouse models with 4 T1 tumors confirmed the anti-tumor properties of VA in vivo and revealed the infiltration of VA-induced cytotoxic T cells into the tumors. These data suggest that VA is an effective agonist of STING and provides a new perspective for cancer immunotherapy.
Collapse
Affiliation(s)
- Man Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoyu Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an 710061, China
| | - Zeren Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhengyan Gong
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an 710061, China
| | - Wenjuan Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yu Hu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an 710061, China
| | - Cheng Cheng
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an 710061, China
| | - Hongying Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ammar Sarwar
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yanbin Chen
- Shaanxi Institute of International Trade & Commerce, Xianyang 712046, China; Shaanxi Buchang Pharmaceutical Co. Ltd, Xi'an 710075, China
| | - Feng Liu
- Shaanxi Institute of International Trade & Commerce, Xianyang 712046, China; Shaanxi Buchang Pharmaceutical Co. Ltd, Xi'an 710075, China
| | - Jian Huo
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xuemei Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta Weststreet, #54, Xi'an, Shaanxi Province 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
10
|
Tian Y, Xie T, Sun X. Analysis of the regulatory mechanisms of prognostic immune factors in thyroid cancer. Front Oncol 2022; 12:1059591. [PMID: 36591507 PMCID: PMC9795211 DOI: 10.3389/fonc.2022.1059591] [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: 10/01/2022] [Accepted: 11/24/2022] [Indexed: 12/15/2022] Open
Abstract
Objective To explore the regulatory mechanism of immune prognostic factors in thyroid cancer. Methods Based on the TCGA database and GEO database, this study used bioinformatics methods to study the potential regulatory mechanism of thyroid cancer prognosis, analyzed the differentially expressed genes and differential miRNAs between thyroid cancer and normal paracancerous tissues by R software, and constructed lasso risk factors. The immune prognostic factors of thyroid cancer were obtained from the model, and the miRDB website was used to predict the possibility of differential miRNA target binding of the immune prognostic factors and correlation analysis was performed, and finally verified by cell experiments. Results There were 1413 differentially expressed genes between thyroid cancer and normal paracancerous tissues, among which 21 immune-related genes were prognostic factors with significant differences in expression; lasso risk model obtained AKAP12, APOC1, TIMP3, ADAMTS9, ANK2, HTRA3, SYNDIG1 , ADAMTS5 and DACT1 were nine prognostic factors. A total of 58 differential miRNAs were found in thyroid cancer tissues and non-cancerous tissues. The possibility of differential miRNA targeting and binding of immune prognostic factors on the miRDB website and cell experiments was analyzed. Conclusions The potential miRNA regulatory mechanism of immune prognostic factors in thyroid cancer has been explored.
Collapse
Affiliation(s)
- Yin Tian
- Department of Pediatric Surgery, Jingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, China
| | - Tao Xie
- Department of Anesthesiology, Jingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, China
| | - Xue Sun
- Department of Clinical Nutrition, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
11
|
Jiang Y, Feng D, Wang C, Zhang Y, Zhao C, Li S, Qin Y, Chang AH, Zhu J. Administration of granulocyte-macrophage colony-stimulating factor enhanced chimeric antigen receptor T-cell expansion and cellular immunity recovery without inducing cytokine release syndrome. Front Med (Lausanne) 2022; 9:1042501. [PMID: 36405594 PMCID: PMC9669452 DOI: 10.3389/fmed.2022.1042501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
Background Neutropenia and cytokine release syndrome (CRS) are two major toxicities of chimeric antigen receptor (CAR)-T cell therapy. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is an ideal candidate treatment for neutropenia except for its potential aggravation of CRS. We hypothesized that the optimal timing of supplemental with GM-CSF in a shortage of host immunity and CAR T-cell was chosen as avoidance of CRS. In the study we evaluated the safety and efficacy of GM-CSF intervention post-CAR T-cell therapy while circulating CAR T-cell declined. Materials and methods Nine patients received GM-CSF therapy who displayed moderate neutropenia with absolute neutrophil counts (ANC) < 1,500 cells/mm3 with concomitant declination of circulating CAR T-cell. Results The median duration of GM-CSF intervention was 15 days (4–30). CAR T-cell expansion was observed in peripheral blood (PB) of seven patients (7/9). The median baseline and peak CAR T cells count in PB of the seven patients with CAR T-cell expansion were 0.85 × 106/L (0–50.9) and 6.06 × 106/L (1.43–112.55). And the peaks of CAR T-cell levels in PB appeared in day 7 (2–11) following the initiation of GM-CSF administration with increases of 2.84 × 106/L (0.38–61.65). Also, increased white blood cells in PB were observed in all patients. The median onset and duration time of WBC recovery were 9 (1–14) and 17 (3–53) days. Moreover, the increment of WBC, neutrophil, lymphocyte and CD3-CD16 + CD56 + natural killer cell in PB was observed. In addition, no CRS or fatal infection occurred during GM-CSF treatment. Conclusion This study provides evidence for the clinical feasibility of combining CAR T-cell therapy with the GM-CSF to treat neutropenia patients with concomitant declination of circulating CAR T-cell.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Hematology, Shanghai Zhaxin Traditional Chinese and Western Medicine Hospital, Shanghai, China
| | - Dan Feng
- Department of Hematology, Shanghai Zhaxin Traditional Chinese and Western Medicine Hospital, Shanghai, China
| | - Chun Wang
- Department of Hematology, Shanghai Zhaxin Traditional Chinese and Western Medicine Hospital, Shanghai, China
| | - Yanlei Zhang
- Shanghai YaKe Biotechnology Ltd., Shanghai, China
| | - Chuxian Zhao
- Department of Hematology, Shanghai Zhaxin Traditional Chinese and Western Medicine Hospital, Shanghai, China
| | - Su Li
- Department of Hematology, Shanghai Zhaxin Traditional Chinese and Western Medicine Hospital, Shanghai, China
| | - Youwen Qin
- Department of Laboratory, Shanghai Zhaxin Traditional Chinese and Western Medicine Hospital, Shanghai, China
| | - Alex H. Chang
- Shanghai YaKe Biotechnology Ltd., Shanghai, China
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Alex H. Chang,
| | - Jun Zhu
- Department of Hematology, Shanghai Zhaxin Traditional Chinese and Western Medicine Hospital, Shanghai, China
- *Correspondence: Jun Zhu,
| |
Collapse
|
12
|
Hunt C, Montgomery S, Berkenpas JW, Sigafoos N, Oakley JC, Espinosa J, Justice N, Kishaba K, Hippe K, Si D, Hou J, Ding H, Cao R. Recent Progress of Machine Learning in Gene Therapy. Curr Gene Ther 2021; 22:132-143. [PMID: 34161210 DOI: 10.2174/1566523221666210622164133] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/15/2021] [Accepted: 04/02/2021] [Indexed: 11/22/2022]
Abstract
With new developments in biomedical technology, it is now a viable therapeutic treatment to alter genes with techniques like CRISPR. At the same time, it is increasingly cheaper to do whole genome sequencing, resulting in rapid advancement in gene therapy and editing in precision medicine. Thus, understanding the current industry and academic applications of gene therapy provides an important backdrop to future scientific developments. Additionally, machine learning and artificial intelligence techniques allow for the reduction of time and money spent in the development of new gene therapy products and techniques. In this paper, we survey the current progress of gene therapy treatments for several diseases and explore machine learning applications in gene therapy. We also discuss the ethical implications of gene therapy and the use of machine learning in precision medicine. Machine learning and gene therapy are both topics gaining popularity in various publications, and we conclude that there is still room for continued research and application of machine learning techniques in the gene therapy field.
Collapse
Affiliation(s)
- Cassandra Hunt
- Department of Computer Science, Pacific Lutheran University, Tacoma, WA, United States
| | - Sandra Montgomery
- Department of Physics, Pacific Lutheran University, Tacoma, WA, United States
| | | | - Noel Sigafoos
- Department of Computer Science, Pacific Lutheran University, Tacoma, WA, United States
| | - John Christian Oakley
- Department of Computer Science, Pacific Lutheran University, Tacoma, WA, United States
| | - Jacob Espinosa
- Department of Mathematics, Pacific Lutheran University, Tacoma, WA, United States
| | - Nicola Justice
- Department of Mathematics, Pacific Lutheran University, Tacoma, WA, United States
| | - Kiyomi Kishaba
- Department of Humanities, Pacific Lutheran University, Tacoma, WA, United States
| | - Kyle Hippe
- Department of Computer Science, Pacific Lutheran University, Tacoma, WA, United States
| | - Dong Si
- Division of Computing Software Systems, University of Washington-Bothell, Bothell, WA, United States
| | - Jie Hou
- Department of Computer Science, Saint Louis University, St. Louis, MO, United States
| | - Hui Ding
- School of Life Science and Technology and Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu, China
| | - Renzhi Cao
- Department of Computer Science, Pacific Lutheran University, Tacoma, WA, United States
| |
Collapse
|