1
|
Blitzer GC, Paz C, McCoy SS, Kimple RJ. Radiation-Therapy Related Salivary Dysfunction. Semin Radiat Oncol 2025; 35:278-284. [PMID: 40090753 PMCID: PMC11911547 DOI: 10.1016/j.semradonc.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/16/2025] [Indexed: 03/18/2025]
Abstract
Radiation-induced xerostomia (RIX) is a common and debilitating side effect of head and neck cancer radiotherapy, significantly impacting patients' quality of life. This review comprehensively summarizes the current understanding of RIX, encompassing its clinical quantification, underlying pathophysiology, and established and emerging treatment modalities. We explore various objective and subjective measures used to quantify salivary flow and assess the severity of xerostomia in clinical settings. The pathophysiological mechanisms leading to RIX are elucidated, including radiation damage to salivary glands, alterations in saliva composition, and the role of inflammatory processes. Current treatment strategies, such as saliva substitutes and stimulants, are discussed alongside their limitations. Furthermore, we delve into novel investigational approaches, including gene therapy, stem cell transplantation, and pharmacologic interventions, offering promising avenues for future RIX management. This review provides clinicians and researchers with a comprehensive overview of RIX, highlighting the need for continued research to develop more effective preventative and therapeutic strategies to alleviate this burdensome condition.
Collapse
Affiliation(s)
- Grace C Blitzer
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI.; University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Cristina Paz
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Sara S McCoy
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI.; Department of Medicine, Division of Rheumatology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI.; University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI..
| |
Collapse
|
2
|
Zhao Z, He D, Wang J, Xiao Y, Gong L, Tang C, Peng H, Qiu X, Liu R, Zhang T, Li J. Swertiamarin relieves radiation-induced intestinal injury by limiting DNA damage. Mol Cell Biochem 2025; 480:2277-2290. [PMID: 38795212 DOI: 10.1007/s11010-024-05030-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 05/04/2024] [Indexed: 05/27/2024]
Abstract
Radiotherapy is the conventional treatment for pelvic abdominal tumors. However, it can cause some damage to the small intestine and colorectal, which are very sensitive to radiation. Radiation-induced intestinal injury (RIII) affects the prognosis of radiotherapy, causing sequelae of loss of function and long-term damage to patients' quality of life. Swertiamarin is a glycoside that has been reported to prevent a variety of diseases including but not limited to diabetes, hypertension, atherosclerosis, arthritis, malaria, and abdominal ulcers. However, its therapeutic effect and mechanism of action on RIII have not been established. We investigated whether swertiamarin has a protective effect against RIII. In this article, we use irradiator to create cellular and mouse models of radiation damage. Preventive administration of swertiamarin could reduce ROS and superoxide anion levels to mitigate the cellular damage caused by radiation. Swertiamarin also attenuated RIII in mice, as evidenced by longer survival, less weight loss and more complete intestinal barrier. We also found an increase in the relative abundance of primary bile acids in irradiated mice, which was reduced by both FXR agonists and swertiamarin, and a reduction in downstream interferon and inflammatory factors via the cGAS-STING pathway to reduce radiation-induced damage.
Collapse
Affiliation(s)
- Zhe Zhao
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, China
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Dan He
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, China
| | - Jinyu Wang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Yu Xiao
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, China
| | - Lixin Gong
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, China
| | - Can Tang
- School of Biological Science and Technology, Chengdu Medical College, Chengdu, China
| | - Haibo Peng
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xuemei Qiu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Tao Zhang
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, China.
- School of Biological Science and Technology, Chengdu Medical College, Chengdu, China.
| | - Jingyi Li
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, China.
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China.
| |
Collapse
|
3
|
Huang Y, Li J, Wang S, Tian H, Fan S, Zhao Y. Diselenide-based nanoparticles enhancing the radioprotection to the small intestine of mice. J Nanobiotechnology 2025; 23:236. [PMID: 40119423 PMCID: PMC11929180 DOI: 10.1186/s12951-025-03276-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 02/24/2025] [Indexed: 03/24/2025] Open
Abstract
The widespread application of ionizing radiation (IR) in medicine, while beneficial, also poses potential risks that necessitate effective countermeasures. Both 2-(3-aminopropylamino) ethanethiol (WR-1065) and curcumin are recognized as radioprotective agents; however, their clinical utility is hindered by notable shortcomings that could be addressed through reactive oxygen species (ROS)-responsive amphiphilic nanomaterials. We introduced a newly synthesized poly (ethylene glycol) (PEG)-polycaprolactone (PCL) polymer integrated with diselenide bonds and curcumin (HOOC-SeSe-Cur-PEG-SeSe-Cur-PCL, PEG-Cur-SeSe-PCL). The resulting spherical nanoparticles (NPs), which self-assembled from this polymer, were uniform with an average diameter of 118 nm. As a carrier for WR-1065, these NPs demonstrated a loading capacity of 30.9% and an efficacy of 56.7%. Importantly, the degradation of WR-1065 within the NPs was minimal in gastric fluid, decreasing by only approximately 20% over a 6-hour period. The innovative aspect of these NPs is their design to destabilize in ROS-rich environments, facilitating the release of WR-1065 and curcumin. Indeed, the survival rate of mice increased to 50% when these NPs were orally administered prior to exposure to a lethal dose of whole-body irradiation (8 Gy). The radioprotective impact of WR-1065-loaded NPs was evident in the small intestine of irradiated mice, characterized by the amelioration of radiation-induced epithelial damage, reduction of DNA damage, and inhibition of the apoptotic pathway. Collectively, this oral nanocarrier system for WR-1065 and curcumin holds promise as a potential candidate for the prophylaxis and treatment of acute intestinal injuries induced by IR.
Collapse
Affiliation(s)
- Yichi Huang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Jiaze Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Sen Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Hongqi Tian
- Kechow Pharma, Inc., Shanghai, 200131, China.
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China.
| | - Yu Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China.
| |
Collapse
|
4
|
Liu D, Wei M, Fang Y, Yuan T, Sun Y, Xie H, Yan W, Yuan B, Zhuang B, Jin Y. Oral colon-retentive inulin gels protect against radiation-induced hematopoietic and gastrointestinal injury by improving gut homeostasis. Int J Biol Macromol 2025; 292:139199. [PMID: 39730057 DOI: 10.1016/j.ijbiomac.2024.139199] [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: 08/24/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Ionizing radiation-induced injury often occurs in nuclear accidents or large-dose radiotherapy, leading to acute radiation syndromes characterized by hematopoietic and gastrointestinal injuries even to death. However, current radioprotective drugs are only used in hospitals with unavoidable side effects. Here, we heated the aqueous solution of inulin, a polysaccharide dietary fiber, forming colon-retentive gel as a radiation protector in radiotherapy. Mouse models were established after 60Co γ-ray irradiation of the total body or abdomen. Inulin gels were orally administered to the mice every day from 3 days pre-radiation to 3 days post-radiation. The hematopoietic system was well protected with good blood cell recovery and cell proliferation in the femur and spleen. Oral inulin gels increased the relative abundances of key commensal microorganisms including f_Lachnospiraceae, Akkermansia, Blautia, and short-chain fatty acid metabolites. The secretion of the anti-inflammation cytokines IL-22 and IL-10 in the intestinal cells also increased. Similarly, the expression of the tight junction proteins claudin-1 and occludin in the gut mucosa was affected. In an orthotopic murine colorectal cancer model, oral inulin gels followed by 10-Gy abdomen radiation improved the radiotherapy efficiency with low attenuated radiation injury. Taken the data together, these results suggest that oral inulin gels are a bioactive material against ionizing radiation-induced injury.
Collapse
Affiliation(s)
- Dongdong Liu
- Beijing Institute of Radiation Medicine, Beijing 100850, China; China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing 100068, China
| | - Meng Wei
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yubao Fang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Tianyu Yuan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yingbao Sun
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hua Xie
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wenrui Yan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bochuan Yuan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bo Zhuang
- Institute of NBC Defense, Beijing 102205, China.
| | - Yiguang Jin
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
5
|
Wang Y, Zhao D, Nong X. Artesunate alleviates radiation-induced submandibular gland epithelial cell damage in rats by reducing inflammation and apoptosis. Cell Biol Int 2025; 49:250-261. [PMID: 39607036 DOI: 10.1002/cbin.12261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/09/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024]
Abstract
Salivary hypofunction is a common complication in patients with head and neck cancers following radiotherapy (RT). RT-induced inflammation in salivary gland cells leads to apoptosis and fibrosis. Artesunate (ART) is a bioactive compound with anti-inflammatory and anti-fibrosis properties. This study aimed to investigate the protective effects of ART on X-ray-induced injury of submandibular gland (SMG) epithelial cells in rats. Second-generation SMG epithelial cells were randomly divided into five groups: natural control group (NC), irradiated group (IR), and irradiated groups treated with ART at concentrations of 5, 10, and 20 μM. Cells were harvested 48 h postirradiation for analysis. The results demonstrated that ART attenuated the damage to AQP5, a crucial indicator of salivary gland function, as evidenced by the decreased expression of AQP5 at both mRNA and protein levels. Additionally, ART decreased the expression of inflammatory cytokines: IL-6 and TNF-α. TUNEL staining revealed reduced apoptosis in the ART groups, particularly the IR + 10 μM group. RT-PCR and Western blot analysis of apoptosis cytokines Bax/Bcl-2 and Caspase-3 confirmed these findings. Furthermore, ART inhibited the expression of NF-κB at both mRNA and protein levels. In conclusion, these results suggest that ART may reduce inflammation and apoptosis in SMG epithelial cells following radiation by inhibiting the NF-κB pathway.
Collapse
Affiliation(s)
- Yuchen Wang
- College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Danni Zhao
- College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaolin Nong
- College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Nanning, Guangxi, China
| |
Collapse
|
6
|
Yamaguchi M, Htun KT, Tatara Y, Sato Y, Hosoda M, Kothan S, Kamimura C, Inanami O, Kashiwakura I. Radio-protective effects of ultra-fine bubble hydrogen water and serum protein responses in whole-body radiation-exposed mice. Sci Rep 2025; 15:4447. [PMID: 39910205 PMCID: PMC11799536 DOI: 10.1038/s41598-025-87963-z] [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] [Accepted: 01/23/2025] [Indexed: 02/07/2025] Open
Abstract
Many studies have demonstrated hydrogen's therapeutic and preventive effects on various diseases. Its selective antioxidant properties against hydroxyl radicals, which are responsible for the indirect effects of ionizing radiation, may make it worthy of attention as a new radio-protector. We recently developed new hydrogen water that is more stable and has higher antioxidant activity by using ultra-fine bubbles. In this study, female C57BL/6J mice given ad libitum access to ultra-fine bubble hydrogen water (UBHW) were subjected to whole-body irradiation (WBI) with X-rays, and the radio-protective effect of UBHW was evaluated. WBI with 6.0 Gy (sub-lethal dose) resulted in a 30-day survival rate of 100% in UBHW-fed mice, compared with 37% in control mice. In the case of WBI with 6.5 Gy (lethal dose), while the control mice died out in about 3 weeks, the 30-day survival rate improved to 40% by UBHW due to the high scavenging activity of hydroxy radicals. Twenty-six serum proteins involved in inflammatory and immune responses were significantly identified in UBHW-fed mice by proteomics, and UBHW may enhance and regulate these functions, resulting in reduced damage in mice exposed to WBI. We conclude that UBHW has good potential in radio-protection, with evidence that warrants further research efforts in this field.
Collapse
Affiliation(s)
- Masaru Yamaguchi
- Graduate School of Health Sciences, Hirosaki University, 66-1 Hon-cho, Hirosaki, 036-8564, Aomori, Japan
| | - Khin Thandar Htun
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Youta Tatara
- Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, 036-8562, Aomori, Japan
| | - Yoshiaki Sato
- Norris Comprehensive Cancer Center, University of Southern California, 1450 Biggy Street, NRT Rm 4516, Los Angeles, CA, 90033, USA
| | | | - Suchart Kothan
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chikashi Kamimura
- Information Science Research Institute Ltd, 535-3 Mohno, Ueki-cho, Kita-ku, Kumamoto, Kumamoto, 861-0134, Japan
| | - Osamu Inanami
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Ikuo Kashiwakura
- Graduate School of Health Sciences, Hirosaki University, 66-1 Hon-cho, Hirosaki, 036-8564, Aomori, Japan.
| |
Collapse
|
7
|
Wang K, Yuan B, Zhang F, Li Z, Jia X, Hu Y, Chen Z, Hong J, Du L, Jin Y. A bioadhesive antioxidase-overexpressed probiotic prevents radiation enteritis by scavenging the excess reactive oxygen species. Free Radic Biol Med 2025; 227:485-498. [PMID: 39643134 DOI: 10.1016/j.freeradbiomed.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
The scavenging of the excess reactive oxygen species (ROS) induced by radiation is fundamental for radiation protection. However, directly applying antioxidants results in low bioavailability and side effects. Superoxide dismutase (SOD) and catalase (CAT) have high ROS clearance efficiency, whereas their application is limited by the enzyme inactivation, making it difficult to exhibit significant therapeutic effects. Here, we engineered a probiotic Escherichia coli Nissle 1917 (EcN), i.e., AAEcN, serving as a SOD/CAT vehicle to scavenge ROS for the prevention and treatment of radiation enteritis (RE). The overexpressed Drsod and katE in AAEcN showed 5-fold ROS elimination efficiency compared to the wild EcN. Furthermore, the intestinal retention time of engineered EcN was prolonged through trefoil factor 3 gene (TFF3) modification of curli fibers on the bacterial surface, which contributed to the persistence of antioxidant enzyme activity. We found that AAEcN rapidly eliminated the intracellular ROS induced by radiation. Only a single oral dosing of AAEcN was satisfied to alleviate the radiation damage to the small intestine, colon, and spleen. Moreover, the homeostasis of pro-/anti-inflammatory cytokines was realized. The proliferation of the intestinal stem cells and spleen hematopoietic stem cells was enhanced, while the apoptosis of mucosal cells was inhibited. Our findings suggest valuable insights into the ROS scavenging way in RE, and establish an empirical basis for developing probiotics as an antioxidant enzyme vehicle for the bacteriotherapy of RE.
Collapse
Affiliation(s)
- Ke Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Bochuan Yuan
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Feng Zhang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Zhangyu Li
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Xueli Jia
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yadan Hu
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Ziyuan Chen
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jinyun Hong
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Lina Du
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yiguang Jin
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
8
|
Zhou H, Xu Z, Jiang C, Wu Q, Zhang C, Liu Z, Zhang X, Li W, Pang Y, Zhang J, Pan W, Chen M, Xia X. Ionizing radiation-induced disruption of Rela-Bclaf1-spliceosome regulatory axis in primary spermatocytes causing spermatogenesis dysfunction. Cell Commun Signal 2025; 23:58. [PMID: 39891142 PMCID: PMC11786355 DOI: 10.1186/s12964-025-02067-5] [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: 11/29/2024] [Accepted: 01/27/2025] [Indexed: 02/03/2025] Open
Abstract
INTRODUCTION Ionizing radiation (IR) poses a significant threat to male fertility by inducing substantial changes in the testis, yet the mechanisms underlying IR-induced spermatogenesis disorders remain poorly understood, necessitating the development of more effective radioprotective agents. METHODS We employed Bulk RNA-seq and single-cell RNA-seq (scRNA-seq) on Balb/c mice testes models following IR exposure to assess cellular and transcriptional alterations. Histological examination, sperm concentration and motility analysis, Western blotting (WB), and reverse transcription quantitative PCR (RT-qPCR) were used to evaluate testicular injury. The therapeutic potential of NF-κB agonists was investigated in an IR-induced spermatogenesis disorder model. RESULTS A 6 Gy IR dose induced spermatogenesis disorder and suppressed the spliceosome pathway, predominantly affecting the cell abundance of spermatogonia and primary spermatocytes. Bioinformatics analysis revealed that IR induced splicing disorders in differentiation-related genes, thereby impairing the differentiation ability of primary spermatocytes. Mechanistically, This IR-induced disruption was linked to IR-induced inhibition of NF-κB/Rela and Bclaf1 activity. Notably, NF-κB agonists were found to ameliorate this damage via upregulating Bclaf1 and spliceosome-related genes expression, thereby normalizing splicing patterns and rescuing IR-induced spermatogenesis disorders. CONCLUSION This study reveals a novel IR-mediated Rela-Bclaf1-spliceosome regulatory axis in primary spermatocytes and propose Rela as a potential drug target for mitigating IR-induced spermatogenesis disorders. This study not only provides new insights for further research into IR-induced damage and spermatogenic disorders caused by other factors, but also offers potential therapeutic strategies for developing radioprotective agents in cancer radiotherapy.
Collapse
Affiliation(s)
- Hongjian Zhou
- Institute of Laboratory Medicine, Jinling Hospital, First School of Clinical Medicine, Nanjing University School of Medicine, Southern Medical University, Zhongshan East Road 305, Nanjing, Jiangsu, 210002, China
| | - Zhipeng Xu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Chun Jiang
- Institute of Laboratory Medicine, Jinling Hospital, First School of Clinical Medicine, Nanjing University School of Medicine, Southern Medical University, Zhongshan East Road 305, Nanjing, Jiangsu, 210002, China
| | - Qiuyue Wu
- Institute of Laboratory Medicine, Jinling Hospital, First School of Clinical Medicine, Nanjing University School of Medicine, Southern Medical University, Zhongshan East Road 305, Nanjing, Jiangsu, 210002, China
| | - Chuanyue Zhang
- Institute of Laboratory Medicine, Jinling Hospital, First School of Clinical Medicine, Nanjing University School of Medicine, Southern Medical University, Zhongshan East Road 305, Nanjing, Jiangsu, 210002, China
| | - Zhenyu Liu
- Institute of Laboratory Medicine, Jinling Hospital, First School of Clinical Medicine, Nanjing University School of Medicine, Southern Medical University, Zhongshan East Road 305, Nanjing, Jiangsu, 210002, China
| | - Xiaoxue Zhang
- Institute of Laboratory Medicine, Jinling Hospital, First School of Clinical Medicine, Nanjing University School of Medicine, Southern Medical University, Zhongshan East Road 305, Nanjing, Jiangsu, 210002, China
| | - Weiwei Li
- Institute of Laboratory Medicine, Jinling Hospital, First School of Clinical Medicine, Nanjing University School of Medicine, Southern Medical University, Zhongshan East Road 305, Nanjing, Jiangsu, 210002, China
| | - Yujia Pang
- Institute of Laboratory Medicine, Jinling Hospital, First School of Clinical Medicine, Nanjing University School of Medicine, Southern Medical University, Zhongshan East Road 305, Nanjing, Jiangsu, 210002, China
| | - Jing Zhang
- Institute of Laboratory Medicine, Jinling Hospital, First School of Clinical Medicine, Nanjing University School of Medicine, Southern Medical University, Zhongshan East Road 305, Nanjing, Jiangsu, 210002, China
| | - Wenju Pan
- Institute of Laboratory Medicine, Jinling Hospital, First School of Clinical Medicine, Nanjing University School of Medicine, Southern Medical University, Zhongshan East Road 305, Nanjing, Jiangsu, 210002, China
| | - Min Chen
- Institute of Laboratory Medicine, Jinling Hospital, First School of Clinical Medicine, Nanjing University School of Medicine, Southern Medical University, Zhongshan East Road 305, Nanjing, Jiangsu, 210002, China
| | - Xinyi Xia
- Institute of Laboratory Medicine, Jinling Hospital, First School of Clinical Medicine, Nanjing University School of Medicine, Southern Medical University, Zhongshan East Road 305, Nanjing, Jiangsu, 210002, China.
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu, 210093, China.
| |
Collapse
|
9
|
Gupta J, Almulla AF, Jalil AT, Jasim NY, Aminov Z, Alsaikhan F, Ramaiah P, Chinnasamy L, Jawhar ZH. Melatonin in Chemo/Radiation Therapy; Implications for Normal Tissues Sparing and Tumor Suppression: An Updated Review. Curr Med Chem 2025; 32:511-538. [PMID: 37916636 DOI: 10.2174/0109298673262122231011172100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/20/2023] [Accepted: 09/01/2023] [Indexed: 11/03/2023]
Abstract
Resistance to therapy and the toxicity of normal tissue are the major problems for efficacy associated with chemotherapy and radiotherapy. Drug resistance is responsible for most cases of mortality associated with cancer. Furthermore, their side effects can decrease the quality of life for surviving patients. An enhancement in the tumor response to therapy and alleviation of toxic effects remain unsolved challenges. One of the interesting topics is the administration of agents with low toxicity to protect normal tissues and/or sensitize cancers to chemo/radiotherapy. Melatonin is a natural body hormone that is known as a multitasking molecule. Although it has antioxidant properties, a large number of experiments have uncovered interesting effects of melatonin that can increase the therapeutic efficacy of chemo/radiation therapy. Melatonin can enhance anticancer therapy efficacy through various mechanisms, cells such as the immune system, and modulation of cell cycle and death pathways, tumor suppressor genes, and also through suppression of some drug resistance mediators. However, melatonin may protect normal tissues through the suppression of inflammation, fibrosis, and massive oxidative stress in normal cells and tissues. In this review, we will discuss the distinct effects of melatonin on both tumors and normal tissues. We review how melatonin may enhance radio/chemosensitivity of tumors while protecting normal tissues such as the lung, heart, gastrointestinal system, reproductive system, brain, liver, and kidney.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U.P., India
| | - Abbas F Almulla
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | | | | | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil, Kurdistan Region, Iraq
- Clinical Biochemistry Department, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region, Iraq
| |
Collapse
|
10
|
Li Y, He Y. Therapeutic applications of stem cell-derived exosomes in radiation-induced lung injury. Cancer Cell Int 2024; 24:403. [PMID: 39695650 DOI: 10.1186/s12935-024-03595-9] [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: 06/04/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Radiation-induced lung injury is a common complication of chest tumor radiotherapy; however, effective clinical treatments are still lacking. Stem cell-derived exosomes, which contain various signaling molecules such as proteins, lipids, and miRNAs, not only retain the tissue repair and reconstruction properties of stem cells but also offer improved stability and safety. This presents significant potential for treating radiation-induced lung injury. Nonetheless, the clinical adoption of stem cell-derived exosomes for this purpose remains limited due to scientific, practical, and regulatory challenges. In this review, we highlight the current pathology and therapies for radiation-induced lung injury, focusing on the potential applications and therapeutic mechanisms of stem cell-derived exosomes. We also discuss the limitations of existing stem cell-derived exosomes and outline future directions for exosome-based treatments for radiation-induced lung injury.
Collapse
Affiliation(s)
- Ying Li
- Department of Radiotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yan He
- Department of Radiotherapy, West China Hospital, Sichuan University, Chengdu, China.
- Department of Cancer Center, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
11
|
Grosu-Bularda A, Lita FF, Hodea FV, Bordeanu-Diaconescu EM, Cretu A, Dumitru CS, Cacior S, Marinescu BM, Lascar I, Hariga CS. Navigating the Complexities of Radiation Injuries: Therapeutic Principles and Reconstructive Strategies. J Pers Med 2024; 14:1100. [PMID: 39590592 PMCID: PMC11595796 DOI: 10.3390/jpm14111100] [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: 09/24/2024] [Revised: 10/21/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Radiation injuries, particularly those resulting from therapeutic or accidental exposure, present complex challenges for medical management. These injuries can manifest localized skin damage or extend to deeper tissues, presenting as various clinical entities that require treatment strategies, ranging from conservative management to complex surgical interventions. Radiation treatment constitutes a fundamental component of neoplastic management, with nearly two out of three oncological instances undergoing it as an element of their therapeutic strategy. The therapeutic approach to radiation injury consists of expanding prophylactic measures while maintaining the efficacy of treatment, such as conservative treatment or local debridement followed by reconstruction. The armamentarium of reconstructive methods available for plastic surgeons, from secondary healing to free tissue transfer, can be successfully applied to radiation injuries. However, the unique pathophysiological changes induced by radiation necessitate a careful and specialized approach for their application, considering the altered tissue characteristics and healing dynamics. The therapeutic strategy is guided by both the severity and progression of the injury, with the primary aim of restoring functionality and aesthetic aspects while simultaneously minimizing the risk of complications. This paper explores the various conditions encompassed by the term "radiation injury," reviews both non-surgical and surgical therapeutic strategies for managing these injuries, and highlights the unique challenges associated with treating irradiated tissues within specific oncological contexts.
Collapse
Affiliation(s)
- Andreea Grosu-Bularda
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Flavia-Francesca Lita
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
- Clinical Department Plastic Surgery and Reconstructive Microsurgery, Central Military Emergency University Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Florin-Vlad Hodea
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Eliza-Maria Bordeanu-Diaconescu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Andrei Cretu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Catalina-Stefania Dumitru
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Stefan Cacior
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Bogdan-Mihai Marinescu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinical Department Plastic Surgery and Reconstructive Microsurgery, Central Military Emergency University Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Ioan Lascar
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Cristian-Sorin Hariga
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| |
Collapse
|
12
|
Wen L, Zhang W, Hu J, Chen T, Wang Y, Lv C, Li M, Wang L, Xiao F. Luteolin target HSPB1 regulates endothelial cell ferroptosis to protect against radiation vascular injury. PLoS One 2024; 19:e0311922. [PMID: 39392831 PMCID: PMC11469493 DOI: 10.1371/journal.pone.0311922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/26/2024] [Indexed: 10/13/2024] Open
Abstract
Vascular endothelial damage due to ionizing radiation is the main pathological process of radiation injury and the main cause of damage to various organs in nuclear accidents. Ferroptosis plays an important role in ionizing radiation-induced cell death. We have previously reported that luteolin is highly resistant to ferroptosis. In the present study, body weight, microvessel count, H&E, and Masson staining results showed that luteolin rescued radial vascular injury in vivo. Cell Counting Kit 8 (CCK8), Giemsa staining clarified the anti-ferroptosis ability of luteolin with low toxicity. Malondialdehyde (MDA), superoxide dismutase (SOD), NADP+/NADPH, Fe2+ staining, dihydroethidium (DHE) and MitoTracker assays for ferroptosis-related metrics, we found that luteolin enhances human umbilical vein endothelial cells (HUVECs) antioxidant damage capacity. Drug affinity responsive target stability (DARTS), surface plasmon resonance (SPR), computer simulated docking and western blot showed that heat shock protein beta-1 (HSPB1) is one of the targets of luteolin action. Luteolin inhibits ferroptosis by promoting the protein expression of HSPB1/solute carrier family 7 member 11 (SLC7A11)/ glutathione peroxidase 4 (GPX4). In conclusion, we have preliminarily elucidated the antioxidant damage ferroptosis ability and the target of action of luteolin to provide a theoretical basis for the application of luteolin in radiation injury diseases.
Collapse
Affiliation(s)
- Li Wen
- School of Nursing, Jilin University, Changchun, P. R. China
| | - Weiyuan Zhang
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, P. R. China
- Laboratory of Molecular Diagnosis and Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, P. R. China
| | - Jia Hu
- Department of Cardiovascular, The Sixth Medical Center of Chinese PLA General Hospital, Haidian District, Beijing, China
| | - Tao Chen
- Department of Cardiovascular, The Sixth Medical Center of Chinese PLA General Hospital, Haidian District, Beijing, China
| | - Yiming Wang
- School of Nursing, Jilin University, Changchun, P. R. China
| | - Changchang Lv
- School of Nursing, Jilin University, Changchun, P. R. China
| | - Min Li
- Beijing Institute of Radiation Medicine, Beijing, P. R. China
| | - Lisheng Wang
- School of Nursing, Jilin University, Changchun, P. R. China
- Laboratory of Molecular Diagnosis and Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, P. R. China
| | - Fengjun Xiao
- Beijing Institute of Radiation Medicine, Beijing, P. R. China
| |
Collapse
|
13
|
Paz C, Glassey A, Frick A, Sattar S, Zaorsky NG, Blitzer GC, Kimple RJ. Cancer therapy-related salivary dysfunction. J Clin Invest 2024; 134:e182661. [PMID: 39225092 PMCID: PMC11364403 DOI: 10.1172/jci182661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Salivary gland dysfunction is a common side effect of cancer treatments. Salivary function plays key roles in critical daily activities. Consequently, changes in salivary function can profoundly impair quality of life for cancer patients. We discuss salivary gland anatomy and physiology to understand how anticancer therapies such as chemotherapy, bone marrow transplantation, immunotherapy, and radiation therapy impair salivary function. We discuss approaches to quantify xerostomia in the clinic, including the advantages and limitations of validated quality-of-life instruments and approaches to directly measuring salivary function. Current and emerging approaches to treat cancer therapy-induced dry mouth are presented using radiation-induced salivary dysfunction as a model. Limitations of current sialagogues and salivary analogues are presented. Emerging approaches, including cellular and gene therapy and novel pharmacologic approaches, are described.
Collapse
Affiliation(s)
- Cristina Paz
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Annemarie Glassey
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Abigail Frick
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Sarah Sattar
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Nicholas G. Zaorsky
- University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
- Case Western Reserve University, Cleveland, Ohio, USA
| | - Grace C. Blitzer
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Randall J. Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
14
|
Wallwork RS, Page BR, Wigley FM, Hummers LK, Paik JJ, McMahan Z, Domsic RT, Shah AA. The Impact of Radiation Therapy in Patients with Systemic Sclerosis and Head and Neck Cancer. Pract Radiat Oncol 2024; 14:e334-e343. [PMID: 38704024 PMCID: PMC11983331 DOI: 10.1016/j.prro.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/06/2024]
Abstract
OBJECTIVE Systemic sclerosis (SSc) is considered a relative, or in some cases, absolute contraindication for radiation therapy for various cancers; however, radiation is the standard of care and the best option for tumor control for locally advanced head and neck (H&N) cancer. We present a case series to document postradiation outcomes in patients with SSc and H&N cancer. METHODS Patients with SSc and H&N cancer treated with radiation were identified from the Johns Hopkins Scleroderma Center and the University of Pittsburgh Scleroderma Center research registries. Through chart review, we identified whether patients developed predetermined acute and late side effects or changes in SSc activity from radiation. We further describe therapies used to prevent and treat radiation-induced fibrosis. RESULTS Thirteen patients with SSc who received radiation therapy for H&N cancer were included. Five-year survival was 54%. Nine patients (69%) developed local radiation-induced skin thickening, and 7 (54%) developed reduced neck range of motion. Two patients required long-term percutaneous endoscopic gastrostomy use due to radiation therapy complications. No patients required respiratory support related to radiation therapy. Regarding SSc disease activity among the patients with established SSc before radiation therapy, none experienced interstitial lung disease progression in the postradiation period. After radiation, one patient had worsening skin disease outside the radiation field; however, this patient was within the first year of SSc, when progressive skin disease is expected. Treatment strategies to prevent radiation fibrosis included pentoxifylline, amifostine, and vitamin E, while intravenous immunoglobulin (IVIG) was used to treat it. CONCLUSION Although some patients with SSc who received radiation for H&N cancer developed localized skin thickening and reduced neck range of motion, systemic flares of SSc were uncommon. This observational study provides evidence to support the use of radiation therapy for H&N cancer in patients with SSc when radiation is the best treatment option.
Collapse
Affiliation(s)
- Rachel S Wallwork
- Department of Medicine, Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Brandi R Page
- Department of Radiation Oncology-National Capitol Region, Johns Hopkins Medicine, Bethesda, Maryland
| | - Fredrick M Wigley
- Department of Medicine, Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Laura K Hummers
- Department of Medicine, Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Julie J Paik
- Department of Medicine, Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Zsuzsanna McMahan
- Department of Medicine, Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Robyn T Domsic
- Department of Medicine/Rheumatology, University of Pittsburgh School of Medicine. Pittsburgh, Pennsylvania
| | - Ami A Shah
- Department of Medicine, Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
15
|
Perrucci E, Macchia G, Cerrotta A, Andrulli AD, Autorino R, Barcellini A, Campitelli M, Corrao G, Costantini S, De Sanctis V, Di Muzio J, Epifani V, Ferrazza P, Fodor A, Garibaldi E, Laliscia C, Lazzari R, Magri E, Mariucci C, Pace MP, Pappalardi B, Pastorino A, Piccolo F, Scoglio C, Surgo A, Titone F, Tortoreto F, De Felice F, Aristei C. Prevention and management of radiotherapy-related toxicities in gynecological malignancies. Position paper on behalf of AIRO (Italian Association of Radiotherapy and Clinical Oncology). LA RADIOLOGIA MEDICA 2024; 129:1329-1351. [PMID: 39198369 PMCID: PMC11379782 DOI: 10.1007/s11547-024-01844-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/01/2024] [Indexed: 09/01/2024]
Abstract
Multi-modal therapies for gynecological cancers management may determine a wide range of side effects which depend on therapy-related factors and patient characteristics and comorbidities. Curative or adjuvant pelvic radiotherapy is linked with acute and late toxicity due to irradiation of organs at risk, as small and large bowel, rectum, bladder, pelvic bone, vagina and bone marrow. Successful toxicity management varies with its severity, Radiation Centre practice and experience and skills of radiation oncologists. This position paper was designed by the Italian Association of Radiation and Clinical Oncology Gynecology Study Group to provide radiation oncologists with evidence-based strategies to prevent and manage acute and late toxicities and follow-up recommendations for gynecological cancer patients submitted radiotherapy. Six workgroups of radiation oncologists with over 5 years of experience in gynecologic cancers were setup to investigate radiotherapy-related toxicities. For each topic, PubMed database was searched for relevant English language papers from January 2005 to December 2022. Titles and abstracts of results were checked to verify suitability for the document. Reference lists of selected studies and review papers were added if pertinent. Data on incidence, etiopathogenesis, prevention, treatment and follow-up of acute and late side effects for each organ at risk are presented and discussed.
Collapse
Affiliation(s)
| | - Gabriella Macchia
- Radiation Oncology Unit, Responsible Research Hospital, Campobasso, Italy
| | - Annamaria Cerrotta
- Radiotherapy Unit, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Rosa Autorino
- UOC di Radioterapia, Dipartimento di Scienze Radiologiche, Radioterapiche ed Ematologiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Amelia Barcellini
- Radiation Oncology Unit, Clinical Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
- Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy
| | - Maura Campitelli
- UOC di Radioterapia, Dipartimento di Scienze Radiologiche, Radioterapiche ed Ematologiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giulia Corrao
- Department of Radiotherapy, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Sara Costantini
- Radiation Oncology Centre, Santa Maria Hospital, Terni, Italy
| | - Vitaliana De Sanctis
- Radiotherapy Oncology, Department of Medicine, Surgery and Translational Medicine, St. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Jacopo Di Muzio
- Dipartimento Di Oncologia P.O. S. Anna - SS Radioterapia, A.O.U "Città Della Salute E Della Scienza", Turin, Italy
| | - Valeria Epifani
- Radiation Oncology Section, University of Perugia, Perugia, Italy.
| | | | - Andrei Fodor
- Department of Radiation Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Garibaldi
- Department of Radiotherapy, Ospedale Regionale Parini-AUSL Valle d'Aosta, Aosta, Italy
| | - Concetta Laliscia
- Department of Translational Medicine, Radiation Oncology Division, University of Pisa, Pisa, Italy
| | - Roberta Lazzari
- Department of Radiotherapy, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Elena Magri
- Department of Radiotherapy, Santa Chiara Hospital, Trento, Italy
| | - Cristina Mariucci
- Radiotherapy Department, Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy
| | - Maria Paola Pace
- UOC Radioterapia Oncologica, Ospedale Generale Provinciale di Macerata, AST Macerata, Italy
| | - Brigida Pappalardi
- Radiotherapy Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Federica Piccolo
- Radiotherapy Unit, Ospedale di Circolo Fondazione Macchi, Varese, Italy
| | - Claudio Scoglio
- Radiotherapy Unit, Ospedale Maggiore di Trieste, Trieste, Italy
| | - Alessia Surgo
- Department of Radiation Oncology, General Regional Hospital "F. Miulli", Acquaviva delle Fonti, Bari, Italy
| | - Francesca Titone
- Radiation Oncology Unit, Department of Oncology, "Santa Maria della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | | | - Francesca De Felice
- Department of Radiotherapy, Policlinico Umberto I, Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, University of Perugia and Perugia General Hospital, Perugia, Italy
| |
Collapse
|
16
|
Yang Y, Yang J, Zhu J, Chen X, Zhou L, Ma W, Lin Y. A DNA tetrahedron-based nanosuit for efficient delivery of amifostine and multi-organ radioprotection. Bioact Mater 2024; 39:191-205. [PMID: 38808157 PMCID: PMC11131065 DOI: 10.1016/j.bioactmat.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/30/2024] Open
Abstract
Unnecessary exposure to ionizing radiation (IR) often causes acute and chronic oxidative damages to normal cells and organs, leading to serious physiological and even life-threatening consequences. Amifostine (AMF) is a validated radioprotectant extensively applied in radiation and chemotherapy medicine, but the short half-life limits its bioavailability and clinical applications, remaining as a great challenge to be addressed. DNA-assembled nanostructures especially the tetrahedral framework nucleic acids (tFNAs) are promising nanocarriers with preeminent biosafety, low biotoxicity, and high transport efficiency. The tFNAs also have a relative long-term maintenance for structural stability and excellent endocytosis capacity. We therefore synthesized a tFNA-based delivery system of AMF for multi-organ radioprotection (tFNAs@AMF, also termed nanosuit). By establishing the mice models of accidental total body irradiation (TBI) and radiotherapy model of Lewis lung cancer, we demonstrated that the nanosuit could shield normal cells from IR-induced DNA damage by regulating the molecular biomarkers of anti-apoptosis and anti-oxidative stress. In the accidental total body irradiation (TBI) mice model, the nanosuit pretreated mice exhibited satisfactory alteration of superoxide dismutase (SOD) activities and malondialdehyde (MDA) contents, and functional recovery of hematopoietic system, reducing IR-induced pathological damages of multi-organ and safeguarding mice from lethal radiation. More importantly, the nanosuit showed a selective radioprotection of the normal organs without interferences of tumor control in the radiotherapy model of Lewis lung cancer. Based on a conveniently available DNA tetrahedron-based nanocarrier, this work presents a high-efficiency delivery system of AMF with the prolonged half-life and enhanced radioprotection for multi-organs. Such nanosuit pioneers a promising strategy with great clinical translation potential for radioactivity protection.
Collapse
Affiliation(s)
- Yuting Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, PR China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, PR China
| | - Jinlong Yang
- Department of Neurosurgery of West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Jianwei Zhu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Xingyu Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, PR China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, PR China
| | - Li Zhou
- Histology and Imaging Platform, Core Facility of West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, PR China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, PR China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, PR China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, PR China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| |
Collapse
|
17
|
Chen YS, Jin E, Day PJ. Use of Drug Sensitisers to Improve Therapeutic Index in Cancer. Pharmaceutics 2024; 16:928. [PMID: 39065625 PMCID: PMC11279903 DOI: 10.3390/pharmaceutics16070928] [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: 05/30/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
The clinical management of malignant tumours is challenging, often leading to severe adverse effects and death. Drug resistance (DR) antagonises the effectiveness of treatments, and increasing drug dosage can worsen the therapeutic index (TI). Current efforts to overcome DR predominantly involve the use of drug combinations, including applying multiple anti-cancerous drugs, employing drug sensitisers, which are chemical agents that enhance pharmacokinetics (PK), including the targeting of cellular pathways and regulating pertinent membrane transporters. While combining multiple compounds may lead to drug-drug interactions (DDI) or polypharmacy effect, the use of drug sensitisers permits rapid attainment of effective treatment dosages at the disease site to prevent early DR and minimise side effects and will reduce the chance of DDI as lower drug doses are required. This review highlights the essential use of TI in evaluating drug dosage for cancer treatment and discusses the lack of a unified standard for TI within the field. Commonly used benefit-risk assessment criteria are summarised, and the critical exploration of the current use of TI in the pharmaceutical industrial sector is included. Specifically, this review leads to the discussion of drug sensitisers to facilitate improved ratios of effective dose to toxic dose directly in humans. The combination of drug and sensitiser molecules might see additional benefits to rekindle those drugs that failed late-stage clinical trials by the removal of detrimental off-target activities through the use of lower drug doses. Drug combinations and employing drug sensitisers are potential means to combat DR. The evolution of drug combinations and polypharmacy on TI are reviewed. Notably, the novel binary weapon approach is introduced as a new opportunity to improve TI. This review emphasises the urgent need for a criterion to systematically evaluate drug safety and efficiency for practical implementation in the field.
Collapse
Affiliation(s)
- Yu-Shan Chen
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (Y.-S.C.); (E.J.)
| | - Enhui Jin
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (Y.-S.C.); (E.J.)
| | - Philip J. Day
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK; (Y.-S.C.); (E.J.)
- Department of Medicine, University of Cape Town, Cape Town 7925, South Africa
| |
Collapse
|
18
|
Zhou Y, Li K, Adelson DL. An unmet need for pharmacology: Treatments for radiation-induced gastrointestinal mucositis. Biomed Pharmacother 2024; 175:116767. [PMID: 38781863 DOI: 10.1016/j.biopha.2024.116767] [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: 03/04/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Gastrointestinal mucositis (GIM) continues to be a significant issue in the management of abdominal cancer radiation treatments and chemotherapy, causing significant patient discomfort and therapy interruption or even cessation. This review will first focus on radiotherapy induced GIM, providing an understanding of its clinical landscape. Subsequently, the aetiology of GIM will be reviewed, highlighting diverse contributing factors. The cellular and tissue damage and associated molecular responses in GIM will be summarised in the context of the underlying complex biological processes. Finally, available drugs and pharmaceutical therapies will be evaluated, underscoring their insufficiency, and highlighting the need for further research and innovation. This review will emphasize the urgent need for improved pharmacologic therapeutics for GIM, which is a key research priority in oncology.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia; Zhendong Australia China Centre for Molecular Chinese Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia.
| | - Kun Li
- Beijing Zhendong Guangming Pharmaceutical Research Institute, Beijing 100120, China.
| | - David L Adelson
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia; Zhendong Australia China Centre for Molecular Chinese Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia.
| |
Collapse
|
19
|
Pillai S, Munguia-Lopez JG, Tran SD. Bioengineered Salivary Gland Microtissues─A Review of 3D Cellular Models and their Applications. ACS APPLIED BIO MATERIALS 2024; 7:2620-2636. [PMID: 38591955 DOI: 10.1021/acsabm.4c00028] [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] [Indexed: 04/10/2024]
Abstract
Salivary glands (SGs) play a vital role in maintaining oral health through the production and release of saliva. Injury to SGs can lead to gland hypofunction and a decrease in saliva secretion manifesting as xerostomia. While symptomatic treatments for xerostomia exist, effective permanent solutions are still lacking, emphasizing the need for innovative approaches. Significant progress has been made in the field of three-dimensional (3D) SG bioengineering for applications in gland regeneration. This has been achieved through a major focus on cell culture techniques, including soluble cues and biomaterial components of the 3D niche. Cells derived from both adult and embryonic SGs have highlighted key in vitro characteristics of SG 3D models. While still in its first decade of exploration, SG spheroids and organoids have so far served as crucial tools to study SG pathophysiology. This review, based on a literature search over the past decade, covers the importance of SG cell types in the realm of their isolation, sourcing, and culture conditions that modulate the 3D microenvironment. We discuss different biomaterials employed for SG culture and the current advances made in bioengineering SG models using them. The success of these 3D cellular models are further evaluated in the context of their applications in organ transplantation and in vitro disease modeling.
Collapse
Affiliation(s)
- Sangeeth Pillai
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, 3640 Rue University, Montreal, QC H3A 0C7, Canada
| | - Jose G Munguia-Lopez
- Department of Mining and Materials Engineering, McGill University, 3610 University Street, Montreal, QC H3A 0C5, Canada
| | - Simon D Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, 3640 Rue University, Montreal, QC H3A 0C7, Canada
| |
Collapse
|
20
|
Liu C, Wang L, Zhou Y, Xia W, Wang Z, Kuang L, Hua D. Biogenic crocetin-crosslinked chitosan nanoparticles with high stability and drug loading for efficient radioprotection. Int J Biol Macromol 2024; 265:130756. [PMID: 38462118 DOI: 10.1016/j.ijbiomac.2024.130756] [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: 12/12/2023] [Revised: 02/20/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024]
Abstract
The risk of radiation exposure increases with the development of nuclear energy and technology, and radiation protection receives more and more attention from public health and safety. However, the numerous adverse effects and low drug utilization limit the practical applications of radioprotective agents. In this study, we developed a biogenic crocetin-crosslinked chitosan nanoparticle with high stability and drug loading for efficient radioprotection. In detail, the nanoparticles were prepared using the natural antioxidant crocetin as a cross-linking reagent in amidation reactions of chitosan and mPEG-COOH. The nanoparticles exhibit a quick scavenging ability for common reactive oxygen species and reactive nitrogen in vitro. Meanwhile, cellular experiments demonstrate the good biocompatibility of the nanoparticles and the alleviation of radiation damage by scavenging reactive oxygen species, reducing apoptosis, and inhibiting DNA damage, etc. Importantly, the nanoparticles are effective in mitigating oxidative damage in major organs and maintaining peripheral blood cell content. In addition, they perform better radioprotective properties than free drug due to the significant extension of the blood half-life of crocetin in vivo from 10 min to 5 h. This work proposes a drug-crosslinking strategy for the design of a highly efficient radioprotective agent, which exhibits a promising prospect in the fields of nuclear emergency and public health.
Collapse
Affiliation(s)
- Chang Liu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Lu Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yi Zhou
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Wanyi Xia
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ziyu Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Liangju Kuang
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye & Ear, Harvard Medical School, Boston, MA 02114, USA.
| | - Daoben Hua
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| |
Collapse
|
21
|
Ge YX, Zhang Z, Yan JY, Ma ZC, Wang YG, Xiao CR, Zhuang XM, Gao Y. Prediction of Human Pharmacokinetics of E0703, a Novel Radioprotective Agent, Using Physiologically Based Pharmacokinetic Modeling and an Interspecies Extrapolation Approach. Int J Mol Sci 2024; 25:3047. [PMID: 38474292 DOI: 10.3390/ijms25053047] [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: 02/05/2024] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
E0703, a new steroidal compound optimized from estradiol, significantly increased cell proliferation and the survival rate of KM mice and beagles after ionizing radiation. In this study, we characterize its preclinical pharmacokinetics (PK) and predict its human PK using a physiologically based pharmacokinetic (PBPK) model. The preclinical PK of E0703 was studied in mice and Rhesus monkeys. Asian human clearance (CL) values for E0703 were predicted from various allometric methods. The human PK profiles of E0703 (30 mg) were predicted by the PBPK model in Gastro Plus software 9.8 (SimulationsPlus, Lancaster, CA, USA). Furthermore, tissue distribution and the human PK profiles of different administration dosages and forms were predicted. The 0.002 L/h of CL and 0.005 L of Vss in mice were calculated and optimized from observed PK data. The plasma exposure of E0703 was availably predicted by the CL using the simple allometry (SA) method. The plasma concentration-time profiles of other dosages (20 and 40 mg) and two oral administrations (30 mg) were well-fitted to the observed values. In addition, the PK profile of target organs for E0703 exhibited a higher peak concentration (Cmax) and AUC than plasma. The developed E0703-PBPK model, which is precisely applicable to multiple species, benefits from further clinical development to predict PK in humans.
Collapse
Affiliation(s)
- Yun-Xuan Ge
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zhuo Zhang
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jia-Yi Yan
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zeng-Chun Ma
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yu-Guang Wang
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Cheng-Rong Xiao
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiao-Mei Zhuang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yue Gao
- College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
22
|
Yang Y, Chen Y, Tang H, Zhang Z, Zhou X, Xu W. DTTZ suppresses ferroptosis and reverses mitochondrial dysfunction in normal tissues affected by chemotherapy. Biomed Pharmacother 2024; 172:116227. [PMID: 38335570 DOI: 10.1016/j.biopha.2024.116227] [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: 11/10/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Conventional antineoplastic therapies cause severe normal tissue damage and existing cytoprotectants with acute toxicities or potential tumor protection limit their clinical application. We evaluated the selective cytoprotection of 2,2-dimethylthiazolidine hydrochloride in this study, which could protect normal tissue toxicity without interfering antineoplastic therapies. By using diverse cell lines and A549 xenograft model, we discovered a synthetic aminothiol 2,2-dimethylthiazolidine hydrochloride selectively diminished normal cellular ferroptosis via SystemXc-/Glutathione Peroxidase 4 pathway upon antineoplastic therapies without interfering the anticancer efficacy. We revealed the malignant and non-malignant tissues presenting different energy metabolism patterns. And cisplatin induces disparate replicative stress, contributing to the distinguishable cytoprotection of 2,2-dimethylthiazolidine in normal and tumor cells. The compound pre-application could mitigate cisplatin-induced normal cellular mitochondrial oxidative phosphorylation (OXPHOS) dysfunction. Pharmacologic ablation of mitochondria reversed 2,2-dimethylthiazolidine chemoprotection against cisplatin in the normal cell line. Combined, these results provide a potential therapeutic adjuvant to selectively diminish normal tissue damages retaining antineoplastic efficacy.
Collapse
Affiliation(s)
- Yuwei Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Yuanfang Chen
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Haikang Tang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Ziqi Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Xiaoliang Zhou
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| | - Wenqing Xu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| |
Collapse
|
23
|
Guo F, Xu F, Li S, Zhang Y, Lv D, Zheng L, Gan Y, Zhou M, Zhao K, Xu S, Wu B, Deng Z, Fu P. Amifostine ameliorates bleomycin-induced murine pulmonary fibrosis via NAD +/SIRT1/AMPK pathway-mediated effects on mitochondrial function and cellular metabolism. Eur J Med Res 2024; 29:68. [PMID: 38245795 PMCID: PMC10799491 DOI: 10.1186/s40001-023-01623-4] [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: 01/27/2023] [Accepted: 12/25/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a devastating chronic lung disease characterized by irreversible scarring of the lung parenchyma. Despite various interventions aimed at mitigating several different molecular aspects of the disease, only two drugs with limited clinical efficacy have so far been approved for IPF therapy. OBJECTIVE We investigated the therapeutic efficacy of amifostine, a detoxifying drug clinically used for radiation-caused cytotoxicity, in bleomycin-induced murine pulmonary fibrosis. METHODS C57BL6/J mice were intratracheally instilled with 3 U/kg of bleomycin. Three doses of amifostine (WR-2721, 200 mg/kg) were administered intraperitoneally on days 1, 3, and 5 after the bleomycin challenge. Bronchoalveolar lavage fluid (BALF) was collected on day 7 and day 21 for the assessment of lung inflammation, metabolites, and fibrotic injury. Human fibroblasts were treated in vitro with transforming growth factor beta 1 (TGF-β1), followed by amifostine (WR-1065, 1-4 µg/mL) treatment. The effects of TGF-β1 and amifostine on the mitochondrial production of reactive oxygen species (ROS) were assessed by live cell imaging of MitoSOX. Cellular metabolism was assessed by the extracellular acidification rate (ECAR), the oxygen consumption rate (OCR), and the concentrations of various energy-related metabolites as measured by mass spectrum (MS). Western blot analysis was performed to investigate the effect of amifostine on sirtuin 1 (SIRT1) and adenosine monophosphate activated kinase (AMPK). RESULTS Three doses of amifostine significantly attenuated lung inflammation and pulmonary fibrosis. Pretreatment and post-treatment of human fibroblast cells with amifostine blocked TGF-β1-induced mitochondrial ROS production and mitochondrial dysfunction in human fibroblast cells. Further, treatment of fibroblasts with TGF-β1 shifted energy metabolism away from mitochondrial oxidative phosphorylation (OXPHOS) and towards glycolysis, as observed by an altered metabolite profile including a decreased ratio of NAD + /NADH and increased lactate concentration. Treatment with amifostine significantly restored energy metabolism and activated SIRT1, which in turn activated AMPK. The activation of AMPK was required to mediate the effects of amifostine on mitochondrial homeostasis and pulmonary fibrosis. This study provides evidence that repurposing of the clinically used drug amifostine may have therapeutic applications for IPF treatment. CONCLUSION Amifostine inhibits bleomycin-induced pulmonary fibrosis by restoring mitochondrial function and cellular metabolism.
Collapse
Affiliation(s)
- Feng Guo
- Department of Biochemistry, Health Science Center, Ningbo University, Ningbo, 315041, China
- Central Laboratory of the Medical Research Center, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Feng Xu
- Central Laboratory of the Medical Research Center, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Shujuan Li
- Department of Biochemistry, Health Science Center, Ningbo University, Ningbo, 315041, China
| | - Yun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo University, Ningbo, 315041, China
| | - Dan Lv
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo University, Ningbo, 315041, China
| | - Lin Zheng
- Department of Laboratory Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yongxiong Gan
- Department of Emergency Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Miao Zhou
- Department of Biochemistry, Health Science Center, Ningbo University, Ningbo, 315041, China
| | - Keyu Zhao
- Department of Dermatology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Shuling Xu
- Department of Dermatology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Bin Wu
- Department of Pulmonary and Critical Care Medicine, South China Hospital Affiliated to Shenzhen University, Shenzhen, China
| | - Zaichun Deng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Ningbo University, Ningbo University, Ningbo, 315041, China.
| | - Panfeng Fu
- Department of Biochemistry, Health Science Center, Ningbo University, Ningbo, 315041, China.
- Central Laboratory of the Medical Research Center, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| |
Collapse
|
24
|
Ji L, Cui P, Zhou S, Qiu L, Huang H, Wang C, Wang J. Advances of Amifostine in Radiation Protection: Administration and Delivery. Mol Pharm 2023; 20:5383-5395. [PMID: 37747899 DOI: 10.1021/acs.molpharmaceut.3c00600] [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] [Indexed: 09/27/2023]
Abstract
Amifostine (AMF, also known as WR-2721) is the only approved broad-spectrum small-molecule radiation protection agent that can combat hematopoietic damage caused by ionizing radiation and is used as an antitumor adjuvant and cell protector in cancer chemotherapy and radiotherapy. Amifostine is usually injected intravenously before chemotherapy or radiotherapy and has been used in the treatment of head and neck cancer. However, the inconvenient intravenous administration and its toxic side effects such as hypotension have severely limited its further application in clinic. In order to reduce the toxic and side effects, scientists are trying to develop a variety of drug administration methods and are devoted to developing a wide application of amifostine in radiation protection. This paper reviews the research progress of amifostine for radiation protection in recent years, discusses its mechanism of action, clinical application, and other aspects, with focus on summarizing the most widely studied amifostine injection administration and drug delivery systems, and explored the correlation between various administrations and drug efficacies.
Collapse
Affiliation(s)
- Lihua Ji
- School of Pharmacy, Changzhou University, Changzhou 213164, Jiangsu, China
- School of Petroleum and Chemical Engineering, Changzhou University, Changzhou 213164, Jiangsu, China
| | - Pengfei Cui
- School of Pharmacy, Changzhou University, Changzhou 213164, Jiangsu, China
| | - Shuwen Zhou
- School of Pharmacy, Changzhou University, Changzhou 213164, Jiangsu, China
| | - Lin Qiu
- School of Pharmacy, Changzhou University, Changzhou 213164, Jiangsu, China
| | - Hai Huang
- School of Petroleum and Chemical Engineering, Changzhou University, Changzhou 213164, Jiangsu, China
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, Jiangsu, China
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, Jiangsu, China
| |
Collapse
|
25
|
Yu M, Xie W, Tang Z, Luo J, Liu Y. Radiopaque and X-ray-Responsive Nanomedicine for Preventive Therapy of Radiation-Induced Heart Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303803. [PMID: 37452441 DOI: 10.1002/smll.202303803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/03/2023] [Indexed: 07/18/2023]
Abstract
Radiation-induced heart disease (RIHD) is a common radiotherapy complication. Reducing radiation exposure and post-irradiation antioxidant therapy are promising approaches. Here, a liquid metal-based core-shell nanomedicine (LMN) composed of a gallium core and a multifunctional polymeric shell with radiopaque, X-ray shielding, and X-ray-responsive antioxidation properties for preventive therapy of RIHD is developed. The liquid metal provides radiopaque properties to enhance X-ray and computed tomography imaging and attenuate radiation to prevent primary myocardial damage. Under X-ray radiation, cleavage of the diselenide bond on the polymeric shell results in the release of LMN and controlled antioxidation. In vitro and in vivo experiments have demonstrated that LMN significantly reduces myocardial injury and impaired cardiac function, stabilizes mitochondrial function, and inhibits myocardial fibrosis. This nanomedicine with radiographic contrast, radiation shielding, and responsive features provides a new strategy for the prevention of radiation-related diseases.
Collapse
Affiliation(s)
- Mingchuan Yu
- Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Weichang Xie
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Zhibo Tang
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Jun Luo
- Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Yu Liu
- Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| |
Collapse
|
26
|
Aghajanshakeri S, Salmanmahiny A, Aghajanshakeri S, Babaei A, Alishahi F, Babayani E, Shokrzadeh M. Modulatory effect of amifostine (WR-1065) against genotoxicity and oxidative stress induced by methotrexate in human umbilical vein endothelial cells (HUVECs). Toxicol Mech Methods 2023; 33:755-765. [PMID: 37537746 DOI: 10.1080/15376516.2023.2238069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/11/2023] [Indexed: 08/05/2023]
Abstract
Amifostine is used in chemotherapy and radiotherapy as a cytoprotective adjuvant alongside DNA-binding chemotherapeutic agents. It functions by reducing free radicals and detoxifying harmful metabolites. Methotrexate, as an antimetabolite drug has been considered for treating various cancers and autoimmune diseases. However, the cytotoxic effects of methotrexate extend beyond tumor cells to crucial organs, including the heart. This study applied the HUVEC cell line as a reference in vitro model for researching the characteristics of vascular endothelium and cardiotoxicity. The current study aimed to assess amifostine's potential cytoprotective properties against methotrexate-induced cellular damage. Cytotoxicity was measured using the MTT assay. Apoptotic rates were evaluated by Annexin V-FITC/PI staining via flow cytometry. The genoprotective effect of amifostine was determined using the comet assay. Cells were exposed to various amifostine doses (10-200 μg/mL) and methotrexate (2.5 μM) in pretreatment culture condition. Methotrexate at 2.5 μM revealed cytotoxicity, apoptosis, oxidative stress and genotoxicity while highlighting amifostine's cyto/geno protective properties on HUVECs. Amifostine significantly decreased the levels of ROS and LPO while preserving the status of GSH and SOD activity. Furthermore, it inhibited genotoxicity (tail length, %DNA in tail, and tail moment) in the comet assay. Amifostine markedly attenuated methotrexate-induced apoptotic cell death (early and late apoptotic rates). These findings convey that amifostine can operate as a cytoprotectant agent.
Collapse
Affiliation(s)
- Shaghayegh Aghajanshakeri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amirhossein Salmanmahiny
- Department of Toxicology and Pharmacology, Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahin Aghajanshakeri
- Biological Oncology (Orchid Pharmed) Department, CinnaGen Pharmaceutical Company, Tehran, Iran
| | - Amirhossein Babaei
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Farhad Alishahi
- Department of Toxicology and Pharmacology, Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Erfan Babayani
- Department of Toxicology and Pharmacology, Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Shokrzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
27
|
Fu Z, Gao C, Wu T, Wang L, Li S, Zhang Y, Shi C. Peripheral neuropathy associated with monomethyl auristatin E-based antibody-drug conjugates. iScience 2023; 26:107778. [PMID: 37727735 PMCID: PMC10505985 DOI: 10.1016/j.isci.2023.107778] [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] [Indexed: 09/21/2023] Open
Abstract
Since the successful approval of gemtuzumab ozogamicin, antibody-drug conjugates (ADCs) have emerged as a pivotal category of targeted therapies for cancer. Among these ADCs, the use of monomethyl auristatin E (MMAE) as a payload is prevalent in the development of ADC drugs, which has significantly improved overall therapeutic efficacy against various malignancies. However, increasing clinical observations have raised concerns regarding the potential nervous system toxicity associated with MMAE-based ADCs. Specifically, a higher incidence of peripheral neuropathy has been reported in ADCs incorporating MMAE as payloads. Considering the increasing global use of MMAE-based ADCs, it is imperative to provide an inclusive overview of diagnostic and management strategies for this adverse event. In this review, we examine current information and what future research directions are required to better understand and manage this type of clinical challenge.
Collapse
Affiliation(s)
- Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| | - Chen Gao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| | - Tingting Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| | - Lulu Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| | - Shijun Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| | - Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| |
Collapse
|
28
|
Kumari P, Kumar R, Singh D, Kumar R. N-acetyl-L-tryptophan (NAT) provides protection to intestinal epithelial cells (IEC-6) against radiation-induced apoptosis via modulation of oxidative stress and mitochondrial membrane integrity. Mol Biol Rep 2023; 50:6381-6397. [PMID: 37322322 DOI: 10.1007/s11033-023-08579-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/02/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Ionizing radiation generates oxidative stress in biological systems via inducing free radicals. Gastro-intestinal system has been known for its high radiosensitivity. Therefore, to develop an effective radiation countermeasure for gastrointestinal system, N-acetyl L-tryptophan was evaluated for its radioprotective efficacy using intestinal epithelial cells-6 (IEC-6) cells as the experimental model. METHODS AND RESULTS Cellular metabolic and lysosomal activity of L-NAT and L-NAT treated irradiated IEC-6 cells were assessed by MTT and NRU staining, respectively. ROS and mitochondrial superoxide levels along with mitochondrial disruption were detected using specific fluorescent probes. Endogenous antioxidants (CAT, SOD, GST, GPx) activities were determined using calorimetric assay. Apoptosis and DNA damage were assessed using flow cytometery and Comet assay, respectively. Results of the study were demonstrated that L-NAT pre-treatment (- 1 h) to irradiated IEC-6 cells significantly contribute to ensuring 84.36% to 87.68% (p < 0.0001) survival at 0.1 μg/mL concentration against LD50 radiation dose (LD50; 20 Gy). Similar level of radioprotection was observed with a clonogenic assay against γ radiation (LD50; 5 Gy). L-NAT was found to provide radioprotection by neutralizing radiation-induced oxidative stress, enhancing antioxidant enzymes (CAT, SOD, GST, and GPx), and protecting DNA from radiation-induced damage. Further, significant restoration of mitochondrial membrane integrity along with apoptosis inhibition was observed with irradiated IEC-6 cells upon L-NAT pretreatment.
Collapse
Affiliation(s)
- Pratibha Kumari
- Radiation Biotechnology Group, Division of Chemical, Biological, Radiological & Nuclear Defence (CBRN), Defence Research and Development Organization (DRDO), Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Ravi Kumar
- Radiation Biotechnology Group, Division of Chemical, Biological, Radiological & Nuclear Defence (CBRN), Defence Research and Development Organization (DRDO), Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Darshana Singh
- Radiation Biotechnology Group, Division of Chemical, Biological, Radiological & Nuclear Defence (CBRN), Defence Research and Development Organization (DRDO), Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Raj Kumar
- Radiation Biotechnology Group, Division of Chemical, Biological, Radiological & Nuclear Defence (CBRN), Defence Research and Development Organization (DRDO), Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
29
|
Walker DM, Lazarova TI, Riesinger SW, Poirier MC, Messier T, Cunniff B, Walker VE. WR1065 conjugated to thiol-PEG polymers as novel anticancer prodrugs: broad spectrum efficacy, synergism, and drug resistance reversal. Front Oncol 2023; 13:1212604. [PMID: 37576902 PMCID: PMC10419174 DOI: 10.3389/fonc.2023.1212604] [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: 04/26/2023] [Accepted: 06/27/2023] [Indexed: 08/15/2023] Open
Abstract
The lack of anticancer agents that overcome innate/acquired drug resistance is the single biggest barrier to achieving a durable complete response to cancer therapy. To address this issue, a new drug family was developed for intracellular delivery of the bioactive aminothiol WR1065 by conjugating it to discrete thiol-PEG polymers: 4-star-PEG-S-S-WR1065 (4SP65) delivers four WR1065s/molecule and m-PEG6-S-S-WR1065 (1LP65) delivers one. Infrequently, WR1065 has exhibited anticancer effects when delivered via the FDA-approved cytoprotectant amifostine, which provides one WR1065/molecule extracellularly. The relative anticancer effectiveness of 4SP65, 1LP65, and amifostine was evaluated in a panel of 15 human cancer cell lines derived from seven tissues. Additional experiments assessed the capacity of 4SP65 co-treatments to potentiate the anticancer effectiveness and overcome drug resistance to cisplatin, a chemotherapeutic, or gefitinib, a tyrosine kinase inhibitor (TKI) targeting oncogenic EGFR mutations. The CyQUANT®-NF proliferation assay was used to assess cell viability after 48-h drug treatments, with the National Cancer Institute COMPARE methodology employed to characterize dose-response metrics. In normal human epithelial cells, 4SP65 or 1LP65 enhanced or inhibited cell growth but was not cytotoxic. In cancer cell lines, 4SP65 and 1LP65 induced dose-dependent cytostasis and cytolysis achieving 99% cell death at drug concentrations of 11.2 ± 1.2 µM and 126 ± 15.8 µM, respectively. Amifostine had limited cytostatic effects in 11/14 cancer cell lines and no cytolytic effects. Binary pairs of 4SP65 plus cisplatin or gefitinib increased the efficacy of each partner drug and surmounted resistance to cytolysis by cisplatin and gefitinib in relevant cancer cell lines. 4SP65 and 1LP65 were significantly more effective against TP53-mutant than TP53-wild-type cell lines, consistent with WR1065-mediated reactivation of mutant p53. Thus, 4SP65 and 1LP65 represent a unique prodrug family for innovative applications as broad-spectrum anticancer agents that target p53 and synergize with a chemotherapeutic and an EGFR-TKI to prevent or overcome drug resistance.
Collapse
Affiliation(s)
- Dale M. Walker
- The Burlington HC Research Group, Inc., Jericho, VT, United States
| | | | | | - Miriam C. Poirier
- Carcinogen–DNA Interactions Section, Laboratory of Cellular Carcinogenesis and Tumor Promotion, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Terri Messier
- Department of Pathology and Laboratory Medicine, Redox Biology and Pathology Program, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Brian Cunniff
- Department of Pathology and Laboratory Medicine, Redox Biology and Pathology Program, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Vernon E. Walker
- Department of Pathology and Laboratory Medicine, Redox Biology and Pathology Program, Larner College of Medicine, University of Vermont, Burlington, VT, United States
| |
Collapse
|
30
|
Hariharan A, Tran SD. Localized Drug Delivery Systems: An Update on Treatment Options for Head and Neck Squamous Cell Carcinomas. Pharmaceutics 2023; 15:1844. [PMID: 37514031 PMCID: PMC10385385 DOI: 10.3390/pharmaceutics15071844] [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: 04/13/2023] [Revised: 06/12/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most common cancers in the world, with surgery, radiotherapy, chemotherapy, and immunotherapy being the primary treatment modalities. The treatment for HNSCC has evolved over time, due to which the prognosis has improved drastically. Despite the varied treatment options, major challenges persist. HNSCC chemotherapeutic and immunotherapeutic drugs are usually administered systemically, which could affect the patient's quality of life due to the associated side effects. Moreover, the systemic administration of salivary stimulating agents for the treatment of radiation-induced xerostomia is associated with toxicities. Localized drug delivery systems (LDDS) are gaining importance, as they have the potential to provide non-invasive, patient-friendly alternatives to cancer therapy with reduced dose-limiting toxicities. LDDSs involve directly delivering a drug to the tissue or organ affected by the disease. Some of the common localized routes of administration include the transdermal and transmucosal drug delivery system (DDSs). This review will attempt to explore the different treatment options using LDDSs for the treatment of HNSCC and radiotherapy-induced damage and their potential to provide a better experience for patients, as well as the obstacles that need to be addressed to render them successful.
Collapse
Affiliation(s)
- Arvind Hariharan
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada
| | - Simon D Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada
| |
Collapse
|
31
|
Affandi T, Haas A, Ohm AM, Wright GM, Black JC, Reyland ME. PKCδ regulates chromatin remodeling and DNA repair through SIRT6. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.541991. [PMID: 37292592 PMCID: PMC10245827 DOI: 10.1101/2023.05.24.541991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Protein kinase C delta (PKCδ) is a ubiquitous kinase whose function is defined in part by localization to specific cellular compartments. Nuclear PKCδ is both necessary and sufficient for IR-induced apoptosis, while inhibition of PKCδ activity provides radioprotection in vivo. How nuclear PKCδ regulates DNA-damage induced cell death is poorly understood. Here we show that PKCδ regulates histone modification, chromatin accessibility, and double stranded break (DSB) repair through a mechanism that requires SIRT6. Overexpression of PKCδ promotes genomic instability and increases DNA damage and apoptosis. Conversely, depletion of PKCδ increases DNA repair via non-homologous end joining (NHEJ) and homologous recombination (HR) as evidenced by more rapid formation of NHEJ (DNA-PK) and HR (Rad51) DNA damage foci, increased expression of repair proteins, and increased repair of NHEJ and HR fluorescent reporter constructs. Nuclease sensitivity indicates that PKCδ depletion is associated with more open chromatin, while overexpression of PKCδ reduces chromatin accessibility. Epiproteome analysis revealed that PKCδ depletion increases chromatin associated H3K36me2, and reduces ribosylation of KDM2A and chromatin bound KDM2A. We identify SIRT6 as a downstream mediator of PKCδ. PKCδ-depleted cells have increased expression of SIRT6, and depletion of SIRT6 reverses the changes in chromatin accessibility, histone modification and NHEJ and HR DNA repair seen with PKCδ-depletion. Furthermore, depletion of SIRT6 reverses radioprotection in PKCδ-depleted cells. Our studies describe a novel pathway whereby PKCδ orchestrates SIRT6-dependent changes in chromatin accessibility to increase DNA repair, and define a mechanism for regulation of radiation-induced apoptosis by PKCδ.
Collapse
Affiliation(s)
- Trisiani Affandi
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ami Haas
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angela M. Ohm
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Gregory M. Wright
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Joshua C. Black
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mary E. Reyland
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
32
|
Prades-Sagarra È, Yaromina A, Dubois LJ. Polyphenols as Potential Protectors against Radiation-Induced Adverse Effects in Patients with Thoracic Cancer. Cancers (Basel) 2023; 15:cancers15092412. [PMID: 37173877 PMCID: PMC10177176 DOI: 10.3390/cancers15092412] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Radiotherapy is one of the standard treatment approaches used against thoracic cancers, occasionally combined with chemotherapy, immunotherapy and molecular targeted therapy. However, these cancers are often not highly sensitive to standard of care treatments, making the use of high dose radiotherapy necessary, which is linked with high rates of radiation-induced adverse effects in healthy tissues of the thorax. These tissues remain therefore dose-limiting factors in radiation oncology despite recent technological advances in treatment planning and delivery of irradiation. Polyphenols are metabolites found in plants that have been suggested to improve the therapeutic window by sensitizing the tumor to radiotherapy, while simultaneously protecting normal cells from therapy-induced damage by preventing DNA damage, as well as having anti-oxidant, anti-inflammatory or immunomodulatory properties. This review focuses on the radioprotective effect of polyphenols and the molecular mechanisms underlying these effects in the normal tissue, especially in the lung, heart and esophagus.
Collapse
Affiliation(s)
- Èlia Prades-Sagarra
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
33
|
Liu D, Zhuang B, Wei M, Yuan T, Li J, Deng P, Du L, Yuan B, Jin Y. Oral konjac glucomannan for prevention of ionizing radiation-induced injury by regulating gut microbiota and increasing short chain fatty acids. Int J Biol Macromol 2023; 240:124402. [PMID: 37044326 DOI: 10.1016/j.ijbiomac.2023.124402] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/21/2023] [Accepted: 04/06/2023] [Indexed: 04/14/2023]
Abstract
Ionizing radiation-induced injury commonly happens in radiotherapy, leading to damages of the hematopoietic and gastrointestinal systems. Radioprotective medications are mainly applied in hospitals, although only injections are available and their gut protection is limited. Here, oral konjac glucomannan (KGM), a natural macromolecule and soluble dietary fiber, was used against ionizing radiation-induced injury. The mice were fed with KGM (0.4 g/kg) for 3 days or injected with a clinical medication amifostine before 6.5 Gy γ-ray whole body irradiation (WBI) or 13 Gy whole abdominal irradiation (WAI). In the WBI experiments, KGM improved blood cell recovery and bone marrow cell proliferation in the femur and spleen, though its effect was weaker than or similar to that of amifostine. In the WBI experiments, the gut protection of KGM was similar to or a little better than that of amifostine, involving regenerated crypts numbers, villus length, and gut permeability. Moreover, KGM remarkably enhanced the survival rates of WBI and WAI mice, consistent with amifostine. KGM, as a prebiotic, enhanced gut microbiota abundance, probiotic numbers, and short chain fatty acid production, maintaining gut homeostasis. Moreover, KGM inhibited the apoptosis of irradiated human intestinal epithelial cells. KGM is a promising natural macromolecule against ionizing radiation-induced injury.
Collapse
Affiliation(s)
- Dongdong Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bo Zhuang
- Department of Chemical Defense, Institute of NBC Defense, Beijing 102205, China
| | - Meng Wei
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Tianyu Yuan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jian Li
- Department of Chemical Defense, Institute of NBC Defense, Beijing 102205, China
| | - Pei Deng
- Department of Gastroenterology, Second Clinical Medical College of Beijing University of Chinese Medicine (Dongfang Hospital), Beijing 100078, China
| | - Lina Du
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bochuan Yuan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
34
|
Bruno JG, Sivils JC, Mohan S, Natarajan M. Alpha-thiol deoxynucleotide triphosphates (S-dNTPs) as radioprotective agents: A novel approach. Biochem Biophys Res Commun 2023; 660:6-12. [PMID: 37058844 DOI: 10.1016/j.bbrc.2023.03.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 04/09/2023]
Abstract
In this study, the ability of a mixture of four different alpha-thiol deoxynucleotide triphosphates (S-dNTPs) each at a concentration of 10μM when incorporated into the genomic DNA of proliferating human HL-60 and Mono-Mac-6 (MM-6) cells in vitro to provide protection from 2, 5, and 10 Gy of gamma radiation was investigated. Incorporation of the four different S-dNTPs into nuclear DNA at 10 μM concentration for five days was validated by agarose gel electrophoretic band shift analysis. S-dNTP-treated genomic DNA reacted with BODIPY-iodoacetamide demonstrated a band shift to higher molecular weight to confirm the presence of sulfur moieties in the resultant phosphorothioate DNA backbones. No overt signs of toxicity or obvious morphologic cellular differentiation were noted in the presence of 10 μM S-dNTPs even after 8 days in culture. Significantly reduced radiation-induced persistent DNA damage measured at 24 and 48 h post-exposure by γ-H2AX histone phosphorylation using FACS analysis in S-dNTP incorporated HL-60 and MM6 cells indicated protection against radiation-induced direct and indirect DNA damage. Statistically significant protection by S-dNTPs was noted at the cellular level by CellEvent™ Caspase-3/7 assay, which assess the extent of apoptotic events, and by trypan blue dye exclusion to assed cell viability. The results appear to support an innocuous antioxidant thiol radioprotective effect built into genomic DNA backbones as the last line of defense against ionizing radiation and free radical-induced DNA damage.
Collapse
|
35
|
Guo J, Zhao Z, Shang Z, Tang Z, Zhu H, Zhang K. Nanodrugs with intrinsic radioprotective exertion: Turning the double-edged sword into a single-edged knife. EXPLORATION (BEIJING, CHINA) 2023; 3:20220119. [PMID: 37324033 PMCID: PMC10190950 DOI: 10.1002/exp.20220119] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 02/10/2023] [Indexed: 06/17/2023]
Abstract
Ionizing radiation (IR) poses a growing threat to human health, and thus ideal radioprotectors with high efficacy and low toxicity still receive widespread attention in radiation medicine. Despite significant progress made in conventional radioprotectants, high toxicity, and low bioavailability still discourage their application. Fortunately, the rapidly evolving nanomaterial technology furnishes reliable tools to address these bottlenecks, opening up the cutting-edge nano-radioprotective medicine, among which the intrinsic nano-radioprotectants characterized by high efficacy, low toxicity, and prolonged blood retention duration, represent the most extensively studied class in this area. Herein, we made the systematic review on this topic, and discussed more specific types of radioprotective nanomaterials and more general clusters of the extensive nano-radioprotectants. In this review, we mainly focused on the development, design innovations, applications, challenges, and prospects of the intrinsic antiradiation nanomedicines, and presented a comprehensive overview, in-depth analysis as well as an updated understanding of the latest advances in this topic. We hope that this review will promote the interdisciplinarity across radiation medicine and nanotechnology and stimulate further valuable studies in this promising field.
Collapse
Affiliation(s)
- Jiaming Guo
- Department of Radiation Medicine, College of Naval MedicineNaval Medical UniversityShanghaiChina
| | - Zhemeng Zhao
- Department of Radiation Medicine, College of Naval MedicineNaval Medical UniversityShanghaiChina
- National Engineering Research Center for Marine Aquaculture, Marine Science and Technology CollegeZhejiang Ocean UniversityZhoushanChina
| | - Zeng‐Fu Shang
- Department of Radiation OncologySimmons Comprehensive Cancer Center at UT Southwestern Medical CenterDallasTexasUSA
| | - Zhongmin Tang
- Department of RadiologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Huanhuan Zhu
- Central Laboratory, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiP. R. China
| | - Kun Zhang
- Central Laboratory, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiP. R. China
- National Center for International Research of Bio‐targeting TheranosticsGuangxi Medical UniversityNanningGuangxiP. R. China
- Department of Oncology, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuanP. R. China
| |
Collapse
|
36
|
Yang Q, Qin B, Hou W, Qin H, Yin F. Pathogenesis and therapy of radiation enteritis with gut microbiota. Front Pharmacol 2023; 14:1116558. [PMID: 37063268 PMCID: PMC10102376 DOI: 10.3389/fphar.2023.1116558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/07/2023] [Indexed: 04/03/2023] Open
Abstract
Radiotherapy is widely used in clinic due to its good effect for cancer treatment. But radiotherapy of malignant tumors in the abdomen and pelvis is easy to cause radiation enteritis complications. Gastrointestinal tract contains numerous microbes, most of which are mutualistic relationship with the host. Abdominal radiation results in gut microbiota dysbiosis. Microbial therapy can directly target gut microbiota to reverse microbiota dysbiosis, hence relieving intestinal inflammation. In this review, we mainly summarized pathogenesis and novel therapy of the radiation-induced intestinal injury with gut microbiota dysbiosis and envision the opportunities and challenges of radiation enteritis therapy.
Collapse
Affiliation(s)
- Qilin Yang
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- School of Clinical Medicine of Nanjing Medical University, Nanjing, China
| | - Bingzhi Qin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Weiliang Hou
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Shanghai Cancer Institute, Renji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| | - Huanlong Qin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| | - Fang Yin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| |
Collapse
|
37
|
The Progress in Reconstruction of Mandibular Defect Caused by Osteoradionecrosis. JOURNAL OF ONCOLOGY 2023; 2023:1440889. [PMID: 36968640 PMCID: PMC10033216 DOI: 10.1155/2023/1440889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 03/17/2023]
Abstract
Osteoradionecrosis (ORN) is described as a disease with exposed, nonviable bone that fails to heal spontaneously or by means of conservative treatment after radiotherapy in at least 3 months. Though traditional theories in the early stage including hypoxic-hypocellular-hypovascular and fibro-atrophic in addition to new findings such as ferroptosis were put forward to explain the mechanisms of the osteoradionecrosis, the etiology of ORN is still unclear. With the high rate of occurrence in the head and neck area, especially in the mandible, this disease can disrupt the shape and function of the irradiated area, leading to a clinical presentation ranging from stable small areas of asymptomatic exposed bone to severe progressive necrosis. In severe cases, patients may experience pain, xerostomia, dysphagia, facial fistulas, and even a jaw defect. Consequently, sequence therapy and sometimes extensive surgery and reconstructions are needed to manage these sequelae. Treatment options may include pain medication, antibiotics, the removal of sequesters, hyperbaric oxygen therapy, segmental resection of the mandible, and free flap reconstruction. Microanastomosed free-flaps are considered to be promising choice for ORN reconstruction in recent researches, and new methods including three-dimensional (3-D) printing, pentoxifylline, and amifostine are used nowadays in trying increase the success rates and improve quality of the reconstruction. This review summarizes the main research progress in osteoradionecrosis and reconstruction treatment of osteoradionecrosis with mandibular defect.
Collapse
|
38
|
Molecular Influence of the ATM Protein in the Treatment of Human Cells with Different Radioprotective Drugs: Comparisons between Antioxidative and Pro-Episkevic Strategies. Biomolecules 2023; 13:biom13030524. [PMID: 36979459 PMCID: PMC10046588 DOI: 10.3390/biom13030524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 03/15/2023] Open
Abstract
The radiation protection strategy with chemical agents has long been based on an antioxidative approach consisting in reducing the number of radical oxygen and nitrogen species responsible for the formation of the radiation-induced (RI) DNA damage, notably the DNA double-strand breaks (DSB), whose subset participates in the RI lethal effect as unrepairable damage. Conversely, a DSB repair-stimulating strategy that may be called the “pro-episkevic” approach (from the ancient Greek episkeve, meaning repair) can be proposed. The pro-episkevic approach directly derives from a mechanistic model based on the RI nucleoshuttling of the ATM protein (RIANS) and contributes to increase the number of DSB managed by NHEJ, the most predominant DSB repair and signaling pathway in mammalians. Here, three radioresistant and three radiosensitive human fibroblast cell lines were pretreated with antioxidative agents (N-acetylcysteine or amifostine) or to two pro-episkevic agents (zoledronate or pravastatin or both (ZOPRA)) before X-ray irradiation. The fate of the RI DSB was analyzed by using γH2AX and pATM immunofluorescence. While amifostine pretreatment appeared to be the most efficient antioxidative process, ZOPRA shows the most powerful radiation protection, suggesting that the pro-episkevic strategy may be an alternative to the antioxidative one. Additional investigations are needed to develop some new drugs that may elicit both antioxidative and pro-episkevic properties and to quantify the radiation protection action of both types of drugs applied concomitantly.
Collapse
|
39
|
Luo H, Sun Y, Wang L, Zhao R, James B. Cellular proteomic profiling of esophageal epithelial cells cultured under physioxia or normoxia reveals high correlation of radiation response. RADIATION MEDICINE AND PROTECTION 2023. [DOI: 10.1016/j.radmp.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
|
40
|
Liu L, Liang Z, Ma S, Li L, Liu X. Radioprotective countermeasures for radiation injury (Review). Mol Med Rep 2023; 27:66. [PMID: 36799170 PMCID: PMC9926870 DOI: 10.3892/mmr.2023.12953] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 01/10/2023] [Indexed: 02/09/2023] Open
Abstract
A series of physiological and pathological changes occur after radiotherapy and accidental exposure to ionizing radiation (IR). These changes cause serious damage to human tissues and can lead to death. Radioprotective countermeasures are radioprotective agents that prevent and reduce IR injury or have therapeutic effects. Based on a good understanding of radiobiology, a number of protective agents have achieved positive results in early clinical trials. The present review grouped known radioprotective agents according to biochemical categories and potential clinical use, and reviewed radiation countermeasures, i.e., radioprotectors, radiation mitigators and radiotherapeutic agents, with an emphasis on their current status and research progress. The aim of the present review is to facilitate the selection and application of suitable radioprotectors for clinicians and researchers, to prevent or reduce IR injury.
Collapse
Affiliation(s)
- Lianchang Liu
- National Health Commission Key Laboratory of Radiobiology, School of Public Health of Jilin University, Jilin, Changchun 130021, P.R. China,School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,Department of Intervention, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Zhenzhen Liang
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, P.R. China
| | - Shumei Ma
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou, Zhejiang 325035, P.R. China
| | - Lan Li
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou, Zhejiang 325035, P.R. China,Correspondence to: Professor Lan Li, School of Public Health and Management, Wenzhou Medical University, 1 North Zhongxin Road, Chashan, Wenzhou, Zhejiang 325035, P.R. China, E-mail:
| | - Xiaodong Liu
- National Health Commission Key Laboratory of Radiobiology, School of Public Health of Jilin University, Jilin, Changchun 130021, P.R. China,School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China,South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou, Zhejiang 325035, P.R. China,Professor Xiaodong Liu, National Health Commission Key Laboratory of Radiobiology, School of Public Health of Jilin University, 1163 Xinmin Road, Changchun, Jilin 130021, P.R. China, E-mail:
| |
Collapse
|
41
|
Chmil V, Filipová A, Tichý A. Looking for the phoenix: the current research on radiation countermeasures. Int J Radiat Biol 2023; 99:1148-1166. [PMID: 36745819 DOI: 10.1080/09553002.2023.2173822] [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: 06/07/2022] [Revised: 09/06/2022] [Accepted: 12/26/2022] [Indexed: 02/08/2023]
Abstract
PURPOSE Ionizing radiation (IR) is widely applied in radiotherapy for the treatment of over 50% of cancer patients. IR is also intensively used in medical diagnostics on a daily basis in imaging. Moreover, recent geopolitical events have re-ignited the real threat of the use of nuclear weapons. Medical radiation countermeasures represent one of the effective protection strategies against the effects of IR. The aim of this review was to summarize the most commonly used strategies and procedures in the development of radiation countermeasures and to evaluate the current state of their research, with a focus on those in the clinical trial phase. METHODS Clinical trials for this review were selected in accordance with the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement. The search was performed in the clinicaltrials.gov database as of May 2022. RESULTS Our search returned 263 studies, which were screened and of which 25 were included in the review. 10 of these studies had been completed, 3 with promising results: KMRC011 increased G-CSF, IL-6, and neutrophil counts suggesting potential for the treatment of hematopoietic acute radiation syndrome (H-ARS); GC4419 reduced the number of patients with severe oral mucositis and its duration; the combination of enoxaparin, pentoxifylline, and ursodeoxycholic acid reduced the incidence of focal radiation-induced liver injury. CONCLUSION The agents discovered so far show significant side effects or low efficacy, and hence most of the tested agents terminate in the early stages of development. In addition, the low profitability of this type of drug demotivates the private sector to invest in such research. To overcome this problem, there is a need to involve more public resources in funding. Among the technological opportunities, a deeper use of in silico approaches seems to be prospective.
Collapse
Affiliation(s)
- Vojtěch Chmil
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | - Alžběta Filipová
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | - Aleš Tichý
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
- Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
42
|
Almansoori AA, Hariharan A, Cao UMN, Upadhyay A, Tran SD. Drug Therapeutics Delivery to the Salivary Glands: Intraglandular and Intraductal Injections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1436:119-130. [PMID: 36809639 DOI: 10.1007/5584_2023_765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Salivary gland hypofunction and xerostomia following pathological conditions like Sjogren's syndrome or head and neck radiotherapy usually lead to tremendous impairment of oral health, speech, and swallowing. The use of systemic drugs to alleviate the symptoms of these conditions has been associated with various adverse effects. Techniques of local drug delivery to the salivary gland have grown enormously to address this problem properly. The techniques include intraglandular and intraductal injections. In this chapter, we will provide a review of the literature for both techniques while incorporating our lab experience in using them.
Collapse
Affiliation(s)
- Akram Abdo Almansoori
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - Arvind Hariharan
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - Uyen M N Cao
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - Akshaya Upadhyay
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - Simon D Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
43
|
Lv G, Wu X, Wang Z, Wu K, Ang G, Cui S, Shi Y, Wang Y, Liu D. Higher radiation doses after partial laryngectomy may raise the incidence of pneumonia: A retrospective cohort study. Front Oncol 2022; 12:1072474. [PMID: 36636552 PMCID: PMC9831674 DOI: 10.3389/fonc.2022.1072474] [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: 10/17/2022] [Accepted: 12/05/2022] [Indexed: 12/28/2022] Open
Abstract
Background Currently, studies have shown that a high dose of radiotherapy to the throat have various harmful and adverse effects on the patients' laryngeal function, resulting in the development of pneumonia. This study aimed to explore how radiotherapy dose affected the probability of pneumonia following laryngeal cancer surgery. Materials and methods A retrospective analysis was done on patients diagnosed with laryngeal cancer between 2010 and 2020 and were treated surgically and with postoperative radiotherapy in the same institution. This study included 108 patients in total, 51 of who were in the low-dose group and 57 of whom were in the high-dose group. Age, gender, the location of laryngeal cancer, the presence or absence of lymph node metastasis, and other demographic and clinical characteristics were collected, and the prevalence of postoperative pneumonia was compared between the two groups. Results The total prevalence of postoperative pneumonia was 59.3%, but there was a significant difference between the two groups(high-dose group 71.9% VS low-dose group 45.1%; p=0.005). A total of 9.3% (10/108) of the patients had readmission due to severe pneumonia, and the rate of readmission due to pneumonia was significantly different between the two groups (high-dose group 15.8% VS low-dose group 2.0%, p=0.032). Additionally, the high-dose group's prevalence of Dysphagia was significantly higher than the low-dose group's. According to multivariate logistic modeling, high-dose radiation was a risk factor for pneumonia (OR=4.224, 95%CI =1.603-11.131, p=0.004). Conclusion Pneumonia risk could increase with radiotherapy doses > 50 Gy in the treatment of laryngeal cancer. Therefore, we recommend that when the radiation dose surpasses 50Gy, doctors should pay particular attention to the lung health of patients with laryngeal cancer.
Collapse
Affiliation(s)
- Guoqi Lv
- Department of Otorhinolaryngology Head and Neck Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning, China,Dalian Medical University, Dalian, Liaoning, China
| | - Xiuling Wu
- Department of Otorhinolaryngology Head and Neck Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning, China,Dalian Medical University, Dalian, Liaoning, China
| | - Zhengying Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning, China,China Medical University, Shenyang, Liaoning, China
| | - Kanglong Wu
- Dalian Medical University, Dalian, Liaoning, China,The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Geer Ang
- Department of Otorhinolaryngology Head and Neck Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning, China,Dalian Medical University, Dalian, Liaoning, China
| | - Shulin Cui
- Department of Otorhinolaryngology Head and Neck Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning, China,Dalian Medical University, Dalian, Liaoning, China
| | - Yuqi Shi
- Department of Otorhinolaryngology Head and Neck Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning, China,Dalian Medical University, Dalian, Liaoning, China
| | - Yu Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning, China,Dalian Medical University, Dalian, Liaoning, China
| | - Delong Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning, China,*Correspondence: Delong Liu,
| |
Collapse
|
44
|
Vasin MV, Ushakov IB, Chernov YN, Semenova LA, Afanasyev RV. Radioprotective Properties of Indralin and Essentiale N for Separate and Combined Application under Fractionated γ-Irradiation. BIOL BULL+ 2022. [DOI: 10.1134/s106235902211022x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
|
45
|
Oronsky B, Caroen S, Abrouk N, Reid TR. RRx-001 and the "Right stuff": Protection and treatment in outer space. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:69-75. [PMID: 36336372 DOI: 10.1016/j.lssr.2022.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/26/2022] [Accepted: 05/09/2022] [Indexed: 06/16/2023]
Abstract
From antibiotics to aspirin to antimalarials and to anticancer agents, about half of the world's best-selling drugs are derived from nature. However, accelerating climatic disruption, habitat destruction, pollution, and biodiversity loss all negatively impact the potential of natural sources to continue to serve as repositories of novel pharmaceuticals. On that basis, the final frontier for drug development is perhaps not the rainforests, coral reefs, and other natural habitats but rather the aerospace industry with its virtually unlimited and inexhaustible man-made 'library' of potentially bioactive compounds. The first aerospace-sourced therapeutic to reach the clinic is RRx-001, an inhibitor of the NOD-like receptor - Nucleotide-binding oligomerization domain with Leucine rich Repeat and Pyrin domain (NLRP3) inflammasome in a Phase 3 trial for the treatment of small cell lung cancer (SCLC) and in a soon-to-start Phase 3 trial for protection against chemoradiotherapy-induced severe oral mucositis in first line head and neck cancer. As manned missions to the Moon, Mars, and asteroids as well as space tourism beckon, it is perhaps fitting that a compound like RRx-001, which is derived from 1,3,3-Trinitroazetidine (TNAZ), an explosive propellant for rockets, is a potential "all purpose" option to mitigate the major biomedical effects of space radiation exposures including cancer development and other tissue degenerations both within mission and after mission. This article highlights the promise of RRx-001 to attenuate the acute and late effects of radiation exposure on astronauts including the development of cancer.
Collapse
|
46
|
Yang Z, Zhong W, Yang L, Wen P, Luo Y, Wu C. The emerging role of exosomes in radiotherapy. Cell Commun Signal 2022; 20:171. [PMCID: PMC9620591 DOI: 10.1186/s12964-022-00986-1] [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: 07/29/2022] [Accepted: 10/01/2022] [Indexed: 11/10/2022] Open
Abstract
Presently, more than half of cancer patients receive radiotherapy to cure localized cancer, palliate symptoms, or control the progression of cancer. However, radioresistance and radiation-induced bystander effects (RIBEs) are still challenging problems in cancer treatment. Exosomes, as a kind of extracellular vesicle, have a significant function in mediating and regulating intercellular signaling pathways. An increasing number of studies have shown that radiotherapy can increase exosome secretion and alter exosome cargo. Furthermore, radiation-induced exosomes are involved in the mechanism of radioresistance and RIBEs. Therefore, exosomes hold great promise for clinical application in radiotherapy. In this review, we not only focus on the influence of radiation on exosome biogenesis, secretion and cargoes but also on the mechanism of radiation-induced exosomes in radioresistance and RIBEs, which may expand our insight into the cooperative function of exosomes in radiotherapy.
Video abstract
Collapse
Affiliation(s)
- Zhenyi Yang
- grid.412644.10000 0004 5909 0696Fourth Affiliated Hospital of China Medical University, Liaoning, China
| | - Wen Zhong
- grid.412644.10000 0004 5909 0696Fourth Affiliated Hospital of China Medical University, Liaoning, China
| | - Liang Yang
- grid.412644.10000 0004 5909 0696Fourth Affiliated Hospital of China Medical University, Liaoning, China
| | - Ping Wen
- grid.412644.10000 0004 5909 0696Fourth Affiliated Hospital of China Medical University, Liaoning, China
| | - Yixuan Luo
- grid.412644.10000 0004 5909 0696Fourth Affiliated Hospital of China Medical University, Liaoning, China
| | - Chunli Wu
- grid.412644.10000 0004 5909 0696Fourth Affiliated Hospital of China Medical University, Liaoning, China
| |
Collapse
|
47
|
Chen Y, Yang Y, Tang H, Zhang Z, Zhou X, Xu W. ROS-Responsive and pH-Sensitive Aminothiols Dual-Prodrug for Radiation Enteritis. Antioxidants (Basel) 2022; 11:antiox11112145. [PMID: 36358517 PMCID: PMC9686648 DOI: 10.3390/antiox11112145] [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: 09/27/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 11/30/2022] Open
Abstract
Radiation exposure can immediately trigger a burst of reactive oxygen species (ROS), which can induce severe cell death and long-term tissue damage. Therefore, instantaneous release of sufficient radioprotective drugs is vital to neutralize those accumulated ROS in IR-exposed areas. To achieve this goal, we designed, synthesized, and evaluated a novel oral ROS-responsive radioprotective compound (M1) with high biocompatibility and efficient ROS-scavenging ability to act as a promising oral drug for radiation protection. The compound is stably present in acidic environments and is hydrolyzed in the intestine to form active molecules rich in thiols. M1 can significantly remove cellular ROS and reduce DNA damage induced by γ-ray radiation. An in vivo experiment showed that oral administration of M1 effectively alleviates acute radiation-induced intestinal injury. Immunohistochemical staining showed that M1 improved cell proliferation, reduced cell apoptosis, and enhanced the epithelial integrity of intestinal crypts. This study provides a promising oral ROS-sensitive agent for acute intestinal radiation syndrome.
Collapse
|
48
|
Synthesis of novel benzothiophene derivatives as protectors against cranial irradiation-induced neuroinflammation. Future Med Chem 2022; 14:1527-1539. [DOI: 10.4155/fmc-2022-0203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Cranial irradiation results in many deleterious effects to normal tissues, including neuroinflammation. There is a need to explore radioprotective agents that could be safely used to ameliorate these effects. Method: Nine novel benzothiophene derivatives bearing pyrimidinone, pyrazolidinone, triazole and other active moieties were synthesized and evaluated as antioxidants in an in vitro screening experiment. The most potent compounds were then tested as protectors against radiation-induced neuroinflammation and oxidative stress in rat brains following cranial irradiation. Results: The most potent antioxidant compounds were compounds 3–5 and 10 . P-fluro,p- bromo and pyrido benzothiophene derivatives offered good antioxidant and anti-inflammatory effects. Conclusion: Compounds 3–5 may be introduced as nontoxic candidates for adjuvant therapeutic protocols used in head and neck tumor radiotherapeutic management.
Collapse
|
49
|
Parashar P, Das MK, Tripathi P, Kataria T, Gupta D, Sarin D, Hazari PP, Tandon V. DMA, a Small Molecule, Increases Median Survival and Reduces Radiation-Induced Xerostomia via the Activation of the ERK1/2 Pathway in Oral Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14194908. [PMID: 36230831 PMCID: PMC9562201 DOI: 10.3390/cancers14194908] [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: 06/11/2022] [Revised: 06/24/2022] [Accepted: 07/09/2022] [Indexed: 11/16/2022] Open
Abstract
Survival, recurrence, and xerostomia are considerable problems in the treatment of oral squamous carcinoma patients. In this study, we investigated the role of DMA (5-(4-methylpiperazin-1-yl)-2-[2′-(3,4-dimethoxyphenyl)5″benzimidazoyl]benzimidazole) as a salivary gland cytoprotectant in a patient-derived xenograft mouse model. A significant increase in saliva secretion was observed in the DMA-treated xenograft compared to radiation alone. Repeated doses of DMA with a high dose of radiation showed a synergistic effect on mice survival and reduced tumor growth. The mean survival rate of tumor-bearing mice was significantly enhanced. The increased number of Ki-67-stained cells in the spleen, intestine, and lungs compared to the tumor suggests DMA ablates the tumor but protects other organs. The expression of aquaporin-5 was restored in tumor-bearing mice injected with DMA before irradiation. The reduced expression of αvβ3 integrin and CD44 in DMA alone and DMA with radiation-treated mice suggests a reduced migration of cells and stemness of cancer cells. DMA along with radiation treatment results in the activation of the Ras/Raf/MEK/ERK pathway in the tumor, leading to apoptosis through caspase upregulation. In conclusion, DMA has strong potential for use as an adjuvant in radiotherapy in OSCC patients.
Collapse
Affiliation(s)
- Palak Parashar
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Monoj Kumar Das
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Pragya Tripathi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Tejinder Kataria
- Division of Radiation Oncology, Medanta―The Medicity, Gurgaon 122001, India
| | - Deepak Gupta
- Division of Radiation Oncology, Medanta―The Medicity, Gurgaon 122001, India
| | - Deepak Sarin
- Head and Neck OncoSurgery, Medanta―The Medicity, Gurgaon 122001, India
| | - Puja Panwar Hazari
- Defence Research and Development Organization, Institute of Nuclear Medicine and Allied Sciences, Delhi 110054, India
| | - Vibha Tandon
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
- Correspondence: ; Tel.: +91-11-26742181
| |
Collapse
|
50
|
Kim LN, Rubenstein RN, Chu JJ, Allen RJ, Mehrara BJ, Nelson JA. Noninvasive Systemic Modalities for Prevention of Head and Neck Radiation-Associated Soft Tissue Injury: A Narrative Review. J Reconstr Microsurg 2022; 38:621-629. [PMID: 35213927 PMCID: PMC9402815 DOI: 10.1055/s-0042-1742731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Radiation-associated soft tissue injury is a potentially devastating complication for head and neck cancer patients. The damage can range from minor sequelae such as xerostomia, which requires frequent daily maintenance, to destructive degenerative processes such as osteoradionecrosis, which can contribute to flap failure and delay or reverse oral rehabilitation. Despite the need for effective radioprotectants, the literature remains sparse, primarily focused on interventions beyond the surgeon's control, such as maintenance of good oral hygiene or modulation of radiation dose. METHODS This narrative review aggregates and explores noninvasive, systemic treatment modalities for prevention or amelioration of radiation-associated soft tissue injury. RESULTS We highlighted nine modalities with the most clinical potential, which include amifostine, melatonin, palifermin, hyperbaric oxygen therapy, photobiomodulation, pentoxifylline-tocopherol-clodronate, pravastatin, transforming growth factor-β modulators, and deferoxamine, and reviewed the benefits and limitations of each modality. Unfortunately, none of these modalities are supported by strong evidence for prophylaxis against radiation-associated soft tissue injury. CONCLUSION While we cannot endorse any of these nine modalities for immediate clinical use, they may prove fruitful areas for further investigation.
Collapse
Affiliation(s)
- Leslie N. Kim
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robyn N. Rubenstein
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jacqueline J. Chu
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert J. Allen
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Babak J. Mehrara
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jonas A. Nelson
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|