1
|
Gu W, Dong H, Yuan Y, Yuan Z, Jiang X, Qian Y, Shen Z. Is Acupuncture an Effective Treatment for Radiation-Induced Xerostomia of Patients with Head and Neck Cancer? A Systematic Review and Meta-Analysis. JOURNAL OF INTEGRATIVE AND COMPLEMENTARY MEDICINE 2025; 31:431-444. [PMID: 39600262 DOI: 10.1089/jicm.2023.0781] [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: 11/29/2024]
Abstract
Background: Radiation-induced xerostomia (RIX) stands out as one of the most severe side effects among patients with head and neck cancer (HNC). Given the varied conclusions in previous studies concerning the association between acupuncture, sham acupuncture, or acupuncture combined with standard oral care and therapeutic effects, our aim is to conduct a systematic review to assess the effectiveness and safety of acupuncture in managing RIX in patients with HNC. Methods: Six databases (Cochrane Library, PubMed, EMBASE, China National Knowledge Infrastructure, Chongqing VIP, and WanFang Database) were electronically searched, following the Cochrane manual and adhering to reported Preferred Reporting Item for Systematic Reviews and Meta-Analyses guidelines, from their inception dates to July 1, 2024. Primary randomized clinical trials included in systematic reviews or meta-analyses were identified, with the Xerostomia Questionnaire and Xerostomia Inventory designated as the primary outcomes. Salivary flow rates (unstimulated or stimulated) were defined as secondary outcomes. Results: Eight clinical trials involving 1273 participants were analyzed, with six studies included in the meta-analysis. The results indicate that acupuncture demonstrated a significant improvement in patient-reported xerostomia scores (standardized mean difference [SMD] = -0.20, 95% confidence interval [95% CI] [-0.38, -0.02], I2 = 0%) in comparison to standard care, but did not significantly improve oral dryness symptoms compared with sham acupuncture (SMD = -0.06, 95% CI [-0.29, 0.16], I2 = 25.8%). The merged total showed negative result (SMD = -0.13, 95% CI [-0.27, 0.01], I2 = 8.2%). Additionally, there was no significant difference in stimulated salivary flow rate (SMD = -0.22, 95% CI [-0.58, 0.13], I2 = 0%) and unstimulated salivary flow rate (SMD = -0.19, 95% CI [-0.11, 0.72], I2 = 67.2%). In general, the acupuncture did not cause serious adverse effects. Conclusion: As far as current research is concerned, acupuncture treatment for RIX symptoms in patients with HNC still lacks strong and convincing evidence support. The more scientific research methods and more clinical trials are still needed.
Collapse
Affiliation(s)
- Wenzhe Gu
- Department of Otolaryngology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Hongjun Dong
- Department of Otolaryngology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Yuan Yuan
- Department of Rehabilitation, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Zijiang Yuan
- Department of Otolaryngology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Xiaoting Jiang
- Department of Otolaryngology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Yuhan Qian
- Department of Otolaryngology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Zhengjie Shen
- Department of Oncology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| |
Collapse
|
2
|
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
|
3
|
Pillai S, Munguia-Lopez JG, Liu Y, Gigliotti J, Zeitouni A, Kinsella JM, Tran SD. Development of human salivary gland cell lines for modeling radiation-induced damage in three-dimensional spheroid cultures. J Tissue Eng 2025; 16:20417314251326667. [PMID: 40322739 PMCID: PMC12048756 DOI: 10.1177/20417314251326667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/25/2025] [Indexed: 05/08/2025] Open
Abstract
No permanent cure exists for salivary gland (SG) damage and consequent xerostomia (dry mouth) in patients undergoing radiotherapy for head and neck cancers. The lack of commercially available healthy human SG-derived cell lines has hindered in vitro studies of radiation-induced glandular injury. In this study, we successfully immortalized and characterized two novel human major SG-derived cell lines. Leveraging these cell lines and hyaluronic-acid hydrogels, we bioengineered distinct multicellular SG spheroids and microtissues expressing key acinar, ductal, myoepithelial, and mesenchymal cell markers in long-term cultures. Further, using this platform, we developed a proof-of-concept radiation injury model, demonstrating spheroid disruption characterized by actin depolymerization, DNA damage, apoptosis, and loss of SG-specific markers following radiation exposure. Notably, these detrimental effects were partially mitigated with a radioprotective agent. Our findings demonstrate that the bioengineered SG spheroids provide a scalable and versatile platform with significant potential for disease modeling and drug testing, thereby accelerating the development of targeted therapies for radiation-induced xerostomia.
Collapse
Affiliation(s)
- Sangeeth Pillai
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | | | - Younan Liu
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - Jordan Gigliotti
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - Anthony Zeitouni
- Department of Otolaryngology-Head and Neck Surgery, McGill University Health Center, Montreal, QC, Canada
| | | | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| |
Collapse
|
4
|
Yorgancilar N, Kose O, Rakici SY, Mercantepe T, Akyildiz K, Tumkaya L, Yilmaz A. Preventive effects of melatonin and amifostine on irradiated rats with experimental periodontitis. BMC Oral Health 2024; 24:1453. [PMID: 39614289 DOI: 10.1186/s12903-024-05251-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND The aim of this study was to investigate the preventive effects of amifostine and melatonin oxidatively, biochemically and histomorphometrically in rats with radiotherapy-induced experimental periodontitis. METHODS 40 female Sprague-Dawley rats were divided into 5 groups: Control, experimental periodontitis (Ep), Ep + radiotherapy (Ep + Rt), Ep + Rt + amifostine (Ep + Rt + Ami), Ep + Rt + melatonin (Ep + Rt + Mel). The day after induction of periodontitis by ligature, a single dose of 5 Gy radiotherapy was administered. On the same day, treatments with amifostine (200 mg/kg) for 3 days and melatonin (10 mg/kg) for 15 days were started. By after 23 days of experiment, periodontal bone loss was measured by histomorphometry. RANKL, OPG and Caspase-3 activities were analyzed immunohistochemically and inflammatory cytokine (IL-1β, IL-10, IL-6, TNF-α) levels and oxidative stress (TOS/TAS) were analyzed biochemically in tissue homogenates. RESULTS It was observed that there was a significant difference in many biochemical parameters and oxidative stress levels between the control group and Ep + Rt (p < 0.05). Alveolar bone destruction in the melatonin prophylaxis group was observed to be close to control (p > 0.05). Melatonin significantly improved biochemical, histochemical, apoptotic and bone loss levels in irradiated experimental periodontitis rats (p < 0.05). When comparing the two drug groups (Ep + Rt + Ami and Ep + Rt + Mel), no statistically significant difference was found at any parameter level (p > 0.05). CONCLUSION Both melatonin and amifostine can significantly limit RT-induced periodontal bone loss by suppressing inflammatory stress, apoptotic mechanisms, and RANKL-related osteoclastic activity. Given the limited side effects of melatonin, it may be an alternative to amifostine.
Collapse
Affiliation(s)
- Nur Yorgancilar
- School of Dentistry, Department of Periodontology, Recep Tayyip Erdogan University, Rize, 53100, TR, Turkey
| | - Oguz Kose
- School of Dentistry, Department of Periodontology, Recep Tayyip Erdogan University, Rize, 53100, TR, Turkey.
| | - Sema Yilmaz Rakici
- School of Medicine, Department of Radiation Oncology, Recep Tayyip Erdogan University, Rize, Turkey
| | - Tolga Mercantepe
- School of Medicine, Department of Histology and Embryology, Recep Tayyip Erdogan University, Rize, Turkey
| | - Kerimali Akyildiz
- School of Healh Care Services Vocational, Department of Medical Services and Techniques, Recep Tayyip Erdogan University, Rize, Turkey
| | - Levent Tumkaya
- School of Medicine, Department of Histology and Embryology, Recep Tayyip Erdogan University, Rize, Turkey
| | - Adnan Yilmaz
- School of Medicine, Department of Biochemistry, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
5
|
Farhadi M, Fadavi P, Mohebbi S, Taghizadeh-Hesary F. A new approach to prevent radiation-induced xerostomia using intraglandular injection of mitochondria-boosting agents. BMC Cancer 2024; 24:832. [PMID: 38992600 PMCID: PMC11241784 DOI: 10.1186/s12885-024-12582-2] [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: 12/03/2023] [Accepted: 06/30/2024] [Indexed: 07/13/2024] Open
Abstract
Radiotherapy in patients with head and neck cancer fairly leads to xerostomia, profoundly affecting their quality of life. With limited effective preventive and therapeutic methods, attention has turned to exploring alternatives. This article outlines how intraglandular injection of mitochondria-boosting agents can serve as a potential strategy to reduce salivary acinar damage. This method can contribute to the thoughtful development of study protocols or medications to reduce radiation-induced salivary glands damage.
Collapse
Affiliation(s)
- Mohammad Farhadi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Pedram Fadavi
- Department of Radiation Oncology, Iran University of Medical Sciences, Tehran, Iran
- Breast Health Cancer Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saleh Mohebbi
- Skull Base Research Center, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Department of Radiation Oncology, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
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
|
7
|
Li CX, Tan XR, Wei W, Li MQ, Zhang WN, Gong ZC, Zhang Y, Zhao HR. A radiobiological perspective on radioresistance or/and radiosensitivity of head and neck squamous cell carcinoma. Rep Pract Oncol Radiother 2024; 28:809-822. [PMID: 38515813 PMCID: PMC10954264 DOI: 10.5603/rpor.99355] [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: 07/22/2023] [Accepted: 10/11/2023] [Indexed: 03/23/2024] Open
Abstract
Background This article aimed to compile and summarize clinically relevant literature in radiation therapy, and to discuss the potential in radioresistant and radiosensitive head and neck cancer. Study Design Narrative review. Materials and methods Google Scholar, PubMed and the Cochrane Library were retrieved using combined key words such as "radiotherapy" and "head and neck cancer". Search strings additionally queried were "radioresistant", "radiosensitive", "head and neck region", "squamous cell carcinoma", in combination with Boolean Operators 'AND' and 'OR'. Subsequently, the resulting publications were included for review of the full text. Results Radiotherapeutic response currently in clinical observation referred to HNSCC scoping were selected into this review. The compiled mechanisms were then detailed concerning on the clinical significance, biological characteristics, and molecular function. Conclusions Brachytherapy or/and external-beam radiotherapy are crucial for treating HNSCC, especially the early stage patients, but in patients with locally advanced tumors, their outcome with radiation therapy is poor due to obvious radioresistance. The curative effects mainly depend on the response of radiation therapy, so an updated review is needed to optimize further applications in HNSCC radiotherapy.
Collapse
Affiliation(s)
- Chen-xi Li
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-rong Tan
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Wei Wei
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Mu-qiu Li
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Wei-na Zhang
- Ear, Nose & Throat Department, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhong-cheng Gong
- Department of Oral and Maxillofacial Oncology & Surgery, School/Hospital of Stomatology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yang Zhang
- The First Ward of Oncological Department, Cancer Center, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hua-rong Zhao
- The First Ward of Oncological Department, Cancer Center, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Wei F, Tuong ZK, Omer M, Ngo C, Asiatico J, Kinzel M, Pugazhendhi AS, Khaled AR, Ghosh R, Coathup M. A novel multifunctional radioprotective strategy using P7C3 as a countermeasure against ionizing radiation-induced bone loss. Bone Res 2023; 11:34. [PMID: 37385982 DOI: 10.1038/s41413-023-00273-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/16/2023] [Accepted: 05/28/2023] [Indexed: 07/01/2023] Open
Abstract
Radiotherapy is a critical component of cancer care but can cause osteoporosis and pathological insufficiency fractures in surrounding and otherwise healthy bone. Presently, no effective countermeasure exists, and ionizing radiation-induced bone damage continues to be a substantial source of pain and morbidity. The purpose of this study was to investigate a small molecule aminopropyl carbazole named P7C3 as a novel radioprotective strategy. Our studies revealed that P7C3 repressed ionizing radiation (IR)-induced osteoclastic activity, inhibited adipogenesis, and promoted osteoblastogenesis and mineral deposition in vitro. We also demonstrated that rodents exposed to clinically equivalent hypofractionated levels of IR in vivo develop weakened, osteoporotic bone. However, the administration of P7C3 significantly inhibited osteoclastic activity, lipid formation and bone marrow adiposity and mitigated tissue loss such that bone maintained its area, architecture, and mechanical strength. Our findings revealed significant enhancement of cellular macromolecule metabolic processes, myeloid cell differentiation, and the proteins LRP-4, TAGLN, ILK, and Tollip, with downregulation of GDF-3, SH2B1, and CD200. These proteins are key in favoring osteoblast over adipogenic progenitor differentiation, cell matrix interactions, and shape and motility, facilitating inflammatory resolution, and suppressing osteoclastogenesis, potentially via Wnt/β-catenin signaling. A concern was whether P7C3 afforded similar protection to cancer cells. Preliminarily, and remarkably, at the same protective P7C3 dose, a significant reduction in triple-negative breast cancer and osteosarcoma cell metabolic activity was found in vitro. Together, these results indicate that P7C3 is a previously undiscovered key regulator of adipo-osteogenic progenitor lineage commitment and may serve as a novel multifunctional therapeutic strategy, leaving IR an effective clinical tool while diminishing the risk of adverse post-IR complications. Our data uncover a new approach for the prevention of radiation-induced bone damage, and further work is needed to investigate its ability to selectively drive cancer cell death.
Collapse
Affiliation(s)
- Fei Wei
- Biionix Cluster, and Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Zewen Kelvin Tuong
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Mahmoud Omer
- Biionix Cluster, and Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Christopher Ngo
- Biionix Cluster, and Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Jackson Asiatico
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Michael Kinzel
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Abinaya Sindu Pugazhendhi
- Biionix Cluster, and Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Annette R Khaled
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Ranajay Ghosh
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Melanie Coathup
- Biionix Cluster, and Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
11
|
Liu D, Wei M, Yan W, Xie H, Sun Y, Yuan B, Jin Y. Potential applications of drug delivery technologies against radiation enteritis. Expert Opin Drug Deliv 2023; 20:435-455. [PMID: 36809906 DOI: 10.1080/17425247.2023.2183948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
INTRODUCTION The incidence of abdominal tumors, such as colorectal and prostate cancers, continually increases. Radiation therapy is widely applied in the clinical treatment of patients with abdominal/pelvic cancers, but it often unfortunately causes radiation enteritis (RE) involving the intestine, colon, and rectum. However, there is a lack of suitable treatment options for effective prevention and treatment of RE. AREAS COVERED Conventional clinical drugs for preventing and treating RE are usually applied by enemas and oral administration. Innovative gut-targeted drug delivery systems including hydrogels, microspheres, and nanoparticles are proposed to improve the prevention and curation of RE. EXPERT OPINION The prevention and treatment of RE have not attracted sufficient attention in the clinical practice, especially compared to the treatment of tumors, although RE takes patients great pains. Drug delivery to the pathological sites of RE is a huge challenge. The short retention and weak targeting of conventional drug delivery systems affect the therapeutic efficiency of anti-RE drugs. Novel drug delivery systems including hydrogels, microspheres, and nanoparticles can allow drugs long-term retention in the gut and targeting the inflammation sites to alleviate radiation-induced injury.
Collapse
Affiliation(s)
- Dongdong Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Meng Wei
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Wenrui Yan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hua Xie
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yingbao Sun
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Bochuan Yuan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
12
|
Total Skin Treatment with Helical Arc Radiotherapy. Int J Mol Sci 2023; 24:ijms24054492. [PMID: 36901922 PMCID: PMC10002962 DOI: 10.3390/ijms24054492] [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: 01/14/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 03/02/2023] Open
Abstract
For widespread cutaneous lymphoma, such as mycosis fungoides or leukemia cutis, in patients with acute myeloid leukemia (AML) and for chronic myeloproliferative diseases, total skin irradiation is an efficient treatment modality for disease control. Total skin irradiation aims to homogeneously irradiate the skin of the entire body. However, the natural geometric shape and skin folding of the human body pose challenges to treatment. This article introduces treatment techniques and the evolution of total skin irradiation. Articles on total skin irradiation by helical tomotherapy and the advantages of total skin irradiation by helical tomotherapy are reviewed. Differences among each treatment technique and treatment advantages are compared. Adverse treatment effects and clinical care during irradiation and possible dose regimens are mentioned for future prospects of total skin irradiation.
Collapse
|
13
|
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
|
14
|
Obrador E, Salvador-Palmer R, Villaescusa JI, Gallego E, Pellicer B, Estrela JM, Montoro A. Nuclear and Radiological Emergencies: Biological Effects, Countermeasures and Biodosimetry. Antioxidants (Basel) 2022; 11:1098. [PMID: 35739995 PMCID: PMC9219873 DOI: 10.3390/antiox11061098] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022] Open
Abstract
Atomic and radiological crises can be caused by accidents, military activities, terrorist assaults involving atomic installations, the explosion of nuclear devices, or the utilization of concealed radiation exposure devices. Direct damage is caused when radiation interacts directly with cellular components. Indirect effects are mainly caused by the generation of reactive oxygen species due to radiolysis of water molecules. Acute and persistent oxidative stress associates to radiation-induced biological damages. Biological impacts of atomic radiation exposure can be deterministic (in a period range a posteriori of the event and because of destructive tissue/organ harm) or stochastic (irregular, for example cell mutation related pathologies and heritable infections). Potential countermeasures according to a specific scenario require considering basic issues, e.g., the type of radiation, people directly affected and first responders, range of doses received and whether the exposure or contamination has affected the total body or is partial. This review focuses on available medical countermeasures (radioprotectors, radiomitigators, radionuclide scavengers), biodosimetry (biological and biophysical techniques that can be quantitatively correlated with the magnitude of the radiation dose received), and strategies to implement the response to an accidental radiation exposure. In the case of large-scale atomic or radiological events, the most ideal choice for triage, dose assessment and victim classification, is the utilization of global biodosimetry networks, in combination with the automation of strategies based on modular platforms.
Collapse
Affiliation(s)
- Elena Obrador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Rosario Salvador-Palmer
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Juan I. Villaescusa
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain; (J.I.V.); (A.M.)
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| | - Eduardo Gallego
- Energy Engineering Department, School of Industrial Engineering, Polytechnic University of Madrid, 28040 Madrid, Spain;
| | - Blanca Pellicer
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - José M. Estrela
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Alegría Montoro
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain; (J.I.V.); (A.M.)
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| |
Collapse
|
15
|
Radioprotective effects and mechanism of HL-003 on radiation-induced salivary gland damage in mice. Sci Rep 2022; 12:8419. [PMID: 35589816 PMCID: PMC9120142 DOI: 10.1038/s41598-022-12581-y] [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: 02/02/2022] [Accepted: 05/12/2022] [Indexed: 11/25/2022] Open
Abstract
Ionizing radiation (IR) can cause damage to the structure and function of salivary glands. Our research group independently synthesized the ROS scavenger, HL-003. The aim of this study was to explore the protective effects and underlying mechanisms of HL-003 on radiation-induced salivary gland injury. Salivary flow rate measurement, H&E staining, immunohistochemistry, FRAP, TUNEL, and western blotting were used to evaluate the radioprotective effect on salivary glands. The results showed that HL-003 protected the salivary secretion function by protecting the AQP-5 protein, on the salivary epithelial cell membrane, from IR damage. HL-003 reduced oxidative stress in the salivary gland by regulating the expression of ROS-related proteins NOX4, SOD2, and 8-OHdG. Furthermore, HL-003 downregulated the expression of p-p53, Bax, caspase 3, and caspase 9, and upregulated the expression of Bcl-2, suggesting that it could inhibit the activation of p53 to reduce cell apoptosis. In conclusion, HL-003 is an effective radioprotector that prevents damage of the radiation-induced salivary gland.
Collapse
|
16
|
Park EA, Graves SA, Menda Y. The Impact of Radiopharmaceutical Therapy on Renal Function. Semin Nucl Med 2022; 52:467-474. [DOI: 10.1053/j.semnuclmed.2022.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/13/2022] [Accepted: 02/20/2022] [Indexed: 11/11/2022]
|
17
|
Vasin MV. B-190 (Indralin) in Light of the History of the Formation of Ideas about the Mechanism of Action of Radioprotectors. BIOL BULL+ 2022. [DOI: 10.1134/s1062359021110091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Boutros SW, Zimmerman B, Nagy SC, Lee JS, Perez R, Raber J. Amifostine (WR-2721) Mitigates Cognitive Injury Induced by Heavy Ion Radiation in Male Mice and Alters Behavior and Brain Connectivity. Front Physiol 2021; 12:770502. [PMID: 34867479 PMCID: PMC8637850 DOI: 10.3389/fphys.2021.770502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022] Open
Abstract
The deep space environment contains many risks to astronauts during space missions, such as galactic cosmic rays (GCRs) comprised of naturally occurring heavy ions. Heavy ion radiation is increasingly being used in cancer therapy, including novel regimens involving carbon therapy. Previous investigations involving simulated space radiation have indicated a host of detrimental cognitive and behavioral effects. Therefore, there is an increasing need to counteract these deleterious effects of heavy ion radiation. Here, we assessed the ability of amifostine to mitigate cognitive injury induced by simulated GCRs in C57Bl/6J male and female mice. Six-month-old mice received an intraperitoneal injection of saline, 107 mg/kg, or 214 mg/kg of amifostine 1 h prior to exposure to a simplified five-ion radiation (protons, 28Si, 4He, 16O, and 56Fe) at 500 mGy or sham radiation. Mice were behaviorally tested 2-3 months later. Male mice that received saline and radiation exposure failed to show novel object recognition, which was reversed by both doses of amifostine. Conversely, female mice that received saline and radiation exposure displayed intact object recognition, but those that received amifostine prior to radiation did not. Amifostine and radiation also had distinct effects on males and females in the open field, with amifostine affecting distance moved over time in both sexes, and radiation affecting time spent in the center in females only. Whole-brain analysis of cFos immunoreactivity in male mice indicated that amifostine and radiation altered regional connectivity in areas involved in novel object recognition. These data support that amifostine has potential as a countermeasure against cognitive injury following proton and heavy ion irradiation in males.
Collapse
Affiliation(s)
- Sydney Weber Boutros
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Benjamin Zimmerman
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Sydney C. Nagy
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Joanne S. Lee
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Ruby Perez
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Departments of Neurology and Radiation Medicine, Oregon Health & Science University, Portland, OR, United States
- Division of Neuroscience, Oregon National Primate Research Center, Portland, OR, United States
| |
Collapse
|
19
|
Gkantaifi A, Alongi F, Vardas E, Cuccia F, Hajiioannou J, Kyrodimos E, Christopoulos C, Mauri D, Charalampakis N, Trogkanis N, Tsoukalas N, Iliadis G, Tolia M. Honey Against Radiation-induced Oral Mucositis in Head and Neck Cancer Patients. An Umbrella Review of Systematic Reviews and Meta- Analyses of the Literature. Rev Recent Clin Trials 2021; 15:360-369. [PMID: 32646360 DOI: 10.2174/1574887115666200709140405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/23/2020] [Accepted: 06/05/2020] [Indexed: 11/22/2022]
Abstract
Backround: Oral mucositis (OM) consists of a major side effect of radiotherapy (RT) in head and neck (H-N) cancer patients and natural honey is gaining more and more scientific interest due to its beneficial effects in tissue repair. OBJECTIVE The aim of this review is to better clarify the preventive/therapeutic role of honey in the management of OM in patients with H-N cancer undergoing RT with or without chemotherapy (CT). METHODS We used the PubMed database to retrieve journal articles and the inclusion criteria were only reviews and meta-analyses that illustrated the effective use of honey for either the prevention or treatment of OM in H-N cancer patients receiving either RT alone or in combination with CT. RESULTS Our search resulted in 92 citations, of which 12 eventually fulfilled the inclusion criteria of our study. Decreased incidence and severity of OM, extended time of occurrence of mucositis, less weight loss and less treatment interruptions were occasionally documented with conventional honey use in the included reviews and meta-analyses. In contrast to conventional honey, manuka honey proved to be weak in improving OM management in the small number of included reviews in our search. CONCLUSION Conventional honey might constitute a highly promising natural product against OM attracting much scientific interest due to its easy accessibility and low financial cost. Hence, the lack of studies with high evidence requires further advanced research to enhance the existing knowledge about the potential value of honey in radiation-induced OM in H-N cancer patients.
Collapse
Affiliation(s)
- Areti Gkantaifi
- Radiotherapy Department, Interbalkan Medical Center, 10 Asclepioustr, 55535Pylaia, Thessaloniki, Greece
| | - Filippo Alongi
- Advanced Radiation Oncology Department, IRCCS SacroCuore Don Calabria,Negrar-Verona, Italy
| | - Emmanouil Vardas
- Clinic of Dentistry Hospital, Dental School, National and Kapodistrian University of Athens, 2 Thivonstr, Goudi, 11527Athens, Greece
| | - Francesco Cuccia
- Advanced Radiation Oncology Department, IRCCS SacroCuore Don Calabria,Negrar-Verona, Italy
| | - Jiannis Hajiioannou
- Department of Otolaryngology, Faculty of Medicine, School of Health Sciences, University of Thessaly, University Hospital of Larissa, Biopolis,41110, Larissa, Greece
| | - Efthymios Kyrodimos
- First Department of Otorhinolaryngology, Head and Neck Surgery, Hippocration Hospital, University of Athens, Athens, Greece
| | - Christos Christopoulos
- Radiation Oncology Department, GroupeHospitalierIntercommunal Le Raincy, Montfermeil, 10 Rue du GeneralLeclerc, 93370 Montfermeil, France
| | - Davide Mauri
- Medical Oncology, University of Ioannina, Ioannina, Greece
| | | | - Nikolaos Trogkanis
- National and Kapodistrian University of Athens, Medical School, 2nd Department of Radiology, Radiotherapy Unit, Attikon University Hospital, Athens, Greece
| | - Nikolaos Tsoukalas
- Oncology Department, General Military Hospital 401, 138 Mesogeion & Katechaki Avenue, 11525, Athens, Greece
| | - George Iliadis
- Radiotherapy Department, Interbalkan Medical Center, 10 Asclepioustr, 55535Pylaia, Thessaloniki, Greece
| | - Maria Tolia
- Department of Radiotherapy, Faculty of Medicine, University of Thessaly, School of Health Sciences, Biopolis,41110, Larisa, Greece
| |
Collapse
|
20
|
Kočović DM, Bajuk-Bogdanović D, Pećinar I, Nedeljković BB, Daković M, Andjus PR. Assessment of cellular and molecular changes in the rat brain after gamma radiation and radioprotection by anisomycin. JOURNAL OF RADIATION RESEARCH 2021; 62:793-803. [PMID: 34062561 PMCID: PMC8438266 DOI: 10.1093/jrr/rrab045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/31/2021] [Indexed: 06/12/2023]
Abstract
The objective of the study was to describe cellular and molecular markers of radioprotection by anisomycin, focusing on the changes in rat brain tissue. Two-month-old Wistar rats were exposed to a 60Co radiation source at a dose of 6 Gy, with or without radioprotection with anisomycin (150 mg/kg) administered subcutaneously 30 min before or 3 or 6 h after irradiation. Survivors were analyzed 30 days after treatment. Astroglial and microglial responses were investigated based on the expression of glial markers assessed with immunohistochemistry, and quantitative changes in brain biomolecules were investigated by Raman microspectroscopy. In addition, blood plasma levels of pro-inflammatory (interleukin 6 and tumor necrosis factor α) and anti-inflammatory (interleukin 10) cytokines were assessed. We found that application of anisomycin either before or after irradiation significantly decreased the expression of the microglial marker Iba-1. We also found an increased intensity of Raman spectral bands related to nucleic acids, as well as an increased level of cytokines when anisomycin was applied after irradiation. This suggests that the radioprotective effects of anisomycin are by decreasing Iba-1 expression and stabilizing genetic material by increasing the level of nucleic acids.
Collapse
Affiliation(s)
- Dušica M Kočović
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Studentski Trg 3, 11 000 Belgrade, Serbia
| | - Danica Bajuk-Bogdanović
- Faculty of Physical Chemistry, University of Belgrade, Studentski Trg 12-16, 11 000 Belgrade, Serbia
| | - Ilinka Pećinar
- Faculty of Agriculture, Department for Agrobotany, University of Belgrade, Nemanjina 6, 11 080 Belgrade, Serbia
| | - Biljana Božić Nedeljković
- Institute for Physiology and Biochemistry ``Jean Giaja'', Faculty of Biology, University of Belgrade, Studentski Trg 3, 11 000 Belgrade, Serbia
| | - Marko Daković
- Faculty of Physical Chemistry, University of Belgrade, Studentski Trg 12-16, 11 000 Belgrade, Serbia
| | - Pavle R Andjus
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Studentski Trg 3, 11 000 Belgrade, Serbia
| |
Collapse
|
21
|
Crook A, De Lima Leite A, Payne T, Bhinderwala F, Woods J, Singh VK, Powers R. Radiation exposure induces cross-species temporal metabolic changes that are mitigated in mice by amifostine. Sci Rep 2021; 11:14004. [PMID: 34234212 PMCID: PMC8263605 DOI: 10.1038/s41598-021-93401-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/24/2021] [Indexed: 11/09/2022] Open
Abstract
Exposure to acute, damaging radiation may occur through a variety of events from cancer therapy and industrial accidents to terrorist attacks and military actions. Our understanding of how to protect individuals and mitigate the effects of radiation injury or Acute Radiation Syndrome (ARS) is still limited. There are only a few Food and Drug Administration-approved therapies for ARS; whereas, amifostine is limited to treating low dose (0.7-6 Gy) radiation poisoning arising from cancer radiotherapy. An early intervention is critical to treat ARS, which necessitates identifying diagnostic biomarkers to quickly characterize radiation exposure. Towards this end, a multiplatform metabolomics study was performed to comprehensively characterize the temporal changes in metabolite levels from mice and non-human primate serum samples following γ-irradiation. The metabolomic signature of amifostine was also evaluated in mice as a model for radioprotection. The NMR and mass spectrometry metabolomics analysis identified 23 dysregulated pathways resulting from the radiation exposure. These metabolomic alterations exhibited distinct trajectories within glucose metabolism, phospholipid biosynthesis, and nucleotide metabolism. A return to baseline levels with amifostine treatment occurred for these pathways within a week of radiation exposure. Together, our data suggests a unique physiological change that is independent of radiation dose or species. Furthermore, a metabolic signature of radioprotection was observed through the use of amifostine prophylaxis of ARS.
Collapse
Affiliation(s)
- Alexandra Crook
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Aline De Lima Leite
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Thomas Payne
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Fatema Bhinderwala
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Jade Woods
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
- Armed Forces Radiobiology Research Institute, USUHS, Bethesda, MD, 20814, USA.
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA.
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA.
| |
Collapse
|
22
|
Choi SH, Lee DY, Kang S, Lee MK, Lee JH, Lee SH, Lee HL, Lee HY, Jeong YIL. Caffeic Acid Phenethyl Ester-Incorporated Radio-Sensitive Nanoparticles of Phenylboronic Acid Pinacol Ester-Conjugated Hyaluronic Acid for Application in Radioprotection. Int J Mol Sci 2021; 22:6347. [PMID: 34198522 PMCID: PMC8231778 DOI: 10.3390/ijms22126347] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/31/2021] [Accepted: 06/08/2021] [Indexed: 12/30/2022] Open
Abstract
We synthesized phenylboronic acid pinacol ester (PBPE)-conjugated hyaluronic acid (HA) via thiobis(ethylamine) (TbEA) linkage (abbreviated as HAsPBPE conjugates) to fabricate the radiosensitive delivery of caffeic acid phenetyl ester (CAPE) and for application in radioprotection. PBPE was primarily conjugated with TbEA and then PBPE-TbEA conjugates were conjugated again with hyaluronic acid using carbodiimide chemistry. CAPE-incorporated nanoparticles of HAsPBPE were fabricated by the nanoprecipitation method and then the organic solvent was removed by dialysis. CAPE-incorporated HAsPBPE nanoparticles have a small particle size of about 80 or 100 nm and they have a spherical shape. When CAPE-incorporated HAsPBPE nanoparticles were irradiated, nanoparticles became swelled or disintegrated and their morphologies were changed. Furthermore, the CAPE release rate from HAsPBPE nanoparticles were increased according to the radiation dose, indicating that CAPE-incorporated HAsPBPE nanoparticles have radio-sensitivity. CAPE and CAPE-incorporated HAsPBPE nanoparticles appropriately prevented radiation-induced cell death and suppressed intracellular accumulation of reactive oxygen species (ROS). CAPE and CAPE-incorporated HAsPBPE nanoparticles efficiently improved survivability of mice from radiation-induced death and reduced apoptotic cell death. We suggest that HAsPBPE nanoparticles are promising candidates for the radio-sensitive delivery of CAPE.
Collapse
Affiliation(s)
- Seon-Hee Choi
- Biomedical R&D Center, Pusan National University Yangsan Hospital, Gyeongnam 50612, Korea; (S.-H.C.); (H.-L.L.)
| | - Dong-Yeon Lee
- Department of Radiation Oncology, Dongnam Institute of Radiological & Medical Sciences, Pusan 46033, Korea;
| | - Sohi Kang
- Biomaterial R&BD Center, Chonnam National University, Gwangju 61186, Korea;
| | - Min-Kyung Lee
- Department of Dental Hygiene, Dong-Eui University, Pusan 47340, Korea;
| | - Jae-Heun Lee
- Department of Radiological Science, Dong-Eui University, Pusan 47340, Korea; (J.-H.L.); (S.-H.L.)
| | - Sang-Heon Lee
- Department of Radiological Science, Dong-Eui University, Pusan 47340, Korea; (J.-H.L.); (S.-H.L.)
| | - Hye-Lim Lee
- Biomedical R&D Center, Pusan National University Yangsan Hospital, Gyeongnam 50612, Korea; (S.-H.C.); (H.-L.L.)
| | - Hyo-Young Lee
- Department of Radiological Science, Dong-Eui University, Pusan 47340, Korea; (J.-H.L.); (S.-H.L.)
| | - Young-IL Jeong
- Biomedical R&D Center, Pusan National University Yangsan Hospital, Gyeongnam 50612, Korea; (S.-H.C.); (H.-L.L.)
| |
Collapse
|
23
|
Jurgensen KJ, Skinner WKJ, Oronsky B, Abrouk ND, Graff AE, Landes RD, Culp WE, Summers TA, Cary LH. RRx-001 Radioprotection: Enhancement of Survival and Hematopoietic Recovery in Gamma-Irradiated Mice. Front Pharmacol 2021; 12:676396. [PMID: 33967816 PMCID: PMC8100686 DOI: 10.3389/fphar.2021.676396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/06/2021] [Indexed: 01/03/2023] Open
Abstract
The present studies evaluate the in vivo prophylactic radioprotective effects of 1-bromoacetyl-3, 3-dinitroazetidine (RRx-001), a phase III anticancer agent that inhibits c-myc and downregulates CD-47, after total body irradiation (TBI), in lethally and sublethally irradiated CD2F1 male mice. A single dose of RRx-001 was administered by intraperitoneal (IP) injection 24 h prior to a lethal or sublethal radiation dose. When irradiated with 9.35 Gy, the dose lethal to 70% of untreated mice at 30 days (LD70/30), only 33% of mice receiving RRx-001 (10 mg/kg) 24 h prior to total body irradiation (TBI) died by day 30, compared to 67% in vehicle-treated mice. The same pretreatment dose of RRx-001 resulted in a significant dose reduction factor of 1.07. In sublethally TBI mice, bone marrow cellularity was increased at day 14 in the RRx-001-treated mice compared to irradiated vehicle-treated animals. In addition, significantly higher numbers of lymphocytes, platelets, percent hematocrit and percent reticulocytes were observed on days 7 and/or 14 in RRx-001-treated mice. These experiments provide proof of principle that systemic administration of RRx-001 prior to TBI significantly improves overall survival and bone marrow regeneration.
Collapse
Affiliation(s)
- Kimberly J Jurgensen
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States.,Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University, Bethesda, MD, United States
| | - William K J Skinner
- Department of Radiation Oncology, Walter Reed National Military Medical Center, Bethesda, MD, United States
| | | | | | - Andrew E Graff
- Department of Radiation Oncology, Walter Reed National Military Medical Center, Bethesda, MD, United States
| | - Reid D Landes
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - William E Culp
- Director, Biomedical Instrumentation Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Thomas A Summers
- Department of Pathology, Uniformed Services University, Bethesda, MD, United States
| | - Lynnette H Cary
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
24
|
Wellenberg A, Weides L, Kurzke J, Hennecke T, Bornhorst J, Crone B, Karst U, Brinkmann V, Fritz G, Honnen S. Use of C. elegans as a 3R-compliant in vivo model for the chemoprevention of cisplatin-induced neurotoxicity. Exp Neurol 2021; 341:113705. [PMID: 33753139 DOI: 10.1016/j.expneurol.2021.113705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023]
Abstract
Anticancer therapeutics can provoke severe side effects that impair the patient's quality of life. A frequent dose-limiting side effect of platinum-based anticancer therapy is neurotoxicity. Its pathophysiology is poorly understood, and effective preventive or therapeutic measures are missing. Therefore, elucidation of the molecular mechanism of platinating drug-induced neurotoxicity and the development of preventive strategies is urgently needed. To this end, we aim to use C. elegans as a 3R-compliant in vivo model. The 3R principles were conceived for animal welfare in science concerning animal experiments, which should be replaced, reduced or refined. We can analytically demonstrate dose-dependent uptake of cisplatin (CisPt) in C. elegans, as well as genotoxic and cytotoxic effects based on DNA adduct formation (i.e., 1,2-GpG intrastrand crosslinks), induction of apoptosis, and developmental toxicity. Measuring the impairment of pharyngeal pumping as a marker of neurotoxicity, we found that especially CisPt reduces the pumping frequency at concentrations where basal and touch-provoked movement were not yet affected. CisPt causes glutathione (GSH) depletion and RNAi-mediated knockdown of the glutamate-cysteine ligase GCS-1 aggravates the CisPt-induced inhibition of pharyngeal pumping. Moreover, N-acetylcysteine (NAC) mitigated CisPt-triggered toxicity, indicating that GSH depletion contributes to the CisPt-induced pharyngeal damage. In addition to NAC, amifostine (WR1065) also protected the pharynx of C. elegans from the toxic effects of CisPt. Measuring pharyngeal activity by the electrophysiological recording of neurotransmission in the pharynx, we confirmed that CisPt is neurotoxic in C. elegans and that NAC is neuroprotective in the nematode. The data support the hypothesis that monitoring the pharyngeal activity of C. elegans is a useful surrogate marker of CisPt-induced neurotoxicity. In addition, a low GSH pool reduces the resistance of neurons to CisPt treatment, and both NAC and WR1065 are capable of attenuating platinum-induced neurotoxicity during post-incubation in C. elegans. Overall, we propose C. elegans as a 3R-compliant in vivo model to study the molecular mechanisms of platinum-induced neurotoxicity and to explore novel neuroprotective therapeutic strategies to alleviate respective side effects of platinum-based cancer therapy.
Collapse
Affiliation(s)
- Anna Wellenberg
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Lea Weides
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Jennifer Kurzke
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| | - Till Hennecke
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| | - Julia Bornhorst
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, D-14558 Nuthetal, Germany; Faculty of Mathematics and Natural Sciences, Food Chemistry, University of Wuppertal, Gaußstr. 20, D-42119 Wuppertal, Germany.
| | - Barbara Crone
- Institute of Inorganic and Analytical Chemistry, University of Muenster, Corrensstraße 30, D-48149 Muenster, Germany.
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Muenster, Corrensstraße 30, D-48149 Muenster, Germany.
| | - Vanessa Brinkmann
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Sebastian Honnen
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| |
Collapse
|
25
|
Antioxidative Stress: Inhibiting Reactive Oxygen Species Production as a Cause of Radioresistance and Chemoresistance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6620306. [PMID: 33628367 PMCID: PMC7884184 DOI: 10.1155/2021/6620306] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/07/2021] [Accepted: 01/30/2021] [Indexed: 02/05/2023]
Abstract
Radiotherapy and chemotherapy are the most effective nonsurgical treatments for cancer treatment. They usually induce regulated cell death by increasing the level of reactive oxygen species (ROS) in tumour cells. However, as intracellular ROS concentration increases, many antioxidant pathways are concurrently upregulated by cancer cells to inhibit ROS production, ultimately leading to drug resistance. Understanding the mechanism of antioxidant stress in tumour cells provides a new research direction for overcoming therapeutic resistance. In this review, we address (1) how radiotherapy and chemotherapy kill tumour cells by increasing the level of ROS, (2) the mechanism by which ROS activate antioxidant pathways and the subsequent cellular mitigation of ROS in radiotherapy and chemotherapy treatments, and (3) the potential research direction for targeted treatment to overcome therapeutic resistance.
Collapse
|
26
|
Sarkar AA, Allyn DM, Delay RJ, Delay ER. Cyclophosphamide-Induced Inflammation of Taste Buds and Cytoprotection by Amifostine. Chem Senses 2021; 46:bjab031. [PMID: 34161570 PMCID: PMC8345827 DOI: 10.1093/chemse/bjab031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Taste buds in the oral cavity have a complex immune system regulating normal functions and inflammatory reactions. Cyclophosphamide (CYP), a chemotherapy drug, has wide-ranging disruptive effects on the taste system including loss of taste function, taste sensory cells, and capacity for taste cell renewal. In bladder epithelium, CYP also induces inflammation. To determine if CYP induces inflammation in taste buds, we used immunohistochemistry to examine tumor necrosis factor alpha (TNF-α) (a proinflammatory cytokine) expression over a 72-hour period. Expression of TNF-α increased in a subset of PLCβ2 labeled (Type II) cells, but not SNAP-25 labeled (Type III) cells, between 8 and 24 h postinjection and declined slowly thereafter. This inflammatory response may play an important role in the disruptive effects of CYP on the taste system. Further, pretreatment with amifostine, a sulfhydryl drug known to protect normal tissues during chemo- or radiation therapy, reduced the amount of CYP-induced TNF-α expression in taste buds, suggesting this drug is capable of protecting normal cells of the taste system from adverse effects of CYP. Amifostine, used as a pretreatment to CYP and possibly other chemotherapy drugs, may offer clinical support for preventing negative side effects of chemotherapy on the taste system.
Collapse
Affiliation(s)
- Anish A Sarkar
- Department of Biology and Vermont Chemosensory Group, University of Vermont, 109 Carrigan Drive, Burlington, VT 05405, USA
| | - David M Allyn
- Department of Biology and Vermont Chemosensory Group, University of Vermont, 109 Carrigan Drive, Burlington, VT 05405, USA
- Program of Biotechnology, School of Engineering and Applied Sciences, University of Pennsylvania, 107 Towne Building, 220 South 33rd Street, Philadelphia, PA 19104-6391, USA
| | - Rona J Delay
- Department of Biology and Vermont Chemosensory Group, University of Vermont, 109 Carrigan Drive, Burlington, VT 05405, USA
| | - Eugene R Delay
- Department of Biology and Vermont Chemosensory Group, University of Vermont, 109 Carrigan Drive, Burlington, VT 05405, USA
| |
Collapse
|
27
|
Azzam P, Mroueh M, Francis M, Daher AA, Zeidan YH. Radiation-induced neuropathies in head and neck cancer: prevention and treatment modalities. Ecancermedicalscience 2020; 14:1133. [PMID: 33281925 PMCID: PMC7685771 DOI: 10.3332/ecancer.2020.1133] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Indexed: 12/24/2022] Open
Abstract
Head and neck cancer (HNC) is the sixth most common human malignancy with a global incidence of 650,000 cases per year. Radiotherapy (RT) is commonly used as an effective therapy to treat tumours as a definitive or adjuvant treatment. Despite the substantial advances in RT contouring and dosage delivery, patients suffer from various radiation-induced complications, among which are toxicities to the nervous tissues in the head and neck area. Radiation-mediated neuropathies manifest as a result of increased oxidative stress-mediated apoptosis, neuroinflammation and altered cellular function in the nervous tissues. Eventually, molecular damage results in the formation of fibrotic tissues leading to susceptible loss of function of numerous neuronal substructures. Neuropathic sequelae following irradiation in the head and neck area include sensorineural hearing loss, alterations in taste and smell functions along with brachial plexopathy, and cranial nerves palsies. Numerous management options are available to relieve radiation-associated neurotoxicities notwithstanding treatment alternatives that remain restricted with limited benefits. In the scope of this review, we discuss the use of variable management and therapeutic modalities to palliate common radiation-induced neuropathies in head and neck cancers.
Collapse
Affiliation(s)
- Patrick Azzam
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Manal Mroueh
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Marina Francis
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Alaa Abou Daher
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Youssef H Zeidan
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| |
Collapse
|
28
|
Fuloria S, Subramaniyan V, Karupiah S, Kumari U, Sathasivam K, Meenakshi DU, Wu YS, Guad RM, Udupa K, Fuloria NK. A Comprehensive Review on Source, Types, Effects, Nanotechnology, Detection, and Therapeutic Management of Reactive Carbonyl Species Associated with Various Chronic Diseases. Antioxidants (Basel) 2020; 9:1075. [PMID: 33147856 PMCID: PMC7692604 DOI: 10.3390/antiox9111075] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
Continuous oxidation of carbohydrates, lipids, and amino acids generate extremely reactive carbonyl species (RCS). Human body comprises some important RCS namely hexanal, acrolein, 4-hydroxy-2-nonenal, methylglyoxal, malondialdehyde, isolevuglandins, and 4-oxo-2- nonenal etc. These RCS damage important cellular components including proteins, nucleic acids, and lipids, which manifests cytotoxicity, mutagenicity, multitude of adducts and crosslinks that are connected to ageing and various chronic diseases like inflammatory disease, atherosclerosis, cerebral ischemia, diabetes, cancer, neurodegenerative diseases and cardiovascular disease. The constant prevalence of RCS in living cells suggests their importance in signal transduction and gene expression. Extensive knowledge of RCS properties, metabolism and relation with metabolic diseases would assist in development of effective approach to prevent numerous chronic diseases. Treatment approaches for RCS associated diseases involve endogenous RCS metabolizers, carbonyl metabolizing enzyme inducers, and RCS scavengers. Limited bioavailability and bio efficacy of RCS sequesters suggest importance of nanoparticles and nanocarriers. Identification of RCS and screening of compounds ability to sequester RCS employ several bioassays and analytical techniques. Present review describes in-depth study of RCS sources, types, properties, identification techniques, therapeutic approaches, nanocarriers, and their role in various diseases. This study will give an idea for therapeutic development to combat the RCS associated chronic diseases.
Collapse
Affiliation(s)
- Shivkanya Fuloria
- Faculty of Pharmacy, AIMST University, Kedah, Bedong 08100, Malaysia;
| | - Vetriselvan Subramaniyan
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Kuala Lumpur 42610, Malaysia; (V.S.); (Y.S.W.)
| | - Sundram Karupiah
- Faculty of Pharmacy, AIMST University, Kedah, Bedong 08100, Malaysia;
| | - Usha Kumari
- Faculty of Medicine, AIMST University, Kedah, Bedong 08100, Malaysia;
| | | | | | - Yuan Seng Wu
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Kuala Lumpur 42610, Malaysia; (V.S.); (Y.S.W.)
| | - Rhanye Mac Guad
- Faculty of Medicine and Health Science, University Malaysia Sabah, Kota Kinabalu 88400, Malaysia;
| | - Kaviraja Udupa
- Department of Neurophysiology, NIMHANS, Bangalore 560029, India;
| | | |
Collapse
|
29
|
Neurotoxicity of antineoplastic drugs: Mechanisms, susceptibility, and neuroprotective strategies. Adv Med Sci 2020; 65:265-285. [PMID: 32361484 DOI: 10.1016/j.advms.2020.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 12/22/2019] [Accepted: 04/13/2020] [Indexed: 02/06/2023]
Abstract
This review summarizes the adverse effects on the central and/or peripheral nervous systems that may occur in response to antineoplastic drugs. In particular, we describe the neurotoxic side effects of the most commonly used drugs, such as platinum compounds, doxorubicin, ifosfamide, 5-fluorouracil, vinca alkaloids, taxanes, methotrexate, bortezomib and thalidomide. Neurotoxicity may result from direct action of compounds on the nervous system or from metabolic alterations produced indirectly by these drugs, and either the central nervous system or the peripheral nervous system, or both, may be affected. The incidence and severity of neurotoxicity are principally related to the dose, to the duration of treatment, and to the dose intensity, though other factors, such as age, concurrent pathologies, and genetic predisposition may enhance the occurrence of side effects. To avoid or reduce the onset and severity of these neurotoxic effects, the use of neuroprotective compounds and/or strategies may be helpful, thereby enhancing the therapeutic effectiveness of antineoplastic drug.
Collapse
|
30
|
Clinical observation on prevention and treatment of radiation-induced parotid-gland injury in patients with nasopharyngeal carcinoma by Compound Sarcandra Glabra. RADIATION MEDICINE AND PROTECTION 2020. [DOI: 10.1016/j.radmp.2020.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
31
|
Jensen SB, Vissink A, Limesand KH, Reyland ME. Salivary Gland Hypofunction and Xerostomia in Head and Neck Radiation Patients. J Natl Cancer Inst Monogr 2020; 2019:5551361. [PMID: 31425600 DOI: 10.1093/jncimonographs/lgz016] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The most manifest long-term consequences of radiation therapy in the head and neck cancer patient are salivary gland hypofunction and a sensation of oral dryness (xerostomia). METHODS This critical review addresses the consequences of radiation injury to salivary gland tissue, the clinical management of salivary gland hypofunction and xerostomia, and current and potential strategies to prevent or reduce radiation injury to salivary gland tissue or restore the function of radiation-injured salivary gland tissue. RESULTS Salivary gland hypofunction and xerostomia have severe implications for oral functioning, maintenance of oral and general health, and quality of life. Significant progress has been made to spare salivary gland function chiefly due to advances in radiation techniques. Other strategies have also been developed, e.g., radioprotectors, identification and preservation/expansion of salivary stem cells by stimulation with cholinergic muscarinic agonists, and application of new lubricating or stimulatory agents, surgical transfer of submandibular glands, and acupuncture. CONCLUSION Many advances to manage salivary gland hypofunction and xerostomia induced by radiation therapy still only offer partial protection since they are often of short duration, lack the protective effects of saliva, or potentially have significant adverse effects. Intensity-modulated radiation therapy (IMRT), and its next step, proton therapy, have the greatest potential as a management strategy for permanently preserving salivary gland function in head and neck cancer patients.Presently, gene transfer to supplement fluid formation and stem cell transfer to increase the regenerative potential in radiation-damaged salivary glands are promising approaches for regaining function and/or regeneration of radiation-damaged salivary gland tissue.
Collapse
Affiliation(s)
- Siri Beier Jensen
- Department of Dentistry and Oral Health, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Arjan Vissink
- Department of Oral and Maxillofacial Surgery, University of Groningen, University Medical Center, Groningen, The Netherlands
| | | | - Mary E Reyland
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
32
|
Barazzuol L, Coppes RP, van Luijk P. Prevention and treatment of radiotherapy-induced side effects. Mol Oncol 2020; 14:1538-1554. [PMID: 32521079 PMCID: PMC7332214 DOI: 10.1002/1878-0261.12750] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 01/10/2023] Open
Abstract
Radiotherapy remains a mainstay of cancer treatment, being used in roughly 50% of patients. The precision with which the radiation dose can be delivered is rapidly improving. This precision allows the more accurate targeting of radiation dose to the tumor and reduces the amount of surrounding normal tissue exposed. Although this often reduces the unwanted side effects of radiotherapy, we still need to further improve patients' quality of life and to escalate radiation doses to tumors when necessary. High-precision radiotherapy forces one to choose which organ or functional organ substructures should be spared. To be able to make such choices, we urgently need to better understand the molecular and physiological mechanisms of normal tissue responses to radiotherapy. Currently, oversimplified approaches using constraints on mean doses, and irradiated volumes of normal tissues are used to plan treatments with minimized risk of radiation side effects. In this review, we discuss the responses of three different normal tissues to radiotherapy: the salivary glands, cardiopulmonary system, and brain. We show that although they may share very similar local cellular processes, they respond very differently through organ-specific, nonlocal mechanisms. We also discuss how a better knowledge of these mechanisms can be used to treat or to prevent the effects of radiotherapy on normal tissue and to optimize radiotherapy delivery.
Collapse
Affiliation(s)
- Lara Barazzuol
- Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Rob P. Coppes
- Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Peter van Luijk
- Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| |
Collapse
|
33
|
Alterations in Tissue Metabolite Profiles with Amifostine-Prophylaxed Mice Exposed to Gamma Radiation. Metabolites 2020; 10:metabo10050211. [PMID: 32455594 PMCID: PMC7281564 DOI: 10.3390/metabo10050211] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/02/2022] Open
Abstract
Acute exposure to high-dose ionizing irradiation has the potential to severely injure the hematopoietic system and its capacity to produce vital blood cells that innately serve to ward off infections and excessive bleeding. Developing a medical radiation countermeasure that can protect individuals from the damaging effects of irradiation remains a significant, unmet need and an area of great public health interest and concern. Despite significant advancements in the field of radiation countermeasure development to find a nontoxic and effective prophylactic agent for acute radiation syndrome, no such drug has yet been approved by the Food and Drug Administration. This study focuses on examining the metabolic corrections elicited by amifostine, a potent radioprotector, on tissues of vital body organs, such as the heart, spleen, and kidney. Our findings indicate that prophylaxis with this drug offers significant protection against potentially lethal radiation injury, in part, by correction of radiation-induced metabolic pathway perturbations.
Collapse
|
34
|
Taberna M, Gil Moncayo F, Jané-Salas E, Antonio M, Arribas L, Vilajosana E, Peralvez Torres E, Mesía R. The Multidisciplinary Team (MDT) Approach and Quality of Care. Front Oncol 2020; 10:85. [PMID: 32266126 PMCID: PMC7100151 DOI: 10.3389/fonc.2020.00085] [Citation(s) in RCA: 206] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 01/16/2020] [Indexed: 12/17/2022] Open
Abstract
The core function of a multidisciplinary team (MDT) is to bring together a group of healthcare professionals from different fields in order to determine patients' treatment plan. Most of head and neck cancer (HNC) units are currently led by MDTs that at least include ENT and maxillofacial surgeons, radiation and medical oncologists. HNC often compromise relevant structures of the upper aerodigestive tract involving functions such as speech, swallowing and breathing, among others. The impairment of these functions can significantly impact patients' quality of life and psychosocial status, and highlights the crucial role of specialized nurses, dietitians, psycho-oncologists, social workers, and onco-geriatricians, among others. Hence, these professionals should be integrated in HNC MDTs. In addition, involving translational research teams should also be considered, as it will help reducing the existing gap between basic research and the daily clinical practice. The aim of this comprehensive review is to assess the role of the different supportive disciplines integrated in an MDT and how they help providing a better care to HNC patients during diagnosis, treatment and follow up.
Collapse
Affiliation(s)
- Miren Taberna
- Medical Oncology Department, Catalan Institute of Oncology (ICO), ONCOBELL, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Francisco Gil Moncayo
- Psicooncology Department, Catalan Institute of Oncology (ICO), IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Enric Jané-Salas
- Department of Odontostomatology, Faculty of Medicine and Health Sciences (Dentistry), University of Barcelona, Barcelona, Spain.,Oral Health and Masticatory System Group (Bellvitge Biomedical Research Institute) IDIBELL, L'Hospitalet de Llobregat, University of Barcelona, Barcelona, Spain
| | - Maite Antonio
- Oncogeriatrics Unit, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Lorena Arribas
- Clinical Nutrition Unit, Catalan Institute of Oncology (ICO), IDIBELL, L'Hospitalet de Llobregat, University of Barcelona, Barcelona, Spain
| | - Esther Vilajosana
- Head and Neck Nurse, Head and Neck Functional Unit, Catalan Institute of Oncology (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Elisabet Peralvez Torres
- Expert SLP in Oncologic Patients, Head of SLP's Department, Atos Medical Spain, Barcelona, Spain
| | - Ricard Mesía
- Medical Oncology Department, Catalan Institute of Oncology (ICO), B-ARGO, Barcelona, Spain
| |
Collapse
|
35
|
King M, Joseph S, Albert A, Thomas TV, Nittala MR, Woods WC, Vijayakumar S, Packianathan S. Use of Amifostine for Cytoprotection during Radiation Therapy: A Review. Oncology 2019; 98:61-80. [PMID: 31846959 DOI: 10.1159/000502979] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 08/19/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Radiation therapy is a cornerstone of the therapeutic modalities used in modern oncology. However, it is sometimes limited in its ability to achieve optimal tumor control by radiation-induced normal tissue toxicity. In delivering radiation therapy, a balance must be achieved between maximizing the dose to the tumor and minimizing any injury to the normal tissues. Amifostine was the first Food and Drug Administration (FDA)-approved clinical radiation protector intended to reduce the impact of radiation on normal tissue, lessening its toxicity and potentially allowing for increased tumor dose/control. Despite being FDA-approved almost 20 years ago, Amifostine has yet to achieve widespread clinical use. SUMMARY A thorough review of Amifostine's development, mechanism of action, and current clinical status were conducted. A brief history of Amifostine is given, from its development at Walter Reid Institute of Research to its approval for clinical use. The mechanism of action of Amifostine is explored. The results of a complete literature review of all prospective randomized trials to date involving the use of Amifostine in radiation therapy are presented. The results are arranged by treatment site and salient findings discussed. Side effects and complications to consider in using Amifostine are reviewed. Key Messages: Amifostine has been explored as a radiation protectant in most radiation treatment sites. Studies have demonstrated efficacy of Amifostine in all treatment sites reviewed, but results are heterogeneous. The heterogeneity of studies looking at Amifostine as a clinical radiation protectant has precluded a definitive answer on its efficacy. Complicating its clinical use is its toxicity and delivery requirements. Amifostine has largely fallen out of use with the advent of intensity modulated radiation therapy (IMRT). However, side effects with IMRT remain a challenge and concern. The use of Amifostine in the IMRT era has been poorly explored and is worthy of future study.
Collapse
Affiliation(s)
- Maurice King
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Sanjay Joseph
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ashley Albert
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Toms V Thomas
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Mary R Nittala
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA,
| | - William C Woods
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Srinivasan Vijayakumar
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Satyaseelan Packianathan
- Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
36
|
Cheema AK, Li Y, Girgis M, Jayatilake M, Simas M, Wise SY, Olabisi AO, Seed TM, Singh VK. Metabolomic studies in tissues of mice treated with amifostine and exposed to gamma-radiation. Sci Rep 2019; 9:15701. [PMID: 31666611 PMCID: PMC6821891 DOI: 10.1038/s41598-019-52120-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/14/2019] [Indexed: 02/07/2023] Open
Abstract
Although multiple radioprotectors are currently being investigated preclinically for efficacy and safety, few studies have investigated concomitant metabolic changes. This study examines the effects of amifostine on the metabolic profiles in tissues of mice exposed to cobalt-60 total-body gamma-radiation. Global metabolomic and lipidomic changes were analyzed using ultra-performance liquid chromatography (UPLC) quadrupole time-of-flight mass spectrometry (QTOF-MS) in bone marrow, jejunum, and lung samples of amifostine-treated and saline-treated control mice. Results demonstrate that radiation exposure leads to tissue specific metabolic responses that were corrected in part by treatment with amifostine in a drug-dose dependent manner. Bone marrow exhibited robust responses to radiation and was also highly responsive to protective effects of amifostine, while jejunum and lung showed only modest changes. Treatment with amifostine at 200 mg/kg prior to irradiation seemed to impart maximum survival benefit, while the lower dose of 50 mg/kg offered only limited survival benefit. These findings show that the administration of amifostine causes metabolic shifts that would provide an overall benefit to radiation injury and underscore the utility of metabolomics and lipidomics to determine the underlying physiological mechanisms involved in the radioprotective efficacy of amifostine. This approach may be helpful in identifying biomarkers for radioprotective efficacy of amifostine and other countermeasures under development.
Collapse
Affiliation(s)
- Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, USA
| | - Yaoxiang Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Michael Girgis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Meth Jayatilake
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Madison Simas
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA
| | - Stephen Y Wise
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA
| | - Ayodele O Olabisi
- Armed Forces Radiobiology Research Institute, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA
| | | | - Vijay K Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA.
- Armed Forces Radiobiology Research Institute, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
37
|
Singh VK, Seed TM. The efficacy and safety of amifostine for the acute radiation syndrome. Expert Opin Drug Saf 2019; 18:1077-1090. [PMID: 31526195 DOI: 10.1080/14740338.2019.1666104] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: A radiation countermeasure that can be used prior to radiation exposure to protect the population from the harmful effects of radiation exposure remains a major unmet medical need and is recognized as an important area for research. Despite substantial advances in the research and development for finding nontoxic, safe, and effective prophylactic countermeasures for the acute radiation syndrome (ARS), no such agent has been approved by the United States Food and Drug Administration (FDA). Area covered: Despite the progress made to improve the effectiveness of amifostine as a radioprotector for ARS, none of the strategies have resolved the issue of its toxicity/side effects. Thus, the FDA has approved amifostine for limited clinical indications, but not for non-clinical uses. This article reviews recent strategies and progress that have been made to move forward this potentially useful countermeasure for ARS. Expert opinion: Although the recent investigations have been promising for fielding safe and effective radiation countermeasures, additional work is needed to improve and advance drug design and delivery strategies to get FDA approval for broadened, non-clinical use of amifostine during a radiological/nuclear scenario.
Collapse
Affiliation(s)
- Vijay K Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda , MD , USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| | | |
Collapse
|
38
|
Sucipto U, Waluyo A, Yona S. Phenomenological study: the experiences of patients with nasopharyngeal cancer after undergoing chemoradiation. BMC Nurs 2019; 18:29. [PMID: 31427891 PMCID: PMC6696816 DOI: 10.1186/s12912-019-0356-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background Chemoradiation is a combination therapy of chemotherapy and radiotherapy. Because chemotherapy is given together with radiotherapy, the side effects are heavier and more severe for some patients. For nasopharyngeal cancer patients, the side effects involve nausea, vomiting, anorexia, diarrhea, mucositis, xerostomia, and tasting and hearing loss, which influence their quality of life. The purpose of this study was to explore the experiences of patients with head and neck cancer undergoing chemoradiation. Methods A Phenomenogical desctipve, involving in depth interviewes was conducted during a 6 month study period. Eleven patiets with nasopharyngeal cancer participated in indepth inteviews Colaizzi’s method was used in analyzing data. The selection of participants in this study using purposive sampling method the inclusion criteria were1) the patient had been diagnosed with head and neck cancer, 2) minimum age of 18 years, and 3 had been received external chemoradiation, minimum 14 times of chmemoradiation. The study was conducted at one out-patientradiotherapy department. at Dharmais Cancer Centre Hospital, Jakarta. Result The findings show three main themes: 1) xerostomia was the main physical complaint, 2) patients had decreased social interaction, and 3) having adequate support from the family was important for patients. Conclusion The findings suggest involving family members when patients are undergoing chemoradiation. Adequate family support is needed to help the patients adapt to the side effects of chemoradiation with the best possible response.
Collapse
Affiliation(s)
- Ucip Sucipto
- Faculty of Nursing Universitas Indonesia, Jalan Prof. Dr Bahder Djohan, Kampus UI, Depok, West Java 16424 Indonesia
| | - Agung Waluyo
- Faculty of Nursing Universitas Indonesia, Jalan Prof. Dr Bahder Djohan, Kampus UI, Depok, West Java 16424 Indonesia
| | - Sri Yona
- Faculty of Nursing Universitas Indonesia, Jalan Prof. Dr Bahder Djohan, Kampus UI, Depok, West Java 16424 Indonesia
| |
Collapse
|
39
|
Clémenson C, Liu W, Bricout D, Soyez-Herkert L, Chargari C, Mondini M, Haddad R, Wang-Zhang X, Benel L, Bloy C, Deutsch E. Preventing Radiation-Induced Injury by Topical Application of an Amifostine Metabolite-Loaded Thermogel. Int J Radiat Oncol Biol Phys 2019; 104:1141-1152. [DOI: 10.1016/j.ijrobp.2019.04.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/01/2019] [Accepted: 04/21/2019] [Indexed: 10/26/2022]
|
40
|
Amini P, Mirtavoos-Mahyari H, Motevaseli E, Shabeeb D, Musa AE, Cheki M, Farhood B, Yahyapour R, Shirazi A, Goushbolagh NA, Najafi M. Mechanisms for Radioprotection by Melatonin; Can it be Used as a Radiation Countermeasure? Curr Mol Pharmacol 2019; 12:2-11. [PMID: 30073934 DOI: 10.2174/1874467211666180802164449] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 06/06/2018] [Accepted: 06/28/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Melatonin is a natural body product that has shown potent antioxidant property against various toxic agents. For more than two decades, the abilities of melatonin as a potent radioprotector against toxic effects of ionizing radiation (IR) have been proved. However, in the recent years, several studies have been conducted to illustrate how melatonin protects normal cells against IR. Studies proposed that melatonin is able to directly neutralize free radicals produced by IR, leading to the production of some low toxic products. DISCUSSION Moreover, melatonin affects several signaling pathways, such as inflammatory responses, antioxidant defense, DNA repair response enzymes, pro-oxidant enzymes etc. Animal studies have confirmed that melatonin is able to alleviate radiation-induced cell death via inhibiting pro-apoptosis and upregulation of anti-apoptosis genes. These properties are very interesting for clinical radiotherapy applications, as well as mitigation of radiation injury in a possible radiation disaster. An interesting property of melatonin is mitochondrial ROS targeting that has been proposed as a strategy for mitigating effects in radiosensitive organs, such as bone marrow, gastrointestinal system and lungs. However, there is a need to prove the mitigatory effects of melatonin in experimental studies. CONCLUSION In this review, we aim to clarify the molecular mechanisms of radioprotective effects of melatonin, as well as possible applications as a radiation countermeasure in accidental exposure or nuclear/radiological disasters.
Collapse
Affiliation(s)
- Peyman Amini
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanifeh Mirtavoos-Mahyari
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Dheyauldeen Shabeeb
- Department of Medical Physics & Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, International Campus, Tehran, Iran.,Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
| | - Ahmed Eleojo Musa
- Department of Medical Physics & Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, International Campus, Tehran, Iran.,Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Cheki
- Department of Radiologic Technology, Faculty of Paramedicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Rasoul Yahyapour
- Department of Medical School, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Alireza Shirazi
- Department of Medical Physics & Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, International Campus, Tehran, Iran
| | - Nouraddin Abdi Goushbolagh
- Department of medical Physics, International Campus, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
41
|
Farhood B, Mortezaee K, Motevaseli E, Mirtavoos-Mahyari H, Shabeeb D, Eleojo Musa A, Sanikhani NS, Najafi M, Ahmadi A. Selenium as an adjuvant for modification of radiation response. J Cell Biochem 2019; 120:18559-18571. [PMID: 31190419 DOI: 10.1002/jcb.29171] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 01/06/2023]
Abstract
Ionizing radiation plays a central role in several medical and industrial purposes. In spite of the beneficial effects of ionizing radiation, there are some concerns related to accidental exposure that could pose a threat to the lives of exposed people. This issue is also very critical for triage of injured people in a possible terror event or nuclear disaster. The most common side effects of ionizing radiation are experienced in cancer patients who had undergone radiotherapy. For complete eradication of tumors, there is a need for high doses of ionizing radiation. However, these high doses lead to severe toxicities in adjacent organs. Management of normal tissue toxicity may be achieved via modulation of radiation responses in both normal and malignant cells. It has been suggested that treatment of patients with some adjuvant agents may be useful for amelioration of radiation toxicity or sensitization of tumor cells. However, there are always some concerns for possible severe toxicities and protection of tumor cells, which in turn affect radiotherapy outcomes. Selenium is a trace element in the body that has shown potent antioxidant and radioprotective effects for many years. Selenium can potently stimulate antioxidant defense of cells, especially via upregulation of glutathione (GSH) level and glutathione peroxidase activity. Some studies in recent years have shown that selenium is able to mitigate radiation toxicity when administered after exposure. These studies suggest that selenium may be a useful radiomitigator for an accidental radiation event. Molecular and cellular studies have revealed that selenium protects different normal cells against radiation, while it may sensitize tumor cells. These differential effects of selenium have also been revealed in some clinical studies. In the present study, we aimed to review the radiomitigative and radioprotective effects of selenium on normal cells/tissues, as well as its radiosensitive effect on cancer cells.
Collapse
Affiliation(s)
- Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanifeh Mirtavoos-Mahyari
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Sadat Sanikhani
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
42
|
Bartolini D, Wang Y, Zhang J, Giustarini D, Rossi R, Wang GY, Torquato P, Townsend DM, Tew KD, Galli F. A seleno-hormetine protects bone marrow hematopoietic cells against ionizing radiation-induced toxicities. PLoS One 2019; 14:e0205626. [PMID: 31034521 PMCID: PMC6488049 DOI: 10.1371/journal.pone.0205626] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 03/28/2019] [Indexed: 01/05/2023] Open
Abstract
2,2'-diselenyldibenzoic acid (DSBA) is a chemical probe produced to explore the pharmacological properties of diphenyldiselenide-derived agents with seleno-hormetic activity undergoing preclinical development. The present study was designed to verify in vivo the drug's properties and to determine mechanistically how these may mediate the protection of tissues against stress conditions, exemplified by ionizing radiation induced damage in mouse bone marrow. In murine bone marrow hematopoietic cells, the drug initiated the activation of the Nrf2 transcription factor resulting in enhanced expression of downstream stress response genes. This type of response was confirmed in human liver cells and included enhanced expression of glutathione S-transferases (GST), important in the metabolism and pharmacological function of seleno-compounds. In C57 BL/6 mice, DSBA prevented the suppression of bone marrow hematopoietic cells caused by ionizing radiation exposure. Such in vivo prevention effects were associated with Nrf2 pathway activation in both bone marrow cells and liver tissue. These findings demonstrated for the first time the pharmacological properties of DSBA in vivo, suggesting a practical application for this type of Se-hormetic molecules as a radioprotective and/or prevention agents in cancer treatments.
Collapse
Affiliation(s)
- Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
- * E-mail:
| | - Yanzhong Wang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States of America
| | - Jie Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States of America
| | - Daniela Giustarini
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Ranieri Rossi
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Gavin Y. Wang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States of America
| | - Pierangelo Torquato
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Danyelle M. Townsend
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States of America
| | - Kenneth D. Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States of America
| | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
43
|
Rim CH, Koun S, Park HC, Lee S, Kim CY. Radioprotective effects of mistletoe extract in zebrafish embryos in vivo. Int J Radiat Biol 2019; 95:1150-1159. [PMID: 30836032 DOI: 10.1080/09553002.2019.1590661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Radioprotectors can enhance the efficacy of cancer radiotherapy, but their clinical use remains uncommon. The present study aimed to assess the radioprotective potential of mistletoe extract (commercial name: Abnoba Viscum), a well-known complementary cancer medicine, in zebrafish larvae. Materials and methods: Wild-type AB zebrafish embryos at 4 h-post-fertilization were exposed to 5 Gy 9-MeV electron beam irradiation after being treated for 1 h with 4 mMl/L amifostine or 0.2 mg/ml Abnoba Viscum A, F, M, or Q. Primary endpoints were abnormality-free survival and abnormality-free rates among survivors at 5 days-post-fertilization. Results: The crude abnormality-free survival rates were 33.7%, 49.0%, 38.8%, 43.9%, 38.1%, and 52.6%, whereas abnormality-free rates among survivors were 36.4%, 49.6%, 37.8%, 45.6%, 52.0%, and 62.8% for the control (with no pharmacologic treatment), amifostine, Abnoba Viscum A, F, M, and Q groups, respectively. Abnormality-free survival rates in the amifostine and Abnoba Viscum Q groups were significantly different from those in the control (p = .040 and .012, respectively), with an odds ratio (OR) of 1.90 [95% confidence interval (CI): 1.03-3.51] and 2.20 (95% CI: 1.19-4.08), respectively. Abnormality-free rates among survivors in the amifostine and Abnoba Viscum M and Q groups were significantly different from those in the control group (p = .048, .042, and <.001, respectively), with an OR of 1.79 (95% CI: 1.00-3.20), 1.82 (95% CI: 1.02-3.26), and 2.98 (1.67-5.33), respectively. Conclusion: Abnoba Viscum Q has at least a similar radioprotective effect to that of amifostine. Mistletoe extracts have been clinically applied for a long time and their effectiveness and feasibility have been verified. Abnoba Viscum Q might be a new candidate radioprotectant to enhance cancer radiotherapy efficacy.
Collapse
Affiliation(s)
- Chai Hong Rim
- a Department of Radiation Oncology, Korea University Ansan Hospital , Gyeonggido , Republic of Korea
| | - Soonil Koun
- b Biomedical Research Center Korea University Ansan Hospital , Gyeonggido , Republic of Korea
| | - Hae-Chul Park
- c Laboratory of Neurodevelopmental Genetics, Korea University Graduate School of Medicine , Seoul , Republic of Korea
| | - Suk Lee
- d Department of Radiation Oncology, Korea University Anam Hospital , Seoul , Republic of Korea
| | - Chul Yong Kim
- d Department of Radiation Oncology, Korea University Anam Hospital , Seoul , Republic of Korea
| |
Collapse
|
44
|
Barbosa SCM, Pereira VBM, Wong DVT, Santana APM, Lucetti LT, Carvalho LL, Barbosa CRN, Callado RB, Silva CAA, Lopes CDH, Brito GAC, Alencar NMN, Lima-Júnior RCP. Amifostine reduces inflammation and protects against 5-fluorouracil-induced oral mucositis and hyposalivation. ACTA ACUST UNITED AC 2019; 52:e8251. [PMID: 30810625 PMCID: PMC6393848 DOI: 10.1590/1414-431x20188251] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 11/23/2018] [Indexed: 01/30/2023]
Abstract
Oral mucositis (OM) is a common and dose-limiting side effect of cancer treatment, including 5-fluorouracil (5-FU) and radiotherapy. The efficacy of the therapeutic measures to prevent OM is limited and disease prevention is not fully observable. Amifostine is a cytoprotective agent with a described anti-inflammatory potential. It is clinically used to reduce radiotherapy and chemotherapy-associated xerostomia. This study investigated the protective effect of amifostine on an experimental model of OM. Hamsters were divided into six groups: saline control group (5 mL/kg), mechanical trauma (scratches) of the right cheek pouch; 5-FU (60 and 40 mg/kg, ip, respectively, administered on days 1 and 2); amifostine (12.5, 25, or 50 mg/kg) + 5-FU + scratches. Salivation rate was assessed and the animals were euthanized on day 10 for the analysis of macroscopic and microscopic injury by scores. Tissue samples were harvested for the measurement of neutrophil infiltration and detection of inflammatory markers by ELISA and immunohistochemistry. 5-FU induced pronounced hyposalivation, which was prevented by amifostine (P<0.05). In addition, 5-FU injection caused pronounced tissue injury accompanied by increased neutrophil accumulation, tumor necrosis factor-alpha (TNF-α), and interleukin-1 beta (IL-1β) tissue levels, and positive immunostaining for TNF-α, IL-1β, and inducible nitric oxide synthase (iNOS). Interestingly, amifostine prevented the inflammatory reaction and consequently improved macroscopic and microscopic damage (P<0.05 vs 5-FU group). Amifostine reduced inflammation and protected against 5-FU-associated oral mucositis and hyposalivation.
Collapse
Affiliation(s)
- S C M Barbosa
- Departamento de Fisiologia e Famacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - V B M Pereira
- Departamento de Fisiologia e Famacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - D V T Wong
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - A P M Santana
- Departamento de Fisiologia e Famacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - L T Lucetti
- Departamento de Fisiologia e Famacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - L L Carvalho
- Departamento de Fisiologia e Famacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - C R N Barbosa
- Departamento de Fisiologia e Famacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - R B Callado
- Departamento de Fisiologia e Famacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - C A A Silva
- Departamento de Fisiologia e Famacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - C D H Lopes
- Departamento de Fisiologia e Famacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - G A C Brito
- Departamento de Morfologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - N M N Alencar
- Departamento de Fisiologia e Famacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| | - R C P Lima-Júnior
- Departamento de Fisiologia e Famacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, Brasil
| |
Collapse
|
45
|
Reactive Oxygen Species Drive Epigenetic Changes in Radiation-Induced Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4278658. [PMID: 30881591 PMCID: PMC6381575 DOI: 10.1155/2019/4278658] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/06/2018] [Accepted: 12/12/2018] [Indexed: 12/14/2022]
Abstract
Radiation-induced fibrosis (RIF) develops months to years after initial radiation exposure. RIF occurs when normal fibroblasts differentiate into myofibroblasts and lay down aberrant amounts of extracellular matrix proteins. One of the main drivers for developing RIF is reactive oxygen species (ROS) generated immediately after radiation exposure. Generation of ROS is known to induce epigenetic changes and cause differentiation of fibroblasts to myofibroblasts. Several antioxidant compounds have been shown to prevent radiation-induced epigenetic changes and the development of RIF. Therefore, reviewing the ROS-linked epigenetic changes in irradiated fibroblast cells is essential to understand the development and prevention of RIF.
Collapse
|
46
|
Münstedt K, Momm F, Hübner J. Honey in the management of side effects of radiotherapy- or radio/chemotherapy-induced oral mucositis. A systematic review. Complement Ther Clin Pract 2019; 34:145-152. [DOI: 10.1016/j.ctcp.2018.11.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 11/29/2022]
|
47
|
Tsoutsou P, Montay-Gruel P, Vozenin MC. The Era of Modern Radiation Therapy: Innovations to Spare Normal Tissues. Radiat Oncol 2019. [DOI: 10.1007/978-3-319-52619-5_70-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
48
|
Lee MG, Freeman AR, Roos DE, Milner AD, Borg MF. Randomized double-blind trial of amifostine versus placebo for radiation-induced xerostomia in patients with head and neck cancer. J Med Imaging Radiat Oncol 2018; 63:142-150. [PMID: 30461207 DOI: 10.1111/1754-9485.12833] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/17/2018] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The role of the radioprotector amifostine in ameliorating radiotherapy side effects in head and neck squamous cell carcinoma (HNSCC) is controversial. This trial aimed to determine whether pretreatment with amifostine reduced the incidence of Radiation Therapy Oncology Group grade ≥2 acute and late xerostomia in patients receiving definitive or adjuvant radiotherapy for HNSCC, without reducing tumour control or survival. METHODS Between 14 September 2001 and 8 November 2004, 44 Royal Adelaide Hospital patients were randomized double-blind to receive amifostine (200 mg/m2 IV) or placebo (normal saline IV) 5 days/week, prior to standard radiotherapy (60-70 Gy), each having ≥75% of the parotids treated to ≥40 Gy. Side effects were assessed weekly during treatment, at 3 and 5 months after radiotherapy, then every 6 months until disease progression or death. RESULTS The accrual target was 200 patients over 4-5 years, but the trial closed prematurely when only 44 patients had been randomized after 3 years. Of 41 evaluable patients, 80% (16/20) in the amifostine arm had grade ≥2 acute radiation salivary toxicity versus 76% (16/21) in the placebo arm (P = 1.00). The rate of grade ≥2 late radiation salivary toxicity at 12 months was 66% in the amifostine arm and 82% in the placebo arm (estimated hazard ratio 1.61, 95% confidence interval 0.74-3.49, P = 0.22). Other toxicities tended to be worse in the amifostine arm: acute grade 3-4 skin 35% vs 5% and mucous membrane 40% vs 5%; grade ≥2 vomiting 35% vs 5%, hypocalcaemia 25% vs 5% and fatigue 85% vs 33%, with only the latter retaining statistical significance after adjusting for multiple comparisons. There were no significant differences in failure-free (P = 0.70) or overall survival (P = 0.86), with estimated 4-year rates of 48% vs 54% and 49% vs 59% for the amifostine vs placebo arms respectively. CONCLUSION There was no clear evidence that pretreatment with amifostine made any difference to the incidence of grade ≥2 acute or late xerostomia. Other toxicity tended to be more severe with amifostine. There was no effect on failure-free or overall survival. Acknowledging the low statistical power, these results do not support the use of IV amifostine pre-radiotherapy in HNSCC.
Collapse
Affiliation(s)
- Maverick Gl Lee
- Department of Radiation Oncology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | | | - Daniel E Roos
- Department of Radiation Oncology, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | | | - Martin F Borg
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia.,Formerly of GenesisCare, Adelaide, South Australia, Australia
| |
Collapse
|
49
|
Farhood B, Goradel NH, Mortezaee K, Khanlarkhani N, Salehi E, Nashtaei MS, Mirtavoos-Mahyari H, Motevaseli E, Shabeeb D, Musa AE, Najafi M. Melatonin as an adjuvant in radiotherapy for radioprotection and radiosensitization. Clin Transl Oncol 2018; 21:268-279. [PMID: 30136132 DOI: 10.1007/s12094-018-1934-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022]
Abstract
It is estimated that more than half of cancer patients undergo radiotherapy during the course of their treatment. Despite its beneficial therapeutic effects on tumor cells, exposure to high doses of ionizing radiation (IR) is associated with several side effects. Although improvements in radiotherapy techniques and instruments could reduce these side effects, there are still important concerns for cancer patients. For several years, scientists have been trying to modulate tumor and normal tissue responses to IR, leading to an increase in therapeutic ratio. So far, several types of radioprotectors and radiosensitizers have been investigated in experimental studies. However, high toxicity of chemical sensitizers or possible tumor protection by radioprotectors creates a doubt for their clinical applications. On the other hand, the protective effects of these radioprotectors or sensitizer effects of radiosensitizers may limit some type of cancers. Hence, the development of some radioprotectors without any protective effect on tumor cells or low toxic radiosensitizers can help improve therapeutic ratio with less side effects. Melatonin as a natural body hormone is a potent antioxidant and anti-inflammatory agent that shows some anti-cancer properties. It is able to neutralize different types of free radicals produced by IR or pro-oxidant enzymes which are activated following exposure to IR and plays a key role in the protection of normal tissues. In addition, melatonin has shown the ability to inhibit long-term changes in inflammatory responses at different levels, thereby ameliorating late side effects of radiotherapy. Fortunately, in contrast to classic antioxidants, some in vitro studies have revealed that melatonin has a potent anti-tumor activity when used alongside irradiation. However, the mechanisms of its radiosensitive effect remain to be elucidated. Studies suggested that the activation of pro-apoptosis gene, such as p53, changes in the metabolism of tumor cells, suppression of DNA repair responses as well as changes in biosynthesis of estrogen in breast cancer cells are involved in this process. In this review, we describe the molecular mechanisms for radioprotection and radiosensitizer effects of melatonin. Furthermore, some other proposed mechanisms that may be involved are presented.
Collapse
Affiliation(s)
- B Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - N H Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - K Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - N Khanlarkhani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - E Salehi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - M S Nashtaei
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Infertility Department, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - H Mirtavoos-Mahyari
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - E Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - D Shabeeb
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, International Campus, Tehran, Iran.,Department of Physiology, College of Medicine, University of Misan, Amarah, Iraq
| | - A E Musa
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, International Campus, Tehran, Iran.,Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| | - M Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
50
|
Yang C, Tang H, Wang L, Peng R, Bai F, Shan Y, Yu Z, Zhou P, Cong Y. Dimethyl Sulfoxide Prevents Radiation-Induced Oral Mucositis Through Facilitating DNA Double-Strand Break Repair in Epithelial Stem Cells. Int J Radiat Oncol Biol Phys 2018; 102:1577-1589. [PMID: 30092334 DOI: 10.1016/j.ijrobp.2018.07.2010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 07/20/2018] [Accepted: 07/23/2018] [Indexed: 11/25/2022]
Abstract
PURPOSE Oral mucositis is one of the most prevalent side effects in patients undergoing radiation therapy for head and neck cancers. Current therapeutic agents such as palifermin recombinant human keratinocyte growth factor and amifostine do not efficiently or fully prevent mucositis. Dimethyl sulfoxide (DMSO), a free-radical scavenger, has shown therapeutic benefits in many preclinical and clinical studies. This study aimed to investigate the efficacy of DMSO in a clinically relevant mouse model of acute, radiation-induced oral mucositis. METHODS AND MATERIALS Oral mucositis was induced by a high single and fractioned irradiation of the head and neck area in C57BL/6J mice, and the effects of DMSO (by intraperitoneal injection) were assessed by macroscopic and histopathological examination. Epithelial stem and progenitor cells were analyzed by immunohistochemical staining of p63 and Ki-67, and DNA double-strand breaks (DSBs) were visualized by immunofluorescence detection of γ-H2AX. Tumor xenograft was obtained using CAL-27 cells. RESULTS Pretreatment with DMSO protected the oral mucosa from severe acute radiation injury, reduced the extent of radiation-induced weight loss, and had no significant effects on tumor weight in irradiated or nonirradiated xenograft mice. Furthermore, the efficacy of DMSO was superior to that of recombinant human keratinocyte growth factor and amifostine. DMSO treatment prevented the loss of proliferative lingual epithelial stem and progenitor cells upon irradiation. More interestingly, the average levels of γ-H2AX foci were significantly decreased in p63-positive epithelial stem cells at 6 hours, but not at 2 hours, after irradiation, indicating that DMSO facilitated DNA DSB repair rather than suppressing the indirect action of irradiation. CONCLUSIONS DMSO prevents the loss of proliferative lingual epithelial stem and progenitor cells upon irradiation by facilitating DNA DSB repair, thereby protecting against radiation-induced mucositis without tumor protection. Given its high efficacy and low toxicity, DMSO could be a potential treatment option to prevent radiation-induced oral mucositis.
Collapse
Affiliation(s)
- Chao Yang
- School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, Jiangsu, PR China; Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology, Beijing, PR China
| | - Hongwei Tang
- Department of Gastroenterology, Hospital of Tsinghua Changgung, Beijing, PR China
| | - Limei Wang
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology, Beijing, PR China
| | - Renjun Peng
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology, Beijing, PR China
| | - Fan Bai
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology, Beijing, PR China
| | - Yajun Shan
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology, Beijing, PR China
| | - Zuyin Yu
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology, Beijing, PR China
| | - Pingkun Zhou
- School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, Jiangsu, PR China; Department of Radiation Toxicology and Oncology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing, PR China.
| | - Yuwen Cong
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology, Beijing, PR China.
| |
Collapse
|