• Users Online: 9837
  • Home
  • Print this page
  • Email this page
Home About us Editorial board Ahead of print Current issue Search Archives Submit article Instructions Subscribe Contacts Login 


 
 Table of Contents  
REVIEW ARTICLE
Year : 2021  |  Volume : 7  |  Issue : 1  |  Page : 1-6

Role of radiosensitizers, radioprotectors, and radiation mitigators in radiation therapy


Department of Oral Medicine and Radiology, Rajasthan Dental College and Hospital, Jaipur, Rajasthan, India

Date of Submission28-Aug-2020
Date of Acceptance14-Jan-2021
Date of Web Publication12-Jul-2021

Correspondence Address:
Dr. Kanica Sharma
Vill- Bishangarh, P.O. - Bhogpur, Dehradun, Uttarakhand
India
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/ijohr.ijohr_23_20

Rights and Permissions
  Abstract 

Radiation is used in the treatment of a broad range of malignancies. Exposure of normal tissues to radiation may result in both acute and chronic toxicities that can result in an inability to deliver the intended therapy, a range of symptoms, and a decrease in the quality of life. Radiation therapy kills cancer cells by damaging their deoxyribonucleic acid. There are different types of radiation therapy for different malignancies. Radiation therapy can also deteriorate the normal cells, leading to side effects. A radiation countermeasure that can be used before 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. There are certain compounds that act to increase the radiosensitivity of tumor cells or to protect the normal cells from the effect of radiation termed as radiosensitizers and radioprotectors. Other agents termed mitigators may be used to minimize toxicity even after radiation has been delivered. The aim of this article is to critically review the available compounds used as radiosensitizers, radioprotectors, and mitigators for different types of cancers.

Keywords: Radiation therapy, radioprotector and radiation mitigators, radiosensitizers


How to cite this article:
Sharma K, Patil N, Sareen M, Tyagi N. Role of radiosensitizers, radioprotectors, and radiation mitigators in radiation therapy. Indian J Oral Health Res 2021;7:1-6

How to cite this URL:
Sharma K, Patil N, Sareen M, Tyagi N. Role of radiosensitizers, radioprotectors, and radiation mitigators in radiation therapy. Indian J Oral Health Res [serial online] 2021 [cited 2024 Mar 29];7:1-6. Available from: https://www.ijohr.org/text.asp?2021/7/1/1/321117


  Introduction Top


Radiation therapy is one of the most effective tools against the treatment of cancer. High doses of radiations are used in the radiation therapy to halt growth of tumor. Ionizing radiation (IR), such as X-rays and gamma-rays, is commonly used for the treatment of cancer because it has the ability to pass through the tissues and can break chemical bonds and help in the removal of electrons from the atoms to get ionized. The ionized ions as a result damage cancer cell. Cancer cells are not killed immediately by IR; in fact, substantial time is required for killing of cancer cells.[1]

Local tumor failure is the cause of 40%–60% of cancer deaths and may occur in 60%–80% of cancer patients at the time of death. Efforts to improve the therapeutic ratio have resulted in the development of certain compounds that act to increase the radiosensitivity of tumor cells or to protect the normal cells from the effects of radiation.[2]

Deoxyribonucleic acid (DNA) lesions may result from direct damage, following the interaction of photons or ionizing particles with DNA itself, or indirect damage, which occurs through the generation of reactive-oxygen species (ROS), particularly hydroxyl radicals formed by the radiolysis of water, which may then react with DNA [Figure 1].
Figure 1: Simplified representation of cellular to deoxyribonucleic acid damage lesions induced by ionizing radiation. ROS: Reactive oxygen species, SSB: Single-strand break, DSB: Double-strand break

Click here to view


Based on the intimate relationship between tumors and their normal host tissues and surrounding critical structures and the need to irradiate clinically uninvolved normal tissue margins that are potentially contaminated with microscopic disease, it is anticipated that normal tissue toxicity will remain a concern for therapeutic radiation. An alternative mechanism to reduce normal tissue toxicity is the use of radiation modifiers/protectors, agents that when present before or shortly after radiation exposure alter the response of normal tissues to irradiation. This approach has also been viewed as an attractive countermeasure for possible nuclear/radiological terrorism. To be useful in the radiotherapy clinic, radioprotectors should ideally have several characteristics that relate to the ability of the agent to improve the therapeutic ratio.[3]


  Classification of Agents Top


Radiosensitizers are the agents that sensitize the tumor cells to radiation. These compounds apparently promote fixation of the free radicals produced by radiation damage at the molecular level. The mechanism of action is similar to the oxygen effect, in which biochemical reactions in the damaged molecules prevent repair of the cellular radiation damage. Free radicals such as OH + are captured by the electron affinity of the radiosensitizers, rendering the molecules incapable of repair.[4]

Radioprotectors are the compounds that are designed to reduce the damage in the normal tissues caused by radiation. These compounds are often antioxidants and must be present before or at the time of radiation for effectiveness.[3]

Agents delivered at the time of irradiation or after irradiation are complete, but prior to the manifestation of normal tissue toxicity are described as mitigators of normal tissue injury.[3]

With the increasing demand for better patient care globally, a lot of research is ongoing in the field of oral oncology regarding radio sensitizers and radioprotectors. However, the literature is scanty regarding the same. Hence, this article tries to discuss the various aspects of radio protectors, radiosensitizers, and mitigators such as mechanism of action, uses, and side effects of these agents.


  Radiosensitizers Top


A radiosensitizer is a drug that makes tumor cells more sensitive to radiation therapy. These compounds apparently promote fixation of the free radicals produced by radiation damage at the molecular level. Radiotherapy commonly affects DNA; mainly, it leads to DNA double strand break (DSB). Hence, to target clinically developed DNA DSB repair pathways, many radio-sensitizing agents have been formulated.[1]

Radiosensitizers are compounds that sensitize the tumor cells to radiation during radiotherapy.[2] A list of radio-sensitizing agents are given in [Table 1].
Table 1: List of various Radiosensitizers

Click here to view



  Hyperbaric Oxygen Top


Hyperbaric oxygen (HBO) therapy is the inhalation of 100% oxygen at elevated pressure > 1.5 atmospheres absolute (ATA; 150 kPa), typically 2–3 ATA (200–300 kPa). The hyperbaric chamber is the medical tool that provides those conditions to apply very high doses of oxygen in amounts that cannot be reached by any other means.[5] The physiological effects of HBO include short-term effects such as vasoconstriction and enhanced oxygen delivery, reduction of edema, and phagocytises activation, and it has an anti-inflammatory effect. Long-term effects are neovascularization, osteogenesis as well as stimulation of collagen production by fibroblasts. The clinical results are, therefore, wound healing and recovery of radiation-injured tissue.[6]

Most tumors contain nutrient- and oxygen-deprived compartments. Sterilization of hypoxic tumor cells requires a three times higher radiation dose than for cells at normal oxygen tension. HBO therapy is an effective approach to cope with the phenomenon of hypoxia by increasing the oxygen load of the tumor, and there with to enhance the response to irradiation.[7]


  Mechanism of Action Top


Oxygen is known to increase the radiosensitivity of cells. The reactions of oxygen with aqueous as well as organic-free radicals induced by IR s may lead to the production of very toxic and relatively stable peroxy radicals and hydrogen peroxide resulting in the damage to bio molecules and structures. Therefore, the simplest approach to enhance the radio sensitivity of hypoxic tumor cells would be to increase the oxygen tension in the tumor.[8]

Subsequent studies showed an increase in the 5-year survival of patients with cancers of uterine cervix and head and neck. HBO has been observed to be effective in relatively small tumors, whereas the advanced tumors do not show an increased radio sensitization.[2]


  Carbogen Top


The notion of improving tumor oxygenation by breathing highly oxygenated air has been revived recently by experiments in which participants breathe carbogen, a mixture of 95% oxygen and 5% carbon dioxide that does not produce vasoconstriction associated with breathing 100% oxygen. Breathing carbogen at atmospheric pressure is an attempt to overcome chronic (diffusion limited) hypoxia through much simpler means than the use of hyperbaric chambers.[9]


  Nicotinamide Top


Nicotinamide is the amide of vitamin B3. Acute hypoxia within tumors arises from intermittent closure of blood vessels, resulting in fluctuations in the tumor microcirculation. Nicotinamide overcomes acute hypoxia by reducing these changes in the microcirculation.

Furthermore, when nicotinamide is combined with carbogen, additional tumor sensitivity to radiation has been demonstrated, with overall enhancement ratios of between 1.8 and 2.1 in animal models using clinically relevant dose schedules of 2 Gy day-'. It has therefore been proposed that the combination of carbogen and nicotinamide provides the optimal means of overcoming tumor hypoxia.[10]

Dosage administered in the tablet form to patients with advanced head and neck and non-small cell lung carcinomas. A standard 6 g dose given regardless of body weight after an overnight fast and at least 30 min before breakfast.[11]

After platinum-containing compounds, ruthenium-based complexes represent the group of metallocomplexes that have been most heavily studied for anticancer activity. The cellular mechanism of action for a representative molecule from this class was found to be G1 arrest accompanied by activation of p53/p21 DNA damage signalling pathways 72, and the majority of complexes induce high levels of apoptosis at low micromolar doses.


  Radioprotectors Top


Radioprotectors are the agents that are used to reduce the deterioration of normal cells during irradiation. Antioxidants may also act as radioprotector, and they must be present before or at the time of radiation for effectiveness.[1] Radioprotectors are the chemical compounds that protect the nontumor (normal) cells from radiation during radiotherapy.[2]

A list of radioprotectors is given in [Table 2].
Table 2: List of various Radioprotectors

Click here to view



  Amifostine: A Radioprotector in Use Clinically Top


Amifostine is a phosphorothioate that is not taken into cells until it is dephosphorylated by alkaline phosphatase.[12] Once dephosphorylated the agent freely diffuses into cells and can act as a free radical scavenger. Amifostine has been shown to concentrate more rapidly in normal tissues than in tumor tissues in studies of tumor-bearing animals, which is thought to result from several factors, including the effects of tumor blood flow, the acidosis of tumors, and the lower expression of alkaline phosphatase.[13]


  Mechanism of Action Top


It is actually a pro-drug, which cannot readily permeate cell membranes. Amifostine on administration undergoes metabolism and gets converted into WR-1065, which can readily permeate the cell membrane[14] [Figure 2].
Figure 2: Mechanism of action of amifostine as radioprotector

Click here to view


Other functions of Amifostine-cytotoxic effects of chemotherapeutic agents, nephrotoxicity, ototoxicity and neuropathy associated with cisplatin, hematologic toxicity with cyclophosphamide.[15]

Side effects of this agent include nausea, vomiting, and hypotension. Clinical trials with patients receiving head and neck, thoracic, and pelvic radiation therapy are ongoing.[2]


  Nitroxides Top


Nitroxides are among the most promising agents for future use as radiation protectors. Although a number of these agents are useful in the laboratory as radiation protectors, not all have the requisite characteristics that allow them to be used clinically.[16]

Laboratory studies have shown that stable nitroxide-free radicals and their one-electron reduction products, hydroxyl amines, are recycling antioxidants that protect cells when exposed to oxidative stress, including superoxide and hydrogen peroxide. Similarly, preclinical studies have shown that the oxidized form of a nitroxide is a radio protector in both in vitro (cell survival) and in vivo (lethal total body radiation) models. Although the hydroxylamine exhibits antioxidant activity, it is incapable of protecting against radiation damage. The lead compound from this class for radioprotection is Tempol.[2]

Tempol protects against radiation-induced damage to salivary glands and does not alter tumor growth after irradiation suggesting that delivery of the agent before irradiation would not alter tumor control. Tempol also protected salivary glands from radiation-induced damage, but did not protect the tumor tissue, suggesting that delivery of the agent before irradiation would not alter tumor control.[2]


  Antioxidants as Radio Protectors Top


Antioxidant agents, such as Vitamin A, C, and E, offer protective properties against radiation because deleterious effects of radiation mimic the oxidative damage associated with active oxygen toxicity. Selenium (as Na2SeO3) and Vitamin E have been reported to act alone and in an additive form as radio-protective and chemo-preventive agents.[17] A number of trials have been performed with antioxidants delivered during the course of radiotherapy, with the goal of reducing normal tissue toxicity, in many instances with promising results. For example, antioxidants have been delivered concurrently during the course of radiotherapy to reduce xerostomia, mucositis, pulmonary fibrosis, cystitis, and alopecia.[2]


  Vitamin E Top


Vitamin E (alpha tocopherol) and related analogs are nutraceuticals that can scavenge singlet oxygen and superoxide anion radicals A water-soluble Vitamin E derivative, tocopherol monoglucoside (TMG), was reported to be very effective in protecting DNA both in vitro and also in mice after oral or intraperitoneal administration against gamma irradiation. TMG ([2-a-D-glucopyranosyl] methyl-2, 5, 7, 8-tetramethylchroman-6-ol), was efficient when administered immediately after radiation exposure at a dose of 600 mg/kg. The treatment decreased radiation-induced abnormal metaphases in mouse bone marrow chromosomes and reduced the frequency of micronucleated erythrocytes at 24 h after exposure to radiation.[17]


  Herbal Radio-Protectors Top


The majority of synthetic radio protectors have limited clinical application owing to their side effects and inherent toxicity. Therefore, natural herbal radio protectors, such as Ginkgo biloba, Chagas mushroom, green tea (polyphenols), dithiolthiones, Panax ginseng, Shigoka extract, and Spirulina platensis, have become an important alternative. The proposed radio-protective efficacy of plant products is due to their containing a large number of active constituents, such as antioxidants, immunostimulants, and compounds with antimicrobial activity.[18]

The doses of herbal preparations that were effective in protecting against irradiation were significantly lower than the toxic dose of synthetic compounds, and this is one of the major advantages of these agents compared to synthetic compounds. The use of plants as radio protectors has some disadvantages, such as low to mild efficacy and a short protective time-window (in most cases 30 min to 2 h before irradiation).[2]


  Superoxide Dimutase Top


IR results in the formation of superoxide radicals that are highly reactive and potentially damaging to cells. Superoxide dismutases (SOD) is an enzyme that is naturally present in human cells. It catalyzes the conversion of superoxide to oxygen and hydrogen peroxide and functions as an antioxidant during the normal conditions and after radiation. SOD as a radioprotector has used gene therapy to increase the levels of SOD in tissues to be irradiated to prevent or decrease radiation-induced mucositis, esophagitis, pneumonitis, and fibrosis in animal models.[19],[20]


  Melatonin Top


Melatonin (N-acetyl-5-methyloxytryptamine), a pineal gland hormone involved in regulating the neuroendocrine axis, is a highly efficient free-radical scavenger and antioxidant. Melatonin has been reported to be a direct free-radical scavenger and an indirect antioxidant through its stimulative effects on antioxidant enzymes, such as SOD, GSH-Px, glutathione reductase, and catalase.[17]

The use of melatonin as a radiation sensitizer for tumor cells and as a radioprotector for normal cells was tested clinically in a phase II Radiation Therapy Oncology Group trial. In that study, patients were randomized to either morning-or night-time high-dose melatonin during radiotherapy. Melatonin was continued after radiotherapy until progression or until 6 months.[2]


  Novel Oral Radio protectors Top


For convenience and simple administration, the development of an effective orally administered radio-protective agent is highly desirable. One particular class of immunomodulators that has been relatively well developed and studied is 5-androstene steroids, whose archetype species is 5-androstene-3b, 17b-diol (5-AED), a natural steroid hormone produced in the adrenal cortex. Multiple efficacy studies have demonstrated the hematopoietic stimulation properties of 5-AED administered by subcutaneous injection before or after irradiation. In humans, 5-AED induces elevation of blood neutrophil and platelet counts, and in mice, it causes significant increases in the levels of granulocyte colony-stimulating factor and interleukin 6.[21]


  Radiation Mitigators Top


Radiation-induced late normal tissue toxicity is increasingly being appreciated as a phenomenon of ongoing changes in tissue after radiation but before the manifestation of cascades that can lead to vascular damage, tissue hypoxia, and excessive extracellular matrix deposition.[22] Radiation mitigators can be agents delivered during or shortly after exposure to repopulate a critical cell compartment such as the mucosa or bone marrow. In this instance, the mitigator is used to prevent acute toxicity. For radiologic terrorism and space research, much of the focus of mitigators have been in the field of developing chemo preventatives to reduce carcinogenesis of total body exposures.[2]


  Keratinocyte Growth Factor Top


KGF is a growth factor that stimulates a number of cellular processes such as differentiation, proliferation, DNA repair, and detoxification of ROS.[3]These properties make KGF an attractive method to stimulate the recovery of mucosa after IR. These properties make KGF an attractive method to stimulate the recovery of mucosa after IR. Accordingly, delivery of KGF in animal models prevents radiation-induced xerostomia and mucositis.[3]


  Palifermin Top


Palifermin is a recombinant human KGF that is approved for use in decreasing the incidence and duration of severe oral mucositis in patients with hematologic malignancies who receive high doses of chemotherapy and radiation therapy followed by stem cell rescue. The success of palifermin in patients with mucositis after cytotoxic therapy led to attempts to evaluate its use in patients with head and neck cancers receiving chemoradiotherapy, in whom mucositis can be severe and prolonged. The success of palifermin in patients with mucositis after cytotoxic therapy led to attempts to evaluate its use in patients with head and neck cancers receiving chemoradiotherapy, in whom mucositis can be severe and prolonged.[23]


  Transforming Growth Factor and Halofuginone Top


Transforming growth factor beta (TGF-γ) plays a critical role in the development of radiation-induced fibrosis. It is therefore not surprising that many of the agents that have been used to prevent the development of radiation fibrosis directly or indirectly inhibit the TGF-γ signalling pathway. TGF-γ receptor inhibition has shown the ability to prevent lung fibrosis after radiation exposure in the animal models. An alternative mechanism is the use of halofuginone, a small molecule that inhibits TGF-γ signalling, which has been shown in animal models to inhibit radiation-induced fibrosis.[3]


  Angiotensin Converting Enzyme Inhibitor Top


ACE inhibitors have been shown to mitigate radiation-induced lung injury in the preclinical models. The exact mechanisms of action have not been completely elucidated. ACE inhibitors may decrease the incidence of radiation pneumonitis in patients receiving thoracic radiation. These findings are consistent with preclinical evidence and should be prospectively evaluated.[24]


  Cyclooxygenase-2 Inhibitors/Nonsteroidal Anti-Inflammatory Drugs Top


Cyclooxygenase-2 (COX-2) is associated with ROS production and inflammatory signs in normal tissues. These effects further amplify radiation toxicity in irradiated cells as well as adjacent cells through a phenomenon known as Bystander effect. Increased COX-2 expression in distant nonirradiated tissues causes oxidative DNA damage and elevated cancer risk. Moreover, in tumors, the activation of this enzyme can increase the resistance of malignant cells to radiotherapy. Hence, the inhibition of COX-2 has been proposed for better therapeutic response and amelioration of normal tissues. Celecoxib is one of the most studied COX-2 inhibitors for radio sensitization and radioprotection, while some other inhibitors have shown interesting results.[25]


  Conclusion Top


Radiation therapy is frequently used in the definitive management and palliative care of patients with cancer. These agents termed as radiosensitizers and radioprotectors have a special role in the treatment of malignancies by radiotherapy. The novel agents are exhibiting promising results. In majority of instances, the success rate of radiotherapy is related to radiosensitizers and the patient's quality of life is dependent on the radioprotectors and radiation mitigators. The emerging application of new compounds such as nanoparticles as radiosensitizers and a steroid as a radioprotector and mitigator helps to reduce the cancer death.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

 
  References Top

1.
Ruba T, Tamilselvi R. Radiosensitizers and radioprotectors for effective radiation therapy-A review. Asian J Appl Sci Technol 2018;6:77-86.  Back to cited text no. 1
    
2.
Raviraj J, Bokkasam VK, Kumar VS, Reddy US, Suman V. Radiosensitizers, radioprotectors, and radiation mitigators. Indian J Dent Res 2014;25:83-90.  Back to cited text no. 2
[PUBMED]  [Full text]  
3.
Citrin D, Cotrim AP, Hyodo F, Baum BJ, Krishna MC, Mitchell JB. Radioprotectors and mitigators of radiation-induced normal tissue injury. Oncologist 2010;15:360-71.  Back to cited text no. 3
    
4.
Gosselin-Acomb TK. Principles of radiation therapy. In: Yarbro CH, Goodman M, Frogge MH, editors. Cancer Nursing: Principles and Practice. 6th ed. Sudbury: Jones and Bardett Publishers; 2005. p. 230-49.  Back to cited text no. 4
    
5.
Mayer R, Hamilton-Farrell MR, van der Kleij AJ, Schmutz J, Granström G, Sicko Z, et al. Hyperbaric oxygen and radiotherapy. Strahlenther Onkol 2005;2:114-23.  Back to cited text no. 5
    
6.
Al-Waili NS, Butler GJ, Beale J, Hamilton RW, Lee BY, Lucas P. Hyperbaric oxygen and malignancies: A potential role in radiotherapy, chemotherapy, tumor surgery and phototherapy. Med Sci Monit 2005;11:RA279-89.  Back to cited text no. 6
    
7.
Churchill-Davidson I, Foster CA, Wiernik G, Collins CD, Pizey NC, Skeggs DB, et al. The place of oxygen in radiotherapy. Br J Radiol 1966;39:321-31.  Back to cited text no. 7
    
8.
Henk JM. Late results of a trial of hyperbaric oxygen and radiotherapy in head and neck carcinomas: A rationale for hypoxic cell sensitizers. Internat J Radiat Oncol Biol Phys 1986;12:1339-41.  Back to cited text no. 8
    
9.
Hall EJ, Cox JD. Physical and biological basis of radiation therapy. In: Cox JD, Ang KK, editors. Radiation Oncology: Rationale, Technique, Results. 8th ed.. Missouri: Mosby; 2003. p. 3-62.  Back to cited text no. 9
    
10.
Hoskin PJ, Saunders MI, Phillips H, Cladd H, Powell ME, Goodchild K, et al. Carbogen and nicotinamide in the treatment of bladder cancer with radical radiotherapy. Br J Cancer 1997;76:260-3.  Back to cited text no. 10
    
11.
Bernier J, Stratford MR, Denekamp J, Dennis MF, Bieri S, Hagen F, et al. Pharmacokinetics of nicotinamide in cancer patients treated with accelerated radiotherapy. Radiother Oncol 1998:48:123-33.  Back to cited text no. 11
    
12.
Calabro-Jones PM, Fahey RC, Smoluk GD, Ward JF. Alkaline phosphatase promotes radioprotection and accumulation of WR-1065 in V79-171 cells incubated in medium containing WR-2721. Int J Radiat Biol Relat Stud Phys Chem Med 1985;47:23-7.  Back to cited text no. 12
    
13.
Purdie JW, Inhaber ER, Schneider H, Labelle JL. Interaction of cultured mammalian cells with WR-2721 and its thiol, WR-1065: Implications for mechanisms of radioprotection. Int J Radiat Biol Relat Stud Phys Chem Med 1983;43:517-27.  Back to cited text no. 13
    
14.
Blumberg AL, Nelson DF, Gramkowski M, Glover D, Glick JH, Yuhas JM, et al. Clinical trials of WR-2721 with radiation therapy. Int J Radiat Oncol Biol Phys 1982;8:561-3.  Back to cited text no. 14
    
15.
Brown JM. Sensitizers and protectors in radiotherapy. Cancer 1985;55:2222-8.  Back to cited text no. 15
    
16.
Soule BP, Hyodo F, Matsumoto K, Simone NL, Cook JA, Krishna MC, et al. Therapeutic and clinical applications of nitroxide compounds. Antioxid Redox Signal 2007;9:1731-43.  Back to cited text no. 16
    
17.
Shirazi A, Mihandoost E, Mahdevi RS, Mohseni M. Radioprotective role of antioxidant agents. Oncol Rev 2012;6:130-4.  Back to cited text no. 17
    
18.
Lee TK, Johnke RM, Allison RR, O'Brien KF, Dobbs LJ Jr. Radioprotective potential of ginseng. Mutagenesis 2005;20:237-43.  Back to cited text no. 18
    
19.
Hosseinimehr SJ. Trends in the development of radioprotective agents. Drug Discov Today 2007;12:794-805.  Back to cited text no. 19
    
20.
Guo H, Seixas-Silva JA Jr., Epperly MW, Gretton JE, Shin DM, Bar-Sagi D, et al. Prevention of radiation-induced oral cavity mucositis by plasmid/liposome delivery of the human manganese superoxide dismutase (SOD2) transgene. Radiat Res 2003;159:361-70.  Back to cited text no. 20
    
21.
Bai F, Wang L, Li Z, Yi L, Zuo Z, Zhang C, et al. 17a-Ethinyl-androst-5-ene-3b, 17b-diol, a novel potent oral radioprotective agent, confers radioprotection of hematopoietic stem and progenitor cells in a granulocyte colony-stimulating factoreindependent manner. Int J Radiat Oncol 2019;103:217e228.  Back to cited text no. 21
    
22.
Bentzen SM. Preventing or reducing late side effects of radiation therapy: Radiobiology meets molecular pathology. Nat Rev Cancer 2006;6:702-13.  Back to cited text no. 22
    
23.
Spielberger R, Stiff P, Bensinger W, Gentile T, Weisdorf D, Kewalramani T, et al. Palifermin for oral mucositis after intensive therapy for hematologic cancers. N Engl J Med 2004;351:2590-8.  Back to cited text no. 23
    
24.
Medhora M, Gao F, Jacobs ER, Moulder JE. Radiation damage to the lung: Mitigation by angiotensin-converting enzyme (ACE) inhibitors. Respirology 2012;17:66-71.  Back to cited text no. 24
    
25.
Cheki M, Yahyapour R, Farhood B, Rezaeyan A, Shabeeb D, Amini P, et al. COX-2 in radiotherapy: A potential target for radioprotection and ra-diosensitization. Curr Mol Pharmacol 2018;11:173-83.  Back to cited text no. 25
    


    Figures

  [Figure 1], [Figure 2]
 
 
    Tables

  [Table 1], [Table 2]



 

Top
 
 
  Search
 
Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
 Related articles
Access Statistics
Email Alert *
Add to My List *
* Registration required (free)

 
  In this article
Abstract
Introduction
Classification o...
Radiosensitizers
Hyperbaric Oxygen
Mechanism of Action
Carbogen
Nicotinamide
Radioprotectors
Amifostine: A Ra...
Mechanism of Action
Nitroxides
Antioxidants as ...
Vitamin E
Herbal Radio-Pro...
Superoxide Dimutase
Melatonin
Novel Oral Radio...
Radiation Mitigators
Keratinocyte Gro...
Palifermin
Transforming Gro...
Angiotensin Conv...
Cyclooxygenase-2...
Conclusion
References
Article Figures
Article Tables

 Article Access Statistics
    Viewed4976    
    Printed312    
    Emailed0    
    PDF Downloaded139    
    Comments [Add]    

Recommend this journal


[TAG2]
[TAG3]
[TAG4]