AccScience Publishing / ARNM / Volume 1 / Issue 2 / DOI: 10.36922/arnm.0914
Cite this article
83
Download
1364
Views
Journal Browser
Volume | Year
Issue
Search
News and Announcements
View All
REVIEW

Advancements and challenges in interstitial brachytherapy using iodine-125 seeds

Liting Xiong1† Yuhan Yang1† Mengyuan Li1 Ping Jiang1* Chunxiao Li1* Junjie Wang1*
Show Less
1 Department of Radiation Oncology, Peking University Third Hospital, Institute of Medical Technology, Peking University Health Science Center, Beijing, China
Submitted: 6 May 2023 | Accepted: 21 August 2023 | Published: 3 October 2023
© 2023 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

Radiation therapy has been used for over a century in the treatment of tumors, with interstitial tissue treatment using radioactive seeds playing a key role in this approach. Iodine-125 (I-125) seeds are the most commonly used radioactive sealed source for permanent interstitial brachytherapy. In recent decades, significant advancements have been made in the field of interstitial radiation therapy. The development of three-dimensional (3D)-printed personalized templates, treatment planning systems, and robot-assisted systems have significantly improved the accuracy of I-125 brachytherapy. This review summarizes the advances in technology, radiobiology, physics, and immunology of I-125 brachytherapy. These advancements have improved the accuracy of dose delivery and increased the effectiveness of I-125 interstitial brachytherapy. In particular, the utilization of 3D-printed personalized templates has allowed for customized treatment planning and more precise dose delivery. Robot-assisted systems have also made significant contributions by assisting in the precise placement of radioactive seeds during treatment. However, several challenges persist within the field of interstitial I-125 brachytherapy. One of the current issues is the difficulty in accurately predicting the biological response to radiation therapy in individual patients. Addressing this challenge represents an important area for further research, as it has the potential to improve treatment outcomes and minimize side effects. In addition, there is a need for more research into the utilization of immunotherapy in conjunction with interstitial brachytherapy, as this combination has demonstrated promise in preclinical studies. Overall, this review provides a comprehensive overview of the advances and challenges associated with interstitial brachytherapy using I-125 seeds. These advancements offer a theoretical basis for achieving precise and remote medical care in brachytherapy. As technology continues to evolve, it is likely that interstitial brachytherapy will emerge as an even more effective treatment option for cancer patients.

Keywords
Iodine-125
Brachytherapy
Advancements
Funding
National Natural Science Foundation of China
Natural Science Foundation of Beijing Municipality
Intramural funding from the Beijing University Third Hospital
Special Fund of the National Clinical Key Specialty Construction Program, P. R. China
Cancer Precision Radiotherapy Spark Program of China International Medical Foundation
References
  1. Parker SL, 1997, Cancer statistics, 1997. CA Cancer J Clin, 47: 5–27. https://doi.org/10.3322/canjclin.47.1.5

 

  1. Rosenberg I, 2008, Radiation oncology physics: A handbook for teachers and students. Br J Cancer, 98: 1020. https://doi.org/10.1038/sj.bjc.6604224

 

  1. Yoshioka Y, 2009, Current status and perspectives of brachytherapy for prostate cancer. Int J Clin Oncol, 14: 31–36. https://doi.org/10.1007/s10147-008-0866-z

 

  1. Torp-Pedersen S, Holm HH, Littrup PJ, 1987, Transperineal I-125 seed implantation in prostate cancer guided by transrectal ultrasound. Prog Clin Biol Res, 237: 143–152.

 

  1. Ji Z, Jiang Y, Guo F, et al., 2020, Safety and efficacy of CT-guided radioactive iodine-125 seed implantation assisted by a 3D printing template for the treatment of thoracic malignancies. J Cancer Res Clin Oncol, 146: 229–236. https://doi.org/10.1007/s00432-019-03050-7

 

  1. Ji Z, Jiang Y, Tian S, et al., 2019, The effectiveness and prognostic factors of CT-guided radioactive I-125 seed implantation for the treatment of recurrent head and neck cancer after external beam radiation therapy. Int J Radiat Oncol Biol Phys, 103: 638–645. https://doi.org/10.1016/j.ijrobp.2018.10.034

 

  1. Wang H, Wang J, Jiang Y, et al., 2013, The investigation of 125I seed implantation as a salvage modality for unresectable pancreatic carcinoma. J Exp Clin Cancer Res, 32: 106. https://doi.org/10.1186/1756-9966-32-106

 

  1. Wang J, Yuan H, Ma Q, et al., 2010, Interstitial 125I seeds implantation to treat spinal metastatic and primary paraspinal malignancies. Med Oncol, 27: 319–326. https://doi.org/10.1007/s12032-009-9212-1

 

  1. Yao L, Cao Q, Wang J, et al., 2016, CT-Guided (125)I Seed Interstitial brachytherapy as a salvage treatment for recurrent spinal metastases after external beam radiotherapy. Biomed Res Int, 2016: 8265907. https://doi.org/10.1155/2016/8265907

 

  1. Li Y, Jiang Y, Wang J, 2023, Safety and efficacy of CT-guided radioactive iodine-125 seed implantation as a salvage treatment for recurrent head and neck cancer after two or more courses of radiotherapy. Radiat Oncol, 18: 73. https://doi.org/10.1186/s13014-023-02254-z

 

  1. Qiu Z, Yu C, Qiu X, et al., 2023, Safety and efficacy of CT-guided iodine-125 brachytherapy for portal vein tumor thrombus in hepatocellular carcinoma. Acad Radiol, 30 Suppl 1: S53–S60. https://doi.org/10.1016/j.acra.2023.02.006

 

  1. McLaughlin PW, Troyer S, Berri S, et al., 2005, Functional anatomy of the prostate: Implications for treatment planning. Int J Radiat Oncol Biol Phys, 63: 479–491. https://doi.org/10.1016/j.ijrobp.2005.02.036

 

  1. Takiar V, Pugh TJ, Swanson D, et al., 2014, MRI-based sector analysis enhances prostate palladium-103 brachytherapy quality assurance in a phase II prospective trial of men with intermediate-risk localized prostate cancer. Brachytherapy, 13: 68–74. https://doi.org/10.1016/j.brachy.2013.04.001

 

  1. Register SP, Kudchadker RJ, Levy LB, et al., 2013, An MRI-based dose--reponse analysis of urinary sphincter dose and urinary morbidity after brachytherapy for prostate cancer in a phase II prospective trial. Brachytherapy, 12: 210–216. https://doi.org/10.1016/j.brachy.2012.10.006

 

  1. Lin ZY, Lin J, Lin C, et al., 2012, 1.5T conventional MR-guided iodine-125 interstitial implants for hepatocellular carcinoma: Feasibility and preliminary clinical experience. Eur J Radiol, 81: 1420–1425. https://doi.org/10.1016/j.ejrad.2011.03.043

 

  1. Huang MW, Liu SM, Zheng L, et al., 2012, A digital model individual template and CT-guided 125I seed implants for malignant tumors of the head and neck. J Radiat Res, 53: 973–977. https://doi.org/10.1093/jrr/rrs046

 

  1. Zhang H, Dev D, Yu H, et al., 2019, Feasibility of three-dimensional-printed template-guided (125)I seed brachytherapy and dosimetric evaluation in patients with malignant tumor. J Cancer Res Ther, 15: 793–800. https://doi.org/10.4103/jcrt.JCRT_347_18

 

  1. Qiu B, Jiang Y, Ji Z, et al., 2021, The accuracy of individualized 3D-printing template-assisted I(125) radioactive seed implantation for recurrent/metastatic head and neck cancer. Front Oncol, 11: 664996. https://doi.org/10.3389/fonc.2021.664996

 

  1. Rivard MJ, Coursey BM, DeWerd LA, et al., 2004, Update of AAPM Task Group No. 43 Report: A revised AAPM protocol for brachytherapy dose calculations. Med Phys, 31: 633–674. https://doi.org/10.1118/1.1646040

 

  1. Salembier C, Lavagnini P, Nickers P, et al., 2007, Tumour and target volumes in permanent prostate brachytherapy: A supplement to the ESTRO/EAU/EORTC recommendations on prostate brachytherapy. Radiother Oncol, 83: 3–10. https://doi.org/10.1016/j.radonc.2007.01.014

 

  1. Kunos CA, Resnick MI, Kinsella TJ, et al., 2004, Migration of implanted free radioactive seeds for adenocarcinoma of the prostate using a Mick applicator. Brachytherapy, 3: 71–77. https://doi.org/10.1016/j.brachy.2004.06.002

 

  1. Reed DR, Wallner KE, Merrick GS, et al., 2007, A prospective randomized comparison of stranded vs. loose 125I seeds for prostate brachytherapy. Brachytherapy, 6: 129–134. https://doi.org/10.1016/j.brachy.2007.01.003

 

  1. Nuver TT, Hilgers GC, Kattevilder RA, et al., 2022, Local seed displacement from day 0 to day 30 in I-125 permanent prostate brachytherapy: A detailed, computed tomography-based analysis. Brachytherapy, 21: 208–215. https://doi.org/10.1016/j.brachy.2021.12.009

 

  1. Herbert C, Morris WJ, Hamm J, et al., 2011, The effect of loose versus stranded seeds on biochemical no evidence of disease in patients with carcinoma of the prostate treated with iodine-125 brachytherapy. Brachytherapy, 10: 442–448. https://doi.org/10.1016/j.brachy.2011.01.011

 

  1. Al-Qaisieh B, Carey B, Ash D, et al., 2004, The use of linked seeds eliminates lung embolization following permanent seed implantation for prostate cancer. Int J Radiat Oncol Biol Phys, 59: 397–399. https://doi.org/10.1016/j.ijrobp.2003.10.034

 

  1. Ishiyama H, Satoh T, Kawakami S, et al., 2014, A prospective quasi-randomized comparison of intraoperatively built custom-linked seeds versus loose seeds for prostate brachytherapy. Int J Radiat Oncol Biol Phys, 90: 134–139. https://doi.org/10.1016/j.ijrobp.2014.05.009

 

  1. Ahmed AK, Zygourakis CC, Kalb S, et al., 2019, First spine surgery utilizing real-time image-guided robotic assistance. Comput Assist Surg (Abingdon), 24: 13–17. https://doi.org/10.1080/24699322.2018.1542029

 

  1. Egberts JH, Möller T, Becker T, 2019, Robotic-assisted sleeve lobectomy using the four-arm technique in the DaVinci Si® and Xi® systems. Thorac Cardiovasc Surg, 67: 603–605. https://doi.org/10.1055/s-0038-1660508

 

  1. Rohatgi P, Jafrani RJ, Brandmeir NJ, et al., 2018, Robotic-guided bihippocampal and biparahippocampal depth placement for responsive neurostimulation in bitemporal lobe epilepsy. World Neurosurg, 111: 181–189. https://doi.org/10.1016/j.wneu.2017.10.164

 

  1. Dhaliwal SS, Chettibi T, Wilby S, et al., 2021, Review of clinical and technological consideration for MRI-guided robotic prostate brachytherapy. IEEE Trans Med Robot Bionics, 3: 583–605. https://doi.org/10.1109/TMRB.2021.3097127

 

  1. Seifabadi R, Song SE, Krieger A, et al., 2012, Robotic system for MRI-guided prostate biopsy: Feasibility of teleoperated needle insertion and ex vivo phantom study. Int J Comput Assist Radiol Surg, 7: 181–190. https://doi.org/10.1007/s11548-011-0598-9

 

  1. Salcudean SE, Prananta TD, Morris WJ, et al., 2008, A Robotic Needle Guide for Prostate Brachytherapy. In: 2008 IEEE International Conference on Robotics and Automation.

 

  1. Bodei L, Kassis AI, Adelstein SJ, et al., 2003, Radionuclide therapy with iodine-125 and other auger-electron-emitting radionuclides: Experimental models and clinical applications. Cancer Biother Radiopharm, 18: 861–877. https://doi.org/10.1089/108497803322702833

 

  1. Welt S, Scott AM, Divgi CR, et al., 1996, Phase I/II study of iodine 125-labeled monoclonal antibody A33 in patients with advanced colon cancer. J Clin Oncol, 14: 1787–1797. https://doi.org/10.1200/JCO.1996.14.6.1787

 

  1. Chargari C, Van Limbergen E, Mahantshetty U, et al., 2018, Radiobiology of brachytherapy: The historical view based on linear quadratic model and perspectives for optimization. Cancer Radiother, 22: 312–318. https://doi.org/10.1016/j.canrad.2017.11.011

 

  1. Thames HD, 1985, An “incomplete-repair” model for survival after fractionated and continuous irradiations. Int J Radiat Biol Relat Stud Phys Chem Med, 47: 319–339. https://doi.org/10.1080/09553008514550461

 

  1. Dikomey E, Brammer I, 2000, Relationship between cellular radiosensitivity and non-repaired double-strand breaks studied for different growth states, dose rates and plating conditions in a normal human fibroblast line. Int J Radiat Biol, 76: 773–781. https://doi.org/10.1080/09553000050028922

 

  1. Deore SM, Sarin R, Dinshaw KA, et al., 1993, Influence of dose-rate and dose per fraction on clinical outcome of breast cancer treated by external beam irradiation plus iridium-192 implants: Analysis of 289 cases. Int J Radiat Oncol Biol Phys, 26: 601–606. https://doi.org/10.1016/0360-3016(93)90275-z

 

  1. Steel GG, 1991, The ESTRO Breur lecture. Cellular sensitivity to low dose-rate irradiation focuses the problem of tumour radioresistance. Radiother Oncol, 20: 71–83. https://doi.org/10.1016/0167-8140(91)90140-c

 

  1. Williams JR, Zhang Y, Zhou H, et al., 2008, Overview of radiosensitivity of human tumor cells to low-dose-rate irradiation. Int J Radiat Oncol Biol Phys, 72: 909–917. https://doi.org/10.1016/j.ijrobp.2008.06.1928

 

  1. Baumann M, Dörr W, Petersen C, et al., 2003, Repopulation during fractionated radiotherapy: Much has been learned, even more is open. Int J Radiat Biol, 79: 465–467. https://doi.org/10.1080/0955300031000160259

 

  1. Zhang WF, Jin WD, Li B, et al., 2014, Effect of brachytherapy on NF-kappaB and VEGF in gastric carcinoma xenografts. Oncol Rep, 32; 635–640. https://doi.org/10.3892/or.2014.3255

 

  1. Wang H, Li J, Qu A, et al., 2013, The different biological effects of single, fractionated and continuous low dose rate irradiation on CL187 colorectal cancer cells. Radiat Oncol, 8: 196. https://doi.org/10.1186/1748-717X-8-196

 

  1. Hennequin C, Mazeron JJ, 2013, Radiobiology in brachytherapy. Cancer Radiother, 17: 81–84. https://doi.org/10.1016/j.canrad.2013.03.001

 

  1. Halle C, Andersen E, Lando M, et al., 2012, Hypoxia-induced gene expression in chemoradioresistant cervical cancer revealed by dynamic contrast-enhanced MRI. Cancer Res, 72: 5285–5295. https://doi.org/10.1158/0008-5472.CAN-12-1085

 

  1. Barendsen GW, 1982, Dose fractionation, dose rate and iso-effect relationships for normal tissue responses. Int J Radiat Oncol Biol Phys, 8: 1981–1997. https://doi.org/10.1016/0360-3016(82)90459-x

 

  1. Jani AB, Hand CM, Lujan AE, et al., 2004, Biological effective dose for comparison and combination of external beam and low-dose rate interstitial brachytherapy prostate cancer treatment plans. Med Dosim, 29: 42–48. https://doi.org/10.1016/j.meddos.2003.09.005

 

  1. Antipas V, Dale RG, Coles IP, 2001, A theoretical investigation into the role of tumour radiosensitivity, clonogen repopulation, tumour shrinkage and radionuclide RBE in permanent brachytherapy implants of 125I and 103Pd. Phys Med Biol, 46: 2557–25569. https://doi.org/10.1088/0031-9155/46/10/304

 

  1. Ling CC, Li WX, Anderson LL, 1995, The relative biological effectiveness of I-125 and Pd-103. Int J Radiat Oncol Biol Phys, 32: 373–378. https://doi.org/10.1016/0360-3016(95)00530-C

 

  1. Armpilia CI, Dale RG, Coles IP, et al., 2003, The determination of radiobiologically optimized half-lives for radionuclides used in permanent brachytherapy implants. Int J Radiat Oncol Biol Phys, 55: 378–385. https://doi.org/10.1016/s0360-3016(02)04208-6

 

  1. Manimaran S, 2007, Radiobiological equivalent of low/ high dose rate brachytherapy and evaluation of tumor and normal responses to the dose. Radiat Med, 25: 229–235. https://doi.org/10.1007/s11604-007-0131-9

 

  1. Patankar SS, Tergas AI, Deutsch I, et al., 2015, High versus low-dose rate brachytherapy for cervical cancer. Gynecol Oncol, 136: 534–541. https://doi.org/10.1016/j.ygyno.2014.12.038

 

  1. Cagetti LV, Zemmour C, Salem N, et al., 2019, High-dose-rate vs. low-dose-rate interstitial brachytherapy boost for anal canal cancers. Brachytherapy, 18: 814–822. https://doi.org/10.1016/j.brachy.2019.08.005

 

  1. Nath R, Anderson LL, Luxton G, et al., 1995, Dosimetry of interstitial brachytherapy sources: recommendations of the AAPM Radiation Therapy Committee Task Group No. 43. American Association of Physicists in Medicine. Med Phys, 22: 209–234. https://doi.org/10.1118/1.597458

 

  1. Li R, Ying Z, Yuan Y, et al., 2019, Comparison of two iodine-125 brachytherapy implant techniques for the treatment of lung tumor: Preplanning and intraoperative planning. Brachytherapy, 18: 87–94. https://doi.org/10.1016/j.brachy.2018.08.007

 

  1. Loftus TP, 1984, Exposure standardization of Iodine-125 seeds used for brachytherapy. J Res Natl Bur Stand (1977), 89: 295–303. https://doi.org/10.6028/jres.089.017

 

  1. DeWerd LA, Huq MS, Das IJ, et al., 2004, Procedures for establishing and maintaining consistent air-kerma strength standards for low-energy, photon-emitting brachytherapy sources: Recommendations of the Calibration Laboratory Accreditation Subcommittee of the American Association of Physicists in Medicine. Med Phys, 31: 675–681. https://doi.org/10.1118/1.1645681

 

  1. Perez-Calatayud J, Ballester F, Das RK, et al., 2012, Dose calculation for photon-emitting brachytherapy sources with average energy higher than 50 keV: Report of the AAPM and ESTRO. Med Phys, 39: 2904–2929. https://doi.org/10.1118/1.3703892

 

  1. Nath R, Anderson LL, Meli JA, et al., 1997, Code of practice for brachytherapy physics: Report of the AAPM Radiation Therapy Committee Task Group No. 56. American Association of Physicists in Medicine. Med Phys, 24: 1557–1598.

 

  1. Podder TK, Beaulieu L, Caldwell B, et al., 2014, AAPM and GEC-ESTRO guidelines for image-guided robotic brachytherapy: Report of Task Group 192. Med Phys, 41: 101501. https://doi.org/10.1118/1.4895013

 

  1. Liu S, Wang H, Wang C, et al., 2019, Dosimetry verification of 3D-printed individual template based on CT-MRI fusion for radioactive (125)I seed implantation in recurrent high-grade gliomas. J Contemp Brachytherapy, 11: 235–242. https://doi.org/10.5114/jcb.2019.85729

 

  1. Major T, Polgár C, Jorgo K, et al., 2017, Dosimetric comparison between treatment plans of patients treated with low-dose-rate vs. high-dose-rate interstitial prostate brachytherapy as monotherapy: Initial findings of a randomized clinical trial. Brachytherapy, 16: 608–615. https://doi.org/10.1016/j.brachy.2017.02.003
  2. Zhang H, Liang Y, Qiu G, et al., 2019, Precision dose measurement of 125I seed in solid water through SPECT/ CT detecting. J Cancer Res Ther, 15: 291–297. https://doi.org/10.4103/jcrt.JCRT_522_18

 

  1. Zhang H, Zhao X, Wang J, et al., 2016, Dose distribution detected by SPECT/CT in a patient with prostate cancer treated with 125I seeds: A case report. Brachytherapy, 15: S183–S184. https://doi.org/10.1016/j.brachy.2016.04.337

 

  1. Kono Y, Miyamoto Y, Oohashi S, et al., 2011, Radiation exposure to general public after permanent brachytherapy for prostate cancer. Radiat Prot Dosimetry, 146: 229–230. https://doi.org/10.1093/rpd/ncr156

 

  1. Herrera FG, Bourhis J, Coukos G, 2017, Radiotherapy combination opportunities leveraging immunity for the next oncology practice. CA Cancer J Clin, 67: 65–85. https://doi.org/10.3322/caac.21358

 

  1. Pencik J, Schlederer M, Gruber W, et al., 2015, STAT3 regulated ARF expression suppresses prostate cancer metastasis. Nat Commun, 6: 7736. https://doi.org/10.1038/ncomms8736

 

  1. Formenti SC, Demaria S, 2013, Combining radiotherapy and cancer immunotherapy: A paradigm shift. J Natl Cancer Inst, 105: 256–265. https://doi.org/10.1093/jnci/djs629

 

  1. Finkelstein SE, Salenius S, Mantz CA, et al., 2015, Combining immunotherapy and radiation for prostate cancer. Clin Genitourin Cancer, 13: 1–9. https://doi.org/10.1016/j.clgc.2014.09.001

 

  1. Du E, Wang L, Li CY, et al., 2017, Analysis of immune status after iodine-125 permanent brachytherapy in prostate cancer. Onco Targets Ther, 10: 2561–2567. https://doi.org/10.2147/OTT.S137491

 

  1. Ishihara D, Pop L, Takeshima T, et al., 2017, Rationale and evidence to combine radiation therapy and immunotherapy for cancer treatment. Cancer Immunol Immunother, 66: 281–298. https://doi.org/10.1007/s00262-016-1914-6

 

  1. Cabel L, Loir E, Gravis G, et al., Long-term complete remission with Ipilimumab in metastatic castrate-resistant prostate cancer: Case report of two patients. J Immunother Cancer, 5: 31. https://doi.org/10.1186/s40425-017-0232-7

 

  1. Tumeh PC, Harview CL, Yearley JH, et al., 2014, PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature, 515: 568–571. https://doi.org/10.1038/nature13954

 

  1. Arina A, Karrison T, Galka E, et al., 2017, Transfer of allogeneic CD4+ T cells rescues CD8+ T cells in anti-PD-L1-resistant tumors leading to tumor eradication. Cancer Immunol Res, 5: 127–136. https://doi.org/10.1158/2326-6066.CIR-16-0293

 

  1. Sallin MA, Sakai S, Kauffman KD, et al., 2017, Th1 differentiation drives the accumulation of intravascular, non-protective CD4 T cells during tuberculosis. Cell Rep, 18: 3091–3104. https://doi.org/10.1016/j.celrep.2017.03.007

 

  1. Hou PF, Zhu LJ, Chen XY, et al., Age-related changes in CD4+CD25+FOXP3+ regulatory T cells and their relationship with lung cancer. PLoS One, 12: e0173048. https://doi.org/10.1371/journal.pone.0173048

 

  1. Wang X, Liu Y, Dai L, et al., 2016, Foxp3 downregulation in NSCLC mediates epithelial-mesenchymal transition via NF-kappaB signaling. Oncol Rep, 36: 2282–2288. https://doi.org/10.3892/or.2016.5024

 

  1. Wada S, Harris TJ, Tryggestad E, et al., Combined treatment effects of radiation and immunotherapy: Studies in an autochthonous prostate cancer model. Int J Radiat Oncol Biol Phys, 87: 769–776. https://doi.org/10.1016/j.ijrobp.2013.07.015

 

  1. Nardone V, Botta C, Caraglia M, et al., 2016, Tumor infiltrating T lymphocytes expressing FoxP3, CCR7 or PD-1 predict the outcome of prostate cancer patients subjected to salvage radiotherapy after biochemical relapse. Cancer Biol Ther, 17: 1213–1220. https://doi.org/10.1080/15384047.2016.1235666

 

  1. Harris TJ, Hipkiss EL, Borzillary S, et al., 2008, Radiotherapy augments the immune response to prostate cancer in a time-dependent manner. Prostate, 68: 1319–1329. https://doi.org/10.1002/pros.20794

 

  1. Mittal D, Gubin MM, Schreiber RD, et al., 2014, New insights into cancer immunoediting and its three component phases- -elimination, equilibrium and escape. Curr Opin Immunol, 27: 16–25. https://doi.org/10.1016/j.coi.2014.01.004
Conflict of interest
The authors declare no potential conflicts of interest.
Share
Back to top
Advances in Radiotherapy & Nuclear Medicine, Electronic ISSN: 2972-4392 Published by AccScience Publishing