DOI QR코드

DOI QR Code

Novel biological strategies to enhance the radiation therapeutic ratio

  • Kim, Jae Ho (Department of Radiation Oncology, Henry Ford Hospital) ;
  • Jenrow, Kenneth A. (Department of Psychology/Neuroscience Program, Central Michigan University) ;
  • Brown, Stephen L. (Department of Radiation Oncology, Henry Ford Hospital)
  • Received : 2018.07.16
  • Accepted : 2018.08.29
  • Published : 2018.09.30

Abstract

Successful anticancer strategies require a differential response between tumor and normal tissue (i.e., a therapeutic ratio). In fact, improving the effectiveness of a cancer therapeutic is of no clinical value in the absence of a significant increase in the differential response between tumor and normal tissue. Although radiation dose escalation with the use of intensity modulated radiation therapy has permitted the maximum tolerable dose for most locally advanced cancers, improvements in tumor control without damaging normal adjacent tissues are needed. As a means of increasing the therapeutic ratio, several new approaches are under development. Drugs targeting signal transduction pathways in cancer progression and more recently, immunotherapeutics targeting specific immune cell subsets have entered the clinic with promising early results. Radiobiological research is underway to address pressing questions as to the dose per fraction, irradiated tumor volume and time sequence of the drug administration. To exploit these exciting novel strategies, a better understanding is needed of the cellular and molecular pathways responsible for both cancer and normal tissue and organ response, including the role of radiation-induced accelerated senescence. This review will highlight the current understanding of promising biologically targeted therapies to enhance the radiation therapeutic ratio.

Keywords

References

  1. Citrin DE. Recent Developments in Radiotherapy. N Engl J Med 2017;377:1065-75.
  2. Giaccia AJ. Molecular radiobiology: the state of the art. J Clin Oncol 2014;32:2871-8.
  3. Kim KW, Myers CJ, Jung DK, Lu B. NVP-BEZ-235 enhances radiosensitization via blockade of the PI3K/mTOR pathway in cisplatin-resistant non-small cell lung carcinoma. Genes Cancer 2014;5:293-302.
  4. Chang L, Graham PH, Hao J, et al. Emerging roles of radioresistance in prostate cancer metastasis and radiation therapy. Cancer Metastasis Rev 2014;33:469-96.
  5. Tang L, Wei F, Wu Y, et al. Role of metabolism in cancer cell radioresistance and radiosensitization methods. J Exp Clin Cancer Res. 2018;37:87.
  6. Jaklevic B, Uyetake L, Lemstra W, et al. Contribution of growth and cell cycle checkpoints to radiation survival in Drosophila. Genetics 2006;174:1963-72.
  7. Eshleman JS, Carlson BL, Mladek AC, Kastner BD, Shide KL, Sarkaria JN. Inhibition of the mammalian target of rapamycin sensitizes U87 xenografts to fractionated radiation therapy. Cancer Res 2002;62:7291-7.
  8. Shinohara ET, Cao C, Niermann K, et al. Enhanced radiation damage of tumor vasculature by mTOR inhibitors. Oncogene 2005;24:5414-22.
  9. Albert JM, Kim KW, Cao C, Lu B. Targeting the Akt/mammalian target of rapamycin pathway for radiosensitization of breast cancer. Mol Cancer Ther 2006;5:1183-9.
  10. Nagata Y, Takahashi A, Ohnishi K, et al. Effect of rapamycin, an mTOR inhibitor, on radiation sensitivity of lung cancer cells having different p53 gene status. Int J Oncol 2010;37:1001-10.
  11. Mauceri HJ, Sutton HG, Darga TE, et al. Everolimus exhibits efficacy as a radiosensitizer in a model of non-small cell lung cancer. Oncol Rep 2012;27:1625-9.
  12. Nassim R, Mansure JJ, Chevalier S, Cury F, Kassouf W. Combining mTOR inhibition with radiation improves antitumor activity in bladder cancer cells in vitro and in vivo: a novel strategy for treatment. PLoS One 2013;8:e65257.
  13. Hayman TJ, Williams ES, Jamal M, Shankavaram UT, Camphausen K, Tofilon PJ. Translation initiation factor eIF4E is a target for tumor cell radiosensitization. Cancer Res 2012;72:2362-72.
  14. Hayman TJ, Wahba A, Rath BH, et al. The ATP-competitive mTOR inhibitor INK128 enhances in vitro and in vivo radiosensitivity of pancreatic carcinoma cells. Clin Cancer Res 2014;20:110-9.
  15. Ushijima H, Suzuki Y, Oike T, et al. Radio-sensitization effect of an mTOR inhibitor, temsirolimus, on lung adenocarcinoma A549 cells under normoxic and hypoxic conditions. J Radiat Res 2015;56:663-8.
  16. Kelly CJ, Hussien K, Fokas E, et al. Regulation of O2 consumption by the PI3K and mTOR pathways contributes to tumor hypoxia. Radiother Oncol 2014;111:72-80.
  17. Kalender A, Selvaraj A, Kim SY, et al. Metformin, independent of AMPK, inhibits mTORC1 in a rag GTPase-dependent manner. Cell Metab 2010;11:390-401.
  18. Song CW, Lee H, Dings RP, et al. Metformin kills and radiosensitizes cancer cells and preferentially kills cancer stem cells. Sci Rep 2012;2:362.
  19. Storozhuk Y, Hopmans SN, Sanli T, et al. Metformin inhibits growth and enhances radiation response of non-small cell lung cancer (NSCLC) through ATM and AMPK. Br J Cancer 2013;108:2021-32.
  20. Zannella VE, Dal Pra A, Muaddi H, et al. Reprogramming metabolism with metformin improves tumor oxygenation and radiotherapy response. Clin Cancer Res 2013;19:6741-50.
  21. Brown, SL, Nagaraja, TN, Aryal, MP, et al. MRI-tracked tumor vasculature changes in the hours after single fraction irradiation. Radiat Res 2015;183:713-21.
  22. NRG Oncology. Randomized phase II trial of concurrent chemoradiotherapy +/- metformin HCL in locally advanced NSCLC (NRG-LU001, ClinicalTrials.gov NCT02186847). Philadelphia, PA: NRG Oncology; 2016.
  23. Lew YS, Kolozsvary A, Brown SL, Kim JH. Synergistic interaction with arsenic trioxide and fractionated radiation in locally advanced murine tumor. Cancer Res 2002;62:4202-5.
  24. Kim JH, Lew YS, Kolozsvary A, Ryu S, Brown SL. Arsenic trioxide enhances radiation response of 9L glioma in the rat brain. Radiat Res 2003;160:662-6.
  25. Shiotani B, Zou L. Single-stranded DNA orchestrates an ATMto-ATR switch at DNA breaks. Mol Cell 2009;33:547-58.
  26. Khosravi R, Maya R, Gottlieb T, Oren M, Shiloh Y, Shkedy D. Rapid ATM-dependent phosphorylation of MDM2 precedes p53 accumulation in response to DNA damage. Proc Natl Acad Sci U S A 1999;96:14973-7.
  27. Origanti S, Cai SR, Munir AZ, White LS, Piwnica-Worms H. Synthetic lethality of Chk1 inhibition combined with p53 and/or p21 loss during a DNA damage response in normal and tumor cells. Oncogene 2013;32:577-88.
  28. Bartek J, Lukas J. Chk1 and Chk2 kinases in checkpoint control and cancer. Cancer Cell 2003;3:421-9.
  29. Sorensen CS, Hansen LT, Dziegielewski J, et al. The cellcycle checkpoint kinase Chk1 is required for mammalian homologous recombination repair. Nat Cell Biol 2005;7:195-201.
  30. Tao Y, Leteur C, Yang C, et al. Radiosensitization by Chir-124, a selective CHK1 inhibitor: effects of p53 and cell cycle checkpoints. Cell Cycle 2009;8:1196-205.
  31. Engelke CG, Parsels LA, Qian Y, et al. Sensitization of pancreatic cancer to chemoradiation by the Chk1 inhibitor MK8776. Clin Cancer Res 2013;19:4412-21.
  32. Suzuki M, Yamamori T, Bo T, Sakai Y, Inanami O. MK-8776, a novel Chk1 inhibitor, exhibits an improved radiosensitizing effect compared to UCN-01 by exacerbating radiation-induced aberrant mitosis. Transl Oncol 2017;10:491-500.
  33. Qiu Z, Oleinick NL, Zhang J. ATR/CHK1 inhibitors and cancer therapy. Radiother Oncol 2018;126:450-64.
  34. Morgan MA, Parsels LA, Maybaum J, Lawrence TS. Improving the efficacy of chemoradiation with targeted agents. Cancer Discov 2014;4:280-91.
  35. Prevo R, Fokas E, Reaper PM, et al. The novel ATR inhibitor VE-821 increases sensitivity of pancreatic cancer cells to radiation and chemotherapy. Cancer Biol Ther 2012;13:1072-81.
  36. Hirai H, Iwasawa Y, Okada M, et al. Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumor cells to DNA-damaging agents. Mol Cancer Ther 2009;8:2992-3000.
  37. Sarcar B, Kahali S, Prabhu AH, et al. Targeting radiationinduced G(2) checkpoint activation with the Wee-1 inhibitor MK-1775 in glioblastoma cell lines. Mol Cancer Ther 2011;10:2405-14.
  38. Bridges KA, Hirai H, Buser CA, et al. MK-1775, a novel Wee1 kinase inhibitor, radiosensitizes p53-defective human tumor cells. Clin Cancer Res 2011;17:5638-48.
  39. Old LJ, Boyse EA. Immunology of experimental tumors. Annu Rev Med 1964;15:167-86.
  40. Old LJ, Stockert E, Boyse EA, Kim JH. Antigenic modulation: loss of TL antigen from cells exposed to TL antibody. Study of the phenomenon in vitro. J Exp Med 1968;127:523-39.
  41. Callahan MK, Wolchok JD, Allison JP. Anti-CTLA-4 antibody therapy: immune monitoring during clinical development of a novel immunotherapy. Semin Oncol 2010;37:473-84.
  42. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252-64.
  43. Brahmer JR, Pardoll DM. Immune checkpoint inhibitors: making immunotherapy a reality for the treatment of lung cancer. Cancer Immunol Res 2013;1:85-91.
  44. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology: mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124-8.
  45. Postow MA, Callahan MK, Barker CA, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med 2012;366:925-31.
  46. Golden EB, Demaria S, Schiff PB, Chachoua A, Formenti SC. An abscopal response to radiation and ipilimumab in a patient with metastatic non-small cell lung cancer. Cancer Immunol Res 2013;1:365-72.
  47. Formenti SC, Demaria S. Systemic effects of local radiotherapy. Lancet Oncol 2009;10:718-26.
  48. Dewan MZ, Galloway AE, Kawashima N, et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res 2009;15:5379-88.
  49. Schaue D, Ratikan JA, Iwamoto KS, McBride WH. Maximizing tumor immunity with fractionated radiation. Int J Radiat Oncol Biol Phys 2012;83:1306-10.
  50. McBride WH, Ganapathy E, Lee MH, Nesseler JP, Nguyen C, Schaue D. A perspective on the impact of radiation therapy on the immune rheostat. Br J Radiol 2017;90:20170272.
  51. Koo T, Kim IA. Radiotherapy and immune checkpoint blockades: a snapshot in 2016. Radiat Oncol J 2016;34:250-9.
  52. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell 2010;141:39-51.
  53. Cannarile MA, Weisser M, Jacob W, Jegg AM, Ries CH, Ruttinger D. Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy. J Immunother Cancer 2017;5:53.
  54. Jenrow KA, Brown SL, Lapanowski K, Naei H, Kolozsvary A, Kim JH. Selective inhibition of microglia-mediated neuroinflammation mitigates radiation-induced cognitive impairment. Radiat Res 2013;179:549-56.
  55. Jenrow KA, Brown SL, Kolozsvary AJ, Lapanowski K, Kim JH. Time-dependent inhibition of pan-inflammatory cytokines mitigates radiation-induced skin injury in mice. Radiat Res 2014;182:316-21.
  56. Acharya MM, Green KN, Allen BD, et al. Elimination of microglia improves cognitive function following cranial irradiation. Sci Rep 2016;6:31545.
  57. Vanpouille-Box C, Diamond JM, Pilones KA, et al. $TGF{\beta}$ is a master regulator of radiation therapy-induced antitumor immunity. Cancer Res 2015;75:2232-42.
  58. Ehrhart EJ, Segarini P, Tsang ML, Carroll AG, Barcellos-Hoff MH. Latent transforming growth factor beta1 activation in situ: quantitative and functional evidence after low-dose gamma-irradiation. FASEB J 1997;11:991-1002.
  59. Anscher MS, Marks LB, Shafman TD, et al. Risk of long-term complications after TFG-beta1-guided very-high-dose thoracic radiotherapy. Int J Radiat Oncol Biol Phys 2003;56:988-95.
  60. Tsushima H, Kawata S, Tamura S, et al. High levels of transforming growth factor beta 1 in patients with colorectal cancer: association with disease progression. Gastroenterology 1996;110:375-82.
  61. Morris JC, Tan AR, Olencki TE, et al. Phase I study of GC1008 (fresolimumab): a human anti-transforming growth factorbeta ($TGF{\beta}$) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PLoS One 2014;9:e90353.
  62. Bouquet F, Pal A, Pilones KA, er al. $TGF{\beta}1$ inhibition increases the radiosensitivity of breast cancer cells in vitro and promotes tumor control by radiation in vivo. Clin Cancer Res 2011;17:6754-65.
  63. Formenti SC, Lee P, Adams S, et al. Focal irradiation and systemic $TGF{\beta}$ Blockade in metastatic breast cancer. Clin Cancer Res 2018;24:2493-504.
  64. Liang H, Deng L, Hou Y, et al. Host STING-dependent MDSC mobilization drives extrinsic radiation resistance. Nat Commun 2017;8:1736.
  65. Son CH, Bae JH, Shin DY, et al. Combination effect of regulatory T-cell depletion and ionizing radiation in mouse models of lung and colon cancer. Int J Radiat Oncol Biol Phys 2015;92:390-8.
  66. Kim JH, Jenrow KA, Brown SL. Mechanisms of radiationinduced normal tissue toxicity and implications for future clinical trials. Radiat Oncol J 2014;32:103-15.
  67. Sharlow ER, Leimgruber S, Lira A, et al. A small molecule screen exposes mTOR signaling pathway involvement in radiation-induced apoptosis. ACS Chem Biol 2016;11:1428-37.
  68. Lawrence J, Nho R. The role of the mammalian target of rapamycin (mTOR) in pulmonary fibrosis. Int J Mol Sci 2018;19:E778.
  69. Taniguchi CM, Miao YR, Diep AN, et al. PHD inhibition mitigates and protects against radiation-induced gastrointestinal toxicity via HIF2. Sci Transl Med 2014;6:236ra64.
  70. Olcina MM, Giaccia AJ. Reducing radiation-induced gastrointestinal toxicity: the role of the PHD/HIF axis. J Clin Invest 2016;126:3708-15.
  71. Richardson RB. Ionizing radiation and aging: rejuvenating an old idea. Aging (Albany NY) 2009;1:887-902.
  72. Wang Y, Schulte BA, LaRue AC, Ogawa M, Zhou D. Total body irradiation selectively induces murine hematopoietic stem cell senescence. Blood 2006;107:358-66.
  73. Day RM, Snow AL, Panganiban RA. Radiation-induced accelerated senescence: a fate worse than death? Cell Cycle 2014;13:2011-2.
  74. Wang Y, Boerma M, Zhou D. Ionizing radiation-induced endothelial cell senescence and cardiovascular diseases. Radiat Res 2016;186:153-61.
  75. Pan J, Li D, Xu Y, et al. Inhibition of Bcl-2/xl with ABT-263 selectively kills senescent type II pneumocytes and reverses persistent pulmonary fibrosis induced by ionizing radiation in mice. Int J Radiat Oncol Biol Phys 2017;99:353-61.
  76. Panganiban RA, Day RM. Inhibition of IGF-1R prevents ionizing radiation-induced primary endothelial cell senescence. PLoS One 2013;8:e78589.
  77. Kim JH, Kolozsavry A, Brown SL, et al. Senolytics mitigate radiation-induced normal tissue damage. Proceedings of the 64th Annual Radiation Research Society Meeting; 2018 Sep 23-26; Chicago, IL.
  78. Kirkland JL, Tchkonia T. Cellular senescence: a translational perspective. EBioMedicine 2017;21:21-8.
  79. Fuhrmann-Stroissnigg H, Ling YY, Zhao J, et al. Identification of HSP90 inhibitors as a novel class of senolytics. Nat Commun 2017;8:422.

Cited by

  1. Targeted Therapies and Immune-Checkpoint Inhibition in Head and Neck Squamous Cell Carcinoma: Where Do We Stand Today and Where to Go? vol.11, pp.4, 2019, https://doi.org/10.3390/cancers11040472
  2. Clinical significance of radiotherapy before and/or during nivolumab treatment in hepatocellular carcinoma vol.8, pp.16, 2018, https://doi.org/10.1002/cam4.2570
  3. Clinical Significance of Systemic Inflammation Markers in Newly Diagnosed, Previously Untreated Hepatocellular Carcinoma vol.12, pp.5, 2018, https://doi.org/10.3390/cancers12051300
  4. MnTnHex-2-PyP5+, Coupled to Radiation, Suppresses Metastasis of 4T1 and MDA-MB-231 Breast Cancer via AKT/Snail/EMT Pathways vol.10, pp.11, 2018, https://doi.org/10.3390/antiox10111769