Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 May 3.
Published in final edited form as: Int J Radiat Oncol Biol Phys. 2018 Jun 1;101(2):267–270. doi: 10.1016/j.ijrobp.2017.04.005

Current Opportunities and Future Vision of Precision Medicine in Radiation Oncology

David G Kirsch 1
PMCID: PMC6499377  NIHMSID: NIHMS1025554  PMID: 29726356

At the 2016 ASTRO annual meeting, Dr. Thomas Lynch delivered a keynote address where he presented the remarkable development of targeted therapy for non-small cell lung cancer with activating mutations in the epidermal growth factor receptor (EGFR)1. He described the care of one patient, whose lung cancer had an exceptional response to a first-generation EGFR inhibitor, but ultimately progressed with tumor cells harboring a resistant EGFR mutation. The patient subsequently derived benefit from a next-generation EGFR inhibitor. The use of small molecule inhibitors to treat EGFR mutant lung adenocarcinoma was the culmination of years of basic science and translational research2,3. It was also an example of precision medicine in oncology, where therapy is tailored to a specific gene mutation in an individual patient’s cancer. After the plenary session, a somewhat deflated colleague asked me when this kind of precision medicine would come to radiation oncology.

From one perspective, the current practice of radiation oncology already encompasses the central tenets of precision medicine. During treatment planning, radiation oncologists contour patient-specific tumor and normal tissue anatomy. Beam angles, shapes, and energies are designed to maximize the therapeutic ratio. Radiation oncologists select optimal treatment plans based on patient-specific dose-volume histograms of normal tissues and target volume coverage. Furthermore, individual patient and tumor characteristics factor into determining which patients are offered radiation therapy. For example, the treatment approach for a 75 year-old woman with a T1 ER+/PR+ invasive ductal adenocarcinoma may include lumpectomy and an aromatase inhibitor without adjuvant radiation therapy4, while adjuvant radiation therapy and trastuzumab would routinely be recommended to a 40 year-old woman with Her2 amplified T1 invasive ductal adenocarcinoma after lumpectomy. Similarly, in prostate cancer, the treatment for individual patients can vary from active surveillance to radiation therapy alone to radiotherapy plus hormonal therapy depending on a number of clinical (age, co-morbidities, etc.) and tumor-specific (PSA, Gleason score, stage) parameters.

However, from another perspective, it can feel like radiation oncology is failing to incorporate the tremendous advances in cancer genomics and cancer biology into individual patient treatments. Instead, when a patient is offered radiation therapy, the same radiation dose is typically delivered regardless of the molecular features of the individual cancer.

Others have recently highlighted the opportunity to widen the therapeutic window of radiation therapy in the era of precision medicine by combining technology-driven improvement of treatment conformality with biology-driven approaches5. In this issue of the Red Journal, authors discuss how advances in genomics6, imaging7, and “Big Data”8 can be leveraged to tailor radiation therapy to individual patients in an effort to improve outcomes. While these and other approaches to precision medicine should be tested to try to improve radiation therapy, the limits to personalized cancer medicine that arise from tumor heterogeneity9 must be appreciated to avoid what some have recently concluded is a state of irrational exuberance in precision medicine for cancer10.

Although the concept of performing genomic sequencing to identify an “actionable mutation” that can then be paired with a molecularly targeted drug remains appealing, thus far DNA-guided precision medicine has benefited only a small subset of cancers10. Most responders to date have tumors with a gain-of-function oncogenic mutation or an overexpressed protein driving tumor growth and maintenance. In addition to EGFR mutant lung adenocarcinoma and Her2 amplified breast cancer, successful applications of DNA-guided targeted therapy include EML4-ALK positive lung adenocarcinoma, BRAFV600E mutant melanoma, and c-kit mutant GI stromal tumors (GISTs), which show dramatic responses to crizotinib, vemurafenib, and imatinib respectively. However, in this subgroup of cancers, most patients with macroscopic metastases develop resistance to targeted therapy because of intratumor heterogeneity and the selection of pre-existing resistant subclones9. Remarkably, the presence of a gain-of-function mutation does not always cause sensitivity to a molecularly targeted drug. For example, the presence of the identical BRAFV600E mutation in colorectal cancer does not lead to the same sensitivity to vemurafenib as in melanoma11, apparently because of EGFR signaling12. Furthermore, a randomized controlled trial of patients with metastatic cancer harboring a targetable mutation showed no difference in progression-free survival with matched molecularly targeted therapy compared to physician’s choice of treatment13. Therefore, while the success of DNA-guided targeted therapy in certain cancers with specific driver mutations should be celebrated, the value of genomic sequencing for identifying targeted therapies cannot necessarily be extrapolated to other cancers. The implication for precision medicine in radiation oncology is that specific gene mutations, gene expression patterns14, or imaging biomarkers5 should be studied to try to identify radiation-sensitive and radiation-resistant tumors. However, the identification of a useful biomarker for one type of cancer may not necessarily translate to other cancers. Moreover, tumor heterogeneity has the potential to limit our ability to tailor radiation therapy to individual patients.

Opportunities for Precision Medicine in Radiation Oncology: Selecting Individual Patients for Adjuvant Radiotherapy

Many patients with non-metastatic cancer are treated with surgery and adjuvant radiation therapy. Randomized clinical trials have established the value of adjuvant radiation therapy for specific populations of cancer patients by increasing local control and in some scenarios overall survival. However, in these trials over half of the patients randomized to surgery alone achieve local control1517. For these patients, radiation therapy adds toxicity and cost without any benefit. Therefore, there is an opportunity to personalize the decision to deliver adjuvant radiation therapy to those patients at highest risk for local recurrence. For example, intraoperative imaging of microscopic residual cancer in the tumor bed has the potential to be used to stratify patients for adjuvant radiation therapy18. Such intraoperative imaging technology is now being tested in early phase clinical trials19,20, and could be combined with clinical and/or pathological features to help guide the decision of postoperative radiation therapy. Just as patients with localized breast cancer are now selected for adjuvant chemotherapy based in part on a gene expression score, similar tests of resected breast cancers21 and prostate cancers22 could be used to tailor adjuvant radiation therapy to those patients most likely to benefit. Although these approaches for precision radiation oncology have the potential to refine the selection of individual patients for adjuvant radiation therapy, prospective testing and validation in well-designed clinical trials will be required before they are broadly implemented in the future.

Opportunities for Precision Medicine in Radiation Oncology: Prescribing Radiation Dose Based on Genetic Drivers of Individual Cancers

Genetic drivers of individual cancers also have the potential to be used for prescribing radiation dose. For example, the discovery that oropharyngeal squamous cell carcinomas associated with human papillomavirus (HPV) infection show significantly better rates of progression-free survival after chemo-radiotherapy than other oropharyngeal squamous cell carcinomas23 provides the rationale for ongoing clinical trials of treatment de-escalation for patients with HPV-associated head and neck cancer. HPV infection leads to the expression of several viral genes, including E6. The E6 protein binds to the tumor suppressor p53, which causes p53 degradation that in turn leads to p16 overexpression. Therefore, detecting expression of p16 in oropharyngeal cancer by immunohistochemistry is a surrogate for a tumor associated with HPV infection and identifies a subgroup of patients with better outcomes following chemo-radiotherapy23. While ongoing clinical investigations aim to identify which p16+ head and neck squamous cell carcinomas can safely be treated with less radiation therapy and/or chemotherapy5, HPV-associated head and neck cancer serves as a paradigm for using the genetic drivers of tumor development to tailor radiation therapy to individual patients. Another example of a driver mutation that is correlated with radioresistance is oncogenic KRAS in lung adenocarcinoma24 and in liver metastases25. Remarkably, the 1-year local control rate after stereotactic body radiation therapy (SBRT) for liver metastases with the combination of KRAS and TP53 mutations was only 20% compared to 69% for all other metastases25. These findings suggest that tumor genotype can be a critical determinant of local control. Moreover, these results suggest that to achieve optimal rates of local control in cancers with KRAS and TP53 mutations, treatment intensification may be required. For example, future clinical trials for liver metastases with KRAS and TP53 mutations could test increasing the radiation dose, utilizing alternative therapy such as surgery or radiofrequency ablation, or combining radiotherapy with concurrent chemotherapy or a drug that blocks the Ataxia Telangiectasia Mutated (ATM) kinase to increase radiosensitivity26. In addition to classifying tumor radiation resistance by genotype, gene expression can also be utilized to define a radiation sensitivity index that has the potential to be used to personalize radiotherapy dose prescription27.

Challenges for Precision Medicine in Radiation Oncology

As radiation oncology moves toward a future of precision medicine based in part on genomic features of the tumor, it is important to recognize the challenge of tumor heterogeneity where one cancer is comprised of multiple subclones, each harboring different mutations. When a tumor is biopsied or resected, genomic analysis may reflect the mutations or gene expression present in the dominant clone. It is possible that targeting these pathways with molecularly targeted drugs and radiation therapy will increase local control and cure before additional heterogeneity develops at sties of metastasis. However, a recent study comparing paired medulloblastomas at diagnosis and recurrence after radiotherapy with or without chemotherapy demonstrated substantial genetic divergence of the dominant clone at recurrence28. This study showed that the dominant clone at recurrence arose through clonal selection of a pre-existing minor clone present at diagnosis28. Remarkably, only a minority of the mutations present in the tumor at diagnosis were retained at recurrence. Therefore, to optimize precision medicine in radiation oncology in the future, cancer therapy will not only need to be tailored to treat the dominant clone, but will also need to eradicate minor clones poised to seed local and distant recurrence. One approach to addressing the challenge of tumor heterogeneity is to combine radiation therapy with immunotherapy to try to activate an immune response against different subclones within a cancer. While appreciating the problem of tumor heterogeneity increases the challenge of applying precision medicine to our cancer patients, it will facilitate the development of strategies with the best chance of improving patient outcomes.

Acknowledgements:

I thank Yvonne Mowery, MD, PhD for critically reading this manuscript.

D.G.K. is supported by grant 1R35 CA197616 from the National Cancer Institute.

Footnotes

Conflicts of Interest: DGK is a member of the scientific advisory board and owns stock in Lumicell Diagnostics, a company commercializing intraoperative imaging systems. DGK is a founder and owns stock in XRAD Therapeutics, which is developing radiosensitizers. DGK has research support from XRAD Therapeutics and Merck and has received research support in the past from GlaxoSmithKline and Janssen.

References

  • 1.Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, Louis DN, Christiani DC, Settleman J, Haber DA. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350(21):2129–2139. [DOI] [PubMed] [Google Scholar]
  • 2.Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer. 2007;7(3):169–181. [DOI] [PubMed] [Google Scholar]
  • 3.Wakeling AE, Guy SP, Woodburn JR, Ashton SE, Curry BJ, Barker AJ, Gibson KH. ZD1839 (Iressa): an orally active inhibitor of epidermal growth factor signaling with potential for cancer therapy. Cancer Res. 2002;62(20):5749–5754. [PubMed] [Google Scholar]
  • 4.Hughes KS, Schnaper LA, Bellon JR, Cirrincione CT, Berry DA, McCormick B, Muss HB, Smith BL, Hudis CA, Winer EP, Wood WC. Lumpectomy plus tamoxifen with or without irradiation in women age 70 years or older with early breast cancer: long-term follow-up of CALGB 9343. J Clin Oncol. 2013;31(19):2382–2387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Baumann M, Krause M, Overgaard J, Debus J, Bentzen SM, Daartz J, Richter C, Zips D, Bortfeld T. Radiation oncology in the era of precision medicine. Nat Rev Cancer. 2016;16(4):234–249. [DOI] [PubMed] [Google Scholar]
  • 6.Hall WA, Bergom C, Thompson RF, Baschnagel AM, Vijayakumar S, Willers H, Li XA, Schultz CJ, Wilson GD, West CML, Capala J, Coleman CN, Torres-Roca JF, Weidhaas J, Feng FY. Precision Oncology and Genomically Guided Radiation Therapy: A Report From the American Society for Radiation Oncology/American Association of Physicists in Medicine/National Cancer Institute Precision Medicine Conference. International Journal of Radiation Oncology, Biology, & Physics. 2018;101(2):274–284. [DOI] [PubMed] [Google Scholar]
  • 7.Jaffray DA, Das S, Jacobs PM, Jeraj R, Lambin P. How Advances in Imaging Will Affect Precision Radiation Oncology. International Journal of Radiation Oncology, Biology, & Physics. 2018;101(2):292–298. [DOI] [PubMed] [Google Scholar]
  • 8.McNutt TR, Benedict SH, Low DA, Moore K, Shpitser I, Jiang W, Lakshminarayanan P, Cheng Z, Han P, Hui X, Nakatsugawa M, Lee J, Moore JA, Robertson SP, Shah V, Taylor R, Quon H, Wong J, DeWeese T. Using Big Data Analytics to Advance Precision Radiation Oncology. International Journal of Radiation Oncology, Biology, & Physics. 2018;101(2):285–291. [DOI] [PubMed] [Google Scholar]
  • 9.Tannock IF, Hickman JA. Limits to Personalized Cancer Medicine. N Engl J Med. 2016;375(13):1289–1294. [DOI] [PubMed] [Google Scholar]
  • 10.Voest EE, Bernards R. DNA-Guided Precision Medicine for Cancer: A Case of Irrational Exuberance? Cancer Discov. 2016;6(2):130–132. [DOI] [PubMed] [Google Scholar]
  • 11.Kopetz S, Desai J, Chan E, Hecht JR, O’Dwyer PJ, Maru D, Morris V, Janku F, Dasari A, Chung W, Issa JP, Gibbs P, James B, Powis G, Nolop KB, Bhattacharya S, Saltz L. Phase II Pilot Study of Vemurafenib in Patients With Metastatic BRAF-Mutated Colorectal Cancer. J Clin Oncol. 2015;33(34):4032–4038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, Beijersbergen RL, Bardelli A, Bernards R. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature. 2012;483(7387):100–103. [DOI] [PubMed] [Google Scholar]
  • 13.Le Tourneau C, Delord JP, Goncalves A, Gavoille C, Dubot C, Isambert N, Campone M, Tredan O, Massiani MA, Mauborgne C, Armanet S, Servant N, Bieche I, Bernard V, Gentien D, Jezequel P, Attignon V, Boyault S, Vincent-Salomon A, Servois V, Sablin MP, Kamal M, Paoletti X, investigators S. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol. 2015;16(13):1324–1334. [DOI] [PubMed] [Google Scholar]
  • 14.Eschrich SA, Fulp WJ, Pawitan Y, Foekens JA, Smid M, Martens JW, Echevarria M, Kamath V, Lee JH, Harris EE, Bergh J, Torres-Roca JF. Validation of a radiosensitivity molecular signature in breast cancer. Clin Cancer Res. 2012;18(18):5134–5143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Fisher B, Anderson S, Bryant J, Margolese RG, Deutsch M, Fisher ER, Jeong JH, Wolmark N. Twenty-year follow-up of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. N Engl J Med. 2002;347(16):1233–1241. [DOI] [PubMed] [Google Scholar]
  • 16.Kapiteijn E, Marijnen CA, Nagtegaal ID, Putter H, Steup WH, Wiggers T, Rutten HJ, Pahlman L, Glimelius B, van Krieken JH, Leer JW, van de Velde CJ, Dutch Colorectal Cancer G. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer. N Engl J Med. 2001;345(9):638–646. [DOI] [PubMed] [Google Scholar]
  • 17.Pisters PW, Harrison LB, Leung DH, Woodruff JM, Casper ES, Brennan MF. Long-term results of a prospective randomized trial of adjuvant brachytherapy in soft tissue sarcoma. J Clin Oncol. 1996;14(3):859–868. [DOI] [PubMed] [Google Scholar]
  • 18.Whitley MJ, Weissleder R, Kirsch DG. Tailoring Adjuvant Radiation Therapy by Intraoperative Imaging to Detect Residual Cancer. Semin Radiat Oncol. 2015;25(4):313–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Whitley MJ, Cardona DM, Lazarides AL, Spasojevic I, Ferrer JM, Cahill J, Lee CL, Snuderl M, Blazer DG 3rd, Hwang ES, Greenup RA, Mosca PJ, Mito JK, Cuneo KC, Larrier NA, O’Reilly EK, Riedel RF, Eward WC, Strasfeld DB, Fukumura D, Jain RK, Lee WD, Griffith LG, Bawendi MG, Kirsch DG, Brigman BE. A mouse-human phase 1 co-clinical trial of a protease-activated fluorescent probe for imaging cancer. Sci Transl Med. 2016;8(320):320ra324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.van Dam GM, Themelis G, Crane LM, Harlaar NJ, Pleijhuis RG, Kelder W, Sarantopoulos A, de Jong JS, Arts HJ, van der Zee AG, Bart J, Low PS, Ntziachristos V. Intraoperative tumor-specific fluorescence imaging in ovarian cancer by folate receptor-alpha targeting: first in-human results. Nat Med. 2011;17(10):1315–1319. [DOI] [PubMed] [Google Scholar]
  • 21.Mamounas EP, Liu Q, Paik S, Baehner FL, Tang G, Jeong JH, Kim SR, Butler SM, Jamshidian F, Cherbavaz DB, Sing AP, Shak S, Julian TB, Lembersky BC, Wickerham DL, Costantino JP, Wolmark N. 21-Gene Recurrence Score and Locoregional Recurrence in Node-Positive/ER-Positive Breast Cancer Treated With Chemo-Endocrine Therapy. J Natl Cancer Inst. 2017;109(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhao SG, Chang SL, Spratt DE, Erho N, Yu M, Ashab HA, Alshalalfa M, Speers C, Tomlins SA, Davicioni E, Dicker AP, Carroll PR, Cooperberg MR, Freedland SJ, Karnes RJ, Ross AE, Schaeffer EM, Den RB, Nguyen PL, Feng FY. Development and validation of a 24-gene predictor of response to postoperative radiotherapy in prostate cancer: a matched, retrospective analysis. Lancet Oncol. 2016;17(11):1612–1620. [DOI] [PubMed] [Google Scholar]
  • 23.Ang KK, Harris J, Wheeler R, Weber R, Rosenthal DI, Nguyen-Tan PF, Westra WH, Chung CH, Jordan RC, Lu C, Kim H, Axelrod R, Silverman CC, Redmond KP, Gillison ML. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 2010;363(1):24–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wang M, Han J, Marcar L, Black J, Liu Q, Li X, Nagulapalli K, Sequist LV, Mak RH, Benes CH, Hong TS, Gurtner K, Krause M, Baumann M, Kang JX, Whetstine J, Willers H. Radiation resistance in KRAS-mutated lung cancer is enabled by stem-like properties mediated by an osteopontin-EGFR pathway. Cancer Res. 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hong TS WJ, Borger DR, Yeap BY, McDonnell EI, Willers H, Blaszkowsky LS, Kwak EL, Allen JN, Clark JW et al. Phase II study of proton-based stereotactic body radiation therapy for liver metastases: Importance of tumor genotype J Natl Cancer Inst. 2017;In Press. [DOI] [PubMed] [Google Scholar]
  • 26.Moding EJ, Castle KD, Perez BA, Oh P, Min HD, Norris H, Ma Y, Cardona DM, Lee CL, Kirsch DG. Tumor cells, but not endothelial cells, mediate eradication of primary sarcomas by stereotactic body radiation therapy. Sci Transl Med. 2015;7(278):278ra234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Scott JG, Berglund A, Schell MJ, Mihaylov I, Fulp WJ, Yue B, Welsh E, Caudell JJ, Ahmed K, Strom TS, Mellon E, Venkat P, Johnstone P, Foekens J, Lee J, Moros E, Dalton WS, Eschrich SA, McLeod H, Harrison LB, Torres-Roca JF. A genome-based model for adjusting radiotherapy dose (GARD): a retrospective, cohort-based study. Lancet Oncol. 2017;18(2):202–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Morrissy AS, Garzia L, Shih DJ, Zuyderduyn S, Huang X, Skowron P, Remke M, Cavalli FM, Ramaswamy V, Lindsay PE, Jelveh S, Donovan LK, Wang X, Luu B, Zayne K, Li Y, Mayoh C, Thiessen N, Mercier E, Mungall KL, Ma Y, Tse K, Zeng T, Shumansky K, Roth AJ, Shah S, Farooq H, Kijima N, Holgado BL, Lee JJ, Matan-Lithwick S, Liu J, Mack SC, Manno A, Michealraj KA, Nor C, Peacock J, Qin L, Reimand J, Rolider A, Thompson YY, Wu X, Pugh T, Ally A, Bilenky M, Butterfield YS, Carlsen R, Cheng Y, Chuah E, Corbett RD, Dhalla N, He A, Lee D, Li HI, Long W, Mayo M, Plettner P, Qian JQ, Schein JE, Tam A, Wong T, Birol I, Zhao Y, Faria CC, Pimentel J, Nunes S, Shalaby T, Grotzer M, Pollack IF, Hamilton RL, Li XN, Bendel AE, Fults DW, Walter AW, Kumabe T, Tominaga T, Collins VP, Cho YJ, Hoffman C, Lyden D, Wisoff JH, Garvin JH Jr., Stearns DS, Massimi L, Schuller U, Sterba J, Zitterbart K, Puget S, Ayrault O, Dunn SE, Tirapelli DP, Carlotti CG, Wheeler H, Hallahan AR, Ingram W, MacDonald TJ, Olson JJ, Van Meir EG, Lee JY, Wang KC, Kim SK, Cho BK, Pietsch T, Fleischhack G, Tippelt S, Ra YS, Bailey S, Lindsey JC, Clifford SC, Eberhart CG, Cooper MK, Packer RJ, Massimino M, Garre ML, Bartels U, Tabori U, Hawkins CE, Dirks P, Bouffet E, Rutka JT, Wechsler-Reya RJ, Weiss WA, Collier LS, Dupuy AJ, Korshunov A, Jones DT, Kool M, Northcott PA, Pfister SM, Largaespada DA, Mungall AJ, Moore RA, Jabado N, Bader GD, Jones SJ, Malkin D, Marra MA, Taylor MD. Divergent clonal selection dominates medulloblastoma at recurrence. Nature. 2016;529(7586):351–357. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES