PDF version

Recent advances in technological aspects of radiotherapy have paved the way for personalised treatment as they enable the tailoring of radiation doses to individual patients. However, the current optimisation process is primarily focused on dose conformity without taking into account biological factors and it assumes uniform tumour responses to radiation [1].

In medical oncology, genomic signatures have been incorporated into routine practice with tests such as MammaPrint, Oncotype DX, or PAM50. However, the use of such signatures in radiotherapy has been limited. Until now, there has been scarce information available regarding molecular predictive signatures in the field. For instance, the postoperative radiation therapy outcomes score (PORTOS) consists of 24 genes and is used to forecast the effectiveness of postoperative radiotherapy in prostate cancer [2]. Additionally, there are two other classifiers, which are known as the adjuvant radiotherapy intensification classifier (ARTIC) and the profile for the omission of local adjuvant radiotherapy (POLAR). These incorporate 27 and 16 genes respectively. These classifiers have been developed to predict outcomes of the use of postoperative radiotherapy in breast cancer [3,4].

In contrast to cancer-specific indicators such as these, an alternative measure known as the radiosensitivity index (RSI) has been proposed as a universal marker of cellular radiosensitivity that can be applied across various types of cancer [5]. The RSI is calculated based on the expression levels of ten specific genes (AR, c-JUN, STAT1, PKC, Rel A, cABL, SUMO1, CDK1, HDAC1 and IRF1), which are associated with DNA damage-response mechanisms such as cell-cycle regulation, apoptosis induction and proliferation modulation [5]. To enhance its practical applicability in clinical settings for radiotherapy planning purposes, the genomic-adjusted radiation dose (GARD) concept has been developed through the use of RSI values [6].

As an initial validation step, GARD was evaluated in patient cohorts that had been diagnosed with breast, lung and pancreatic cancer, and glioblastoma [6]. Subsequent studies were conducted on a pooled, retrospective sample that encompassed multiple tumour variations. These studies produced new evidence that demonstrated comprehensively that GARD exhibited a strong correlation with key oncological outcomes, such as time until first recurrence and overall survival rates [7]. In another recent study, the use of GARD was explored to explain the outcomes observed in the Radiation Therapy Oncology Group (RTOG) 0617 trial [8]. This model was designed to determine an optimal radiation dose for each patient. In other research that utilised prospectively collected tissue, a correlation was found between low RSI values (which indicate higher radiosensitivity) and increased immune infiltration and activation [9]. Furthermore, a reanalysis of publicly available datasets demonstrated that RSI was associated with immune-related characteristics and could be used to predict responses to blockade therapy that was targeted at the programmed cell-death protein 1 (PD-1) [10-12]. In contrast, another analysis suggested that the application of RSI did not impact survival rates and should not be used to adjust radiation doses [13]. Instead, it has been proposed that evaluation of the tumour clones that remain after radiotherapy may provide better predictions regarding treatment outcomes [14].

Recent studies have furthered the integration of genomic signatures into radiotherapy decision-making (see Table 1). The European Organisation for Research and Treatment of Cancer has recognised the importance of RSI/GARD studies and has prioritised them for phase III clinical trials in radiotherapy [15]. A notable example of such an approach is the ongoing GARD-based trial, the aim of which is to optimise radiotherapy for triple-negative breast cancer (NCT05528133). Further evaluation is necessary to assess the clinical utility of these approaches by integrating molecular data into prospective clinical trials and routine practice.

 

 

 

Table 1. Summary of clinical studies that have investigated the use of radiosensitivity-predicting genomic signatures (adapted from [16], with permission). Key: 95%CI – 95% confidence interval, HR – hazard ratio, sHR – sub-distribution hazard ratio.

 

Study

Cancer type

Sample size

Main findings

Zhao et al., 2016 [2]

Prostate cancer

526 patients (196 and 330 in training and validation cohorts, respectively)

24-gene predictor of response to postoperative radiotherapy. High PORTOS score predicts a lower incidence of distant metastases in both training (HR 0.12, 95% CI 0.03-0.41, p<0.0001) and validation (HR 0.15, 95% CI 0.04-0.60, p=0.002) cohorts.

Tang et al., 2017 [17]

Sarcomas

253 patients from The Cancer Genome Atlas (TCGA)

26-gene radiosensitivity signature. Predicted radiosensitive patients had better overall survival than predicted non-radiosensitive patients (HR 0.07, p<0.001).

Cui et al., 2018 [18]

Breast cancer

948 and 1439 patients in training and validation cohorts, respectively (Molecular Taxonomy of Breast Cancer International Consortium)

34-gene radiosensitivity signature. Patients administered radiotherapy had better disease-specific survival than did those who did not in the radiation-sensitive group (HR 0.68, p=0.059); a reverse effect was observed in the radiation-resistant group (HR 1.53, p=0.059).

4-gene immune signature predictive of radiotherapy benefit. Patients who were administered radiotherapy had significantly better disease-specific survival in the immune-effective group (HR 0.46, p=0.0076), with no difference in disease-specific survival in the immune-defective group (HR 1.27, p=0.16).

Sjöström et al., 2019 [3]

Breast cancer

748 patients from the Swedish breast cancer group 91 radiotherapy (SweBCG91-RT) trial

ARTIC comprised 27 genes and patient age was prognostic for locoregional recurrence in breast cancer patients who were administered radiotherapy (HR 3.4, 95% CI 2.0-5.9, p<0.001) and was predictive of radiotherapy benefit (p=0.005). Patients with low ARTIC scores had a greater benefit from radiotherapy (HR 0.33, 95% CI 0.21-0.52, p<0.001) than those with high ARTIC scores (HR 0.73, 95% CI 0.44-1.2, p=0.23).

Kim et al., 2020 [19]

Head and neck squamous cell carcinomas that were negative for human papillomavirus

203 patients from TCGA cohort

Use of 41-gene radiosensitivity signature predicted reduced five-year recurrence-free survival in the radioresistant group versus the radiosensitive group (57.8% vs. 80.1%; p=0.035)

Scott et al., 2021 [20]

Various types (breast, head and neck, non-small-cell lung, pancreatic and endometrial cancers, melanoma and glioma)

1615 patients, of whom 1298 (982 and 316 with and without radiotherapy, respectively) were assessed for time to first recurrence and 677 (424 and 253 with and without radiotherapy, respectively) for overall survival

GARD was associated with time to first recurrence (HR 0.98, 95% CI 0.97-0.99, p=0.0017) and overall survival (HR 0.97, 95% CI 0.95-0.99, p=0.0007). The effect on overall survival was dependent on radiotherapy use (p=0.011).

Feng et al., 2021 [21]

Prostate cancer

486 of 760 patients randomised in NRG Oncology/RTOG 9601 trial

22-gene genomic classifier (Decipher Biosciences Inc) was associated with distant metastases (HR 1.17, 95% CI 1.05-1.32, p=0.006), prostate cancer-specific mortality (HR 1.39, 95% CI 1.20-1.63, p<0.001) and overall survival (HR 1.17, 95% CI 1.06-1.29, p=0.002).

Dal Pra et al., 2022 [22]

Prostate cancer

226 of 350 patients randomised in the Swiss Group for Clinical Cancer Research (SAKK) 09/10 trial

22-gene genomic classifier (Decipher Biosciences Inc) was associated with biochemical progression (HR 2.26, 95% CI 1.42-3.60, p<0.001), clinical progression (HR 2.29, 95% CI 1.32-3.98, p=0.003) and use of hormone therapy (sHR 2.99, 95% CI 1.55-5.76, p=0.001). Patients with high and low classifier scores had 45% and 71% five-year freedom from biochemical progression, respectively.

Wu et al., 2022 [23]

Gliomas

1395 from Chinese Glioma Genome Atlas and TCGA

12-gene radiosensitivity predictive index had better predictive capacity than the traditional World Health Organization classification system (concordance index: 0.842 vs. 0.787, p≤2.2 × 10−16).

Sjöström et al., 2023 [4]

Breast cancer

729 patients from the SweBCG91-RT trial and Princess Margaret Hospital

16-gene POLAR signature used. POLAR low-risk patients did not benefit from the use of adjuvant radiotherapy (HR 1.1, 95% CI 0.39-3.40, p=0.81; HR 1.5, 95% CI 0.14-16, p=0.74). POLAR high-risk patients had a significantly lower risk of locoregional recurrence when radiotherapy was applied (HR 0.43, 95% CI 0.24-0.78, p=0.006; HR 0.25, 95% CI 0.07-0.92, p=0.038).

Spratt et al., 2023 [24]

Prostate cancer

215 patients from NRG Oncology/RTOG 0126

22-gene genomic classifier (Decipher Biosciences Inc) was independently prognostic for disease progression (sHR 1.12, 95% CI 1.00-1.26, p=0.04), biochemical failure (sHR 1.22, 95% CI 1.10-1.37, p<0.001), distant metastasis (sHR 1.28, 95% CI 1.06-1.55, p=0.01), and prostate cancer-specific mortality (sHR 1.45, 95% CI 1.20-1.76, p<0.001).

 

References:

  1. Price JM, Prabhakaran A and West CML (2023) Predicting Tumour Radiosensitivity to Deliver Precision Radiotherapy. Nature Reviews. Clinical Oncology 20, 83–98.
  2. Zhao SG, Chang SL, Spratt DE, Erho N, Yu M, Ashab HA-D, 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 and Feng FY (2016) Development and Validation of a 24-Gene Predictor of Response to Postoperative Radiotherapy in Prostate Cancer: A Matched, Retrospective Analysis. The Lancet. Oncology 17, 1612–1620.
  3. Sjöström M, Chang SL, Fishbane N, Davicioni E, Zhao SG, Hartman L, Holmberg E, Feng FY, Speers CW, Pierce LJ, Malmström P, Fernö M and Karlsson P (2019) Clinicogenomic Radiotherapy Classifier Predicting the Need for Intensified Locoregional Treatment After Breast-Conserving Surgery for Early-Stage Breast Cancer. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 37, 3340–3349.
  4. Sjöström M, Fyles A, Liu F-F, McCready D, Shi W, Rey-McIntyre K, Chang SL, Feng FY, Speers CW, Pierce LJ, Holmberg E, Fernö M, Malmström P and Karlsson P (2023) Development and Validation of a Genomic Profile for the Omission of Local Adjuvant Radiation in Breast Cancer. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 41, 1533–1540.
  5. Eschrich SA, Pramana J, Zhang H, Zhao H, Boulware D, Lee J-H, Bloom G, Rocha-Lima C, Kelley S, Calvin DP, Yeatman TJ, Begg AC and Torres-Roca JF (2009) A Gene Expression Model of Intrinsic Tumor Radiosensitivity: Prediction of Response and Prognosis after Chemoradiation. International Journal of Radiation Oncology, Biology, Physics 75, 489–496.
  6. 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 and Torres-Roca JF (2017) A Genome-Based Model for Adjusting Radiotherapy Dose (GARD): A Retrospective, Cohort-Based Study. The Lancet. Oncology 18, 202–211.
  7. Scott JG, Sedor Geoffrey, Ellsworth P, Scarborough JA, Ahmed KA, Oliver DE, Eschrich SA, Kattan MW and Torres-Roca JF (2021) Pan-Cancer Prediction of Radiotherapy Benefit Using Genomic-Adjusted Radiation Dose (GARD): A Cohort-Based Pooled Analysis. The Lancet. Oncology 22, 1221–1229.
  8. Scott JG, Sedor Geoff, Scarborough JA, Kattan MW, Peacock J, Grass GD, Mellon EA, Thapa R, Schell M, Waller A, Poppen S, Andl G, Teer JK, Eschrich SA, Dilling TJ, Dalton WS, Harrison LB, Fox T and Torres-Roca JF (2021) Personalizing Radiotherapy Prescription Dose Using Genomic Markers of Radiosensitivity and Normal Tissue Toxicity in NSCLC. Journal of Thoracic Oncology: Official Publication of the International Association for the Study of Lung Cancer 16, 428–438.
  9. Grass GD, Alfonso JCL, Welsh E, Ahmed KA, Teer JK, Pilon-Thomas S, Harrison LB, Cleveland JL, Mulé JJ, Eschrich SA, Enderling H and Torres-Roca JF (2022) The Radiosensitivity Index Gene Signature Identifies Distinct Tumor Immune Microenvironment Characteristics Associated With Susceptibility to Radiation Therapy. International Journal of Radiation Oncology, Biology, Physics 113, 635–647.
  10. Bin Lim S, Chua MLK, Yeong JPS, Tan SJ, Lim W-T and Lim CT (2019) Pan-Cancer Analysis Connects Tumor Matrisome to Immune Response. NPJ precision oncology 3, 15.
  11. Weinstein JN, Collisson EA, Mills GB, Shaw KM, Ozenberger BA, Ellrott K, Shmulevich I, Sander C and Stuart JM (2013) The Cancer Genome Atlas Pan-Cancer Analysis Project. Nature Genetics 45, 1113–1120.
  12. Dai Y-H, Wang Y-F, Shen P-C, Lo C-H, Yang J-F, Lin C-S, Chao H-L and Huang W-Y (2021) Radiosensitivity Index Emerges as a Potential Biomarker for Combined Radiotherapy and Immunotherapy. NPJ genomic medicine 6, 40.
  13. Mistry HB (2023) Radiosensitivity Index Is Not Fit to Be Used for Dose Adjustments: A Pan-Cancer Analysis. Clinical Oncology (Royal College of Radiologists (Great Britain)) S0936-6555(23)00084-5.
  14. Alfonso JCL and Berk L (2019) Modeling the Effect of Intratumoral Heterogeneity of Radiosensitivity on Tumor Response over the Course of Fractionated Radiation Therapy. Radiation Oncology (London, England) 14, 88.
  15. Thomas G, Eisenhauer E, Bristow RG, Grau C, Hurkmans C, Ost P, Guckenberger M, Deutsch E, Lacombe D, Weber DC, and European Organisation for Research and Treatment of Cancer (EORTC) (2020) The European Organisation for Research and Treatment of Cancer, State of Science in Radiation Oncology and Priorities for Clinical Trials Meeting Report. European Journal of Cancer (Oxford, England: 1990) 131, 76–88.
  16. Tomasik B, Garbicz F, Braun M, Bieńkowski M and Jassem J (2023). Heterogeneity in precision oncology. Cambridge Prisms: Precision Medicine, 1-37.
  17. Tang Z, Zeng Q, Li Y, Zhang X, Ma J, Suto MJ, Xu B and Yi N (2017) Development of a Radiosensitivity Gene Signature for Patients with Soft Tissue Sarcoma. Oncotarget 8, 27428–27439.
  18. Cui Y, Li B, Pollom EL, Horst KC and Li R (2018) Integrating Radiosensitivity and Immune Gene Signatures for Predicting Benefit of Radiotherapy in Breast Cancer. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research 24, 4754–4762.
  19. Kim SI, Kang JW, Noh JK, Jung HR, Lee YC, Lee JW, Kong M and Eun Y-G (2020) Gene Signature for Prediction of Radiosensitivity in Human Papillomavirus-Negative Head and Neck Squamous Cell Carcinoma. Radiation Oncology Journal 38, 99–108.
  20. Scott JG, Sedor Geoffrey, Ellsworth P, Scarborough JA, Ahmed KA, Oliver DE, Eschrich SA, Kattan MW and Torres-Roca JF (2021) Pan-Cancer Prediction of Radiotherapy Benefit Using Genomic-Adjusted Radiation Dose (GARD): A Cohort-Based Pooled Analysis. The Lancet. Oncology 22, 1221–1229.
  21. Feng FY, Huang H-C, Spratt DE, Zhao SG, Sandler HM, Simko JP, Davicioni E, Nguyen PL, Pollack A, Efstathiou JA, Dicker AP, Todorovic T, Margrave J, Liu YS, Dabbas B, Thompson DJS, Das R, Dignam JJ, Sweeney C, Attard G, Bahary J-P, Lukka HR, Hall WA, Pisansky TM, Shah AB, Pugh SL, Shipley WU and Tran PT (2021) Validation of a 22-Gene Genomic Classifier in Patients With Recurrent Prostate Cancer: An Ancillary Study of the NRG/RTOG 9601 Randomized Clinical Trial. JAMA oncology 7, 544–552.
  22. Dal Pra A, Ghadjar P, Hayoz S, Liu VYT, Spratt DE, Thompson DJS, Davicioni E, Huang H-C, Zhao X, Liu Y, Schär C, Gut P, Plasswilm L, Hölscher T, Polat B, Hildebrandt G, Müller A-C, Pollack A, Thalmann GN, Zwahlen D and Aebersold DM (2022) Validation of the Decipher Genomic Classifier in Patients Receiving Salvage Radiotherapy without Hormone Therapy after Radical Prostatectomy - an Ancillary Study of the SAKK 09/10 Randomized Clinical Trial. Annals of Oncology: Official Journal of the European Society for Medical Oncology 33, 950–958.
  23. Wu S, Xu J, Li G and Jin X (2022) Integrating Radiosensitivity Gene Signature Improves Glioma Outcome and Radiotherapy Response Prediction. Medicina (Kaunas, Lithuania) 58, 1327.
  24. Spratt DE, Liu VYT, Michalski J, Davicioni E, Berlin A, Simko JM, Efstathiou JA, Tran PT, Sandler HM, Hall WA, Thompson DJ, Parliament MB, Dayes IS, Correa RJM, Robertson JM, Gore EM, Doncals DE, Vigneault E, Souhami L, Karrison TG and Feng FY (2023) Genomic Classifier Performance in Intermediate-Risk Prostate Cancer: Results from NRG Oncology/RTOG 0126 Randomized Phase III Trial. International Journal of Radiation Oncology, Biology, Physics 117, 370-377.

Bartek Tomasik

Radiation oncologist

Department of Oncology and Radiotherapy, Faculty of Medicine

Medical University of Gdańsk

Gdańsk, Poland

bartlomiej.tomasik@gumed.edu.pl