Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 May 1.
Published in final edited form as: Int J Radiat Oncol Biol Phys. 2014 May 1;89(1):7–9. doi: 10.1016/j.ijrobp.2014.01.045

Acceler-Dated Fractionation – the End of the Era of the Large ‘One size Fits All’ Trial for Locally Advanced Head and Neck Cancer

Quynh-Thu Le 1,*, Mitchell Machtay 2
PMCID: PMC3986724  NIHMSID: NIHMS562575  PMID: 24725684

Repair, accelerated repopulation, re-oxygenation and re-assortment (cell cycle) are the four tenets of classical radiation biology. They form the cornerstones for designing and optimizing radiation fractionation schemas in head and neck squamous cell carcinoma, particularly locally advanced head and neck cancer (LAHNC), in the last century. Accelerated fractionation targets tumor repopulation whereas classical hyperfractionation aims to increase the total dose while taking advantage of normal tissue repair. Because of the scientific rationale and positive results reported in the literature, the Radiation Therapy Oncology Group (RTOG) launched RTOG 9003, one of the largest randomized trials in LAHNC ever performed. This study tested three different altered fractionation regimens against standard fractionation RT alone (SFX)1.

Early results of the trial showed that both hyperfractionation (HFX) and accelerated fractionation with concomitant boost (AFX-C) yielded significantly better locoregional control (LRC) and a trend for improved disease-free survival (DFS) when compared to SFX1. Both were associated with more acute toxicity but without a significant increase of physician-assessed late effects. In this latest update of RTOG 9003 (Beitler et al in the same issue), only HFX remained significantly associated with improved LRC, although a trend for higher LRC was still noted for AFX-C. Composite severe Grade 3–5 late toxicity appeared to trend higher for six versus seven weeks of treatment, though the difference was not statistically significant. Patient reported outcomes were not included in RTOG 9003.

What have we learned from the long-term update of RTOG 9003? First, the study confirms that late failures are uncommon in this population. Only 31 additional LRFs were recorded between the last report (data cut-point August 1999) and the current report (October 2012), justifying the practice of censoring the locoregional failure endpoint at 5 years. Beyond 5 years, a large number of deaths occurred from causes other than the original cancer; therefore, overall survival curves showed no significant benefit to altered fractionation over standard fractionation. This confirms data from meta-analyses showing that the benefit from altered fractionation over SFX is very modest, smaller than the benefit from concurrent chemoradiotherapy (CCRT), with the exception of pure HFX2. In an unselected population of LAHNC, accelerated fractionation alone, even with dose escalation, is probably inferior to CCRT; randomized trials from GORTEC and the German Cancer Society would appear to confirm that 3,4. Finally, late toxicity continues to plague our patients. About 8% (13 of 166) of 5-year survivors without disease are dependent on feeding tube use. An exploratory analysis suggested that 6-week (accelerated) treatment courses had greater late toxicity than 7-week treatment courses. Although late toxicity in RTOG 9003 appears to be somewhat lower than after CCRT, the rates are still considerable.

Given the LRC benefit of CCRT and altered fractionation and the survival benefit of CCRT, the next question is whether further gains can be achieved by combining both strategies. This was addressed in GORTEC 99-02, where accelerated RT (70 Gy in 6 weeks) plus 2 cycles of concurrent carboplatin-fluorouracil was compared to SFX (70 Gy in 7 weeks) plus three cycle of concurrent carboplatin-fluorouracil. AFX plus concurrent chemotherapy was not better; in fact the highest survival rate was noted for the SFX plus chemotherapy arm3. Similarly, RTOG 0129 compared SFX plus 3 cycles of high dose cisplatin to an experimental arm of AFX-C plus 2 cycles of cisplatin5. At a mean follow-up of 7.9 years, there was no demonstration of improved outcome for any endpoint6. Although AFX-C for one week may have compensated for the lower total cisplatin dose, accelerated RT plus concurrent chemotherapy was not shown to be superior to SFX plus concurrent RT. Based on these data, it is unlikely that future trials will compare various radiation fractionation schedules during CCRT in unselected groups of patients. Individual clinicians may opt to use AFX-C with slightly less chemotherapy than SFX plus a full-course chemotherapy – but they should recognize that the primary reason is for logistical grounds rather than improved outcomes.

Where are we going from here for LAHNC? Recent advances in molecular biology and next generation sequencing have taught us much about these tumors. We know now that there is a growing entity of human papilloma virus (HPV) related oropharyngeal carcinomas (OPC), and that they have a better prognosis than other LAHNC5. Many HPV-positive OPCs respond very well to standard chemoradiation therapy and may warrant treatment de-escalation (in the setting of clinical trials). This may include altered fractionation RT alone, or with less intense concurrent systemic therapy.

In addition, analogous to breast cancer, HNSCC might soon be classifiable into different subtypes (classical, mesenchymal, basal and atypical) with different behaviors based on their gene expression profiles7. We are also learning that tumor behavior is often influenced by the tumor microenvironment and the immune function within the tumor and the host8. Moreover, certain tumors have mutations (such as FGFR2-3 and PIK3K mutations) and aberrant pathway function (such as the PIK3K pathway) that may in the future be successfully targeted pharmacologically911.

Based on this newfound knowledge, risk stratification using a combination of clinical and biological features now forms the corner stones of RTOG clinical trials for HNSCC. As previously mentioned, for certain good risk HPV-positive OPCs (small-size T1-T3 tumors with limited nodal involvement in patients with no or minimal smoking history), the goal is for treatment de-intensification to minimize late toxicity while maintaining an outstanding cure rate. Clinical trials are being designed to address this intent. For the intermediate risk HPV-positive tumors (large T3-4 tumors or with extensive nodal involvement or in patients with more than 10 pack-year of cigarette use), the disease progression rate is still high (32% at 5-years for RTOG 0129 and 0522) 5,12. In these patients, the addition of an immune check point inhibitor (e.g. against CTLA4 or PD1) to CCRT has the theoretical benefit of overcoming the immune exhaustion that is often seen in these patients13 while eliciting antitumor immune response to antigens that are unmasked by RT14.

Immune checkpoint targeting may likewise benefit patients with HPV-negative LAHNC, since immune dysfunction is common in these patients15. In addition, targeting a specific active pathway or mutation in combination with CCRT may benefit the subsets of patients harboring such defect. Novel approaches at DNA damage, such as PARP inhibition, may be an option for patients who do not have an obviously ‘actionable’ molecular target. However, one needs to keep in mind that many HPV-negative HNSCC patients are quite frail and any treatment intensification needs to take into account the potential for severe acute and late treatment-related toxicity.

In summary, we are approaching a new paradigm in the management of SCCHN. Altered fractionation alone may continue to play a role in small groups of patients such as those who cannot tolerate concomitant chemotherapy or those with good risk HPV-positive tumor enrolled on clinical trials. The remaining patients will be partitioned into different risk groups based on clinical and biological features and will be treated with different strategies. New treatment paradigms need to take into consideration the late toxicity profile that may be experienced by our patients and new research to mitigate such late toxicity is needed to improve the quality of life in long-term survivors.

Acknowledgments

Supported by grants RTOG U10-CA21661, CCOP U10-CA37422 from the National Cancer Institute. This manuscript's contents are the sole responsibility of the authors and do not necessarily represent the official views of the NCI.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflicts of Interest: None

References

  • 1.Fu KK, Pajak TF, Trotti A, et al. A Radiation Therapy Oncology Group (RTOG) phase III randomized study to compare hyperfractionation and two variants of accelerated fractionation to standard fractionation radiotherapy for head and neck squamous cell carcinomas: first report of RTOG 9003. Int J Radiat Oncol Biol Phys. 2000;48:7–16. doi: 10.1016/s0360-3016(00)00663-5. [DOI] [PubMed] [Google Scholar]
  • 2.Bourhis J, Overgaard J, Audry H, et al. Hyperfractionated or accelerated radiotherapy in head and neck cancer: a meta-analysis. Lancet. 2006;368:843–854. doi: 10.1016/S0140-6736(06)69121-6. [DOI] [PubMed] [Google Scholar]
  • 3.Bourhis J, Sire C, Graff P, et al. Concomitant chemoradiotherapy versus acceleration of radiotherapy with or without concomitant chemotherapy in locally advanced head and neck carcinoma (GORTEC 99-02): an open-label phase 3 randomised trial. Lancet Oncol. 2012;13:145–153. doi: 10.1016/S1470-2045(11)70346-1. [DOI] [PubMed] [Google Scholar]
  • 4.Budach V, Stuschke M, Budach W, et al. Hyperfractionated accelerated chemoradiation with concurrent fluorouracil-mitomycin is more effective than dose-escalated hyperfractionated accelerated radiation therapy alone in locally advanced head and neck cancer: final results of the radiotherapy cooperative clinical trials group of the German Cancer Society 95-06 Prospective Randomized Trial. J Clin Oncol. 2005;23:1125–1135. doi: 10.1200/JCO.2005.07.010. [DOI] [PubMed] [Google Scholar]
  • 5.Ang KK, Harris J, Wheeler R, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. The New England journal of medicine. 2010;363:24–35. doi: 10.1056/NEJMoa0912217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Nguyen-Tan PF, Zhang Q, Wheeler RH, et al. A Randomized PhaseIII Trial to Test Accelerated Versus Standard Fractionation in Combination With Concurrent Cisplatin for Head and Neck Carcinomas (RTOG 0129): Long-term Report of Efficacy and Toxicity [Abstract] Int J Radiat Oncol Biol Phys. 2013;87:S56–S57. [Google Scholar]
  • 7.Chung CH, Parker JS, Karaca G, et al. Molecular classification of head and neck squamous cell carcinomas using patterns of gene expression. Cancer Cell. 2004;5:489–500. doi: 10.1016/s1535-6108(04)00112-6. [DOI] [PubMed] [Google Scholar]
  • 8.Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19:1423–1437. doi: 10.1038/nm.3394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lui VW, Hedberg ML, Li H, et al. Frequent mutation of the PI3K pathway in head and neck cancer defines predictive biomarkers. Cancer Discov. 2013;3:761–769. doi: 10.1158/2159-8290.CD-13-0103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Keck MK, Zuo Z, Khattri A, et al. Genomic profiling of kinase genes in head and neck squamous cell carcinomas to identify potentially targetable genetic aberrations in FGFR1/2, DDR2, EPHA2, and PIK3CA [Abstract] J Clin Oncol. 2013;31 (suppl; abstr 6010) [Google Scholar]
  • 11.Hayes DN, Grandis JR, El-Naggar AK. The Cancer Genome Atlas: Integrated analysis of genome alterations in squamous cell carcinoma of the head and neck [Abstract] J Clin Oncol. 2013;31 (suppl; abstr 6009) [Google Scholar]
  • 12.Gillison ML, Zhang Q, Jordan R, et al. Tobacco smoking and increased risk of death and progression for patients with p16-positive and p16-negative oropharyngeal cancer. J Clin Oncol. 2012;30:2102–2111. doi: 10.1200/JCO.2011.38.4099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Gildener-Leapman N, Lee J, Ferris RL. Tailored immunotherapy for HPV positive head and neck squamous cell cancer. Oral Oncol. 2013 doi: 10.1016/j.oraloncology.2013.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Postow MA, Callahan MK, Barker CA, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med. 2012;366:925–931. doi: 10.1056/NEJMoa1112824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Molling JW, Langius JA, Langendijk JA, et al. Low levels of circulating invariant natural killer T cells predict poor clinical outcome in patients with head and neck squamous cell carcinoma. J Clin Oncol. 2007;25:862–868. doi: 10.1200/JCO.2006.08.5787. [DOI] [PubMed] [Google Scholar]

RESOURCES