49Radiotherapy dose fractionation Third edition
7.
Lung cancer
Background
Overall survival has increased in lung cancer in the past ten years, with the vast majority of
the gains occurring in disease stages I–III. There has been very little, if any improvement
seen in outcomes for stage IV patients.
1,2
Several publications have looked at access to
radiotherapy treatments (Level 2a).
3–5
Although many of these do not distinguish between
radical and palliative treatment, it appears that the proportion of lung cancer patients in the
UK accessing radiotherapy remains lower than expected.
Lung cancer staging has improved with routine use of positron emission tomography-
computed tomography (PET-CT) and endobronchial ultrasound (EBUS). Routine use
of intravenous (IV) contrast in planning has improved mediastinal target delineation.
Signicant technological advances have taken place in the delivery of radiotherapy. For
radical radiotherapy, four-dimensional computed tomography (4DCT) planning is replacing
three-dimensional conformal radiotherapy (3DCRT) as the standard of care. Bulky tumours
in certain anatomical locations, such as the paravertebral gutter, have improved dosimetry
with intensity-modulated radiotherapy (IMRT) and can more often meet normal tissue
constraints (NTC) than those planned conformally (Level 2c).
5–7
However, as with many
tumour types, there is insucient evidence to determine the ecacy of IMRT (Level 4).
5,7,8
Non-small cell lung cancer (NSCLC): curative therapy
Background
For patients with stage I and II lung cancer, anatomically based surgical resection remains
the treatment of choice. There is an emerging body of literature to support ablative
therapies in node-negative patients, of which stereotactic ablative radiotherapy (SABR)
has the most mature evidence base. There are, as yet, no completed randomised studies.
The two international randomised studies, which closed due to poor accrual, have been
published in pooled form (Level 2b).
5,9
There are a number of multi-institutional prospective
as well as retrospective series. Most concentrate on medically inoperable patients who are,
by denition, less well than their surgical counterparts. Published outcomes both in terms
of overall survival (OS) and disease-free survival (DFS) approach surgical series. Two-year
survival has been reported as 70% and ve-year survival 43%.
10,11
For medically inoperable patients with node negative tumours less than 5 centimetres (cm)
and in a favourable anatomical position, stereotactic ablative radiotherapy (SABR) is the
treatment of choice. The best outcomes occur when the tumour receives >100 Gray (Gy)
equivalent dose in 2 Gy per fraction EQD2 biologically equivalent dose (BED). Treatment
should be delivered with an interfraction interval of greater than 40 hours but less than four
days (Level 2a).
5,12
Stage III NSCLC is an extremely heterogeneous group in terms of tumour size and extent
of nodal involvement. Concurrent chemoradiotherapy has been demonstrated in meta-
analyses to give superior outcomes when compared with sequential chemoradiotherapy
or radiotherapy alone, but the optimum dose fractionation schedule has yet to be dened
(Level 1a).
5,13–15
Concurrent schedules have an increased incidence of grade three
oesophageal toxicities (Level 1b) and elderly patients with good performance status and
few co-morbidities derive as much benet from concurrent therapy as their younger
counterparts (Level 1b).
5,16
Although trimodality therapy remains an option, there is no evidence of benet over
denitive chemoradiotherapy. The only tumour group where there is some evidence to
support the use of trimodality therapy is Pancoast tumours (Level 1b).
5,17
There is no evidence of benet for chemotherapy delivered either neoadjuvantly or
adjuvantly to those receiving concurrent regimes (Level 1b).
5
50Radiotherapy dose fractionation Third edition
Dose escalation has been investigated in many studies. The recently published Radiation
Therapy Oncology Group (RTOG) 0617 trial did not demonstrate a survival benet in the
escalated arm. This trial has received signicant interest and review of individual data. The
quality assurance of the radiotherapy delivered may have been the cause of the lack of a
positive outcome so it is likely that this issue will be revisited (Level 1b).
5,18
For those unable to tolerate concurrent chemoradiotherapy, a sequential approach
demonstrates survival benet over radiotherapy alone.
15
The optimum therapy schedule
has yet to be dened (Level 1a).
5
Patients unt for systemic therapy should be treated with radiotherapy alone. Accelerated
fractionation schedules seem to improve outcomes (Level 1b) and can be safely combined
with concurrent and neoadjuvant approaches (Level 1b).
5,15,19–22
Recommendations
Medically inoperable T1–3 (≤5 cm) N0:
SABR using:
54 Gy in 3 fractions over 5–8 days (Grade B)
55 Gy in 5 fractions over 10–14 days (Grade B)
60 Gy in 5 fractions over 10–14 days (Grade B)
60 Gy in 8 fractions over 10–20 days (Grade B)
Medically inoperable stage I and II:
54 Gy in 36 fractions treating thrice daily over 12 consecutive days continuous,
hyperfractionated, accelerated radiotherapy (CHART) (Grade A)
55 Gy in 20 fractions (Grade C)
STAGE III:
Concurrent:
55 Gy in 20 fractions over 4 weeks with cisplatin and vinorelbine (Grade A)
60 Gy in 30 fractions over 6 weeks with cisplatin and etoposide (Grade A)
66 Gy in 33 fractions over 6.5 weeks with cisplatin and etoposide (Grade A)
Sequential:
55 Gy in 20 fractions over 4 weeks (Grade A)
60 Gy in 30 fractions over 6 weeks (Grade B)
66 Gy in 33 fractions over 6.5 weeks (Grade B)
54 Gy in 36 fractions treating thrice daily over 12 consecutive days (CHART) (Grade B)
Radiotherapy alone:
54 Gy in 36 fractions treating thrice daily over 12 consecutive days (CHART) (Grade A)
66 Gy in 33 fractions over 6.5 weeks (Grade A)
55Gy in 20 fractions over 4 weeks (Grade B)
Pancoast tumours (T3–4 N0–1):
45 Gy in 25 fractions over 5 weeks with cisplatin and etoposide followed by surgery
(Level 2b)
The types of evidence and the grading of recommendations used within this review are based on
those proposed by the Oxford Centre for Evidence-based Medicine.
5
51Radiotherapy dose fractionation Third edition
Non-small cell lung cancer (NSCLC): palliative radiotherapy
Background
The early trials were undertaken predominantly in patients unexposed to chemotherapy.
Current practice would see a signicant proportion of patients receiving sequential
chemoradiotherapy, with good performance status (PS) stage III patients managed with
radical concurrent chemoradiotherapy.
Overall the trials demonstrate that short-course radiotherapy can palliate intrathoracic
symptoms as well as long-course, but for those with good PS, higher doses confer a
moderate survival advantage at the expense of extra toxicity (Level 1a).
5,23
Recommendations
For those with good PS:
39 Gy in 13 fractions over 2.5 weeks with cord dose limited to 36 Gy (Grade A)
36 Gy in 12 fractions over 2.5 weeks (Grade A)
30 Gy in 10 fractions over 2 weeks (Grade A)
20 Gy in 5 fractions over 1 week (Grade A)
For those with poor PS:
17 Gy in 2 fractions over 8 days (Grade A)
10 Gy in 1 fractions (Grade A)
The types of evidence and the grading of recommendations used within this review are based on
those proposed by the Oxford Centre for Evidence-based Medicine.
5
Small cell lung cancer (SCLC)
Background
The evidence base now favours integration of chemotherapy and radiotherapy at all disease
stages (Level 1a).
5
Concurrent chemoradiotherapy (stages I–III)
For patients with T1–4 and N0–3 SCLC, there is evidence for concurrent
chemoradiotherapy with radiotherapy starting no later than day one cycle three of
chemotherapy (Level 1a).
5,24
The UK-led phase III Concurrent Once-Daily Versus Twice Daily
Radiotherapy (CONVERT) trial has compared the internationally accepted standard of 45
Gy in 30 fractions treating twice daily over three weeks with 66 Gy in 33 daily fractions over
six weeks, nding no dierence between the two schedules.
25,26
In addition, a US intergroup
study is currently recruiting, which compares three fractionation schedules (45 Gy in 30
fractions treating twice daily; 70 Gy in 35 daily fractions and 61.2 Gy over ve weeks treating
once daily until day 21 and twice daily thereafter) (Level 1b).
5
One trial of early versus late concurrent thoracic radiotherapy used 40 Gy in 15 daily
fractions using a simple parallel opposed pair with cord shielding (Level 1b).
5,24
This can
shield the tumour and, in the modern era, cord constraints would be met using 3DCRT.
52Radiotherapy dose fractionation Third edition
Sequential chemoradiotherapy (stages I–III)
For those patients who, due to tumour size or co-morbidities, cannot be treated with
concurrent chemoradiotherapy, sequential chemoradiotherapy is the best alternative (Level
1a).
5
There is no denitive evidence to indicate the optimal schedule in this patient group,
although many use 40 Gy in 15 daily fractions (Level 2b).
5
Recommendations
Concurrent chemoradiotherapy with cisplatin and etoposide should be
delivered with either:
45 Gy in 30 fractions treating twice daily over 3 weeks (Grade A)
66 Gy in 33 fractions over 6.5 weeks (Grade A)
40 Gy in 15 fractions over 3 weeks (Grade B)
Sequential chemoradiotherapy:
40 Gy in 15 daily fractions over 3 weeks (Grade B)
50 Gy in 25 fractions over 5 weeks (Grade B)
The types of evidence and the grading of recommendations used within this review are based on
those proposed by the Oxford Centre for Evidence-based Medicine.
5
SCLC: palliative thoracic radiotherapy
Background
A recent European Organisation for Research and Treatment of Cancer (EORTC) trial
randomised 498 patients with metastatic SCLC, who had not progressed during primary
chemotherapy to prophylactic cranial irradiation (PCI), with or without thoracic radiotherapy
with 30 Gy in ten daily fractions in addition. The trial did not meet its primary endpoint of
improved OS at one year, but OS at two years was in favour of mediastinal consolidation
(Level 1b).
5,27
Further data analysis has conrmed the OS and DFS benets are limited to
those with persistent intrathoracic disease (Level 1b).
5,28
Recommendation
Those patients with metastatic SCLC who respond to primary chemotherapy but
have persistent intrathoracic disease or thoracic symptoms should be considered for
thoracic consolidation radiotherapy with 30 Gy in 10 fractions over 2 weeks (Grade A).
The types of evidence and the grading of recommendations used within this review are based on
those proposed by the Oxford Centre for Evidence-based Medicine.
5
53Radiotherapy dose fractionation Third edition
Prophylactic cranial irradiation (PCI) (stages I–III)
Meta-analysis of patients with stages I–III SCLC in complete or near complete thoracic
remission following primary chemoradiotherapy have an increased OS and decreased
incidence of intracerebral relapse when PCI is delivered (Level 1a).
5,29,30
25 Gy in ten fractions over 14 days carries the same disease relapse rate but lower mortality
when compared with 36 Gy in 18 fractions over 24 days (Level 1a).
5,30
Recommendations
Selected patients with locally advanced metastatic SCLC who respond to
primary chemotherapy should be oered PCI. Acceptable schedules are:
20 Gy in 5 fractions over 1 week (Grade A)
30 Gy in 10 fractions over 2 weeks (Grade A)
25 Gy in 10 fractions over 2 weeks (Grade A)
30 Gy in 12 fractions over 2.5 weeks (Grade A)
The types of evidence and the grading of recommendations used within this review are based on
those proposed by the Oxford Centre for Evidence-based Medicine.
5
Prophylactic cranial irradiation (PCI) (stage IV)
Patients with stage IV SCLC who had any response to primary chemotherapy were
randomised to either PCI with one of ve schedules (20–30 Gy in 5–12 daily fractions) or no
PCI. The treatment arms had an increased OS and reduced symptomatic incidence of brain
metastases (Level 1b).
5
85% of patients were treated with either 30 Gy in ten fractions or 20
Gy in ve fractions. Two thirds received 20 Gy in ve fractions. The trial excluded patients
above 75 years of age.
31
Recommendations
Selected patients with metastatic SCLC who respond to primary chemotherapy
should be oered PCI. Acceptable schedules are:
20 Gy in 5 fractions over 1 week (Grade A)
30 Gy in 10 fractions over 2 weeks (Grade A)
25 Gy in 10 fractions over 2 weeks (Grade A)
30 Gy in 12 fractions over 2.5 weeks (Grade A)
The types of evidence and the grading of recommendations used within this review are based on
those proposed by the Oxford Centre for Evidence-based Medicine.
5
54Radiotherapy dose fractionation Third edition
Mesothelioma
Background
The use of prophylactic irradiation of tracts of pleural interventions has been thought to
reduce the incidence of chest wall recurrence. Three small randomised studies have
been reported, one demonstrating benet, two not (Level 1b).
5,32–34
Currently in the UK, two
studies are addressing this issue. The Prophylactic Irradiation of Tracts (PIT) trial (closed
to recruitment in December 2015) randomises those with a visible scar following minor
pleural interventions between 21 Gy in three daily fractions using electrons or no treatment.
The Simultaneous Modulated Accelerated Radiation Therapy (SMART) trial randomised
those with larger pleural interventions between immediate radiotherapy with 21 Gy in
three daily fractions or treatment deferred until tract metastases occurred. The SMART
trial has been verbally presented (January 2016), with no benet of immediate radiotherapy
demonstrated.
35
For those patients with a diagnosis of mesothelioma and chest wall pain, controversy exists
about the utility of radiotherapy, especially where the pain is poorly localised. A recently
published non-randomised study demonstrates a 35% response rate when chest wall
radiotherapy is delivered to patients with localised pain (Level 2c).
5,36
Recommendation
Routine prophylactic irradiation of tracts is not recommended (Level 1b)
Selected patients with chest wall pain may benet from radiotherapy with either:
20 Gy in 5 fractions over 1 week (Grade C)
36 Gy in 6 fractions treating twice per week (Grade D)
The types of evidence and the grading of recommendations used within this review are based on
those proposed by the Oxford Centre for Evidence-based Medicine.
5
55Radiotherapy dose fractionation Third edition
1. www.hqip.org.uk/ncapop-library/#cancer (last accessed 28/9/16)
2. National Cancer Intelligence Network Survival by stage (NCIN) 2014:
www.ncin.org.uk/publications/survival_by_stage (last accessed 28/9/16)
3. Koning CC, Aarts MJ, Struikmans H et al. Mapping use of radiotherapy for patients with non-small cell
lung cancer in the Netherlands between 1997 and 2008. Clin Oncol (R Coll Radiol) 2012; 24(2): e46–
e53.
4. Delaney GP and Barton MB. Evidence-based estimates of the demand for radiotherapy. Clin Oncol (R
Coll Radiol) 2015; 27(2): 70–76.
5. www.cebm.net/oxford-centre-evidence-based-medicine-levels-evidence-march-2009
(last accessed 26/9/16)
6. Harris JP, Murphy JD, Hanlon AL, Le QT, Loo BW Jr, Diehn M. A population-based comparative
eectiveness study of radiation therapy techniques in stage III non-small cell lung cancer. Int J Radiat
Oncol Biol Phys 2014; 88(4): 872–884.
7. Chan C, Lang S, Rowbottom C et al. Intensity-modulated radiotherapy for lung cancer: current status
and future developments. J Thorac Oncol 2014; 9(11): 1598–1608.
8. Bezjak A, Rumble RB, Rodrigues G, Hope A, Warde P, Members of the IMRT Indications Expert Panl.
Intensity-modulated radiotherapy in the treatment of lung cancer. Clin Oncol (R Coll Radiol) 2012;
24(7): 508–520.
9. Chang JY, Senan S, Paul MA et al. Stereotactic ablative radiotherapy versus lobectomy for operable
stage I non-small-cell lung cancer: a pooled analysis of two randomised trials. Lancet Oncol 2016;
16(6): 630–637.
10. Zheng X, Schipper M, Kidwell K et al. Survival outcome after stereotactic body radiation therapy and
surgery for stage I non-small cell lung cancer: a meta-analysis. Int J Radiat Oncol Biol Phys 2011; 90(3):
603–611.
11. Shirvani SM, Jiang J, Chang JY et al. Comparative eectiveness of 5 treatment strategies for early-stage
non-small cell lung cancer in the elderly. Int J Radiat Oncol Biol Phys 2012; 84(5): 1060–1070.
12. SABR UK Consortium. Stereotactic ablative radiotherapy (SABR): A resource. Version 5.0. Manchester:
SABR UK Consortium, 2015.
13. Aupérin A, Le Péchoux C, Pignon JP et al. Concomitant radio-chemotherapy based on platin
compounds in patients with locally advanced non-small cell lung cancer (NSCLC): a meta-analysis of
individual data from 1764 patients. Ann Oncol 2006; 17(3): 473–483.
14. O’Rourke N, Roqué i Figuls M, Farré Bernadó N, Macbeth F. Concurrent chemoradiotherapy in non-
small cell lung cancer. Cochrane Database Syst Rev 2010; 16(6): CD002140.
15. Maguire J, Khan I, McMenemin R et al. SOCCAR: A randomised phase II trial comparing sequential
versus concurrent chemotherapy and radical hypofractionated radiotherapy in patients with
inoperable stage III non-small cell lung cancer and good performance status. Eur J Cancer 2014;
50(17): 2939–2949.
16. Jalal SI, Riggs HD, Melnyk A et al. Updated survival and outcomes for older adults with inoperable
stage III non-small-cell lung cancer treated with cisplatin, etoposide, and concurrent chest radiation
with or without consolidation docetaxel: analysis of a phase III trial from the Hoosier Oncology Group
(HOG) and US Oncology. Ann Oncol 2012; 805(23): 1730–1738.
References
56Radiotherapy dose fractionation Third edition
17. Rusch VW, Giroux DJ, Kraus MJ et al. Induction chemoradiation and surgical resection for superior
sulcus non-small-cell lung carcinomas: long-term results of Southwest Oncology Group Trial 9416
(Intergroup trial 0160). J Clin Oncol 2007; 25(3): 313–318.
18. Bradley JD, Paulus R, Komaki R et al. Standard-dose versus high-dose conformal radiotherapy with
concurrent and consolidation carboplatin plus paclitaxel with or without cetuximab for patients with
stage IIIA or IIIB non-small cell lung cancer (RTOG 0617): a randomised, two-by-two factorial phase 3
study. Lancet Oncol 2015; 16 (2): 187–199.
19. Saunders M, Dische S, Barrett A, Harvey A, Gibson D, Palmar M. Continuous hyperfractionated
accelerated radiotherapy (CHART) versus conventional radiotherapy in non-small cell lung cancer: a
randomised multicentre trial. CHART Steering Committee. Lancet 1997; 350(9072): 161–165.
20. Mauguen A, Le Péchoux C, Saunders MI et al. Hyperfractionated or accelerated radiotherapy in lung
cancer: an individual patient data meta-analysis. J Clin Oncol 2102; 30(22): 2788–2297.
21. Hatton M, Nankivell M, Lyn E et al. Induction chemotherapy and continuous hyperfractionated
accelerated radiotherapy (CHART) for patients with locally advanced inoperable non-small-cell lung
cancer: the MRC INCH randomized trial. Int J Radiat Oncol Biol Phys 2011; 81(3): 712–718.
22. Price A, Yellowlees A, Keerie C et al. Radical radiotherapy with or without gemcitabine in patients with
early stage medically inoperable non-small cell lung cancer. Lung Cancer 2012; 77(3): 532–536.
23. Lester JF, Macbeth F, Toy E, Coles B. Palliative radiotherapy regimens for non-small-cell lung cancer.
Cochrane Database Syst Rev 2006; 18(4): CD002143.
24. Murray N, Coy P, Pater JL et al. Importance of timing for thoracic irradiation in the combined modality
treatment of limited-stage small-cell lung cancer. The National Cancer Institute of Canada Clinical
Trials Group. J Clin Oncol 1993; 11(2): 336–344.
25. Faivre-Finn C, Snee M, Ashcroft L et al. CONVERT: An international randomised trial of concurrent
chemo-radiotherapy (cCTRT) comparing twice-daily (BD) and once-daily (OD) radiotherapy schedules
in patients with limited stage small cell lung cancer (LS-SCLC) and good performance status (PS). J
Clin Oncol 2016; 34: (suppl; abstr 8504).
26. Turrisi AT 3rd, Kim K, Blum R et al. Twice-daily compared with once-daily thoracic radiotherapy in
limited small cell lung cancer treated concurrently with cisplatin and etoposide. N Engl J Med 1999;
340(4): 265–271.
27. Slotman BJ, van Tinteren H, Praag JO et al. Use of thoracic radiotherapy for extensive stage small-cell
lung cancer: a phase 3 randomised controlled trial. Lancet 2015; 385(9962): 36–42.
28. Slotman BJ, van Tinteren H, Praag JO et al. Which patients with extensive-stage small cell lung cancer
(ES-SCLC) are most likely to benet from routine thoracic radiotherapy (TRT) following chemotherapy?
Lancet (in press).
29. Aupérin A, Arriagada R, Pignon JP et al. Prophylactic cranial irradiation for patients with small-cell lung
cancer in complete remission. Prophylactic Cranial Irradiation Overview Collaborative Group. N Engl J
Med 1999; 341(7): 476–484.
30. Le Péchoux C, Dunant A, Senan S et al. Prophylactic Cranial Irradiation (PCI) Collaborative Group.
Standard-dose versus higher-dose prophylactic cranial irradiation (PCI) in patients with limited-stage
small-cell lung cancer in complete remission after chemotherapy and thoracic radiotherapy (PCI 99-
01, EORTC 22003-08004, RTOG 0212, and IFCT 99-01): a randomised clinical trial. Lancet Oncol 2009;
10(5): 467–474.
References
57Radiotherapy dose fractionation Third edition
31. Slotman B, Faivre-Finn C, Kramer G et al. Prophylactic cranial irradiation in extensive small-cell lung
cancer. N Engl J Med 2007; 357(7): 664–672.
32. Boutin C, Rey F, Viallat JR. Prevention of malignant seeding after invasive diagnostic procedures in
patients with malignant pleural mesothelioma. A randomised trial of local radiotherapy. Chest 1995;
108(3): 754–758.
33. Bydder S, Phillips M, Joseph DJ et al. A randomised trial of single dose radiotherapy to prevent
procedure tract metastases by malignant pleural mesothelioma. Br J Cancer 2004; 91(1): 9–10.
34. O’Rourke N, Garcia JC, Paul J, Lawless C, mcMenemin R, Hill J. A randomised controlled trial of
intervention site radiotherapy in malignant pleural mesothelioma. Radiother Oncol 2007; 84(1): 18–22.
35. Clive AO, Wilson P, Taylor H et al. Protocol for the surgical and large bore procedures in malignant
pleural mesothelioma and radiotherapy trial (SMART Trial): an RCT evaluating whether prophylactic
radiotherapy reduces the incidence of procedure tract metastases. BMJ Open 2015; 5(1): e006673.
36. MacLeod N, Chalmers A, O’Rourke N et al. Is radiotherapy useful for treating pain in mesothelioma?: A
phase II trial. J Thorac Oncol 2015; 10(6): 944–950.