pdf hosted at the radboud repository of the radboud ... · phil bosmans dit geldt zeker voor...

136
PDF hosted at the Radboud Repository of the Radboud University Nijmegen The following full text is a publisher's version. For additional information about this publication click this link. http://hdl.handle.net/2066/30924 Please be advised that this information was generated on 2017-12-05 and may be subject to change.

Upload: others

Post on 19-Jan-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

PDF hosted at the Radboud Repository of the Radboud University

Nijmegen

The following full text is a publisher's version.

For additional information about this publication click this link.

http://hdl.handle.net/2066/30924

Please be advised that this information was generated on 2017-12-05 and may be subject to

change.

Page 2: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Radiotherapy in Non-Small-Cell Lung Cancer

Changing perspectives in palliative care

Gijsbert Willem Pieter Marie Kramer

Page 3: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Radiotherapy in Non-Small-Cell Lung Cancer

Changing perspectives in palliative care

Bestraling voor het niet-kleincellige longcarcinoom

Veranderende vooruitzichten in de palliatieve zorg

Gijsbert Willem Pieter Marie Kramer

Page 4: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

ISBN: 978-90-9022399-5 Printed by: Universal Press Copyright © 2007 erven G.W.P.M. Kramer, Elst (Gelderland) Cover & Layout Design: G.W.P.M. Kramer

Page 5: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

RADBOUD UNIVERSITEIT NIJMEGEN

Radiotherapy in Non-Small-Cell Lung Cancer

Changing perspectives in palliative care

Een wetenschappelijke proeve op het gebied van de Medische Wetenschappen

Proefschrift

ter verkrijging van de graad van doctor aan de Radboud Universiteit Nijmegen

op gezag van de Rector Magnificus, prof. mr. S.C.J.J. Kortmann, volgens besluit van het College van Decanen

in het openbaar te verdedigen op dinsdag 27 november 2007, om 17.15 uur precies

in naam van

Gijsbert Willem Pieter Marie Kramer

geboren op 17 november 1956 te Venlo overleden op 6 mei 2006 te Elst

Page 6: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Promotores: Prof. dr. J.W.H. Leer Prof. dr. J.P. van Meerbeeck (Universiteit Gent, België) Manuscriptcommissie: Prof. dr. C.J.A. Punt Prof. dr. E.M. Noordijk (LUMC) Prof. dr. K.C.P. Vissers

Page 7: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Je bent als mens een verliezer. Je zou een gelukkig moment willen doen voortduren, Een mooie dag nooit zien eindigen, Maar je verliest, Je moet afgeven, Elke avond een dag achterlaten. Phil Bosmans

Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten zij, gesteund door familie, vrienden, en kennissen belangeloos mee.

Uit dank en respect, draag ik dit proefschrift op aan deze patiënten.

Page 8: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten
Page 9: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Table of contents Chapter 1 Introduction

9

Chapter 2 Hypofractionated external beam radiotherapy as retreatment for symptomatic non-small-cell lung carcinoma: an effective treatment? G.W.P.M. Kramer, S. Gans, E. Ullmann, J.P. van Meerbeeck, C.L. Legrand, J.W.H. Leer Int J Rad Oncol Biol Phys 2004; 58:1388–1393.

17

Chapter 3 Randomized controlled trial of resection versus radiotherapy after induction chemotherapy in stage IIIA-N2 non-small-cell lung cancer (EORTC 08941). J.P. van Meerbeeck, G.W.P.M. Kramer, P.E.Y. van Schil, C. Legrand, E.F. Smit, F. Schramel, V.C. Tjan-Heijnen, B. Biesma, C. Debruyne, N. van Zandwijk, T.A.W. Splinter, G. Giaccone. J Nat Cancer Inst 2007; 99(6): 442-450.

27

Chapter 4 Gemcitabine and Cisplatin as induction regimen for patients with biopsy-proven stage IIIA N2 Non-Small-Cell Lung Cancer: a phase II study of the European organization for research and treatment of cancer Lung Cancer Cooperative group (EORTC 08955). N. Van Zandwijk, E. F. Smit, G.W.P. Kramer, F. Schramel, S. Gans, J. Festen, A. Termeer, N. J. J. Schlosser, C. Debruyne, D. Curran, G. Giaccone. J Clin Oncol 2000;18: 2658–2664.

47

Chapter 5 Quality Assurance of Thoracic Radiotherapy in EORTC 08941: A randomised trial of Surgery versus Thoracic Radiotherapy in patients with stage IIIA Non-Small-Cell Lung Cancer (NSCLC) after response to induction chemotherapy. G.W.P.M. Kramer , C.L. Legrand, P. van Schil, L. Uitterhoeve, E.F. Smit, F.Schramel, B. Biesma, V. Tjan-Heijnen, N. van Zandwijk, T. Splinter, G.Giaccone, J.P. van Meerbeeck, on behalf of EORTC-Lung Cancer Group. Eur J of Cancer 2006; 42:1391-1398.

59

Page 10: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Chapter 6 The results of the Dutch National Study to the palliative effect of irradiation comparing two different treatment schemes in Non-Small-Cell Lung Cancer (NSCLC). G.W.P.M. Kramer , S.L. Wanders , E.M. Noordijk, E.J.A. Vonk, H.C. van Houwelingen, W.B. van den Hout, R.B. Geskus, M. Scholten, J.W.H. Leer. J Clin Oncol 2005; 23: 2962–2970.

73

Chapter 7 Cost-Utility analysis of short- versus long-course palliative radiotherapy in patients with Non-Small-Cell lung cancer. W.B. van den Hout, G.W.P.M. Kramer, E.M. Noordijk, J.W.H. Leer J Nat Cancer Institute 2006; 98(24):1786-1794.

89

Chapter 8 General discussion

105

Summary

121

Samenvatting

123

List of Publications

127

Curriculum vitae

131

Dankwoord

133

Page 11: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Chapter 1

INTRODUCTION

Page 12: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

10

Chapter 1 Non-small-cell lung cancer (NSCLC) is a prominent cause of cancer death in many Western countries.1 Of the 8000 new cases of lung cancer diagnosed yearly in the Netherlands, 80% has NSCLC.2 Only 25% of these patients are candidates for a curative treatment option.3 Surgery is the primary choice of treatment for these patients. For those who cannot or will not undergo surgery, radiotherapy is an alternative.4 For the majority of patients only palliative treatment options are possible. Despite the various treatment options, many of these patients will eventually suffer a recurrence with severe symptoms from local tumour growth before demise.5 It is therefore a challenge to search for new perspectives in palliative cancer care. The clinical studies described in this thesis focus on the following options: 1. Feasibility of repeat radiotherapy as part of palliative care, 2. New treatment combinations to gain better results in palliative care and life

expectancy, finally resulting in a more radical approach. 3. Optimalization of palliative irradiation schemes for those patients who only

can be treated with radiotherapy. Chapter 2 A common problem in the late 1980’s were complaints of a recurrent tumor in patients, treated with radiotherapy before. In these days in only 12% of patients complete tumor regression could be obtained with radiotherapy as a single modality treatment, even after doses of 60 Gy.6 Therefore, a local tumour recurrence rate of 50 – 65% in patients treated with external beam radiotherapy was seen.5 In these patients, with symptoms as a result of renewed tumour progression, data on repeat radiotherapy were scarce.7,8 The number of cases in these studies was limited and the radiotherapy schemes used varied. Chemotherapy was no alternative9, as effective chemotherapeutic agents only became available in more recent years.10 Radiation oncologists were reluctant to retreat these patients with external beam irradiation, afraid to exceed the tolerance treshhold dose of normal tissues such as lung parenchyma11, the spinal cord12-14 and the esophagus.15 The limits in tolerance dose because of normal tissues, were not known for repeat radiotherapy. To reduce the possibility of complications, the dose to normal tissues had to be limited. Therefore, techniques were used to limit the retreated volume to the site of tumour recurrence only. Endoscopic techniques, as brachytherapy, laser ablation, or photodynamic therapy, each have their own limitations, but have in common that they are only suitable for patients with centrally located tumors with no extension beyond the trachea or bronchus.16-19 Repeat external beam radiotherapy has the advantage, that it can also cover the tumour burden around trachea and bronchus and in the mediastinum, but the disadvantage that a larger volume is irradiated. As life expectancy is limited, the duration of the retreatment is preferably as short as possible. The effectiveness and safety of hypofractionation was shown in palliative settings for primary treatment19, and at the time had to be proven for repeat radiotherapy. In repeat radiotherapy data of palliative response rates were available7,8 but the duration of such response was not known. All this prompted us to initiate in 1991 a prospective study of hypofractionated radiotherapy for patients with recurrent intrathoracic NSCLC after prior curative external beam treatment. Response was measured as the maximum relief of

Page 13: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

11

symptoms. We focused on response rates with regard to the different symptoms and toxicity, but also on the duration of the palliative effect. The study was discontinued in 1999 when chemotherapy became an alternative and subsequently accrual declined.20 However, we demonstrated that a reasonable palliative effect could be obtained with two fractions of 8 Gy. Chapter 3 In this - and the following two chapters, we describe the results of EORTC study 08941 which was initiated in 1994. In this study loco-regional control and survival were compared for induction chemotherapy followed by either radiotherapy or surgery. Patients had histological or cytological proven unilateral mediastinal lymphnode metastasis of a non-resectable NSCLC stage IIIA(N2). All were treated with induction chemotherapy. Only patients with a tumour response were randomized between external beam irradiation and surgery. Up-front, they had to be fit enough to undergo pneumonectomy. This study was initiated because of a couple of changes in the treatment of lung cancer in the previous years. Firstly, chemotherapy regimens showed to be beneficial in reducing tumor burden locally and at distance.20 Chemotherapy as sensitizer to radiotherapy showed a better local control and a decrease in distant metastases, both resulting in a better survival.21 Secondly, as in the early 1990’s, a curative resection was seldom performed in case of ipsilateral mediastinal lymphnode metastasis (stage IIIA N2), the treatment consisted of radiotherapy or a wait and see policy, considering the treatment had to be only palliative.22 The treatment goal in this category of patients is to postpone or relieve tumor related symptoms as long as possible. Gradually however doses up to 60 Gy in 6 weeks were used and 2-year survival rates of 17% were reported.23 Although this survival rate is less than is achieved with curative surgical or radiation treatments4, it still implies a chance of cure. Because of these two developments it was considered that maybe combined modality may be better, and that radiotherapy only would no longer be acceptable.Consequently, treatment intention for this particular group of patients was redefined in 1997 from palliative to radical22 and finally in 2002 in curative.24

As surgery still was the primary treatment option in curative settings, the study evaluated surgery as an alternative for radical radiotherapy after a response to induction chemotherapy. Surgery was expected to offer a better chance of loco-regional control and survival than radiotherapy.25-27 A hypothesis which was not confirmed, based on the results of our study. Chapter 4 The choice of chemotherapeutic regimens was also open for debate. The response rates of the chemotherapy regimens used in those days was 30%.28 The search for more effective regimens continued. New chemotherapeutic agents became available, such as Gemcitabine. In EORTC study 08955 the objective was to better define the efficacy/feasibility of Gemcitabine/cisplatin (GC) as induction chemotherapy within the ongoing EORTC study 08941. The results of this EORTC study such were that good, that this combination became one of the new standard regimens.

Page 14: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

12

Chapter 5 Quality assurance is an important issue in the EORTC. 29 In 1987 the EORTC Radiotherapy Group introduced a series of procedures to document systematic errors, e.g. by dummy runs, made in single institutions. This dummy run gives also information about accuracy of the design and the application of phase III study protocols. In 1991 Koning30 recommended immediate monitoring of treatment parameters and uniform assessment of patient data in clinical trials. However, at the start in 1994 of the EORTC study 08941 these quality measures on radiotherapy treatment were not yet implemented. Questions about the quality of radiotherapy data came up halfway the trial31, as not all investigators adhered to prescribed treatment schemes. At site visits, the radiotherapy treatment fields and set up appeared to be correct, but compliance to the protocol was subject to improvement. Therefore, in May 1999 the protocol was amended detailing the radiotherapy prescriptions.32 The radiotherapy prescriptions were tightened and updated as follows: planning-CT became standard for the whole treatment procedure, time frames and dose specifications were formulated more precisely, questionnaires revised, and participants instructed. The target volumes were redefined in accordance with the specifications of the ICRU 50 report.33 The radiotherapy data before and after the amendment are presented. This amendment resulted in an improvent of the quality of and the compliance to the radiotherapy and an enhancement of the reliability of the final results of the study. Chapter 6 In this and the next chapter the clinical and socio-economic results of the prospective National Study on the effect of irradiation with different treatment schemes in Non-Small-Cell Lung Cancer (NSCLC) (OG98/009) are presented. There remains a group of patients not fit enough to undergo either chemotherapy or combined treatment, but still with a need to be palliated. Radiotherapy has been shown to be an effective palliative treatment, but the radiation schedule is still controversial.34 Due to the contradictory reports in the literature, radiation oncologists in the Netherlands were unable to reach a consensus on the palliative schedule for patients with poor prognosis NSCLC. Because of subgroup analysis results within the MRC study19, we expected for patients with poor prognostic factors an equal outcome for two Dutch treatment schemes with regard to palliation and survival: 10 x 3 Gy at 4 – 5 fractions a week, and 2 x 8 Gy at day 1 and 8. Because of the relative shortage of radiotherapy facilities, 2 x 8 Gy for palliation would be preferred. Specifically, seven tumor related symptoms were scored and response measured over 52 weeks. The sum of these scores is calculated over time. We argue this total symptom score gives a better view of the overall palliative effect than represented by each separate symptom. It covers not only diminishing symptoms, not responding symptoms, but also intensification of symptoms, because of toxicity. The fluctuation in time of the total symptom scores was compared between both treatment arms, representing the duration of the palliative effect. The 10 x 3-Gy scheme resulted not only in a more protracted duration of palliation, but also in a small survival gain.

Page 15: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

13

Chapter 7 Not only therapeutic, but also socio-economic reasons could affect the treatment choice. Therefore, a cost-effectiveness assessment for both treatment arms was performed. A pragmatic model was constructed to estimate lifelong costs and quality adjusted life years. Patients were followed for up to one year, with symptom palliation as primary outcome measure. Utility was measured using the EuroQol classification system.35 Societal costs were assessed during the initial 12 weeks, using cost questionnaires used in 3 of the 11 participating centers. Despite the higher costs, partly because of the increased survival for the 10 x 3-Gy scheme, the cost-utility ratio is still acceptable according the Dutch standards.36 Based on this conclusion and the better palliative effect, the 10 x 3-Gy scheme has to be the standard. References 1. Landis SH, Murray T, Bolden S, et al. Cancer statistics. CA Cancer J Clin

1998;48:6–9. 2. Dijck JAAM van, Coebergh JWW, Siesling S, Visser O (ed). Trends of cancer in

the Netherlands 1989-1998. Utrecht: Vereniging van Integrale Kankercentra, 2002.

3. Van Meerbeeck JP, Koning CCE, Tjan-Heijnen VCG (ed). Richtlijn “Niet-kleincellig longcarcinoom: stadiëring en behandeling”, 2004. Utrecht: Vereniging van Integrale Kankercentra.

4. Qiao X, Tullgren O, Lax I, et al. The role of radiotherapy in treatment of stage I non-small cell lung cancer. Lung Cancer 2003;41:1–11.

5. Perez CA, Pajak TF, Rubin P, et al. Long-term observations of the patterns of failure in patients with unresectable non-oat cell carcinoma of the lung treated with definitive radiotherapy: Report by the Radiation Therapy Oncology Group. Cancer 1987;59:1874–1881.

6. Arriagada R, Le Chevalier T, Quoix E, et al. for the GETCB, FNCLCC and the CEBI trialists. ASTRO plenary –– Effect of chemotherapy on locally advanced non-small cell lung carcinoma: A randomized study of 353 patients. Int J Radiat Oncol Biol Phys 1991;20:1183–1190.

7. Green N, Melbye RW. Lung cancer: Retreatment of local recurrence after definitive irradiation. Cancer 1982;49:865–868.

8. Jackson MA, Ball DL. Palliative retreatment of locally recurrent lung cancer after radical radiotherapy. Med J Aust 1987;147:391–394.

9. Ruckdeschel JC. Is chemotherapy for metastatic NSCLC worth it? a Review. J Clin Oncol 1990;8:1293–1296.

10. Emami B, Lyman J, Brown A, et al.Tolerance of normal tissue to therapeutic irradiation. Int J Radiat Oncol Biol Phys 2001;50:899–908.

11. Schultheiss T.E. and Stephens L.C. Invited Review: Permanent radiation myelopathy. The British Journal of Radiology 1992;65:737–753.

12. Fowler JF, Bentzen SM, Bond SJ, et al. Clinical radiation doses for spinal cord: the 1998 international questionnaire. Radiother Oncol 2000;55:295–300.

13. Ang KK, Jiang G-L, Feng Y, et al. Extent and kinetics of recovery of occult spinal cord injury. Int J Radiat Oncol Biol Phys 2001;50:1013–1020.

Page 16: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

14

14. Morris CD, Budd JM, Godette KD, et al. Palliative management of malignant

airway obstruction. Ann Thorac Surg 2002;74:1928–1932. 15. Poltinnikov IM, Fallon K, Xiao Y et al. Combination of longitudinal and

circumferential three-dimensional esophageal dose distribution predicts acute esophagitis in hypofractionated reirradiation of patients with non-small-cell lung cancer treated in stereotactic body frame. Int J Radiat Oncol Biol Phys 2005;62:652–658.

16. Metz JM, Friedberg JS. Endobronchial photodynamic therapy for the treatment of lung cancer. Chest Surg Clin North Am 2001;11:829–839.

17. Schray MF, McDougall JC, Martinez A, et al. Management of malignant airway comprise with laser and low dose rate brachytherapy. Chest 1988;93:264–269.

18. Speiser B, Spratling L. Intermediate dose rate remote afterloading brachytherapy for intraluminal control of bronchogenic carcinoma. Int J Radiat Oncol Biol Phys 1990;18:1443–1448.

19. Bleehen NM, Girling DJ, Fayers PM, et al. Inoperable non-small-cell lung cancer (NSCLC): a Medical Research Council randomized trial of palliative radiotherapy with two fractions or ten fractions. Br J Cancer 1991;63:265–270.

20. Thatcher N. Chemotherapy for advanced NSCLC. A Review. Lung Cancer 2001;34(S2):S171–175.

21. Schaake-Koning C, van den Bogaert W, Dalesio O, et al. Radiosensitization by cytotoxic drugs. The EORTC experience by the Radiotherapy and Lung Cancer Cooperative Groups. Lung Cancer. 1994;10 Suppl 1:S263–270.

22. Festen J. Guideline “Radiotherapie in non-small-cell lung carcinoma. Working Group, National Organization for Quality Assurance in Hospitals, Netherlands Ned Tijdschr Geneeskd. 1998;142:2248–2251. Review (Dutch)

23. Van Dijck JA, Festen J, de Kleijn EM, et al. Treatment and survival of patients with non-small cell lung cancer Stage IIIA diagnosed in 1989–1994: a study in the region of the Comprehensive Cancer Centre East, The Netherlands. Lung Cancer 2001;34:19–27.

24. van Meerbeeck JP, Koning CC, Tjan-Heijnen, et al. Guideline on ‘non-small-cell lung carcinoma; staging and treatment’. NTvG 2005;149:72–77.

25. Burkes RL, Ginsberg RJ, Shepherd FA et al. Induction chemotherapy with mitomycin, vindesine, and cisplatin for stage III unresectable non-small-cell lung cancer: results of the Toronto Phase II Trial. J Clin Oncol 1992;10:580–586.

26. Vokes EE. Sequential combined modality therapy for stage III non-small cell lung cancer. Hematol Oncol Clin North Am 1990;6:1133–1142.

27. Murren JR, buzaied AC, Hait WN. Critical analysis of neoadjuvant therapy for stage IIIa non-small cell lung cancer (corrected). Am Rev Respir Dis. 1991;143:889–894.

28. T.A.W. Splinter, P.E. van Schil, G.W.P.M. Kramer, et al. Randomized Trial of Surgery Versus Radiotherapy in Patients with Stage IIIA (N2) Non–Small-Cell Lung Cancer After a Response to Induction Chemotherapy. EORTC 08941. Clin Lung Cancer 2000;2:69–72.

29. Kouloulias VE, Poortmans PM, Bernier J, et al. The Quality Assurance Programme of the Radiotherapy Group of the European Organization for Research and Treatment of Cancer (EORTC): a critical appraisal of 20 years of continuous efforts. Eur. J. of Cancer 2003;39:430–437.

Page 17: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

15

30. Schaake-Koning C, Kirkpatrick A, Kroger R, et al. The need for immediate monitoring of treatment parameters and uniform assessment of patient data in clinical trials. A quality control study of the EORTC Radiotherapy and Lung Cancer Cooperative Groups. Eur J Cancer. 1991;27:615–619.

31. van Zandwijk N, Smit EF, Kramer GWP, et al. Gemcitabine and cisplatin as induction regimen for patients with biopsy-proven stage IIIA N2 non-small-cell lung cancer: a phase II study of the european organization for research and treatment of cancer. Lung Cancer Cooperative Group (EORTC 08955). J Clin Oncol 2000;18:2658–2664.

32. Bolla M, Bartelink H, Garavaglia G, et al. EORTC guidelines for writing protocols for clinical trials of radiotherapy. Radiother Oncol 1995;36:1–8.

33. ICRU Report 50. Prescribing, recording and reporting photon beam therapy. International Commission on Radiation Units and Measurements. 7910 Woodmont Avenue, Bethesda, Maryland 20814, USA.

34. Macbeth F, Toy E, Coles B, et al. Palliative radiotherapy regimens for non-small cell lung cancer. Cochrane. Database Syst Rev 1:CD002143,2002.

35. Dolan P. Modeling valuations for EuroQol health states. Med Care 1997;35(11):1095–1108.

36. Oostenbrink JB, Koopmanschap MA, Rutten FF. Standardisation of costs: the Dutch Manual for Costing in economic evaluations. Pharmacoeconomics 2002;2:443–454.

Page 18: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Chapter 2

HYPOFRACTIONATED EXTERNAL BEAM RADIOTHERAPY

AS RETREATMENT FOR SYMPTOMATIC NON-SMALL-CELL

LUNG CARCINOMA: AN EFFECTIVE TREATMENT?

Gijsbert W.P.M. Kramer, M.D.,* Steven Gans, M.D.,† Eric Ullmann, M.D., ‡ Jan P. van Meerbeeck, M.D., Ph.D.,§ Catherine C. Legrand, M.Sc.,|| and Jan-Willem H. Leer,

M.D., Ph.D.*

*Radiotherapy Institute ARTI, Joint Centre of Oncology RADIAN, Arnhem, The Netherlands; †Department of Pulmonology, St. Jansdal hospital, Harderwijk, The

Netherlands; ‡ Department of Pulmonology, Rijnstate hospital, Arnhem, the Netherlands; §Department of Pulmonology, Erasmus Medical Center, Rotterdam, the

Netherlands; || European Organization for Research and Treatment of Cancer Data Center, Brussels, Belgium

International Journal Radiation Oncology Biology Physics, 2004;58:1388–1393.

Page 19: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

17

Summary Purpose: To evaluate prospectively efficacy, toxicity, and duration of the palliative effect of retreatment with external beam radiotherapy in symptomatic patients with recurrent non-small-cell lung cancer. Patients and Methods: Twenty-eight symptomatic patients with local recurrence of non-small-cell lung cancer underwent repeated treatment after previous radiotherapy (equivalent dose, 46 – 60 Gy). Reirradiation consisted of two fractions of 8 Gy on days 1 and 8 with 2 opposed beams using 6-18-MV photon beams at the site of pulmonary recurrence. The physician scored symptom resolution. Results: Relief of hemoptysis and superior vena cava syndrome could be obtained in all assessable cases (100%). Treatment was less effective for coughing (67%) and dyspnea (35%). The overall median duration of this palliative effect was 4 months. Palliation in almost all patients lasted more than one-half of their remaining life span. The Karnofsky performance score improved in 45% of assessable cases. One patient had Grade 2 esophagitis. Complications consisted of tumor-related fatal hemoptysis in 5 patients (17%) and 1 death from bronchoesophageal fistula (4%). Conclusion: External beam hypofractionated reirradiation can be effective as a palliative treatment for local complaints in non-small-cell lung cancer. The complication rate of reirradiation was acceptably low. Key words: Non-Small-Cell Lung Cancer, Repeated RT, Radiotherapy, Palliation.

Page 20: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

18

Introduction Non-small-cell lung cancer (NSCLC) is a prominent cause of cancer death in many Western countries, including The Netherlands.1,2 Surgery is the first option for the curative treatment of patients with NSCLC. However, many patients are no longer candidates for this type of treatment at the time of primary diagnosis. For patients who are medically inoperable or who have recurrent or unresectable NSCLC, radiotherapy (RT) is an alternative to surgery. More recently, a combined modality approach with chemoradiotherapy or chemotherapy has been advocated as an alternative. However, many patients are also unable to undergo chemotherapy or combined treatment.3 Therefore, a substantial number of patients are still treated with primary RT either with radical (high dose) or palliative (low dose) intent. However, even after radical irradiation for NSCLC, a complete response is only obtained in a few patients.4 Local tumor recurrence occurs in 50 – 65 % of the patients. In 25-30% this recurrence will be localized. These patients will eventually have severe symptoms from local tumor regrowth before final demise.5 Whether repeated RT of these patients is possible and useful is uncertain and published data are scarce. Repeated RT with brachytherapy, laser ablation, or photodynamic therapy may be considered for some patients but will not be suitable for all. For all these treatment modalities, the tumor burden should be low and the tumor extension beyond the trachea or bronchus should be limited.6,7 Laser treatment can be as effective as brachytherapy, but the combination of laser ablation and brachytherapy can yield a two- to threefold prolongation of the palliation duration compared with laser therapy alone.8 Palliation in recurrent NSCLC is achieved in 67% of the patients treated with brachytherapy alone.9 Retreatment with external beam RT (EBRT) might be another option but the dose might be greater than the tolerance of the normal tissues such as lung parenchyma and spinal cord.10-14 We prospectively studied 28 patients who underwent repeated RT for proven recurrent locoregional NSCLC with special attention to the efficacy on relief of symptoms and the toxicity of such a treatment. We anticipated that about 50-60 patients would be suitable for reirradiation in the period of the study. However, the strict entry criteria, a slow accrual at the start, and a decreasing accrual at the end of the study owing to an increasing popularity of chemotherapy, resulted in a relatively small number of patients. Still, this number exceeds those of similar studies treated with a uniform schedule.10-14 Methods and materials Between August 1991 and April 1999, 28 patients with recurrent NSCLC were entered in this prospective study in which the value of retreatment with EBRT was tested. The patient characteristics are summarized in Table 1. All patients gave written informed consent before entry into the study. Patients were eligible for the study if the following criteria were met. Recurrent NSCLC had been proven by either positive histologic and/or cytologic findings. Also patients with visible recurrent tumor at bronchoscopy and/or evident tumor progression at chest-CT-scan were accepted.

Page 21: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

19

Abbreviations: KPS = Karnofsky performance score; NSCLC = Non-small-cell lung cancer; RT = radiotherapy; EBRT = external beam RT. Data in parentheses are RT regimens.

Recurrence was histologically confirmed in 19 patients. In 5 patients, tumor recurrence was clinically evident at bronchoscopy. In another 4 patients, obvious progression of tumor growth was seen on consecutive chest CT scans. All patients underwent a chest X-ray and/or chest CT to visualize tumor regrowth at their initial site to establish the diagnosis of recurrent disease. Additionally, patients had had superior vena cava syndrome (SVCS), dyspnea, hemoptysis, cough, or a combination of these. An overview of all diagnostic procedures and complaints is presented in table 2.

Abbreviation: n= number of patients; SVCS = superior vena cava syndrome.

Table 1. Patient characteristics

Gender (n) Male Female

27 1

Age (y) Median Range

68 52 – 83

KPS Median Range

80 40 - 90

Histologic type NSCLC

Initial RT (n) RT for recurrence after surgery (n) Adjuvant RT after surgery (n) RT as primary treatment (n) Repeated EBRT (n)

28

28 2 (55.2 Gy in 24fx 6 wk) 1 (46.0 Gy in 20 fx in 4 wk) 25 (36-60 Gy in12-30 fx in 3-6 wk) 28 (2 x 8 Gy, Days 1 and 8)

Interval to re-RT (mo) Median Range

17 6 – 72

Table 2. Complaints and diagnostic procedures

Diagnostic procedure

Complaint n Bronchoscopy Biopsy Chest X-ray Chest CT

SVCS 5 3 2 5 5

Hemoptysis 13 12 9 10 5

Dyspnea 19 17 16 19 16

Coughing 8 7 6 6 6

24 19 24 19

Page 22: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

20

All patients had been treated with EBRT in the past. Three patients had undergone a surgical resection as first-line treatment consisting of a lobectomy in two and bilobectomy in one patient. One of these patients subsequently underwent RT to the mediastinum to a dose of 46 Gy in 20 fractions during 4 weeks immediately postoperatively. The other 2 patients underwent RT to a dose of 55.2 Gy in 24 fractions during 6 weeks at 23 and 35 months after surgery because of a first recurrence. All other patients underwent primary RT to the tumor with a 10–20-mm margin and a field extending to the adjacent mediastinum. The total dose to the primary tumor varied from 36 to 60 Gy given in four to five fractions weekly in 2–3 Gy fractions. The dose to the mediastinum was 46 Gy, applied using the same fractionation scheme. One of these patients was treated three times for a local recurrence: first with brachytherapy, 8 months later with repeated EBRT, and another 8 months later with laser vaporization. All patients had been treated with 10- or 18-MV energy photon beams. The minimal interval from the initial RT course to the repeated EBRT had to be 6 months (median interval 17 months). Most patients had received radical RT as first treatment, during which the generally accepted spinal cord tolerance dose of 50 Gy had already been reached (15). Although, on basis of recent animal studies, the spinal cord can recover from injury (16), the lack of such data at the time of this study precluded repeated RT of the cord after previous RT up to 50 Gy. No special limitations were given according to the volume of lung parenchyma, esophagus, or heart to be reirradiated. At repeated RT, we tried to spare the contralateral lung as much as possible and took care with the volume of the total lung that was reirradiated to reduce the possible risk of radiation pneumonitis. Because the life expectancy for this population was limited, no special attention was paid to cardiac dose. Field sizes varied from 6 x 7 to 14 x 15 cm (mean surface area 104 cm2 and median field size 9.5 x 11 cm). Repeated RT was delivered at the site of the recurrence using two opposing oblique fields to avoid the spinal cord. Two fractions of 8.00 Gy were given on Days 1 and 8 with 6–18-MV photon beams. Chemotherapy during repeated RT was not allowed. No restrictions were made concerning concurrent medications, including steroids. Patients were followed according to the protocol for response, side effects, and survival from start of repeated RT at 6 and 12 weeks and every 3 months thereafter until death. At each follow-up visit, the relief of complaints, Karnofsky performance score (KPS), toxicity score according to the World Health Organization toxicity scale and complications were evaluated. To score the symptom response, we created our own scoring system because no validated system was available (17). This system takes into account the general subjective perception of symptom relief by the patient. The physician scored relief of complaints according to patient’s statement for each symptom as vanished (complete resolution of the symptom), as diminished (any improvement without complete resolution), as stabilized (no change), or progressive (deterioration). The best response at any time was reported. We also performed physical examination and chest X-ray followed by bronchoscopy and/or chest CT in case of abnormal clinical findings or progression of complaints. If a patient died at home, the date of death was checked with the general practitioner. Nevertheless, the cause of death remained unknown for 3 patients. The date of death, however, was retrieved for all, including the patient withdrawn from follow-up 4 months after treatment. The evaluation of symptoms could be assessed for 25 patients. We defined the symptom response period as the number of months between the start of repeated RT and assessment of progression of the specific symptom or at the time of death. The median and range of the duration of symptom response were calculated.

Page 23: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

21

The proportion of patients with symptom relief is presented, and 95% confidence intervals (Cis) were computed on the basis of the exact binominal distribution. Overall survival was measured in days from the start of repeated RT to the date of death, and the overall survival curve was estimated using the Kaplan-Meier method (18). Results Table 3 shows the palliative effect of the retreatment. The symptom response was computed up to progression of the symptom considered in each line of Table 3.

Abbreviations: V = vanished (complete resolution of the symptom); D = diminished (any improvement without complete resolution); S = stabilized (no change); P = progressive (deterioration); SVCS = superior vena cava syndrome; KPS = Karnofsky performance score. Numbers in parentheses are the range. Thirteen patients had only one symptom, 14 had two symptoms, and 1 had three symptoms at presentation. The relief of symptoms could not be scored in 3 patients because 1 patient was lost to follow-up, and 2 died before a response could be measured. Overall, 71% of the patients (95% CI, 51–87%) had partial or complete symptom relief of one or more of their symptoms. All assessable patients with SVCS showed improvement. Dyspnea was relieved in 6 (35%) of 17 assessable patients (95% CI, 14–62%) and stabilized in 10. In all 13 patients with hemoptysis, it disappeared or diminished. Coughing was relieved in 4 (67%) of 6 patients (95% CI, 22–96%) and stabilized in 1 patient. Among the 20 patients for whom change in KPS was known, symptom relief resulted in an improvement of the KPS in 9 (45%; 95%CI, 23–69%). In 8 patients, the KPS was stable. In 3 patients, the KPS deteriorated. The complete vanishing of symptoms was only seen in patients who had one symptom before repeated RT. The overall median duration of symptom relief was 4 months (range, 1-19 months). As is shown in figure 1, the palliative effect lasted in most patients until the last month of their life. However, those who lived >6 months had recurrent complaints during their remaining lifespan.

Table 3. Overview of symptom response after reirradiation

Symptom response (n)

Symptom

Not assessable

(n) V D S P

Persistent palliation

(n)

Median response

duration (mo)

SVCS 1 3 1 2 4 (2 –12)

Hemoptysis 12 1 8 5 (1 –14)

Dyspnea 2 4 2 10 1 6 4 (1 –19)

Cough 2 4 1 1 0 5 (2 –10)

KPS 8 9 8 3

(improved) (stabilized) (deteriorated)

Page 24: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

22

0 5 10 15 20 25

123456789

10111213141516171819202122232425

patie

nt n

umbe

r

months

SVCSdyspneahemoptysiscoughlife span

Figure 1. Relation between duration of symptom control and life span Esophagitis toxicity grade 3-4 was not seen. One patient, who had undergone brachytherapy for his first recurrence 8 months earlier, had Grade 2 esophagitis according to the World Health Organization toxicity scale. Radiation pneumonitis occurred in 1 patient. Treatment-related death occurred in 1 patient who died of a bronchoesophageal fistula following laser treatment 2 months after repeated RT. The overall survival is shown in Fig. 2. The median survival was 5.6 months (95 % CI, 4.4–8.8). Only 3 patients were still alive after 1 year. At the time of analysis, all patients had died. The interval between the primary treatment and repeated RT exceeded the survival time after repeated RT in all patients treated. None of the patients with an interval of <10 months survived >5 months. All patients surviving >1 year had a tumor-free interval of >1 year.

Page 25: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

23

Months after reirradiation

0 3 6 9 12 15 18 21 0

10

20

30

40

50

60

70

80

90

100%

0 N Number of patients at risk : 28 28 21 13 9 5 1 1 1

Figure 2. Overall survival

The cause of death could not be determined in 2 patients who died shortly (2 and 4 months) after retreatment, nor in the patient who withdrew from follow-up after 4 months. One patient probably died as a result of the retreatment. This patient was treated for hemoptysis with a field size of 8 x 9 cm. Two months later, because of renewed complaints of hemoptysis, laser treatment was given. A bronchoesophageal fistula occurred with pulmonary infection and subsequent demise. In the remaining 24 patients, local or distant metastasis occurred. Of the 20 patients with local tumor progression, 16 died as a result and 4 died of cachexia. Of the 4 patients without local tumor regrowth, 2 died of cachexia and 2 of cardiac failure. Five of the 13 patients referred for hemoptysis died after retreatment, after a period of symptom relief, owing to fatal hemoptysis from recurrent tumor growth (3, 5, 6, 11 and 11 months after repeated RT). The field size in these patients varied from 8 x 8 to 11 x 13 cm. Discussion The issue how to palliate patients with local tumor regrowth after previous radical RT is controversial. Only 2–3 % of the patients are amenable to additional radical treatment by reoperation or radical repeated RT. Repeated RT after previous radical RT has hazards, and only limited data are available regarding its effectiveness.10-14,19 The radiation dose reported to palliate symptoms by repeated EBRT varies from 6 to 60 Gy.10-14 Symptom improvement is reported to occur in 48–75% of patients.10-14 We reirradiated 28 patients to palliate symptoms of tumor regrowth with two 8-Gy fractions. In 71% of our patients, this treatment resulted in symptom relief. This schedule was most effective in palliating hemoptysis and SVCS (90–100%), but was less effective in palliating dyspnea and coughing (45%). The 2 x 8-Gy schedule has the advantage of causing little discomfort to the patient and being less dependent on the availability of radiation equipment and time, and the results seem equal to those obtained with more intensive schedules.12 The median survival of 5.6 months in our

Page 26: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

24

group is comparable with the median survival time in retrospective repeated RT studies (range, 3 – 5.4 months).10-14 The lack of specific criteria to define symptom response and palliation makes it difficult, or even impossible, to interpret and compare results across various studies.17 A recent study has validated the Edmonton Symptom Assessment Scale; however, it pertains to symptoms at 2 days and for pain only.20 Because we used a general subjective scoring of symptom response, the influence of co-medication could not be fully excluded. However, it is likely that the repeated RT mostly contributed to this symptom relief and its duration, because these patients were referred for repeated RT owing to insufficient symptom relief with best supportive care. The duration of palliation in relation to the remaining survival time is the most important factor in measuring the effectiveness of the retreatment. In none of the mentioned studies about repeated RT were details given about the duration of palliation. In 22 of our 25 patients, the percentage of their remaining life span during which palliation could be achieved was ≥ 50%. Most patients were palliated until a few weeks before they died. Symptom relief resulted in an improved or stabilized KPS in almost all assessable patients. These results are comparable with the findings of Bleehen et al 21 after primary palliative RT for NSCLC. Renewed progression of tumor growth after repeated RT finally resulted in the death of most patients, mostly within a few months. The observation of a relation between the interval of primary treatment and retreatment to overall survival time can be helpful in estimating the prognosis of the patients sent for retreatment. Death from massive hemoptysis is particularly distressing for patients and families. Nevertheless, repeated RT could not prevent this in 5 of 13 patients with hemoptysis. In 4 of these patients, renewed tumor progression was proven by bronchoscopy. The field sizes were comparable to those of the rest of the study population. Because none of the patients without hemoptysis at the time of repeated RT died of fatal hemoptysis, we consider these events primarily tumor related and not treatment related. The acute toxicity of this schedule was acceptable. Acute esophagitis was very mild and persisted only for a few weeks. The period between repeated RT and laser therapy could be of importance in reducing the risk of bronchoesophageal fistula.22 One patient who underwent laser treatment 8 months after repeated RT did not develop bronchoesophageal fistula. In contrast, the interval between laser treatment and repeated RT was only 2 months in the patient who did develop a fatal bronchoesophageal fistula. Data on the risk of radiation pneumonitis after repeated RT are scarce because the relation to dose and field sizes is rarely stated. Our treatment fields encompassed only the recurrent tumor burden to prevent unnecessary complications because of the high fractionation dose. The overall incidence in the literature of repeated RT varies from 0% to 60% (22 of a total of 138 patients) (10-14). Of these, Okamoto et al14 reported Grade 2 and 3 radiation pneumonitis in 19 of 34 patients who received either palliative or radical treatment. No relation to dose or treatment volume was given. Montebello et al12 had 1 patient with radiation pneumonitis after 51 Gy in 17 fractions during 28 days through 169 cm2 AP-PA fields. Gressen et al13 reported 1 of 23

Page 27: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

25

patients with fatal radiation pneumonitis after 30 Gy with concurrent and post-repeated RT chemotherapy. That we saw only 1 patient with radiation pneumonitis in our group after a field size of 8x9 cm suggests that 2 x 8 Gy is safe in this respect. Conclusions Repeated EBRT with 2 x 8 Gy can be very effective as palliative treatment for localized complaints due to tumor regrowth, especially for hemoptysis and SVCS. The complication rate was acceptably low. Despite a short median duration of the palliative effect, most patients were palliated during a substantial part of their survival. The palliation lasted in almost all patients > 50% of their remaining lifespan. Repeated RT, therefore, was an effective and safe treatment in the case of an isolated recurrent intrathoracic tumor with symptoms. References 1. Visser O, Coebergh JWW, Schouten LJ, et al, editors. Incidence of cancer in

the Netherlands 1997. Utrecht: Vereniging van Integrale Kankercentra; 2001. 2. Landis SH, Murray T, Bolden S, et al. Cancer statistics. CA Cancer J Clin

1998;48:6–9. 3. American Society of Clinical Oncology. Special Article: clinical practice

guidelines for treatment of unresectable non-small cell lung cancer. J Clin Oncol 1997;15:2996–3018.

4. Arriagada R, Le Chevalier T, Quoix E, et al. for the GETCB, FNCLCC and the CEBI trialists. ASTRO plenary –– Effect of chemotherapy on locally advanced non-small cell lung carcinoma: A randomized study of 353 patients. Int J Radiat Oncol Biol Phys 1991;20:1183–1190.

5. Perez CA, Pajak TF, Rubin P, et al. Long-term observations of the patterns of failure in patients with unresectable non-oat cell carcinoma of the lung treated with definitive radiotherapy: Report by the Radiation Therapy Oncology Group. Cancer 1987;59:1874–1881.

6. Morris CD, Budd JM, Godette KD, et al. Palliative management of malignant airway obstruction. Ann Thorac Surg 2002;74:1928–1932.

7. Metz JM, Friedberg JS. Endobronchial photodynamic therapy for the treatment of lung cancer. Chest Surg Clin North Am 2001;11:829–839.

8. Schray MF, McDougall JC, Martinez A, et al. Management of malignant airway comprise with laser and low dose rate brachytherapy. Chest 1988;93:264–269.

9. Speiser B, Spratling L. Intermediate dose rate remote afterloading brachytherapy for intraluminal control of bronchogenic carcinoma. Int J Radiat Oncol Biol Phys 1990;18:1443–1448.

10. Green N, Melbye RW. Lung cancer: Retreatment of local recurrence after definitive irradiation. Cancer 1982;49:865–868.

11. Jackson MA, Ball DL. Palliative retreatment of locally recurrent lung cancer after radical radiotherapy. Med J Aust 1987;147:391–394.

12. Montebello JF, Aron BS, Manatunga AK, et al. The reirradiation of recurrent bronchogenic carcinoma with external beam irradiation. Am J Clin Oncol 1993;16:482–488.

Page 28: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

26

13. Gressen EL, Werner-Wasik M, Cohn J, et al. Thoracic reirradiation for symptomatic relief after prior radiotherapeutic management for lung cancer. Am J Clin Oncol 2000;23:160–163.

14. Okamoto Y, Murakami M, Yoden E, et al. Reirradiation for locally recurrent lung cancer previously treated with radiation therapy. Int J Radiat Oncol Biol Phys 2002;52:390–396.

15. Fowler JF, Bentzen SM, Bond SJ, et al. Clinical radiation doses for spinal cord: the 1998 international questionnaire Radiother Oncol 2000;55:295–300.

16. Ang KK, Jiang G-L, Feng Y, et al. Extent and kinetics of recovery of occult spinal cord injury. Int J Radiat Oncol Biol Phys 2001;50:1013–1020.

17. Stephens RJ, Hopwood P, Girling DJ. Defining and analyzing symptom palliation in cancer clinical trials: a deceptively difficult exercise. Br J Cancer 1999;79:538–544.

18. Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc 1958;53:457–481.

19. Voltolini L, Paladini P, Luzzi L, et al. Iterative surgical resections for local recurrent and second primary bronchogenic carcinoma. Eur J Cardiothorac Surg 2000;18:529–534.

20. Chang VT, Hwang SS, Feuerman M. Validation of the Edmonton Symptom Assessment Scale. Cancer 2000;88:2164–2171.

21. Bleehen NM, Girling DJ, Fayers PM, et al. Inoperable non-small-cell lung cancer (NSCLC): a Medical Research Council randomised trial of palliative radiotherapy with two fractions or ten fractions. Br J Cancer 1991;63:265–270.

22. Khanavkar B, Stern P, Alberti W, et al. Complications associated with brachytherapy alone or with laser in lung cancer. Chest 1991;99:1062–1065.

Page 29: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Chapter 3

RANDOMIZED CONTROLLED TRIAL OF RESECTION

VERSUS RADIOTHERAPY AFTER INDUCTION

CHEMOTHERAPY IN STAGE IIIA-N2 NON-SMALL CELL

LUNG CANCER

Jan P. van Meerbeeck M.D., Ph.D.a*, Gijsbert W.P.M. Kramer M.D. b, Paul van Schil M.D., Ph.D.c, Catherine L. Legrand Sc.D. d, Egbert F. Smit M.D., Ph.D. e, Franz

Schramel M.D. f, Bonne Biesma M.D. PhD g, Vivianne Tjan-Heijnen M.D.PhD h, Nico van Zandwijk M.D. PhD i, Ted Splinter M.D. Ph.D.j, Giuseppe Giaccone M.D. PhD k,

l, on behalf of EORTC-Lung Cancer Group.

a Department of Respiratory Diseases, University Hospital, Ghent, Belgium; b

Radiotherapeutic Institute ARTI, Arnhem, The Netherlands; c Department of Thoracic and Vascular Surgery University Hospital of Antwerp, Edegem, Belgium; d EORTC

(European Organization for Research and Treatment of Cancer) Data Centre, Brussels, Belgium; e Department of Pulmonology Vrije Universiteit Medical Centre,

Amsterdam, The Netherlands; f St. Antonius hospital, Nieuwegein, The Netherlands; g

Jeroen Bosch Ziekenhuis ‘s-Hertogenbosch, The Netherlands; h Department of Medical Oncology University Medical Centre Nijmegen-St. Radboud, The

Netherlands; k Department of Pulmonology NKI Amsterdam, the Netherlands; j Erasmus University Rotterdam, The Netherlands; k Department of Medical Oncology

Vrije Universiteit Medical Centre, Amsterdam, The Netherlands, l.

J Nat Cancer Inst 2007; 99(6): 442-450

Page 30: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

28

Abstract

Background: Induction chemotherapy before surgical resection increases survival compared with surgical resection alone in patients with stage IIIA non–small-cell lung cancer (NSCLC). We hypothesized that following a response to induction chemotherapy, surgical resection was superior to thoracic radiotherapy as locoregional therapy. Methods: Patients with histologic or cytologic proven stage IIIA-N2 NSCLC were given three cycles of platinum-based induction chemotherapy. Responding patients were subsequently randomly assigned to surgical resection or radiotherapy. Survival curves were estimated using Kaplan-Meier analyses. Results: Induction chemotherapy resulted in a response rate of 61% (95% confidence interval=57 to 65%) among the 579 eligible patients. A total of 167 patients were allocated to resection and 165 to radiotherapy. Of the 154 (92%) patients who underwent surgery, 14% had an exploratory thoracotomy, 50% a radical resection, 42% a pathologic downstaging, and 5% a pathologic complete response; 4% died after surgery. Postoperative radiotherapy was administered to 62 (40%) of patients in the surgery arm. Among the 154 (93%) irradiated patients, overall compliance to the radiotherapy prescription was 55%, and grade 3/4 acute and late esophageal and pulmonary toxicities occurred in 4% and 7%; one patient died of radiation pneumonitis. Median and 5-year overall survival for patients randomly assigned to resection versus radiotherapy were 16.4 versus 17.5 months and 15.7% versus 14%, respectively (hazard ratio =1.06, 95% confidence interval=0.84 to 1.35). Rates of progression-free survival were also similar in both groups. Conclusion: In selected patients with pathologically proven stage IIIA-N2 NSCLC and a response to induction chemotherapy, surgical resection did not improve overall or progression-free survival compared with radiotherapy. In view of its low morbidity and mortality, radiotherapy is to be considered the preferred locoregional treatment for these patients.

Page 31: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

29

An estimated 1.2 million new cases of lung cancer occur yearly worldwide, resulting in annual fatalities of 183,000 in Europe and 160,000 in the United States (1). Eighty percent of all lung cancers are non–small-cell lung cancer (NSCLC), and approximately 15% of patients with NSCLC are diagnosed with stage IIIA-N2 disease (2). This subgroup is heterogeneous, with lymph nodes that are only microscopically invaded to those that are radiologically visible with bulky ipsilateral mediastinal lymph node involvement (3). Surgical resection in selected patients results in 5-year survival rates of 7–24% (4). Preoperative chemotherapy has been shown to increase 5-year survival to 17–36% (5-7). The combination of platinum-based chemotherapy and thoracic radiotherapy yielded a 5-year survival rate of 15% in a meta-analysis (8), and this combination has since been considered standard treatment for patients with unresectable stage IIIA disease. In a prospective study (9), concurrent chemoradiotherapy resulted in a 5-year survival rate of 20%. Based on the results of these previous studies, we hypothesized that surgery may be superior to radiotherapy following induction chemotherapy. Therefore we performed a large multicenter prospective randomized trial to compare surgery with radiotherapy in patients with stage IIIA-N2 NSCLC who showed a response to induction chemotherapy. Patients and Methods Eligible patients had to have cytologic or histologic proof of unresectable stage IIIA-N2 NSCLC (2). Staging included physical examination, a computed tomography (CT) scan of the thorax, and ultrasound or CT scan of the upper abdomen. Guidelines for unresectability were as follows: 1) any N2 involvement by a non-squamous carcinoma; 2) in case of squamous cell carcinoma, any N2 nodal involvement exceeding level 4R for a right-sided tumor and level 5 and 6 for a left-sided tumor. N2 found only at thoracotomy after a negative staging mediastinoscopy was not necessarily considered unresectable. Tumors and/or any involved mediastinal lymph node(s) had to be unidimensionally measurable on CT scan. Patients had to be older than 18 years, have a World Health Organization (WHO) performance status of 0–2, show no evidence of pulmonary fibrosis, and be considered fit for the combined modality treatment by their local multidisciplinary team. Patients with pre-existing neurotoxicity, with infections, who had had prior therapy for NSCLC, or had concurrent or prior malignancy were excluded. Patients had to give written informed consent for participation. Induction chemotherapy consisted of three cycles of cisplatin, at a dose of at least 80 mg/m² per cycle, or carboplatin, at a target area under the curve (AUC) of at least 5 per cycle, combined with at least one other chemotherapy drug. Three phase 2 trials investigating the activity of novel drug combinations (10-12) were nested within the present study. Response was evaluated with CT scan after at least two cycles of induction chemotherapy and scored according to WHO criteria (13), but confirmation was not required. Eligibility was reassessed before random assignment. Only patients showing a response (complete, partial, or minor) to induction chemotherapy were eligible for random assignment to either surgery or radiotherapy after stratification for type of response, histologic subtype, and institution (14). Locoregional therapy (i.e., surgery or radiotherapy) had to start within 6 weeks of random assignment. The standards, definitions, and criteria of radiotherapy and surgery have been reported previously (15, 16). A compliance score was defined based on the following radiotherapy

Page 32: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

30

prescription requirements: 1) an interval from the last day of induction chemotherapy until the start of radiotherapy of less than 10 weeks, 2) the use of three-dimensional planning, 3) the use of lung tissue correction, 4) a dosage administered to the primary tumor and involved mediastinum of 60–62.5 Gray (Gy) and to the uninvolved mediastinum of 40–46 Gy, 5) a fractionation size of 1.95 to 2.05 Gy, 6) a number of fractions of 30 to 32, and 7) a total treatment duration of 40 to 46 days (17). Mediastinal downstaging was defined as the absence of tumor on pathologic examination of the resected mediastinal lymph nodes. A resection was considered complete if, following review of the surgery and pathology report by one of the authors (PVS), both surgical margins and the highest mediastinal lymph node were found to be free of tumor. Postoperative radiotherapy consisting of 56 Gy in once-daily fractions of 2 Gy was recommended in cases of incomplete resection and had to start between the 4th and 10th postoperative week. Pulmonary function was measured at least once before and after surgery or radiotherapy. The management of treatment related toxicities and complications was left to the local investigator. Surgical complications were recorded descriptively. Radiotherapy toxicity was scored according to the National Cancer Institute of Canada Clinical Trials Group Expanded Common Toxicity Criteria (18). Patients underwent follow-up visits every 3 months for 2 years and every 6 months thereafter, which included clinical evaluation, a chest-X-ray, and additional investigations when clinically indicated. Statistical analysis and trial conduct The primary endpoint of the trial was overall survival measured from the day of random assignment and analyzed on all randomly assigned patients according to the intention-to-treat principle. Secondary endpoints were progression-free survival and safety. Progression-free survival was defined as time to progression or death, whichever came first, and both progression and death were considered as events. Assuming a 15% 5-year survival with radiotherapy, a two-sided type 1 error of 5%, and a power of 80%, 292 events were necessary to detect an increase to 25% in the 5-year survival with surgery. Assuming 8 years of accrual and 2 further years of follow-up, 358 patients had to be randomly assigned. At an historically assumed 1:1 randomization rate of 56%, 640 patients had to be accrued. The trial was closed after randomization of slightly fewer patients (332) than initially foreseen, after statistician’s advice that this early closure would not interfere with its power, because the observed number of deaths for the primary endpoint (279) was close to that required at the time of the analysis. The posterior power as computed with EAST v4.1, (Cytel Software Corporation, Cambridge, MA, USA) was approximately 75%. Randomization was performed by a central computer–generated random allocation sequence at the European Organisation for Research and Treatment of Cancer (EORTC) Data Center. Both arms were compared using a two-sided log rank test, and survival curves were estimated using the Kaplan-Meier technique. The statistical significance level was set at 0.05, and all comparisons were two-sided. Uni- and multivariable analyses of prognostic factors using the Cox proportional hazards model were conducted on all randomly assigned patients and per treatment arm. Proportionality was checked visually based on the Kaplan-Meier survival curves. Because there was no suggestion of nonproportional hazards, a formal test was not applied.

Page 33: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

31

Two interim analyses on all outcomes of the study were conducted after 80 and 146 deaths. Stopping rules for efficacy were specified a priori in the protocol, considering an alpha-spending function with O'Brien and Fleming boundary. The results of these interim analyses were declared to the members of the Independent Data Monitoring Committee only, who recommended continuing the trial. Following an audit of radiotherapy quality control and according to recommendations given during the first Independent Data Monitoring Committee review, the protocol was amended in 1999 as follows: 1) Radiotherapy prescription needed to be better detailed and adhered to (14, 17); 2) only the following induction chemotherapy regimens were allowed: cisplatin-docetaxel, cisplatin-gemcitabine, carboplatin-paclitaxel, cisplatin-vindesin (with or without ifosfamide), carboplatin-etoposide and cisplatin-ifosfamide-mitomycin; 3) uniform definitions of unresectability and complete resection were introduced and the available data reviewed accordingly; 4) a formal process of reporting serious adverse events and late treatment-related side effects was implemented. The study protocol and all amendments were approved by the Protocol Review Committee of the EORTC and the Ethical Committees of all participating institutions. This study was registered at www.clinicaltrials.gov with number NCT00002623.

Results A total of 582 patients were registered from December 1, 1994, to December 1, 2002, from 41 institutions of the EORTC-Lung Cancer Group. The study was prematurely closed due to lower than planned accrual at a slightly lower (n= 332) than the required number of randomly assigned patients. This study was analyzed in December 2005 after a median follow-up of approximately 6 years, and no losses to follow-up for overall survival occurred in the randomized patients. The CONSORT diagram is shown in Fig. 1. Patient, tumor, and induction chemotherapy characteristics of the 579 eligible and the 332 randomly assigned patients are provided in Table 1. Induction chemotherapy consisted mainly of a platinum/gemcitabine (40%) or a platinum/taxane combination (21%) (19). Eighty-seven percent of patients received three cycles of induction chemotherapy; nine patients never started induction chemotherapy. An overall response rate of 61% (95% confidence interval [CI] = 57 to 65%) was observed. Thirty-eight responding patients were not randomly assigned because of refusal (n=14), inoperability (n=15), unresectability (n=5), toxicity of induction chemotherapy (n=2), or for unknown causes (n=2). Retrospective analysis by response type in the 332 randomly assigned patients showed that 316 had an objective response, 12 had stable disease, and four progressed on induction chemotherapy. Of the 16 non-responding patients above, six were allocated to surgery and 10 to radiotherapy. The patient and tumor characteristics of the randomly assigned patients and the distribution of the observed responses to induction chemotherapy were well balanced. One hundred fifty-four (93%) of the 165 patients in the radiotherapy arm were irradiated (Table 2). The results of the radiotherapy quality assurance have been published (17). The overall compliance to the radiotherapy prescribed was 55%. The median overall treatment time from start of induction chemotherapy to the end of radiotherapy was 145 days (range=91–194). Acute grade 3–4 esophageal and pulmonary toxicities were observed in one (<1%) and five patients (4%), respectively. Late pulmonary and esophageal fibrosis occurred in 11 (7%) and 1 (<1%) patients, respectively, and one patient died of radiation pneumonitis.

Page 34: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

32

Fig. 1. Trial design and CONSORT diagram. This flow chart describes the different populations included in the study analysis: registered (n=582), eligible (n=579), randomized (n=332). Patients were allocated and actually treated as per protocol with either surgery or radiotherapy. One hundred fifty-four (92%) of the 167 patients in the surgery arm underwent surgery (Table 3). The results of the operated patients have been reported previously (15). Briefly, 14% had an exploratory thoracotomy, 50% a radical resection, 42% a pathologic downstaging, and 5% a pathologic complete response; 4% died within 30 days following surgery. Resection was performed after a median of 49 days (range=22–86) after the last cycle of induction chemotherapy. Postoperative radiotherapy was administered to 62 (40%) patients, among whom two cases of grade 3 acute esophagitis and one case of grade 4 acute pneumonitis was observed. Median overall survival, estimated from the time of registration in all 579 eligible patients, was 15.4 months (95% CI= 14 2 to 17.3). The results of overall and progression-free survival, estimated from the day of randomization, were similar in the 332 patients allocated to both treatment arms (Table 4). Median survival time and

No. of patients registered: 582 3 patients excluded because of lack of informed consent

No. of patients randomized: 332 (57%) Stratified for type of response, histological subtype and institution

No. allocated to Surgery: 167 No. allocated to Radiotherapy: 165

No. not irradiated: 11 (7%) - operated : 3 - unfit for Radiotherapy : 8

No. not operated: 13 (8%) - irradiated : 5 - unfit for Surgery : 8

Surgery: 154 (92%) Radiotherapy: 154 (93%)

No. of included patients: 579

No. of patients off study: 247(43%) - progressive or stable disease or death: 175 - toxicity or refusal: 37 - other: 35

Page 35: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

33

Fig. 2 Overall survival rates estimated from time of randomization using Kaplan-Meier analyses. P value (tw-sided) was calculated using the log rank test. 0=number of deaths; N=number of patients. Hazard ratio=1.06, 95% confidence interval=0.84 to 1.35; P=.596.

Fig. 3 Progression-free survival rates estimated from time of randomization using Kaplan-Meier analyses. P value (two-sided) was calculated using the log rank test. 0=number of deaths; N=number of patients. Hazard ratio=1.06, 95% confidence interval=0.85 to 1.33; P= .605.

Page 36: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

34

5-year overall survival percentages were 17.5 months and 14% in the radiotherapy arm and 16.4 months and 15.7% in the surgery arm, respectively. Kaplan-Meier analyses showed that overall survival (Fig. 2) and progression-free survival (Fig. 3) were similar among patients in both arms. Site of first relapse was more often locoregional in the radiotherapy arm and more often distant in the surgery arm (Table 4). Of all the baseline characteristics and stratification factors of the randomly assigned patients that were included in the multivariable analyses, only histologic subtype was prognostic, with a higher risk of death for patients with non-squamous histologies. After adjusting for histologic subtype, the treatment comparison was till not statistically significant. In the radiotherapy arm, none of the patient or treatment characteristics –including randomization before or after the protocol amendment- was prognostic. In the surgery arm, extent and type of resection were prognostic factors (for lobectomy versus pneumonectomy, hazard ratio [HR] = 0.59, 95% CI=0.40 to 0.87 and for complete resection versus non-radical resection, HR= 0.46, 95% CI=0.32 to 0.67). Unplanned exploratory subgroup analyses were performed in the 154 surgery-arm patients (Table 5). Patients who underwent a (bi-) lobectomy, had a complete resection, had pathologically proven mediastinal clearance had a statistically significantly better outcome than patients who underwent a pneumonectomy, had incomplete resection, or had no mediastinal clearance. Patients who received postoperative radiotherapy had similar overall survival outcomes as patients who did not. Multivariable analysis of survival showed that extent and completeness of resection, mediastinal clearance and postoperative radiotherapy were all statistically significant factors. However, due to their small number of patients these results should be considered with caution.

Page 37: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

35

Table 1. Characteristics of patients, tumors, and induction chemotherapy*

*This table describes the characteristics of the patients, their tumors, and their response to the induction chemotherapy in the whole group of eligible patients and in both randomized subgroups, regardless whether they actually received the allocated treatment. All percentages are rounded. VATS = Video Assisted Thoracic Surgery; cT= clinical Tumor stage. NA = not available. †At registration. ‡Including transbronchial and transthoracic fine needle aspiration or core biopsy.

Population Randomly assigned patients Variable

All registered patients (n=579)

Radiotherapy arm (n=165)

Surgery arm (n=167)

Sex, n (%) Male Female

427 (74) 152 (26)

127 (77) 38 (23)

119 (71) 48 (29)

Age, median (range)† 61 (29–78) 62 (33–76) 61 (29–78) Caucasian ethnicity, n % 552 (95) 160 (97) 162 (97) Histologic subtype, n (%)

Squamous carcinoma Adenocarcinoma Large cell carcinoma Other

NA NA NA

66 (40) 46 (28) 46 (28)

7 (4)

65 (39) 57 (34) 41 (25)

4 (2)

Proof of N2 by, n (%) Histology Cytology Clinically only

Type of biopsy, n (%) Mediastinoscopy VATS Thoracotomy Needle procedure‡

528 (91) 41 (7) 10 (2)

478 (83)

6 (1) 34 (6)

56 (10)

152 (92)

8 (5) 5 (3)

135 (82)

1 (1) 13 (8) 13 (8)

155 (93)

11 (7) 1 (1)

141 (84)

4 (2) 8 (5)

14 (8)

cT at registration, n (%) cT1 cT2 cT3 cT4 other/unspecified

67 (12) 396 (68) 100 (17) 13 (2) 3 (1)

21 (13) 119 (72) 25 (15)

0 1

20 (12)

120 (73) 25 (16)

1 0

Patients receiving three cycles of induction chemotherapy, N (%)

503 (87) 165 (100) 166 (99)

Median interval between registration and random assignment, days (range)

NA 71 (0–113) 73 (0–117)

Response on induction chemotherapy, N (%)

Complete response Partial and minor response Stable disease Disease progression

22 (4) 332 (57) 105 (18) 80 (14)

8 (5) 147 (89)

8 (5) 2 (1)

12 (7) 149 (89)

4 (2) 2 (1)

Page 38: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

36

Table 2. Characteristics of locoregional treatment in the thoracic radiotherapy arm (N = 154)*

Protocol prescription No. of patients treated according to prescription (%)

No. of fractions given = 30–32 131 (85) Fraction size = 1.95–2.05 Gy 134 (87) Duration = 40–46 days 123 (80) Interval between

Day 1 of last cycle induction chemotherapy and day 1 of radiotherapy within 10 weeks

Day 1 of last cycle induction chemotherapy and random assignment within 4 weeks

Random assignment and day 1 of radiotherapy within 6 weeks

133 (86)

93 (60)

136 (82)

Planning CT scan performed with lung correction

141 (92) 138 (90)

Dose and location of radiation primary tumor (60–62.5 Gy) mediastinum

involved (60–62.5 Gy) uninvolved (40–46 Gy)

129 (84)

115 (75) 134 (87)

*These figures relate to all patients who were allocated to the radiotherapy arm and were actually irradiated. CT= computed tomography; Gy=Gray.

Page 39: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

37

Table 3. Characteristics of locoregional treatment in the surgical resection arm (N = 154)* Characteristic No. of patients with this characteristic

(%) Extent of resection Pneumonectomy

right-sided left-sided

(Bi-)lobectomy Exploratory thoracotomy Other (remediastinoscopy)

72 (47) 38 (25) 33 (22) 59 (38) 22 (14) 1 (<1)

Type of resection Complete Pathologically complete

77 (50) 8 (5)

Downstaging ypN0 ypN1 ypN2 ypN3 missing

39 (25) 25 (16) 86 (56) 1 (1) 3 (2)

Operative mortality within 30 daysoverall after pneumonectomy after right-sided pneumonectomy after left-sided pneumonectomy after lobectomy after exploratory thoracotomy

6 (4) 5 (7) 2 (5) 3 (9) 0 (0) 1 (5)

PORT dosage (Gy), median (range) no. of fractions, median (range)

62 (40) 50 (0–60) 25 (8–30)

*These figures relate to all patients who were allocated to the surgery arm and were actually operated on. Mortality rates are expressed as number of deceased within 30 days/ number with the specific type of resection. Gy=Gray; PORT=postoperative radiotherapy; ypN=pathologic N after induction therapy.

Page 40: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

38

Table 4. Outcome after random assignment* Outcome Radiotherapy arm

(N= 165) Surgery arm

(N= 167) P†

Median follow-up, months 73 67 No. deceased (%) 141 (86) 138 (83) Overall survival (95% CI)

Median, months 2 year, % 5 year, %

Hazard ratio (95% CI)

17.5 (15.8 to 23.2)

41 (33 to 47.9) 14 (9 to 20) 1 (referent)

16.4 (13.3 to 19.0)

35 (28 to 42) 15.7 (10 to 22)

1.06 (0.84 to 1.35)

.6 Progression No. progressing (%) Site of first relapse

Locoregional Distant Both

130 (79)

71 (55)‡ 50 (39)‡ 9 (7)‡

115 (69)

37 (32)‡ 70 (61)‡ 8 (7)‡

Progression-free survival (95% CI)

Median, months 2 year, %

Hazard ratio (95% CI)

11.3 (8.9 to 12.7) 24 (18 to 31) 1 (referent)

9 (7.6 to 11.2) 27 (20 to 33)

1.06 (0.85 to 1.33)

.6 *These figures relate to all randomly assigned patients on intention-to-treat basis. CI=confidence interval. †P values were calculated using a two-sided log rank test. ‡Percentage of the patients progressing in either arm. Table 5. Exploratory analyses in 154 patients in the resection surgery arm* Subgroup N Median OS,

months (95%CI)

5-year OS, %

P, Univariate

analysis

P, Multivariate

analysis

Extent of resection

(bi-)lobectomy pneumonectomy

58 72

25.4 (17.7 to 48.9) 13.4 (11.1 to 19.5)

27 12

.009 .03

Mediastinal status

ypN0–1 ypN2

64 86

22.7 (17.6 to 42.7) 14.9 (11.2 to 18.5)

29 7

<.001 .04

Type of resection complete incomplete

77 76

24.1 (16.7 to 42.4) 12.1 (9.5 to 17.1)

27 7

<.001

.01

No PORT PORT

92 62

14.1 (11.2 to 19.9) 18.0 (15.0 to 25.9)

19 13

.6 .004

*OS=overall survival; PORT= postoperative radiotherapy; CI = confidence interval; ypN = pathologic N after induction therapy. P values were calculated using a two-sided log rank test.

Page 41: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

39

Discussion This large randomized multicenter study demonstrated that surgery did not improve survival after a radiologic response to induction chemotherapy in patients with unresectable stage IIIA-N2 NSCLC as compared with radiotherapy. These results are important because several centers routinely use induction chemotherapy followed by surgery to treat patients with this stage of disease based on small randomized studies that showed that surgery alone is inferior to perioperative chemotherapy and surgery in stage IIIA patients (5-7). The results of this study are robust in terms of numbers and mature in terms of follow-up and are similar to those observed in trials with a similar hypothesis but with a different design (20, 21) The North American Intergroup Trialists recently reported a median survival rate of 23.6 months and 22.2 months for induction chemoradiotherapy with or without resection, respectively.. The best treatment option for patients with stage IIIA-N2 has been debated. Part of the debate is caused by the heterogeneity of presentation at diagnosis, varying from intracapsular involvement of a single lymph node, in which surgery is indicated, to bulky mediastinal invasion, in which radiotherapy has been standard treatment until approximately a decade ago. A strength of our study is its requirement of pathologic proof of mediastinal involvement and the randomiz assignment of responders only. Although the definition of non-resectable tumor was left to the judgement of the local surgeon, clear guidelines were provided, and surgery and pathology reports were reviewed centrally. The limitations of this study rely mainly on evolving staging and treatment standards in the course of its accrual. We strongly feel that these do not influence the conclusions. One possible limitation is stage migration; this, will affect patient selection but is equally distributed over both arms by the process of randomization. Another is that changes in treatment standards occurred in both arms. However, their net magnitude on the outcome is likely to be limited. The complete resection rate in this series can be considered low in comparison with others (20, 21), but it should be interpreted with the definition that became the later standard (22). The feasibility of a sequential trimodality approach in patients with N2-disease, which was proven by mediastinoscopy, was explored in CALGB trial 8935 (23) In that trial, 74 patients were initially treated by two cycles of induction chemotherapy consisting of cisplatin and vinblastine. In patients with response or stable disease, surgical resection was performed followed by sequential adjuvant chemoradiotherapy. Sixty-three patients in CALGB 8935 underwent an exploratory thoracotomy with 46 (75%) having resectable lesions. Applying the same criteria for complete resection as in our study, 23 patients or 36.5% of those undergoing thoracotomy had a complete resection. In only 10 patients was the disease pathologically downstaged, and there were no pathologic complete responses. Operative mortality was 3.2%, and 7-year overall survival was 10% (24). Thus, in CALGB 8935 the complete resection rate was less than 40% after two cycles of induction chemotherapy, with only a small fraction of patients having a pathologic downstaging. In the present study applying the same criteria, three cycles of induction chemotherapy yielded a higher resectability rate. The survival of the not–completely resected patients in the present study is worse than that of patients in the radiotherapy arm. This difference may be explained by the fact that there is within the radiotherapy-arm a subgroup of patients with pathologically complete response and a subgroup of patients with mediastinal downstaging. Several series have shown that pathologic complete resection and mediastinal downstaging

Page 42: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

40

are stronger predictive factors than complete resection (25, 26). The number of patients in these two subgroups was not determined but is estimated to be similar to that of the corresponding subgroups in the surgery arm by the process of randomization after induction therapy. The observed high rate of pneumonectomies in this trial was likely the result of the prevailing opinion among the surgical community during its initial years—i.e., in patients with N2 disease the whole lung had to be removed. Later, lobectomy with systematic nodal dissection was regarded by most surgeons to be adequate treatment, provided that a complete resection was performed (15, 27). In subgroup analysis, we observed a statistically significantly better outcome after (bi-)lobectomy than after pneumonectomy, and this difference has also been observed in the North American Intergroup Trial (21). Pneumonectomy as such is also a known independent negative prognostic factor (28, 29), due in part to the higher stage of the disease in patients who require this extensive operation and also to an increased operative mortality associated with this procedure, especially for right-sided tumors. Surgical expertise and hospital volume are determinants of outcome and complication rate of lung cancer surgery (30). However, we observed a low post-operative mortality rate in our study, even among patients who underwent right-sided pneumonectomy. Patient selection may have contributed to this observation (247 patients were excluded for surgery or radiation therapy due to inadequate response). Furthermore, post-operative mortality is markedly high after chemoradiation (21), suggesting a possible detrimental effect of this induction combination. Surgery is highly unlikely to be beneficial when persistent mediastinal involvement is present after induction treatment, as shown in this study and by others (25, 26). Whether these patients should be offered further radical radiotherapy is debatable, because their disease will relapse mainly as distant metastases. Hence, the assessment of mediastinal clearance becomes very important. Fluoro-desoxy-glucose positron emission tomography scan, endoscopic ultrasound, and remediastinoscopy all claim a higher accuracy than CT-scan in restaging after induction therapy, and the techniques may complement each other (31). Whether only patients with mediastinal clearance should be offered surgery remains uncertain. Only an adequately powered trial, in which patients with proven mediastinal clearance are randomly assigned between surgery and radiotherapy, can truly address this question (32). The EORTC-LCG is considering such a study (van Meerbeeck, personal communication). The induction treatment used in this study was platinum-based chemotherapy. Although several different regimens were used, nearly all randomly assigned patients received three cycles, and major responses were achieved in more than 50% of them, similar to the results obtained in other series of induction chemotherapy (33-34). The rate of pathologic complete remission is lower with induction chemotherapy alone when compared with chemoradiotherapy (9, 35). Although pathologically complete response has predictive importance in other tumor types, it is still unknown whether response has relevance for long term survival in NSCLC. Furthermore, although chemoradiation per se does not increase the pneumonectomy rate, it is associated with higher rates of Adult Respiratory Distress Syndrome, bronchopleural fistula, and empyema (15). In a multicenter trial among IIIA-N2 NSCLC patients in Germany (36), the addition of twice-daily chemoradiation to induction chemotherapy, followed by resection, had no impact on outcome compared with induction chemotherapy and resection only but contributed to a statistically significantly higher rate of grade 3/4 esophagitis. In the US Intergroup Trial (21) the rate of severe post-operative complications and operative mortality was higher than in our study. A Swiss co-

Page 43: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

41

operative group study (26) reported a pathologic remission rate of 19% using cisplatin and docetaxel induction chemotherapy and observed a low operative morbidity and mortality and high survival rates, suggesting that third-generation induction chemotherapy might be superior to second-generation concurrent chemoradiotherapy. However, we could not reproduce the high response rate of this combination in a multicenter nested phase 2 trial performed within our trial (12). It is to be expected that improvements in radiotherapy timing, planning, and fractionation, together with the use of better tolerated chemotherapy agents or targeted agents, will result in better compliance and reduce the complication rate of concurrent chemoradiation (37). The role of the intensity of the induction treatment is the subject of an ongoing randomized US trial (38). During the 8 years that this trial was ongoing, radiotherapy standards underwent substantial improvements. This change in practice was addressed by a formal protocol-specified amendment to the prescription requirements. The tumor volume irradiated encompassed the pre-chemotherapy tumor areas with a margin that can be considered as optimal. Geographic misses have been reported to occur in reconstructing pre-chemotherapy target volumes in 26% of patients (39). This can result in a lower locoregional control, but few data from randomized trials on treating pre- or post-chemotherapy target volumes exist (40). Of more concern may be the interval between induction chemotherapy and the start of radiotherapy. Although 86% of patients in the radiotherapy arm started their radiotherapy within the prescribed 70 days of their last induction chemotherapy cycle, it has been shown that 41% of tumors progress when a mean interval of 80 days between induction chemotherapy and radiotherapy (41). Although only five patients were not treated after randomization because of clinical deterioration or tumor progression, this delay could be responsible for the high observed local relapse rate in the radiotherapy arm. Similar considerations apply with regard to the overall treatment time. The use of concurrent chemoradiotherapy has been reported to result in lower overall treatment times and is associated with better short-term survival results at the risk of higher toxicity (42). Our observed median overall treatment time of 145 days in the radiotherapy arm could be alone responsible for the observed difference in survival with the concurrent chemoradiotherapy arm of the Intergroup Trial (21) but other explanations are possible. Survival in the Intergroup Trial was counted from time of randomization, and thus a median of 2.5 months has to be added to compare survival times in this trial with those of studies with upfront randomization (20, 21, 36). Furthermore, most studies also combined induction treatment with postoperative chemotherapy (5-7, 9, 20, 21), and the latter has recently been associated with improved outcome, which may also bias the comparison (43). In conclusion, our study demonstrates that surgery does not improve survival compared with chest radiotherapy in patients with unresectable stage IIIA-N2 lung cancer after response to induction chemotherapy. In view of its low morbidity and mortality, radiotherapy is to be considered the preferred locoregional treatment for these patients. Properly designed studies to investigate whether surgery has a role in patients with confirmed mediastinal clearance are needed.

Page 44: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

42

Notes This publication was supported by grants number 5U10CA11488-24 through 5U10CA11488-34 from the National Cancer Institute (Bethesda, Maryland,USA). Its contents are solely the responsibility of the authors and do not necessarily represent the official views of the National Cancer Institute. The three nested phase 2 studies were supported by unrestricted educational grants of Eli Lilly, Bristol-Myers Squibb and Aventis. Nico van Zandwijk has received research support from E. Lilly and Company and has participated in their advisory board meetings and in their speaker’s bureau. We thank the patients and their families for their precious confidence and collaboration, as well as the following investigators and data managers for their collaboration in the successful conduct of this study: Anne Kirkpatrick, Sonia Dussenne, Annelore Dehoorne, Ann Gregor, Pierre Rocmans, Alexandre Passioukov. List of institutions and investigators in descending order of their respective accrual (in rounded %) to the 332 randomized patients: Free University Medical Center, Amsterdam, NL (E Smit, G Giaccone, J van Mourik, P. Postmus): 11; Erasmus MC, Rotterdam NL, (T Splinter, A Maat, S Senan, M de Jonge, J van Meerbeeck): 9; St. Antonius Ziekenhuis Nieuwegein NL (F. Schramel): 8; ARTI Arnhem, NL (G Kramer, H Smit, S Gans): 6; Radboud University MC, Nijmegen NL (J Festen, V Tjan Heijnen, J. Termeer): 6; Jeroen Bosch Ziekenhuis, ‘s Hertogenbosch NL (B. Biesma): 6; Netherland Cancer Institute, Amsterdam NL (N van Zandwijk, P Baas, J. Belderbos): 5; LUMC Leiden, NL (L. Willems): 5; UZ Gasthuisberg, Leuven B (J. Vansteenkiste, P. de Leyn): 4; UMCU, Utrecht NL (N. Schlösser, SY Elsharouni): 4; UZA Edegem B (P Van Schil, J van Meerbeeck): 3; Isala, Zwolle NL (J Stigt): 3; AZ Middelheim Antwerp B (D. Galdermans, K. Goor, P. De Roover): 2; OLV Gasthuis, Amsterdam NL (B. Kwa): 2; AZ St.Jan Brugge, B ( D. Van Renterghem): 2; Klinicni Center Golnik SL (A. Debeljak): 2; UCL Mont Godinne, Yvoir B (L. Delaunois): 2; Royal Marsden Hospital Sutton UK (I Smith, M. O’Brien, P. Goldstraw): 2; Ospedale SL Gonzaga, Torino, I (G. Scagliotti): 1; Spaarne Ziekenhuis, Haarlem, NL (H. van Breukelen): 1.; National Cancer Institute, Genoa, I (A. Ardizzoni ): 1; Slotervaartziekenhuis, Amsterdam NL (P. van Spiegel): 1; Maria Sklodowska-Curie MCC, Warsaw PL (M. Krzakowski): 1; Lievensberg Ziekenhuis, Bergen op Zoom NL (H. Liem); Franciscusziekenhuis, Roosendaal NL (A. van Boxem) ; UZGent B (P. Pinson, G. Joos); Weston Park Hospital, Sheffield UK (M. Hatton); Royal Infirmary, Leicester UK, (K. O’Byrne); Policlino Monteluce, Perugia I (S. Darwish); Clinique Ste. Elisabeth, Namur B (A. Baudoux); AZ Zusters Barmhartigheid Ronse B (J. Verschuere); CHR-St.Jozeph, Mons B (JP d’Odemont); Istituto Nazionale, Napoli I (C. Gridelli); Elkerliek Ziekenhuis, Helmond NL (P. van Valenberg); Ziekenhuis De Heel Zaandam, NL (A. van Bochove); Leyenburg Ziekenhuis, Den Haag NL (E. Lammers); Hopital de Jolimont, Haine- St. Paul, B (M. Beauduin); Twee Steden, Tilburg NL (H. Goey); Western General Hospital, Edinburgh UK (A. Price); AZ Hoge Beuken, Antwerpen B (W. Moorkens); Stedelijk Ziekenhuis, Roeselare B (W. Verbeke): all <1.

Page 45: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

43

References 1. http://www-dep.iarc.fr/ Globocan 2002 (accessed May 14, 2006) 2. Mountain CF. Revisions in the International System for Staging Lung Cancer.

Chest; 1997; 111:1710-7. 3. Grunenwald D, Le Chevalier T. Stage IIIA category of non-small cell lung cancer:

a new proposal. J Natl Cancer Inst 1997; 89:88–9. 4. Andre F, Grunenwald D, Pignon JP, Dujon A, Pujol JL, Brichon PY et al.

Survival of patients with resected N2 non-small-cell lung cancer: evidence for a subclassification and implications. J Clin Oncol. 2000; 18:2981-9.

5. Rosell R, Gomez-Codina J, Camps C, Maestre J, Padille J, Canto A et al. A randomized trial comparing preoperative chemotherapy plus surgery with surgery alone in patients with non-small-cell lung cancer. N Engl J Med 1994; 330:1153-8.

6. Roth JA, Fossella F, Komaki R, Ryan MB, Putnam JB Jr, Lee JS et al. A randomized trial comparing perioperative chemotherapy and surgery with surgery alone in resectable stage IIIA non-small- cell lung cancer. J Natl Cancer Inst 1994; 86: 673-80

7. Pass HI, Pogrebniak HW, Steinberg SM, Mulshine J, Minna J. Randomized trial of neoadjuvant therapy for lung cancer: interim analysis. Ann Thorac Surg 1992;53:992-8.

8. NSCLC Collaborative Group. Chemotherapy in non-small cell lung cancer: a meta-analysis using updated data on individual patients from 52 randomised clinical trials. Brit Med J 1995; 311:899-909.

9. Albain KS, Rusch VW, Crowley JJ, et al. Concurrent cisplatin/etoposide plus chest radiotherapy followed by surgery for stages IIIA (N2) and IIIB non-small cell lung cancer: mature results of Southwest Oncology Group phase II study 8805. J Clin Oncol 1995; 13: 1880-92.

10. Van Zandwijk N, Smit EF, Kramer GW, Schramel F, Gans S, Festen J et al. Gemcitabine and cisplatin as induction regimen for patients with biopsy-proven stage IIIA N2 non-small-cell lung cancer: a phase II study of the European Organization for Research and Treatment of Cancer Lung Cancer Cooperative Group (EORTC 08955). J Clin Oncol. 2000; 18:2658-64.

11. O'Brien ME, Splinter T, Smit EF, Biesma B, Krzakowski M, Tjan-Heijnen VC et al. Carboplatin and paclitaxel (Taxol) as an induction regimen for patients with biopsy-proven stage IIIA N2 non-small cell lung cancer. an EORTC phase II study (EORTC 08958). Eur J Cancer. 2003; 39:1416-22.

12. Biesma B, Manegold C, Smit HJM, Willems L, Legrand C, Passioukov A et al. Docetaxel and cisplatin as induction chemotherapy in patients with pathologically-proven stage IIIA N2 non-small cell lung cancer: a phase II study of the European Organization for Research and Treatment of Cancer (EORTC 08984). Eur J Cancer 2006; 42 (10): 1399-06.

13. WHO handbook for reporting results of cancer treatment. Geneva (Switzerland): World Health Organization Offset Publication No. 48; 1979.

14. Splinter TAW, Van Schil PE, Kramer GWPM, van Meerbeeck JP, Gregor A, Rocmans P et al. A. Randomised trial of surgery versus radiotherapy in patients with stage IIIA (N2) non-small cell lung cancer after a response to induction-chemotherapy EORTC 08941. A study of the EORTC Lung Cancer Cooperative Group. Clinical Lung Cancer 2000; 2:69-72.

Page 46: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

44

15. Van Schil P, van Meerbeeck JP, Kramer G, Splinter T, Legrand C, Giaccone G et al. Surgery after induction chemotherapy: morbidity and mortality in the surgery arm of EORTC 08941 trial. Eur Resp J 2005; 26: 192-8.

16. ICRU Report 50: prescribing, recording and reporting photon beam therapy. International Commission on Radiation Units and Measurements. 7910 Woodmont Avenue, Bethesda. 1993; ISBN 0-913394-48-3.

17. Kramer GWPM, Legrand CL, Van Schil P, Uitterhoeve L, Smit EF, Schramel F et al. Quality Assurance of Thoracic Radiotherapy in EORTC 08941: A randomised trial of Surgery versus Thoracic Radiotherapy in patients with stage IIIA Non-Small-Cell Lung Cancer (NSCLC) after response to induction chemotherapy. Eur J Cancer 2006; 42 (10): 1391-8.

18. Williams J, Chen Y, Rubin P, Finkelstein J, Okunieff P. The biological basis of a comprehensive grading system for the adverse effects of cancer treatment. Semin Radiat Oncol 2003; 13:182-8.

19. Van Meerbeeck JP, Kramer G, Van Schil P, Legrand C, O’Brien M, Manegold C et al. Induction chemotherapy (CT) in stage IIIA-N2 non-small cell lung cancer (NSCLC): an analysis of different regimens used in EORTC 08941. Lung Cancer 2003; 41: S(2)79, 273.

20. Johnstone DW, Byhardt RW, Ettinger D, Scott CB. Phase III study comparing chemotherapy and radiotherapy with preoperative chemotherapy and surgical resection in patients with non-small-cell lung cancer with spread to mediastinal lymph nodes (N2); final report of RTOG 89-01. Radiation Therapy Oncology Group. Int J Radiat Oncol Biol Phys. 2002; 54:365-9.

21. Albain KS, Swann RS, Rusch VR, Turrisi AT, Shepherd FA, Smith CJ et al. Phase III study of concurrent chemotherapy and radiotherapy (CT/RT) vs CT/RT followed by surgical resection for stage IIIA (pN2) non-small cell lung cancer (NSCLC): Outcomes update of North American Intergroup 0139 (RTOG 9309). J Clin Oncol 2005; 23: 7014.

22. Rami-Porta R, Wittekind C, Goldstraw P. Complete resection in lung cancer surgery: proposed definition. Lung Cancer 2005; 49(1):25-33.

23. Sugarbaker DJ, Herndon J, Kohman LJ, Krasna MJ, Green MR. Results of cancer and leukemia group B protocol 8935. A multi institutional phase II trimodality trial for stage IIIA (N2) non-small-cell lung cancer. Cancer and Leukemia group B Thoracic Surgery Group. J Thorac Cardiovasc Surg 1995; 109:473-85.

24. Kumar P, Herndon J, Sugarbaker D, Kohman L, Elias A, Green M. Long term survival outcome of a sequential trimodality trial in pathologically staged IIIA (N2) non-small cell lung cancer (NSCLC): final results of Cancer and Leukemia Group B (CALGB) protocol 8935. J Clin Oncol 2000; 19S:1939.

25. Bueno R, Richards WG, Swanson SJ, Jaklitsch MT, Lukanich JM, Mentzer SJ et al. Nodal stage after induction therapy for stage IIIA lung cancer determines patient survival. Ann Thorac Surg 2000; 70: 1826-31.

26. Betticher DC, Hsu Schmitz SF, Totsch M, Hansen E, Joss C, von Briel C et al. Mediastinal lymph node clearance after docetaxel-cisplatin neoadjuvant chemotherapy is prognostic of survival in patients with stage IIIA pN2 non-small-cell lung cancer: a multicenter phase II trial. J Clin Oncol 2003; 21:1752-9.

27. Liptay MJ, Fry WA. Complications from induction regimens for thoracic malignancies. Perioperative considerations. Chest Surg Clin N Am. 1999; 9(1):79-95.

Page 47: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

45

28. Van Meerbeeck JP, Damhuis RA, Vos de Wael ML. High postoperative risk after pneumonectomy in elderly patients with right-sided lung cancer. Eur Respir J 2002; 19:141-5

29. Martin J, Ginsberg RJ, Abolhoda A, Bains MS, Downey RJ, Korst RJ et al. Morbidity and mortality after neoadjuvant therapy for lung cancer: the risks of right pneumonectomy. Ann Thorac Surg 2001; 72: 1149–54

30. Birkmeyer JD. Surgeon volume and operative mortality in the United States. N Engl J Med 2003; 349: 2117-27.

31. Leyn P de, Stroobants S, Wever W de, Lerut T, Coosemans W, Decker G et al. Prospective Comparative Study of Integrated Positron Emission Tomography-Computed Tomography Scan Compared With Remediastinoscopy in the Assessment of Residual Mediastinal Lymph Node Disease After Induction Chemotherapy for Mediastinoscopy- Proven Stage IIIA-N2 Non–Small-Cell Lung Cancer: A Leuven Lung Cancer Group Study. J Clin Oncol. 2006;24(21):3333-9

32. Rami-Porta R. Induction Chemotherapy: A surgeon’s perspective. J Thor Oncol 2006; 1 (7): 605-6

33. Pisters KW, Ginsberg RJ, Giroux DJ, Putnam JB, Kris MG, Johnson DH et al. Induction chemotherapy before surgery for early-stage lung cancer: a novel approach. J Thorac Cardiovasc Surg 2000; 119: 429-39

34. Depierre A, Milleron B, Moro-Sibilot D, Chevret S, Quoix E, Lebeau B et al. Preoperative chemotherapy followed by surgery compared with primary surgery in resectable stage I (except T1N0), II, and IIIa non-small-cell lung cancer. J Clin Oncol. 2002; 1: 247-53

35. Eberhardt W, Wilke H, Stamatis G, Stuschke M, Harstrick A, Menker H et al. Preoperative chemotherapy followed by concurrent chemoradiation therapy based on hyperfractionated accelerated radiotherapy and definitive surgery in locally advanced non-small-cell lung cancer: mature results of a phase II trial. J Clin Oncol 1998;16(2):622-34.

36. Thomas M, Macha HN, Ukena D, Hamm M, Deppermann M, Semik M et al. Cisplatin / etoposide (PE) followed by twice-daily chemoradiation (hfRT/CT) versus PE alone before surgery in stage III non-small cell lung cancer (NSCLC): a randomised phase III trial of the German Lung Cancer Cooperative Group (GLCCG). J Clin Oncol 2004; 22S: 7004

37. Bonner JA, Harari PM, Giralt J, Azarnia N, Shin DM, Cohen RB et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med. 2006 Feb 9;354(6):567-78.

38. http://www.cancer.gov/clinicaltrials/RTOG-0412 (accessed February 25, 2006) 39. Lagerwaard FJ, van de Vaart PJ, Voet PW, Nijssen-Visser MR,Schuchhard-

Schipper RH, Joosten HP et al. Can errors in reconstructing pre-chemotherapy target volumes contribute to the inferiority of sequential chemoradiation in stage III non-small cell lung cancer (NSCLC)? Lung Cancer 2002;38:297–301

40. Liengswangwong V, Bonner JA, Shaw EG, Foote RL, Frytak S, Eagan RT et al. Limited-Stage Small-Cell Lung Cancer: Patterns of Intrathoracic Recurrence and the Implications for Thoracic Radiotherapy. J Clin Oncol 1994; 12:496-502

41. ElSharouni SY, Kal HB, Battermann JJ. Accelerated regrowth of non-small-cell lung tumours after induction chemotherapy. Br J Cancer 2003;89:2184–9.

Page 48: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

46

42. Rowell NP, O'Rourke NP. Concurrent chemoradiotherapy in non-small cell lung cancer. The Cochrane Database of Systematic Reviews 2004, Issue 4. Art. No.: CD002140.pub2.

43. Winton T, Livingston R, Johnson D, Rigas J, Johnston M, Butts C et al. National Cancer Institute of Canada Clinical Trials Group; National Cancer Institute of the United States Intergroup JBR.10 Trial Investigators. A prospective randomized trial of adjuvant vinorelbine (VIN) and cisplatine (CIS) in completely resected stage 1B and II non small cell lung cancer (NSCLC) Intergroup JBR.10. N Engl J Med 2005; 352: 2589-97.

Page 49: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Chapter 4

GEMCITABINE AND CISPLATIN AS INDUCTION REGIMEN

FOR PATIENTS WITH BIOPSY-PROVEN STAGE IIIA N2 NON-

SMALL-CELL LUNG CANCER: A PHASE II STUDY OF THE

EUROPEAN ORGANIZATION FOR RESEARCH AND

TREATMENT OF CANCER LUNG CANCER COOPERATIVE

GROUP (EORTC 08955)

N. Van Zandwijk, * E. F. Smit, * G. W. P. Kramer,† F. Schramel, † S. Gans, ‡ J. Festen,§ A. Termeer,§ N. J. J. Schlosser,|| C. Debruyne,|| D. Curran, || G. Giaccone||

*From the Netherlands Cancer Institute and Free University, Amsterdam; †Arnhem

Page 50: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

48

Summary Purpose: Our objective was to better define the activity/feasibility of gemcitabine/cisplatin (GC) as induction chemotherapy in patients with stage IIIA N2 non–small-cell lung cancer (NSCLC) followed by surgery or radiotherapy within a large, ongoing comparative study (EORTC 08941). Patients and Methods: Forty-seven chemotherapy-naive patients with NSCLC, median age of 58 years, stage IIIA N2 disease, World Health Organization performance status of 0 or 1, and the ability to tolerate a pneumonectomy received gemcitabine 1,000 mg/m2 on days 1, 8, and 15 and cisplatin 100 mg/m2 on day 2, every 4 weeks. Patients received induction chemotherapy (three cycles) before re-evaluation and randomization to surgery or radiotherapy. Results: Grade 3/4 thrombocytopenia, the main hematologic toxicity, occurred in 60% of patients but was not associated with bleeding. Full-dose gemcitabine was given in 48% of the courses. Severe nonhematologic toxicity was uncommon. Two patients with preexisting, autoimmune pulmonary fibrosis had deterioration of pulmonary

function after radiotherapy. Thirty-three (70.2%; 95% confidence interval, 55.1% to 82.7%) of the 47 eligible patients had objective responses (three complete responses and 30 partial responses). Mediastinal nodes were tumor-free after induction therapy in 53% of cases. Resections were considered complete in 71% of the patients who underwent thoracotomy after induction therapy. Median survival for all recruited patients (N = 53) was 18.9 months, with an estimated 1-year survival rate of 69%. Conclusion: In patients with N2 stage IIIA NSCLC, GC is a highly active and well-tolerated induction regimen. GC should be explored in combination with surgery or radiotherapy in stage I and II patients.

Page 51: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

49

Introduction The rationale for using induction chemotherapy in locally advanced non–small-cell lung cancer (NSCLC) is based on the considerations that chemotherapy may prevent the growth of systemic disease and, at the same time, shrink the locoregional macroscopic disease, which may then be adequately treated with surgery, radiotherapy, or both.1 Several groups have shown that this approach is feasible in patients with stage III disease, who usually have better response rates than patients with stage IV disease.2,3 Moreover, three small randomized trials comparing surgery alone with a combined program of induction therapy followed by surgery have shown prolonged survival in the combined-modality arms.4-6 In many centers, induction therapy followed by surgery and/or radiotherapy has thus become standard therapy for selected groups of patients with stage III disease.7 Gemcitabine (2',2'-difluorodeoxycytidine), an analog of deoxycytidine with established activity in NSCLC, is an anticancer drug with novel properties and mechanism of action.8 The combination of gemcitabine and cisplatin (GC) has been shown to be synergistic in preclinical studies.9,10 In patients with advanced NSCLC, this combination produced objective response rates of more than 50%.11,12 In three randomized studies in the same category of patients, GC was more active than cisplatin alone13 and produced a higher response rate as well as a longer response duration and time to progressive disease than the etoposide-cisplatin combination.14 In addition, GC produced a higher response rate than the three-drug combination of mitomycin, ifosfamide, and cisplatin.15 The early suggestions of increased efficacy of the GC combination prompted the European Organization for the Research and Treatment of Cancer (EORTC) Lung Cancer Cooperative Group to initiate a phase II trial to better define the toxicity and activity of this combination as an induction regimen for patients with stage IIIA NSCLC. This trial was the first of a series of phase II studies using new chemotherapy combinations within the setting of one large, ongoing randomized trial in patients with stage IIIA N2 disease (EORTC 08941) comparing surgery with radiotherapy after induction chemotherapy. Patients and Methods Patient Selection Eligibility criteria for study entry included the following: stage IIIA NSCLC with biopsy proof of positive N2 nodes, World Health Organization (WHO) performance status score of 0 to 1, and an adequate baseline organ function defined as a WBC count of at least 3,000/µL, liver and renal function within normal limits, and spirometric values allowing pneumonectomy. Patients who had previous chemotherapy, radiotherapy, or presence of active infection were excluded from the study. All patients gave written informed consent, and the study was approved by the EORTC Protocol Review Committee (PRC) and by the ethics committees of the participating centers.

Page 52: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

50

Treatment Plan Gemcitabine 1,000 mg/m2 was administered as a 30-minute intravenous infusion on days 1, 8, and 15 of a 28-day treatment cycle. On day 2, cisplatin 100 mg/m2 was infused over 30 minutes after intravenous hyperhydration with 1,500 mL saline (plus potassium chloride) and administration of standard antiemetics. The doses of gemcitabine and/or cisplatin were adjusted to the outcomes of blood counts and blood chemistry immediately before the infusion(s), and toxicity was recorded during treatment. Full doses of chemotherapy were given if WBC and platelet counts on the day of treatment were 3 x 109/L and 100 x 109/L, respectively. Chemotherapy was administered at 75% of the planned dose in the event of grade 2 hematologic toxicity and at 50% of the planned dose in the presence of grade 2 neurotoxicity or a creatinine clearance rate less than 60 mL/min. Dose reductions were handled the same way for both agents. Doses were withheld if myelotoxicity or neurotoxicity exceeded grade 2 or if creatinine clearance dropped below 40 mL/min. Three cycles of chemotherapy were administered unless progressive disease or intolerable toxicity was recorded. Patient Evaluation A complete history, physical examination, complete blood cell count with differential, serum biochemistry, urinalysis, spirometry, bronchoscopy, computed tomography (CT) scan of the chest and upper abdomen, and ECG were obtained at baseline. Patients were monitored throughout treatment by recording history, toxic events, and complete blood cell counts with differential (weekly); serum chemistry determinations were repeated just before the start of each chemotherapy cycle. Tumor response was evaluated by CT scan after three cycles of treatment. CT examinations were reviewed by an independent radiologist, the study chairman, and the responsible physician. A complete response (CR) was defined as the complete disappearance of all abnormalities on the CT scan. A partial response (PR) was defined as a reduction of more than 50% in the sum of the products of two perpendicular diameters of the primary tumor and hilar and mediastinal nodes (if enlarged). Stable disease was defined as a less than 50% reduction or a less than 25% increase in the sum of the products of two perpendicular diameters per site. Response rate calculations were based on all eligible patients (N = 47). Almost all patients with responsive disease gave consent to be randomized to thoracic surgery or radical radiotherapy (ongoing trial EORTC 08941). Repeated mediastinoscopy/tomy and spirometry were not included in the postchemotherapy evaluation. The combined-modality approach of this study did not allow the formal 4-week confirmation of response to chemotherapy alone according to WHO criteria. Overall, survival was measured from the date of registration to the date of death or last follow-up examination and was estimated using

the Kaplan-Meier method.16 Results Patient Characteristics From March 1996 to August 1997, 53 patients were enrolled onto the study. Six patients were ineligible: three patients had stage IIIB disease and another three patients had started protocol treatment before registration.

Page 53: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

51

Table 1. Patient characteristics

Characteristic No. of patients %

Total recruited 53 NA

Total eligible 47 NA Sex Male 33 70 Female 14 30 Age, years Median 58 Range 44-78 TNM stage

T1N2M0 3 6 T2N2M0 40 85 T3N2M0 4 9 Histology

Squamous cell 20 43 Adenocarcinoma 10 21 Undifferentiated 15 32 Large-cell 2 4 WHO performance status

0 28 60 1 19 40

Abbreviations: NA, not applicable; TNM, tumor-node-metastasis system. Characteristics of the 47 eligible patients (listed in Table 1) included a median age of 58 years (range, 44 to 78years), WHO performance status of 0 in 60% of the patients, and squamous cell carcinoma as the predominant histologic subtype (43%). All patients had N2 disease. In one case, the N2 involvement was found by thoracotomy (open-close): one of the mediastinal nodes appeared less than 1 cm on the CT scan and was found to be adherent to the superior caval vein; thus no primary surgery was attempted. In all other cases, mediastinal lymph node involvement was confirmed by mediastinoscopy/tomy (42 cases) or needle biopsy/aspiration cytology (four cases). In 32 of the 47 patients, baseline CT scans provided an estimate of the largest diameter of the N2 nodes. This diameter was less than 10 mm in one case, between 10 and 20 mm in 10 cases, between 20 and 30 mm in 11 cases, and more than 30 mm in 10 cases. Among the 15 remaining patients in whom CT did not allow an exact

measurement, there were at least five patients with mediastinal lymph node enlargement on plain-chest roentgenograms. Hematologic Toxicity During 127 treatment cycles with GC, grade 3/4 neutropenia was observed in 38.3% of the patients. Only one patient (2.1%) had febrile neutropenia. Grade 3/4 anemia was seen in 14.9% of the patients. Thrombocytopenia was the predominant hematologic toxicity: grade 3/4 thrombocytopenia was seen in 46% of the treatment

cycles (59.6% of the patients; Table 2); however, thrombocytopenia was not associated with an increased number of bleeding events, and no platelet transfusions were administered. Overall, hematologic toxicity frequently caused dose omission (34%) or reduction (23%) of the gemcitabine doses on day 15.

Page 54: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

52

Table 2. Hematologic Toxicity (N = 47)

nadir*

grade 0 grade 1 grade 2 grade 3 grade 4

No. of pts

% No. of pts

% No. of pts

% No. of pts

% No. of pts

%

Leukocytes 7 14.9 10 21.3 16 34.0 14 29.8 –

Neutrophils 16 34.0 2 4.3 11 23.4 8 17.0 10 21.3

Thrombocytes 2 4.3 4 8.5 13 27.7 11 23.4 17 36.2

Hemoglobin – 13 27.7 27 57.5 6 12.8 1 2.2

Abbreviations: pts = patients. *National Cancer Institute common toxicity criteria. Nonhematologic Toxicity Nonhematologic adverse events were typical of conventional cisplatin-based

combinations used in patients with NSCLC (Table 3). Nausea and vomiting occurred frequently but reached grade 3 in only a minority of patients (14.8% and 10.6%, respectively). Grade 3/4 neurotoxicity mainly involved ototoxicity (four patients). Sensory neuropathy never exceeded grade 1.

Table 3. Number of patients with nonhematologic toxicities (N = 47)

Grade*

0 1 2 3 4 3/4 (%)

Nausea 10 13 17 7 — 14.8

Vomiting 14 10 18 5 — 10.6

Headache 36 4 1 0 0 0

Sensory neuropathy

37 9 0 0 0 0

Other neurotoxicity†

23 8 8 3 2 10.6

Alopecia 19 8 14 5 — 10.6

Lethargy 12 16 16 3 — 6.4

Creatinine 35 9 1 — 1 2.1

Febrile neutropenia

46 — — 1 — 2.1

Infection 37 — 3 1 — 2.1

Diarrhea 38 6 2 1 — 2.1

Hemorrhage 29 11 1 1 — 2.1

Weight loss 38 4 1 1 — 2.1

Shortness of breath

36 1 — 1 — 2.1

* National Cancer Institute common toxicity criteria. † Other neurotoxicity excludes constipation, headache, and sensory neurotoxicity.

Page 55: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

53

Flu-like symptoms were occasionally seen, and alopecia (grade 2 or more) was noted in 40% of patients. In two patients with known seropositive rheumatoid arthritis and pulmonary fibrosis, pulmonary dysfunction deteriorated after radiotherapy. One patient died 1 month after the radiotherapy. The autopsy confirmed a complete remission but also revealed the presence of organizing diffuse alveolar damage in addition to pulmonary fibrosis. The other patient died 7 months after radiotherapy with signs of pulmonary dysfunction in addition to recurrent disease that was diagnosed 5 months earlier. Dose-Intensity Cisplatin was administered within the 90% to 110% range of the intended dose in 92% of the courses. As a result of omissions and dose reductions, only 48% of the courses were at the intended dose of gemcitabine, which translated into 259 infusions at the intended dose, 66 infusions at reduced dose, and 55 omitted infusions. Thrombocytopenia on day 15 was the main reason for dose reductions or withholding of gemcitabine. Because of a miscalculation of body-surface area, one patient received more than 110% of the intended doses of cisplatin and gemcitabine during cycle 2. Response to Induction Therapy and Additional Treatment The CT scans of responding patients were reviewed and all were independently confirmed, with the exception of one patient for whom CT scans could not be obtained. A summary of treatment and responses is presented in Fig 1. Of the 38 patients who received the planned induction therapy of three cycles of GC, 33 patients responded (three CRs and 30 PRs), for an overall response rate of 70.2% (95% confidence interval, 55.1% to 82.7%). In addition, there were three patients with no change (minor response in two) and two with progressive disease. Among the nine patients who did not receive the planned induction therapy of GC, seven discontinued GC therapy because of toxicity (ototoxicity in three cases). Three of these seven patients achieved PR with other gemcitabine combinations and were subsequently randomized to additional treatment with surgery or radiotherapy per EORTC 08941. Thus a total of 35 patients were randomized to additional surgery (n = 17) or radiotherapy (n = 18) per EORTC 08941. In the group of patients who underwent thoracotomy, minor (n = 1), partial (n = 15), and complete (n = 1) clinical responses were confirmed. The surgeon decided to perform an open-close thoracotomy in one case and resection in 16 cases. Resections were judged to be complete in 71% of the patients. After induction therapy, the mediastinal nodes of 53% of the cases were tumor-free. Twelve patients were not randomized to additional surgery or radiotherapy. Reasons for nonrandomization included cerebral infarction for one patient, lack of response for five patients (one was later corrected to PR by review panel) for patients who received three cycles of GC, toxicity for two patients who received two cycles of GC, and toxicity, progression, and second primary tumor in two, one, and one patient(s), respectively, who received one cycle of GC.

Page 56: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

54

Fig 1. Summary of treatment and response. Abbreviations: NC, no change; MR, minor response. aPatients not randomized to radiotherapy or surgery (n = 12). bOne patient was judged to have stable disease by the attending physician, but the review panel documented a PR. After induction treatment, one patient received laser therapy (second primary tumor) and nine received radiotherapy. Two patients, who stopped protocol treatment with GC, continued with other chemotherapy combinations followed by surgery. Survival At the time of this analysis, after a median follow-up period of 19 months, 29 of the 53 patients have died. In 27 cases, the deaths were related to the malignant diagnosis. One patient died from a cardiovascular cause. In another case, a (viral) infection was noted as the cause of death. The median duration of survival was 18.9 months, and the estimated 1-year survival rate was 69% (95% confidence interval, 56% to 82%). The survival curve is depicted in Fig 2.

Surgery N = 17

(1 CR, 15 PR, 1 MR)

Eligible patients N = 47

GC followed by other

gemcitabine combinations

3 cycles N = 3 (3 PR)

GC 3 cycles N = 38

GC 3 cycles N = 6a

(2 PRb, 2 NC, 2 PD)

GC 2 cycles N = 2b

GC 1 cycle N = 4 a

(1 PD)

N = 32 (3 CR, 28 PR,

1 NC)

N = 6 radiotherapy

1 radiotherapy 1 chemotherapy

+ surgery + radiotherapy

2 radiotherapy 1 chemotherapy

+ surgery 1 lasertherapy

Radiotherapy N = 18

Induction Chemotherapy

Randomized N = 35 Per ongoing trial EORTC 08941

Page 57: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

55

(months)

0 3 6 9 12 15 18 21 24 27 30 33

0

10

20

30

40

50

60

70

80

90

100

O N Number of patients at risk :

29 53 53 47 40 36 27 15 11 3 2 1 All patients

Fig 2. Duration of survival. Discussion In the last 10 to 15 years, many phase II trials have investigated multimodality treatment strategies that included induction chemotherapy and surgical resection in patients with stage III NSCLC. The substantial variability in the design of these trials makes interpretation of the results difficult. However, it has been well recognized that patients with stage III NSCLC are consistently more sensitive to preoperative chemotherapy than patients with stage IV disease.17 Therefore, the EORTC Lung Cancer Cooperative Group decided to further study new chemotherapy combinations

within the framework of a comparative study in patients with NSCLC IIIA N2, evaluating the role of surgery and radiotherapy after induction therapy. GC was selected as the first combination tested. Preclinical data led this research group to adopt the GC schedule developed by Crinò et al,11 in which gemcitabine is given on days 1, 8, and 15 and cisplatin on day 2. This regimen allows incorporation

of gemcitabine in the DNA before cisplatin is given, which in experimental studies seems to favor synergistic interaction.9,10 In this context, it is worth mentioning that reanalysis of clinical (phase II) studies also suggested better efficacy of cisplatin when given after gemcitabine.18,19 When comparing the toxicities of different GC schedules, the administration of cisplatin on day 2 has been associated with more pronounced thrombocytopenia than that observed with cisplatin administration on day 15. The results of this study, in which almost 60% of patients had grade 3/4 thrombocytopenia, are in line with this observation. Fortunately, this thrombocytopenia was not associated with an increase in bleeding events.

Page 58: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

56

Nonhematologic toxicities were typical for a cisplatin-containing combination. The worsening of pulmonary function that occurred after radiotherapy in two patients with preexisting pulmonary fibrosis reinforces the fact that radiation pneumonitis is seen

more often after combined-modality treatment. Gemcitabine is a potent radiosensitizer that has been associated with severe radiation pneumonitis when given at full dose concurrently with radiotherapy.20,21 On the other hand, the data showing the feasibility

of combining gemcitabine at lower doses with concurrent radiotherapy in patients with stage III NSCLC22 indicate that the preexisting fibrotic pulmonary lesions must have contributed to the pulmonary dysfunction observed. Patients with preexistent fibrotic pulmonary disorders must, therefore, be excluded from the combination of GC and radiotherapy until the interactions with pulmonary fibrosis are better understood. Overall, the clinical results of this study confirm previous observations that the GC combination has acceptable toxicity. The high response rate produced by three courses of GC is accounted for by the inclusion of all eligible patients and the exclusion of responding patients completing fewer than three courses of GC. The response rate of 70.2% is far greater than the percentages demonstrated in previous phase II and III studies in patients with advanced or metastatic NSCLC. This difference is likely related at least in part to the increased sensitivity to chemotherapy of patients with stage III disease who have a good performance status. On the other hand, this response rate, which is among the highest for preoperative chemotherapy, occurred despite significant tumor bulk; in fact, the large majority of patients had significant N2 enlargement on CT and a relatively low exposure to gemcitabine. It may be theorized, therefore, that a day 2

cisplatin schedule clearly exhibits synergism, resulting in greater activity and increased hematologic toxicity. In the past, it has been difficult to prove the superiority of one chemotherapy regimen against NSCLC over another. Taking into account the activity of the GC combination as induction treatment in this trial, there is a good reason to consider GC for induction therapy in patients with earlier-stage disease. A preliminary report also indicates that this group of patients may benefit from induction chemotherapy.3 Of the 17 patients

randomized to the surgical arm after induction therapy, 94% were operable; in 71% of these cases, the resection was considered complete. Especially important is the fact that mediastinal lymph nodes were rendered tumor-free in 53% of cases. This figure is somewhat higher than that obtained in the Memorial Sloan-Kettering Cancer Center experience, but exactly the same as that reported after the trimodality approach (concurrent chemotherapy and irradiation followed by surgery) used by the Southwest Oncology Group (SWOG). Nodal downstaging is of particular interest, as it was the only significant prognostic factor in a multivariate analysis of this SWOG (8805) trial.23 Other data in this SWOG study, including a 46% major pathologic response rate in patients with stable disease, suggested that chemotherapy given concurrently

with radiotherapy may be more effective than chemotherapy alone. Interestingly, the percentage of patients who underwent complete resection in this study was identical (71%) to that of the SWOG study, and median survival was similar (18.9 months in EORTC 08955 v 15 months in SWOG 8805). The question of whether radiotherapy is necessary in the induction regimen24 remains important, and it is expected that the outcomes of this study will encourage investigations optimizing the addition of radiotherapy to GC.22 Concerning the GC schedule, the present experience indicates that slight modifications may be advisable. While keeping dose-intensity largely unchanged, the schedule can be limited to 3 weeks by giving gemcitabine (1,250 mg/m2) on days 1

Page 59: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

57

and 8 only and cisplatin (75 mg/m2) on day 2. An almost identical schedule has already been shown to be feasible.14 In conclusion, our data show that GC with cisplatin on day 2 is highly active in patients with stage IIIA N2 NSCLC. The combination of GC with surgery and radiotherapy was feasible, and the present results encourage the exploration of this combination in patients with resectable, early-stage disease. Acknowledgments Supported in full by the EORTC Lung Cancer Cooperative Group. References 1. Lilenbaum RC, Green M: Multimodality therapy for non-small cell lung

cancer. Oncology 8:25–31,1994. 2. Martini N, Kris MG, Flehinger BJ, et al: Preoperative chemotherapy for stage

IIIa (N2) lung cancer: The Sloan-Kettering experience with 136 patients. Ann Thorac Surg 55:1365–1373,1993[Abstract].

3. Depierre A, Milleron B, Moro D, et al: Phase III trial of neo-adjuvant chemotherapy (NCT) in resectable stage I (except T1N0), II, IIIa non-small cell lung cancer (NSCLC): The French experience. Proc Am Soc Clin Oncol 18:465a,1999 (abstr 1792).

4. Rosell R, Gomez-Codina J, Camps C, et al: A randomized trial comparing preoperative chemotherapy plus surgery with surgery alone in patients with non-small cell lung cancer. N Engl J Med 330:153–158,1994.

5. Roth JA, Fossella F, Komaki R, et al: A randomized trial comparing perioperative chemotherapy and surgery with surgery alone in resectable stage IIIA non-small cell lung cancer. J Natl Cancer Inst 86:673–680, 1994[Abstract].

6. Pass HI, Pogrebniak HW, Steinberg SM, et al: Randomized trial of neoadjuvant therapy for lung cancer: Interim analysis. Ann Thorac Surg 53:992–998,1992[Abstract].

7. American Society of Clinical Oncology: Clinical practice guidelines for the treatment of unresectable non-small cell lung cancer: Adopted on May 16, 1997 by the American Society of Clinical Oncology. J Clin Oncol 15:2996–3018,1997[Abstract].

8. Luud B, Kristjanson PEG, Hansen HH: Clinical and preclinical activity of 2',2'-difluorodeoxycytidine (gemcitabine). Cancer Treat Rev 19:45–55,1994.

9. Bergman AM, Ruiz van Haperen VWT, Veerman G, et al: Synergistic interaction between cisplatin and gemcitabine in vitro. Clin Cancer Res 2:521–530,1996[Abstract].

10. Peters GJ, Ruiz van Haperen VWT, Bergman AM, et al: Preclinical combination therapy with gemcitabine and mechanisms of resistance. Semin Oncol 23:16–24,1996.

11. Crinò L, Scagliotti G, Marangolo M, et al: Cisplatin-gemcitabine combination in advanced non-small cell lung cancer: A phase II study. J Clin Oncol 15:297–303,1997[Abstract].

Page 60: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

58

12. Abratt RP, Bezwoda WR, Goedhals L, et al: Weekly gemcitabine with monthly cisplatin: Effective chemotherapy for advanced non-small cell lung cancer. J Clin Oncol 15:744–749,1997[Abstract].

13. Sandler A, Nemunaitis J, Dehnam C, et al: Phase III study of cisplatin with or without gemcitabine in patients with advanced NSCLC. Proc Am Soc Clin Oncol 17:454,1998 (abstr 1747).

14. Cardenal F, Paz López-Cabrerizo M, Antón A, et al: Randomized phase III study of gemcitabine-cisplatin versus etoposide-cisplatin in the treatment of locally advanced or metastatic non-small cell lung cancer. J Clin Oncol 17:12–18,1999.

15. Crinò L, Conte P, De Marinis F, et al: A randomized trial of gemcitabine cisplatin versus mitomycin, ifosfamide and cisplatin (MIC) in advanced non-small cell lung cancer (NSCLC): A multicenter phase III study. Proc Am Soc Clin Oncol 17:455,1998 (abstr 1750).

16. Kaplan EL, Meier P: Nonparametric estimation from incomplete observations. J Am Stat Assoc 53:457–481,1958.

17. Lynch TJ: Treatment of stage III non-small-cell lung cancer. ASCO Educational Book, Spring:265–275,1998.

18. Shepherd FA, Anglin G, Abratt R, et al: Influence of gemcitabine (GEM) and cisplatin (CP) schedule on response and survival in advanced non-small cell lung cancer (NSCLC). Proc Am Soc Clin Oncol 17:472a,1998 (abstr 1816).

19. Abratt RP, Sandler A, Crinò L: Combined cisplatin and gemcitabine for non-small cell lung cancer: Influence of scheduling on toxicity and drug delivery. Semin Oncol 25:35–43,1998.

20. Raach M, Fandara DR, Yuo M-S, et al: Radiation pneumonitis following combined modality therapy for lung cancer: Analysis of prognostic factors. J Clin Oncol 13:2606–2612,1995.

21. Goor C, Scalliet P, Van Meerbeeck J, et al: A phase II study combining gemcitabine and radiotherapy in stage III NSCLC. Ann Oncol 7:S101,1996 (suppl 5) (abstr).

22. Vokes EE, Leopols KA, Herndon JE, et al: A randomized phase II study of gemcitabine or paclitaxel or vinorelbine with cisplatin as induction chemotherapy and concomitant chemoradiotherapy for unresectable stage III non-small cell lung cancer (NSCLC) (CALGB study 9431). Proc Am Soc Clin Oncol 18:459a,1999 (abstr 1771).

23. Albain KS, Rusch VW, Crowley JJ, et al: Concurrent cisplatin/etoposide plus chest radiotherapy followed by surgery for stages IIIA (N2) and IIIB non-small-cell lung cancer: Mature results of Southwest Oncology Group phase II study 8805. J Clin Oncol 13:1880–1892,1995.

24. Albain KS: Induction chemotherapy with or without radiation followed by surgery in stage III non-small cell lung cancer: Update and perspectives. Oncology 11:S51–S57,1997 (suppl 9).

Page 61: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Chapter 5

QUALITY ASSURANCE OF THORACIC RADIOTHERAPY IN

EORTC 08941: A RANDOMISED TRIAL OF SURGERY VERSUS

THORACIC RADIOTHERAPY IN PATIENTS WITH STAGE IIIA

NON-SMALL-CELL LUNG CANCER (NSCLC) AFTER

RESPONSE TO INDUCTION CHEMOTHERAPY.

Gijsbert W.P.M. Kramer M.D. a*, Catherine L. Legrand Sc.D. b, Paul van Schil M.D., Ph.D.c, Lon Uitterhoeve M.D.d, Egbert F. Smit M.D., Ph.D. e, Franz Schramel M.D. f, Bonne Biesma M.D. PhD g, Vivianne Tjan-Heijnen M.D.PhD h, Nico van Zandwijk M.D. PhD i, Ted Splinter M.D. Ph.D.j, Giuseppe Giaccone M.D. PhD k, Jan P. van

Meerbeeck M.D., Ph.D.l, on behalf of EORTC-Lung Cancer Group.

a Radiotherapeutic Institute ARTI, Arnhem, The Netherlands; b EORTC (European Organization for Research and Treatment of Cancer) Data Centre, Brussels, Belgium;

c Department of Thoracic and Vascular Surgery University Hospital of Antwerp, Edegem, Belgium; d Department of Radiotherapy Academic Medical Centre,

Amsterdam, The Netherlands; e Department of Pulmonology Vrije Universiteit Medical Centre, Amsterdam, The Netherlands; f St. Antonius hospital, Nieuwegein, The Netherlands; g Jeroen Bosch Ziekenhuis ‘s-Hertogenbosch, The Netherlands; h

Department of Medical Oncology University Medical Centre Nijmegen-St. Radboud, The Netherlands; k Department of Pulmonology NKI Amsterdam, the Netherlands; j

Erasmus University Rotterdam, The Netherlands; k Department of Medical Oncology Vrije Universiteit Medical Centre, Amsterdam, The Netherlands, l Department of

Respiratory Diseases, University Hospital, Ghent, Belgium.

Eur J of Cancer 2006; 42:1391-1398.

Page 62: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

60

Summary

Purpose: To investigate the improvement of quality of radiotherapy and compliance to the protocol amendment of EORTC study 08941. Patients and Methods: The radiotherapy-specific data were analyzed of 154 patients with stage IIIA-N2 Non-Small-Cell Lung Cancer who were actually irradiated after response to 3 cycles of platinum-based induction chemotherapy. The parameters of quality assessed in 93 patients before and in 61 after protocol amendment included: time interval between last chemotherapy course and start of thoracic radiotherapy, the use of a 3-D planning CT, dose and fractionation scheme to the primary tumour, the involved – and uninvolved mediastinum, duration of radiotherapy and toxicity. Results: A significant improvement of all quality parameters was noted, except for the overall treatment time, which decreased slightly. Conclusions: Protocol amendment resulted in an improvement of quality and compliance of parameters, at the cost of some increase in overall treatment time. The latter reflects logistic problems rather than poor compliance. Keywords: Non-Small Cell Lung Cancer, Quality Assurance, Radiotherapy

Page 63: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

61

Introduction Three meta-analyses show that the addition of cisplatin containing combination induction chemotherapy followed by radiotherapy results in an absolute survival gain of 4% at 2 years in stage III non-small-cell lung cancer.1-3

In 1994 the European Organisation for Research and Treatment of Cancer (EORTC) Lung Cancer Group launched a randomised trial (08941) in patients with irresectable stage IIIA N2 NSCLC to address the question which locoregional treatment was superior following a response to systemic induction with platinum-based combination chemotherapy: surgical resection or thoracic radiotherapy.4 Quality assurance is an important issue in the EORTC.5 In 1987 the EORTC Radiotherapy Group introduced a series of procedures to document systematic errors, dummy runs, made in single institutions. This dummy run gives also information about accuracy of the design and the application of phase III study protocols. In 1991 Koning 6 recommended immediate monitoring of treatment parameters and uniform assessment of patient data in clinical trials. However, at the start in 1994 of the present randomised study these quality controls on radiotherapy treatment were not yet implemented. Questions about the quality of radiotherapy data came up halfway the trial, as not all investigators adhered to prescribed treatment schemes. At site visits, the radiotherapy treatment fields and set up appeared to be correct, but compliance to the protocol was subject of improvement. In May 1999, the protocol was amended with detailing and tighthening of the radiotherapy prescriptions. The aim was to improve the quality of and the compliance to the radiotherapy and to enhance the reliability of the final results of the study. This report focuses on the radiotherapy quality data before and after this protocol amendment. Patients and methods In EORTC 08941, patients with proven stage IIIA-N2 NSCLC were treated with 3 cycles of platinum-based induction chemotherapy; responding patients were then randomised between thoracic radiotherapy or surgical resection.7 The EORTC Protocol Review Committee (PRC) and the local ethical committees approved the protocol and its amendment. All patients had to give informed consent prior to randomisation according to local practice. In the surgery arm, 165 patients were included and 40% was referred for post-operative radiotherapy because of presumed irradical resection according to the preset definition. These postoperative radiotherapy data are not the subject of this paper. Protocol Amendment for improving Radiotherapy Quality In the protocol amendment of May 1999, most of the guidelines for writing protocols published in 1995 by the EORTC Radiotherapy Group8 were implemented. The protocol was revised with the target volumes redefined according the specifications of the ICRU 50 report.9 The radiotherapy prescriptions were tightened and updated as follows: planning-CT scan became standard for the whole treatment, timeframes and

Page 64: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

62

dose specifications were formulated more precisely, case report forms revised, and radiotherapists instructed (Table 1).

Table 1: Protocol prescriptions before and after the amendment

Before the amendment After the amendment

1. CT-based treatment planning mandatory for boost only

1. 3-D planning CT mandatory from start of treatment

2. No time intervals stated for ICT-Rand and Rand-RT

2. Time intervals prescribed (see Table 3)

3. Involved fields: 60 – 62.5 Gy in 30 fractions

3 Involved fields: 60 Gy in 30 fractions 5x per week

4. Uninvolved regions: 40 – 45 Gy in 20 – 23 fractions

4 Uninvolved regions 40 – 46 Gy in 20 – 23 fractions 5x per week

5. Fraction size at least 1.80 Gy 5 Fraction size 2.00 Gy

6. Tumor margins ≥ 2 cm for involved areas, the margins of the uninvolved areas were not specified

6 GTV 1 (prechemo involved areas) with ≥ 1 cm margin; GTV 2 (uninvolved areas) without margins

7. Implementation of ICRU 50 guidelines

Abbreviations: ICT = induction chemotherapy; Rand = randomisation; RT = thoracic radiotherapy; GTV = gross tumour volume. The primary protocol requirements were specified as follows: The primary tumour and areas of radiological nodal involvement had to be treated with at least 2 cm margin to a dose of 60-62.5Gy using daily fractions of 2 Gy. The lymphatic drainage areas of the uninvolved mediastinum on CT scan of the thorax should have received a dose of 40-45 Gy in daily fractions of 2 Gy. The total radiation dose should have been 60 Gy with lung tissue correction (0.3 or CT-based), given in 30 daily fractions of 2 Gy for 5 treatments per week. The fractionation dose should have been calculated for the target volume and should not have been less than 1.8 Gy. The overall radiotherapy treatment time should not have exceeded 7 weeks. Megavoltage equipment with photon energies of at least 4 MV was required. For fixed Source Skin Distance (SSD) techniques a treatment distance of 100 cm had to be used. In addition to dose distribution in the central axis plane, central dose at upper and lower limits of target volume should have been calculated and wedged filters used as compensators if greater than 10% inhomogeneity resulted from varying tissue thickness. CT –based treatment planning was mandatory for the boost technique only. Treatment could be started with conventional APPA opposed treatment fields. Simulator films and treatment plans with specified target volume and dose distributions had to be available for review. In the amendment, volumes of interest and organs at risk were more precisely described according ICRU 50 specifications.9 CT-based 3D- planning with lung tissue correction became mandatory for the whole radiation treatment. Dose inhomogeneity was accepted between +/- 10 %. Dose prescriptions were described more precisely and alternative treatment schemes were no longer allowed. Minor deviations from the prescribed dose, were defined as tumour dose between 55 – 66 Gy, dose to the involved mediastinum between 55 – 66 Gy, and fraction dose

Page 65: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

63

between 1.8 - 2.20 Gy. Major protocol deviations were defined as a fraction size of >2.20 Gy and/or fraction number <28. Data collection on delivered radiotherapy The data on the quality of the delivered radiotherapy were retrospectively collected for the period before the introduction of the amendment and prospectively for the period after the amendment. Site visits to participating institutes were performed in 1998 and 1999 by 2 experienced radiation-oncologists of the EORTC Lung Cancer Group. Data checked on site were: field sizes, margins, treatment techniques, and dose delivery. Toxicity was scored using the National Cancer Institute of Canada Clinical Trials Group Expanded Common Toxicity Criteria.10 Acute toxicity was scored for white blood cell counts, platelets, upper gastro-intestinal, pharynx and oesophagus, and lung toxicity. Only grade 3 – 4 toxicities will be discussed in this analysis. The case report forms were revised according the new prescriptions. All investigators, and specifically the radiotherapists, were instructed of the changes in protocol guidelines initiated in May 1999. Further, intensification of the data control at the EORTC Data Centre was performed. Endpoints and statistics To study the impact of this amendment, we defined a measure of compliance based on 1. interval scored from the last day of induction chemotherapy (ICT) till the start of thoracic radiotherapy (≤10 weeks), 2. the use of 3-D planning, 3. the use of lung tissue corrections, 4. the dosages administered to the primary tumour (60 – 62.5 Gy), the involved – (60 – 62.5 Gy) and uninvolved (40 – 46 Gy) mediastinum, 5. the fractionation size (1.95 – 2.05) and numbers (30 – 32), and 6. the duration of the radiotherapy treatment (40 – 46 days). We also report data on the interval between last day of ICT and randomisation, and from randomisation until start of thoracic radiotherapy, as well as toxicity data. Indeed, a reduction in toxicity rate was expected as standard dose prescriptions had to be followed and hypofractionated treatment schemes were not any longer allowed. Descriptive statistics are presented for these variables. All analyses are to be considered as exploratory and unplanned in the protocol. Results Patient inclusion EORTC study 08941accrued 578 patients between December 1994 and December 2002; of these 332 were randomised.7 Among the 165 patients randomised to the radiotherapy arm, 154 patients started their allocated protocol treatment and are the subject of this analysis. Of these, 93 patients were randomised before and 61 after May 1999, period at which the protocol amendment was implemented. A summary of the characteristics of the patients allocated to and actually treated with radiotherapy is given in table 2. Distribution of gender, T status and histological subtypes are similar between both randomisation periods.

Page 66: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

64

Analysis of portal films and planning data during the site visits revealed no major deviations in target volumes and margins from protocol prescriptions.

Table 2: Baseline characteristics at registration of patients

randomised to the radiotherapy arm

Randomisation period

Before amendment (N = 93)

After amendment ( N = 61)

Variable N (%) N (%) Sex

Male Female

71 22

(76) (24)

47 14

(77) (23)

cT T1 T2 T3 Tx

10 68 14 1

(11) (73) (15) (1)

10 42 9 0

(16) (69) (15) (0)

Histological subtype Squamous cell Non-squamous cell

38 55

(41) (59)

25 36

(41) (59)

Randomised in The Netherlands Yes No

66 27

(71) (29)

53 8

(87) (13)

Compliance to radiotherapy protocol prescription Table 3 shows data on compliance to protocol prescriptions for both periods. Response evaluation after the last chemotherapy course was done within the required time period of 4 weeks or less in 53% of patients before and 72% after the amendment respectively. The number of patients starting thoracic radiotherapy within the required 10 weeks or less after the last induction chemotherapy course, however, decreased from 91% before to 79% after the amendment. The median interval increased from 50 (range 17 – 113) to 53 (range 20-84) days. There is a trend towards longer time interval after randomisation till thoracic irradiation for patients in The Netherlands than for those patients treated outside The Netherlands (Table 3). Before protocol amendment 11% respectively 4% of the patients was not treated within 6 weeks after randomisation. After protocol amendment this number increased to 29% respectively 12% of the patients.

Page 67: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

65

Abbreviations: ICT = induction chemotherapy; Rand = randomisation; RT = thoracic radiotherapy * includes all items except interval ICT-Rand and Rand-RT Planning-CT was in the initial protocol mandatory for the boost dose only, and after protocol amendment mandatory from start of the treatment. All patients were treated with various photon beams of 6-18 MV. The compliance for dose delivery to the primary tumour, the involved and the uninvolved mediastinum, improved from 75 to 97%, 66 to 89% and 86 to 89% respectively. In general the compliance to the protocol improved from 48% (45/93 patients) to 66% (40/61 patients). For all 154 patients, 85 (55%) fulfilled all protocol prescriptions. Minor deviations from the prescribed dose, were observed in 140 patients (91%), 127 (83%), and 139 patients (90%) patients respectively. Major deviations were observed in 16 patients (10%), 14 before and 2 after protocol amendment (see Table 4). In 2/16 patients (12%) treatment scheme was changed because distant metastasis occurred during radiotherapy treatment. Toxicity of radiotherapy The Biological Effective Dose (BED) for tumour and late toxicity is given (Table 4) for the aberrant treatment schemes.

Table 3: Overview of compliance to protocol prescription

Before amendment After amendment N=93 N=61

Protocol prescription Number (%) pts

Interval

ICT-RT ≤10 wks ICT-Rand ≤ 4 wks Rand-RT ≤ 6 wks - in The Netherlands - outside The Netherlands

85 (91) 49 (53) 85 (91) 59 (89) 26 (96)

48 (79) 44 (72) 49 (80) 40 (71) 7 (88)

Planning-CT

Performed With lung correction

82 (88) 80 (86)

59 (97) 58 (95)

Dose (Gy):

Tumour (60 – 62.5) Mediastinum - Involved (60 – 62.5) - Uninvolved (40 – 46)

70 (75)

61 (66) 80 (86)

58 (97)

54 (89) 54 (89)

Fraction- numbers

30 – 32

72 (77)

59 (97)

Fraction size (Gy)

1.95 – 2.05 75 (81) 59 (97)

Duration 40 – 46 (days) 67 (72) 56 (92)

Compliance to protocol* 45 (48) 40 (66)

Page 68: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

66

Table 4: Overview of major protocol deviations used in individual patients:

before protocol amendment

Duration (days)

Dose primary (Gy)

Fraction numbers

Fraction dose BED tumour

BED for late toxicity

23* 35.0 16 2.18 * *

40 56.0 24 2.33 69.0 121

34 55 22 2.50 62.5 124

43 48.0 18 2.66 60.8 112

22 48.0 16 3.00 62.4 120

22 45.0 15 3.00 58.5 112

22 45.0 15 3.00 58.5 112

22 45.0 15 3.00 52.2 112

21 45.0 15 3.00 58.5 112

22 45.0 15 3.00 58.5 112

22 45.0 15 3.00 58.5 112

22 45.0 15 3.00 58.5 112

15 40.0 8 5.00 72 200

16 40.0 8 5.00 72 200

After protocol amendment

31* 45.0 20 2.25 * *

39 56.0 23 2.43 69.6 124

Prescribed dose

40 60.0 30 2.00 72 120

Formula for reference treatment: BEDref = Dose ref [1 + fraction dose ref /(α/β)] α/β for tumour = 10, and for late toxicity α/β = 2 * Patients stopped because of distant metastasis Only 5 (5%) patients before the amendment and 2 (3%) patients after the protocol amendment had toxicity grade 3-4 in any of these variables (Table 5). Two patients with pre-existent lungfibrosis died because of a radiation pneumonitis after induction chemotherapy with gemcitabine/cisplatin.

Page 69: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

67

Table 5: Acute toxicity in thoracic radiotherapy after induction chemotherapy

Randomisation period

Variable Before protocol amendment (N = 93)

After protocol amendment (N = 61)

WBC (109/l) ≥ 4 2 - <4 <2 unknown/missing

69 8 0 16

(74) (9)

(17)

46 4 0 11

(75) (7)

(18)

Platelets (109/l) ≥ 100 50 - <100 <50 unknown/missing

39 1 1 52

(42) (1) (1) (56)

48 1 0 12

(79) (2) (0) (20)

Upper GI toxicity Degree 0 Degree 1-2 Degree 3-4

59 34 0

(63) (37)

42 19 0

(69) (31)

Pharynx and Oesophagus toxicity Degree 0 Degree 1-2 Degree 3-4

19 74 0

(20) (80)

15 45 1

(25) (74) (2)

Lung toxicity Degree 0 Degree 1-2 Degree 3-4 Missing

46 42 4 1

(50) (45) (4) (1)

26 34 1 0

(43) (56) (2)

Other acute toxicity No Yes

80 13

(86) (14)

49 12

(80) (20)

Abbreviations: WBC = white blood count Discussion In 1999, quality control of EORTC 08941 showed limited protocol compliance mainly due to the use of hypofractionated treatment schemes. That was the reason for a protocol amendment with more strict prescriptions on the radiotherapy delivery. We observed that due to this amendment overall treatment parameters as dose, fractionation, and overall treatment time improved substantially. The stricter regulations in the amendment resulted in a gain in overall compliance to the protocol of 17%.

Page 70: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

68

Unexpectedly, we also observed that despite the more strict protocol prescription, the proportion of patients starting with radiotherapy within 10 weeks of the last cycle of chemotherapy actually declined from 91% to 79%. This was mainly due to an increased delay between randomisation and start of radiotherapy and not due to a further delay between chemotherapy and randomisation. Apparently, chest physicians and medical oncologists became now also aware of the importance to improve their time schedule. The use of a planning-CT, which became mandatory for the whole treatment course, was probably the main reason the number of patients started with thoracic radiotherapy within 10 weeks after finishing chemotherapy decreased. Planning-CT is a more time-consuming technique than conventional simulation.11 Furthermore, the shortage of radiation equipment12 could be responsible for lengthening of the time interval, as a large majority (77%) of the patients in this study were treated in the Netherlands (Table 2 and 3). However, we consider the gain in quality control with the introduction of a planning-CT for the whole treatment to be more important on the main outcomes of this trial than the increase of 3 days of the median time before and after the amendment. The Planning Target Volume (PTV) prescribed by the protocol amendment, had a margin of ≥1 cm around the pre-chemotherapy tumour areas and pathological hilar or mediastinal nodes (Gross Tumour Volume or GTV). All patients in this study had at least a response at randomisation. Shih et al13 showed for fast CT-scan at shallow free breathing that internal margins (expansion margins) of 13 mm are required to approximate the composite GTV in 95% of cases. Van Sörnsen de Koste et al14 showed that margins of 15 mm around GTV in immobile tumours using conventional planning-CT are adequate, but in mobile tumours the coverage is still inadequate in 11% of the cases. Lagerwaard et al15 recently showed in reconstructing pre-chemotherapy target volumes clinicians may fail to treat the actual pre-chemotherapy tumour volume in 26% of the cases. This can result in lower local control, but no data exist of randomised trials in treating pre- or post-chemotherapy target volumes. Therefore, we consider the margin of ≥1 cm around prechemotherapy tumour areas and mediastinal nodes used in the present protocol as adequate. The majority of patients were planned using a 3-D planning-CT during this study, reaching 99% after protocol amendment. Thus, based on the abovementioned findings and the pre-amendment site visits, we believe that no major concerns exist about the quality of and the compliance to treatment planning volumes. Before protocol amendment, a major protocol violation in the administered tumour dose was present in 15% of patients with doses to primary tumour of less than 55 Gy or in excess of 66 Gy. These findings were worse than found in an earlier study wherein incorrectly given radiation doses were reported in only 7% of the cases.6 Although the protocol already recommended fraction doses of 1.8 - 2.0 Gy at initiation of the study, capacity problems in several hospitals led to the introduction of hypofractionated treatment schemes. After amendment of the protocol, it was stressed that dose prescriptions had to be strictly followed. This resulted in a substantial improvement of the protocol compliance. A reduction in overall treatment time to 20-24 days using hypofractionated schemes on loco-regional control is evident. Lester16 treated stage I-III NSCLC patients to 50-55 Gy in 15-20 fractions over 3-4 weeks. This resulted in a favourable outcome compared to standard radical radiotherapy data. Cheung17 used 48 Gy in 12 once-daily fractions for early stage non-small-cell lung cancer patients with a recurrence free survival after 2 years of 40%. The CHART study18 showed a reduction of overall treatment time to 12 consecutive days with a hyperfractionated treatment scheme of

Page 71: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

69

thirty-six fractions of 1.5 Gy given three times per day, resulting in a gain in two-year survival of 9%, compared to 30 fractions of 2 Gy to a total dose of 60 Gy in 6 weeks. On the other hand, hypofractionation can increase the risk of late toxicity. Abratt et al19 showed that the biological effect of radiation on tumours is increased as overall treatment time is shortened, but this is not true for late-reacting normal tissue. However, for patients with stage III tumours treated with combined modality more conservative hypofractionation regimens are warranted. The BED for late toxicity in only 2 hypofractionation schemes (BED 200) is much higher than in the standard treatment scheme (BED 120).20 Therefore, no differences in toxicity over both periods were to be expected. The two patients, who died after induction chemotherapy followed by radiotherapy, were both treated according to the initial protocol prescriptions, but suffered from interstitial lung fibrosis.21 Maas et al22 showed the combination of gemcitabine/cisplatin as part of combined modality treatment, especially radiotherapy, resulted in an decline of pulmonary function and could be responsable for an increase of toxicity. However, van Zandwijk et al21 showed the combination of gemcitabine/cisplatin is safe as induction treatment. Therefore, patients with interstitial lung fibrosis were hence excluded from further inclusion in 08941. This trial was running over a period of more than 8 years. During this period, radiotherapy techniques changed to the actual standards. The progress in simulation techniques urged for standard 3-D planning techniques. The improvement of protocol compliance after the amendment supports the earlier recommendation of immediate monitoring of treatment parameters and uniform assessment of patient data in clinical trials. For future trials we would recommend to use routine checks on the quality of (any) delivered treatment as this would provide the possibility of corrections in the protocol shortly after start of a study and guarantee compliance. Conclusion After protocol amendment, a substantial improvement of protocol compliance was observed, despite some increase in overall treatment time. The latter is thought to reflect logistic problems rather than poor compliance. The use of hypofractionated treatment schemes is of some concern with respect to the outcome of the trial. Our data support the use of strict radiotherapy specifications, dummy runs, and quality control from the start of a large study and frequent quality assessments during it. References 1. NSCLC Collaborative Group. Chemotherapy in non-small-cell lung cancer: a

meta-analysis using updated data on individual patients from 52 randomised clinical trials. BMJ 1995;311:899–909.

2. Marino P, Preatoni A, Cantoni A. Randomised trials of radiotherapy alone versus combined chemotherapy and radiotherapy in stage IIIa and IIIb non-small-cell lung cancer. A meta-analysis. Cancer 1995;76:593–601.

3. Pritchard RS, Anthony SP. Chemotherapy plus radiotherapy compared with radiotherapy alone in the treatment of locally advanced, unresectable, non-small-cell lung cancer. A meta-analysis. Ann Intern Med 1996;125:23–29.

Page 72: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

70

4. Splinter TAW, van Schil P, Kramer GWPM et al.Randomized Trial of Surgery Versus Radiotherapy in Patients with Stage IIIA (N2) Non–Small-Cell Lung Cancer After a Response to Induction Chemotherapy. EORTC 08941. Clin Lung Cancer 2000;2:69–72; discussion 73.

5. Kouloulias VE, Poortmans PM, Bernier J, et al. The Quality Assurance Programme of the Radiotherapy Group of the European Organization for Research and Treatment of Cancer (EORTC): a critical appraisal of 20 years of continuous efforts. Eur. J. of Cancer 2003;39:430–437.

6. Schaake-Koning C, Kirkpatrick A, Kroger R, van Zandwijk N, Bartelink H. The need for immediate monitoring of treatment parameters and uniform assessment of patient data in clinical trials. A quality control study of the EORTC Radiotherapy and Lung Cancer Cooperative Groups. Eur J Cancer 1991;27:615–619.

7. van Meerbeeck JP, Kramer GWP, Van Schil PE et al. A randomised trial of radical surgery (S) versus thoracic radiotherapy (TRT) in patients (pts) with stage IIIA-N2 non-small-cell lung cancer (NSCLC) after response to induction chemotherapy (ICT) EORTC 08941. J Clin Oncol 2005;23(16S):1095S,#7015.

8. Bolla M, Bartelink H, Garavaglia G et al. EORTC guidelines for writing protocols for clinical trials of radiotherapy. Radiother Oncol 1995;36:1–8.

9. ICRU Report 50 (1993): Prescribing, Recording and Reporting Photon Beam Therapy. ISBN 0-913394-48-3.

10. Williams J, Chen Y, Rubin P et al. The biological basis of a comprehensive grading system for the adverse effects of cancer treatment. Semin Radiat Oncol 2003;13:182–188.

11. Perez CA, Purdy JA, Harms W et al. Three-dimensional treatment planning and conformal radiation therapy: preliminary evaluation. Radiother Oncol 1995;36:34–43.

12. Slotman BJ, Leer JW. Infrastructure of radiotherapy in the Netherlands: evaluation of prognoses and introduction of a new model for determining the needs. Radiother Oncol. 2003;66:345–349.

13. Shih HA, Jiang SB, Aljarrah KM et al. Internal target volume determined with expansion margins beyond composite gross tumour volume in three-dimensial conformal radiotherapy for lung cancer. Int J Radiat Oncol Biol Phys 2004;60:613–622.

14. van Sörnsen de Koste JR, Lagerwaard FJ, Schuchhard-Schipper RH et al. Dosimetric consequences of tumour mobility in radiotherapy of stage I non-small cell lung cancer – an analysis of data generated using ‘slow’CT scans. Radiother Oncol. 2001;61:93–99.

15. Lagerwaard FJ, van de Vaart PJ, Voet PW et al. Can errors in reconstructing pre-chemotherapy target volumes contribute to the inferiority of sequential chemoradiation in stage III non-small cell lung cancer (NSCLC)? Lung Cancer 2002;38:297–301.

16. Lester JF, Macbeth FR, Brewster AE et al. CT-planned accelerated hypofractionated radiotherapy in the radical treatment of non-small-cell lung cancer. Lung Cancer 2004:45;237–242.

17. Cheung PC, Yeung LT, Basrur V et al. Accelerated hypofractionation for early-stage non-small-cell cancer. Int J Radiat Oncol Biol Phys. 2003:54;1014–23.

18. Saunders M, Dische S, Barrett A et al. Continuous hyperfractionated accelerated radiotherapy (CHART) versus conventional radiotherapy in non-small-cell lung

Page 73: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

71

cancer: a randomised multicentre trial. CHART Steering Committee. Lancet 1997:350;161–165.

19. Abratt RP, Bogart JA, Hunter A. Hypofractionated irradiation for non-small cell lung cancer. Lung Cancer 2002;36:225–233.

20. Joiner M.C., van der Kogel A.J. The linear-quadratic approach to fractionation and calculation of isoeffect relationships. In: Basic Clinical Radiobiology, 2nd Edition (Editor G.G. Steel), Edward Arnold, Kent, UK. Chapter 13, pp. 106–122 (1977).

21. Van Zandwijk N, Smit E.F., Kramer G.W.P. et al. Gemcitabine and Cisplatin as Induction Regimen for Patients with Biopsy-Proven Stage IIIA N2 Non-Small-Cell Lung Cancer: A Phase II Study of the European Organization for Research and Treatment of Cancer Lung Cancer Cooperative Group (EORTC 08955). J Clin Oncol 2000;18:2658–64.

22. Maas KW, van der Lee I, Bolt K et al. Lung function changes and pulmonary complications in patients with stage III non-small cell lung cancer treated with gemcitabine/cisplatin as part of combined modality treatment. Lung Cancer 2003;41:345–51.

Page 74: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

72

Acknowledgement This publication was supported by grants number 5U10CA11488-24 through 5U10CA11488-34 from the National Cancer Institute (Bethesda, Maryland, USA). Its contents are solely the responsibility of the authors and do not necessarily represent the official views of the National Cancer Institute. The authors thank the following physicians and institutes of the EORTC Lung Cancer Group who are as follows. The Netherlands: B-H. Liem (Lievensberg Ziekenhuis, Bergen Op Zoom); B. Biesma (Bosch Medicentrum-Groot Ziekengasthuis, ‘s-Hertogenbosch); P. van Valenberg (Elkerliek Ziekenhuis, Helmond); A. Termeer (Canisius-Wilhelmina Ziekenhuis, Nijmegen); T. van Boxem (Ziekenhuis Franciscus, Roosendaal); H. Goey (Maria Ziekenhuis, Tilburg); F. Schramel (St. Antonius Ziekenhuis/Diakonessenhuis, Nieuwegein-Utrecht); A. van Bochove (Ziekenhuis de Heel, Zaandam); P.I. van Spiegel (Slotervaart Ziekenhuis, Amsterdam); N. van Zandwijk (Antoni van Leeuwenhoekhuis, Amsterdam); J. van Meerbeeck /T.A.W. Splinter/S. Senan/ A. Maat (Erasmus M.C., Rotterdam); F. van Breukelen (Spaarne Ziekenhuis, Haarlem); V. Tjan-Heijnen (St. Radboud University Hospital, Nijmegen); N.J.J. Schlosser (Universitair Medisch Centrum, Utrecht); G. Giaccone (Academisch Ziekenhuis der Vrije Universiteit, Amsterdam); H.B. Kwa (Onze Lieve Vrouw Gasthuis, Amsterdam); L. Willems (Academisch Ziekenhuis Leiden, Leiden); B.M. Drenth (Atrium Medisch Centrum, Heerlen); J. Stigt (Sophia Ziekenhuis, Zwolle); E. Lammers (Leyenburg Ziekenhuis, Den Haag); G.W.P. Kramer/S. Gans (Arnhem’s Radiotherapeutisch Instituut/Ziekenhuis St. Jansdal, Arnhem). Belgium: P. Pinson (Universitair Ziekenhuis, Gent); D. van Renterghem (A.Z. St. Jan, Brugge); D. Galdermans (Algemeen Ziekenhuis Middelheim, Antwerpen); P. van Schil (Universitair Ziekenhuis, Antwerpen); L. Delaunois (Cliniques Universitaires de Mont Godinine, Yvoir); J. Vansteenkiste (U.Z. Gasthuisberg, Leuven); M. Beauduin (Hopital de Jolimont, Haine St. Paul); A. Baudoux (Clinique Sainte Elisabeth, Namur); W. Moorkens (AZ Hoge Beuken, Antwerpen); J. Verschuere (AZ Zusters van Barmhasrtigheid, Ronse); J-P. D’Odemont (Clinique Saint Joseph, Mons); W. Verbeke (Stedeljik Ziekenhuis, Aalst). Italy: S. Darwish (Policlinicl Monteluce, Perugia); A. Ardizzoni (Istittuto Nazionale Per la Ricerca Sul Cancro, Genova); G. Scagliotti (Ospedale S. Luigi Gonzaga–Universita di Torino, Torino); and C. Gridelli (Istittuto Nazionale Per lo Studio e la Cura Dei Tumori, Napoli). Poland: T. Lewinski (Oncology Centre Institute, Warsaw). Slovenia: A. Debeljak (Klinicni Center Ljubljana Golnik, Golnik). UK: M. Hatton (Weston Park Hospital, Sheffield); M. O’Brien (Royal Marsden Hospital, Sutton); A. Price (Western General Hospital, Edinburgh); and K. O’Byrne (Leicester Royal Infirmary, Leicester).

Page 75: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Chapter 6

RESULTS OF THE DUTCH NATIONAL STUDY OF THE

PALLIATIVE EFFECT OF IRRADIATION USING TWO

DIFFERENT TREATMENT SCHEMES FOR NON-SMALL-CELL

LUNG CANCER (NSCLC)

Gijsbert W.P.M. Kramer *, Stofferinus L. Wanders †, Ed M. Noordijk ‡,

Ernest J.A. Vonk §, Hans C. van Houwelingen║, Wilbert B. van den Hout ║, Ronald B. Geskus║, Mirjam Scholten*, Jan-Willem H. Leer*

*Department of Radiotherapy, RADIAN, location Arnhems Radiotherapeutic Institute (ARTI), Arnhem; †MAASTRO clinic Maastricht/Heerlen; ‡ Department of Clinical Oncology, Leiden University Medical Center (LUMC); §Radiotherapeutic Institute Stedendriehoek en Omstreken (RISO) Deventer, the Netherlands; ║Department of medical decision making, Leiden University Medical Center (LUMC), Leiden, the

Netherlands.

J Clin Oncol 2005;23:2962–2970

Page 76: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

74

Summary Purpose: A national multicenter randomized study compared the efficacy of 2 x 8 Gy versus our standard 10 x 3 Gy in patients with inoperable stage IIIA/B (with Eastern Cooperative Oncology Group score of 3 to 4 and/or substantial weight loss) and stage IV non-small-cell lung cancer. Patients and Methods: Between January 1999 and June 2002, 297 patients were eligible and randomized to receive either 10 x 3 Gy or 2 x 8 Gy by external-beam irradiation. The primary endpoint was a patient-assessed score of treatment effect on seven thoracic symptoms using an adapted Rotterdam Symptom Checklist. Study sample size was determined based upon an average total symptom score difference of more than one point over the initial 39 weeks post-treatment. The time course of symptom scores were also evaluated, and other secondary endpoints were toxicity and survival. Results: Both treatment arms were equally effective, as the average total symptom score over the initial 39 weeks did not differ. However, the pattern in time of these scores differed significantly (P <0.001). Palliation in the 10 x 3-Gy arm was more prolonged (until week 22) with less worsening symptoms than in 2 x 8-Gy. Survival in the 10 x 3-Gy arm was significantly (P = .03) better than in the 2 x 8-Gy arm with 1-year survival of 19.6% (95%CI, 14.1 to 27.3%) versus 10.9% (95%CI, 6.9 to 17.3%). Conclusion: The 10 x 3-Gy radiotherapy schedule is preferred over the 2 x 8-Gy schedule for palliative treatment, as it improves survival and results in a longer duration of the palliative response. Keywords: Randomized trial, non-small-cell lung cancer, irradiation, palliative treatment, hypofractionation

Page 77: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

75

Introduction The treatment of non-small-cell lung cancer (NSCLC) is still a challenge for the oncologist. In the past decade, combined-modality treatment in stage III disease and chemotherapy for stage IV disease improved the survival and quality of life in NSCLC patients.1 Unfortunately, many patients are unfit to undergo these intensive treatments.2 Patients who undergo palliative chemotherapy may eventually require treatment for locoregional complaints. Symptoms such as hemoptysis, chest pain, dysphagia, and dyspnea in NSCLC can all be effectively palliated using different radiation treatment schemes.3 The effectiveness of these palliative schemes varies for each symptom, but is most effective for hemoptysis and chest pain in patients with Eastern Cooperative Oncology Group (ECOG) score of 2 to 3.4 The optimal dose for palliation remains controversial.5 In the first Medical Research Council (MRC) trial reported in 1991, no differences in palliative effect or in survival were seen between 13 x 3 Gy and 2 x 8.5 Gy.6 In a second MRC study (1992)7 for poor prognostic patients only, no differences in palliation or survival were seen between 10 Gy single dose and 2 x 8.5 Gy. However in a study by Bezjak et al 8 a difference in survival between 20 Gy in five fractions and a 10-Gy single dose was demonstrated in favor of the multifractionation treatment. In a subgroup-analysis, however, this survival advantage was not seen in patients with an ECOG score ≥ 2. Later data suggested a survival advantage of 13 x 3 Gy over 2 x 8.5 Gy in patients with good performance status.9. For patients in a poor general condition it still has to be proven whether 2 x 8.5 Gy is equally effective as 10 x 3 Gy. Due to the contradictory literature, radiation oncologists in the Netherlands were unable to reach a consensus on the palliative schedule for patients with poor- prognosis NSCLC. The present study was designed for patients with stage IV NSCLC (ECOG 0 to 1) in whom chemotherapy options were exhausted. Patients with ECOG 2, stage IV disease were also eligible, as chemotherapy for this group of patients was not general practice at the start of this study.10 We focused on patients with poor general condition and/or significant weight loss with stage III or stage IV NSCLC, comparing 10 x 3 Gy with 2 x 8 Gy. Because of the results of the MRC studies mentioned here, we expected both treatment arms to have an equal outcome for palliation as for survival.6

Patients and Methods Patient population Patient characteristics are presented in table 1. The diagnosis NSCLC had to be cytologically or histologically confirmed. Tumor stage was IIIA to IV in combination with performance status 3 to 4 and/or a weight loss of more than 5% in 3 months or greater than 10% in 6 months before diagnosis. Patients with stage IV, ECOG stage 0 to 2 disease could also be included if, in the opinion of the treating pulmonologist, no further chemotherapeutic options were left after prior chemotherapy, or a substantial weight loss and/or comorbidity prevented the use of chemotherapy. Patients had to have a minimum patient-assessed total symptom score of 8, indicating at least some burden on at least one of the complaints caused by the tumor itself (Fig 1). Furthermore, the patient should be physically and mentally fit enough to participate in this study. Patients having superior vena cava syndrome (SVCS) at

Page 78: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

76

presentation, prior radiotherapy to the chest, and/or other malignant diseases in the past or concurrent chemotherapy were excluded. Study Endpoints and Data Collection Primary endpoint of this study were palliation of thoracic symptoms measured over 26 questionnaires (39 weeks) after random assignment. Secondary endpoints were toxicity and survival. Patients were also asked to provide information about quality of life and costs. These data will be published separately in a cost-utility analysis. Thoracic symptoms were measured using a total symptom score that measured the extent to which patients were bothered by seven tumor-related symptoms (Fig 1). Existing validated questionnaires were considered either too burdensome11 or not specific enough for our patient population.11,12 Based on symptoms previously observed in irradiated patients with NSCLC,13 we instead adapted the Rotterdam Symptom Checklist.11 We maintained the symptoms lack of appetite and dyspnea, and added chest pain, coughing, hemoptysis and hoarseness. In a different format, patients also reported their level of dysphagia.14 Using four-point Likert scales for each symptom, the total symptom score could range from 7 (not bothered at all on any symptom) to 28 (bothered very much on all symptoms). Palliation was defined as an average total score below the baseline score.

Page 79: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

77

Abbreviations: SD, standard deviation; ECOG, Eastern Cooperative Oncology Group.

Table 1: Baseline patient characteristics

10 x 3 Gy 2 x 8 Gy

No. of patients

%

No. of patients

%

No. of patients 148 149

Sex, No.

Male Female

118 30

119 30

Age, years

Median SD

68.9 8.7

69.3 10.5

Time since diagnosis, years

Median Range

0.08 0.01-7.31

0.08 0.00-8.28

Baseline symptom score

Median SD

12.71 2.42

12.88 2.52

Tumor stage, No. of patients

IIIA IIIB IV

16 56 76

24 59 66

Tumor type, No. of patients

Sqamous cell carcinoma Adenocarcinoma Large cell carcinoma Other

64 37 39 8

60 27 53 9

Diagnosis by, No. of patients

Histology Cytology

114 34

114 35

Treatment indication, No. of patients Primary lesion

Recurrent lesion (no prior irradiation)

142 6

147

2

Prior chemotherapy 18 20

Other treatment at start irradiation:

Pain medication Other medications

Antibiotics Not specified

Supplemental oxygen

80 54 50 5

49 9

98 67 60 5

58 8

Substantial weight loss (%) 104 70.2 109 73.1

ECOG score

0 1 2 3 4

9 45 60 33 1

6.1 30.5 40.6 22.3 0.1

5 48 44 51 1

3.4 32.2 29.5 34.2 0.1

Page 80: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

78

In addition, information about acute toxicity as nausea, vomiting, and radiation esophagitis-induced dysphagia was collected, using the National Cancer Institute of Canada Clinical Trials Group Expanded Common Toxicity Criteria.15

Figure 1. Thoracic symptom checklist (originally in Dutch). The data collection schedule for follow-up is presented in Table 2. All symptoms were entirely patient-based scored. A maximum of 33 questionnaires would be sent in case follow-up would be sent in case follow-up could be completed. On average, 17.4 sets (53%) per patient were received because a substantial number of patients did not survive long enough or stayed fit enough to fill in all the questionnaires over 52 weeks. Of the 4,369 questionnaires sent by the data manager to the individual patient, 4,306 were returned. The first questionnaire was completed prior to random assignment, the last questionnaire in the 52nd week after random assignment. Follow-up after 52 weeks was continued by the data manager, who made three-monthly inquiries to establish the survival.

To what extent during the past week have you been bothered by complaints that may arise from your disease? Please put a cross by the answer that is most applicable to you. Example: Have you been bothered, during the past week, by: Not at all A little Quite a bit Very much

Fatigue � � � �

Have you been bothered, during the past week, by:

Not at all A little Quite a bit Very much

Lack of appetite

Shortness of breath

Pain in the chest

Coughing

Coughing up blood

Hoarseness

� � � � � �

� � � � � �

� � � � � �

� � � � � �

Problems with swallowing: food won't go down

� able to eat solid food; food goes down easily

� only able to eat semi-solid food; food goes down with plenty of liquid

� only able to eat liquid food

� not able to eat liquid food

Page 81: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

79

* Only once 2 months after the start of the treatment. † Only together with questionnaires 2 to 9. ‡ Only together with questionnaires 2, 4, 6, 8, 10 and 12. Statistical analysis The study was designed as an equivalence study. We assumed as null hypothesis that the response score of the 2 x 8 Gy to be at least 10% lower compared to our standard 10 x 3 Gy. We expected a response rate of 65% with the standard treatment. We compared the average total symptom score for all present symptoms during 39 weeks. With this design, 134 patients per treatment arm were needed to obtain, at a 5% significance level, a power of .90 of correctly concluding equivalent effectiveness. Expecting an ineligibility and withdrawal of patients of 10% a total number of 300 patients were needed. A response was defined as a reduction in the total symptom score by more than one point compared to the baseline. Progression was defined as an increase of the total symptom score by more than one point, if re-treatment was given, or in case the patient did not return the questionnaire. A difference in palliative effect between the two schedules was defined as a difference of at least one point in the average total symptom score for at least one of the symptoms. Data are presented over the initial 39 weeks, as too few patients remained alive to allow for meaningful analysis.

Table 2: overview of data collection

Measurements Before Randomization

Follow-up measurements After Randomization

Doctor

Data Manager

Patient

2 Months After Start and Every 2 months

Thereafter for a Maximum of 12

months (data manager)

Weekly and After 3 Months every 2 Weeks for a maximum of 52

weeks (patient)

Date of randomization X Performance status X Weight loss X TNM - classification X X Histology X Medication X X

Follow-Up assessment after randomization

Quality of Life X X

Tumor related thoracic symptom score

X X X

Toxicity X†

Radiotherapy delivered according to protocol

X *

Date of histology X Retreatment with

radiotherapy X

Patients follow up/ survival

X

Costs X‡

Page 82: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

80

A secondary analysis was performed in which average total symptom score was corrected for the initial total symptom score at the start of the treatment and time of follow-up during this period. The latter was performed as initially higher total symptom score could result in a higher chance of a declining total symptom score and would suggest a better outcome. Also, differences in the course of the total symptom score between each treatment arm over the first 39 weeks were analyzed using the random effects model using the natural splines function in S-PLUS 6.2 professional edition.16

A secondary analysis was performed in which average total symptom score was corrected for the initial total symptom score at the start of the treatment and time of follow-up during this period. The latter was performed as initially higher total symptom score could result in a higher chance of a declining total symptom score and would suggest a better outcome. Also, differences in the course of the total symptom score between each treatment arm over the first 39 weeks were analyzed using the random effects model using the natural splines function in S-PLUS 6.2 professional edition.16

The overall survival of both treatments was described using the Kaplan-Meier survival curves.17 The log-rank test was used to compare overall survival between both treatments. Fisher’s exact test was used to compare the rates of toxicity between both groups. All reported P values are two-sided, and significance level was set at .05. All data were evaluated according to the intention-to-treat principle. Radiation treatment Patients were randomized, without stratification, either to the multiple fractionation scheme of 10 x 3 Gy, four to five times per week, or the hypofractionated scheme of 2 x 8 Gy, given on day 1 and day 8. Irradiation was given using two opposing anterior-posterior fields with 6- to 18-MV photon beams. The treatment portals encompassed the tumor, with a margin of 1 ½ - 2 cm including adjacent pathological lymph nodes. No limitations were set for the target volume. Dose calculation was not corrected for tissue inhomogeneities. Comedication, including corticosteroids and analgetics, and supplemental oxygen were allowed and documented. Ethical issues The study fulfilled all criteria as prescribed by the Dutch Law on Medical Research. Approval of the protocol by the Central Medical Committee of Ethics was obtained and each participating radiotherapy center had also a separate approval by their Local Medical Committee of Ethics. Random assignment took place after receiving informed consent of the patient. Results From the January 1, 1999, to May 31, 2002, 303 patients from 13 radiotherapy centers were randomized. Six patients were not eligible because of stage III disease with WHO performance scores 0 to 1 and no weight loss. Of the 297 patients who were

Page 83: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

81

eligible, 148 patients received 10 x 3 Gy and 149 patients received 2 x 8 Gy. All patients’ survival and toxicity data were available and analyzed. Baseline patient characteristics were well balanced between the two groups (Table 1). No major differences were seen between the two treatment groups for age, histology, stage, and prior chemotherapy. Requirements for comedications and supplemental oxygen were well balanced between the 2 treatment arms. Most patients had a poor general condition, as 213 patients (71.7%) had substantial weight loss at randomization. Also, 190 patients (63.9%) presented with an ECOG score of 2 or more. Only a slight, but statistically not significant (P = .145), imbalance in ECOG 2 and 3 in favor of the 10 x 3-Gy arm was recorded.

Table 3: Overview of the Patient-Assessed Symptom Score at Start per Treatment Arm

Symptom Score Patients with

Symptom Score >1 1. not at all 2. a little 3. quite a bit 4. very much

Symptom/ Treatment arm

N % N % N % N % N %

Lack of appetite 181 61 10 x 3 Gy 61 41.2 43 29.1 30 20.3 14 9.5 2 x 8 Gy 55 36.9 40 26.8 49 32.9 5 3.4

Dyspnea 259 87 10 x 3 Gy 21 14.2 43 29.1 58 39.1 26 17.6 2 x 8 Gy 17 11.4 51 34.2 60 40.2 21 14.1

Chest Pain 142 48 10 x 3 Gy 83 56.1 31 21.0 25 16.9 9 6.1 2 x 8 Gy 72 48.3 42 28.2 24 16.1 11 7.4

Cough 249 84 10 x 3 Gy 24 16.2 74 50.0 46 31.1 4 2.7 2 x 8 Gy 24 16.1 72 48.3 48 32.2 5 3.4

Hemoptysis 114 38 10 x 3 Gy 92 62.1 39 26.4 13 8.8 4 2.7 2 x 8 Gy 91 61.1 39 26.2 16 10.7 3 2.0

Hoarseness 93 31 10 x 3 Gy 103 69.6 21 14.2 14 9.5 10 6.8 2 x 8 Gy 101 67.8 21 14.1 17 11.4 10 6.8

Dysphagea 29 10 10 x 3 Gy 132 89.2 14 9.5 2 1.4 2 x 8 Gy 136 91.3 6 4.0 6 4.0 1 0.7

Page 84: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

82

Most patients suffered from dyspnea (87%) and cough (84%), followed by lack of appetite (61%), chest pain (48%), hemoptysis (38%), hoarseness (31%), and dysphagia (10%). Distribution and intensity of symptoms were well balanced across both treatments groups (Table 3). The difference in the start of the treatment after random assignment between 10 x 3 Gy and 2 x 8 Gy was less than 2 days. After random assignment, 35 patients did not receive the prescribed treatment scheme. In the 2 x 8-Gy arm, 14 patients did not finish their treatment. In the 10 x 3-Gy arm 18 patients received less than 10 fractions of 3.0 Gy, one patient had one additional fraction of 3.0 Gy, and two patients received 16 Gy in two fractions.

Figure 2: Changes in Total Symptom Score (with confidence interval) over time. Solid

line: 10 x 3 Gy. Dotted line: 2 x 8 Gy. The overall results show that both treatments were equally effective, as they differ less than one point in average total symptom score. Symptom scores for both arms did not differ at start of the treatment, being 12.71 (95% CI, 10.28 to 15.14) and 12.87 (95% CI, 10.35 to 15.39) for the 10 x 3-Gy and 2 x 8-Gy arms respectively (Fig 2). The average total symptom score over the initial 39 weeks was in the 10 x 3-Gy arm 12.71 (95% CI, 12.28 to 13.12) and 13.07 (95% CI, 12.66 to 13.49) in the 2 x 8-Gy arm, with a difference of 0.37 (95% CI, -0.22 to 0.96; P = .22). Corrected for initial total symptom score and for time of follow up, the difference remained less than 1 point, with respectively 0.26 (95% CI, –0.23 to 0.75; P = .299) and 0.24 (95% CI, –0.30 to 0.78; P = .388). Analysis of each symptom showed some tendencies in response patterns during treatment (Fig 3). For cough (P = .84) and dyspnea (P = .84) no significant differences were seen. For the other symptoms, numbers were too small to compare these differences for each separate symptom and particularly for the changes in intensity per symptom between both treatment arms. All symptoms, except hemoptysis, initially intensified as the number of the patients suffering from that

Page 85: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

83

Figure 3: Patterns of symptom response during treatment per treatment arm. Symptom

score measures the experienced level of burden: white, not at all; gray, a little; dark gray, quite a bit; black = very much.

particular symptom rose and/or the severity of the symptom increased as a result of acute toxicity. The incidence of dysphagia initially rose to 30 – 40% of the patients, and was finally hardly palliated. All other symptoms were eventually palliated in comparison to the pretreatment situation. An overview of the evolution of total symptom score pattern for both treatment arms over the initial 39 weeks is presented in Figure 2. In both arms, total symptom scores initially rose as a result of treatment-related side effects. This rise seems to be earlier and more intense in the 2 x 8-Gy arm reaching significance in week 1 to 2. Palliation

Page 86: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

84

was reached in week 5 in the 2 x 8-Gy arm and in week 7 in the 10 x 3-Gy arm, as the average total symptom score was below the initial score. The palliative effect was longer in the 10 x 3-Gy arm, persisting until week 22, and was also less progressive in intensity than in the 2 x 8-Gy arm. This difference over time was significant (P < .001). No differences in severe (grade 3 to 5) toxicity between both arms influenced the different pattern of palliation. Five patients experienced severe acute toxicity of dyspnea, malaise, and/or nausea, four of whom were treated in the 10 x 3-Gy arm and one in the 2 x 8-Gy arm. No patient developed a myelopathy in our study. No significant differences in the usage and kind of comedication existed between both treatment arms to explain the observed differences. Reirradiation took place in eight patients (5%) in the 10 x 3-Gy arm. One of these patients was even two times reirradiated. In the 2 x 8-Gy arm nine patients (6%) were retreated with irradiation. A subsequent treatment with chemotherapy after randomization was not registered. In a retrospective subgroup-analysis of a sample of 105 patients equally assigned to both treatment arms, four patients retrieved re-treatment with chemotherapy. These four patients were all previously treated according the 10 x 3 Gy-arm.

Figure 4: Kaplan Meier survival curve. Black line: 10 x 3 Gy. Grey

line: 2 x 8 Gy.

Somewhat unexpectedly, the survival analysis revealed a significantly better survival in the 10 x 3-Gy arm (P < .0299; Fig 4). The 1-year survival was 19.6% (95% CI, 14.1 to 27.3%) in the 10 x 3-Gy arm (26 of 148 patients) and only 10.9 % (95% CI, 6.9 to 17.3%) in the 2 x 8-Gy arm (15 of 149 patients). Even after 3 years, the survival rate in the 10 x 3-Gy arm was 5.6% (95% CI, 2.8 to 11.3) (four of 148) v 1.6% (95%

Page 87: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

85

CI, 0.4 to 6.1) (two of 149) in the 2 x 8-Gy arm. This difference in survival existed from week 15 and further and varied between 4 and 11%. Discussion Our study comparing 10 x 3-Gy with 2 x 8-Gy palliative irradiation in NSCLC revealed a superiority of the former over the hypofractionation scheme. Thoracic symptom scores for both schemes were similar over the initial 39 weeks. However, the palliative effect differed significantly over time between treatment arms. In the 10 x 3-Gy arm, the onset of the palliative effect occurred later and persisted longer. Furthermore, the 1-year survival was better in patients treated using the 10 x 3-Gy arm. We observed a reduction of symptoms in line with several other randomized studies, which found no differences in palliative effect using different fractionation schemes.6-

8,18-20 Only one report thus far showed a better palliative effect with 45 Gy in 18 fractions in 3.5 weeks compared with 31.2 Gy in four fractions over 4 weeks. 21 Our total symptom score gives no information on symptom relief per symptom, but is an indicator of the overall palliative effect. Symptom relief will vary depending the type, number, and intensity of the complaints. 4,22,23 An equal distribution for prognostic factors over both treatment arms is also important. Although in our study no stratification for all these factors was performed, all items were well balanced over both treatment arms. Bezjak et al8 proposed to restrict measurements of palliation only to the index symptom, because response of individual symptoms will be less than that of the index symptom. These larger changes in an individual patient will therefore not be seen at the group level when the average change was reported. As a result, analyzing the index symptom may be more sensitive to change at the expense of providing inadequate information about the overall palliative effect. Stephens et al22 proposed that palliation should not only measure reduction of complaints, but also stabilization and occurrence of new complaints due to tumor and/or treatment. Furthermore the duration of palliation should be stated. All these aspects are included in our scoring system of the palliative effect. We observed a significantly different pattern in palliation over a period of 39 weeks between the two treatment arms. The method for assessing palliation appears crucial. Hopwood et al24 showed that differences in missing information between studies can lead to incorrect conclusions, which may be a consequence of differing patterns of change in symptoms during treatment. The differences in the pattern of symptom reduction between both our treatment arms can support this finding. Measuring once or only a few times, or weekly as in our study, will produce differences in outcome. Therefore, comparing studies is difficult, as frequency and duration of measurements varies substantially.5 Combining the sum of the palliative effect by symptom reduction and the symptoms induced by the treatment in our total symptom score can be responsible for visualizing these different effects in time and results in a more precise view on the overall effect of these particular treatments. Our results are consistent with those of Macbeth et al9, who also observed that hypofractionation resulted in a quicker reduction of complaints, whereas hyperfractionation resulted in a longer duration of palliation. Our data gave insufficient support to the assumption that the difference in pattern we found could occur because of differences in the radiobiological equivalent dose

Page 88: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

86

(BED).25,26 The 10 x 3-Gy arm was less toxic, and a steep and significant rise in total symptom scores was seen in the first weeks post-treatment after 2 x 8 Gy. The relation between tumor control and radiation dose with these palliative treatment schedules is uncertain. Notably, in this poor prognostic group, local tumor growth could be the major cause of complaints and possibly local tumor progression,and also the major cause of death. As follow-up in our study was based only on questionnaires, no data about objective tumor response were available. An earlier MRC study6 found no differences in radiological responses, using radiation doses comparable with that used in our study. Even with higher doses, data about tumor control are conflicting. Sundstrøm et al20 presented no relation between tumor dose and local control on chest x-ray, whereas Nestle et al19 and Reinfuss et al27 found a tendency of better control with higher radiation doses. The observation of a survival gain in the 10 x 3-Gy arm in our study was the most surprising finding, and it must be interpreted with caution, as difference in survival were not detected in most other comparable studies4,6,7,18,19,20 and especially not in patients with poor general condition.6,8 The difference in survival seen in patients treated with a multiple fractionation scheme was only seen in those having a good performance score.8,9,27 In a subgroup analysis we also found that the advantage in survival in the 10 x 3 Gy-arm over the 2 x 8-Gy arm was statistically significant in patients with ECOG 0 to 1, but not in patients with ECOG 2 to 4. All patients with ECOG 0 to 1 had stage IV disease. Although there was a tendency, no significant difference in survival was seen in subgroup analysis between both treatment arms for stage IV. The fact that most patients had also a substantial weight loss could be responsible for this finding. The definition of a poor prognosis patient population is unclear and remains to be elucidated. From a statistical point of view, chance could be another explanation for finding a difference in survival between both treatment arms, although the level of significance that was detected is considered sufficient.28 Stephens29 has critical reviewed the risks of obtaining false positive results with sample sizes, and this factor cannot be fully excluded. As such, conformation of a survival advantage for the 10 x 3 Gy scheme will be necessary as it appears to be significant, and clinically relevant. Despite the prolongation in symptom relief and better survival, hardly any re-treatment, including chemotherapy, was observed. This can be a result of our selection, as patients were at referral no (longer) candidates for chemotherapy. Secondly, the performance state will not automatically improve, despite a symptom response. And finally, only recently indications for PS ECOG 2 are formulated.10 In patients with short expected survival 2 x 8 Gy can be the treatment of choice as most acute symptoms induced by toxicity can be treated or prevented with steroids and/or analgetics.30 However, based on the criteria of this study, we could not define this particular group of patients. Therefore, although the overall palliative effect is equal to that of 2 x 8 Gy, we still recommend 10 x 3 Gy because of a longer duration of the palliative response and a significant benefit in survival.

Acknowledgements We thank following colleagues for participating in this study A.L.J. Uitterhoeve (AMC, Amsterdam), J. Bussink (RADIAN, location UMC St. Radboud, Nijmegen), A.H.D. van de Leest (AZG, Groningen), C. Niël (MCH, The Hague), M.J.C. van der Sangen (Catharina hospital, Eindhoven), E.H.J.M. Rutten

Page 89: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

87

(MCA, Alkmaar), M.F.H. Dielwart (ZRTI, Vlissingen), P. van der Maden (Sophia hospital/Isala, Zwolle), J. Pomp (RDGG, Delft) This study was financially supported by the Dutch Health Care Insurance Board (College voor Zorgverzekeringen) as CKVO (Clinical Trial Review Committee) study OG98/009. References 1. Non-Small Cell Lung Cancer Collaborative Group. Chemotherapy in non-small

cell lung cancer: a meta-analysis using updated data on individual patients from 52 randomised clinical trials. Br Med J 1995;311:899–909.

2. American Society of Clinical Oncology. Special Article: clinical practice guidelines for treatment of unresectable non-small cell lung cancer. J Clin Oncol 1997;15:2996–3018.

3. Brundage MD, Bezjak A, Dixon P et al. The role of palliative thoracic radiotherapy in non-small cell lung cancer. Can J Oncol 1996;6(Suppl. 1):25–32.

4. Rees GJG, Devrell CE, Barley VL et al. Palliative Radiotherapy for Lung Cancer: Two Versus Five Fractions. Clinical Oncology 1997;9:90–95.

5. Macbeth F, Toy E, Coles B et al. Palliative radiotherapy regimens for non-small cell lung cancer. Cochrane Database Syst Rev 1:CD002143,2002.

6. Bleehen NM, Girling DJ, Fayers PM et al. Inoperable non-small-cell lung cancer (NSCLC): a Medical Research Council randomized trial of palliative radiotherapy with two fractions or ten fractions. Br J Cancer 1991;63:265–270.

7. Bleehen NM, Girling DJ, Machin D et al. A Medical Research Council (MRC) randomized trial of palliative radiotherapy with two fractions or a single fraction in patients with inoperable non-small-cell lung cancer (NSCLC) and poor performance status. Br J Cancer 1992;65:934–941.

8. Bezjak A, Dixon P, Brundage M, et al. Randomized study of single versus fractionated radiotherapy (RT) in the palliation of non-small cell lung cancer. Int J Radiat Oncol Biol Phys 2002;54:719–728.

9. Macbeth FR, Bolger JJ, Hopwood P et al. Randomized trial of palliative two-fraction versus more intensive 13-fraction radiotherapy for patients with inoperable non-small cell lung cancer and good performance status. Clinical Oncology 1996;8:167–175.

10. Lilenbaum R. Management of advance non-small-cell lung cancer in patients with a performance status of 2. Clin Lung Cancer 2004;5:209–213.

11. De Haes JCJM, van Knippenberg FCE, Neijt JP. Measuring psychological and physical distress in cancer patients: structure and application of the Rotterdam Symptom Checklist. Br J Cancer 1990;62;1034–1038.

12. Bergman OB, Aaronson NK, Ahmedzai S, et al. The EORTC QLQ-LC13: a modular supplement to the EORTC Core Quality of Life Questionnaire (QLQ-C30) for use in lung cancer clinical trials. EORTC Study Group on Quality of Life. Eur.J.Cancer 1994; 30A:635–42.

13. Noordijk EM. Classification, Staging and radiotherapy for bronchuscarcinoma (PhD-thesis in Dutch, page 156), Leiden University Medical Center, The Netherlands, 1983.

Page 90: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

88

14. Caspers RJ, Welvaart K, Verkes RJ, et al. The effect of radiotherapy on dysphagia and survival in patients with esophageal cancer. Radiother Oncol 1988;12:15–23.

15. Williams J, Chen Y, Rubin P et al. The biological basis of a comprehensive grading system for the adverse effects of cancer treatment. Semin Radiat Oncol 2003;13:182–8.

16. S-PLUS 6 for Windows Guide to Statistics, Volume 1, Insightful Corporation, Seattle, WA USA.

17. Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc 1958;53:457–481.

18. Abratt R, Shepherd L, Salton D et al. Palliative radiation for stage 3 non-small cell lung cancer – A prospective study of two moderately high dose regimens. Lung Cancer 1995;13:137–143.

19. Nestle U, Nieder C, Walter K et al. A palliative accelerated irradiation regimen for advanced non-small cell lung cancer vs. conventionally fractionated 60 Gy: results of a randomized equivalence study. Int J Rad Onc Biol Phys 2000;48:95–103.

20. Sundstrøm S, Bremnes R, Aasebø U et al. Hypofractionated Palliative Radiotherapy (17 Gy per two fractions) in Advanced Non-Small-Cell Lung Carcinoma Is Comparable to Standard Fractionation for Symptom Control and Survival: A National Phase III Trial. J Clin Onc 2004;22:801–810.

21. Teo P, Tay T, Choy D et al. A randomized study on palliative radiation therapy for inoperable non-small cell carcinoma of the lung. Int J Rad Onc Biol Phys 1987;14:867–871.

22. Stephens RJ, Hopwood P, Girling DJ. Defining and analyzing symptom palliation in cancer clinical trials: a deceptively difficult exercise. Br J Cancer 1999;79:538–544.

23. Kramer GW, Gans S, Ullmann E et al. Hypofractionated external beam radiotherapy as retreatment for symptomatic non-small cell lung carcinoma: an effective treatment? Int J Rad Onc Biol Phys 2004;58:1388–1399.

24. Hopwood P, Stephens RJ, Machin D. Approaches to the analysis of quality of life data: experiences gained from a Medical Research Council Lung Cancer Working Party palliative chemotherapy trial. Quality of Life Research 1994;3:339–352.

25. Fowler JF. The linear-quadratic formula and progress in fractionated radiotherapy. Br J Radiology 1989;62:679–694.

26. Singer JM, Price P, Dale RG. Radiobiological prediction of normal tissue toxicities and tumour response in the radiotherapy of advanced non-small-cell lung cancer. British Journal of Cancer 1998;78:1629–1633.

27. Reinfuss M, Glinski B, Kowalska T. Radiothérapie du cancer bronchique non à petites cellules de stade III, inopérable, asymptomatique. Résultats définitifs d’un essai prospectif randomize (240 patients). Cancer/Radiotherap 1999;3:475–479.

28. Sterne A., Smith G: Sifting the evidence – what’s wrong with significance tests? BMJ 2001;322:226–231.

29. Stephens R. The need for a world strategy for clinical trials. Lung Cancer 2003;41(S3): 96.

30. Michalowski A. On radiation damage to normal tissue and its treatment. II: anti-inflammatory drugs. Acta Oncol 1994;33:139–157.

Page 91: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Chapter 7

COST-UTILITY ANALYSIS OF SHORT- VERSUS LONG-COURSE

PALLIATIVE RADIOTHERAPY IN PATIENTS WITH

NON-SMALL-CELL LUNG CANCER

Wilbert B. van den Hout1, Gijsbert W.P.M. Kramer2, Ed M. Noordijk3, Jan Willem H. Leer2

1 Department of Medical Decision Making, Leiden University Medical Center,

Leiden, The Netherlands. 2 Department of Radiotherapy, RADIAN, Arnhems Radiotherapeutic Institute,

Arnhem, The Netherlands. 3 Department of Clinical Oncology, Leiden University Medical Center, Leiden, The

Netherlands.

J Nat Cancer Institute 2006; 98(24):1786-1794.

Page 92: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

90

Summary Background: In poor-prognosis patients with non-small-cell lung cancer (NSCLC), radiotherapy can effectively palliate complaints. Controversy remains on whether short-course or more protracted schedules provide better value for money. Methods: A societal cost-utility analysis was conducted, alongside a Dutch multi-center randomized trial with one-year follow-up, comparing radiotherapy in either a 10×3 Gy or a 2×8 Gy schedule in 297 patients. Lifelong quality adjusted life years (QALYs) were estimated using the EuroQol questionnaire (EQ-5D). Lifelong societal costs were estimated using a model estimated from cost questionnaires filled out by a subset of patients. Results: QALYs were more favorable for the 10×3 Gy schedule than the 2×8 Gy schedule (20.0 versus 13.2 weeks, P = 0.04), which was mainly due to the difference in survival (38.1 versus 27.4 weeks, P = 0.03) as opposed to the difference in the average valuation of health (0.41 versus 0.37, P = 0.27). Radiotherapy (related) costs were estimated at $ 5236 versus $ 2512 (P < 0.001). The 39% difference in life expectancy rendered an estimated 30% difference in survival-related costs. The cost-utility ratio was estimated at $ 40,900 per QALY. Conclusions: In our group of poor-prognosis NSCLC patients, the estimated cost-utility ratio for the 10×3 Gy schedule was acceptable according to current economic standards. The additional costs for the protracted schedule were justified not by improved quality of life, but by longer survival. Keywords: cost-utility analysis; non-small-cell lung cancer; radiotherapy; palliation

Page 93: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

91

Introduction In patients with non-small-cell lung cancer (NSCLC), palliative radiotherapy can effectively reduce chest pain and hemoptysis, and also dysphagia and dyspnea.1-3

Combined-modality treatment for patients with stage III disease and chemotherapy for patients with stage IV disease have resulted in improved quality of life and survival.4 However, patients are frequently too ill for these more intensive treatments.5 Moreover, even after palliative chemotherapy, locoregional treatment may eventually be required to palliate locoregional complaints. Controversy remains on whether or not more protracted radiotherapy schedules provide better results than short-course.6 So far, none of the trials comparing different schedules have shown a difference in palliation.7– 10 On survival the results have been inconclusive: two Medical Research Counsel trials have shown no difference7,8, whereas two later trials by Macbeth and Bezjak9,10 did show a difference in especially patients with a good prognosis. So far, no economic evaluations on this subject have been published. To reach an evidence-based consensus on the preferred palliative schedule for patients with poor-prognosis NSCLC, a Dutch randomized trial was started in 1999.11 Included patients were either stage IV patients or stage IIIA/B patients with weight loss or a poor performance score (Eastern Cooperative Oncology Group (ECOG) score ≥ 2).2 They were randomized to receive radiotherapy consisting of either ten fractions of 3 Gy (10×3 Gy) or two fractions of 8 Gy (2×8 Gy). The study showed no difference in average symptom control over the initial 9 months. However, there was a statistically significant difference in time pattern, suggesting prolonged palliation in the 10×3 Gy group with less worsening of symptoms than in the 2×8 Gy group. In addition, 1-year survival was significantly more favorable in the 10×3 Gy group (19.6% versus 10.9%). It was therefore concluded that, also for patients with poor-prognosis NSCLC, the 10×3 Gy schedule was preferred over the 2×8 Gy schedule. However, in this conclusion the difference in costs between both treatments was not taken into account: the protracted 10×3 Gy schedule results in higher medical and patient costs and, also, the gain in survival leads to continued costs. As a result, controversy remained on whether the better effectiveness of the 10×3 Gy schedule justified the additional costs. Here we will present the full cost-utility analysis of our study. Effectiveness is measured by quality-adjusted life years (QALYs), which is the overall valuation of the lifelong health of the patients in the study. Effectiveness is compared to the total costs to society, including not only the medical costs of radiotherapy but also costs of other health care and costs incurred by the patients. Aim of the analysis is to show, from a societal perspective, which radiotherapy schedule provides better value for money. Methods From January 1999 until May 2002, a total of 303 NSCLC patients from 13 out of 21 Dutch radiotherapy centers were included in the study. Table 1 shows the inclusion and exclusion criteria. Six stage III patients with ECOG scores 0 or 1 and no weight loss were later excluded because they did not satisfy the inclusion criteria. The remaining 297 patients were randomly assigned to schedules with either 10×3 Gy (in

Page 94: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

92

four or five fractions per week, n = 148) or 2×8 Gy (one week apart, n = 149). Patients were followed for one year. Primary outcome measure of the study was a patient-assessed symptom score, measuring total burden on seven symptoms: loss of appetite, dyspnea, chest pain, cough, hemoptysis, hoarseness, and dysphagia (each rated on a four-point scale ranging from bothered not at all to bothered very much).

* An Eastern Cooperative Oncology Group score of 2 denotes that the patient is ambulatory and capable of all self-care, unable to carry out any work activities, and up and about more than 50% of waking hours (www.ecog.org/general/perf_stat.html)

† Weight loss was defined as a decrease of at least 5% in 3 months or at least 10% in 6 months before inclusion.

‡ Eligibility for chemotherapy was assessed by the treating pulmonologist, based on prior chemotherapy, weight loss and co-morbidity.

At 5% significance level, a total of 268 patients were needed for the primary outcome measure to obtain a power of 90% for correctly concluding equivalent response.11 The study was approved by the Medical Ethics Committees of all participating institutions and random assignment took place after receiving informed consent of the patient. Assessment of quality adjusted life years In July 2002 the last systematic assessment of survival was performed. At that time 269 (91%) patients had died. Until February 2004, 12 (4%) more patients were reported to have died. To obtain individual survival and QALY data for each patient, the remaining 16 (5%) patients were assumed to have died in February 2004, with survival ranging from 21 to 60 months. This underestimation of the survival time led to less favorable estimated results for the treatment schedule with better survival. During the one-year follow-up, starting before random assignment, patients filled out 13 weekly and 21 biweekly mailed questionnaires containing questions on specific symptoms and on quality of life measured using the EuroQol classification system (EQ-5D). The EQ-5D assesses general health status, by five questions on mobility, self-care, usual activities, pain/discomfort, and anxiety/depression.12 From the EQ-5D classification system, the EQ-5D utility was calculated.13 This utility reflects the valuation by the general public of the reported health states, ranging from 1.00 (optimal health), through 0.00 (as bad as death) to −0.594 (worse than death). In addition, patients directly evaluated their health on a 100 mm horizontal visual

Table 1. Eligibility criteria for participation in the trial

Inclusion criteria - cytologically or histologically confirmed NSCLC - either stage IV patients or stage IIIA/B patients with ECOG* score ≥ 2 or weight loss† - at least some self-reported burden due to the tumor on at least one of seven complaints:

loss of appetite, dyspnea, chest pain, cough, hemoptysis, hoarseness, dysphagia - physically and mentally fit enough to participate in the study

Exclusion criteria

- stage IV patients with ECOG* score ≤ 2 that were still eligible for chemotherapy‡ - concurrent chemotherapy - prior radiotherapy to the chest - superior vena cava syndrome (SVCS) at presentation - other malignant diseases in the past

Page 95: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

93

analogue scale (VAS), ranging from worst imaginable health to perfect health (0.0 to 1.0). For each patient the quality adjusted life years (QALYs) were calculated as the lifelong area under the utility curve. Utility was discounted at 3% per year. Utility measurements were only available during the initial year. Moreover, especially shortly before their time of death, patients tended to stop returning their questionnaires. Of, on average, 17.3 possible lifetime EQ-5D measurements, 3.0 (17%) were missing, of which 1.1 (6%) in the last month before dying. Because the values of the missing utility measurements are likely to be worse than the non-missing measurements, neglecting the missing data or carrying forward the last measurement would both lead to an overestimation of utilities. For that reason, the missing EQ-5D utilities were imputed, using a decreasing curve from the last available measurement (estimated from the data shown in Figure 2b using non-linear regression: imputed EQ-5D utility = constant × (1 + 50 / remaining lifetime) −1, with the constant fitted to the patient's last available EQ-5D utility, R2 = 0.23). This utility model provides results that are lower than carrying forward the last available utility measurement, but higher than linear descent. Assessment of costs For each patient, lifelong costs were estimated from a societal perspective. Estimated costs included the medical costs of radiotherapy, the non-medical costs of radiotherapy (time and travel costs incurred by the patients), other medical costs (like hospitalizations), health-related non-medical costs (like informal care) and survival-related costs.14 The medical costs of radiotherapy were estimated using a previously published department model15, in which costs of different types of staff, equipment, material, housing, and overhead were obtained from three radiotherapy institutions and combined to generate a typical Dutch radiotherapy department. To estimate the costs of different radiotherapy schedules, each cost item was assigned to either of three allocation bases: treatments (for cost items independent of the treatment schedule), fractions (for cost items proportional to the number of fractions given), or Gy (for cost items proportional to the dose delivered). This way, unit cost prices were estimated at $ 1898 per treatment, plus $ 109 per fraction, plus $ 9 per Gy, resulting in medical costs of $ 3260 and $ 2261 for the 10×3 Gy and the 2×8 Gy schedules, respectively. For divergent schedules and repeated radiotherapy, the actual schedules were used to estimate costs. Average reported time spent per fraction was 126 minutes and 68% of the patients reported a travel companion. Valued at $ 11 per hour16, average time costs per fraction were estimated at $ 38. Patients reported their means of transportation as car (42%), taxi (50%), ambulance (6%) or walking (3%). The average travel distance was 20 kilometers and average travel costs were estimated at $ 65 per fraction. Non-radiotherapy health care costs during the initial 12 weeks were estimated using six bi-weekly mailed cost questionnaires. For practical reasons and to limit the burden to the patients, these questionnaires were filled out by a subsample of the patients from three different radiotherapy institutes. Patients could refuse to participate without being excluded from the effectiveness study. Of 113 consecutive patients that were asked to fill out the cost questionnaires, 56 (50%) consented. Of, on average, 4.6 possible lifetime cost questionnaires, 0.9 (19%) were missing, of which 0.4 (9%) in the last month before dying. In case of missing cost measurements, the previous available measurement was carried forward. In the three-page, mostly closed-format, cost questionnaires, patients reported consultations (GP, specialists, and paramedical

Page 96: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

94

professionals), hospitalizations, purchased medication, home (nursing) care, paid domestic help, informal care, and out-of-pocket expenses (due to radiotherapy and otherwise).17 Most cost prices were obtained from Dutch standard prices that were designed to reflect societal costs and to standardize economic analyses.18,19 Medical costs of consultations varied from $ 10 per telephone GP consultation to $ 82 per neurologist consultation. Hospitalizations were valued at $ 545 per day for clinical admission, $ 218 per day for daycare, and $ 163 per day for nursing care. Reported health care costs include the patients' time and travel costs. Purchased medication was valued according to the Pharmacotherapeutic Compass20, plus $ 6 per non-drugstore purchase. Home nursing care and other home care were valued at $ 38 and $ 21 per hour, respectively. Other paid domestic care and informal care were both valued at $ 11 per hour.16 Out-of-pocket expenses were valued as reported by the patients. Lifelong non-radiotherapy health care costs for the entire sample were estimated using a regression model estimated from the data obtained from the cost questionnaires. Using linear multilevel analysis (MLwiN version 1.1), adjusting for repeated measures, significant predictors of total non-radiotherapy costs were selected (stepwise selection: enter 0.05, remove 0.10) from the following lifelong-available variables: time since randomization, time until death, age, gender, EQ-5D utility, and VAS utility. Also, two separate dummies for costs during the initial four weeks in both randomization groups were included, to capture the costs related to the initial radiotherapy. Because of the low labor participation among these patients, differences in productivity costs were assumed to be negligible.15 In a sensitivity analysis, consumption costs were included21, to the amount of $ 43 per day (average consumer expenditures for persons aged 65-74 years, excluding healthcare, insurance and pension, www.bls.gov/cex). Costs were discounted at 3%, updated to price level 2005 euros using the price index rate for the Dutch health care sector (www.cbs.nl) and converted to U.S. dollars using the July 2005 Dutch purchasing power parity index (€ 1 = $ 1.09, www.oecd.org). Analysis In the cost-utility analysis, the difference in lifelong QALYs was compared to the difference in lifelong societal costs. Whether a particular treatment is more cost-effective than another depends on the willingness-to-pay (WTP) per QALY. The average net benefit (WTP × QALYs − Costs) in both randomization groups was compared, for a range of WTP values. Significantly rejecting equality of the net benefit demonstrates the superior cost-effectiveness of the more favorable schedule. The net benefit approach gets around the problems associated with uninterpretable negative cost-utility ratios. All outcome measures were estimated on an intention-to-treat basis. For all outcome measures, differences were tested using double-sided non-parametric bootstrapping22, with 1,000,000 replications and 0.05 significance threshold. Reported confidence intervals (CIs) are the corresponding symmetric 95% trimmed intervals. Bootstrapping explicitly compares the means in both groups without making distributional assumptions, thus allowing for skewed distributed costs. Statistical analysis only represents uncertainty due to the random selection of the patient population. Uncertainty due to modeling assumptions was analyzed using univariate sensitivity analyses on the assumed lifetime for patients with unknown date

Page 97: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

95

of death (February 2004 versus five year after inclusion), the utility measure (EQ-5D versus VAS), and the included cost categories. Results

* Symptom score ranging from 7 to 28, measuring total burden on seven symptoms (each

rated on a four-point scale ranging from bothered not at all to bothered very much). Table 2 shows the baseline characteristics of the 297 patients included in the study. Baseline characteristics of the subsample of 56 patients that filled out the cost questionnaires did not differ significantly from the rest of the sample (data not

Table 2. Baseline patient characteristics

10×3 Gy (n = 148)

2×8 Gy (n = 149)

Age (median, range) 69 (48 – 85) 69 (41 – 91)

Sex (n, %) - male - female

118 (80) 30 (20)

119 (80) 30 (20)

Total symptom score* (median, SD) EQ-5D utility (median, SD) VAS(median, SD)

13 (2.4) 0.62 (0.35) 0.52 (0.27)

13 (2.5) 0.52 (0.35) 0.59 (0.26)

Tumor stage (n, %) - IIIA - IIIB - IV

16 (11) 56 (38) 76 (51)

24 (16) 59 (40) 66 (44)

Eastern Cooperative Oncology Group score (n, %)- score 0 - score 1 - score 2 - score 3 - score 4

9 (6)

45 (30) 60 (41) 33 (22)

1 (1)

5 (3)

48 (32) 44 (30) 51 (34)

1 (1)

Substantial weight loss (n, %) 104 (70) 109 (73)

Tumor type (n, %) - squamous cell carcinoma - adenocarcinoma - large cell carcinoma - other

64 (43) 37 (25) 39 (26)

8 (5)

60 (40) 27 (18) 53 (36)

9 (6)

Months since diagnosis (median, range) 1 (0 – 88) 1 (0 – 99)

Treatment indication (n, %) - primary lesion - recurrent lesion (no prior radiotherapy)

142 (96)

6 (4)

147 (99)

2 (1)

Prior chemotherapy (n, %) 18 (12) 20 (13)

Other treatment at start radiotherapy (n, %) - pain medication - antibiotics - other medication - supplemental oxygen

80 (54) 54 (36)

5 (3) 49 (33)

9 (6)

98 (66) 67 (45)

5 (3) 58 (39)

8 (5)

Page 98: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

96

shown), except that it contained more stage IV patients (59% versus 48%, P = 0.05) and more patients with squamous cell carcinoma (57% versus 42%, P = 0.04). Quality adjusted life years In accordance with the poor prognosis of the patients, the survival time was known for 95% of the patients. The solid lines in Figure 1 show the survival curves, assuming that the remaining 5% died in February 2004.

0,0

0,2

0,4

0,6

0,8

1,0

0 1 2 3 4 5

Years since randomization

Surv

ival

Baseline analysis

Missings set at 5 years

10x3 Gy

2x8 Gy

Figure 1. Survival, by randomization group The corresponding life expectancy was estimated at 38.1 weeks for the 10×3 Gy group and 27.4 weeks in the 2×8 Gy group (Table 3), with a statistically significant difference of 10.7 weeks (P = 0.03, CI 0.9 to 20.6 weeks).

* The 5% missing survival times set at 5 years, instead of February 2004

Table 3. Quality adjusted life expectancy, in weeks (averages and SD)

10×3 Gy (n = 148)

2×8 Gy (n = 149)

Difference (95% CI)

P value

Base-case analysis - Life expectancy - QALYs

38.1 (49.3) 20.0 (33.9)

27.4 (35.7) 13.2 (23.3)

10.7 (0.9 to 20.6) 6.8 (0.1 to 13.5)

0.03 0.05

Sensitivity analyses - Life expectancy* - QALYs* - QALYs based on VAS

44.4 (67.1) 23.1 (44.1) 21.6 (33.9)

30.5 (47.9) 15.0 (31.0) 15.2 (24.6)

13.9 (0.6 to 27.3) 8.1 (-0.7 to 16.8) 6.4 (-0.4 to 13.2)

0.04 0.07 0.06

Page 99: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

97

Figure 2a shows the valuation of health obtained from the EQ-5D, averaged over the measurements from those patients that were still alive. The utility improved during the initial months, but later on stabilized.

0,0

0,2

0,4

0,6

0,8

1,0

0 13 26 39 52Weeks since randomization

EQ-5

D u

tility

10x3 Gy

2x8 Gy

Figure 2a. EQ-5D utility, at time since randomization or time until death. Figure 2b shows the EQ-5D utility as a function of the remaining lifetime, instead of as a function of the time since randomization. Towards the end of life the EQ-5D utility markedly decreased, to values even below zero. In fact, 10% of all utility measurements were negative, indicating health states valued worse than death.

0,0

0,2

0,4

0,6

0,8

1,0

013263952Weeks until date of death

EQ-5

D u

tility

10x3 Gy

2x8 Gy

Utility model

Figure 2b. EQ-5D utility as a function of the remaining lifetime. In line with Figure 2a, average utility over the remaining lifetime was somewhat more favorable in the 10×3 Gy group, but not statistically significantly so (0.41 versus 0.37, P = 0.27). Combined with the longer survival, quality adjusted life years were significantly more favorable in the 10×3 Gy group (20.0 versus 13.2 weeks, P = 0.05), with an estimated difference of 6.8 weeks (CI 0.1 to 13.5 weeks).

Page 100: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

98

Costs of radiotherapy Average medical costs of the initial radiotherapy were estimated at $ 3120 for the 10×3 Gy group and $ 2173 for the 2×8 Gy group (Table 4). The actual radiotherapy schedule differed from the randomization schedule for 35 patients (14% versus 10%, P = 0.28), mostly due to early treatment withdrawal. Apart from travel expenses, out-of-pocket costs (e.g. for menthol cream and additional telephone costs) were low. Time and travel costs made up 22% and 9% of the costs of the initial radiotherapy, respectively. Because a considerable part of the medical costs are independent of the schedule, time and travel costs even made up 43% of the difference in costs. Repeated radiotherapy was equally frequent in both groups (5% versus 6%, P = 0.84), and the difference in costs of repeated radiotherapy was negligible ($ 150 versus $ 148, P = 0.98). The total radiotherapy costs were estimated at $ 4182 for the 10×3 Gy group and $ 2512 for the 2×8 Gy group, with an estimated difference of $ 1670 (CI $ 1459 to $ 1881).

Societal costs Health care utilization and costs, estimated from the cost questionnaires filled out by a subset of patients, showed no significant differences between both randomization groups (Table 5, P ≥ 0.16). Two-thirds of the estimated costs consisted of hospitalization costs. Although, over the initial 12-week period, the cost questionnaire did not show a difference between the randomization groups, the data did provide an opportunity to construct a model to estimate the difference in survival-related costs. Omitting statistically non-significant predictors (P ≥ 0.30), the model showed a relationship between costs and EQ-5D utility (P = 0.04) and increased costs during the initial four weeks in the 10×3 Gy group (P = 0.001), mainly due to hospitalizations. In the

Table 4. Lifelong societal costs per patient (averages in $ and SD)

10×3 Gy (n = 148)

2×8 Gy (n = 149)

Difference (95% CI)

P value

Initial radiotherapy

- medical costs - time costs - travel costs - out-of-pocket costs

Repeated radiotherapy Total radiotherapy costs

3120 308 591 14

150 4182

2173 63

121 6

148 2512

947 245 470

8 2

1670

<0.001 <0.001 <0.001

0.51 0.98

<0.001

Non-radiotherapy costs - related to radiotherapy - related to survival

Total non-radiotherapy costs

1054 11254 12308

0

8651 8651

1054 2602 3656

<0.001

0.09 0.02

Total societal costs 16490 11164 5326 <0.001

Page 101: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

99

2×8 Gy group, costs in the initial four weeks were not statistically significantly increased (P = 0.96, CI $-179 to $ 189). The model estimated the undiscounted weekly non-radiotherapy costs at $ 517, minus $ 395 times the EQ-5D utility, plus $ 276 during the initial four weeks in the 10×3 Gy group (χ2(2) = 14.7, Figure 3). After the initial four weeks, weekly non-radiotherapy costs according to this model vary from $ 122 for patients in perfect health (i.e. $ 517 – 1.0 × $ 395) to $ 752 for patients in worst possible health (i.e. $ 517 + 0.5940 × $ 395).

Applying the estimated cost model to all patients, the inclusion of radiotherapy-related costs, during the initial four weeks, increased the cost difference between both randomization groups by $ 1054 ($ 1054 versus $ 0, Table 4). Together with the costs of the initial and repeated radiotherapy, the radiotherapy (related) costs were estimated at $ 5236 versus $ 2512 (P < 0.001, difference $ 2724, CI $ 2501 to $ 2947). In addition, the 39% difference in survival led to a 30% difference in related non-radiotherapy costs (P = 0.09, difference $ 2602, CI $ −350 to $ 5555). Including both the radiotherapy (related) costs and the survival-related costs, the total difference in lifelong societal costs was estimated at $ 5326, in favor of the 2×8 Gy schedule (P < 0.001, CI $ 2330 to $ 8323).

Table 5. Non-radiotherapy costs per patient during the first 12 weeks (averages in $)

10×3 Gy (n = 31) 2×8 Gy (n = 25)

Utilization Costs Utilization Costs P value General practitioners Lung specialist Radiotherapist Other specialists Paramedical professionals Hospitalization Analgesics Antibiotics Other medication

3.4 1.6 1.2 0.5 1.0

39% 52% 13% 68%

126 164 101 50 35

2726 76 5

143

4.8 2.5 1.1 0.8 1.9

16% 56% 4% 48%

142 247 94 67 71

1157 32 1

103

0.72 0.25 0.83 0.63 0.48 0.23 0.16 0.18 0.53

Home (nursing) care Paid domestic help Informal care Out-of-pocket expenses

4.1 hr 0.0 hr 7.3 hr

3%

114 0 78 1

0.6 hr 3.1 hr 18 hr 12%

12 33

193 11

0.30 0.22 0.19 0.44

Total non-radiotherapy costs 3617 2162 0.30

Page 102: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

100

0

500

1000

1500

1 2 3 4 5 6Cost questionnaire

Non

-rad

ioth

erap

y co

sts

per t

wo

wee

ks (i

n $)

Observed, 10x3 GyObserved, 2x8 GyModel, 10x3 GyModel, 2x8 Gy

Figure 3. Non-radiotherapy costs, by randomization group. Cost-utility analysis Compared to the 2×8 Gy schedule, the 10×3 Gy schedule provides significantly more QALYs, but at significantly higher costs. Aim of the cost-utility analysis is to show whether or not the additional costs of the 10×3 Gy schedule are justified by the additional QALYs, which depends on the relative value of QALYs and money: for high willingness-to-pay the 10×3 Gy schedule is preferred, whereas for low willingness-to-pay the 2×8 Gy schedule is preferred. The break-even point at which both are equally preferred is at willingness-to-pay equal to the cost-utility ratio of $ 40,900 per QALY. For willingness-to-pay below $ 19,400 per QALY and above $ 1,100,000 per QALY, the preference was statistically significantly in favor of the short- and the long-course schedule, respectively (P ≤ 0.05). Sensitivity analyses Sensitivity analyses were performed to explore the impact of various assumptions. The interrupted lines in Figure 1 show the survival curves, assuming that the 5% patients with unknown date of death survived for five years. These curves are almost identical to the Kaplan-Meier curves (11). Because survival was more favorable in the 10×3 Gy group, also more patients in this group had unknown survival times (7% versus 3%, P = 0.12). Although this concerns only a small part of the patients, assuming longer survival times compared to the base-case analysis had a considerable impact on outcome: the difference in life expectancy between both groups increased from 10.7 to 13.9 weeks (CI 0.6 to 27.3 weeks) and the quality adjusted life expectancy increased from 6.8 to 8.1 weeks (CI –0.7 to 16.8). However, because also

Page 103: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

101

the costs increased with life expectancy, the cost-utility ratio decreased by only 0.1%, to $ 40,800 per QALY. Using the patients' assessment of their health instead of society's assessment (VAS instead of EQ-5D) increased the estimated QALYs, but reduced their difference by 6% (6.4 instead of 6.8 weeks, Table 3). Accordingly, the estimated cost-utility ratio increased by 6% to $ 43,300 per QALY. Non-radiotherapy costs were estimated using a cost model in which the increased costs related to the initial radiotherapy during the initial four weeks in the 2×8 Gy group were not statistically significant and were therefore excluded. Including these costs, despite the fact that they were not statistically significant, rendered a cost model in which the undiscounted weekly non-radiotherapy costs were estimated at $ 515, minus $ 393 times the EQ-5D utility, plus $ 276 and $ 5 during the initial four weeks in the 10×3 Gy group and the 2×8 Gy group, respectively. Using this model, the cost-utility ratio would decrease by only 0.7% to $ 40,600 per QALY. All cost categories were in favor of the short-course radiotherapy, and, in contrast to the other sensitivity analyses, the extent to which the various cost categories were included in the analysis did have a considerable impact on the cost-utility ratio. Excluding survival-related costs reduced the cost-utility ratio from $ 40,900 to $ 20,900 per QALY. Instead, including consumption costs at $ 43 per day increased the cost differences by $ 3078 and the cost-utility ratio to $ 64,500 per QALY. Excluding all non-radiotherapy costs reduced the cost-utility ratio to $ 12,800 per QALY. Discussion In the palliative irradiation of poor-prognosis NSCLC patients, compared to the short-course 2×8 Gy schedule, the long-course 10×3 Gy schedule has been shown to provide prolonged palliation and better survival.11 In the cost-utility analysis presented here, effectiveness was measured by quality-adjusted life expectancy, aiming to capture both length and quality of life. Similar to palliation, also the quality of life was valued better in the 10×3 Gy group, but non-significantly so. Combined with the significant 10.7 week difference in life expectancy, the significant difference in QALYs was estimated at 6.8 weeks. The difference in lifelong societal costs was estimated at $ 5326, of which 31% were radiotherapy costs, 20% were other radiotherapy-related costs during the initial four weeks and 49% were survival-related costs. The cost-utility ratio was estimated at $ 40,900 per QALY. Judged by the, often quoted, acceptability threshold of $ 50,000 per QALY23,24, the 10×3 Gy schedule therefore provides better value for money than the 2×8 Gy schedule. In the Netherlands this has confirmed the consensus that, indeed, the 10×3 Gy schedule should be the preferred standard treatment for poor-prognosis NSCLC patients. A number of remarks are in order concerning the economic preference for the long-course radiotherapy schedule. Firstly, its interpretation should be that long-course radiotherapy need not be withheld because of costs, since the additional costs are acceptable. It should not be concluded that long-course radiotherapy should be imposed because of costs, if, for good reasons, a patient prefers the shorter schedule. On the contrary, long-course radiotherapy was estimated to increase costs by $ 5326. Secondly, in case of restricted radiotherapy capacity and pending the decision to increase that capacity, it may still be more efficient to use the limited capacity for

Page 104: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

102

treatment of other patients with larger expected health gains. And, thirdly, treatment preference in settings other than that in our study can be influenced by differences in economic climate and treatment patterns. A matter of debate among health economists is to what extent future costs due to improved survival should be included in economic evaluations. In our base-case analysis we included both related and unrelated health care costs, because the QALY measure also included the health gain obtained from unrelated health care.14 Both practically and theoretically, it would have been impossible to identify and exclude unrelated effectiveness. Some argue that only related health care costs should be included, because otherwise it would be inconsistent to exclude other future costs of subsistence.25 In our analysis, this would have reduced the estimated cost-utility ratio from the base-case $ 40,900 per QALY to $ 20,900 per QALY, confirming the preference for the 10×3 Gy schedule. Others argue that not only all future health care costs should be included, but also future production and consumption costs.21 In general, this would favor treatment for the young, because their consumption is compensated by productivity. In our analysis, assuming a negligible difference in productivity, including consumption costs would have increased the cost-utility ratio to $ 64,500 per QALY for the 10×3 Gy schedule. This ratio should be judged by other than normal standards but does not seem unacceptably high. A second matter of debate among health economists is how health care costs are related to survival. Traditionally, health care models have described costs in relation to age. Recently it has been argued that costs are more related to the time until death, which would reduce the impact of ageing on costs.26,27 In our analysis we found that both age and time until death were not statistically significant predictors of cost. Instead, the EQ-5D utility measure was predictive, leading to increased costs towards death and a less than proportional relationship between survival time and costs. If the results in our terminally ill patient population are generalizable, then perhaps age is more predictive for chronic diseases, where quality of life is determined by disease progression, whereas time until death is more predictive in economic models for more acute conditions.

Because both radiotherapy schedules provided roughly similar palliation, the conclusion of our economic evaluation strongly hinges on the survival gain. In our study the survival gain was statistically significant, but this has not been a consistent finding in other studies. In their Cochrane review of ten randomized trials6, Macbeth et al conclude that there is evidence for a modest survival gain after higher dose radiotherapy only in patients with better performance status. Kramer et al compared and discussed these results in detail.11 Several remarks can be made concerning the validity of our research methods. Perhaps most importantly, because we had not anticipated the survival difference, long-term costs related to the difference in survival had to be estimated from cost data obtained in the initial 12 weeks. Although our model identified short-term costs related to the initial radiotherapy that should not be extrapolated and although during the initial 12 weeks already more than one-third of the patients had died, the cost model may still not have been representative for long-term health care costs associated with living and dying. Without gold standard we had no means to test the validity of the model. A second matter of concern is the used EQ-5D utility measure that is frequently used in economic evaluations from a societal perspective, but does not include quality of life domains that are specifically relevant in the valuation of end-of-life care.15,28 Unfortunately no valuation instrument exists that incorporates

Page 105: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

103

such issues. Nevertheless, the distinct decrease of the EQ-5D utility towards the end of life demonstrates that the instrument is responsive to the changing health status of these patients and the results were very similar to results from other research in terminally ill cancer patients.15 These, and other, issues were addressed in the sensitivity analyses, which showed that they had very little influence on the estimated cost-utility ratio, thus suggesting that the obtained results are robust.

In conclusion, the inclusion of survival-related costs in our economic evaluation led to a higher but still acceptable cost-utility ratio for the 10×3 Gy schedule. In our group of poor-prognosis NSCLC patients, the additional costs of the protracted schedule were justified not by improved quality of life, but by longer survival. Funding: This study was funded by the Dutch Investigative Medicine Fund of the Health Insurance Executive Board, as CVKO study OG98/009.

References 1. Brundage MD, Bezjak A, Dixon P et al. The role of palliative thoracic

radiotherapy in non-small cell lung cancer. Can J Oncol 1996;6 Suppl 1:25–32. 2. Rees GJ, Devrell CE, Barley VL, Newman HF. Palliative radiotherapy for lung

cancer: two versus five fractions. Clin Oncol 1997; 9(2):90–95. 3. Konski A, Feigenberg S, Chow E. Palliative radiation therapy. Semin Oncol 2005;

32(2):156–164. 4. Non-small Cell Lung Cancer Collaborative Group. Chemotherapy in non-small

cell lung cancer: a meta-analysis using updated data on individual patients from 52 randomised clinical trials. BMJ 1995; 311(7010):899–909.

5. American Society of Clinical Oncology. Clinical practice guidelines for the treatment of unresectable non-small-cell lung cancer. J Clin Oncol 1997; 15(8):2996–3018.

6. Macbeth F, Toy E, Coles B, Melville A, Eastwood A. Palliative radiotherapy regimens for non-small cell lung cancer. Cochrane Database Syst Rev 2001;(3):CD002143.

7. Medical Research Council Lung Cancer Working Party. Inoperable non-small-cell lung cancer (NSCLC): a Medical Research Council randomised trial of palliative radiotherapy with two fractions or ten fractions. Br J Cancer 1991; 63(2):265–270.

8. Medical Research Council Lung Cancer Working Party. A Medical Research Council (MRC) randomised trial of palliative radiotherapy with two fractions or a single fraction in patients with inoperable non-small-cell lung cancer (NSCLC) and poor performance status. Br J Cancer 1992; 65(6):934–941.

9. Bezjak A, Dixon P, Brundage M, Tu D, Palmer MJ, Blood P et al. Randomized phase III trial of single versus fractionated thoracic radiation in the palliation of patients with lung cancer (NCIC CTG SC.15). Int J Radiat Oncol Biol Phys 2002; 54(3):719–728.

10. Macbeth FR, Bolger JJ, Hopwood P, Bleehen NM, Cartmell J, Girling DJ et al. Randomized trial of palliative two-fraction versus more intensive 13-fraction radiotherapy for patients with inoperable non-small cell lung cancer and good

Page 106: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

104

performance status. Medical Research Council Lung Cancer Working Party. Clin Oncol 1996; 8(3):167–175.

11. Kramer GW, Wanders SL, Noordijk EM, Vonk EJ, van Houwelingen HC, Van den Hout WB et al. Results of the Dutch National study of the palliative effect of irradiation using two different treatment schemes for non-small-cell lung cancer. J Clin Oncol 2005; 23(13):2962–2970.

12. The EuroQol Group. EuroQol - a new facility for the measurement of health-related quality of life. Health Policy 1990; 16(3):199–208.

13. Dolan P. Modeling valuations for EuroQol health states. Med Care 1997; 35(11):1095–1108.

14. Nyman JA. Should the consumption of survivors be included as a cost in cost-utility analysis? Health Econ 2004; 13(5):417–427.

15. van den Hout WB, van der Linden YM, Steenland E, Wiggenraad RGJ, Kievit J, de Haes H et al. Single- versus multiple-fraction radiotherapy in patients with painful bone metastases: cost-utility analysis based on a randomized trial. J Natl Cancer Inst 2003; 95(3):222–229.

16. van den Berg B, Brouwer W, van Exel J, Koopmanschap M. Economic valuation of informal care: the contingent valuation method applied to informal caregiving. Health Econ 2005; 14(2):169–183.

17. van den Brink M, van den Hout WB, Stiggelbout AM, Putter H, van de Velde CJ, Kievit J. Self-reports of health care utilization: diary or questionnaire? Int J Technol Assess Health Care 2004.

18. Oostenbrink JB, Koopmanschap MA, Rutten FFH. Manual for cost analyses, methods and standard prices for economic evaluations in health care. Amstelveen: Dutch Health Insurance Executive Board, 2000.

19. Oostenbrink JB, Koopmanschap MA, Rutten FF. Standardisation of costs: the Dutch Manual for Costing in economic evaluations. Pharmacoeconomics 2002; 20(7):443–454.

20. Dutch Health Insurance Executive Board. Pharmacotherapeutic Compass 2000/2001. Amstelveen: 2000.

21. Meltzer D. Accounting for future costs in medical cost-effectiveness analysis. J Health Econ 1997; 16(1):33–64.

22. Desgagné A, Castilloux AM, Angers JF, LeLorier J. The use of the bootstrap statistical method for the pharmacoeconomic cost analysis of skewed data. Pharmacoeconomics 1998; 13(5):487–497.

23. Earle CC, Chapman RH, Baker CS, Bell CM, Stone PW, Sandberg EA et al. Systematic overview of cost-utility assessments in oncology. J Clin Oncol 2000; 18(18):3302–3317.

24. Ubel PA, Hirth RA, Chernew ME, Fendrick AM. What is the price of life and why doesn't it increase at the rate of inflation? Arch Intern Med 2003; 163:1637–1641.

25. Gold MR, Siegel JE, Russell LB, Weinstein MC. Cost-effectiveness in Health and Medicine. New York: Oxford University Press, 1996.

26. Seshamani M, Gray AM. A longitudinal study of the effects of age and time to death on hospital costs. J Health Econ 2004; 23(2):217–235.

27. Gandjour A, Lauterbach KW. Does prevention save costs? Considering deferral of the expensive last year of life. J Health Econ 2005; 24(4):715–724.

28. Singer PA, Martin DK, Kelner M. Quality end-of-life care: patients' perspectives. J Am Med Assoc 1999; 281(2):163–168.

Page 107: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

Chapter 8

GENERAL DISCUSSION

Page 108: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

106

Chapter 1 Palliative care is still an important and recurring issue in Non-Small-Cell lung cancer patients.1 Two of our studies were initiated in the nineties, but the results became available only recently, as accrual in these studies was relatively slow and less events in time occurred than expected upfront. However, despite changes in technical possibilities and in treatment strategies, the conclusions drawn and which we will discuss are still valid. Chapter 2 Even nowadays, the majority of patients eventually will face complaints from tumour recurrence or metastases, as even after concommittant chemoradiation complete tumour response is seen in only 26% of the patients.2 In a prospective study, we retreated patients with external beam radiotherapy of 2 x 8 Gy. Those patients were primarely irradiated more than 6 months before and had complaints of an intrathoracical recurrence of a Non-Small-Cell Lung Carcinoma (NSCLC). We conclude, that this approach appears not only to be safe but is also effective in reducing complaints. Palliation persists more than 50% of the remaining life span in the majority of our patients. The effectiveness and safety of retreatment with external beam radiotherapy was also demonstrated in several other studies.3-9 The prediction of success and risks on toxicity of the treatment depends on the type of complaint and treatment scheme.3-9 All these studies mention only the response rates, but none of them the duration of this palliative effect. In our opinion, the latter is an important issue in decision making if it is worthwhile or not offering this palliative treatment to patients. The schedule of 2 x 8 Gy in our study was chosen, because this was not only frequently used for recurrent NSCLC in brachytherapy settings10, but was also introduced as a safe and effective schedule for palliative treatment with external beam irradiation.11 Based on our recent findings (chapter 6) and other studies12-14, a multifractionated treatment scheme however appears to be more effective. Especially, for patients with a recurrence after primary irradiation, without metastasis and in a good general physical condition, Wu et al.8 showed that this approach has to be considered. Certainly, because we have overestimated the risk in exceeding normal tissue tolerances after retreatment, in the past.15-17 To decide on the appropriate schedule, life-expectancy and physical condition are important factors. An estimation of the life-expectancy can be made based on our study, as none of our patients lived longer than the interval between primary treatment and start of the retreatment, a finding also reported by Tada et al.9 Patients with Eastern Cooperative Oncology Group (ECOG) score 3 are no candidates for high dose reirradiation.9 Nevertheless, as numbers of patients are too small for randomized studies and the selection criteria vary in the different studies3-9, the question which treatment scheme has to be preferred, will remain unanswered and has to be individualised. As chemotherapeutic regimens with better response rates and less toxicity became available, the role of reirradiation as palliative treatment became less important, but is still not negligible.

Page 109: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

107

Chapter 3 In the 1990's chemotherapy was also recognized as an effective modality for the treatment of patients with stage IIIA(N2) NSCLC. For long, primary radiotherapy in locally advanced Non-Small-Cell Lung Cancer was the only treatment option, but the results were unsatisfactory.18 During the National Dutch Consensus Meeting in 1997 a national survey showed that most pulmonologists regarded radiotherapy in stage III Non-Small-Cell lung cancer patients as a palliative treatment.19 This attitude was reflected by the substantial number (22%) of patients with stage IIIA NSCLC not referred for any therapy at all.20 Conversely, most of the radiation oncologists were convinced radiotherapy could still have a curative potential for these patients. At that time the term “radical radiotherapy” was introduced.19 The primary intention of this radiation treatment is to obtain persistent local tumour control, with a small chance of cure. After radiotherapy doses up to 60 Gy in 6 weeks, complete remissions were seen in about 12% 21 but the majority of patients developed distant metastases too.22 Meanwhile, chemotherapy had proven to be effective, not only in combination with radiotherapy as senzitizer (EORTC study 08844)23 but also as induction chemotherapy before surgery (the Toronto phase II trial).24 The patients treated with these combined modalities not only had better tumour response rates, but also a gain in survival. It was therefore that the European Organization for the Research and Treatment of Cancer (EORTC) already launched study 08941 in 1994, a randomized phase III trial of surgery versus radiotherapy in patients with stage IIIA Non-Small Cell Lung Cancer (NSCLC) after a response to induction-chemotherapy. Selected patients with histological or cytological proven non-resectable stage IIIA(N2) NSCLC were given 3 cycles of platinum-based induction chemotherapy. Responding patients were then randomized between surgery, i.e. radical resection with lymph node dissection and optional postoperative radiotherapy, or thoracic radiotherapy, i.e. at least 40 Gy in 2 Gy daily fractions on the mediastinum with a boost up to at least 60 Gy on the involved area. In order to observe an increase of 5-year overall survival from 15 (radiotherapy arm) to 25% (surgery arm), 292 events out of 358 randomized patients had to be observed. Secondary endpoints were progression free survival and toxicity.25 The main conclusion of this study is that no significant differences in overall survival nor progression free survival in both treatment arms were seen. Consequently, radiotherapy after induction chemotherapy remained the standard treatment. Especially, in case of ‘no downstaging’ (ypN2) radiotherapy proved to be even superior to surgery.26 So, these patients with ypN2 disease are no longer candidates for surgical intervention. Also for those patients who had to undergo a pneumonectomy, one should always be very reluctant to perform surgery as mortality rates will increase.27-31 A further reason to prefer radiotherapy above surgery after induction chemotherapy is that the toxicity of surgery is probably underestimated as even after curative surgery in stage I and II NSCLC the quality of life is significantly impaired.32 A last reason to prefer radiotherapy above surgery after induction chemotherapy is that after surgery radicality was obtained in only 50% of the resected patients.27 Of those irradically resected, 40% received postoperative radiotherapy (PORT). PORT results in additional toxicity, whereas an influence on overall survival has not been proven and an influence on the disease free survival is debatable, according to the results of the PORT study.33,34 Regrettably, no data exist about QOL after randomisation between surgery and radiotherapy after induction chemotherapy.

Page 110: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

108

A reason for the less favourable outcome of surgery in our study could be that surgery was less effective because of a suboptimal selection. The amount of tumourload in patients with upfront non-resectable stage IIIA(N2) disease varies substantially. This heterogeneity could be the reason of the low radical resection rate in our study even after a response to neoadjuvant treatment.35 With a better selection the number of radical resections could be increased. The subgroup of patients down-staged to ypN0/N1 disease, are considered good candidates for surgery by some authors. This preference for surgery for this subgroup is based on several phase II studies and two small phase-III studies.25 But this could not be confirmed in our study26 and the RTOG study.28-30 Esophageal ultrasound and endobroncheal ultrasound are techniques recently introduced for mediastinal staging.36 Presently the non-invasive PET/CT scans, changes in SUV's (standardized uptake values) from two serial PET/CT scans before and after three chemotherapy cycles or later, could allow prediction of histopathological response in the primary tumor and mediastinal lymph nodes and have prognostic value with a sensitivity and specificity of 73% and 89% in prediction of ypN0 mediastinal lymph nodes.37 As we performed no remediastinoscopy and these new techniques were not available at the start of our study, further studies are needed to elucidate selection criteria and to define the subgroups for which surgery still might play a role in the treatment of stage IIIA(N2) NSCLC. In these studies esophageal ultrasound (EUS), endobroncheal ultrasound (EBUS), remediastinoscopy and/or PET/CT can be used to predict ypN0 patients and these patients should then be randomised for surgical intervention or radiotherapy in combined modality treatments.36,37 In case such a randomised study in downstaged IIIA(N2) NSCLC patients is considered, the radiotherapy treatment has to be revised too. More recent technical developments in planning systems and radiotherapy equipment make it possible to deliver higher treatment dosages. The prescribed tumour dose in our study, might be relatively low and could result in a lower local control. Sibley et al.38 reported high-dose conformal radiotherapy to result in a 2-year survival rate of 37% and a low morbidity rate. These results compare favorably with trials of chemoradiation or conventional radiotherapy. But local tumor progression still remains a problem in 64% of their patients. Socinsky et al.39 in a phase I study showed that dose escalation in combination with chemotherapy to 90 Gy was feasible. The one-year survival rate was 73%. Mean lung dose and V(20), the total volume of the lung receiving more than 20 Gy are the limiting factors in case of dose escalation.40 Bradley et al.40 showed that dose escalation to 83.8 Gy in fractions of 2.15 Gy is safe for a V(20) < 25%, and to 77.4 Gy for a V(20) between 25 and 36% using three-dimensional conformal radiotherapy techniques. With a V20 >37% an excessive risk for the development of pneumonitis was seen. No concurrent chemotherapy was allowed in their study. Based on the results in sequential chemoradiotherapy, dose-intensification in stage IIIA NSCLC patients to 77.4 Gy could be considered in patients with a V(20) less than 36%. The results of the radiotherapy in our study could also be negatively influenced, because of the relatively long interval of 10 weeks (70 days) between the last induction chemotherapy course and the start of the thoracic radiotherapy. This could have had a detrimental effect on the final results. El Sharouni et al.41 showed that with a mean interval between end of chemotherapy and start of radiotherapy of 80.3 days (range 29 – 141 days), 41% of the tumours progressed before the start of the radiotherapy treatment. The estimated mean value of the tumour doubling time in their study was 46 days (range 8.3 – 171 days). O’Rourke et al.42 found that 21% of

Page 111: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

109

their patients became incurable after a delay of 35 – 187 days. In our study the mean interval between last chemotherapy and start of the thoracic radiotherapy was 52 days (17 – 113 days). Only 5 patients (3%) were not treated after randomisation, because of tumour progression or clinical deterioration of the patient. However, no comparison between planning-CT scan and chemotherapy-evaluation-CT scan was done in our study. As long as the tumour stage according to the radiation oncologist remained IIIA, patients were still considered eligible for the study. Therefore, the number of patients with progression of the locoregional tumour after chemotherapy may be underestimated. Also a long overall treatment time could negatively influence the results of the radiotherapy. Continuous Hyperfractionated Accelerated Radiotherapy study (CHART) reduces the overall treatment time to 12 days, which resulted with a lower total dose in a survival rate comparable to that of induction chemotherapy followed by conventional radiotherapy.43 Bonner et al.44 showed a small advantage of HART (hyperfractionated accelerated radiotherapy) with twice-daily fractions till 60 Gy with or without chemotherapy compared with conventional radiotherapy till 60 Gy in 6 weeks. However, the overall treatment time in the HART scheme was as long as in the conventional arm because of a interval of 2 weeks after 30 Gy reducing the potential benefit of the schedule. Belani et al.45 focused on shortening of the overall time in sequential chemoradiation. After two induction cycles of carboplatin-paclitaxel chemotherapy patients were randomized between HART (hyperfractionated accelerated radiotherapy) tid 1.5 Gy till 57.0 Gy in 2.5 weeks and 64 Gy in 32 fractions over 6.5 weeks. No significant difference between both treatment schemes could be detected although there was a trend in favour of the HART scheme. With HART 2- and 3 year survival rates were 44% and 34%, and 24% and 14% in the conventional arm. However, toxicity was more intense in the HART scheme. All these studies support the hypothesis that a further reduction in overall treatment time also of chemoradiotherapy and/or an intensification of the radiation dose also might improve the overall results. A way to obtain a shorter overall treatment time and to reduce the interval between chemotherapy and radiotherapy is to use these modalities concomitantly. In three recent studies46-48 with concurrent chemoradiation, all with different chemotherapeutic regimens and radiation schemes (see Table 1), results appeared superior to sequential chemoradiation treatment. Only Fournel et al.49 showed no significant difference between concurrent chemoradiation (Vineralbine/Cisplatin and conventional radiotherapy with a dose of 66 Gy in 6½ weeks) and sequential chemoradiation. The median survival after sequential chemoradiotherapy was 12–15 months and 15–18 months after concurrent chemoradiotherapy. The acute toxicity however was substantially more severe in the concurrent arm. WHO grade III/IV leucopenia and thrombopenia occurs in sequential chemoradiotherapy in 5 –10 % of the patients and in concurrent chemoradiotherapy in 40–60 %. Acute oesophagitis occurred during sequential chemoradiation in 4% of the patients and during concurrent chemoradiation in 25–30 %. In a metaanalysis50, including stage I-II patients, concurrent chemoradiotherapy reduced the risk of death at two years with 14% compared to sequential chemoradiotherapy, and with 7% compared to radiotherapy alone at the expense of an increase in acute esophagitis. Therefore, Rowell et al.50 advises to be cautious in adopting concurrent chemoradiotherapy as the standard of care because of uncertainties about the true magnitude of benefit in

Page 112: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

110

Abbreviations: N = number of patients; MVP = Mitomycin-Vindesine-cisplatin; VP = Vinblastine-cisplatin; EP = Etoposide-cisplatin; VrP = Vinorelbine-cisplatin; CT = chemotherapeutic regimen; RT = radiotherapeutic regimen. comparison with sequential chemoradiotherapy. With short follow up and uncertainties about toxicity the optimal chemoradiotherapy regimen remains uncertain. The role of surgery after concurrent chemoradiation in resectable stage III NSCLC patients has been investigated in one randomized and one non-randomized study.30,51 The necessity of adding surgery in this setting is also still an item for debate, as no significant differences in survival were seen, but the toxicity in the surgery arm is still too high. Subgroup analysis in the South-West Oncology Group (SWOG) study28,30 revealed that patients who underwent a lobectomy have a better prognosis when adding surgery. However, the majority of patients in the RTOG study had no 3-D planning-CT and no stratification has been done for this type of surgery. So, also these data have to be interpreted with caution. Chapter 4 The survival in both treatment arms of EORTC study 08941 could be less than expected, because of the use of less effective chemotherapy schemes at the start of the study. During the study new chemotherapeutic agents became available with higher response rates. The early suggestions of increased efficacy of the Gemcitabine and cisplatin combination prompted the EORTC Lung Cancer Cooperative Group to initiate a phase II trial to better define the toxicity and activity of this combination as an induction regimen for patients with stage IIIA NSCLC.52 This trial was the first of a series of phase II studies using new chemotherapy combinations within the setting of EORTC study 08941. With this and other new chemotherapeutic agents the overall response rates increased and these regimens have been standard treatment since.53 Nevertheless, the use of all these different induction chemotherapy schemes are of no

Table 1. Sequential chemoradiation compared with concurrent chemoradiation

Survival Toxicity Study N CT RT Schedule

Median % (yr) Hematological Oesophageal

Furuse46 314 MVP 56 Gy/5 ½ wks

Sequential Concurrent

13 17

9 (5) 19 (5)

76 98

Curran47 610 VP

EP

63 Gy/6wks 63 Gy/6 wks 69.2 Gy/bid

Sequential Concurrent Concurrent

14.6 17

15.2

12 (4) 21 (4) 17 (4)

4 25 44

Zatloukal48 102 VrP 60 Gy/6 wk Sequential Concurrent

13 20.4

12 (2) 42 (2)

39.6 64.7

4.2 17.6

Fournel49 205 VrP 66 Gy/6 ½ wks

Sequential

Concurrent

14.5

16.3

26 (2)

19 (3)

14 (4)

39 (2)

25 (3)

21 (4)

88

(6 tox. death)

75

(10 tox. death)

3

32

Page 113: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

111

influence on the final comparison between surgery and radiotherapy after induction chemotherapy, as they were equally divided over both treatment arms. Chapter 5 During the EORTC 08941 study questions were raised about the quality of the data of the radiotherapy. A quality control was not not yet applied in the EORTC Lung Cancer Group at the start of EORTC study 08941 in 1994, and guidelines for writing protocols for clinical trials in radiotherapy were only published in 1995.54 At site visits halfway the study it was concluded, the radiotherapy treatment fields and set up appeared to be correct, but compliance to the protocol was subject to improvement. With the protocol amendment in 1999, the prescribed radiotherapy techniques fulfilled the criteria of modern radiotherapy and compliance improved. After the amendment no major protocol violations were seen. Overall, in the majority of patients treatment showed no or only minor protocol violations. Therefore, we are convinced that the quality of the radiotherapy treatment itself will hardly have any negative impact on the final results of the EORTC study. But our data support the use of strict radiotherapy specifications, dummy runs and quality control from the start of a large study and frequent quality assessments throughout to optimize the compliance. Conclusion chapters 3, 4 and 5 The question whether there will be a subgroup of patients who might profit from surgery after induction chemoradiotherapy remains to be clarified. The higher toxicity of the combination treatments used today, have to be accepted as today these treatments are considered to be curative.55 Presently, radiotherapy after induction chemotherapy with second generation chemotherapy as Gemcitabine in combination with cisplatin is considered to be the standard daily practice for stage IIIA(N2) patients with a good performance score. Chapter 6 For those patients who are no (longer) candidates for curative treatment or for palliative chemotherapy, palliative radiotherapy may be required. The balance between gain, in terms of a reduction of the complaints, and risks, in terms of the increase in toxicity, is the most essential aspect of palliative treatment. In 1999 a national multicenter randomized study compared the efficacy of 2 x 8 Gy versus our standard 10 x 3 Gy in patients with irresectable stage IIIA/B (with Eastern Cooperative Oncology Group score of 3 to 4 and/or substantial weight loss) and stage IV non-small-cell lung cancer. Two hundred ninety-seven patients were eligible and randomized to receive either 10 x 3 Gy or 2 x 8 Gy by external-beam irradiation. The primary endpoint was a patient-assessed score of treatment effect on seven thoracic symptoms. Study sample size was determined based upon an average total symptom score difference of more than one point over the initial 39 weeks post-treatment. The time course of symptom scores were also evaluated, and other secondary endpoints were toxicity and survival. We concluded, the 10 x 3-Gy radiotherapy schedule is preferred over the 2 x 8-Gy schedule for palliative treatment, as it results in a longer duration of the palliative response and improves survival. The assessment of palliation is not standardized. Therefore, interpretation of the results between different studies is difficult. Symptom relief can vary depending the

Page 114: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

112

type, number, and intensity of the complaints.56-58 Therefore stratification and analysis by symptom can be useful. However, stratification for all factors will require large numbers of patients and create practical problems in running such trials. For this reason this is not implemented in our study. We introduced the patient-assessed total symptom score as a new method of how symptoms can be scored. This method has the disadvantage that it provides no information of symptom relief per symptom. Bezjak et al.12 proposed to restrict measurements of palliation only to the index symptom, being the most important complaint to initiate the treatment. The response of the index symptom will be stronger than that of other individual symptoms. These larger changes in an individual patient will therefore not be seen at the group level when the average change was reported. As a result, analyzing the index symptom may be more sensitive to change at the expense of providing inadequate information about the overall palliative effect. Our method has the advantage that it is a good indicator for the overall palliative effect.58 The sum of the score of 7 different symptoms was measured for each patient. We measured not only the reduction of complaints, but also stabilization and occurrence of new complaints due to tumor and/or treatment. Furthermore it was possible to report the duration of palliation for each separate treatment arm. Therefore the frequency in which symptoms are scored is also crucial. Hopwood et al.59 showed that differences in missing information between studies can lead to incorrect conclusions, which may be a consequence of different patterns of change in symptoms during treatment. The differences in the pattern of symptom reduction between both our treatment arms can support this finding. Measuring once or only a few times, or weekly during the first 12 weeks and 2-weekly in the 26 weeks thereafter as in our study, will produce differences in outcome. Therefore, comparing studies is also difficult because the frequency and duration of measurements varies substantially.60 Most striking is the gain in survival for the more protracted treatment scheme in this poor prognostic patient group. This can be a matter of chance, although the level of significance that was detected is considered sufficient.61 What is missing is a comparison of the palliative effect obtained by radiotherapy and by chemotherapy. However, it is not very likely that such a study will ever be performed. Chapter 7 This gain in survival and prolongation of the palliative effect is important for the individual patient, but the costs for society have to be acceptable too. In our cost-effectivity study an estimated gain of 10.7 weeks in life expectancy and 6.8 weeks in QALYs in favour of 10 x 3 Gy is seen. These findings are comparable with the results of chemotherapy in palliative care for patients with PS 2. 62 The extra costs to be made for the multifractionated radiotherapy are not only acceptable from a socio-economical perspective, but are also nearly half of the costs of those to be made by chemotherapy ($ 5236 vs $ 9450). 63 However, the estimation of costs is complex. The conclusion of our economic evaluation strongly hinges on the survival gain, but this has not been a consistent finding in other studies.60 In our analysis we corrected for differences in survival, survival at study closure and last available EuroQol measure, as neglecting the missing data would lead to bias and an underestimation of the costs. We included both related and unrelated health care costs in our base-case analysis, because the QALY measure also included the health gain obtained from unrelated health care. 64 Excluding unrelated health care costs 65, or including not only all future

Page 115: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

113

health care costs, but also future production and consumption costs 66, would still support the preference for the 10×3 Gy schedule. In this study we found that both age and time until death were not statistically significant predictors of cost, while others argued, that costs are more related to the time until death, than to age. 67, 68 Instead, the EQ-5D utility measure was predictive, leading to increased costs towards death and a less than proportional relationship between survival time and costs. If the results in our terminally ill patient population are generalizable, then perhaps age is more predictive for chronic diseases, where quality of life is determined by disease progression, whereas time until death is more predictive in economic models for more acute conditions. Several remarks can be made concerning the validity of our research methods. Perhaps most importantly, because we had not anticipated the survival difference, long-term costs related to the difference in survival had to be estimated from cost data obtained in the initial 12 weeks. Although our model identified short-term costs related to the initial radiotherapy that should not be extrapolated and although during the initial 12 weeks already more than one-third of the patients had died, the cost model may still not have been representative for long-term health care costs associated with living and dying. Without gold standard we had no means to test the validity of the model. A second matter of concern is the used EQ-5D utility measure that is frequently used in economic evaluations from a societal perspective, but does not include quality of life domains that are specifically relevant in the valuation of end-of-life care.69, 70 Unfortunately no valuation instrument exists that incorporates such issues. Nevertheless, the distinct decrease of the EQ-5D utility towards the end of life demonstrates that the instrument is responsive to the changing health status of these patients and the results were very similar to results from other research in terminally ill cancer patients.69 These issues were addressed in the sensitivity analyses, which showed that they had very little influence on the estimated cost-utility ratio, thus suggesting that the obtained results are robust. In conclusion, because of the better treatment outcome in the 10 x 3-Gy arm and acceptable costs, 10 x 3 Gy has become the standard in palliative care in lung cancer in the Netherlands. For a subgroup of patients with minimal survival, 2 x 8 Gy could be an alternative. However, we were not able to elucidate the criteria for this subgroup of patients. Conclusions The best palliative treatment for each individual patient will be merely based on selection criteria. Many patients will not be fit enough to undergo combined treatment modalities, especially not in a concurrent setting. For future trials, the gain in survival will be as important, as will be the reduction of toxicity. Chemotherapy as single modality treatment will be an option for many patients. But even then, loco-regional tumour growth can give complaints and palliative irradiation will be indicated. The changes over time for the various indications evaluated in this thesis are summarized in the table 2.

Page 116: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

114

In this thesis we demonstrated that combined modality of induction chemotherapy followed by radiotherapy has to be the standard treatment for patients ECOG 0–2 non-resectable stage IIIA NSCLC. The role for surgery in this type of patients has still to be defined based on improved selection criteria and response measurements. Strict palliative care for this type of patients is no longer warranted. Based on our study using a total symptom score, we concluded that for those patients not fit enough to undergo chemotherapy with stage III-IV NSCLC and in a bad general condition, the multifractionated treatment scheme of 10 fractions of 3 Gy 4–5 times a week remains the standard treatment in the Netherlands. Using the total symptom score, a significant different pattern in time between the hypofractionated and multifractionated treatment scheme could be shown. The duration of the palliative effect was in favour of the multifractionated scheme, as was the significant gain in survival and for quality-of-life corrected survival. This resulted in significantly higher costs in the multifractionated arm compared to the hypofractiated arm, but still within acceptable costs according to the Dutch standards.

Table 2. Changing perspectives for patients

in palliative care Non-Small-Cell Lung Cancer

Time period, treatment intention and - options

≈1990 ≈1995 ≈2000

Indication Palliative treatment Radical treatment Curative treatment

Stage IIIA (pN2) Expectative,

Palliative radiotherapy;

Start of combined modality trials

(with 2nd generation chemotherapeutics)

Combined modality chemoradiotherapy:

sequential

Combined modality (with 3rd generation chemotherapeutics) chemoradiotherapy:

Sequential, or concurrent Chemo(radiotherapy)-

surgery? Palliative treatment Palliative treatment Palliative treatment

Stage IIIB,IV with complaints of loco-regional tumour (no candidate for chemotherapy)

Best supportive care,

Multifractionated or hypofractionated

irradiation.

Hypofractionated or multifractionated

treatment schemes,

Best supportive care.

Multifractionated treatment schemes,

Best supportive care.

Palliative treatment Palliative treatment Palliative treatment

Recurrent tumour (after primary radiotherapy)

Expectative, Repeat radiotherapy

(seldom)

Repeat radiotherapy (2x8 Gy),

or chemotherapy

Chemotherapy, or repeat radiotherapy with conventional planning

(2 x 8 Gy) or in combination with 3-D

planning (30 x 2 Gy)

Page 117: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

115

For those patients, who develop a recurrent intrathoracical tumour after radical radiation treatment, we showed that repeat external beam treatment 2 x 8 Gy is a safe and effective palliative treatment. With the treatment equipment and planning-systems of today, higher dosages in repeat radiotherapy have to be considered. References 1. American Society of Clinical Oncology. Special article: clinical practice

guidelines for treatment of unresectable non-small cell lung cancer. J Clin Oncol 1997;15:2996–3018.

2. Eberhardt W, Wilke H, Stamatos M, et al. Preoperative chemotherapy followed by concurrent chemoradiotherapy based on hyperfractionated accelerated radiotherapy and definitive surgery in locally advanced non-small-cell lung cancer: mature results of a phase II trial. J Clin Oncol 1998;16:622–634.

3. Green N, Melbye RW. Lung cancer: Retreatment of local recurrence after definitive irradiation. Cancer 1982;49:865–868.

4. Jackson MA, Ball DL. Palliative retreatment of locally recurrent lung cancer after radical radiotherapy. Med J Aust 1987;147:391–394.

5. Montebello JF, Aron BS, Manatunga AK, et al. The reirradiation of recurrent bronchogenic carcinoma with external beam irradiation. Am J Clin Oncol 1993;16:482–488.

6. Gressen EL, Werner-Wasik M, Cohn J, et al. Thoracic reirradiation for symptomatic relief after prior radiotherapeutic management for lung cancer. Am J Clin Oncol 2000;23:160–163.

7. Okamoto Y, Murakami M, Yoden E, et al. Reirradiation for locally recurrent lung cancer previously treated with radiation therapy. Int J Radiat Oncol Biol Phys 2002;52:390–396.

8. Wu K-L, Jiang G-L, Qian H et al. Three-dimensional conformal radiotherapy for locoregionally recurrent lung carcinoma after external beam irradiation: a prospective phase I-II clinical trial. Int J Radiat Oncol Biol Phys 2003;57:1345–1350.

9. Tada T, Fukuda H, Matsui K et al. Non-small-cell lung cancer: reirradiation for loco-regional relapse previously treated with radiation therapy. Int J Clin Oncol 2005;10:247–250.

10. Speiser B, Spratling L. Intermediate dose rate remote afterloading brachytherapy for intraluminal control of bronchogenic carcinoma. Int J Radiat Oncol Biol Phys 1990;18:1443–1448.

11. Bleehen NM, Girling DJ, Fayers PM, et al. Inoperable non-small-cell lung cancer (NSCLC): a Medical Research Council randomised trial of palliative radiotherapy with two fractions or ten fractions. Br J Cancer 1991;63:265–270.

12. Bezjak A, Dixon P, Brundage M, et al. Randomized study of single versus fractionated radiotherapy (RT) in the palliation of non-small cell lung cancer. Int J Radiat Oncol Biol Phys 2002;54:7194–728.

13. Macbeth FR, Bolger JJ, Hopwood P, et al. Randomized trial of palliative two-fraction versus more intensive 13-fraction radiotherapy for patients with inoperable non-small cell lung cancer and good performance status. Clinical Oncology 1996;8:167–175.

Page 118: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

116

14. Reinfuss M, Glinski B, Kowalska T. Radiothérapie du cancer bronchique non à petites cellules de stade III, inopérable, asymptomatique. Résultats définitifs d’un essai prospectif randomize (240 patients). Cancer/Radiotherap 1999;3:475–479.

15. Ang KK, Jiang G-L, Feng Y, et al. Extent and kinetics of recovery of occult spinal cord injury. Int J Radiat Oncol Biol Phys 2001;50:1013–1020.

16. Fowler JF, Bentzen SM, Bond SJ, et al. Clinical radiation doses for spinal cord: the 1998 international questionnaire. Radiother Oncol 2000;55:295–300.

17. Poltinnikov IM, Fallon K, Xiao Y, et al. Combination of longitudinal and circumferential three-dimensial esophageal dose distribution predicts acute esophagitis in hypofractionated reirradiation of patients with non-small-cell lung cancer treated in stereotactic body frame. Int J Radiat Oncol Biol Phys 2005;62:652–658.

18. Janssen-Heijnen MLG, Dijck JAAM van, Siesling S, et al. Lung cancer in the Netherlands in the period 1989-1997: the epidemic is not over yet. (original in Dutch). Ned Tijdschr Geneesk 2001;145:419–23.

19. Festen J. Guideline ‘Radiotherapy in non-small-cell lung carcinoma. Working Group, National Organization for Quality Assurance in Hospitals, Netherlands’. Ned Tijdschr Geneeskd. 1998;142:2248–51.

20. van Dijck JA, Festen J, de Kleijn EM, Kramer GW, et al. Treatment and survival of patients with non-small cell lung cancer Stage IIIA diagnosed in 1989-1994: a study in the region of the Comprehensive Cancer Centre East, The Netherlands. Lung Cancer 2001;34:19–27.

21. Arriagada R, Le Chevalier T, Quoix E, et al. for the GETCB, FNCLCC and the CEBI trialists. ASTRO plenary –– Effect of chemotherapy on locally advanced non-small cell lung carcinoma: A randomized study of 353 patients. Int J Radiat Oncol Biol Phys 1991;20:1183–1190.

22. Perez CA, Pajak TF, Rubin P, et al. Long-term observations of the patterns of failure in patients with unresectable non-oat cell carcinoma of the lung treated with definitive radiotherapy: Report by the Radiation Therapy Oncology Group. Cancer 1987;59:1874–1881.

23. Schaake-Koning C, van den Bogaert W, Dalesio O, et al. Radiosensitization by cytotoxic drugs. The EORTC experience by the Radiotherapy and Lung Cancer Cooperative Groups. Lung Cancer. 1994;10(Suppl 1):S263–70.

24. Burkes RL, Ginsberg RJ, Shepherd FA et al. Induction chemotherapy with mitomycin, vindesine, and cisplatin for stage III unresectable non-small-cell lung cancer: results of the Toronto Phase II Trial. J Clin Oncol 1992;10:580–586.

25. Splinter T, van Schil P, Kramer G, et al. Randomized trial of surgery versus radiotherapy in patients with stage IIIA(N2) non-small cell lung cancer after a response to induction chemotherapy. EORTC 08941. Clin Lung Cancer 2000;2:69–72.

26. van Meerbeeck J, Kramer G, Legrand C, et al. Does downstaging in patients (pts) with IIIA-N2 non-small cell lung cancer (NSCLC) and a response to induction chemotherapy (ICT) influence outcome with surgery (S) or radiotherapy (RT)? An exploratory analysis of EORTC 08941. Proc. American Society of Clinical Oncology (ASCO), Atlanta, Georgia, U.S.A., June 2-6, 2006 (abs. 7047).

Page 119: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

117

27. van Schil P, van Meerbeeck J, Kramer G, et al. Morbidity and Mortality in the Surgery Arm of EORTC 08941 Trial. European Respiratory Journal,2005;26:1–6.

28. Albain KS. Proc Am Soc Clin Oncol 2005;624S (abstract 7014). 29. Rusch V. Lung Cancer 2005;49(32):S15 (abstract O-035). 30. Albain KS, Rusch VW, Crowley JJ, et al. Concurrent cisplatin/etoposide plus

chest radiotherapy followed by surgery for stages IIIA (N2) and IIIB non-small-cell lung cancer: mature results of Southwest Oncology Group phase II study 8805. J Clin Oncol. 1995;13:1880–1892.

31. Doddoli C, Barlesi F, Trousse D, et al. One hundred consecutive pneumonectomies after induction therapy for non-small cell lung cancer: an uncertain balance between risks and benefits. J Thorac Cardiovasc Surg 2005;130:416–25.

32. Handy JR, Asaph JW, Skokan L, et al. What happens to patients undergoing Lung Cancer surgery? Outcomes and quality of life before and after surgery Chest 2002;122:21–30.

33. PORT meta-analysis Trialists Group. Postoperative radiotherapy in non-small cell lung cancer: systematic review and meta-analysis of individual patient data from nine randomized controlled trials. Lancet 1998;352:257–263.

34. Burdett S, Stewart L. on behalf of the PORT Meta-analysis Group. Postoperative radiotherapy in non-small-cell lung cancer: update of an individual patient data meta-analysis. Lung Cancer 2005;47:81–83.

35. Andre F, Grunenwald D, Pignon JP, et al. Survival of patients with resected N2 non-small-cell lung cancer: evidence for a subclassification and implications. J Clin Oncol 2000;18:2981–2989.

36. Herth FJF. Mediastinal staging – the role of endobronchial and endo-oesophageal sonographic guided needle aspiration. Lung Cancer 2004;45(S2):S63–S67.

37. Pottgen C, Levegrun S, Theegarten D, Marnitz S, Grehl S, Pink R, Eberhardt W, Stamatis G, Gauler T, Antoch G, Bockisch A, Stuschke M. Value of 18F-Fluoro-2-Deoxy-D-Glucose-Positron Emission Tomography/Computed Tomography in Non-Small-Cell Lung Cancer for Prediction of Pathologic Response and Times to Relapse after Neoadjuvant Chemoradiotherapy. Clin Cancer Res. 2006;12:97–106.

38. Sibley GS, Mundt AJ, Shapiro MD, et al. The treatment of stage III non-small cell lung cancer using high dose conformal radiotherapy. Int J Rad Onc Biol Phys 1995;33:1001–1007.

39. Socinski MA, Morris DE, Halle JS, et al. Induction and concurrent chemotherapy with high-dose thoracic conformal radiation therapy in unresectable stage IIIA and IIIB non-small-cell lung cancer: a dose-escalation phase I trial. J Clin Oncol 2004;22:4341–4350.

40. Bradley J, Graham MV, Winter K, et al. Toxicity and outcome results of RTOG 9311: a phase I-II dose-escalation study using three-dimensial conformal radiotherapy in patients with inoperable non-small-cell lung carcinoma. Int J Rad Oncol Biol Phys. 2005;61:318–328.

41. El Sharouni SY, Kal HB, Battermann JJ. Accelerated regrowth of non-small-cell lung tumours after induction chemotherapy. Br J Cancer 2003;89:2184–9.

42. O’Rourke N, Edwards R. Lung cancer treatment waiting times and tumour growth. Clin Oncol (R Coll Radiol) 2000;12:141–4.

Page 120: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

118

43. Saunders M, Dische S, Barrett A, et al. Continuous hyperfractionated, accelerated radiotherapy (CHART) versus conventional radiotherapy in non-small cell lung cancer: mature data from the randomised multicentre trial. CHART steering committee. Radiother Oncol. 1999;52:137–148.

44. Bonner JA, McGinnis WL, Stella PJ, et al. The possible advantage of hyperfractionated thoracic radiotherapy in the treatment of locally advanced non small cell lung carcinoma: results of a North Central Cancer Treatment Group Phase III study. Cancer 1998;82:1037–1048.

45. Belani CP, Wang W, Johnson DH, et al. Phase III Study of the Eastern Cooperative Oncology Group (ECOG 2597): Induction Chemotherapy Followed by Either Standard Thoracic Radiotherapy or hyperfractionated accelerated radiotherapy for patients with unresectable stage IIIA and B Non-Small-Cell Lung Cancer.J Clin Oncol 2005;23:1–8.

46. Furuse K, for the West Japan Lung Cancer Group. Phase III study of concurrent versus sequential thoracic radiotherapy in combination with mitomycin, vindesine, and cisplatin in unresectable stage III non-small cell lung cancer. J Clin Oncol 1999;17:2692–2699.

47. Curran WJ, Scott CB, Langer CJ. Long-term benefit is observed in a phase III comparison of sequential vs concurrent chemoradiation for patients with unresected stage III NSCLC: RTOG 9410. Proc Am Soc Clin Oncol 2003:2499.

48. Zatloukal P, Petruzelka L, Zemanova M., et al. Concurrent versus sequential radiochemotherapy with cisplatin and vinorelbine in locally advanced non-small cell lung cancer: a randomized study. Lung Cancer 2004;46:87–98.

49. Fournel P, Robinet G, Thomas P, et al. Randomized phase III trial of sequential chemoradiotherapy compared with concurrent chemoradiotherapy in locally advanced non-small-cell lung cancer: Groupe Lyon-Saint-Etienne d'Oncologie Thoracique-Groupe Francais de Pneumo-Cancerologie NPC 95-01 Study. J Clin Oncol 2005;23:5910–5917.

50. Rowell NP, O’Rourke NP. Concurrent chemoradiotherapy in non-small cell lung cancer. Cochrane Database Syst Rev. 2004 Oct 18;(4):CD002140.

51. Taylor NA, Liao ZX, Cox JD, et al. Equivalent outcome of patients with clinical stage IIIA non-small-cell lung cancer treated with concurrent chemoradiation compared with induction chemotherapy followed by surgical resection. Int J Radiat Oncol Biol Phys 2004;58:204–212.

52. Cardenal F, Lopez-Cabrerizo MP, Anton A et al. Randomized phase III study of gemcitabine-cisplatin versus etoposide-cisplatin in the treatment of locally advanced or metastatic non-small-cell lung cancer. J Clin Oncol 1999;17:12–18.

53. van Meerbeeck JP, Kramer GWPM, van Schil P et al. Induction chemotherapy (CT) in stage IIIA-N2 non-small cell lung cancer (NSCLC): an analysis of different regimens used in EORTC 08941. Lung Cancer 2003;41:S79.

54. Bolla M, Bartelink H, Garavaglia G, et al. EORTC guidelines for writing protocols for clinical trials of radiotherapy. Radiother Oncol 1995;36:1–8.

55. van Meerbeeck JP, Koning CC, Tjan-Heijnen VC, et al. Guideline on ‘non-small cell lung carcinoma; staging and treatment’. (originally in Dutch). Ned tijdschr Geneeskd. 2005;149:72–77.

56. Rees GJG, Devrell CE, Barley VL, et al. Palliative Radiotherapy for Lung Cancer: Two Versus Five Fractions. Clinical Oncology 1997;9:90–95.

Page 121: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

119

57. Kramer GW, Gans S, Ullmann E, et al. Hypofractionated external beam radiotherapy as retreatment for symptomatic non-small-cell lung carcinoma: an effective treatment? Int J Rad Onc Biol Phys 2004;58:1388–1399.

58. Stephens RJ, Hopwood P, Girling DJ. Defining and analyzing symptom palliation in cancer clinical trials: a deceptively difficult exercise. Br J Cancer 1999;79:538–544.

59. Hopwood P, Stephens RJ, Machin D, et al. Approaches to the analysis of quality of life data: Experiences gained from a Medical research Council Lung Cancer Working Party palliaiteve chemotherapy trial. Qual Life Res. 1994;3:339–352.

60. Macbeth F, Toy E, Coles B, et al. Palliative radiotherapy regimens for non-small cell lung cancer: Cochrane Database Syst. Rev 1:CD002143,2002.

61. Sterne A., Smith G: Sifting the evidence – what’s wrong with significance tests? BMJ 2001;322:226–231.

62. Blackhall FH, Bhosle J, Thatcher N. Chemotherapy for advanced non-small cell lung cancer patients with performance status 2. Curr Opin Oncol. 2005;17:135–139.

63. Maslove L, Gower N, Spiro S, Rudd R, Stephens R, West P. Estimation of the additional costs of chemotherapy for patients with advanced non-small cell lung cancer.Thorax. 2005 Jul;60(7):564–9.

64. Nyman JA. Should the consumption of survivors be included as a cost in cost-utility analysis? Health Econ 2004;13:417–427.

65. Gold MR,Siegel JE, Russell LB et al. Cost-effectiveness in health and medicine. New York: Oxford University Press, 1996.

66. Meltzer D. Accounting for future costs in medical cost-effectiveness analysis. J Health Econ 1997;16:33–64.

67. Seshamani M, Gray AM. A longitudinal study of the effects of age and time to death on hospital costs. J Health Econ 2004;23:217–235.

68. Gandjour A, Lauterbach KW. Does prevention save costs? Considering deferral of the expensive last year of life. J Health Econ 2005;24:715–724.

69. van den Hout WB, van der Linden YM, Steenland E, et al. Single – versus multiple-fraction radiotherapy in patients with painful bone metastasis: cost-utility analysis base don a randomized trial. J Natl Cancer Inst 2003;95:222–229.

70. Singer PA, Martin DK, Kelner M. Quality end-of-life care: patients’perspectives. J Am Med Assoc 1999;281:163–168.

Page 122: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten
Page 123: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

121

Summary In the Introduction of this thesis the poor outlook for patients with a Non-Small-Cell Lung Cancer (NSCLC) is discussed. Most patients are only candidates for a palliative treatment approach. Treatment options are limited, because of former treatments, poor general condition of the patient, or tumour load and/or tumour stage. Despite former radiotherapy, repeated external beam irradiation is an option in case complaints are caused by tumor regrowth (chapter 2). Relief of hemoptysis and Superior Vena Cava Syndrome (SVCS) could be obtained in all evaluable cases. This treatment was less effective for coughing (67%) and dyspnea (35%). The overall median duration of these effects was 4 months. Although a relatively short time of palliation, it meant the complaints are precluded over more than 50% of the patients remaining lifespan. Acute toxicity is very mild. Complications were more frequent with 5 tumor related fatal hemoptysis (17%) and one death due to a broncho-esophageal fistula (4%). Despite these findings, repeated external beam radiotherapy is effective as a palliative treatment for local complaints taking into account the poor prognosis and limited treatment options for this particular population. Nowadays, this treatment option is a part of our regular practice and is implemented in our treatment protocol. For patients in good general condition, chemotherapy gives further opportunities in palliative treatment. Patients with non-resectable stage IIIA(N2) were treated after a response on induction chemotherapy with radiotherapy or surgery (chapter 3). After induction chemotherapy, a reduction in distant metastasis was expected and therefore locoregional control became a major issue. For stage I and II, surgery is the standard above radiotherapy based on historical data. For stage III, radiotherapy was the standard in the Netherlands. It was expected, that surgery after a response to induction chemotherapy showed a decrease of locoregional failure and a better chance of survival compared to radiotherapy. However, this could not be supported by the findings in our study, as no differences in overall, nor progression free survival were found. An underlying cause could be the inhomogeneity of tumor load in stage IIIA(N2) NSCLC patients, as well as the lack of adequate restaging procedures. After surgery only 50% of the resections proved to be radical and 40% of the irradically resected patients were referred for postoperative radiotherapy. Therefore, in stage IIIA(N2) radiotherapy after induction chemotherapy has become the standard treatment in the Netherlands. Search for new treatment combinations led to the use of Gemcytabin® and Cisplatin as induction chemotherapy in patients with stage IIIA(N2) NSCLC with a high response rate of 70.2% (chapter 4). The toxicity was acceptable and mainly hematological. Because of the better response rate compared to former chemotherapy regimens and acceptable toxicity, the Gemcitabine®-Cisplatin regimen has since become common practice in combination treatments in the region of the Comprehensive Cancer Centre East. This gain in effectiveness could explain the better overall survival for the patients in our study. However, the kind of chemotherapy scheme will be of no influence on the comparison between surgery and radiotherapy after induction chemotherapy as these schemes were equally divided over both treatment arms.

Page 124: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

122

Quality assurance of radiotherapy descriptions at start of EORTC study 08941 was not performed. In 1999 radiotherapy data were tightened for: time interval between last chemotherapy course and start of thoracic radiotherapy, the use of a 3-D planning CT, dose and fractionation scheme to the primary tumour, the involved - and uninvolved mediastinum, and duration of radiotherapy. A significant improvement of all quality parameters was noted, except for the interval between last chemotherapy course and start of thoracic radiotherapy, which decreased slightly. The latter reflects logistic problems rather than poor compliance (chapter 5). For future trials, the EORTC guidelines for writing protocols for clinical trials for radiotherapy of the EORTC Radiotherapy Group have to be used. Control of protocol prescriptions by dummy runs will enhance protocol compliance. The relatively low total tumour dose, and the allowed interval of 10 weeks after last induction chemotherapy course, could cause a decrease in overall survival or progression free survival. Patients, who are no candidates for any chemotherapeutic regimen, can be referred for palliative radiotherapy. Many of these patients will have a poor general condition. Because of the relatively shortage of radiotherapy facilities, it was assumed for this particular group of patients that no differences would be found between 10 x 3 Gy and 2 x 8 Gy for palliation as for prognosis (chapter 6). Both treatment arms were equally effective, as the average total symptom score over the initial 39 weeks did not differ. However, the pattern of these scores over time differed significantly. Palliation in the 10 x 3-Gy arm was more prolonged (until week 22) with less worsening symptoms than in the 2 x 8-Gy arm. Survival in the 10 x 3 Gy-arm was significantly better than in the 2 x 8 Gy-arm with a 1-year survival of 19.6% versus 10.9%. Life Expectance (LE) and for Quality of Life corrected survival (QALE) differed significantly in advantage of the 10 x 3 Gy-scheme (chapter 7). Despite the fact that costs for the 10 x 3-Gy arm were significantly higher, they were still acceptable according to the Dutch Standard. In the Netherlands for patients with NSCLC in poor general condition, and not eligible for chemotherapy, 10 x 3 Gy has become the standard. For the poor prognostic group with minimal survival, no standard care has yet determined.

Page 125: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

123

Samenvatting In de Inleiding van dit proefschrift wordt de slechte prognose belicht van patiënten met een niet-kleincellige vorm van longkanker. De meeste patiënten komen slechts voor palliatieve (op klachtengerichte, en niet op levensverlenging beogende) behandeling in aanmerking. De behandelingsmogelijkheden zijn beperkt, hetzij door reeds gegeven behandelingen, hetzij door de slechte algemene conditie van de patiënt, hetzij door de uitbreiding van de kanker zelf. Ondanks eerdere bestralingsbehandeling, is herbestraling te overwegen in geval dat een hernieuwde groei van het gezwel gepaard gaat met klachten (hoofdstuk 2). Klachten als bloed ophoesten (hemoptoë) en het vena cava superior syndroom (VCSS), te herkennen door stuwing van de vaten in hals en gelaat, ontstaan door druk op de lichaamsader in de borstholte door het gezwel zelf en/of door uitzaaiingen in de lymfklieren, konden in de meeste gevallen goed worden bestreden. Deze behandeling had minder succes bij de bestrijding van hoestklachten (67%) en kortademigheid ofwel dyspnoe (35%). De mediane duur van klachtenvermindering was 4 maanden. Hoewel deze duur relatief kort is, betekent dit toch dat hernieuwde klachten konden worden voorkomen gedurende meer dan 50% van de resterende levensduur van de patiënten. De acute bijwerkingen waren mild. Complicaties traden op in de vorm van 5 fatale bloedingen (17%), in combinatie met hernieuwde uitgroei van het kankergezwel, en 1 patient overleed ten gevolge van een fistel, die tussen de luchtpijp en de slokdarm ontstond (4%) zonder dat sprake was van hernieuwde groei van het kankergezwel. Ondanks deze bevindingen beschouwen we herbestraling als een effectieve en veilige behandeling voor locale klachten ten gevolge van een hernieuwde groei van het kankergezwel, gelet op de slechte prognose en de beperkte behandelingsmogelijkheden voor deze patiëntengroep. Deze behandelmogelijkheid maakt tegenwoordig vast onderdeel uit van onze behandelingsrichtlijn. Chemotherapie geeft extra mogelijkheden voor patiënten in een goede algemene conditie. Indien de tumor kleiner is geworden na behandeling met chemotherapie (inductiechemotherapie genoemd) bij patiënten met een stadium IIIA(N2) niet-kleincellige longkanker (dwz alleen aan dezelfde zijde als waar de tumor is gelegen in de borstholte zijn ook uitzaaiingen in de lymfklieren aanwezig) werd aansluitend geloot tussen óf een chirurgische, óf een bestralingsbehandeling (hoofdstuk 3). Door de inductiechemotherapie zouden microscopisch kleine (niet met verder onderzoek zichtbare) uitzaaiingen elders in het lichaam mogelijk alsnog kunnen worden bestreden. Omdat de patiënt dan minder kans heeft om te overlijden aan uitzaaiingen, is het van des te meer belang dat het gezwel en de lymfklieruitzaaiingen blijvend worden uitgeroeid. Voor kleine tumoren zonder of met beperkte lymfklieruitzaaiingen (stadium I en II) vormt chirurgie de standaardbehandeling en vindt radiotherapie alleen plaats als chirurgie niet mogelijk of niet gewenst is. Voor stadium III was radiotherapie de standaardbehandeling in Nederland. Deze opties zijn hoofdzakelijk gebaseerd op historische gegevens en niet gestaafd met gerandomiseerd (via loting bepaald) onderzoek. Wij verwachtten met ons onderzoek aan te tonen dat een chirurgische behandeling vergeleken met een bestralingsbehandeling, na een tumorverkleining ten gevolge van de inductiechemotherapie, een grotere kans op het definitief uitroeien van de longkanker zou geven en daardoor ook de kans op overleving zou doen toenemen.

Page 126: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

124

Ons onderzoek wees echter anders uit, omdat geen verschil in hernieuwde uitgroei van de longkanker, noch verlenging van de ziektevrije periode konden worden aangetoond. Omdat de hoeveelheid en uitbreiding van de kanker in dit stadium enorm kan verschillen, zou dit één van de redenen kunnen zijn dat we geen verschil hebben gevonden. Ook het ontbreken van goede herstadiëring (het preciezer in kaart brengen van de tumorreactie na inductiechemotherapie) kan daarbij een rol hebben gespeeld. Na chirurgie bleek slechts bij 50% van de patiënten het gezwel volledig verwijderd te kunnen worden. Tevens onderging 40% van deze geopereerde patiënten alsnog een bestraling. Bovendien overleden 8% van de patiënten bij wie de gehele long moest worden verwijderd in aansluiting op de operatie. Op basis van deze gegevens lijkt bestraling aansluitend aan inductiechemotherapie bij patiënten met stadium IIIA, niet-kleincellig type, longkanker nog steeds de beste behandeloptie. Onderzoek naar nieuwe geneesmiddelen leidde tot het gebruik van Gemcytabine® en Cisplatinum voor inductiechemotherapie behandeling bij patiënten met stadium IIIA, niet-kleincellig type, longkanker (hoofdstuk 4). Een verkleining van het gezwel in 70% van de behandelde patiënten was nog niet eerder waargenomen. De bijwerkingen waren acceptabel en beperkten zich hoofdzakelijk tot een verandering in het bloedbeeld. Sindsdien wordt deze combinatie standaard gebruikt in onze IKO (Integraal Kankercentrum Oost-Nederland) regio. Deze verbeterde effectiviteit zou kunnen leiden tot een betere overleving van de patiënten in onze studie. Omdat deze behandeling gelijkelijk over beide behandelingsmogelijkheden (chirurgie en bestraling) is verdeeld, zal dit geen effect hebben op het aantonen van een eventueel verschil tussen de effectiviteit van deze beide behandelingen. Kwaliteitscontroles op de bestralingsuitvoering en –voorschriften werden bij de start in 1994 van EORTC studie 08941 nog niet standaard uitgevoerd. In 1999 werden de bestralingsvoorschriften verscherpt voor: het tijdinterval tussen de laatste chemotherapiebehandeling en de start van de bestraling op de borstholte, het gebruik van 3-dimensionale (3-D) planning systemen, doserings- en fractioneringsschema zowel op de longtumor zelf, als de aangedane en niet-aangedane lymfklierregio’s in de borstholte, en de totale duur van de bestralingsbehandeling. Een duidelijke verbetering van alle kwaliteitskenmerken werd bereikt met uitzondering van de beperking van het tijdinterval tussen chemotherapie en start van de bestralingsbehandeling. Dit laatste was eerder te wijten aan logistieke problemen dan aan het niet willen voldoen aan de studievoorschriften (hoofdstuk 5). Bij toekomstige studies zullen de richtlijnen van de EORTC voor het schrijven van protocollen ten behoeve van klinische studies nauwgezet worden gevolgd. Het opvolgen van deze voorschriften wordt gecontroleerd door allereerst een beoordeling van een proefbehandeling (dummy run). Dit zal de betrouwbaarheid van de resultaten ten goede komen. De relatief lage dosis toegepast op het longgezwel en het toestaan van een interval van 10 weken tussen het einde van de inductiechemotherapie en start van de bestraling, zouden een afname van de overlevingskansen en de ziektevrije periode kunnen hebben bewerkstelligd. Indien patiënten geen chemotherapeutische behandeling kunnen ondergaan, kunnen zij worden verwezen voor een palliatieve bestralingsbehandeling. De meeste van deze patiënten zullen een slechte algemene conditie hebben. Omdat er een tekort aan bestralingscapaciteit aanwezig was, meenden wij dat voor deze specifieke groep patiënten een behandeling met 2 hoge bestralingsfracties (2x8Gy) net zo goed zou zijn

Page 127: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

125

als 10 relatief lage bestralingsfracties(10x3 Gy), zowel voor de klachtenbestrijding (palliatie) als voor de levensverwachting (hoofdstuk 6). Beide behandelingsmogelijkheden bleken inderdaad even effectief in het doen afnemen van de klachten, aangezien de door ons gebruikte totale symptoomscore gemiddeld over 39 weken geen verschil gaf te zien. Opvallend was echter wel, dat de afname in de 10x3 Gy behandeling beduidend langer aanhield (tot week 22) en de toename van de symptomen na aanvang van de behandeling beperkter bleef. Ook de kans om 1 jaar te overleven in de 10x3 Gy arm (19,6%) was beduidend langer dan in de 2x8 Gy arm (10,9%). De levensverwachting (LE) en de voor kwaliteit van leven gecorrigeerde overleving (QALE) verschilden beduidend in het voordeel van de 10x3 Gy behandeling (hoofdstuk 7). Ondanks de beduidend hogere kosten van de 10x3Gy behandeling, bleven deze acceptabel gemeten naar de in Nederland geldende normen. Voor Nederland geldt nu dat de 10x3 Gy behandeling de standaardbehandeling dient te zijn. De selectiecriteria voor patiënten in een zeer slechte conditie en minimale overlevingskansen, die eventueel toch met 2x8 Gy behandeld zouden kunnen worden, kunnen we op basis van onze studie niet aangeven.

Page 128: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten
Page 129: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

127

OVERIGE PUBLICATIES Artikelen Kazem I, Jongerius CM, Laquet LK, Kramer GWPM, Huygen P: Evaluation of short course preoperative irradiation in the treatment of resectable bronchuscarcinoma; long term analysis of a randomised pilot study. Int.J.Rad.Onc.Biol.Phys. 1984;10:981-985. Kramer GWPM, Szabo BG: Radiotherapie bij longkanker, Regionaal Nieuwsblad IKN, 5e jaargang, sept. 1986. Kramer GWPM, Rutten E, Slooff J: Zeldzame oorzaak van een postsacrale zwelling, Acta Belgica Chirurgica 1987;87:259-261. Kramer GWPM, Rutten E, Slooff J: Subcutaneous sacrococcygeal myxopapillary ependymoma with lymphnode metastasis, a case report. J. Neurosurgery 1988;68: 474-477. Kramer G, Gans S, Rijnders A, Leer J. Long Term Survival of a patiënt with malignant pleural mesothelioma as a late complication of radiotherapy for Hodgkin’s disease treated with 90Yttrium-silicate. Lung Cancer 2000;27:205-208. Splinter TAW, van Schil PE, Kramer GW, van Meerbeeck JP, Gregor A, Rocmans P, Kirkpatrick A. Randomized Trial of Surgery Versus Radiotherapy in Patients with Stage IIIA (N2) Non–Small-Cell Lung Cancer After a Response to Induction Chemotherapy. EORTC 08941. Clin Lung Cancer 2000;2:69-72; discussion 73 Van Dijck JA, Festen J, de Kleijn EM, Kramer GW, Tjan-Heijnen VC, Verbeek AL; Working Group on Lung Cancer of the Comprehensive Cancer Centre East. Treatment and survival of patients with non-small cell lung cancer Stage IIIA diagnosed in 1989-1994: a study in the region of the Comprehensive Cancer Centre East, The Netherlands. Lung Cancer 2001;34:19-27 De Rijke JM, Schouten LJ, Ten Velde GP, Wanders SL, Bollen EC, Lalisang RI, Van Dijck JA, Kramer GW, van den Brandt PA. Influence of age, comorbidity and performance status on the choice of treatment for patients with non-small cell lung cancer; results of a population-based study. Lung Cancer 2004;46:233-45. Van Schil PE, van Meerbeeck JP, Kramer GW, Splinter TA, Legrand CL, Giaccone G, Manegold C, van Zandwijk N. Morbidity and mortality in the surgery arm of EORTC 08941 trial. Eur Respir J. 2005;26:192-197. Steenbakkers RJ, Duppen JC, Fitton I, Deurloo KE, Zijp L, Uitterhoeve AL, Rodrigus PT, Kramer GW, Bussink J, Jaeger KD, Belderbos JS, Hart AA, Nowak PJ, Herk M, Rasch CR. Observer variation in target volume delineation of lung cancer related to radiation oncologist-computer interaction: A 'Big Brother' evaluation.Radiother Oncol. 2005;77:182-190.

Page 130: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

128

Steenbakkers RJ, Duppen JC, Fitton I, Deurloo KE, Zijp LJ, Comans EF, Uitterhoeve AL, Rodrigus PT, Kramer GW, Bussink J, De Jaeger K, Belderbos JS, Nowak PJ, van Herk M, Rasch CR. Reduction of observer variation using matched CT-PET for lung cancer delineation: a three-dimensional analysis. Int J Radiat Oncol Biol Phys 2006;64:435-448. Abstracts Kramer GWPM, Gans SJ, Ullmann EF. “External re-irradiation of non small cell lung cancer (NSCLC); a pilot study”. Lung Cancer 1994;11(S1):135,517#. van Zandwijk N, Crino L, Kramer GW et al. Phase II study of gemcitabine plus cisplatin as induction regimen for patientw with stage IIIA non-small cell lung cancer by the EORTC Lung Cancer Cooperative Group (EORTC 08955) Lung Cancer 1998;17(S2):486a,1799#. Kramer GWPM, Gans SJ, Ullmann EF, Leer J-W. Reirradiation with external beam treatment of Non-Small Cell Lung Carcinoma. Radiotherapy & Oncol 2002;64:S255,P859. Kramer GWPM. Eindrapportage van de landelijke studie naar het effect van bestraling met verschillende bestralingsschema’s voor het niet-kleincellig longcarcinoom. Verenigingsverslag NTvG 2003;147(20):992. Kramer GWPM, Wanders SL, Noordijk EM, Vonk EJA, Uitterhoeve ALJ, Bussink J, van de Leest AHD,. van Houwelingen JC, van den Hout WB, Leer J-WH. Randomized Dutch National study of the effect of irradiation with different treatment schemes in the palliation of Non-Small-Cell Lung-Cancer (NSCLC). Lung Cancer 2003;41(S2):S38. P. van Schil E, van Meerbeeck JP, . KrameGr, Splinter T, Legrand C, O’brien M, Giaccone G, Manegold C, van Zandwijk N. Surgery after induction chemotherapy: Morbidity and mortality in the first 100 patients of the surgery arm of EORTC 08941 trial. Lung Cancer 2003;Vol 41(S2):S45. van Meerbeeck JP, Kramer GWPM, van Schil PEY, Legrand C, O’brien M, Manegold C, Giaccone G, van Zandwijk N, Splinter TAW. Induction chemotherapy (CT) in stage IIIA-N2 non-small cell lung cancer (NSCLC): an analysis of different regimens used in EORTC 08941. Lung Cancer 2003;41(S2):S79. Kramer G, van Meerbeeck J, van Schil P, Uitterhoeve L, Smit E, Schramel F, Biesma B, Tjan-Heijnen V, Legrand C, van Zandwijk N, Splinter T, Giaccone G, on behalf of EORTC-Lung Cancer Group. Quality Assurance Review of Thoracic Radiotherapy in EORTC 08941: A randomized trial of Surgery versus Thoracic Radiotherapy in patients with stage IIIA Non-Small-Cell Lung Cancer (NSCLC) after response to induction chemotherapy. Lung Cancer 2005;49(S2):S79 PD-045.

Page 131: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

129

van Meerbeeck J, Van Schil P, Kramer G, Legrand C, O’Brien M, Manegold C, Passioukov A, Giaccone G, van Zandwijk N, Splinter T. A randomized trial of radical surgery versus thoracic radiotherapy in patients with stage IIIA-N2 non-small cell lung cancer (NSCLC) after response to induction chemotherapy (EORTC 08941). Lung Cancer;49(S2):S4 Pr5. Voordrachten: Van den Hout WB, van Houwelingen JC, Noordijk EM, Kramer GW, Leer JW. Extrapolating utility and cost data in terminally ill patients: a pragmatic modeling approach. 26th Annual meeting of the Society for medical decision making, 17-20.10.2004

Page 132: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten
Page 133: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

131

Curriculum Vitae De auteur van dit proefschrift werd op 17 november 1956 geboren te Venlo. Na het behalen van het diploma gymnasium β aan het St. Thomascollege te Venlo, volgde hij vanaf september 1975 de studie geneeskunde aan de Radboud Universiteit Nijmegen. In het 3e en 4e jaar van de studie was hij werkzaam als studentonderzoeker op de afdeling radiotherapie naar het effect van bestralingen op tumoren van de neusbijholten (J. Biesta en prof. dr. I. Kazem). Het artsenexamen werd behaald in januari 1982. Van maart 1982 tot maart 1986 volgde hij de opleiding tot radiotherapeut in Nijmegen (Hoofd prof. dr. I. Kazem en prof. dr. W.A.J. van Daal). Tijdens deze opleiding werd in het Christie Hospital te Manchester U.K. (Hoofd dr. Pointon) nog een brachytherapie stage gevolgd. Vanaf maart 1986 tot september 1987 was hij werkzaam als radiotherapeut in het Radiotherapeutisch Instituut Friesland (RIF) en vanaf september 1987 tot heden in het Arnhems Radiotherapeutisch Instituut (ARTI) als radiotherapeut en vanaf 1997 tevens als chef de policlinique. Vanaf het begin heeft hij als aandachtsgebied longkanker gehad. Hij had zitting in de longkanker werkgroepen van het IKN (1986–1987) en het IKO (vanaf 1987). Hij was vice-voorzitter en later voorzitter van de IKO longwerkgroep van 1998-2002. In de periode 1992–1995 was hij lid van de commissie “Landelijke consensus behandeling Longtumoren” namens de Nederlandse Vereniging van Radiotherapie en Oncologie (NVRO) en van 2002 tot 2004 lid van de commissie “richtlijnontwikkeling Longkanker” namens de NVRO. Vanaf 1992 is hij actief lid van de EORTC Lung Cancer Group (LCG) en van daaruit in studies participerend en als coördinator van verschillende studies binnen de regio actief. Vanaf 1995 is hij lid van de Dutch Lung Cancer Group (de “Heelsumgroep”), het platform voor de ontwikkeling van nieuwe longtrials en beleidslijnen binnen Nederland. Van 2001–2006 als secretaris lid van het bestuur van de “Heelsumgroep”. Tijdens de afronding van dit proefschrift en in afwachting van de publicaties van de artikelen die beschreven zijn in de hoofdstukken 3 en 7, overleed de promovendus onverwacht op 6 mei 2006 tijdens het sporten. Op verzoek van zijn naaste familie, met hun uitdrukkelijke medewerking en met instemming van de leden van de manuscriptcommissie en de decaan van de Faculteit Geneeskunde en Gezondheidswetenschappen, is besloten de promotie postuum te laten plaatsvinden.

Page 134: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten
Page 135: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

133

Dankwoord Ik had een droom: onderzoek te doen naar het verbeteren van de kansen voor patiënten met longkanker en daarop te promoveren. Daartoe moesten echter eerst de nodige voorbereidingen worden getroffen en nog een lange reis worden afgelegd. Een reis, waarvoor ik dank verschuldigd ben aan allen, die mijn pad kruisten/volgden op deze reis, en zonder wie ik deze reis niet had kunnen volbrengen. Mijn reis begon in 1987 toen Albertine van Dijck en Jan Werre mij verzochten vanuit het hoge Noorden in het ARTI te komen werken en daarbij onderzoek op te zetten. Het eerste reisdoel was daarbij nog bescheiden gericht op de regio zelf. Omdat patiënten bij wie eerdere radiotherapie was toegepast en die een recidief ontwikkelden geen behandelopties meer hadden, werd in samenwerking met de longartsen in de regio de studie voor herbestraling opgestart. We hadden echter grootsere, Europese, aspiraties. Daartoe werd samenwerking gezocht met de EORTC longkanker werkgroep. Op een gedenkwaardig weekend in 1990 toog ik tesamen met Eric Ullmann, met een nauwelijks te beteugelen enthousiasme, en Peter de Bruyn, nuchter en zakelijk, naar Parijs voor onze eerste kennismaking met dit gezelschap. Onze eerste vingeroefeningen met het opzetten van studies nam daarmee een aanvang, en bleek een lange hobbelige weg te zijn. Later ontmoette ik in Praag Jan van Meerbeeck en Paul van Schil, welke voor mij nadien belangrijke medereizigers op deze lange reis werden. Het was echter met Ted Splinter met wie ik, samen als missionarissen op zendingswerk, niet naar het donkere Afrika, maar in 1994 naar het Hollandse Breda toog. Daar hebben we onze Nederlandse collega longartsen en radiotherapeuten in een achteraf zaaltje moeten overtuigen van het feit, dat de behandeling voor het stadium IIIA longkanker intensiever moest gebeuren, een revolte toentertijd. Gelukkig was onze reisbrochure bij aanvang al dusdanig overtuigend, dat het lukte landelijk voldoende longartsen en radiotherapeuten aan te monsteren, zodat het schip koers kon kiezen. De data invoer van alle patiënten die inscheepten werd voor mij verricht door de datamanagers van het IKO (Jeannine Akkermans en Linda Mol). Hand- en spandiensten werden verricht door alle administratieve medewerkers, laboranten en collega radiotherapeuten op het ARTI, zodat ik de mogelijkheid had aan mijn reisverslag te werken. Het ordenen en bewerken van alle documenten was daarbij in goede handen bij Carla van Cingel en Esther van Nuland. Een groot probleem moest echter nog worden opgelost: Om ons schip zo goed mogelijk te laten varen, moest nog de meest geschikte samenstelling van de brandstof (chemotherapie) worden achterhaald. De toenmalige mengsels hadden slechts een rendement van hooguit 40%. Na verschillende mengsels te hebben getest in EORTC verband, bracht onze samenwerking met Nico van Zandwijk en Egbert Smit uiteindelijk het juiste mengsel aan het licht en kon dit rendement worden opgevoerd naar 70%. Deze chemotherapie wordt sindsdien in onze regio als standaard gebruikt. In de regio werd ik ook gesteund door de longartsen in Rijnstate (Alysis), een veilige thuishaven. Maar ik kon ook volop vertrouwen in onze drukke buitenpost Harderwijk op de longartsen Dolf Hendriks en Steven Gans en de chirurgen Ronald Beck en Wilfried Lackin. Helaas moest Ted om persoonlijke redenen het schip tussentijds verlaten, maar bleef op afstand betrokken bij deze reis. Jan van Meerbeeck volgde Ted als kapitein op en deed dit met veel verve. Ons schip wist koers te houden, doordat onze navigatoren in Brussel, Anne Kirkpatrick, Channa Debruyne, Catherine Legrand en Sonia Dussenne ons tijdig waarschuwden als we van de koers dreigden af te wijken. Eenmaal moest de koers tussentijds drastisch worden aangepast, vanwege stralingsgevaar, als gemeld in het reisverslag. Paul van Schil, bewaakte daarbij dat

Page 136: PDF hosted at the Radboud Repository of the Radboud ... · Phil Bosmans Dit geldt zeker voor degenen die participeerden in onze onderzoeken. Ondanks hun sombere vooruitzichten werkten

134

niet te veel van het parcours werd afgesneden, zodat we uiteindelijk veilig de haven konden bereiken. In 1998 ontmoette ik Jan-Willem Leer, met wie ik samen het plan smeedde om de reisverslagen te bundelen tot een volwaardig reisdocument. Daartoe was het noodzakelijk dat ik nog een reis zou ondernemen, waarbij ik zou onderzoeken of patiënten in een slechte conditie met longkanker, beter af waren met een langere reisduur op een langzaam oplopend pad, dan wel via de binnenbocht met een korte steile weg omhoog. Dat de patiënten beter af waren met een rustige reis kon worden achterhaald door de nimmer aflatende inzet van Mirjam Scholten. Steeds wist zij de afzonderlijke wandelaars weer te traceren en hun persoonlijke reisbevindingen te ontfutselen, een huzarenstukje! Dat geldt ook voor onze rekenmeesters uit Leiden, Hans van Houwelingen, Wilbert van den Hout, en Ronald Geskus. Zij moesten een boel rekenwerk verrichten, en kwamen daarbij tot de conclusie dat dit lange reisje zeker meer kost dan het korte reisje, maar dan heb je ook wat, of niet? Deze lange reis kende ook zijn eigen prijs. Mijn vaste reisgenote gedurende vele jaren, Mieke, heeft in het zicht van de haven, besloten haar bestemming te wijzigen en is haar eigen koers gaan varen. Zonder haar jarenlange inzet voor ons gezin was het mij niet mogelijk geweest deze reis zo ver te volbrengen, en ik wil haar daar toch ook voor danken. Door de steun van mijn kinderen (Stijn, Cindy, en Brenda), mijn ouders, buren en vrienden, lukte het mij toch deze zware reis alsnog tot een goed einde te brengen. Mijn beste vriend René Verheyen in het verre Amsterdam wil ik daarbij zeker niet vergeten. In de woelige wateren heeft hij menige reddingsboei toegeworpen. Daarnaast was hij een trouw lezer van mijn reisverslagen, waarbij hij geregeld een glimlach niet kon onderdrukken bij het lezen van mijn “Engelse” teksten. De conditie werd op peil gehouden met steun van mijn tennismaten, welke alle wel en wee rond het publiceren van de reisverslagen gezellig bij de borrel na het zoveelste partijtje tennis, moesten verdragen. Deze last zal vanavond echter vast en zeker gecompenseerd worden! Bijzonder moedig zijn mijn vrienden Joost Verbunt en Hans Jansen, die het zelfs aandurfden mij hier vandaag te seconderen op deze allerlaatste stap. Deze stap wil ik echter niet zetten zonder ook degenen te danken die, ondanks hun slechte vooruitzichten, trouw onze spreekuren bleven bezoeken en belangeloos hun bijdrage leverden: onze patiënten. Voor hen betekent het goed ten einde brengen van deze reis het bewijs dat hun inzet niet tevergeefs is geweest en dat we weer een kleine stap in de goede richting hebben gezet. Dit stuk van mijn reis zit erop. Een deel van mijn droom is uitgekomen, waarvoor nogmaals dank aan allen die mij daarbij hebben geholpen. De realiteit laat echter zien dat we nog steeds een lange weg hebben te gaan, voordat longkanker kan worden genezen. “Hora est!”: Het is nu tijd om even te rusten en te slapen, op zoek naar nieuwe dromen. Gijs Kramer Post Scriptum Gijs is helaas nooit meer toegekomen aan nieuwe dromen. Toch heb ik besloten zijn oorspronkelijk dankwoord in zijn geheel te handhaven. Jan Willem Leer