Scolaris Content Display Scolaris Content Display

乳房温存術後の女性に対する腫瘍床ブースト照射

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Budapest {published data only}

Polgar C, Fodor J, Orosz Z, Major T, Sulyok Z, Takacsi‐Nagy Z, et al. Electron and brachytherapy boost in the conservative treatment of stage I‐II breast cancer: 5‐year results of the randomized Budapest boost trial. Radiotherapy and Oncology 2002;64((Suppl 1)):S15. CENTRAL
Polgár C, Fodor J, Orosz Z, Major T, Takácsi‐Nagy Z, Mangel L, et al. Electron and high‐dose‐rate brachytherapy boost in the conservative treatment of stage I‐II breast cancer. Strahlentherapie und Onkologie 2002;178:615‐23. CENTRAL

EORTC {published data only}

Antonini N, Jones H, Horiot J, Poortmans P, Struikmans H, Van den Bogaert W, et al. Effect of age and radiation dose on local control after breast conserving treatment: EORTC trial 22881‐10882. Radiotherapy and Oncology 2007;82:265‐71. CENTRAL
Bartelink H, Horiot J, Poortmans P, Struikmans H, Van den Bogaert W, Barillot I, et al. for the European Organisation for Research and Treatment of Cancer Radiotherapy and Breast Cancer Groups. Recurrence rates after treatment of breast cancer with standard radiotherapy with or without additional radiation. New England Journal of Medicine 2001;345:1378‐87. CENTRAL
Bartelink H, Horiot J, Poortmans P, Struikmans H, Van den Bogaert W, Fourquet A, et al. Impact of a higher radiation dose on local control and survival in breast‐conserving therapy of early breast cancer: 10‐year results of the randomized boost versus no boost EORTC 22881‐10882 trial. Journal of Clinical Oncology 2007;25:3259‐65. CENTRAL
Bartelink H, Litiere S, Collette S, Collette L, Bogaerts J, Rutgers E, et al. Update of three major phase III randomized trials from the EORTC breast and radiotherapy group. Radiotherapy and Oncology 2011;99:S80‐1. CENTRAL
Bartelink H, Maingon P, Poortmans P, Weltens C, Fourquet A, Jager J, et al. 17 year results of the randomized boost versus no boost EORTC 22881‐10882 trial in early breast cancer. Radiotherapy and Oncology 2014;111:S190‐1. CENTRAL
Bartelink H, Maingon P, Poortmans P, Weltens C, Fourquet A, Jager J, et al. on behalf of the European Organisation for Research and Treatment of Cancer Radiation Oncology and Breast Cancer Groups. Whole‐breast irradiation with or without a boost for patients treated with breast‐conserving surgery for early breast cancer: 20‐year follow‐up of a randomised phase 3 trial. Lancet Oncology 2015;16:47‐56. CENTRAL
Bartelink H, Maingon, Poortmans P, Weltens C, Fourquet A, Jager J, et al. Correction to Lancet Oncol 2015;16:52. Lancet Oncology 2015;16:e6. CENTRAL
Bartelink H, Poortmans P, Struikmans H, Borger J, Jager J, Hoogenraad W, et al. Recurrence rates after treatment of breast cancer with standard radiotherapy with or without additional radiation. [Minder lokale recidieven van borstkanker door extra lokale bestralingsdosis na mammasparende chirurgie; 5‐jaarsresultaten van een klinische trial.]. Nederlands Tijdschrift voor Geneeskunde 2002;146:416‐23. CENTRAL
Collette S, Collette L, Budiharto T, Horiot J, Poortmans P, Struikmans H, et al. EORTC Radiation Oncology Group. Predictors of the risk of fibrosis at 10 years after breast conserving therapy for early breast cancer ‐ a study based on the EORTC trial 22881‐10882 'boost versus no boost'. European Journal of Cancer 2008;44:2587‐99. CENTRAL
Guinot J. More questions about the European Organisation for Research and Treatment of Cancer 22881‐10882 boost trial. Journal of Clinical Oncology 2011;29:e212. CENTRAL
Immink J, Putter H, Bartelink H, Cardoso J, Cardoso M, van der Hulst‐Vijgen M, et al. Long‐term cosmetic changes after breast‐conserving treatment of patients with stage I‐II breast cancer and included in the EORTC 'boost versus no boost' trial. Annals of Oncology 2012;23:2591‐8. CENTRAL
Jones H, Antonini N, Hart A, Peterse J, Horiot J, Collin F, et al. Impact of pathological characteristics on local relapse after breast‐conserving therapy: a subgroup analysis of the EORTC boost versus no boost trial. Journal of Clinical Oncology 2009;27:4939‐47. CENTRAL
Poortmans P, Ataman F, Davis J, Bartelink H, Horiot J, Pierart M, et al. Quality assurance in the EORTC phase III randomised 'boost vs. no boost' trial for breast conserving therapy: comparison of the results of two individual case reviews performed early and late during the accrual period. Radiotherapy and Oncology 2005;76:278‐84. CENTRAL
Poortmans P, Bartelink H, Horiot J, Struikmans H, Van den Bogaert W, Fourquet A, et al. on behalf of the EORTC radiotherapy and breast cancer groups. The influence of the boost technique on local control in breast conserving treatment in the EORTC 'boost versus no boost' randomised trial. Radiotherapy and Oncology 2004;72:25‐33. CENTRAL
Poortmans P, Collette L, Bartelink H, Struikmans H, Van den Bogaert W, Fourquet A, et al. on behalf of the EORTC Radiation Oncology and Breast Cancer Groups. The addition of a boost dose on the primary tumour bed after lumpectomy in breast conserving treatment for breast cancer. A summary of the results of EORTC 22881‐10882 "boost versus no boost" trial. Cancer Radiothérapie 2008;12:565‐70. CENTRAL
Vrieling C, Bartelink H, Collette L, Fourquet A, Horiot J, Jager J, et al. Impact of a boost dose of 16 Gy on local control in patients with early stage breast cancer older than 50 years of age [abstract]. European Journal of Cancer 2002;38:S149. CENTRAL
Vrieling C, Collette L, Bartelink E, Borger J, Brenninkmeyer S, Horiot J, et al. on behalf of the EORTC radiotherapy and breast cancer cooperative groups. Validation of the methods of cosmetic assessment after breast‐conserving therapy in the EORTC "boost versus no boost" trial. International Journal of Radiation Oncology, Biology, Physics 1999;45:667‐76. CENTRAL
Vrieling C, Collette L, Fourquet A, Hoogenraad W, Horiot J, Jager J, et al. on behalf of the EORTC Radiotherapy and Breast Cancer Cooperative Groups. The influence of patient, tumor and treatment factors on the cosmetic results after breast‐conserving therapy in the EORTC 'boost vs. no boost' trial. Radiotherapy and Oncology 2000;55:219‐32. CENTRAL
Vrieling C, Collette L, Fourquet A, Hoogenraad W, Horiot J, Jager J, et al. on behalf of the EORTC Radiotherapy and Breast Cancer Cooperative Groups. The influence of the boost in breast‐conserving therapy on cosmetic outcome in the EORTC "boost versus no boost" trial. International Journal of Radiation Oncology, Biology, Physics 1999;45:677‐85. CENTRAL
van Tienhoven G, Mijnheer B, Barterlink H, González D. Quality assurance of the EORTC trial 22881/10882: boost versus no boost in breast conserving therapy. Strahlentherapie und Onkologie 1997;173:201‐7. CENTRAL
van Tienhoven G, van Bree N, Mijnheer B, Bartelink H. Quality assurance of the EORTC trial 22881/10882: "assessment of the role of the booster dose in breast conserving therapy": the Dummy Run. EORTC radiotherapy cooperative group. Radiotherapy and Oncology 1991;22:290‐8. CENTRAL

Lyon {published data only}

Romestaing P, Lehingue Y, Carrie C, Coquard R, Montbarbon X, Ardiet J, et al. Role of a 10‐Gy boost in the conservative treatment of early breast cancer: results of a randomized clinical trial in Lyon, France. Journal of Clinical Oncology 1997;15:963‐8. CENTRAL

Nice {published data only}

Teissier E, Henry M, Ramaioli A, Lagrange JL, Courdi A, Bensadoun RJ, et al. Boost in conservative treatment: 6 years results of randomized trial. Breast Cancer Research and Treatment 1998;50:287. CENTRAL

SGW {published data only}

Graham P, Browne L, Capp A, Fox C, Delaney G, Kearsley H, et al. Randomized trial shows reduced whole breast dose negates benefit of lumpectomy radiotherapy boost. Radiotherapy and Oncology 2010;95(Suppl 1):S145. CENTRAL
Graham P, Browne L, Capp A, Fox C, Delaney G, Kearsley J, et al. The St George, Wollongong and Liverpool breast boost trial: 1st planned analysis at 6‐year mean follow‐up. Australasian Radiology 2007;51(Suppl 3):A85. CENTRAL
Graham P, Capp A, Fox C, Nasser E, Delaney G, Ahern V, et al. Why a breast boost should remain a controversial aspect of routine breast conservation management in Australia and New Zealand in 2002. Australasian Radiology 2003;47:44‐9. CENTRAL
Hau E, Browne L, Capp A, Delaney G, Fox C, Kearsley J, et al. The impact of breast cosmetic and functional outcomes on quality of life: long‐term results from the St. George and Wollongong randomized breast boost trial. Breast Cancer Research and Treatment 2013;139:115‐23. CENTRAL
Hau E, Browne L, Khanna S, Cail S, Cert G, Chin Y, et al. Radiotherapy breast boost with reduced whole‐breast dose is associated with improved cosmesis: the results of a comprehensive assessment from the St. George and Wollongong randomized breast boost trial. International Journal of Radiation Oncology, Biology, Physics 2012;82:682‐9. CENTRAL
Lee D, Hau E, Browne L, Chin Y, Lee J, Szwajcer A, et al. Breast irradiation causes hypo‐pigmentation of nipple‐areolar complex in women with Celtic skin type in St. George and Wollongong randomised breast boost trial. Journal of Medical Imaging and Radiation Oncology 2013;57:77. CENTRAL
Lee Y, Hau E, Browne L, Chin Y, Lee J, Szwajcer A, et al. Breast irradiation causes pallor in the nipple‐areolar complex in women with Celtic skin type (results from the St. George and Wollongong randomised breast boost trial). Journal of Medical Imaging and Radiation Oncology 2014;58:229‐36. CENTRAL
Millar E, Graham P, O'Toole S, McNeil C, Browne L, Morey A, et al. Prediction of local recurrence, distant metastases, and death after breast‐conserving therapy in early‐stage invasive breast cancer using a five‐biomarker panel. Journal of Clinical Oncology 2009;27:4701‐8. CENTRAL

References to studies excluded from this review

Cost‐effectiveness {published data only}

Hayman J, Hillner B, Harris J, Pierce L, Weeks J. Cost‐effectiveness of adding an electron‐beam boost to tangential radiation therapy in patients with negative margins after conservative surgery for early‐stage breast cancer. Journal of Clinical Oncology 2000;18:287‐95. CENTRAL
Hayman J, Hillner B, Harris J, Weeks J. Cost‐effectiveness of routine radiation therapy following conservative surgery for early‐stage breast cancer. Journal of Clinical Oncology 1998;16:1022‐9. CENTRAL

References to ongoing studies

ChiCTR‐IOR‐17010342 {published data only}

ChiCTR‐IOR‐17010342. Prospective randomized controlled clinical trial of external beam radiotherapy + interstitial brachytherapy for breast cancer after breast‐conserving surgery. apps.who.int/trialsearch/Trial2.aspx?TrialID=ChiCTR‐IOR‐17010342 (first received 6 January 2017). CENTRAL

Bahadur 2012

Bahadur YA, Constantinescu CT. Tumor bed boost radiotherapy in breast cancer. A review of current techniques. Saudi Medical Journal 2012;33(4):353‐66.

Bartelink 2007

Bartelink H, Horiot JC, Poortmans PM, Struikmans H, Van den Bogaert W, Fourquet A, et al. Impact of a higher radiation dose on local control and survival in breast‐conserving therapy of early breast cancer: 10‐year results of the randomized boost versus no boost EORTC 22881‐10882 trial. Journal of Clinical Oncology 2007;25(22):3259‐65.

Bartelink 2015

Bartelink H, Maignon P, Poortmans P, Weltens C, Fourquet A, Jager J, et al. on behalf of the European Organisation for Research and Treatment of Cancer Radiation Oncology and Breast Cancer Groups. Whole‐breast irradiation with or without a boost for patients treated with breast‐conserving surgery for early breast cancer: 20‐year follow‐up of a randomised phase 3 trial. Lancet Oncology 2015;16(1):47‐56.

Canavan 2014

Canavan J, Truong PT, Smith SL, Lu L, Lesperance M, Olivotto IA. Local recurrence in women with stage I breast cancer: declining rates over time in a large, population‐based cohort. International Journal of Radiation Oncology, Biology, Physics 2014;88:80‐6.

Collette 2008

Collette S, Collette L, Budiharto T, Horiot JC, Poortmans PM, Struikmans H, et al. Predictors of the risk of fibrosis at 10 years after breast conserving therapy for early breast cancer: a study based on the EORTC trial 22881–10882 ’boost versus no boost'. European Journal of Cancer 2008;44(17):2587–99.

Cox 1995

Cox J, Stetz J, Pajak T. Toxicity criteria of the Radiation Therapy Oncology Group (RTOG) and the European Organisation for Research and Treatment of Cancer (EORTC). International Journal of Radiation Oncology, Biology, Physics 1995;31:1341‐6.

Darby 2011

Darby S, McGale P, Correa C, Taylor C, Arriagada R, Clarke M, et al. Effect of radiotherapy after breast‐conserving surgery on 10‐year recurrence and 15‐year breast cancer death: meta‐analysis of individual patient data for 10,801 women in 17 randomised trials. Lancet 2011;378(9804):1707‐16.

DerSimonian 1986

DerSimonian R, Laird N. Meta‐analysis in clinical trials. Controlled Clinical Trials 1986;7(3):177‐88.

EBCTCG 2005

Early Breast Cancer Trialists' Collaborative Group (EBCTCG). Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15‐year survival: an overview of the randomized trials. Lancet 2005;365:1687‐717.

EORTC QoL

EORTC Quality of Life Department. European Organisation for Research and Treatment of Cancer Quality of Life questionnaire ‐ Core 30. www.eortc.be/qol/files/SCManualLG‐C30.pdf 1997 (accessed 2 December 2015).

Fisher 2002

Fisher B, Anderson S, Bryant J, Margolese RG, Deutsch M, Fisher ER, et al. Twenty‐year follow‐up of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. New England Journal of Medicine 2002;347(16):1233‐41.

Fowler 1989

Fowler JF. The linear‐quadratic formula and progress in fractionated radiotherapy. British Journal of Radiology 1989;62(740):679‐94.

Globocan

Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, et al. GLOBOCAN 2012 v1.1, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11. Lyon, France: International Agency for Research on Cancer; 2014. http://globocan.iarc.fr/Pages/fact_sheets_cancer.aspx (accessed 27 October 2017).

GRADEpro [Computer program]

Evidence Prime, Inc. GRADEpro Guideline Development Tool. McMaster University, 2015.

Graham 2007

Graham P, Browne L, Capp A, Fox C, Delaney G, Kearsley J, et al. The St George, Wollongong and Liverpool breast boost trial: 1st planned analysis at 6‐year mean follow‐up. Australasian Radiology 2007;51 (Suppl 3):A85.

Harris 1979

Harris JR, Levene MB, Svensson G, Hellman S. Analysis of cosmetic results following primary radiation therapy for stages I and II carcinoma of the breast. International Journal of Radiation Oncology, Biology, Physics 1979;5(2):257‐61.

Hennequin 2013

Hennequin C, Dubray B. Alpha/beta ratio revisited in the era of hypofractionation. Cancer Radiothérapie 2013;17(5‐6):344‐8.

Higgins 2011

Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Holland 1985

Holland R, Veling SH, Mravunac M, Hendriks JH. Histologic multifocality of Tis, T1‐T2 breast carcinomas: implication for clinical trials of breast‐conserving surgery. Cancer 1985;56(5):979‐90.

Kellerer 1972

Kellerer AM, Rossi HH. The theory of dual radiation action. Current Topics in Radiation Research 1972;8:85‐158.

Kindts 2016

Kindts I, Laenen A, Peeters S, Janssen H, Depuydt T, Neven P, et al. Evaluation of a breast cancer nomogram to predict ipsilateral breast relapse after breast‐conserving therapy. Radiotherapy and Oncology 2016;119(1):45‐51.

Kuerer 2004

Kuerer HM, Julian TB, Strom EA, Lyerly HK, Giuliano AE, Mamounas EP, et al. Accelerated partial breast irradiation after conservative surgery for breast cancer. Annals of Surgery 2004;239(3):338‐51.

Litière 2012

Litière S, Werutsky G, Fentiman IS, Rutgers E, Christiaens MR, Van Limbergen E, et al. Breast conserving therapy versus mastectomy for stage I‐II breast cancer: 20 year follow‐up of the EORTC 10801 phase 3 randomised trial. Lancet Oncology 2012;13(4):412‐9.

Malvezzi 2013

Malvezzi M, Bertuccio P, Levi F, La Vecchia C, Negri E. European cancer mortality predictions for the year 2013. Annals of Oncology 2013;24(3):792‐800.

Millar 2009

Millar E, Graham P, O'Toole S, McNeil C, Browne L, Morey A, et al. Prediction of local recurrence, distant metastases, and death after breast‐conserving therapy in early‐stage invasive breast cancer using a five‐biomarker panel. Journal of Clinical Oncology 2009;27:4701‐8.

NCCN

Gradishan WJ, Anderson BO on behalf of the NCCN Breast Cancer Panel Members. National Comprehense Cancer Network Guidelines Breast Cancer. https://www.nccn.org/professionals/physician_gls/pdf/breast.pdf version 2. 2016 2016 (accessed 2 January 2017).

NCI‐CTCAE v4.0

National Cancer Institute. Common Terminology Criteria for Adverse Events. evs.nci.nih.gov/ftp1/CTCAE/About.html 2009 (accessed 2 December 2015).

Parmar 1998

Parmar MK, Torri V, Stewart L. Extracting summary statistics to perform meta‐analyses of the published literature for survival endpoints. Statistics in Medicine 1998;17(24):2815‐34.

Perez 1996

Perez CA, Taylor ME, Halverson K, Garcia D, Kuske RR, Lockett MA. Brachytherapy or electron beam boost in conservation therapy of carcinoma of the breast: a nonrandomized comparison. International Journal of Radiation Oncology, Biology, Physics 1996;34:995‐1007.

Polgár 2001

Polgár C, Fodor J, Major T, Orosz Z, Németh G. The role of boost irradiation in the conservative treatment of stage I‐II breast cancer. Pathology and Oncology Research 2001;7(4):241‐50.

RevMan [Computer program]

The Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager 5 (RevMan). Version 5.3. Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Sanghani 2010

Sanghani M, Truong PT, Raad RA, Niemierko A, Lesperance M, Olivotto IA, et al. Validation of a web‐based predictive nomogram for ipsilateral breast tumor recurrence after breast conserving therapy. Journal of Clinical Oncology 2010;28(5):718‐22.

Senkus 2015

Senkus E, Kyriakides S, Ohno S, Penault‐Llorca F, Poortmans P, Rutgers E, et al. on behalf of the ESMO Guidelines Working Group. Primary breast cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow‐up. Annals of Oncology 2015;26(Suppl 5):v8‐v30.

START 2008

The START Trialists' Group. The UK Standardisation of Breast Radiotherapy (START) Trial A of radiotherapy hypofractionation for treatment of early breast cancer: a randomised trial. Lancet Oncology 2008;9(4):331‐41.

Tierney 2007

Tierney JF, Stewart LA, Ghersi D, et al. Practical methods for incorporating summary time‐to‐event data into meta‐analysis. Trials 2007;8:16.

Vaidya 2010

Vaidya JS, Joseph DJ, Tobias JS, Bulsara M, Wenz F, Saunders C, et al. Targeted intraoperative radiotherapy versus whole breast radiotherapy for breast cancer (TARGIT‐A trial): an international, prospective, randomised, non‐inferiority phase 3 trial. Lancet 2010;376(9735):91‐102.

van Laar 2013

van Laar C, van der Sangen MJ, Poortmans PM, Nieuwenhuijzen GA, Roukema JA, Roumen RM, et al. Local recurrence following breast‐conserving treatment in women aged 40 years or younger: trends in risk and the impact on prognosis in a population‐based cohort of 1143 patients. European Journal of Cancer 2013;49(15):3093‐101.

van Werkhoven 2011

van Werkhoven E, Hart G, Tinteren Hv, Elkhuizen P, Collette L, Poortmans P, et al. Nomogram to predict ipsilateral breast relapse based on pathology review from the EORTC 22881‐10882 boost versus no boost trial. Radiotherapy and Oncology 2011;100(1):101‐7.

Verhoeven 2015

Verhoeven K, Kindts I, Laenen A, Peeters S, Janssen H, Van Limbergen E, et al. A comparison of three different radiotherapy boost techniques after breast conserving therapy for breast cancer. Breast 2015;24(4):391‐6.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Budapest

Methods

Accrual: 1995 to 1998

Single‐centre
Phase III RCT
Budapest, Hungary
Median follow‐up: 60 months

Participants

  • 627 women with stage I‐II breast cancer.

  • Age from first 207 patients with at least 3 years' follow‐up: 54 (range 34 to 79).

  • Stage for first 207 patients with at least 3 years' follow‐up: T1 61%, T2 39%, N0 76%, N1 24%.

  • Surgical margins of first 207 patients with at least 3 years' follow‐up: positive 8%.

  • Notable exclusion criteria: bilateral breast cancer.

  • Receptor status from the first 207 patients with at least 3 years' follow‐up: ER+ 54%, ER‐ 38%, ER unknown 8%.

  • Menstrual status: not reported.

  • Other adjuvant treatments: from the first 207 patients with at least 3 years' follow‐up: 18% received hormonal therapy alone, 18% chemotherapy alone, and 6% hormonal therapy and chemotherapy. No significant differences between boost and no‐boost groups.

Interventions

ARM 1:
Intervention details (n = 307): WBI 50 Gy in 25 fractions in 5 weeks + 16‐gray boost (n = 238) or 12‐ to 14.25‐gray HDR‐BT boost (n = 65). BED = 87.8 Gy.

ARM 2:
Comparator details (n = 320): WBI 50 Gy in 25 fractions in 5 weeks. BED = 66.7 Gy.

Outcomes

Primary outcomes

  1. Local control.

  2. Relapse‐free survival.

Secondary outcomes

  1. Cancer‐specific survival.

  2. Late side effects of skin and subcutaneous tissues scored by the RTOG/EORTC late radiation morbidity scheme.

  3. Cosmetic results, scored on a 4‐grade scale (excellent, good, fair, poor), as suggested by Perez and colleagues (Perez 1996).

Notes

  • Boost technique was based on the treating radiation oncologist’s preference.

  • Brachytherapy implants were performed 3 weeks after WBI.

  • For the first 207 patients with at least 3 years' follow‐up: the HDR fractionation schedules were calculated using the Linear Quadratic model with an alpha/beta ratio of 4 Gy for late and 10 Gy for early effects. For the first 19 patients, the total boost dose was calculated to be equivalent to late effects of 20 Gy Low‐dose‐rate. Since serious side effects were not observed, the total boost dose was increased for the next 33 patients to be equivalent to the early side effects of 20 Gy LDR. So the prescribed HDR doses (calculated to 100% isodose surface) consisted of 3 fractions of 4 Gy (n = 19) and 4.75 Gy (n = 33) over 3 days for a total boost dose of 12 Gy and 14.25 Gy, respectively.

  • For women with pN0 or pN1a, regional nodal irradiation was omitted. All others received RNI by an anterior supraclavicular/axillary field.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote "Randomization was done by the study coordinator (S.P.)" and "patients were randomly allocated to treatment options by a sealed‐envelope system in blocks of 20".Budapest 2002, page 616, paragraph 2.

Allocation concealment (selection bias)

Unclear risk

Quote "patients were randomly allocated to treatment options by a sealed‐envelope system in blocks of 20". Budapest 2002, page 616, paragraph 2.

Allocation concealment appears to have been done, although the description was incomplete, which contributed to the judgement of unclear risk of bias.

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

Participants and personnel: not mentioned, unlikely to have been done.

We judged this domain as at low risk of bias.

Blinding of participants and personnel (performance bias)
Subjective outcomes

Low risk

Participants and personnel: not mentioned, unlikely to have been done.

We judged this domain as at low risk of bias.

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Participants: not mentioned, unlikely to have been done.

Personnel: not done, but in view of prespecified protocol of follow‐up with regular mammography, unlikely to have introduced bias.

Quote "Patients were seen every 3 months in the first 2 years after RT, and every 6 months thereafter. Mammography, chest X‐ray, breast and abdominal ultrasound examinations, bone scan, and blood tests were performed at least annually. Local recurrence was defined ... proven by histological examination." Budapest 2002, page 618, paragraph 4.

Unlikely to be a source of bias in view of the prespecified schedule for follow‐up visits and investigations. Local recurrence required biopsy information, which would reduce the risk of bias in evaluation of this outcome.

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

Quote "Secondary endpoints were ... late side effects of skin and subcutaneous tissues scored by the RTOG/EORTC late radiation morbidity scheme, and cosmetic results scored by a 4‐grade scale (excellent/good/fair/poor), as suggested by Perez et al." Budapest 2002, page 616, paragraph 4. Outcomes were measured by the treating physician.

Because the assessment of subjective outcomes was not blinded, we judged this domain as at high risk of bias.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Exclusions: 2 (ineligibility: early distant metastasis).

Quote "One patient refused boost irradiation, but was analysed according to the assigned treatment group." Budapest 2002, page 616, paragraph 4.

Postrandomisation exclusions are detailed without information on treatment arm, but with reasons. We deemed this outcome as at low risk of bias.

Selective reporting (reporting bias)

Unclear risk

Outcomes in methods section:

Primary: ipsilateral breast tumour recurrence; occurrence of local, regional, or distant relapse (whichever came first) for relapse‐free survival.

Secondary: death from breast cancer, late side effects of skin and subcutaneous tissues scored by the RTOG/EORTC late radiation morbidity scheme, and cosmetic results scored on a 4‐grade scale (excellent/good/fair/poor), as suggested by Perez and colleagues (Perez 1996).

Outcomes reported in paper:

  1. Ipsilateral breast tumour recurrence, location of recurrence, local recurrence as first event, (mean time to) isolated local recurrence followed by subsequent metastases, mean time between isolated local relapse and distant relapse.

  2. 5‐year probability of local tumour control.

  3. 5‐year probability of relapse‐free survival.

  4. 5‐year probability of cancer‐specific survival.

  5. Actuarial 5‐year local recurrence rate for women younger and older than 40 years and for women with negative versus positive or close surgical margins.

  6. RTOG/EORTC late radiation side effects.

  7. Cosmetic results scored on a 4‐grade scale.

We did not have access to the study protocol, so judged this domain as at unclear risk of bias.

Other bias

Unclear risk

Because this was an interim report (prospectively planned however), we judged it as at unclear risk of bias.

EORTC

Methods

Accrual: 1989 to 1996

Multicentre: 31 centres in 9 countries
Phase III RCT
9 countries: the Netherlands, Belgium, France, Germany, Switzerland, the United Kingdom, Israel, Austria, Spain
Median follow‐up: 17.2 years

Participants

5569 participants randomised in the trial. 5318 participants with stage I or II (T1‐2, N0‐1, M0) breast cancer who had undergone macroscopically complete surgical removal of the tumour load and axillary dissection, with microscopically completely excised tumour.

  • Less than 70 years old, median 56.

  • Stage: T1 52%, T2 48%, T3 < 1%. N0 90%, N1‐2 7%, Nx 3%.

  • Indication of percentage of participants with positive surgical margins: only patients with negative section margins were included.

  • Notable exclusion criteria: pure carcinoma in situ, multiple tumour foci in more than 1 quadrant, tumourectomy more than 9 weeks before the start of radiotherapy or more than 6 months before the start of radiotherapy in case of chemotherapy.

  • Receptor status: ER+PR+ 39%, ER+PR‐ 10%, ER‐PR+ 5%, ER‐PR‐ 13%, unknown 33%.

  • Menstrual status: premenopausal 38%, postmenopausal 62%, unknown < 1%.

  • Other adjuvant treatments: participants with axillary lymph node involvement received adjuvant systemic therapy; premenopausal women received chemotherapy and postmenopausal women received tamoxifen.

Interventions

ARM 1: Intervention details (n = 2661): WBI 50 Gy in 25 fractions in 5 weeks + boost dose of 16 Gy aimed at the tumour bed (in 8 fractions with fast electrons or tangential photon fields; or 15 Gy by means of an iridium‐192 implant at a dose rate of 0.5 Gy per hour). BED 87.8 Gy.

ARM 2: Comparator details (n = 2657): WBI 50 Gy in 25 fractions in 5 weeks. BED 66.7 Gy.

Outcomes

Primary outcomes:

  1. Overall survival defined as the time from randomisation to death from any cause during follow‐up or to last visit.

Secondary outcomes:

  1. Local control defined as the time from randomisation until the development of any local recurrence during follow‐up (time‐to‐event outcome). We define local recurrence as recurrence in the ipsilateral breast (i.e. the breast in which cancer had been diagnosed), the skin and parenchyma. Ipsilateral breast tumour recurrence as the first treatment failure was the event of interest and any other treatment failure as first event (including death) was considered a competing risk.

  2. Cosmesis: panel score and BCCT.core score.

  3. Fibrosis. Cumulative incidence of severe fibrosis was counted from the entry to the date of first report of severe fibrosis or until the last visit before a mastectomy. Mastectomy and death without severe fibrosis were competing risks for this analysis. pBRA was measured.

Exploratory endpoints:

  1. Breast cancer mortality.

  2. Time to distant metastasis defined as the time from randomisation to the first report of distant metastases (event), death without metastases (competing risk), or last visit (censored observation).

  3. Disease‐free survival was the time from day of first report of local ipsilateral breast tumour recurrence, to the day of first report of either a subsequent local failure or a failure at another site or to death of any cause.

  4. Disease‐free survival and overall survival after local failure: time to event was counted from the first ipsilateral breast tumour recurrence for all patients with local ipsilateral breast tumour recurrence as first failure.

Notes

  • Target area for the additional dose of radiation was the site of the primary tumour, with a margin of 1.5 cm around the primary tumour after microscopically complete excision and of 3 cm in case of invasive cancer with extensive ductal carcinoma in situ.

  • Section margins were assessed for the presence of invasive carcinoma, but not for ductal carcinoma in situ.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote "Patients were centrally randomised at the EORTC Headquarters according to a minimisation algorithm (variance method) in a 1:1 ratio." "Factors used in the minimization were age, menopausal status, presence of extensive ductal carcinoma in situ (ten or more ducts involved), clinical tumour size, nodal status and institute where the patient received treatment." Bartelink 2015, page 48, paragraph 3‐4.

We judged this domain as at low risk of bias.

Allocation concealment (selection bias)

Low risk

Quote "Patients were centrally randomised at the EORTC Headquarters according to a minimisation algorithm (variance method) in a 1:1 ratio." "Neither patients nor investigators were masked to treatment allocation" Bartelink 2015, page 48, paragraph 3‐4.

We judged this domain as at low risk of bias.

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

Participants and personnel: not mentioned, unlikely to have been done.

We judged this domain as at low risk of bias.

Blinding of participants and personnel (performance bias)
Subjective outcomes

Low risk

Participants and personnel: not mentioned, unlikely to have been done.

We judged this domain as at low risk of bias.

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Participants: not mentioned, unlikely to have introduced bias.

Personnel: not done, but in view of prespecified protocol of follow‐up with regular mammography, unlikely to have introduced bias.

Quote "patients were followed up two or three times per year for 5 years, then once per year by mammography and clinical examination." Bartelink 2015, page 48, paragraph 8.

Local recurrence is defined as any recurrence within the treated breast, confirmed by histology or cytology (protocol).

Unlikely to be a source of bias in view of the prespecified schedule for follow‐up visits and investigations.

Blinding of outcome assessment (detection bias)
Subjective outcomes

Unclear risk

Quote "The cosmetic outcome and assessment of therapy sequelae will be scored regularly. Therefore colour print photographs or slides have to be taken immediately after surgery (before radiotherapy), and thereafter every 3 years. ... An external review panel will score qualitatively the cosmetic result, based on the pictures, using a 4 point grading scale." Protocol, page 26, paragraph 2.

Quote "At each visit except baseline, the physician (not blinded for the treatment arm) scored the grade of fibrosis (none/minimal/moderate/severe) for the whole breast and for the boost area" Collette 2008, page 2588, paragraph 8.

We would have judged this domain as at low risk of bias based on the use of a grading scale and the blinding of assessors; however, since the physician was not blinded to the treatment arm to score fibrosis, we assessed it as at unclear risk of bias.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Exclusions: 0

Attritions: 0

Intention‐to‐treat analysis

Selective reporting (reporting bias)

Low risk

Outcomes in the protocol

  1. Local recurrence rate.

  2. Cosmetic results.

  3. Survival.

Outcomes in the results

  1. Overall survival.

  2. Breast cancer mortality.

  3. Distant metastases.

  4. Second primary tumour in the contralateral breast.

  5. Overall incidence of breast cancer‐related events.

  6. Disease‐free survival.

  7. Time to any recurrence.

  8. Local recurrence.

  9. Locoregional recurrence.

  10. Second primary ipsilateral breast cancers.

  11. Fibrosis.

  12. Cosmesis.

We had access to the study protocol and all prespecified endpoints were reported, so we judged this domain as at low risk of bias.

Other bias

Low risk

No other sources of bias identified.

Lyon

Methods

Accrual: 1986 to 1992

2 centres: Hospices civils de Lyon and Centre Léon Bérard
Phase III RCT
France
Median follow‐up: 3.3 years

Participants

  • 1028 women with invasive ductal adenocarcinoma ≤ 3 cm and "free" pathological margins (absence of detectable cancer cells at the inked margin M0).

  • Less than 70 years old, mean 53.

  • T ≤ 2 cm 81%, > 2 cm 19%; N0 73%, N1 27%.

  • Free margins 98%.

  • Notable exclusion criteria: > 70 years old.

  • ER+: not reported.

  • Menopausal state: not reported.

  • Other adjuvant treatments: chemotherapy in 22%, endocrine therapy in 30%. N+ in premenopausal women: adjuvant chemotherapy. N+ in postmenopausal women: tamoxifen, sometimes even if N0.

Interventions

ARM 1: Intervention details (n = 521): WBI 50 Gy cobalt 60/20 fractions of 2.5 Gy 4 per week + boost 10 Gy 9 or 12 MeV 4 fractions 1 week. BED 86.4 Gy.

ARM 2: Comparator details (n = 503): WBI 50 Gy cobalt 60/20 fractions of 2.5 Gy 4 per week. BED 72.1 Gy.

Outcomes

Primary outcomes

  1. Ipsilateral breast tumour recurrence, defined as any recurrence within the irradiated breast. Time to local recurrence, with death from any cause apart from metastases considered censored events.

Secondary outcomes

  1. Overall survival.

  2. Disease‐free survival.

  3. Local, regional, or metastatic relapse.

  4. Toxicity: telangiectasia, assessed by clinician.

  5. General cosmetic score, assessed by clinician and participant.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Quote "Stratification factors were the pathologic stages of the tumour (T1 v T2) and node (N0 v N1‐2)." Lyon, page 964, paragraph 2.

We did not have access to the study protocol, so judged this domain as at unclear risk.

Allocation concealment (selection bias)

Unclear risk

Quote "Patients were entered onto the study by the radiation oncologist when first evaluated after surgery, and were randomised at the start of irradiation." Lyon, page 964, paragraph 2.

We did not have access to the study protocol, so judged this domain as at unclear risk.

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

Participants and personnel: not mentioned, unlikely to have been done.

We judged this domain as at low risk of bias.

Blinding of participants and personnel (performance bias)
Subjective outcomes

Low risk

Participants and personnel: not mentioned, unlikely to have been done.

We judged this domain as at low risk of bias.

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Participants: not mentioned, unlikely to have been done.

Personnel: not done, but in view of prespecified protocol of follow‐up with regular mammography, unlikely to have introduced bias.

Quote "Clinical follow‐up evaluation was performed every 4 months during the first 2 years, every 6 months up to the fifth year, and then annually. Bilateral mammography was performed every year." Lyon, page 964, paragraph 2.

Local recurrence is defined as any recurrence within the treated breast, without further specifying if this required biopsy information.

Unlikely to be a source of bias in view of the prespecified schedule for follow‐up visits and investigations.

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

Quote "Toxicity was evaluated only as teleangiectasias, assessed regularly by the clinical oncologist during follow‐up evaluation as follows: 0, absent; 1, a few visible areas of teleangiectasia in the tumour bed; 2, teleangiectasia that covered one quadrant of the breast; and 3, obvious teleangiectasia over more than one quadrant. The general cosmetic score, assessed by both the clinical oncologist and the patients themselves, was as follows: 1, excellent; 2, good; 3, fair; 4, poor." Lyon, page 965, paragraph 2.

Because the assessment of subjective outcomes was not blinded, we judged this domain as at high risk of bias.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Quote "Two patients presented with bilateral synchronous breast cancer, which corresponded to four cases. Since this situation had not been planned for in the initial protocol, these two patients were excluded after randomization. ... Forty patients had not received the assigned treatment: 31 in the control group who had erroneously received a boost, and nine in the experimental group who had not received the assigned boost. The analysis was performed on an intent‐to‐treat basis." Lyon, page 965, paragraph 4.

Quote "Thirty‐two patients (17 in the control group and 15 in the experimental group) were lost to follow‐up after treatment. In 4 years, 194 patients were lost to follow‐up for local recurrence in the control group and 171 in the experimental group." Lyon, page 965, paragraph 6.

Postrandomisation exclusions are detailed without information on treatment arm, but with reasons. There was no postrandomisation attrition. We judged this domain as at low risk of bias.

Selective reporting (reporting bias)

Unclear risk

Outcomes in methods section

Primary outcome: time to local recurrence (any recurrence within the irradiated breast).

Secondary outcomes: overall survival; disease‐free survival; local, regional, or metastatic relapse; telangiectasia; general cosmetic score.

Outcomes reported in paper

  1. Time to local recurrence.

  2. Distant treatment failure as first relapse.

  3. Disease‐free survival.

  4. Overall survival.

  5. Telangiectasia.

  6. Cosmetic score.

We did not have access to the study protocol, so judged this domain as at unclear risk of bias.

Other bias

Low risk

No other sources of bias identified.

Nice

Methods

Accrual: 1987 to 1994

RCT

Nice, France
Median follow‐up: 73 months

Participants

  • 664 women with invasive breast cancer, treated by conservative surgery.

  • Tumour‐free margins.

  • Axillary dissection.

  • Systemic therapy by hormonotherapy or chemotherapy, or both, was possible.

Interventions

ARM 1: Intervention details: WBI 50 Gy/25 fractions + boost 10 Gy/5 fractions. BED 79.9 Gy.

ARM 2: Comparator details: WBI 50 Gy/25 fractions. BED 66.7 Gy.

Outcomes

Primary outcome: local control.

Notes

Abstract

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

We did not have access to the study protocol, so judged this domain as at unclear risk.

Allocation concealment (selection bias)

Unclear risk

We did not have access to the study protocol, so judged this domain as at unclear risk.

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

Participants and personnel: not mentioned, unlikely to have been done.

We judged this domain as at low risk of bias.

Blinding of participants and personnel (performance bias)
Subjective outcomes

Low risk

Participants and personnel: not mentioned, unlikely to have been done.

We judged this domain as at low risk of bias.

Blinding of outcome assessment (detection bias)
Objective outcomes

High risk

We did not have access to this information in the abstract, so judged this domain as at high risk of bias.

Blinding of outcome assessment (detection bias)
Subjective outcomes

High risk

We did not have access to this information in the abstract, so judged this domain as at high risk of bias.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No exclusions or attrition of participants reported. We would have judged this domain as at low risk of bias, however since only the abstract was available, we assessed it as at unclear risk of bias.

Selective reporting (reporting bias)

High risk

Only the results on local control are described in the abstract. Since no protocol was available and no further results on local recurrence were reported, we judged this domain as at high risk of bias.

Other bias

Unclear risk

Because this was an interim report and only the abstract was available, we judged this domain as at unclear risk of bias.

SGW

Methods

Accrual: 1996 to not reported

Multicentre: St George and Wollongong hospitals, NSW and Liverpool Hospital, Sydney
RCT
Australia
Median follow‐up: 8.5 years

Participants

  • 688 women with in situ tumours, T1, T2 N0‐3 breast cancer after lumpectomy.

  • Mean 59 years old, range 24 to 85.

  • Mean tumour size 18 mm, extensive intraductal carcinoma in 10%, N+: 24%.

  • Indication of percentage of women with positive surgical margins: only women with negative section margins were included.

  • Notable exclusion criteria: gross multifocal disease, involvement of margins, bilateral breast cancer.

  • ER+: 76%.

  • 20% premenopausal, 74% postmenopausal, 6% perimenopausal.

  • Other adjuvant treatments: chemotherapy in 20%, endocrine therapy in 39%.

Interventions

ARM 1: Intervention details: WBI 45 Gy/25 fractions + boost 16 Gy/8 fractions. BED 78.6 Gy.

ARM 2: Comparator details: WBI 50 Gy/25 fractions. BED 66.7 Gy.

Outcomes

Primary outcomes

  1. Local control.

Secondary outcomes

  1. Cosmesis:

    • Women were asked to compare the treated with the untreated breast and score the overall result. Results were dichotomised into "normal/excellent/good" and "marked/severe".

    • 5‐person panel evaluated digital photographs. The treated breast was compared with the untreated breast and scored using a 4‐point scale developed by Harris and colleagues. Nipple‐areolar complex hypopigmentation was scored.

    • At each follow‐up the clinician was asked to assess the cosmetic scores. Fibrosis and oedema were scored as "nil", "mild", "marked", and "severe". Overall cosmesis was scored as "excellent", "good", "fair", "poor".

    • Objective BRA, pBRA.

    • BCCT.core software cosmesis score.

  2. Quality of life: EORTC QLQ‐C30 (version 2.0) + functional assessment by asking the participant to score the degree of arm swelling, arm pain, limitation of arm movement, loss of feeling in fingers.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

We did not have access to the study protocol, so judged this domain as at unclear risk.

Allocation concealment (selection bias)

Unclear risk

We did not have access to the study protocol, so judged this domain as at unclear risk.

Blinding of participants and personnel (performance bias)
Objective outcomes

Low risk

Participants and personnel: not mentioned, unlikely to have been done.

We judged this domain as at low risk of bias.

Blinding of participants and personnel (performance bias)
Subjective outcomes

Low risk

Participants and personnel: not mentioned, unlikely to have been done.

We judged this domain as at low risk of bias.

Blinding of outcome assessment (detection bias)
Objective outcomes

Low risk

Quote "Digital photographs were taken at 1, 2 and 3 years as well as at 5 and 10 years post‐surgery by investigator P.G." SGW 2014, page 230, paragraph 3.

Quote "the relative breast retraction assessment was performed by a single observer using another custom written computer software." SGW 2012, page 685, paragraph 2. Also, the BCCT.core software was used.

Tumour recurrences were classified as ipsilateral in‐breast recurrence. Quote "Patients were assessed 6 weeks after radiotherapy, every 6 months for 2 years, then annually thereafter with annual breast imaging.

We judged this domain as at low risk of bias.

Blinding of outcome assessment (detection bias)
Subjective outcomes

Unclear risk

Concerning digital photographs: quote "A five‐person panel (two radiation oncologists, one breast surgeon, one breast clinical nurse consultant, one radiotherapy clinical nurse specialist; four women, one man) all with experience in the treatment of breast cancers were asked to evaluate the images" "The treated breast was compared with the untreated breast and scored using a 4‐point scale developed by Harris et al." Evaluation was done independently. SGW 2012, page 683, paragraph 5.

Quote "Functional assessments were performed by asking the patient to score the degree of arm swelling, arm pain, limitation of arm movement, loss of feeling in fingers, breast sensitivity and breast tenderness. For the cosmetic assessment, the patient was asked to compare the treated with the untreated breast and score the overall result. SGW 2013, page 116, paragraph 5. EORTC QLQ‐C30 was used for quality of life.

Quote "At each follow‐up visit the clinician was asked to assess the cosmetic outcomes. Fibrosis and edema were scored as nil, mild, marked and severe and overall cosmesis was scored as excellent, good, fair and poor. The patient‐self‐assessment .... normal, excellent, good, fair, poor." SGW 2012, page 684, paragraph 2; page 685, paragraph 1.

As we had no information on the blinding of assessors, we judged this domain as at unclear risk of bias.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

346 women allocated to the experimental group, 342 to the control group. At 5 years 253 and 260 were alive, respectively, at the St George Hospital and for 183 and 202, respectively, digital photographs were available. (Figure 1) SGW 2012, page 683.

Quote: "... randomized 688 patients into the boost and no boost arms. 609, 580, and 428 patients had baseline, 5 and 10 years cosmetic data available, respectively."

No exclusions or attrition of participants reported. We judged this domain as at low risk of bias.

Selective reporting (reporting bias)

High risk

Outcomes in methods section

  1. Photographs: panel assessment, BCCT.core score, pBRA measurements, nipple‐areolar complex alterations.

  2. Participant and clinician live assessment.

  3. Quality of life.

Outcomes in results

  1. Photographs: panel assessment, BCCT.core score, pBRA measurements, nipple‐areolar complex alterations.

  2. Participants and clinician live assessment.

  3. Quality of life.

The results on local control are only briefly described in Millar 2009 and Graham 2007. Since no protocol was available and no further results on local recurrence were reported, we judged this domain as at high risk of bias.

Other bias

Low risk

No other sources of bias identified.

BED: biologically equivalent dose
BRA: breast retraction assessment
ER: oestrogen receptor
HDR‐BT: high‐dose‐rate brachytherapy
MeV: megaelectron volt
N: lymph node
pBRA: percentage breast retraction assessment
PR: progesterone receptor
RCT: randomised controlled trial
RT: radiation therapy
RTOG/EORTC: Radiation Therapy Oncology Group/European Organisation for Research and Treatment of Cancer
RNI: regional nodal irradiation
WBI: whole‐breast irradiation

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Cost‐effectiveness

Not a randomised controlled trial

Characteristics of ongoing studies [ordered by study ID]

ChiCTR‐IOR‐17010342

Trial name or title

Prospective randomized controlled clinical trial of external beam radiotherapy + interstitial brachytherapy for breast cancer after breast‐conserving surgery

Methods

Accrual: 2017 to 2022

Single‐centre
Phase III randomised controlled trial
Changchun, Jilin, China

Participants

  • 100 women with stage DCIS‐I‐II breast cancer with tumour < 3 cm.

  • Age 30 to 75.

Interventions

ARM 1: Intervention details (n = 50): WBI + interstitial brachytherapy boost.

ARM 2: Comparator details (n = 50): WBI.

Outcomes

Primary outcomes

  1. Local control.

  2. Overall survival.

  3. Recurrence‐free survival.

  4. Distant metastasis‐free survival.

  5. Quality of life.

Secondary outcomes

  1. Safety.

  2. Cosmetic effect.

Starting date

1 February 2017

Contact information

[email protected]

Notes

DCIS: ductal carcinoma in situ
WBI: whole‐breast irradiation

Data and analyses

Open in table viewer
Comparison 1. Tumour bed boost radiotherapy versus no boost

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Local control Show forest plot

5

8315

Hazard Ratio (Fixed, 95% CI)

0.64 [0.55, 0.75]

Analysis 1.1

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 1 Local control.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 1 Local control.

2 Local control ‐ sensitivity analysis Show forest plot

3

6963

Hazard Ratio (Fixed, 95% CI)

0.62 [0.52, 0.73]

Analysis 1.2

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 2 Local control ‐ sensitivity analysis.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 2 Local control ‐ sensitivity analysis.

3 Local control ‐ subgroup +40 years Show forest plot

2

5058

Hazard Ratio (Fixed, 95% CI)

0.65 [0.53, 0.81]

Analysis 1.3

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 3 Local control ‐ subgroup +40 years.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 3 Local control ‐ subgroup +40 years.

4 Local control ‐ subgroup low boost dose Show forest plot

2

1352

Hazard Ratio (Fixed, 95% CI)

0.95 [0.55, 1.62]

Analysis 1.4

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 4 Local control ‐ subgroup low boost dose.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 4 Local control ‐ subgroup low boost dose.

5 Overall survival Show forest plot

2

6342

Hazard Ratio (Fixed, 95% CI)

1.04 [0.94, 1.14]

Analysis 1.5

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 5 Overall survival.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 5 Overall survival.

6 Disease‐free survival Show forest plot

3

6549

Hazard Ratio (Fixed, 95% CI)

0.94 [0.87, 1.02]

Analysis 1.6

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 6 Disease‐free survival.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 6 Disease‐free survival.

7 Late toxicity, pBRA Show forest plot

2

1526

Mean Difference (IV, Fixed, 95% CI)

0.38 [‐0.18, 0.93]

Analysis 1.7

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 7 Late toxicity, pBRA.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 7 Late toxicity, pBRA.

8 Cosmesis, Panel Show forest plot

2

1116

Odds Ratio (M‐H, Fixed, 95% CI)

1.41 [1.07, 1.86]

Analysis 1.8

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 8 Cosmesis, Panel.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 8 Cosmesis, Panel.

9 Cosmesis, Physician Show forest plot

2

592

Odds Ratio (M‐H, Fixed, 95% CI)

1.58 [0.93, 2.69]

Analysis 1.9

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 9 Cosmesis, Physician.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 9 Cosmesis, Physician.

Illustration of the breast retraction assessment (BRA) measurements as reported by EORTC. BRA = √((a1 ‐ b1)² + (a2 ‐ b2)²); reference length = √(b1² + b2²); percentage breast retraction assessment = (BRA/reference length) x 100.
Figuras y tablas -
Figure 1

Illustration of the breast retraction assessment (BRA) measurements as reported by EORTC. BRA = √((a1 ‐ b1)² + (a2 ‐ b2)²); reference length = √(b1² + b2²); percentage breast retraction assessment = (BRA/reference length) x 100.

Study flow diagram.
Figuras y tablas -
Figure 2

Study flow diagram.

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 3

'Risk of bias' summary: review authors' judgements about each risk of bias item for each included study.

Forest plot of comparison: 1 Tumour bed boost radiotherapy versus no boost, outcome: 1.1 Local control.
Figuras y tablas -
Figure 4

Forest plot of comparison: 1 Tumour bed boost radiotherapy versus no boost, outcome: 1.1 Local control.

Forest plot of comparison: 1 Tumour bed boost radiotherapy versus no boost, outcome: 1.5 Overall survival.
Figuras y tablas -
Figure 5

Forest plot of comparison: 1 Tumour bed boost radiotherapy versus no boost, outcome: 1.5 Overall survival.

Forest plot of comparison: 1 Tumour bed boost radiotherapy versus no boost, outcome: 1.8 Cosmesis, Panel.
Figuras y tablas -
Figure 6

Forest plot of comparison: 1 Tumour bed boost radiotherapy versus no boost, outcome: 1.8 Cosmesis, Panel.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 1 Local control.
Figuras y tablas -
Analysis 1.1

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 1 Local control.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 2 Local control ‐ sensitivity analysis.
Figuras y tablas -
Analysis 1.2

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 2 Local control ‐ sensitivity analysis.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 3 Local control ‐ subgroup +40 years.
Figuras y tablas -
Analysis 1.3

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 3 Local control ‐ subgroup +40 years.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 4 Local control ‐ subgroup low boost dose.
Figuras y tablas -
Analysis 1.4

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 4 Local control ‐ subgroup low boost dose.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 5 Overall survival.
Figuras y tablas -
Analysis 1.5

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 5 Overall survival.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 6 Disease‐free survival.
Figuras y tablas -
Analysis 1.6

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 6 Disease‐free survival.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 7 Late toxicity, pBRA.
Figuras y tablas -
Analysis 1.7

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 7 Late toxicity, pBRA.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 8 Cosmesis, Panel.
Figuras y tablas -
Analysis 1.8

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 8 Cosmesis, Panel.

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 9 Cosmesis, Physician.
Figuras y tablas -
Analysis 1.9

Comparison 1 Tumour bed boost radiotherapy versus no boost, Outcome 9 Cosmesis, Physician.

Summary of findings for the main comparison. Boost compared to no boost in breast cancer radiotherapy

Boost compared to no boost in breast cancer radiotherapy

Patient or population: breast‐conserving radiotherapy for breast cancer
Setting: radiotherapy centres
Intervention: boost
Comparison: no boost

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk without boost

Risk with boost

Local control: recurrence at 5 years

Study population

HR 0.64
(0.55 to 0.75)

8315
(5 RCTs)

⊕⊕⊝⊝
LOW 1 2

Nice did not report 5‐year local control.

70 per 1000

45 per 1000
(39 to 53)

Overall survival: mortality at 5 years

Study population

HR 1.04
(0.94 to 1.14)

6342
(2 RCTs)

⊕⊕⊕⊝
MODERATE 3

91 per 1000

94 per 1000
(86 to 103)

Disease‐free survival: disease progression/mortality at 5 years

Study population

HR 0.94
(0.87 to 1.02)

6549
(3 RCTs)

⊕⊕⊝⊝
LOW 4 5

EORTC did not report 5‐year disease‐free survival.

221 per 1000

209 per 1000
(195 to 224)

Late toxicity, pBRA

Mean pBRA 8.17, range 7.55 to 10

Mean pBRA 8.55,
range 8.26 to 9.4

MD 0.38 higher

(0.18 lower to 0.93 higher)

1526
(2 RCTs)

⊕⊝⊝⊝
VERY LOW 2 5

Cosmesis, panel scored: fair or poor

Study population

OR 1.41
(1.07 to 1.85)

1116
(2 RCTs)

⊕⊕⊝⊝
LOW 1 2

202 per 1000

263 per 1000
(213 to 319)

Cosmesis, physician‐scored: fair or poor

Study population

OR 1.58
(0.93 to 2.69)

592
(2 RCTs)

⊕⊝⊝⊝
VERY LOW 1 5 6

85 per 1000

128 per 1000
(80 to 200)

Sensitivity analysis: local control ‐ recurrence at 5 years

Study population

HR 0.62
(0.52 to 0.73)

6963
(3 RCTs)

⊕⊕⊕⊕
HIGH

74 per 1000

47 per 1000
(39 to 55)

Subgroup analysis: local control ‐ > 40 years old, recurrence at 5 years

Study population

HR 0.65
(0.53 to 0.81)

5058
(2 RCTs)

⊕⊕⊕⊕
HIGH

59 per 1000

39 per 1000
(32 to 48)

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; HR: hazard ratio; MD: mean difference; OR: odds ratio; pBRA: percentage breast retraction assessment; RCT: randomised controlled trial

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: We are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low quality: Our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of the effect.

1High risk of selective reporting in one study.
2There was considerable clinical heterogeneity with respect to radiotherapy dose and the use of quality assurance procedures.
3One of the two studies has a very wide confidence interval.
4There was considerable clinical heterogeneity on statistical testing, but not on visual inspection.
5Both studies have wide confidence intervals.
6High risk of bias for blinding of outcome assessment in one study.

Figuras y tablas -
Summary of findings for the main comparison. Boost compared to no boost in breast cancer radiotherapy
Table 1. RTOG/EORTC late radiation morbidity scoring scheme

Organ tissue

0

Grade 1

Grade 2

Grade 3

Grade 4

Skin

None

Slight atrophy; pigmentation change; some hair loss

Patchy atrophy; moderate telangiectasia; total hair loss

Marked atrophy; gross telangiectasia

Ulceration

Subcutaneous tissue

None

Slight induration (fibrosis) and loss of subcutaneous fat

Moderate fibrosis but asymptomatic; slight field contracture; < 10% linear reduction

Severe induration and loss of subcutaneous tissue; field contracture > 10% linear measurement

Necrosis

RTOG/EORTC: Radiation Therapy Oncology Group/European Organisation for Research and Treatment of Cancer

Figuras y tablas -
Table 1. RTOG/EORTC late radiation morbidity scoring scheme
Table 2. Harvard cosmetic score

Cosmetic score

Excellent

Good

Fair

Poor

Figuras y tablas -
Table 2. Harvard cosmetic score
Comparison 1. Tumour bed boost radiotherapy versus no boost

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Local control Show forest plot

5

8315

Hazard Ratio (Fixed, 95% CI)

0.64 [0.55, 0.75]

2 Local control ‐ sensitivity analysis Show forest plot

3

6963

Hazard Ratio (Fixed, 95% CI)

0.62 [0.52, 0.73]

3 Local control ‐ subgroup +40 years Show forest plot

2

5058

Hazard Ratio (Fixed, 95% CI)

0.65 [0.53, 0.81]

4 Local control ‐ subgroup low boost dose Show forest plot

2

1352

Hazard Ratio (Fixed, 95% CI)

0.95 [0.55, 1.62]

5 Overall survival Show forest plot

2

6342

Hazard Ratio (Fixed, 95% CI)

1.04 [0.94, 1.14]

6 Disease‐free survival Show forest plot

3

6549

Hazard Ratio (Fixed, 95% CI)

0.94 [0.87, 1.02]

7 Late toxicity, pBRA Show forest plot

2

1526

Mean Difference (IV, Fixed, 95% CI)

0.38 [‐0.18, 0.93]

8 Cosmesis, Panel Show forest plot

2

1116

Odds Ratio (M‐H, Fixed, 95% CI)

1.41 [1.07, 1.86]

9 Cosmesis, Physician Show forest plot

2

592

Odds Ratio (M‐H, Fixed, 95% CI)

1.58 [0.93, 2.69]

Figuras y tablas -
Comparison 1. Tumour bed boost radiotherapy versus no boost