Z Gastroenterol 2019; 57(03): 336-418
DOI: 10.1055/a-0833-5712
Leitlinie
© Georg Thieme Verlag KG Stuttgart · New York

S3-Leitlinie – Diagnostik und Therapie der Plattenepithelkarzinome und Adenokarzinome des Ösophagus

Langversion 2.0 – Dezember 2018 AWMF-Registernummer: 021/023OL
Rainer Porschen
1   Klinik für Innere Medizin, Klinikum Bremen Ost, Bremen
,
Wolfgang Fischbach
2   Medizinische Klinik II, Klinikum Aschaffenburg, Aschaffenburg
,
Ines Gockel
3   Klinik und Poliklinik für Viszeralchirurgie, Universitätsklinikum, Leipzig
,
Stephan Hollerbach
4   Klinik für Innere Medizin, Allgemeines Krankenhaus, Celle
,
Arnulf Hölscher
5   Klinik für Allgemein- und Viszeralchirurgie, Agaplesion Markus Krankenhaus, Frankfurt
,
Petra Lynen Jansen
6   Deutsche Gesellschaft für Verdauungs- und Stoffwechselkrankheiten, Berlin
,
Stephan Miehlke
7   Magen-Darm-Zentrum, Hamburg-Eppendorf
,
Oliver Pech
8   Klinik für Gastroenterolgie und Interventionelle Endoskopie, Krankenhaus Barmherzige Brüder, Regensburg
,
Michael Stahl
9   Klinik für Internistische Onkologie und Hämatologie, Kliniken Essen-Mitte, Essen
,
Peter Thuss-Patience
10   Klinik für Hämatologie und Onkologie, Charité Campus Virchow Klinikum, Berlin
,
Udo Vanhoefer
11   Zentrum für Innere Medizin, Marienkrankenhaus, Hamburg
,
und die Mitarbeiter der Leitlinienkommission,
Collaborators:
› Author Affiliations
Further Information

Publication History

Publication Date:
12 March 2019 (online)

Wesentliche Neuerungen durch die Aktualisierung der Leitlinie (Version 2.0, 2018)

In Rahmen der Aktualisierung wurden alle Empfehlungen auf Aktualität geprüft. Hierzu erfolgte eine systematische Recherche für priorisierte Themen sowie eine Befragung der beteiligten Fachexperten. Im Rahmen des Aktualisierungsprozesses wurden die unten aufgeführten Empfehlungen überarbeitet. Hierbei wurden teilweise lediglich die Einschätzungen bzgl. der Evidenzklassifikation aufgrund neuer Studiendaten verändert (mit einem * markiert). Eine detaillierte Übersicht der Änderungen befindet sich in [Tab. 15].

*Statement 4.2. (Alkohol als Risikofaktor)

*Statement 4.7. (Gastroösophagealer Reflux als Risikofaktor)

Empfehlungen 6.4.und 6.5. (Chromoendoskopie (Lugol’sche Lösung) oder computergestützte digitale (Filter-)Verfahren als erweiterte Diagnostik)

*Empfehlung 6.6. (Endoskopischer Ultraschall als Bestandteil des Stagings)

Empfehlung 6.13. (PET/CT bei lokal fortgeschrittenen Tumoren)

Empfehlung 6.16. (Diagnostische Laparoskopie)

Empfehlung 6.18. (Pathologische Zweitmeinung bei histologischer Diagnose einer IEN/Dysplasie)

*Empfehlung 6.23. (Aussagen zum Regressions-Score im pathologischen Befund)

Empfehlung 8.10. (Resektionsausmaß)

 
  • Literatur

  • 1 RKI and GEKID. Krebs in Deutschland 2009/2010, in Beiträge zur Gesundheitsberichterstattung des Bundes. Berlin: Robert Koch-Institut, Gesellschaft der epidemiologischen Krebsregister in Deutschland e.V; 2013
  • 2 Leitlinienprogramm Onkologie, S3-Leitlinie Diagnostik und Therapie der Plattenepithelkarzinome und Adenokarzinome des Ösophagus, Langversion 1.0. 2015 https://www.leitlinienprogramm-onkologie.de/leitlinien/oesophaguskarzinom/ Deutsche Krebsgesellschaft, Deutsche Krebshilfe, AWMF: AWMF-Registernummer: 021/023OL
  • 3 Bayley MT. et al. Where to build the bridge between evidence and practice?: results of an international workshop to prioritize knowledge translation activities in traumatic brain injury care. J Head Trauma Rehabil 2014; 29: 268-276
  • 4 Dear RF. et al. Identifying and prioritising gaps in colorectal cancer trials research in Australia. Med J Aust 2012; 197: 507-511
  • 5 Miake-Lye IM. et al. What is an evidence map? A systematic review of published evidence maps and their definitions, methods, and products. Syst Rev 2016; 5: 28
  • 6 Clavisi O. et al. Effective stakeholder participation in setting research priorities using a Global Evidence Mapping approach. J Clin Epidemiol 2013; 66: 496-502.e2
  • 7 Tramacere I, La VC, Negri E. Tobacco smoking and esophageal and gastric cardia adenocarcinoma: a meta-analysis. Epidemiology 2011; 22: 344-349
  • 8 Lee CH. et al. Anatomical subsite discrepancy in relation to the impact of the consumption of alcohol, tobacco and betel quid on esophageal cancer. Int J Cancer 2007; 120: 1755-1762
  • 9 Pandey N. et al. Gastro-oesophageal reflux symptoms and the risks of oesophageal cancer: are the effects modified by smoking, NSAIDs or acid suppressants?. Gut 2010; 59: 31-38
  • 10 Rafiq R. et al. Secondhand Smoking and the Risk of Esophageal Squamous Cell Carcinoma in a High Incidence Region, Kashmir, India: A Case-control-observational Study. Medicine (Baltimore) 2016; 95: e2340
  • 11 Sewram V. et al. Tobacco and alcohol as risk factors for oesophageal cancer in a high incidence area in South Africa. Cancer Epidemiol 2016; 41: 113-121
  • 12 Islami F. et al. Alcohol drinking and esophageal squamous cell carcinoma with focus on lightdrinkers and never-smokers: a systematic review and meta-analysis. Int J Cancer 2011; 129: 2473-2484
  • 13 Bagnardi V. et al. Light alcohol drinking and cancer: a meta-analysis. AnnOncol 2013; 24: 301-308
  • 14 Freedman ND. et al. Alcohol intake and risk of oesophageal adenocarcinoma: a pooled analysis from the BEACON Consortium. Gut 2011; 60: 1029-1037
  • 15 Pandeya N. et al. Alcohol consumption and the risks of adenocarcinoma and squamous cell carcinoma of the esophagus. Gastroenterology 2009; 136: 1215-1224, e1–2
  • 16 Tramacere I. et al. A meta-analysis on alcohol drinking and esophageal and gastric cardia adenocarcinoma risk. Ann Oncol 2012; 23: 287-297
  • 17 Ji J, Sundquist J, Sundquist K. Associations of alcohol use disorders with esophageal and gastric cancers: a population-based study in Sweden. Eur J Cancer Prev 2017; 26: 119-124
  • 18 Moehler M. et al. S3-Leitlinie „Magenkarzinom“. Z Gastroenterol 2011; 49: 461-531
  • 19 Guh DP. et al. The incidence of co-morbidities related to obesity and overweight: a systematic review and meta-analysis. BMC Public Health 2009; 9: 88
  • 20 Ryan AM. et al. Obesity, metabolic syndrome and esophageal adenocarcinoma: epidemiology, etiology and new targets. Cancer Epidemiol 2011; 35: 309-319
  • 21 Turati F. et al. A meta-analysis of body mass index and esophageal and gastric cardia adenocarcinoma. Ann Oncol 2013; 24: 609-617
  • 22 O’Doherty MG. et al. A prospective cohort study of obesity and risk of oesophageal and gastric adenocarcinoma in the NIH-AARP Diet and Health Study. Gut 2012; 61: 1261-1268
  • 23 Thota PN. et al. Influence of body mass index on the prevalence and progression of dysplasia in Barrett’s esophagus: a retrospective analysis (.). Scand J Gastroenterol 2016; 51: 1288-1293
  • 24 Krishnamoorthi R. et al. Rates and predictors of progression to esophageal carcinoma in a large population-based Barrett’s esophagus cohort. Gastrointest Endosc 2016; 84: 40-46.e7
  • 25 Leeuwenburgh I. et al. Long-term esophageal cancer risk in patients with primary achalasia: a prospective study. Am J Gastroenterol 2010; 105: 2144-2149
  • 26 Zendehdel K. et al. Risk of esophageal adenocarcinoma in achalasia patients, a retrospective cohort study in Sweden. Am J Gastroenterol 2011; 106: 57-61
  • 27 Ji J, Hemminki K. Familial risk for esophageal cancer: an updated epidemiologic study from Sweden. Clin Gastroenterol Hepatol 2006; 4: 840-845
  • 28 Morton LM. et al. Risk of treatment-related esophageal cancer among breast cancer survivors. Ann Oncol 2012; 23: 3081-3091
  • 29 Zablotska LB. et al. Increased risk of squamous cell esophageal cancer after adjuvant radiation therapy for primary breast cancer. Am J Epidemiol 2005; 161: 330-337
  • 30 Levi F. et al. Increased risk of esophageal cancer after breast cancer. Ann Oncol 2005; 16: 1829-1831
  • 31 Roychoudhuri R. et al. Radiation-induced malignancies following radiotherapy for breast cancer. Br J Cancer 2004; 91: 868-872
  • 32 Okonta KE, Tettey M, Abubakar U. In patients with corrosive oesophageal stricture for surgery, is oesophagectomy rather than bypass necessary to reduce the risk of oesophageal malignancy?. Interact Cardiovasc Thorac Surg 2012; 15: 713-715
  • 33 Lagergren J. et al. Symptomatic gastroesophageal reflux as a risk factor for esophageal adenocarcinoma. N Engl J Med 1999; 340: 825-831
  • 34 Velanovich V. et al. Relationship of gastroesophageal reflux disease with adenocarcinoma of the distal esophagus and cardia. Dig Surg 2002; 19: 349-353
  • 35 Wu AH, Tseng CC, Bernstein L. Hiatal hernia, reflux symptoms, body size, and risk of esophageal and gastric adenocarcinoma. Cancer 2003; 98: 940-948
  • 36 Chak A. et al. Gastroesophageal reflux symptoms in patients with adenocarcinoma of the esophagus or cardia. Cancer 2006; 107: 2160-2166
  • 37 Rubenstein JH, Taylor JB. Meta-analysis: the association of oesophageal adenocarcinoma with symptoms of gastro-oesophageal reflux. Aliment Pharmacol Ther 2010; 32: 1222-1227
  • 38 Hazelton WD. et al. The Role of Gastroesophageal Reflux and Other Factors during Progression to Esophageal Adenocarcinoma. Cancer Epidemiol Biomarkers Prev 2015; 24: 1012-1023
  • 39 Hvid-Jensen F. et al. Incidence of adenocarcinoma among patients with Barrett’s esophagus. N Engl J Med 2011; 365: 1375-1383
  • 40 Wani S. et al. Patients with nondysplastic Barrett’s esophagus have low risks for developing dysplasia or esophageal adenocarcinoma. Clin Gastroenterol Hepatol 2011; 9: 220-227 ; quiz e26
  • 41 Anaparthy R. et al. Association between length of Barrett’s esophagus and risk of high-grade dysplasia or adenocarcinoma in patients without dysplasia. Clin Gastroenterol Hepatol 2013; 11: 1430-1436
  • 42 Gaddam S. et al. Persistence of nondysplastic Barrett’s esophagus identifies patients at lower risk for esophageal adenocarcinoma: results from a large multicenter cohort. Gastroenterology 2013; 145: 548-553.e1
  • 43 Desai TK. et al. The incidence of oesophageal adenocarcinoma in non-dysplastic Barrett’s oesophagus: a meta-analysis. Gut 2012; 61: 970-976
  • 44 Solaymani-Dodaran M, Card TR, West J. et al. Cause-specific mortality of people with Barrett’s esophagus compared with the general population: a population-based cohort study. Gastroenterology 2013; 144: 1375-1383, 1383.e1
  • 45 Koop H. et al. S2k guideline: gastroesophageal reflux disease guided by the German Society of Gastroenterology: AWMF register no. 021-013. Z Gastroenterol 2014; 52: 1299-1346
  • 46 Cooper S. et al. Risk factors for the development of oesophageal adenocarcinoma in Barrett’s oesophagus: a UK primary care retrospective nested case-control study. United European Gastroenterol J 2014; 2: 91-98
  • 47 Nguyen T. et al. The Annual Risk of Esophageal Adenocarcinoma Does Not Decrease Over Time in Patients With Barrett’s Esophagus. Am J Gastroenterol 2017; 112: 1049-1055
  • 48 Masclee GM. et al. The incidence of Barrett’s oesophagus and oesophageal adenocarcinoma in the United Kingdom and The Netherlands is levelling off. Aliment Pharmacol Ther 2014; 39: 1321-1330
  • 49 Kestens C. et al. Patients With Barrett’s Esophagus and Persistent Low-grade Dysplasia Have an Increased Risk for High-grade Dysplasia and Cancer. Clin Gastroenterol Hepatol 2016; 14: 956-962.e1
  • 50 Steinberg J. et al. Koinzidenz von Plattenepithelkarzinomen der Speiseröhre und Kopf-Hals-Karzinomen: Risiko und Früherkennung. Tumor Diagn u Ther 2008; 29: 35-39
  • 51 Moschler O. et al. Chromoendoscopy is a valuable tool for screening of high-risk patients with head and neck cancer for early detection of esophageal cancer. Digestion 2006; 73: 160-166
  • 52 Ljung R, Martin L, Lagergren J. Oral disease and risk of oesophageal and gastric cancer in a nationwide nested case-control study in Sweden. Eur J Cancer 2011; 47: 2128-2132
  • 53 Allum WH. et al. Guidelines for the management of oesophageal and gastric cancer. Gut 2011; 60: 1449-1472
  • 54 Rothwell PM. et al. Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomised trials. Lancet 2011; 377: 31-41
  • 55 Algra AM, Rothwell PM. Effects of regular aspirin on long-term cancer incidence and metastasis: a systematic comparison of evidence from observational studies versus randomised trials. Lancet Oncol 2012; 13: 518-527
  • 56 Cuzick J. et al. Estimates of benefits and harms of prophylactic use of aspirin in the general population. Ann Oncol 2015; 26: 47-57
  • 57 Cook NR. et al. Alternate-day, low-dose aspirin and cancer risk: long-term observational follow-up of a randomized trial. Ann Intern Med 2013; 159: 77-85
  • 58 Cardwell CR. et al. Exposure to oral bisphosphonates and risk of cancer. Int J Cancer 2012; 131: E717-E725
  • 59 Green J. et al. Oral bisphosphonates and risk of cancer of oesophagus, stomach, and colorectum: case-control analysis within a UK primary care cohort. BMJ 2010; 341: c4444
  • 60 Oh YH, Yoon C, Park SM. Bisphosphonate use and gastrointestinal tract cancer risk: meta-analysis of observational studies. World J Gastroenterol 2012; 18: 5779-5788
  • 61 Haber SL, McNatty D. An evaluation of the use of oral bisphosphonates and risk of esophageal cancer. Ann Pharmacother 2012; 46: 419-423
  • 62 Andrici J, Tio M, Eslick GD. Meta-analysis: oral bisphosphonates and the risk of oesophageal cancer. Aliment Pharmacol Ther 2012; 36: 708-716
  • 63 Busby J. et al. The effect of medications which cause inflammation of the gastro-oesophageal tract on cancer risk: a nested case-control study of routine Scottish data. Int J Cancer 2017; 140: 1828-1835
  • 64 Bjelakovic G. et al. Antioxidant supplements for preventing gastrointestinal cancers. Cochrane Database Syst Rev 2008; 3: CD004183
  • 65 Vigen C, Bernstein L, Wu AH. Occupational physical activity and risk of adenocarcinomas of the esophagus and stomach. Int J Cancer 2006; 118: 1004-1009
  • 66 Stehle P. Deutsche Gesellschaft für Ernährung. 12. Ernährungsbericht 2012. European Journal of Nutrition an Food Safety 2012; 4: 2347-2564
  • 67 Liu J. et al. Intake of fruit and vegetables and risk of esophageal squamous cell carcinoma: a meta-analysis of observational studies. Int J Cancer 2013; 133: 473-485
  • 68 Choi Y. et al. Consumption of red and processed meat and esophageal cancer risk: metaanalysis. World J Gastroenterol 2013; 19: 1020-1029
  • 69 Keszei AP. et al. Red and processed meat consumption and the risk of esophageal and gastric cancer subtypes in The Netherlands Cohort Study. Ann Oncol 2012; 23: 2319-2326
  • 70 Huang W. et al. Red and processed meat intake and risk of esophageal adenocarcinoma: a meta-analysis of observational studies. Cancer Causes Control 2013; 24: 193-201
  • 71 Jakszyn P. et al. Meat and heme iron intake and esophageal adenocarcinoma in the European Prospective Investigation into Cancer and Nutrition study. Int J Cancer 2013; 133: 2744-2750
  • 72 Salehi M. et al. Meat, fish, and esophageal cancer risk: a systematic review and dose- response meta-analysis. Nutr Rev 2013; 71: 257-267
  • 73 Palmer JB, Drennan JC, Baba M. Evaluation and treatment of swallowing impairments. Am Fam Physician 2000; 61: 2453-2462
  • 74 Behrens A. et al. How safe is sedation in gastrointestinal endoscopy? A multicentre analysis of 388,404 endoscopies and analysis of data from prospective registries of complications managed by members of the Working Group of Leading Hospital Gastroenterologists (ALGK). Z Gastroenterol 2013; 51: 432-436
  • 75 Hori K. et al. Lugol-voiding lesions are an important risk factor for a second primary squamous cell carcinoma in patients with esosphageal cancer or head and neck cancer. Am J Gastroenterol 2011; 106: 858-866
  • 76 Qumseya BJ. et al. Advanced imaging technologies increase detection of dysplasia and neoplasia in patients with Barrett’s esophagus: a meta-analysis and systematic review. Clin Gastroenterol Hepatol 2013; 11: 1562-1570.e1–2
  • 77 Curvers WL. et al. Endoscopic tri-modal imaging is more effective than standard endoscopy in identifying early-stage neoplasia in Barrett’s esophagus. Gastroenterology 2010; 139: 1106-1114
  • 78 Chung CS. et al. Image-enhanced endoscopy for detection of second primary neoplasm in patients with esophageal and head and neck cancer: A systematic review and meta-analysis. Head Neck 2016; 38: E2343-E2349
  • 79 Uedo N. et al. Role of narrow band imaging for diagnosis of early-stage esophagogastric cancer: current consensus of experienced endoscopists in Asia-Pacific region. Dig Endosc 2011; 23: 58-71
  • 80 Curvers WL. et al. Low-grade dysplasia in Barrett’s esophagus: overdiagnosed and underestimated. Am J Gastroenterol 2010; 105: 1523-1530
  • 81 Sharma P. et al. Standard endoscopy with random biopsies versus narrow band imaging targeted biopsies in Barrett’s oesophagus: a prospective, international, randomised controlled trial. Gut 2013; 62: 15-21
  • 82 Canto MI. et al. In vivo endomicroscopy improves detection of Barrett’s esophagus-related neoplasia: a multicenter international randomized controlled trial (with video). Gastrointest Endosc 2014; 79: 211-221
  • 83 Fugazza A. et al. Confocal Laser Endomicroscopy in Gastrointestinal and Pancreatobiliary Diseases: A Systematic Review and Meta-Analysis. Biomed Res Int 2016; 2016: 4638683
  • 84 Gupta A. et al. Utility of confocal laser endomicroscopy in identifying high-grade dysplasia and adenocarcinoma in Barrett’s esophagus: a systematic review and meta-analysis. Eur J Gastroenterol Hepatol 2014; 26: 369-377
  • 85 Coletta M. et al. Acetic acid chromoendoscopy for the diagnosis of early neoplasia and specialized intestinal metaplasia in Barrett’s esophagus: a meta-analysis. Gastrointest Endosc 2016; 83: 57-67.e1
  • 86 Qumseya B. et al. Advanced imaging technologies increase detection of dysplasia and neoplasia in patients with Barrett’s esophagus: a meta-analysis and systematic review (Provisional abstract). Clinical Gastroenterology and Hepatology 2013; 11: 1562-1570.e2
  • 87 Lim H. et al. Clinical significance of early detection of esophageal cancer in patients with head and neck cancer. Gut Liver 2015; 9: 159-165
  • 88 Katada C. et al. Risk of superficial squamous cell carcinoma developing in the head and neck region in patients with esophageal squamous cell carcinoma. Laryngoscope 2012; 122: 1291-1296
  • 89 Chow TL. et al. Prediction of simultaneous esophageal lesions in head and neck squamous cell carcinoma: a multivariate analysis. Arch Otolaryngol Head Neck Surg 2009; 135: 882-885
  • 90 Muto M. et al. Association of multiple Lugol-voiding lesions with synchronous and metachronous esophageal squamous cell carcinoma in patients with head and neck cancer. Gastrointest Endosc 2002; 56: 517-521
  • 91 Gupta A. et al. Utility of confocal laser endomicroscopy in identifying high-grade dysplasia and adenocarcinoma in Barrett’s esophagus: a systematic review and meta-analysis (Provisional abstract). Database of Abstracts of Reviews of Effects 2014; 26: 369-377
  • 92 Thosani N. et al. Diagnostic accuracy of EUS in differentiating mucosal versus submucosal invasion of superficial esophageal cancers: a systematic review and meta-analysis. Gastrointest Endosc 2012; 75: 242-253
  • 93 Puli SR. et al. Staging accuracy of esophageal cancer by endoscopic ultrasound: a metaanalysis and systematic review. World J Gastroenterol 2008; 14: 1479-1490
  • 94 van Vliet EP. et al. Staging investigations for oesophageal cancer: a meta-analysis. Br J Cancer 2008; 98: 547-557
  • 95 Tranchemontagne J. Stadification initiale du cancer de l’oesophage: revue systematique sur la performance des methodes diagnostiques. Initial staging of oesophageal cancer: systematic review of the performance of diagnostic methods. Agence d’evaluation des technologies et des modes d’intervention en sante (AETMIS). ETMIS 5(6). Montreal: 2009
  • 96 Russell I. et al. Cancer of oesophagus or gastricus: New assessment of technology of endosonography (COGNATE): Report of pragmatic randomised trial. Health technology assessment 2013; 17: 1-13 . doi:10.3310/hta17390
  • 97 Luo LN. et al. Endoscopic Ultrasound for Preoperative Esophageal Squamous Cell Carcinoma: a Meta-Analysis. PLoS One 2016; 11: e0158373
  • 98 Takizawa K. et al. Lymph node staging in esophageal squamous cell carcinoma: a comparative study of endoscopic ultrasonography versus computed tomography. J Gastroenterol Hepatol 2009; 24: 1687-1691
  • 99 Stiles BM. et al. Clinical T2-T3N0M0 esophageal cancer: the risk of node positive disease. Ann Thorac Surg 2011; 92: 491-496 ; discussion 496–498
  • 100 Oldenburg A, Albrecht T. Sonografische Leberdiagnostik bei Tumorpatienten ohne und mit Kontrastmittel. Ultraschall in Med 2008; 29: 488-498
  • 101 Kinkel K. et al. Detection of hepatic metastases from cancers of the gastrointestinal tract by using noninvasive imaging methods (US, CT, MR imaging, PET): a meta-analysis. Radiology 2002; 224: 748-756
  • 102 Piscaglia F. et al. Real time contrast enhanced ultrasonography in detection of liver metastases from gastrointestinal cancer. BMC Cancer 2007; 7: 171
  • 103 Dietrich CF. et al. Assessment of metastatic liver disease in patients with primary extrahepatic tumors by contrast-enhanced sonography versus CT and MRI. World J Gastroenterol 2006; 12: 1699-1705
  • 104 Albrecht T. et al. Improved detection of hepatic metastases with pulse-inversion US during the liver-specific phase of SHU 508A: multicenter study. Radiology 2003; 227: 361-370
  • 105 Hanle MM. et al. Screening for liver metastases in women with mammary carcinoma: comparison of contrast-enhanced ultrasound and magnetic resonance imaging. Clin Imaging 2011; 35: 366-370
  • 106 Seitz K. et al. Contrast-Enhanced Ultrasound (CEUS) for the characterization of focal liver lesions prospective comparison in clinical practice: CEUS vs. CT (DEGUM multicenter trial). Parts of this manuscript were presented at the Ultrasound Dreilandertreffen 2008, Davos. Ultraschall in Med 2009; 30: 383-389
  • 107 Seitz K. et al. Contrast-enhanced ultrasound (CEUS) for the characterization of focal liver lesions in clinical practice (DEGUM Multicenter Trial): CEUS vs. MRI--a prospective comparison in 269 patients. Ultraschall in Med 2010; 31: 492-499
  • 108 Strobel D. et al. Tumor-specific vascularization pattern of liver metastasis, hepatocellular carcinoma, hemangioma and focal nodular hyperplasia in the differential diagnosis of 1,349 liver lesions in contrast-enhanced ultrasound (CEUS). Ultraschall in Med 2009; 30: 376-382
  • 109 Westwood M. et al. Contrast-enhanced ultrasound using SonoVue(R) (sulphur hexafluoride microbubbles) compared with contrast-enhanced computed tomography and contrastenhanced magnetic resonance imaging for the characterisation of focal liver lesions and detection of liver metastases: a systematic review and cost-effectiveness analysis. Health Technol Assess 2013; 17: 1-243
  • 110 Doldi SB. et al. Ultrasonographic evaluation of the cervical lymph nodes in preoperative staging of esophageal neoplasms. Abdom Imaging 1998; 23: 275-257
  • 111 Lerut T. et al. Clinical Practice Guidelines Upper Gastrointestinal Cancer update. 2012
  • 112 Schreurs LM. et al. Current relevance of cervical ultrasonography in staging cancer of the esophagus and gastroesophageal junction. Eur J Radiol 2008; 67: 105-111
  • 113 Van Overhagen H. et al. Improved assessment of supraclavicular and abdominal metastases in oesophageal and gastro-oesophageal junction carcinoma with the combination of ultrasound and computed tomography. Br J Radiol 1993; 66: 203-208
  • 114 van Vliet EP. et al. Detection of distant metastases in patients with oesophageal or gastric cardia cancer: a diagnostic decision analysis. Br J Cancer 2007; 97: 868-876
  • 115 Omloo JM. et al. Additional value of external ultrasonography of the neck after CT and PET scanning in the preoperative assessment of patients with esophageal cancer. Dig Surg 2009; 26: 43-49
  • 116 van Overhagen H. et al. Assessment of distant metastases with ultrasound-guided fine-needle aspiration biopsy and cytologic study in carcinoma of the esophagus and gastroesophageal junction. Gastrointest Radiol 1992; 17: 305-310
  • 117 van Overhagen H. et al. Supraclavicular lymph node metastases in carcinoma of the esophagus and gastroesophageal junction: assessment with CT, US, and US-guided fine-needle aspiration biopsy. Radiology 1991; 179: 155-158
  • 118 Mori S. et al. Preoperative assessment of resectability for carcinoma of the thoracic esophagus. Part I. Esophagogram and azygogram. Ann Surg 1979; 190: 100-105
  • 119 Kumbasar B. Carcinoma of esophagus: radiologic diagnosis and staging. Eur J Radiol 2002; 42: 170-180
  • 120 Fiore D. et al. Multimodal imaging of esophagus and cardia cancer before and after treatment. Radiol Med 2006; 111: 804-817
  • 121 Moreto M. Diagnosis of esophagogastric tumors. Endoscopy 2005; 37: 26-32
  • 122 Kim TJ. et al. Multimodality assessment of esophageal cancer: preoperative staging and monitoring of response to therapy. Radiographics 2009; 29: 403-421
  • 123 Pech O. et al. The Impact of Endoscopic Ultrasound and Computed Tomography on the TNM Staging of Early Cancer in Barrett’s Esophagus. Am J Gastroenterol 2006; 101: 2223-2229
  • 124 Quint LE. Thoracic complications and emergencies in oncologic patients. Cancer Imaging 2009; 9 Spec No A: S75-S82
  • 125 Makarawo TP. et al. Water as a contrast medium: a re-evaluation using the multidetectorrow computed tomography. Am Surg 2013; 79: 728-733
  • 126 Kamel IR, Fishman EK. Recent advances in CT imaging of liver metastases. Cancer J 2004; 10: 104-120
  • 127 Network S.I.G. Scottish Intercollegiate Guidelines Network Management of oesophageal and gastric cancer. A national clinical guideline. 2006
  • 128 Gollub MJ. et al. Pelvic CT in patients with esophageal cancer. Am J Roentgenol 2005; 184: 487-490
  • 129 Ba-Ssalamah A. et al. Accuracy of hydro-multidetector row CT in the local T staging of oesophageal cancer compared to postoperative histopathological results. Eur Radiol 2011; 21: 2326-2335
  • 130 Takizawa K. et al. Lymph node staging in esophageal squamous cell carcinoma: a comparative study of endoscopic ultrasonography versus computed tomography. J Gastroenterol Hepatol 2009; 24: 1687-1691
  • 131 Choi J. et al. Comparison of endoscopic ultrasonography (EUS), positron emission tomography (PET), and computed tomography (CT) in the preoperative locoregional staging of resectable esophageal cancer. Surg Endosc 2010; 24: 1380-1386
  • 132 Lowe VJ. et al. Comparison of positron emission tomography, computed tomography, and endoscopic ultrasound in the initial staging of patients with esophageal cancer. Mol Imaging Biol 2005; 7: 422-430
  • 133 Heger U. et al. Is preoperative chemotherapy followed by surgery the appropriate treatment for signet ring cell containing adenocarcinomas of the esophagogastric junction and stomach?. Ann Surg Oncol 2014; 21: 1739-1748
  • 134 Blank S. et al. A reliable risk score for stage IV esophagogastric cancer. Eur J Surg Oncol 2013; 39: 823-830
  • 135 Blank S. et al. Preoperative therapy of esophagogastric cancer: the problem of nonresponding patients. Langenbecks Arch Surg 2013; 398: 211-220
  • 136 Blank S. et al. Impact of pretherapeutic routine clinical staging for the individualization of treatment in gastric cancer patients. Langenbecks Arch Surg 2012; 397: 45-55
  • 137 Sohn KM. et al. Comparing MR imaging and CT in the staging of gastric carcinoma. Am J Roentgenol 2000; 174: 1551-1557
  • 138 Anzidei M. et al. Diagnostic performance of 64-MDCT and 1.5-T MRI with high-resolution sequences in the T staging of gastric cancer: a comparative analysis with histopathology. Radiol Med 2009; 114: 1065-1079
  • 139 Lauenstein TC. et al. Whole-body MR imaging: evaluation of patients for metastases. Radiology 2004; 233: 139-148
  • 140 Wong R, Malthaner R. Esophageal cancer: a systematic review. Curr Probl Cancer 2000; 24: 297-373
  • 141 Weber MA. et al. Assessment of diffusion-weighted MRI and 18F-fluoro-deoxyglucose PET/CT in monitoring early response to neoadjuvant chemotherapy in adenocarcinoma of the esophagogastric junction. J Gastrointestin Liver Dis 2013; 22: 45-52
  • 142 Quint LE, Bogot NR. Staging esophageal cancer. Cancer Imaging 2008; 8 Spec No A: S33-S42
  • 143 Medical Services Advisory Committee. Positron emission tomography for oesophageal and gastric cancer: assessment report/prepared by the Medical Services Advisory Committee with the assistance of Silke Walleser et al. Australia: 2008
  • 144 Ballini L. et al. Criteria for appropriate use of FDG-PET in esophageal cancer. Dossier 209-2011 ISSN 1591-223X. 2011. 2011 Bologna: Agenzia sanitaria e sociale regionale, Regione Emilia-Romagna (ASSR). . ORIentamenti 4
  • 145 Choi JY. et al. Improved detection of individual nodal involvement in squamous cell carcinoma of the esophagus by FDG PET. J Nucl Med 2000; 41: 808-815
  • 146 Flamen P. et al. Utility of positron emission tomography for the staging of patients with potentially operable esophageal carcinoma. J Clin Oncol 2000; 18: 3202-3210
  • 147 Downey RJ. et al. Whole body 18FDG-PET and the response of esophageal cancer to induction therapy: results of a prospective trial. J Clin Oncol 2003; 21: 428-432
  • 148 Heeren PA. et al. Detection of distant metastases in esophageal cancer with (18)F-FDG PET. J Nucl Med 2004; 45: 980-987
  • 149 Noble F. et al. Impact of integrated PET/CT in the staging of oesophageal cancer: a UK population-based cohort study. Clin Radiol 2009; 64: 699-705
  • 150 Kato H, Nakajima M. The efficacy of FDG-PET for the management of esophageal cancer: review article. Ann Thorac Cardiovasc Surg 2012; 18: 412-419
  • 151 Hsu WH. et al. Positron emission tomography-computed tomography in predicting locoregional invasion in esophageal squamous cell carcinoma. Ann Thorac Surg 2009; 87: 1564-1568
  • 152 Barber TW. et al. 18F-FDG PET/CT has a high impact on patient management and provides powerful prognostic stratification in the primary staging of esophageal cancer: a prospective study with mature survival data. J Nucl Med 2012; 53: 864-871
  • 153 Cervino AR. et al. Positron emission tomography/computed tomography and esophageal cancer in the clinical practice: How does it affect the prognosis?. J Cancer Res Ther 2012; 8: 619-625
  • 154 Varghese Jr TK. et al. The society of thoracic surgeons guidelines on the diagnosis and staging of patients with esophageal cancer. Ann Thorac Surg 2013; 96: 346-356
  • 155 Findlay JM. et al. Pragmatic staging of oesophageal cancer using decision theory involving selective endoscopic ultrasonography, PET and laparoscopy. Br J Surg 2015; 102: 1488-1499
  • 156 IQWIG. Abschlussbericht Nr.172 zur "Positronen-Emissionstomografie (PET) und (PET/CT) bei Ösophaguskarzinom. 2013
  • 157 CMS final decision on PET in solid tumors. J Nucl Med 2013; 54: 11n
  • 158 Osugi H. et al. Bronchoscopic ultrasonography for staging supracarinal esophageal squamous cell carcinoma: impact on outcome. World J Surg 2003; 27: 590-594
  • 159 Wakamatsu T. et al. Usefulness of preoperative endobronchial ultrasound for airway invasion around the trachea: esophageal cancer and thyroid cancer. Respiration 2006; 73: 651-657
  • 160 Omloo JM. et al. Value of bronchoscopy after EUS in the preoperative assessment of patients with esophageal cancer at or above the carina. J Gastrointest Surg 2008; 12: 1874-1879
  • 161 Riedel M. et al. Preoperative bronchoscopic assessment of airway invasion by esophageal cancer: a prospective study. Chest 1998; 113: 687-695
  • 162 Baisi A, Bonavina L, Peracchia A. Bronchoscopic staging of squamous cell carcinoma of the upper thoracic esophagus. Arch Surg 1999; 134: 140-143
  • 163 Imadahl A. et al. Is bronchoscopy a useful additional preoperative examination in esophageal carcinoma?. Langenbecks Arch Chir 1990; 375: 326-329
  • 164 Nieveen van Dijkum EJ. et al. The efficacy of laparoscopic staging in patients with upper gastrointestinal tumors. Cancer 1997; 79: 1315-1359
  • 165 de Graaf GW. et al. The role of staging laparoscopy in oesophagogastric cancers. Eur J Surg Oncol 2007; 33: 988-992
  • 166 Nath J. et al. Peritoneal lavage cytology in patients with oesophagogastric adenocarcinoma. Br J Surg 2008; 95: 721-726
  • 167 Krasna MJ. et al. Thoracoscopic staging of esophageal cancer: a prospective, multiinstitutional trial. Cancer and Leukemia Group B Thoracic Surgeons. Ann Thorac Surg 1995; 60: 1337-1340
  • 168 Krasna MJ. et al. CALGB 9380: a prospective trial of the feasibility of thoracoscopy/laparoscopy in staging esophageal cancer. Ann Thorac Surg 2001; 71: 1073-1079
  • 169 Luketich JD. et al. Evaluation of distant metastases in esophageal cancer: 100 consecutive positron emission tomography scans. Ann Thorac Surg 1999; 68: 1133-1136 ; discussion 1136–1137
  • 170 Wang GQ. et al. Histological precursors of oesophageal squamous cell carcinoma: results from a 13 year prospective follow up study in a high risk population. Gut 2005; 54: 187-192
  • 171 Kuwano H. Peculiar histopathologic features of esophageal cancer. Surg Today 1998; 28: 573-575
  • 172 Bosman F. et al. WHO Classification of Tumors of the Digestive System. 4th. edition Lyon: IARC; 2010
  • 173 Schnell TG. et al. Long-term nonsurgical management of Barrett’s esophagus with highgrade dysplasia. Gastroenterology 2001; 120: 1607-1619
  • 174 Reid BJ. et al. Observer variation in the diagnosis of dysplasia in Barrett’s esophagus. Hum Pathol 1988; 19: 166-178
  • 175 Skacel M. et al. The diagnosis of low-grade dysplasia in Barrett’s esophagus and its implications for disease progression. Am J Gastroenterol 2000; 95: 3383-3387
  • 176 Montgomery E. et al. Reproducibility of the diagnosis of dysplasia in Barrett esophagus: a reaffirmation. Hum Pathol 2001; 32: 368-378
  • 177 Ormsby AH. et al. Observer variation in the diagnosis of superficial oesophageal adenocarcinoma. Gut 2002; 51: 671-676
  • 178 Kerkhof M. et al. Grading of dysplasia in Barrett’s oesophagus: substantial interobserver variation between general and gastrointestinal pathologists. Histopathology 2007; 50: 920-927
  • 179 Wani S. et al. Greater interobserver agreement by endoscopic mucosal resection than biopsy samples in Barrett’s dysplasia. Clin Gastroenterol Hepatol 2010; 8: 783-788
  • 180 Weusten B. et al. Endoscopic management of Barrett’s esophagus: European Society of Gastrointestinal Endoscopy (ESGE) Position Statement. Endoscopy 2017; 49: 191-198
  • 181 Shaheen NJ. et al. ACG Clinical Guideline: Diagnosis and Management of Barrett’s Esophagus. Am J Gastroenterol 2016; 111: 30-50 ; quiz 51
  • 182 Bennett C. et al. Consensus statements for management of Barrett’s dysplasia and earlystage esophageal adenocarcinoma, based on a Delphi process. Gastroenterology 2012; 143: 336-346
  • 183 Brown IS, Whiteman DC, Lauwers GY. Foveolar type dysplasia in Barrett esophagus. Mod Pathol 2010; 23: 834-843
  • 184 Odze RD. What the gastroenterologist needs to know about the histology of Barrett’s esophagus. Curr Opin Gastroenterol 2011; 27: 389-396
  • 185 Rucker-Schmidt RL. et al. Nonadenomatous dysplasia in barrett esophagus: a clinical, pathologic, and DNA content flow cytometric study. Am J Surg Pathol 2009; 33: 886-893
  • 186 Mahajan D. et al. Grading of gastric foveolar-type dysplasia in Barrett’s esophagus. Mod Pathol 2010; 23: 1-11
  • 187 Demicco EG. et al. The dichotomy in carcinogenesis of the distal esophagus and esophagogastric junction: intestinal-type vs cardiac-type mucosa-associated adenocarcinoma. Mod Pathol 2011; 24: 1177-1190
  • 188 Chandrasoma P. et al. A proposal for a new validated histological definition of the gastroesophageal junction. Hum Pathol 2006; 37: 40-47
  • 189 Sharma P. et al. Dysplasia and cancer in a large multicenter cohort of patients with Barrett’s esophagus. Clin Gastroenterol Hepatol 2006; 4: 566-572
  • 190 Gatenby PA. et al. Treatment modality and risk of development of dysplasia and adenocarcinoma in columnar-lined esophagus. Dis Esophagus 2009; 22: 133-142
  • 191 Weston AP. et al. p53 protein overexpression in low grade dysplasia (LGD) in Barrett’s esophagus: immunohistochemical marker predictive of progression. Am J Gastroenterol 2001; 96: 1355-1362
  • 192 Reid BJ. et al. Predictors of progression to cancer in Barrett’s esophagus: baseline histology and flow cytometry identify low- and high-risk patient subsets. Am J Gastroenterol 2000; 95: 1669-1676
  • 193 Buttar NS. et al. Chemoprevention of esophageal adenocarcinoma by COX-2 inhibitors in an animal model of Barrett’s esophagus. Gastroenterology 2002; 122: 1101-1112
  • 194 Montgomery E. et al. Are ulcers a marker for invasive carcinoma in Barrett’s esophagus? Data from a diagnostic variability study with clinical follow-up. Am J Gastroenterol 2002; 97: 27-31
  • 195 Harrison R. et al. Detection of intestinal metaplasia in Barrett’s esophagus: an observational comparator study suggests the need for a minimum of eight biopsies. Am J Gastroenterol 2007; 102: 1154-1161
  • 196 Wittekind C. 2010 TNM system: on the 7th edition of TNM classification of malignant tumors. Pathologe 2010; 31: 331-332
  • 197 Fritz A. et al. International Classification of Diseases for oncology (ICD-O). Geneva: WHO; 2000 3rd ed.
  • 198 Wittekind C, Asamura H, LH S. TNM Atlas. Illustrated guide to the TNM Classification of Malignant Tumours. 6th. edition Oxford: Wiley Blackwell; 2014
  • 199 Wittekind C. TNM-Supplement. Erläuterungen zur einheitlichen Anwendung. 4.. Auflage. Wiley- Blackwell [Übersetzung der Englischen Ausgabe: Wittekind Ch, Compton CC, Brierley J, Sobin LH (eds.) TNM Supplement. Commentaries on uniform use. 4th ed.] 2013
  • 200 Kodama M, Kakegawa T. Treatment of superficial cancer of the esophagus: a summary of responses to a questionnaire on superficial cancer of the esophagus in Japan. Surgery 1998; 123: 432-429
  • 201 Bollschweiler E. et al. Staging of esophageal carcinoma: length of tumor and number of involved regional lymph nodes. Are these independent prognostic factors?. J Surg Oncol 2006; 94: 355-363
  • 202 Rice TW. et al. Esophageal carcinoma: depth of tumor invasion is predictive of regional lymph node status. Ann Thorac Surg 1998; 65: 787-792
  • 203 Davies AR. et al. Tumor stage after neoadjuvant chemotherapy determines survival after surgery for adenocarcinoma of the esophagus and esophagogastric junction. J Clin Oncol 2014; 32: 2983-2990
  • 204 ©, T.A.H.M.D.C., The American Heritage® Medical Dictionary Copyright ©. Houghton Mifflin Company. 2007
  • 205 Siewert JR, Stein HJ. Classification of adenocarcinoma of the oesophagogastric junction. Br J Surg 1998; 85: 1457-1459
  • 206 Kato H. et al. Evaluation of the new (1987) TNM classification for thoracic esophageal tumors. Int J Cancer 1993; 53: 220-223
  • 207 Enzinger PC, Mayer RJ. Esophageal cancer. N Engl J Med 2003; 349: 2241-2252
  • 208 Gospodarowicz MK, O’Sullivan B, Sobin LH. International Union Against Cancer (UICC). Prognostic Factors in Cancer. 3rd. ed. New York: Wiley; 2006
  • 209 Roder JD. et al. Ratio of invaded to removed lymph nodes as a predictor of survival in squamous cell carcinoma of the oesophagus. Br J Surg 1994; 81: 410-413
  • 210 Hermanek P. Tumors of the gastrointestinal tract and the pancreas: histopathology, staging and prognosis. Anticancer Res 1999; 19: 2393-2396
  • 211 Dexter SP. et al. Circumferential resection margin involvement: an independent predictor of survival following surgery for oesophageal cancer. Gut 2001; 48: 667-670
  • 212 Khan OA. et al. Prognostic significance of circumferential resection margin involvement following oesophagectomy for cancer. Br J Cancer 2003; 88: 1549-1552
  • 213 Brucher BL. et al. Achalasia and esophageal cancer: incidence, prevalence, and prognosis. World J Surg 2001; 25: 745-749
  • 214 Sarbia M. et al. p53 protein expression and prognosis in squamous cell carcinoma of the esophagus. Cancer 1994; 74: 2218-2223
  • 215 Mandard AM. et al. Pathologic assessment of tumor regression after preoperative chemoradiotherapy of esophageal carcinoma. Clinicopathologic correlations. Cancer 1994; 73: 2680-2686
  • 216 Becker K. et al. Histomorphology and grading of regression in gastric carcinoma treated with neoadjuvant chemotherapy. Cancer 2003; 98: 1521-1530
  • 217 Langer R. et al. Prognostic significance of histopathological tumor regression after neoadjuvant chemotherapy in esophageal adenocarcinomas. Mod Pathol 2009; 22: 1555-1563
  • 218 Becker K. et al. Significance of histopathological tumor regression after neoadjuvant chemotherapy in gastric adenocarcinomas: a summary of 480 cases. Ann Surg 2011; 253: 934-939
  • 219 Langer R. et al. A multifactorial histopathologic score for the prediction of prognosis of resected esophageal adenocarcinomas after neoadjuvant chemotherapy. Ann Surg Oncol 2014; 21: 915-921
  • 220 Wu TT. et al. Excellent interobserver agreement on grading the extent of residual carcinoma after preoperative chemoradiation in esophageal and esophagogastric junction carcinoma: a reliable predictor for patient outcome. Am J Surg Pathol 2007; 31: 58-64
  • 221 Mirza A. et al. Assessment of Histopathological Response in Gastric and Gastro-Oesophageal Junction Adenocarcinoma following Neoadjuvant Chemotherapy: Which Scoring System to Use?. ISRN Pathology 2012; 2012: 8
  • 222 Karamitopoulou E. et al. Assessment of tumor regression of esophageal adenocarcinomas after neoadjuvant chemotherapy: comparison of 2 commonly used scoring approaches. Am J Surg Pathol 2014; 38: 1551-1556
  • 223 Ancona E. et al. Only pathologic complete response to neoadjuvant chemotherapy improves significantly the long term survival of patients with resectable esophageal squamous cell carcinoma: final report of a randomized, controlled trial of preoperative chemotherapy versus surgery alone. Cancer 2001; 91: 2165-2174
  • 224 Rohatgi P. et al. Characterization of pathologic complete response after preoperative chemoradiotherapy in carcinoma of the esophagus and outcome after pathologic complete response. Cancer 2005; 104: 2365-2372
  • 225 Schneider PM. et al. Histomorphologic tumor regression and lymph node metastases determine prognosis following neoadjuvant radiochemotherapy for esophageal cancer: implications for response classification. Ann Surg 2005; 242: 684-692
  • 226 Meredith KL. et al. Pathologic response after neoadjuvant therapy is the major determinant of survival in patients with esophageal cancer. Ann Surg Oncol 2010; 17: 1159-1167
  • 227 Chirieac LR. et al. Posttherapy pathologic stage predicts survival in patients with esophageal carcinoma receiving preoperative chemoradiation. Cancer 2005; 103: 1347-1355
  • 228 Bang YJ. et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010; 376: 687-697
  • 229 Bang Y. Pathological features of advanced gastric cancer (GC): Relatationship to human epidermal growth factor receptor 2 (HER2) positivity in the global screening programme of the ToGA trial. J Clin Oncol 2009; 27: 15s. 2009
  • 230 Satoh T. et al. Quality of life in the trastuzumab for gastric cancer trial. Oncologist 2014; 19: 712-719
  • 231 Weimann A. et al. Significance of preoperative weight loss for perioperative metabolic adaptation and surgical risk in patients with tumors of the upper gastrointestinal tract. Langenbecks Arch Chir 1992; 377: 45-52
  • 232 Saito T. et al. Factors related to malnutrition in patients with esophageal cancer. Nutrition 1991; 7: 117-121
  • 233 Falkner D, Plato R, Weimann A. Die Wertigkeit der Feinnadelkatheterjejunostomie in der postoperativen enteralen Ernährung nach Ösophagusresektion. Deutsche Gesellschaft für Chirurgie. 131. Kongress der Dt. Gesellschaft für Chirurgie. Berlin 25.-28.03.2014
  • 234 van Stijn MF. et al. Preoperative nutrition status and postoperative outcome in elderly general surgery patients: a systematic review. JPEN J Parenter Enteral Nutr 2013; 37: 37-43
  • 235 Weimann A. et al. Clinical nutrition in surgery. Guidelines of the German Society for Nutritional Medicine. Chirurg 2014; 85: 320-326
  • 236 Arends et al. S3-Leitlinie der Deutschen Gesellschaft für Ernährungsmedizin. Klinische Ernährung in der nicht-chirurgischen Onkologie. Aktuel Ernährungsmed in press. 2015
  • 237 Pech O. et al. Comparison between endoscopic and surgical resection of mucosal esophageal adenocarcinoma in Barrett’s esophagus at two high-volume centers. Ann Surg 2011; 254: 67-72
  • 238 Prasad GA. et al. Endoscopic and surgical treatment of mucosal (T1a) esophageal adenocarcinoma in Barrett’s esophagus. Gastroenterology 2009; 137: 815-823
  • 239 Ell C. et al. Endoscopic mucosal resection of early cancer and high-grade dysplasia in Barrett’s esophagus. Gastroenterology 2000; 118: 670-677
  • 240 Ell C. et al. Curative endoscopic resection of early esophageal adenocarcinomas (Barrett’s cancer). Gastrointest Endosc 2007; 65: 3-10
  • 241 Pech O. et al. Long-term results and risk factor analysis for recurrence after curative endoscopic therapy in 349 patients with high-grade intraepithelial neoplasia and mucosal adenocarcinoma in Barrett’s oesophagus. Gut 2008; 57: 1200-1206
  • 242 Pech O. et al. Long-term efficacy and safety of endoscopic resection for patients with mucosal adenocarcinoma of the esophagus. Gastroenterology 2014; 146: 652-660 e1
  • 243 Chennat J. et al. Complete Barrett’s eradication endoscopic mucosal resection: an effective treatment modality for high-grade dysplasia and intramucosal carcinoma--an American singlecenter experience. Am J Gastroenterol 2009; 104: 2684-2692
  • 244 Moss A. et al. Endoscopic resection for Barrett’s high-grade dysplasia and early esophageal adenocarcinoma: an essential staging procedure with long-term therapeutic benefit. Am J Gastroenterol 2010; 105: 1276-1283
  • 245 Pouw RE. et al. Stepwise radical endoscopic resection for eradication of Barrett’s oesophagus with early neoplasia in a cohort of 169 patients. Gut 2010; 59: 1169-1177
  • 246 Pouw RE. et al. Efficacy of radiofrequency ablation combined with endoscopic resection for barrett’s esophagus with early neoplasia. Clin Gastroenterol Hepatol 2010; 8: 23-29
  • 247 van Vilsteren FG. et al. Stepwise radical endoscopic resection versus radiofrequency ablation for Barrett’s oesophagus with high-grade dysplasia or early cancer: a multicentre randomised trial. Gut 2011; 60: 765-773
  • 248 Manner H. et al. Early Barrett’s carcinoma with “low-risk” submucosal invasion: long-term results of endoscopic resection with a curative intent. Am J Gastroenterol 2008; 103: 2589-2597
  • 249 Manner H. et al. Efficacy, safety, and long-term results of endoscopic treatment for early stage adenocarcinoma of the esophagus with low-risk sm1 invasion. Clin Gastroenterol Hepatol 2013; 11: 630-635 ; quiz e45
  • 250 Alvarez Herrero L. et al. Risk of lymph node metastasis associated with deeper invasion by early adenocarcinoma of the esophagus and cardia: study based on endoscopic resection specimens. Endoscopy 2010; 42: 1030-1036
  • 251 Fitzgerald RC. et al. British Society of Gastroenterology guidelines on the diagnosis and management of Barrett’s oesophagus. Gut 2014; 63: 7-42
  • 252 Guo HM. et al. Endoscopic submucosal dissection vs endoscopic mucosal resection for superficial esophageal cancer. World J Gastroenterol 2014; 20: 5540-5547
  • 253 Neuhaus H. et al. Endoscopic submucosal dissection plus radiofrequency ablation of neoplastic Barrett’s esophagus. Endoscopy 2012; 44: 1105-1113
  • 254 Kagemoto K. et al. Clinical outcomes of endoscopic submucosal dissection for superficial Barrett’s adenocarcinoma. Gastrointest Endosc 2014; 80: 239-245
  • 255 Probst A. et al. Endoskopische Submukosadissektion (ESD) im Ösophagus. Z Gastroenterol 2013; 51: K233
  • 256 Feith M, Stein HJ, Siewert JR. Pattern of lymphatic spread of Barrett’s cancer. World J Surg 2003; 27: 1052-1057
  • 257 Holscher AH. et al. Prognostic impact of upper, middle, and lower third mucosal or submucosal infiltration in early esophageal cancer. Ann Surg 2011; 254: 802-807 ; discussion 807–808
  • 258 Zemler B. et al. Early Barrett’s carcinoma: the depth of infiltration of the tumour correlates with the degree of differentiation, the incidence of lymphatic vessel and venous invasion. Virchows Arch 2010; 456: 609-614
  • 259 Buskens CJ. et al. Prediction of appropriateness of local endoscopic treatment for high-grade dysplasia and early adenocarcinoma by EUS and histopathologic features. Gastrointest Endosc 2004; 60: 703-710
  • 260 Westerterp M. et al. Outcome of surgical treatment for early adenocarcinoma of the esophagusor gastro-esophageal junction. Virchows Arch 2005; 446: 497-504
  • 261 Ancona E. et al. Prediction of lymph node status in superficial esophageal carcinoma. Ann Surg Oncol 2008; 15: 3278-3288
  • 262 Liu L. et al. Significance of the depth of tumor invasion and lymph node metastasis in suerficially invasive (T1) esophageal adenocarcinoma. Am J Surg Pathol 2005; 29: 1079-1085
  • 263 Sepesi B. et al. Are endoscopic therapies appropriate for superficial submucosal esophageal adenocarcinoma? An analysis of esophagectomy specimens. J Am Coll Surg 2010; 210: 418-427
  • 264 Badreddine RJ. et al. Depth of submucosal invasion does not predict lymph node metastasis and survival of patients with esophageal carcinoma. Clin Gastroenterol Hepatol 2010; 8: 248-253
  • 265 Griffin SM, Burt AD, Jennings NA. Lymph node metastasis in early esophageal adenocarcinoma. Ann Surg 2011; 254: 731-736 ; discussion 736–737
  • 266 Leers JM. et al. The prevalence of lymph node metastases in patients with T1 esophageal adenocarcinoma a retrospective review of esophagectomy specimens. Ann Surg 2011; 253: 271-278
  • 267 Lorenz D. et al. Prognostic risk factors of early esophageal adenocarcinomas. Ann Surg 2014; 259: 469-476
  • 268 May A. et al. Local endoscopic therapy for intraepithelial high-grade neoplasia and early adenocarcinoma in Barrett’s oesophagus: acute-phase and intermediate results of a new treatment approach. Eur J Gastroenterol Hepatol 2002; 14: 1085-1091
  • 269 Gossner L. et al. Photodynamic ablation of high-grade dysplasia and early cancer in Barrett’s esophagus by means of 5-aminolevulinic acid. Gastroenterology 1998; 114: 448-455
  • 270 Pech O. et al. Long-term results of photodynamic therapy with 5-aminolevulinic acid for superficial Barrett’s cancer and high-grade intraepithelial neoplasia. Gastrointest Endosc 2005; 62: 24-30
  • 271 Overholt BF. et al. Five-year efficacy and safety of photodynamic therapy with Photofrin in Barrett’s high-grade dysplasia. Gastrointest Endosc 2007; 66: 460-468
  • 272 Shaheen NJ. et al. Radiofrequency ablation in Barrett’s esophagus with dysplasia. N Engl J Med 2009; 360: 2277-2288
  • 273 Shaheen NJ. et al. Durability of radiofrequency ablation in Barrett’s esophagus with dysplasia. Gastroenterology 2011; 141: 460-468
  • 274 Ganz RA. et al. Circumferential ablation of Barrett’s esophagus that contains high-grade dysplasia: a U.S. Multicenter Registry. Gastrointest Endosc 2008; 68: 35-40
  • 275 Phoa KN. et al. Remission of Barrett’s esophagus with early neoplasia 5 years after radiofrequency ablation with endoscopic resection: a Netherlands cohort study. Gastroenterology 2013; 145: 96-104
  • 276 Orman ES. et al. Intestinal metaplasia recurs infrequently in patients successfully treated for Barrett’s esophagus with radiofrequency ablation. Am J Gastroenterol 2013; 108: 187-195 ; quiz 196
  • 277 Van Laethem JL. et al. Argon plasma coagulation in the treatment of Barrett’s high-grade dysplasia and in situ adenocarcinoma. Endoscopy 2001; 33: 257-261
  • 278 Ragunath K. et al. Endoscopic ablation of dysplastic Barrett’s oesophagus comparing argon plasma coagulation and photodynamic therapy: a randomized prospective trial assessing efficacy and cost-effectiveness. Scand J Gastroenterol 2005; 40: 750-758
  • 279 Kelty CJ. et al. Endoscopic ablation of Barrett’s oesophagus: a randomized-controlled trial of photodynamic therapy vs. argon plasma coagulation. Aliment Pharmacol Ther 2004; 20: 1289-1296
  • 280 Dumot JA. et al. An open-label, prospective trial of cryospray ablation for Barrett’s esophagus high-grade dysplasia and early esophageal cancer in high-risk patients. Gastrointest Endosc 2009; 70: 635-644
  • 281 Shaheen NJ. et al. Safety and efficacy of endoscopic spray cryotherapy for Barrett’s esophagus with high-grade dysplasia. Gastrointest Endosc 2010; 71: 680-685
  • 282 Ishihara R. et al. Comparison of EMR and endoscopic submucosal dissection for en bloc resection of early esophageal cancers in Japan. Gastrointest Endosc 2008; 68: 1066-1072
  • 283 Cao Y. et al. Meta-analysis of endoscopic submucosal dissection versus endoscopic mucosal resection for tumors of the gastrointestinal tract. Endoscopy 2009; 41: 751-757
  • 284 Srivastava A. et al. Extent of low-grade dysplasia is a risk factor for the development of esophageal adenocarcinoma in Barrett’s esophagus. Am J Gastroenterol 2007; 102: 483-493 ; quiz 694
  • 285 Phoa KN. et al. Radiofrequency ablation vs endoscopic surveillance for patients with Barrett esophagus and low-grade dysplasia: a randomized clinical trial. Jama 2014; 311: 1209-1217
  • 286 McCann P. et al. The safety and effectiveness of endoscopic and non-endoscopic approaches to the management of early esophageal cancer: a systematic review (Structured abstract). Cancer Treatment Reviews 2011; 37: 11-62
  • 287 Haidry RJ. et al. Radiofrequency ablation for early oesophageal squamous neoplasia: outcomes form United Kingdom registry. World J Gastroenterol 2013; 19: 6011-6019
  • 288 Min BH. et al. Feasibility and efficacy of argon plasma coagulation for early esophageal squamous cell neoplasia. Endoscopy 2013; 45: 575-578
  • 289 Tahara K. et al. Argon plasma coagulation for superficial esophageal squamous-cell carcinoma in high-risk patients. World J Gastroenterol 2012; 18: 5412-5417
  • 290 Peters FP. et al. Endoscopic treatment of high-grade dysplasia and early stage cancer in Barrett’s esophagus. Gastrointest Endosc 2005; 61: 506-514
  • 291 Metzger R. et al. High volume centers for esophagectomy: what is the number needed to achieve low postoperative mortality?. Dis Esophagus 2004; 17: 310-314
  • 292 Holscher AH. et al. High-volume centers – effect of case load on outcome in cancer surgery. Onkologie 2004; 27: 412-416
  • 293 Coupland VH. et al. Hospital volume, proportion resected and mortality from oesophageal and gastric cancer: a population-based study in England, 2004–2008. Gut 2013; 62: 961-966
  • 294 Brusselaers N, Mattsson F, Lagergren J. Hospital and surgeon volume in relation to longterm survival after oesophagectomy: systematic review and meta-analysis. Gut 2014; 63: 1393-1400
  • 295 Derogar M. et al. Hospital and surgeon volume in relation to survival after esophageal cancer surgery in a population-based study. J Clin Oncol 2013; 31: 551-557
  • 296 Reames BN, Shubeck SP, Birkmeyer JD. Strategies for reducing regional variation in the use of surgery: a systematic review. Ann Surg 2014; 259: 616-627
  • 297 Schroder W. et al. Preoperative risk analysis--a reliable predictor of postoperative outcome after transthoracic esophagectomy?. Langenbecks Arch Surg 2006; 391: 455-460
  • 298 Bollschweiler E. et al. Preoperative risk analysis in patients with adenocarcinoma or squamous cell carcinoma of the oesophagus. Br J Surg 2000; 87: 1106-1110
  • 299 Lagarde SM. et al. Evaluation of O-POSSUM in predicting in-hospital mortality after resection for oesophageal cancer. Br J Surg 2007; 94: 1521-1526
  • 300 Hodari A. et al. Assessment of morbidity and mortality after esophagectomy using a modified frailty index. Ann Thorac Surg 2013; 96: 1240-1245
  • 301 Pottgen C, Stuschke M. Radiotherapy versus surgery within multimodality protocols for esophageal cancer – a meta-analysis of the randomized trials. Cancer Treat Rev 2012; 38: 599-604
  • 302 Markar SR. et al. Assessment of short-term clinical outcomes following salvage esophagectomy for the treatment of esophageal malignancy: systematic review and pooled analysis. Ann Surg Oncol 2014; 21: 922-931
  • 303 Hofstetter WL. Salvage esophagectomy. J Thorac Dis 2014; 6: S341-S349
  • 304 Wu J. et al. Prognostic significance of positive circumferential resection margin in esophageal cancer: a systematic review and meta-analysis. Ann Thorac Surg 2014; 97: 446-453
  • 305 Holscher AH. et al. How safe is high intrathoracic esophagogastrostomy?. Chirurg 2003; 74: 726-733
  • 306 Holscher AH. et al. Laparoscopic ischemic conditioning of the stomach for esophageal replacement. Ann Surg 2007; 245: 241-246
  • 307 Li B. et al. Comparison of Ivor-Lewis vs Sweet esophagectomy for esophageal squamous cell carcinoma: a randomized clinical trial. JAMA Surg 2015; 150: 292-298
  • 308 Schroder W. et al. The resection of the azygos vein – necessary or redundant extension of transthoracic esophagectomy?. J Gastrointest Surg 2008; 12: 1163-1167
  • 309 Boone J. et al. The effect of azygos vein preservation on mediastinal lymph node harvesting in thoracic esophagolymphadenectomy. Dis Esophagus 2008; 21: 226-229
  • 310 Siewert JR. et al. Cardia cancer: attempt at a therapeutically relevant classification. Chirurg 1987; 58: 25-32
  • 311 Kutup A. et al. What should be the gold standard for the surgical component in the treatment of locally advanced esophageal cancer: transthoracic versus transhiatal esophagectomy. Ann Surg 2014; 260: 1016-1022
  • 312 Omloo JM. et al. Extended transthoracic resection compared with limited transhiatal resection for adenocarcinoma of the mid/distal esophagus: five-year survival of a randomized clinical trial. Ann Surg 2007; 246: 992-1000 ; discussion 1000–1001
  • 313 Workum F. et al. Improved Functional Results After Minimally Invasive Esophagectomy: intrathoracic Versus Cervical Anastomosis. Annals of thoracic surgery 2017; 103: 267-273 . doi:10.1016/j.athoracsur.2016.07.010
  • 314 Sasako M. et al. Left thoracoabdominal approach versus abdominal-transhiatal approach for gastric cancer of the cardia or subcardia: a randomised controlled trial. Lancet Oncol 2006; 7: 644-651
  • 315 Kurokawa Y. et al. Ten-year follow-up results of a randomized clinical trial comparing left thoracoabdominal and abdominal transhiatal approaches to total gastrectomy for adenocarcinoma of the oesophagogastric junction or gastric cardia. British journal of surgery 2015; 102: 341-348 . doi:10.1002/bjs.9764
  • 316 Moehler M. et al. German S3-guideline “Diagnosis and treatment of esophagogastric cancer”. Z Gastroenterol 2011; 49: 461-531
  • 317 Peyre CG. et al. The number of lymph nodes removed predicts survival in esophageal cancer: an international study on the impact of extent of surgical resection. Ann Surg 2008; 248: 549-556
  • 318 Peyre CG. et al. Predicting systemic disease in patients with esophageal cancer after esophagectomy: a multinational study on the significance of the number of involved lymph nodes. Ann Surg 2008; 248: 979-985
  • 319 Lerut T. et al. Three-field lymphadenectomy for carcinoma of the esophagus and gastroesophageal junction in 174 R0 resections: impact on staging, disease-free survival, and outcome: a plea for adaptation of TNM classification in upper-half esophageal carcinoma. Ann Surg 2004; 240: 962-972 ; discussion 972–974
  • 320 Rizk NP. et al. Optimum lymphadenectomy for esophageal cancer. Ann Surg 2010; 251: 46-50
  • 321 Fujita H. et al. Optimal lymphadenectomy for squamous cell carcinoma in the thoracic esophagus: comparing the short- and long-term outcome among the four types of lymphadenectomy. World J Surg 2003; 27: 571-579
  • 322 Bollschweiler E. et al. Influence of neoadjuvant chemoradiation on the number and size of analyzed lymph nodes in esophageal cancer. Ann Surg Oncol 2010; 17: 3187-3194
  • 323 Bekkar S. et al. The impact of preoperative radiochemotherapy on survival in advanced esophagogastric junction signet ring cell adenocarcinoma. Ann Thorac Surg 2014; 97: 303-310
  • 324 Wittekind C, Meyer HJ. TNM: Klassifikation maligner Tumoren. 7.. Auflage Wiley; 2012: 295
  • 325 Gutschow CA. et al. Merendino procedure with preservation of the vagus for early carcinoma of the gastroesophageal junction. Zentralbl Chir 2004; 129: 276-281
  • 326 Stein HJ. et al. Limited resection for early adenocarcinoma in Barrett’s esophagus. Ann Surg 2000; 232: 733-742
  • 327 Zapletal C. et al. Quality of life after surgical treatment of early Barrett’s cancer: a prospective comparison of the Ivor-Lewis resection versus the modified Merendino resection. World J Surg 2014; 38: 1444-1452
  • 328 Schroder W. et al. Ivor-Lewis esophagectomy with and without laparoscopic conditioning of the gastric conduit. World J Surg 2010; 34: 738-743
  • 329 Urschel JD. et al. Pyloric drainage (pyloroplasty) or no drainage in gastric reconstruction after esophagectomy: a meta-analysis of randomized controlled trials. Dig Surg 2002; 19: 160-164
  • 330 Gaur P, Swanson SJ. Should we continue to drain the pylorus in patients undergoing an esophagectomy?. Dis Esophagus 2014; 27: 568-573
  • 331 Vallbohmer D. et al. Diaphragmatic hernia after conventional or laparoscopic-assisted transthoracic esophagectomy. Ann Thorac Surg 2007; 84: 1847-1852
  • 332 Price TN. et al. A comprehensive review of anastomotic technique in 432 esophagectomies. Ann Thorac Surg 2013; 95: 1154-1160 ; discussion 1160–1161
  • 333 Erkmen CP. et al. Laparoscopic repair of hiatal hernia after esophagectomy. J Gastrointest Surg 2013; 17: 1370-1374
  • 334 Avery K. et al. The feasibility of a randomized controlled trial of esophagectomy for esophageal cancer – The ROMIO (Randomized Oesophagectomy: Minimally Invasive or Open) study: Protocol for a randomized controlled trial. Trials 2014; 15 DOI: 10.1186/1745-6215-15-200.
  • 335 Biere SS. et al. Minimally invasive versus open oesophagectomy for patients with oesophageal cancer: a multicentre, open-label, randomised controlled trial. Lancet 2012; 379: 1887-1892
  • 336 Maas KW. et al. Quality of Life and Late Complications After Minimally Invasive Compared to Open Esophagectomy: Results of a Randomized Trial. World J Surg 2015; 39: 1986-1993
  • 337 Straatman J. et al. Minimally Invasive Versus Open Esophageal Resection: Three-year Follow-up of the Previously Reported Randomized Controlled Trial: the TIME Trial. Ann Surg 2017; 266: 232-236
  • 338 Luketich JD. et al. Outcomes after minimally invasive esophagectomy: review of over 1000 patients. Ann Surg 2012; 256: 95-103
  • 339 Smithers BM. et al. Comparison of the outcomes between open and minimally invasive esophagectomy. Ann Surg 2007; 245: 232-240
  • 340 Briez N. et al. Effects of hybrid minimally invasive oesophagectomy on major postoperative pulmonary complications. Br J Surg 2012; 99: 1547-1553
  • 341 Briez N. et al. Is minimally invasive oesophagectomy for cancer decreasing pulmonary complications-Results from a case-control study. J Clin Oncol 2010; 28: 15s
  • 342 Bonavina L. et al. Early outcome of thoracoscopic and hybrid esophagectomy: Propensity-matched comparative analysis. Surgery 2016; 159: 1073-1081
  • 343 Straatman J. et al. Techniques and short-term outcomes for total minimally invasive Ivor Lewis esophageal resection in distal esophageal and gastroesophageal junction cancers: pooled data from six European centers. Surgical endoscopy and other interventional techniques 2017; 31: 119-126 . doi:10.1007/s00464-016-4938-2
  • 344 Seesing MFJ. et al. A Propensity Score Matched Analysis of Open Versus Minimally Invasive Transthoracic Esophagectomy in the Netherlands. Ann Surg 2017; 266: 839-846
  • 345 Schmidt HM. et al. Defining Benchmarks for Transthoracic Esophagectomy: A Multicenter Analysis of Total Minimally Invasive Esophagectomy in Low Risk Patients. Ann Surg 2017; 266: 814-821
  • 346 Bronson NW. et al. The incidence of hiatal hernia after minimally invasive esophagectomy. J Gastrointest Surg 2014; 18: 889-893
  • 347 Willer BL. et al. Incidence of diaphragmatic hernias following minimally invasive versus open transthoracic Ivor Lewis McKeown esophagectomy. Hernia 2012; 16: 185-190
  • 348 Low DE. et al. International Consensus on Standardization of Data Collection for Complications Associated With Esophagectomy: Esophagectomy Complications Consensus Group (ECCG). Ann Surg 2015; 262: 286-294
  • 349 Low DE. et al. Benchmarking Complications Associated with Esophagectomy. Ann Surg 2019; 269: 291-298
  • 350 Erhunmwunsee L. et al. Impact of pretreatment imaging on survival of esophagectomy after induction therapy for esophageal cancer: who should be given the benefit of the doubt?: esophagectomy outcomes of patients with suspicious metastatic lesions. Ann Surg Oncol 2015; 22: 1020-1025
  • 351 Shimoji H. et al. Induction chemotherapy or chemoradiotherapy followed by radical esophagectomy for T4 esophageal cancer: results of a prospective cohort study. World J Surg 2013; 37: 2180-2188
  • 352 Weimann A. et al. ESPEN Guidelines on Enteral Nutrition: Surgery including organ transplantation. Clin Nutr 2006; 25: 224-244
  • 353 Weimann A, Breitenstein S, Breuer JP. et al. S3-Leitlinie der Deutschen Gesellschaft für Ernährungsmedizin: Klinische Ernährung in der Chirurgie. Aktuell Ernährungsmedizin 2013; 38: e155-e197
  • 354 Kondrup J. et al. ESPEN guidelines for nutrition screening 2002. Clin Nutr 2003; 22: 415-421
  • 355 Schwegler I. et al. Nutritional risk is a clinical predictor of postoperative mortality and morbidity in surgery for colorectal cancer. Br J Surg 2010; 97: 92-97
  • 356 Sorensen J. et al. EuroOOPS: an international, multicentre study to implement nutritional risk screening and evaluate clinical outcome. Clin Nutr 2008; 27: 340-349
  • 357 Kuppinger D. et al. Nutritional screening for risk prediction in patients scheduled for abdominal operations. Br J Surg 2012; 99: 728-737
  • 358 Khuri SF. et al. Risk adjustment of the postoperative mortality rate for the comparative assessment of the quality of surgical care: results of the National Veterans Affairs Surgical Risk Study. J Am Coll Surg 1997; 185: 315-327
  • 359 Hennessey DB. et al. Preoperative hypoalbuminemia is an independent risk factor for the development of surgical site infection following gastrointestinal surgery: a multi-institutional study. Ann Surg 2010; 252: 325-329
  • 360 Ligthart-Melis GC. et al. Dietician-delivered intensive nutritional support is associated with a decrease in severe postoperative complications after surgery in patients with esophageal cancer. Dis Esophagus 2013; 26: 587-593
  • 361 Ellrichmann M. et al. Prospective evaluation of malignant cell seeding after percutaneous endoscopic gastrostomy in patients with oropharyngeal/esophageal cancers. Endoscopy 2013; 45: 526-531
  • 362 Mabvuure NT, Roman A, Khan OA. Enteral immunonutrition versus standard enteral nutrition for patients undergoing oesophagogastric resection for cancer. Int J Surg 2013; 11: 122-127
  • 363 Osland E. et al. Effect of timing of pharmaconutrition (immunonutrition) administration on outcomes of elective surgery for gastrointestinal malignancies: a systematic review and metaanalysis. JPEN J Parenter Enteral Nutr 2014; 38: 53-69
  • 364 Jie B. et al. Impact of preoperative nutritional support on clinical outcome in abdominal surgical patients at nutritional risk. Nutrition 2012; 28: 1022-1027
  • 365 Hill GL. Impact of nutritional support on the clinical outcome of the surgical patient. Clin Nutr 1994; 13: 331-340
  • 366 Burden S. et al. Pre-operative nutrition support in patients undergoing gastrointestinal surgery. Cochrane Database Syst Rev 2012; 11: Cd008879
  • 367 Bozzetti F. et al. Perioperative total parenteral nutrition in malnourished, gastrointestinal cancer patients: a randomized, clinical trial. JPEN J Parenter Enteral Nutr 2000; 24: 7-14
  • 368 Perioperative Total Parenteral Nutrition in Surgical Patients. New England Journal of Medicine 1991; 325: 525-532
  • 369 Andersen HK, Lewis SJ, Thomas S. Early enteral nutrition within 24 h of colorectal surgery versus later commencement of feeding for postoperative complications. Cochrane Database Syst Rev 2006; Cd004080
  • 370 Lewis SJ, Andersen HK, Thomas S. Early enteral nutrition within 24h of intestinal surgery versus later commencement of feeding: a systematic review and meta-analysis. J Gastrointest Surg 2009; 13: 569-575
  • 371 Osland E. et al. Early versus traditional postoperative feeding in patients undergoing resectional gastrointestinal surgery: a meta-analysis. JPEN J Parenter Enteral Nutr 2011; 35: 473-487
  • 372 Sica GS. et al. Needle catheter jejunostomy at esophagectomy for cancer. J Surg Oncol 2005; 91: 276-279
  • 373 Han-Geurts IJ. et al. Randomized clinical trial comparing feeding jejunostomy with nasoduodenal tube placement in patients undergoing oesophagectomy. Br J Surg 2007; 94: 31-35
  • 374 Gerritsen A. et al. Efficacy and complications of nasojejunal, jejunostomy and parenteral feeding after pancreaticoduodenectomy. J Gastrointest Surg 2012; 16: 1144-1151
  • 375 Markides GA, Alkhaffaf B, Vickers J. Nutritional access routes following oesophagectomy--a systematic review. Eur J Clin Nutr 2011; 65: 565-573
  • 376 Theologou T. et al. The impact of positive circumferential margin on survival following oesophagectomy using the new 7th TNM classification. Eur J Cardiothorac Surg 2013; 44: 855-859
  • 377 O’Farrell NJ. et al. Lack of independent significance of a close (<1 mm) circumferential resection margin involvement in esophageal and junctional cancer. Ann Surg Oncol 2013; 20: 2727-2733
  • 378 Fok M. et al. Postoperative radiotherapy for carcinoma of the esophagus: a prospective, randomized controlled study. Surgery 1993; 113: 138-147
  • 379 Teniere P. et al. Postoperative radiation therapy does not increase survival after curative resection for squamous cell carcinoma of the middle and lower esophagus as shown by a multicenter controlled trial. French University Association for Surgical Research. Surg Gynecol Obstet 1991; 173: 123-130
  • 380 Bao Y. et al. Three-dimensional conformal radiotherapy with concurrent chemotherapy for postoperative recurrence of esophageal squamous cell carcinoma: clinical efficacy and failure pattern. Radiat Oncol 2013; 8: 241
  • 381 Jingu K. et al. Long-term results of radiotherapy combined with nedaplatin and 5-fluorouracil for postoperative loco-regional recurrent esophageal cancer: update on a phase II study. BMC Cancer 2012; 12: 542
  • 382 Ma DY. et al. Concurrent three-dimensional conformal radiotherapy and chemotherapy for postoperative recurrence of mediastinal lymph node metastases in patients with esophageal squamous cell carcinoma: a phase 2 single-institution study. Radiat Oncol 2014; 9: 28
  • 383 Fakhrian K. et al. Salvage radiotherapy in patients with recurrent esophageal carcinoma. Strahlenther Onkol 2012; 188: 136-142
  • 384 Sudo K. et al. Locoregional failure rate after preoperative chemoradiation of esophageal adenocarcinoma and the outcomes of salvage strategies. J Clin Oncol 2013; 31: 4306-4310
  • 385 Oppedijk V. et al. Patterns of recurrence after surgery alone versus preoperative chemoradiotherapy and surgery in the CROSS trials. J Clin Oncol 2014; 32: 385-391
  • 386 Malthaner RA. et al. Neoadjuvant or adjuvant therapy for resectable esophageal cancer: a systematic review and meta-analysis. BMC Med 2004; 2: 35
  • 387 Arnott SJ. et al. Preoperative radiotherapy for esophageal carcinoma. Cochrane Database Syst Rev 2005; Cd001799
  • 388 Schwer A. et al. Survival effect of neoadjuvant radiotherapy before esophagectomy for patients with esophageal cancer: a surveillance, epidemiology, and end-results study. Int J Radiat Oncol Biol Phys 2009; 73: 449-455
  • 389 Ychou M. et al. Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol 2011; 29: 1715-1721
  • 390 Cunningham D. et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med 2006; 355: 11-20
  • 391 Kelsen DP. et al. Chemotherapy followed by surgery compared with surgery alone for localized esophageal cancer. N Engl J Med 1998; 339: 1979-1984
  • 392 Boonstra JJ. et al. Chemotherapy followed by surgery versus surgery alone in patients with resectable oesophageal squamous cell carcinoma: long-term results of a randomized controlled trial. BMC Cancer 2011; 11: 181
  • 393 Allum WH. et al. Long-term results of a randomized trial of surgery with or without preoperative chemotherapy in esophageal cancer. J Clin Oncol 2009; 27: 5062-5067
  • 394 Medical Research Council Oesophageal Cancer Working Group. Surgical resection with or without preoperative chemotherapy in oesophageal cancer: a randomised controlled trial. Lancet 2002; 359: 1727-1733
  • 395 Law S. et al. Preoperative chemotherapy versus surgical therapy alone for squamous cell carcinoma of the esophagus: a prospective randomized trial. J Thorac Cardiovasc Surg 1997; 114: 210-217
  • 396 Al-Batran SE. et al. Perioperative chemotherapy with docetaxel, oxaliplatin, and fluorouracil/leucovorin (FLOT) versus epirubicin, cisplatin, and fluorouracil or capecitabine (ECF/ECX) for resectable gastric or gastroesophageal junction (GEJ) adenocarcinoma (FLOT4-AIO): A multicenter, randomized phase 3 trial. Journal of Clinical Oncology 2017; 35: 4004-4004
  • 397 Sjoquist KM. et al. Survival after neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal carcinoma: an updated meta-analysis. Lancet Oncol 2011; 12: 681-692
  • 398 Markar SR. et al. Role of neoadjuvant treatment in clinical T2N0M0 oesophageal cancer: results from a retrospective multi-center European study. Eur J Cancer 2016; 56: 59-68
  • 399 Speicher PJ. et al. Induction therapy does not improve survival for clinical stage T2N0 esophageal cancer. J Thorac Oncol 2014; 9: 1195-1201
  • 400 Crabtree TD. et al. Evaluation of the reliability of clinical staging of T2 N0 esophageal cancer: a review of the Society of Thoracic Surgeons database. Ann Thorac Surg 2013; 96: 382-390
  • 401 Thuss-Patience P, Vecchione L, Keilholz U. Should cT2 esophageal cancer get neoadjuvant treatment before surgery?. J Thorac Dis 2017; 9: 2819-2823
  • 402 Cunningham D, Allum W, Weeden S. Perioperative chemotherapy in operable gastric and lower oesophageal cancer: a randomised, controlled trial of the UK NCRI Upper GI Clinical Studies Group (the MAGIC trial, ISRCTN 93793971) [abstract]. European journal of cancer 2003; 1: S18
  • 403 Clark PI. Medical Research Council (MRC) randomised phase III trial of surgery with or without pre-operative chemotherapy in resectable cancer of the oesophagus. British journal of cancer 2000; 83: 1
  • 404 Moehler M. et al. International comparison of the German evidence-based S3-guidelines on the diagnosis and multimodal treatment of early and locally advanced gastric cancer, including adenocarcinoma of the lower esophagus. Gastric Cancer 2015; 18: 550-563
  • 405 Fiorica F. et al. Preoperative chemoradiotherapy for oesophageal cancer: a systematic review and meta-analysis. Gut 2004; 53: 925-930
  • 406 Burmeister BH. et al. Surgery alone versus chemoradiotherapy followed by surgery for resectable cancer of the oesophagus: a randomised controlled phase III trial. Lancet Oncol 2005; 6: 659-668
  • 407 Lee JL. et al. A single institutional phase III trial of preoperative chemotherapy with hyperfractionation radiotherapy plus surgery versus surgery alone for resectable esophageal squamous cell carcinoma. Ann Oncol 2004; 15: 947-954
  • 408 Bosset JF. et al. Chemoradiotherapy followed by surgery compared with surgery alone in squamous-cell cancer of the esophagus. N Engl J Med 1997; 337: 161-167
  • 409 Tepper J. et al. Phase III trial of trimodality therapy with cisplatin, fluorouracil, radiotherapy, and surgery compared with surgery alone for esophageal cancer: CALGB 9781. J Clin Oncol 2008; 26: 1086-1092
  • 410 Stahl M. et al. Phase III comparison of preoperative chemotherapy compared with chemoradiotherapy in patients with locally advanced adenocarcinoma of the esophagogastric junction. J Clin Oncol 2009; 27: 851-816
  • 411 van Hagen P. et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 2012; 366: 2074-2084
  • 412 Urba SG. et al. Randomized trial of preoperative chemoradiation versus surgery alone in patients with locoregional esophageal carcinoma. J Clin Oncol 2001; 19: 305-313
  • 413 Conroy T. et al. Definitive chemoradiotherapy with FOLFOX versus fluorouracil and cisplatin in patients with oesophageal cancer (PRODIGE5/ACCORD17): final results of a randomised, phase 2/3 trial. Lancet Oncol 2014; 15: 305-314
  • 414 van Hagen P. et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 2012; 366: 2074-2084
  • 415 Herskovic A. et al. Combined chemotherapy and radiotherapy compared with radiotherapy alone in patients with cancer of the esophagus. N Engl J Med 1992; 326: 1593-1598
  • 416 Cao XF. et al. Effects of neoadjuvant radiochemotherapy on pathological staging and prognosis for locally advanced esophageal squamous cell carcinoma. Dis Esophagus 2009; 22: 477-481
  • 417 Walsh TN. et al. A comparison of multimodal therapy and surgery for esophageal adenocarcinoma. N Engl J Med 1996; 335: 462-467
  • 418 Walsh TN, Grannell M, Mansoor S. Predictive factors for success of neo-adjuvant therapy in upper gastrointestinal cancer. J Gastroenterol Hepatol 2002; 17: S172-S175
  • 419 Walsh TN. et al. Neoadjuvant treatment of advanced stage esophageal adenocarcinoma increases survival. Dis Esophagus 2002; 15: 121-124
  • 420 Apinop C, Puttisak P, Preecha N. A prospective study of combined therapy in esophageal cancer. Hepatogastroenterology 1994; 41: 391-393
  • 421 Le Prise E. et al. A randomized study of chemotherapy, radiation therapy, and surgery versus surgery for localized squamous cell carcinoma of the esophagus. Cancer 1994; 73: 1779-1784
  • 422 Kumagai K. et al. Meta-analysis of postoperative morbidity and perioperative mortality in patients receiving neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal and gastro-oesophageal junctional cancers. Br J Surg 2014; 101: 321-338
  • 423 Ronellenfitsch U. et al. Perioperative chemo(radio)therapy versus primary surgery for resectable adenocarcinoma of the stomach, gastroesophageal junction, and lower esophagus. Cochrane Database of Systematic Reviews 2013; DOI: 10.1002/14651858.CD008107.pub2.
  • 424 Lone GN. et al. Role of preoperative chemotherapy in squamous cell carcinoma of esophagus in kashmir, a cancer belt a pilot study. Asian Pac J Cancer Prev 2011; 12: 465-470
  • 425 Zhao Y. et al. Perioperative versus Preoperative Chemotherapy with Surgery in Patients with Resectable Squamous Cell Carcinoma of Esophagus: A Phase III Randomized Trial. Journal of thoracic oncology: official publication of the International Association for the Study of Lung Cancer 2015; 10: 1349-1356 . doi:10.1097/JTO.0000000000000612
  • 426 Al-Batran SE. et al. Histopathological regression after neoadjuvant docetaxel, oxaliplatin, fluorouracil, and leucovorin versus epirubicin, cisplatin, and fluorouracil or capecitabine in patients with resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4-AIO): results from the phase 2 part of a multicentre, open-label, randomised phase 2/3 trial. Lancet Oncol 2016; 17: 1697-1708
  • 427 Sjoquist KM. et al. Survival after neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal carcinoma: an updated meta-analysis. Lancet Oncol 2011; 12: 681-692
  • 428 Kranzfelder M. et al. Meta-analysis of neoadjuvant treatment modalities and definitive non-surgical therapy for oesophageal squamous cell cancer. Br J Surg 2011; 98: 768-783
  • 429 Ando N. et al. A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus (JCOG9907). Ann Surg Oncol 2012; 19: 68-74
  • 430 Lordick F. Optimizing neoadjuvant chemotherapy through the use of early response evaluation by positron emission tomography. Recent Results Cancer Res 2012; 196: 201-211
  • 431 Weber WA. et al. Prediction of response to preoperative chemotherapy in adenocarcinomas of the esophagogastric junction by metabolic imaging. J Clin Oncol 2001; 19: 3058-3065
  • 432 Ott K. et al. Metabolic imaging predicts response, survival, and recurrence in adenocarcinomas of the esophagogastric junction. J Clin Oncol 2006; 24: 4692-4698
  • 433 Ilson DH. Cancer of the gastroesophageal junction: combined modality therapy. Surg Oncol Clin N Am 2006; 15: 803-824
  • 434 Wieder HA. et al. Comparison of changes in tumor metabolic activity and tumor size during chemotherapy of adenocarcinomas of the esophagogastric junction. J Nucl Med 2005; 46: 2029-2034
  • 435 Lordick F. et al. PET to assess early metabolic response and to guide treatment of adenocarcinoma of the oesophagogastric junction: the MUNICON phase II trial. Lancet Oncol 2007; 8: 797-805
  • 436 zum Buschenfelde CM. et al. (18)F-FDG PET-guided salvage neoadjuvant radiochemotherapy of adenocarcinoma of the esophagogastric junction: the MUNICON II trial. J Nucl Med 2011; 52: 1189-1196
  • 437 Gillham CM. et al. (18)FDG uptake during induction chemoradiation for oesophageal cancer fails to predict histomorphological tumour response. Br J Cancer 2006; 95: 1174-1179
  • 438 Klaeser B. et al. Limited predictive value of FDG-PET for response assessment in the preoperative treatment of esophageal cancer: results of a prospective multi-center trial (SAKK 75/02). Onkologie 2009; 32: 724-730
  • 439 Malik V. et al. Early repeated 18F-FDG PET scans during neoadjuvant chemoradiation fail to predict histopathologic response or survival benefit in adenocarcinoma of the esophagus. J Nucl Med 2010; 51: 1863-1869
  • 440 van Heijl M. et al. Fluorodeoxyglucose positron emission tomography for evaluating early response during neoadjuvant chemoradiotherapy in patients with potentially curable esophageal cancer. Ann Surg 2011; 253: 56-63
  • 441 Chang DT. et al. Treatment of esophageal cancer based on histology: a surveillance epidemiology and end results analysis. Am J Clin Oncol 2009; 32: 405-410
  • 442 Karran A. et al. Propensity score analysis of oesophageal cancer treatment with surgery or definitive chemoradiotherapy. Br J Surg 2014; 101: 502-510
  • 443 al-Sarraf M. et al. Progress report of combined chemoradiotherapy versus radiotherapy alone in patients with esophageal cancer: an intergroup study. J Clin Oncol 1997; 15: 277-284
  • 444 Crehange G. et al. Phase III trial of protracted compared with split-course chemoradiation for esophageal carcinoma: Federation Francophone de Cancerologie Digestive 9102. J Clin Oncol 2007; 25: 4895-4901
  • 445 Kawaguchi Y. et al. Patterns of failure associated with involved field radiotherapy in patients with clinical stage I thoracic esophageal cancer. Jpn J Clin Oncol 2011; 41: 1007-1012
  • 446 Wong R, Malthaner R. Combined chemotherapy and radiotherapy (without surgery) compared with radiotherapy alone in localized carcinoma of the esophagus. Cochrane Database Syst Rev 2006; Cd002092
  • 447 Yamada K. et al. Treatment results of chemoradiotherapy for clinical stage I (T1N0M0) esophageal carcinoma. Int J Radiat Oncol Biol Phys 2006; 64: 1106-1111
  • 448 Kat H. et al. A phase II trial of chemoradiotherapy for stage I esophageal squamous cell carcinoma: Japan Clinical Oncology Group Study (JCOG9708). Jpn J Clin Oncol 2009; 39: 638-643
  • 449 Kuwano H. et al. Guidelines for diagnosis and treatment of carcinoma of the esophagus. Esophagus 2008; 5: 117-132
  • 450 Gkika E. et al. Long-term results of definitive radiochemotherapy in locally advanced cancers of the cervical esophagus. Dis Esophagus 2014; 27: 678-684
  • 451 Grass GD. et al. Cervical esophageal cancer: A population-based study. Head Neck 2015; 37: 808-814
  • 452 Burmeister BH. et al. Thirty-four patients with carcinoma of the cervical esophagus treated with chemoradiation therapy. Arch Otolaryngol Head Neck Surg 2000; 126: 205-208
  • 453 Ajani JA. et al. Esophageal and esophagogastric junction cancers. J Natl Compr Canc Netw 2011; 9: 830-887
  • 454 Gao X. Treatment guideline of radiotherapy for Chinese esophageal carcinoma (draft). Chin J Cancer 2010; 29: 855-859
  • 455 Fenkell L. et al. Dosimetric comparison of IMRT vs. 3D conformal radiotherapy in the treatment of cancer of the cervical esophagus. Radiother Oncol 2008; 89: 287-291
  • 456 Ma JB. et al. Feasibility of involved-field conformal radiotherapy for cervical and upper-thoracic esophageal cancer. Onkologie 2011; 34: 599-604
  • 457 Ma JB. et al. Feasibility of Involved-Field Conformal Radiotherapy for Cervical and Upper- Thoracic Esophageal Cancer. Oncology Research and Treatment 2011; 34: 599-604
  • 458 Tai P. et al. Improving the consistency in cervical esophageal target volume definition by special training. Int J Radiat Oncol Biol Phys 2002; 53: 766-774
  • 459 Ott K. et al. Limited resection and free jejunal graft interposition for squamous cell carcinoma of the cervical oesophagus. Br J Surg 2009; 96: 258-266
  • 460 Panhofer P. et al. Influence of resection extent on morbidity in surgery for squamous cell cancer at the pharyngoesophageal junction. Langenbecks Arch Surg 2013; 398: 221-230
  • 461 Kranzfelder M. et al. Littoral cell angioma and angiosarcoma of the spleen: report of two cases in siblings and review of the literature. J Gastrointest Surg 2012; 16: 863-867
  • 462 Bedenne L. et al. Chemoradiation followed by surgery compared with chemoradiation alone in squamos cancer of the esophagus: FFCD 9102. J Clin Oncol 2007; 25: 1160-1168
  • 463 Minsky BD. et al. INT 0123 (Radiation Therapy Oncology Group 94-05) phase III trial of combined-modality therapy for esophageal cancer: high-dose versus standard-dose radiation therapy. J Clin Oncol 2002; 20: 1167-1174
  • 464 Ajani JA. et al. Esophageal and esophagogastric junction cancers. J Natl Compr Canc Netw 2011; 9: 830-887
  • 465 Crosby T. et al. Chemoradiotherapy with or without cetuximab in patients with oesophageal cancer (SCOPE1): a multicentre, phase 2/3 randomised trial. Lancet Oncol 2013; 14: 627-637
  • 466 Meerten E. et al. Definitive concurrent chemoradiation (CRT) with weekly paclitaxel and carboplatin for patients (pts) with irresectable esophageal cancer: A phase II study. in ASCO Annual Meeting Proceedings 2010
  • 467 Suntharalingam M. et al. The Initial Report of Local Control on RTOG 0436: A Phase 3 Trial Evaluating the Addition of Cetuximab to Paclitaxel, Cisplatin, and Radiation for Patients With Esophageal Cancer Treated Without Surgery. International Journal of Radiation Oncology Biology Physics 2014; 90: S3
  • 468 Stahl M. et al. Chemoradiation with and without surgery in patients with locally advanced squamous cell carcinoma of the esophagus. J Clin Oncol 2005; 23: 2310-2317
  • 469 Kole TP. et al. Comparison of heart and coronary artery doses associated with intensity-modulated radiotherapy versus three-dimensional conformal radiotherapy for distal esophageal cancer. Int J Radiat Oncol Biol Phys 2012; 83: 1580-1586
  • 470 Lee HK. et al. Postoperative pulmonary complications after preoperative chemoradiation for esophageal carcinoma: correlation with pulmonary dose-volume histogram parameters. Int J Radiat Oncol Biol Phys 2003; 57: 1317-1322
  • 471 Ruol A. et al. Interval between neoadjuvant chemoradiotherapy and surgery for squamous cell carcinoma of the thoracic esophagus: does delayed surgery have an impact on outcome?. Ann Surg 2010; 252: 788-796
  • 472 Kim JY. et al. Does the timing of esophagectomy after chemoradiation affect outcome?. Ann Thorac Surg 2012; 93: 207-212 ; discussion 212–213
  • 473 Tachimori Y. Role of salvage esophagectomy after definitive chemoradiotherapy. Gen Thorac Cardiovasc Surg 2009; 57: 71-78
  • 474 Tomimaru Y. et al. Factors affecting the prognosis of patients with esophageal cancer undergoing salvage surgery after definitive chemoradiotherapy. J Surg Oncol 2006; 93: 422-428
  • 475 Marks JL. et al. Salvage esophagectomy after failed definitive chemoradiation for esophageal adenocarcinoma. Ann Thorac Surg 2012; 94: 1126-1132 ; discussion 1132–1133
  • 476 Taketa T. et al. Propensity-based matching between esophagogastric cancer patients who had surgery and who declined surgery after preoperative chemoradiation. Oncology 2013; 85: 95-99
  • 477 Ariga H. et al. Prospective comparison of surgery alone and chemoradiotherapy with selective surgery in resectable squamous cell carcinoma of the esophagus. Int J Radiat Oncol Biol Phys 2009; 75: 348-356
  • 478 Zheng B. et al. Role of adjuvant chemoradiotherapy in treatment of resectable esophageal carcinoma: a meta-analysis. Chin Med J (Engl) 2013; 126: 1178-1182
  • 479 Thallinger CM. et al. Pre- and postoperative treatment modalities for esophageal squamous cell carcinoma. Anticancer Res 2012; 32: 4609-4627
  • 480 Xu Y. et al. Neoadjuvant versus adjuvant treatment: which one is better for resectable esophageal squamous cell carcinoma?. World J Surg Oncol 2012; 10: 173
  • 481 Lv J. et al. Long-term efficacy of perioperative chemoradiotherapy on esophageal squamous cell carcinoma. World J Gastroenterol 2010; 16: 1649-1654
  • 482 Cao XF. et al. A prospective comparison between surgery alone and postoperative chemoradiotherapy for locally advanced esophageal squamous cell carcinoma. Zhonghua Zhong Liu Za Zhi 2010; 32: 452-455
  • 483 Ohri N. et al. Who benefits from adjuvant radiation therapy for gastric cancer? A meta-analysis. Int J Radiat Oncol Biol Phys 2013; 86: 330-335
  • 484 Bamias A. et al. A randomized phase III study of adjuvant platinum/docetaxel chemotherapy with or without radiation therapy in patients with gastric cancer. Cancer Chemother Pharmacol 2010; 65: 1009-1021
  • 485 Lee J. et al. Phase III trial comparing capecitabine plus cisplatin versus capecitabine plus cisplatin with concurrent capecitabine radiotherapy in completely resected gastric cancer with D2 lymph node dissection: the ARTIST trial. J Clin Oncol 2012; 30: 268-273
  • 486 Macdonald JS. et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med 2001; 345: 725-730
  • 487 Kwon HC. et al. Adjuvant chemoradiation versus chemotherapy in completely resected advanced gastric cancer with D2 nodal dissection. Asia Pac J Clin Oncol 2010; 6: 278-285
  • 488 Paoletti X. et al. Benefit of adjuvant chemotherapy for resectable gastric cancer: a meta-analysis. Jama 2010; 303: 1729-1737
  • 489 Abate E. et al. Recurrence after esophagectomy for adenocarcinoma: defining optimal follow-up intervals and testing. J Am Coll Surg 2010; 210: 428-435
  • 490 Mantziari S. et al. Gastroesophageal cancer: an update on diagnosis and treatment. Rev Med Suisse 2014; 10: 1331-1336
  • 491 Ouattara M. et al. Body mass index kinetics and risk factors of malnutrition one year after radical oesophagectomy for cancer. Eur J Cardiothorac Surg 2012; 41: 1088-1093
  • 492 Haverkort EB. et al. Suboptimal intake of nutrients after esophagectomy with gastric tube reconstruction. J Acad Nutr Diet 2012; 112: 1080-1087
  • 493 Ryan AM. et al. Post-oesophagectomy early enteral nutrition via a needle catheter jejunostomy: 8-year experience at a specialist unit. Clin Nutr 2006; 25: 386-393
  • 494 Mishra SI. et al. Exercise interventions on health-related quality of life for people with cancer during active treatment. Cochrane Database Syst Rev 2012; 8: Cd008465
  • 495 Bourke L. et al. Interventions to improve exercise behaviour in sedentary people living with and beyond cancer: a systematic review. Br J Cancer 2014; 110: 831-841
  • 496 Rock CL. et al. Nutrition and physical activity guidelines for cancer survivors. CA Cancer J Clin 2012; 62: 243-274
  • 497 Mishra SI. et al. Exercise interventions on health-related quality of life for cancer survivors. Cochrane Database Syst Rev 2012; 8: Cd007566
  • 498 Cunningham D. et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med 2008; 358: 36-46
  • 499 Webb A. et al. Randomized trial comparing epirubicin, cisplatin, and fluorouracil versus fluorouracil, doxorubicin, and methotrexate in advanced esophagogastric cancer. J Clin Oncol 1997; 15: 261-267
  • 500 Ross P. et al. Prospective randomized trial comparing mitomycin, cisplatin, and protracted venous-infusion fluorouracil (PVI 5-FU) With epirubicin, cisplatin, and PVI 5-FU in advanced esophagogastric cancer. J Clin Oncol 2002; 20: 1996-2004
  • 501 Van Cutsem E. et al. Phase III study of docetaxel and cisplatin plus fluorouracil compared with cisplatin and fluorouracil as first-line therapy for advanced gastric cancer: a report of the V325 Study Group. J Clin Oncol 2006; 24: 4991-4997
  • 502 Ajani JA. et al. Multicenter phase III comparison of cisplatin/S-1 with cisplatin/infusional fluorouracil in advanced gastric or gastroesophageal adenocarcinoma study: the FLAGS trial. J Clin Oncol 2010; 28: 1547-1553
  • 503 Al-Batran SE. et al. Phase III trial in metastatic gastroesophageal adenocarcinoma with fluorouracil, leucovorin plus either oxaliplatin or cisplatin: a study of the Arbeitsgemeinschaft Internistische Onkologie. J Clin Oncol 2008; 26: 1435-1442
  • 504 Kang YK. et al. Capecitabine/cisplatin versus 5-fluorouracil/cisplatin as first-line therapy in patients with advanced gastric cancer: a randomised phase III noninferiority trial. Ann Oncol 2009; 20: 666-673
  • 505 Okines AF. et al. Meta-analysis of the REAL-2 and ML17032 trials: evaluating capecitabine-based combination chemotherapy and infused 5-fluorouracil-based combination chemotherapy for the treatment of advanced oesophago-gastric cancer. Ann Oncol 2009; 20: 1529-1534
  • 506 Lutz MP. et al. Weekly infusional high-dose fluorouracil (HD-FU), HD-FU plus folinic acid (HD- FU/FA), or HD-FU/FA plus biweekly cisplatin in advanced gastric cancer: randomized phase II trial 40953 of the European Organisation for Research and Treatment of Cancer Gastrointestinal Group and the Arbeitsgemeinschaft Internistische Onkologie. J Clin Oncol 2007; 25: 2580-2585
  • 507 Al-Batran SE. et al. Biweekly fluorouracil, leucovorin, oxaliplatin, and docetaxel (FLOT) for patients with metastatic adenocarcinoma of the stomach or esophagogastric junction: a phase II trial of the Arbeitsgemeinschaft Internistische Onkologie. Ann Oncol 2008; 19: 1882-1887
  • 508 NCCN Clinical Practice Guidelines in Oncology: Esophageal and Esophagastric Junction Cancers. National Comprehensive Cancer Network. 2011
  • 509 Xiang XJ. et al. A phase II study of capecitabine plus oxaliplatin as first-line chemotherapy in elderly patients with advanced gastric cancer. Chemotherapy 2012; 58: 1-7
  • 510 Catalano V. et al. A phase II study of modified FOLFOX as first-line chemotherapy for metastatic gastric cancer in elderly patients with associated diseases. Gastric Cancer 2013; 16: 411-419
  • 511 Al-Batran SE. et al. The feasibility of triple-drug chemotherapy combination in older adult patients with oesophagogastric cancer: a randomised trial of the Arbeitsgemeinschaft Internistische Onkologie (FLOT65+). Eur J Cancer 2013; 49: 835-842
  • 512 Hall PS. et al. A randomised phase II trial and feasibility study of palliative chemotherapy in frail or elderly patients with advanced gastroesophageal cancer (321GO). Br J Cancer 2017; 116: 472-478
  • 513 Grunberger B. et al. Palliative chemotherapy for recurrent and metastatic esophageal cancer. Anticancer Res 2007; 27: 2705-2714
  • 514 NCCN practice guidelines for the management of psychosocial distress. National Comprehensive Cancer Network. 1999
  • 515 Adam R. et al. Hepatic resection for noncolorectal nonendocrine liver metastases: analysis of 1,452 patients and development of a prognostic model. Ann Surg 2006; 244: 524-535
  • 516 Shiono S. et al. Disease-free interval length correlates to prognosis of patients who underwent metastasectomy for esophageal lung metastases. J Thorac Oncol 2008; 3: 1046-1049
  • 517 Ichikawa H. et al. Operative treatment for metachronous pulmonary metastasis from esophageal carcinoma. Surgery 2011; 149: 164-170
  • 518 Ichida H. et al. Pattern of postoperative recurrence and hepatic and/or pulmonary resection for liver and/or lung metastases from esophageal carcinoma. World J Surg 2013; 37: 398-407
  • 519 Miyata H. et al. Salvage esophagectomy after definitive chemoradiotherapy for thoracic esophageal cancer. J Surg Oncol 2009; 100: 442-446
  • 520 Morita M. et al. Clinical significance of salvage esophagectomy for remnant or recurrent cancer following definitive chemoradiotherapy. J Gastroenterol 2011; 46: 1284-1291
  • 521 Schieman C. et al. Salvage resections for recurrent or persistent cancer of the proximal esophagus after chemoradiotherapy. Ann Thorac Surg 2013; 95: 459-463
  • 522 Thuss-Patience PC. et al. Survival advantage for irinotecan versus best supportive care as second-line chemotherapy in gastric cancer – a randomised phase III study of the Arbeitsgemeinschaft Internistische Onkologie (AIO). Eur J Cancer 2011; 47: 2306-2314
  • 523 Hironaka S. et al. Randomized, open-label, phase III study comparing irinotecan with paclitaxel in patients with advanced gastric cancer without severe peritoneal metastasis after failure of prior combination chemotherapy using fluoropyrimidine plus platinum: WJOG 4007 trial. J Clin Oncol 2013; 31: 4438-4444
  • 524 Ford HE. et al. Docetaxel versus active symptom control for refractory oesophagogastric adenocarcinoma (COUGAR-02): an open-label, phase 3 randomised controlled trial. Lancet Oncol 2014; 15: 78-86
  • 525 Fuchs CS. et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 2014; 383: 31-39
  • 526 Wilke H. et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol 2014; 15: 1224-1235
  • 527 Thallinger CM, Raderer M, Hejna M. Esophageal cancer: a critical evaluation of systemic second-line therapy. J Clin Oncol 2011; 29: 4709-4714
  • 528 Thuss-Patience PC. et al. Trastuzumab emtansine versus taxane use for previously treated HER2-positive locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma (GATSBY): an international randomised, open-label, adaptive, phase 2/3 study. Lancet Oncol 2017; 18: 640-653
  • 529 Amdal CD. et al. Patient-reported outcomes evaluating palliative radiotherapy and chemotherapy in patients with oesophageal cancer: a systematic review. Acta Oncol 2013; 52: 679-690
  • 530 Sgourakis G. et al. Survival after chemotherapy and/or radiotherapy versus self-expanding metal stent insertion in the setting of inoperable esophageal cancer: a case-control study. BMC Cancer 2012; 12: 70
  • 531 Javed A. et al. Palliative stenting with or without radiotherapy for inoperable esophageal carcinoma: a randomized trial. J Gastrointest Cancer 2012; 43: 63-69
  • 532 Amdal CD. et al. Palliative brachytherapy with or without primary stent placement in patients with oesophageal cancer, a randomised phase III trial. Radiother Oncol 2013; 107: 428-433
  • 533 Homs MY. et al. Single-dose brachytherapy versus metal stent placement for the palliation of dysphagia from oesophageal cancer: multicentre randomised trial. Lancet 2004; 364: 1497-1504
  • 534 Rosenblatt E. et al. Adding external beam to intra-luminal brachytherapy improves palliation in obstructive squamous cell oesophageal cancer: a prospective multi-centre randomized trial of the International Atomic Energy Agency. Radiother Oncol 2010; 97: 488-494
  • 535 Sgourakis G. et al. The use of self-expanding stents in esophageal and gastroesophageal junction cancer palliation: a meta-analysis and meta-regression analysis of outcomes. Dig Dis Sci 2010; 55: 3018-3030
  • 536 Sabharwal T. et al. A randomised prospective comparison of the Flamingo Wallstent and Ultraflex stent for palliation of dysphagia associated with lower third oesophageal carcinoma. Gut 2003; 52: 922-926
  • 537 Shenfine J. et al. A pragmatic randomised controlled trial of the cost-effectiveness of palliative therapies for patients with inoperable oesophageal cancer. Health Technol Assess 2005; 9: 1-121
  • 538 Vakil N. et al. A prospective, randomized, controlled trial of covered expandable metal stents in the palliation of malignant esophageal obstruction at the gastroesophageal junction. Am J Gastroenterol 2001; 96: 1791-1796
  • 539 Miyayama S. et al. Malignant esophageal stricture and fistula: palliative treatment with polyurethane-covered Gianturco stent. J Vasc Interv Radiol 1995; 6: 243-248
  • 540 Alexander EP. et al. Evolving management and outcome of esophageal cancer with airway involvement. Ann Thorac Surg 2001; 71: 1640-1644
  • 541 Conio M. et al. A randomized prospective comparison of self-expandable plastic stents and partially covered self-expandable metal stents in the palliation of malignant esophageal dysphagia. Am J Gastroenterol 2007; 102: 2667-2677
  • 542 Homs MY. et al. Esophageal stents with antireflux valve for tumors of the distal esophagus and gastric cardia: a randomized trial. Gastrointest Endosc 2004; 60: 695-702
  • 543 Shim CS. et al. Management of malignant stricture of the esophagogastric junction with a newly designed self-expanding metal stent with an antireflux mechanism. Endoscopy 2005; 37: 335-339
  • 544 Wenger U. et al. An antireflux stent versus conventional stents for palliation of distal esophageal or cardia cancer: a randomized clinical study. Surg Endosc 2006; 20: 1675-1680
  • 545 Nagaraja V, Cox MR, Eslick GD. Safety and efficacy of esophageal stents preceding or during neoadjuvant chemotherapy for esophageal cancer: a systematic review and meta-analysis. J Gastrointest Oncol 2014; 5: 119-126
  • 546 Mariette C. et al. Self-expanding covered metallic stent as a bridge to surgery in esophageal cancer: impact on oncologic outcomes. J Am Coll Surg 2015; 220: 287-296
  • 547 Bergquist H. et al. Stent insertion or endoluminal brachytherapy as palliation of patients with advanced cancer of the esophagus and gastroesophageal junction. Results of a randomized, controlled clinical trial. Dis Esophagus 2005; 18: 131-139
  • 548 Wenger U. et al. Health economic evaluation of stent or endoluminal brachytherapy as a palliative strategy in patients with incurable cancer of the oesophagus or gastro-oesophageal junction: results of a randomized clinical trial. Eur J Gastroenterol Hepatol 2005; 17: 1369-1377
  • 549 Bergquist H. et al. Combined stent insertion and single high-dose brachytherapy in patients with advanced esophageal cancer--results of a prospective safety study. Dis Esophagus 2012; 25: 410-415
  • 550 Carazzone A. et al. Endoscopic palliation of oesophageal cancer: results of a prospective comparison of Nd:YAG laser and ethanol injection. Eur J Surg 1999; 165: 351-356
  • 551 Spencer GM. et al. Laser augmented by brachytherapy versus laser alone in the palliation of adenocarcinoma of the oesophagus and cardia: a randomised study. Gut 2002; 50: 224-227
  • 552 Norberto L. et al. Endoscopic palliation of esophageal and cardial cancer: neodymium-yttrium aluminum garnet laser therapy. Dis Esophagus 1999; 12: 294-296
  • 553 Loizou LA. et al. A prospective comparison of laser therapy and intubation in endoscopic palliation for malignant dysphagia. Gastroenterology 1991; 100: 1303-1310
  • 554 Carter R, Smith JS, Anderson JR. Laser recanalization versus endoscopic intubation in the palliation of malignant dysphagia: a randomized prospective study. Br J Surg 1992; 79: 1167-1170
  • 555 Barr H. et al. Prospective randomised trial of laser therapy only and laser therapy followed by endoscopic intubation for the palliation of malignant dysphagia. Gut 1990; 31: 252-258
  • 556 Manner H. et al. The tissue effect of second generation argon plasma coagulation (VIO APC) in comparison to standard APC and Nd:YAG laser in vitro. Acta Gastroenterol Belg 2007; 70: 352-356
  • 557 Eickhoff A. et al. Prospective nonrandomized comparison of two modes of argon beamer (APC) tumor desobstruction: effectiveness of the new pulsed APC versus forced APC. Endoscopy 2007; 39: 637-642
  • 558 Manner H. et al. Prospective evaluation of a new high-power argon plasma coagulation system (hp-APC) in therapeutic gastrointestinal endoscopy. Scand J Gastroenterol 2007; 42: 397-405
  • 559 Rupinski M. et al. Randomized comparison of three palliative regimens including brachytherapy, photodynamic therapy, and APC in patients with malignant dysphagia (CONSORT 1a) (Revised II). The American journal of gastroenterology 2011; 106: 1612-1620
  • 560 Dempster M. et al. Psychological distress among survivors of esophageal cancer: the role of illness cognitions and coping. Dis Esophagus 2012; 25: 222-227
  • 561 Djärv T, Lagergren P. Six-month postoperative quality of life predicts long-term survival after oesophageal cancer surgery. European journal of cancer 2011; 47: 530-535
  • 562 Heijl M. et al. Preoperative and Early Postoperative Quality of Life Predict Survival in Potentially Curable Patients with Esophageal Cancer. Annals of Surgical Oncology 2010; 17: 23-30
  • 563 Gockel I. et al. Long-term survivors of esophageal cancer: Disease-specific quality of life, general health and complications. Journal of Surgical Oncology 2010; 102: 516-522
  • 564 Anstee S. et al. Developing a matrix to identify and prioritise research recommendations in HIV prevention. BMC Public Health 2011; 11: 381
  • 565 Herschbach P, Weis J. Screeningverfahren in der Psychoonkologie.Testinstrumente zur Identifikation betreuungsbedürftiger Krebspatienten. Dt. Krebsgesellschaft; 2010 http://www.krebsgesellschaft.de/download/pso_broschuere.pdf
  • 566 Mehnert A. et al. Assessment of psychosocial distress and resources in oncology – a literature review about screening measures and current developments. Psychother Psychosom Med Psychol 2006; 56: 462-479
  • 567 Mitchell AJ. Short screening tools for cancer-related distress: a review and diagnostic validity meta-analysis. J Natl Compr Canc Netw 2010; 8: 487-494
  • 568 Vodermaier A, Linden W, Siu C. Screening for emotional distress in cancer patients: a systematic review of assessment instruments. J Natl Cancer Inst 2009; 101: 1464-1488
  • 569 Vodermaier A, Millman RD. Accuracy of the Hospital Anxiety and Depression Scale as a screening tool in cancer patients: a systematic review and meta-analysis. Support Care Cancer 2011; 19: 1899-1908
  • 570 Weis J, Schumacher A, Blettner G. Psychoonkologie: Konzepte und Aufgaben einer jungen Fachdisziplin. Der Onkologe 2007; 13: 185-194
  • 571 Faller H. et al. Effects of psycho-oncologic interventions on emotional distress and quality of life in adult patients with cancer: systematic review and meta-analysis. J Clin Oncol 2013; 31: 782-793
  • 572 Ärztliches Zentrum für Qualität in der Medizin (Hrsg.). Kompendium Q-M-A. 3. überarbeitete und erweiterte Auflage. Köln: Deutscher Ärzte-Verlag; 2008
  • 573 Donabedian A. Evaluating the quality of medical care. Milbank Mem Fund Q 1966; 44: 166-206
  • 574 Ärztliches Zentrum für Qualität in der Medizin (ÄZQ), Manual Qualitätsindikatoren. 2009 äzq Schriftenreihe: Berlin
  • 575 Vlayen J. et al. Quality indicators for the management of upper gastrointestinal cancer – synthesis. Good Clinical Practice (GCP). Brussels: Belgian Health Care Knowledge Centre (KCE); 2013 KCE Reports 200 Cs. D/2013/10.273/16
  • 576 Walters DM. et al. Understanding mortality as a quality indicator after esophagectomy. Ann Thorac Surg 2014; 98: 506-511 ; discussion 511–512
  • 577 Talsma AK. et al. The 30-day versus in-hospital and 90-day mortality after esophagectomy as indicators for quality of care. Ann Surg 2014; 260: 267-273