Skip to main content
Erschienen in: Clinical and Translational Oncology 12/2015

Open Access 01.12.2015 | Clinical Guides in Oncology

Seom guidelines for the treatment of gastric cancer 2015

verfasst von: M. Martin-Richard, A. Custodio, C. García-Girón, C. Grávalos, C. Gomez, P. Jimenez-Fonseca, J. L. Manzano, C. Pericay, F. Rivera, A. Carrato

Erschienen in: Clinical and Translational Oncology | Ausgabe 12/2015

Abstract

Gastric cancer is the fourth cause of death by cancer in Spain and a significant medical problem. Molecular biology results evidence that gastroesophageal junction tumors and gastric cancer should be considered as two independent entities with a different prognosis and treatment approach. Endoscopic resection in very early tumors is feasible. Neoadjuvant and adjuvant therapy in locally advanced resectable tumor increase overall survival and should be considered standard treatments. In stage IV tumors, platinum–fluoropyrimidine-based schedule, with trastuzumab in HER2-overexpressed tumors, is the first-line treatment. Different therapies in second line have demonstrated in randomized studies their clear benefit in survival improvement.
Hinweise
An erratum to this article can be found at http://​dx.​doi.​org/​10.​1007/​s12094-016-1491-3.

Introduction

Gastric cancer (GC) is a major contributor to the global cancer burden. In 2012, there were an estimated 951,000 new cases worldwide, 6.8 % of the total, making GC the fifth most common malignancy [1]. GC is the third leading cause of cancer death in both sexes worldwide with 723,000 deaths, 8.8 % of the total. In Spain, GC was the sixth most common malignancy, with 7810 new cases and 5389 deaths [2]. Geographically, almost two-thirds occur in Asia, with 43 % of total global cases in China. Incidence rates are low in developed regions such as North America and Western Europe. GC is more frequent among males than in females (11.0 versus 5.1 per 100,000) and its incidence increases with age (peak presentation is between 65- and 74-year old). Mortality age-standardized rate is 9.7 in men and 4.6 in women [3]. The most important familial cancer syndrome is hereditary diffuse gastric cancer syndrome [mutations in the germline E-cadherin mutations (CDH1) gene] [1].
Although often reported as a single entity, GC can generally be classified [4] into two topographical categories: (1) cardia gastric cancer (CGC) arising in the area of the stomach adjoining the esophageal–gastric junction [5], and (2) non-CGC (NCGC) arising from more distal regions of the stomach. Risk factors for CGC include obesity, gastro-esophagus reflux disease (GORD) and Barrett’s esophagus. NCGC, however, is strongly associated with Helicobacter pylori infection, socioeconomic status (SES), excess nitrates, and atrophic gastritis. Both types are influenced by cigarette smoking and by low intake of fruits and vegetables. Given these differences, there is increasing interest in describing the worldwide burden of CGC and NCGC subsites separately [6]. CGC: there were 260,000 new cases, comprising 27 % of total gastric cancer worldwide (30 % in men and 21 % in women). Age-standardized rate was 3.3 per 100,000 (5.3 versus 1.6 in men and women, respectively). NCGC: there were 691,000 new cases, 70 % in men and 79 % in women. Age-standardized rate was 8.8 per 100,000 (12.1 versus 5.9 in men and women, respectively). In most countries, CGC incidence rates are lower than NCGC rates (91 % of 184 GLOBOCAN countries).
These clinical guidelines aim to offer succinct, practical recommendations for diagnosis, treatment, and follow-up of GC.

Molecular classification of gastric cancer

Gastric cancer is known as a heterogeneous disease, which may be divided into subgroups based on histological, anatomical, epidemiological, and also molecular classifications [712].
Despite such incredible advance in our knowledge of gastric cancer genomics, there is little overlap between the published classifications, and the reproducibility of the different studies is very poor [13]. Therefore, it is difficult to compare expression data from different studies and such inconsistency limits our ability to develop robust and reliable molecular models.

Molecular prognosis of gastric cancer

There is no one single molecular alteration that has been universally accepted as independent prognostic factor in gastric cancer. Many gene expression signatures have been able to classify tumors into intrinsic subtypes and predict the survival of GC patients. Some studies have showed that gene expression profiling can predict patients with a high risk for recurrence after curative surgery [1416]. Other studies using gene expression data with proper clinical information have developed predictive models that could identify long survivors, either by identifying specific gene signatures that correlated with the overall survival [1719] or by identification of Genomic signatures that could successfully predict the relapse of GC, specially in form of peritoneal relapse [20].
Despite all these profits in the development of prognostic and predictive molecular models for GC, they are at an early stage and in need of substantial improvement to be ready for its clinical implementation.

Diagnosis and staging

Diagnosis

Gastroscopy and biopsy of suspicious lesions are the basis for definitive diagnosis. Histology is reported according to World Health Organization criteria [21] and Lauren classification (intestinal and diffuse). Immunohistochemistry (IHC) determines HER2 overexpression in advanced disease, according to GC-specific criteria [22], to decide trastuzumab treatment.

Staging

WHO performance status (PS), nutritional status, and comprehensive geriatric assessment in the elderly, as well as physical exam, liver, renal and blood tests, and tumor stage should be considered before choosing treatment;
Locoregional disease:
  • Endoscopic ultrasound (EUS) and ultrasound-guided fine-needle aspiration of suspicious lymph nodes inform about locoregional disease spread and are optimal to distinguish T2–4 staging [IIA] [23] or
  • Esophagus-gastro-duodenal transit, when endoscopy cannot be performed.
Distant disease:
  • Computed tomography (CT) is standard to confirm metastases (IA).
  • Laparoscopy, peritoneal washings, and cytology is mandatory in locally advanced gastric (T3–4 and/or node-positive disease) and esophagogastric junction cancer [IIA] [24]
  • The value of integrated PET/CT in patients who are offered curative surgery is a subject to debate, although it may be convenient in large tumor size, non-signet ring cell, non-mucinous, non-diffuse carcinoma type, and glucose transporter 1-positive expression on immunohistochemistry [25].
  • Combinations of CEA, CA19-9, and CA72-4 are the most effective serum tumor markers for staging, detection of recurrence, or evaluation of the response [26].
  • Staging is performed according to the 2010 AJCC TNM classification, 7th edition (Table 1) [4]. Four major groups are considered for clinical management purposes (Table 2).
    Table 1
    Tumor stage of gastric cancer according to AJCC 2010
    Tx
    Primary tumor cannot be assessed
      
    T0
    No evidence of primary tumor
      
    Tis
    Carcinoma in situ: intraepithelial tumor without invasion of the lamina propria
      
    T1
    Tumor invades lamina propria, muscularis mucosae, or submucosa
      
    T1a
    Tumor invades lamina propria or muscularis mucosae
      
    T1b
    Tumor invades submucosa
      
    T2
    Tumor invades muscularis propriaa
      
    T3
    Tumor penetrates subserosal connective tissue without invasion of visceral peritoneum or adjacent structuresb
      
    T4
    Tumor invades serosa (visceral peritoneum) or adjacent structuresb
      
    T4a
    Tumor invades serosa (visceral peritoneum)
      
    T4b
    Tumor invades adjacent structures
      
    Nx
    Regional lymph node(s) cannot be assessed
      
    N0
    No regional lymph node metastasisc
      
    N1
    Metastasis in 1–2 regional lymph nodes
      
    N2
    Metastasis in 3–6 regional lymph nodes
      
    N3
    Metastasis in seven or more regional lymph nodes
      
    N3a
    Metastasis in 7–15 regional lymph nodes
      
    N3b
    Metastasis in 16 or more regional lymph nodes
      
    Mx
    Distant metastasis cannot be assessed
      
    M0
    No distant metastasis
      
    M1
    Distant metastasis
      
    Stage
    T
    N
    M
    Anatomic stage/prognostic groups
    0
    Tis
    N0
    M0
    IA
    T1
    N0
    M0
    IB
    T2
    N0
    M0
    T1
    N1
    M0
    IIA
    T3
    N0
    M0
    T2
    N1
    M0
    T1
    N2
    M0
    IIB
    T4a
    N0
    M0
    T3
    N1
    M0
    T2
    N2
    M0
    T1
    N3
    M0
    IIIA
    T4a
    N1
    M0
    T3
    N2
    M0
    T2
    N3
    M0
    IIIB
    T4b
    N0
    M0
    T4b
    N1
    M0
    T4a
    N2
    M0
    T3
    N3
    M0
    IIIC
    T4b
    N2
    M0
    T4b
    N3
    M0
    T4a
    N3
    M0
    IV
    Any T
    Any N
    M1
    cTNM is the clinical classification, pTNM is the pathologic classification
    Primary tumor (T), Regional lymph nodes (N)
    aA tumor may penetrate the muscularis propria with extension into the gastrocolic or gastrohepatic ligaments, or into the greater or lesser omentum, without perforation of the visceral peritoneum covering these structures. In this case, the tumor is classified T3. If there is perforation of the visceral peritoneum covering the gastric ligaments or the omentum, the tumor should be classified T4
    bThe adjacent structures of the stomach include the spleen, transverse colon, liver, diaphragm, pancreas, abdominal wall, adrenal gland, kidney, small intestine, and retroperitoneum. Intramural extension to the duodenum or esophagus is classified by the depth of the greatest invasion in any of these sites, including the stomach
    cA designation of pN0 should be used if all examined lymph nodes are negative, regardless of the total number removed and examined
    Table 2
    Prognosis and treatment options
    Groups
    Early resectable disease (10 %)
    Locally advanced resectable disease
    Locally advanced unresectable disease (20 %)
    Metastatic disease (30 %)
    Stages
    Stages 0–I;
    II–IIIC
    Some IIIB–IIIC
    IV
    5-year/median OS
    70 %
    30–40 %
    12–14 months
    9–11 m with CT
    4 m without CT
    Treatment
    Surgery or Endoscopic resection
    Perioperative, Neoadjuvant o Adjuvant ttm.
    CT
    CT
    CT chemotherapy, ttm treatment

Treatment

Early gastric cancer

Endoscopic resection

Early GC (T1a) may be amenable to endoscopic resection if it is well differentiated, <2 cm, confined to the mucosa, and non-ulcerated. Intestinal Lauren histology and no evidence of lympho-vascular invasion also indicate mucosectomy in: intramucosal cancers without ulceration, regardless of tumor size; ulcerated intramucosal cancers <3 cm, or cancers with early invasion into the submucosa measuring <3 cm, endoscopic submucosal dissection has proven more effective than endoscopic mucosal resection, but requires greater skills and instrumentation and entails significant risk of complications, including perforation [27]. The risk of lymph node metastases following endoscopic resection by experts remains low in these tumors [28]. In less experienced centers, limited surgery is an alternative. T1A GC not meeting criteria for endoscopic treatment will require less extensive surgery than IB-III tumors and lymph node dissection can be limited to peri-gastric and local nodes (Table 3).
Table 3
Treatment recommendations
Stage
Details
Treatment
Early stage: Tis
 T1a
 T1a
 T1b
 T1b
Well dif./<2 cm/non-ulcerated/intestinal
Others non-ulcerated
<3 cm
Others
Endoscopic resection
Endosc. resect mucosectomy/surgery
Submucos. resect/surgery
Surgery
Stage I
 
Surgery
Locally Advanced (Stage II–III)
Cardias GC
Neoadjuvant CT (IB)
Or CRT (IA)
Or Perioperative CT (IB)
Or Adjuvant CT (IA)
Or Adjuvant CRT (IB)
Non-cardias GC
Perioperative CT (IB)
Or Adjuvant CT (IA)
Or Adjuvant CRT (IB)
Advanced disease (Stage IV)
First-line CT
 HER2+
Cisplatin–Fluorop (IB)
 HER2 negative
PFluorop or EPFluorop (IA)
Or TCF (IB)
Or FOLFIRI or IF (IB)
Second-line CT
Irinotecan (IA)
Or Docetaxel (IA)
Or Paclitaxel (IB)
Or Ramucirumab (IB)
Or Paclitaxel–Rramucirum (IB)
Fluorop. 5Fu or Capecitabine, P cisplatin or oxaliplatin, TCF taxotere + cisplatin + 5FU

Locally advanced disease

Surgery

Complete resection with adequate margins remains the cornerstone of curative treatment. The type of resection in GC, subtotal versus total gastrectomy, depends on the anatomic location of primary tumor. For esophagogastric junction (EGJ) cancers, a total esophagectomy with a partial gastrectomy or an extended gastrectomy is generally performed.
Extent of lymph node dissection remains a subject of controversy. In eastern Asia, gastrectomy with D2 lymph node dissection is the standard treatment for curable GC. In Western countries, two large randomized trials failed to demonstrate a significant survival benefit for D2 over D1 lymph node dissection. However, mature 15-year follow-up data from the Dutch trial showed a lower locoregional recurrence rate and fewer GC-related death with D2 lymphadenectomy [29]. A recent meta-analysis of 12 randomized clinical trials confirmed no OS benefit for D2 lymphadenectomy, although a benefit was seen among patients who had resection without splenectomy and/or pancreatectomy [30]. There is uniform consensus that lymphadenectomy must include at least 15 lymph nodes. Gastrectomy with D2 lymph node dissection is a recommended procedure (2B), but should be performed by experienced surgeons in high-volume centers. Routine pancreatectomy and splenectomy are no longer recommended with D2 lymph node dissection.

Neoadjuvant treatment

Chemotherapy (CT)
A study by the EORTC (40954) in patients with locally advanced cancer of the stomach or EGJ found a significantly higher rate of R0 resection among patients receiving neoadjuvant CT; however, no statistically significant difference in survival was reached [31]. Some meta-analysis in GC has been conduced with conflicting results. A meta-analysis in EG cancers, where several EGJ were included [32], showed a survival benefit for neoadjuvant CT. Currently, we should consider neoadjuvant chemotherapy in gastroesophageal cancer (IB).
Chemoradiotherapy (CRT)
The phase III study POET compared preoperative CT with preoperative CRT in patients with locally advanced adenocarcinoma of the EGJ. Patients in the CRT group had a significant higher pathologic complete response, but statistical significance was not achieved for overall survival (OS) [33]. The phase III CROSS trial compared neoadjuvant CRT versus surgery alone in patients with squamous cell carcinoma and adenocarcinoma of the esophagus or EGJ, with a significant increase of OS in the neoadjuvant group [34]. The meta-analysis published by Sjoquist et al. [32] supports a survival benefit for neoadjuvant CRT compared to surgery alone for esophageal and EGJ cancer. Preoperative combined CRT is now the preferred approach for localized EGJ and gastric cardia cancers (IB). However, it is still an experimental procedure in potentially resectable non-cardia gastric adenocarcinomas (2B).

Perioperative treatment

The theoretical advantages of perioperative treatment are the potential increase of the complete resection (R0) rate, a better tolerability profile and increased probability of treatment compliance, early systemic treatment of micrometastatic disease, and an ideal scenario to assess the efficacy of scheduled treatment, as well as new agents.
Two phase III, adequately powered trials in Western countries, the MAGIC [35] and the FNLCC/FFCD 9703 [36], and some recent meta-analysis [37] have shown that perioperative chemotherapy (CT) significantly increases R0 rates, disease-free survival (DFS), and OS, and does not significantly increase perioperative complications or mortality with tolerable grade 3–4 toxicity rates (Table 1). These results have led to the adoption of perioperative CT as a standard approach for medically fit patients with resectable locally advanced (cT2 or higher, any N) distal esophageal, esophagogastric junction, or gastric tumors [IA] throughout most of European countries and other parts of the world.
Regarding the role of targeted agents in the perioperative setting, two phase II trials have tested the combination of trastuzumab and CT in HER-2-positive patients showing promising R0 rates and pathologic complete responses (pCR). However, the addition of bevacizumab to a perioperative regimen of epirubicin, cisplatin, and capecitabine (ECX) did not improve survival [38].
The potential benefit of adding postoperative or preoperative CRT to standard perioperative CT is being evaluated in two trials.

Adjuvant treatment

Chemoradiation
In patients with resected gastric or GEJ adenocarcinoma stages IB–IV (M0), the INT-0116 trial reported better OS (HR 1.35; p = 0.005) and DFS (HR 1.52; p < 0.001) with the MacDonald regimen (5FU/LV plus radiotherapy) versus surgery alone [39] (IB). In the 13-year follow-up, the benefit of adjuvant CRT was maintained and not substantial long-term toxicities were reported [40]. The CALGB 80101 compared the INT-0116 regimen with ECF (epirubicin, cisplatin, 5FU before and after 5FU/RT) in resected GEJ or gastric cancer without observing differences in 3-year OS (52 and 50 % for ECF and 5FU/LV, respectively). In HER2-positive tumors, the phase II TOXAG trial will analyze the safety of adjuvant oxaliplatin, capecitabine', and trastuzumab with radiotherapy.
Chemotherapy
The benefit of adjuvant CT has also been reported. An absolute increment of 6 % in OS (HR 0.82; p < 0.001) and a better DFS (HR 0.82; p < 0.001) were published in a large, individual patient-level meta-analysis of adjuvant 5FU-based chemotherapy versus surgery alone in resected GC [41] [I,A]. However, the preferred combination chemotherapy could not be determined.
In addition, the ACTS-GC randomized phase III trial showed a significantly better 3-year OS with S-1 for 1 year than with observation in D2-resection stage II or III GC patients [42] [IB]. In the update after 5 years of follow-up, the benefit was maintained in the S-1 group [43]. In the same type of patients, the XELOX regimen was superior to surgery alone in the CLASSIC phase III trial. With a median 5-year follow-up, the estimated 5-year DFS was 68 versus 53 % and the estimated 5-year OS was 78 versus 69 % in the XELOX and the surgery-only groups, respectively [44].
Adjuvant chemoradiotherapy vs chemotherapy
The ARTIST trial compared CRT [cisplatin and capecitabine (XP), with capecitabine and radiation concurrently] versus chemotherapy alone (XP × 6 cycles) in patients with at least D2 lymphadenectomy and R0 resection [45]. With a 7-year follow-up, the DFS (HR 0.74; p = 0.092) and the OS (HR 1.13; p = 0.527) were similar between the groups. Subgroup analyses showed that CRT significantly improved DFS in node-positive disease and with intestinal-type GC [46]. Finally, the Korean ARTIST II randomized phase III trial is currently comparing adjuvant S-1 versus S-1 plus oxaliplatin (SOX), with or without radiotherapy.

Advanced disease

First-line treatment

Several randomized studies and a meta-analysis [47], comparing palliative CT with the best supportive care (BSC), have demonstrated that CT increases median overall survival and improves the quality of life of patients with advanced gastric cancer [I,A].
Cisplatin-based chemotherapy
In the late nineties, it was proven that regimes based on cisplatin were superior to other older regimes without cisplatin. Both CF (cisplatin–5FU) and ECF (epirubicin–cisplatin–5FU) can be considered standard combinations (IA). Although a meta-analysis suggests better OS with ECF, there are no randomized head-to-head comparison studies
Chemotherapy with docetaxel
In the Phase III study V-325 [48], docetaxel, cisplatin, and 5FU (DCF) was compared with CF. DCF was superior in TTP, OS, and response rate but was also more toxic. Triplets with Docetaxel can be considered in selected patients (IB).
Oxaliplatin-based chemotherapy
Two phase III studies [49, 50] have suggested that oxaliplatin has similar efficacy and less toxicity than cisplatin and can replace it in this setting (IA).
Oral fluoropyrimidines
Two phase III studies [49, 51] have observed no inferiority in efficacy and a more favorable toxic profile, when replacing 5FU with capecitabine. Another phase III study observed non-inferiority in efficacy and better convenience and toxicity with cisplatin-S1 versus CF [52]. Based on these data, oral fluoropyrimidines (capecitabine or S-1) can replace 5FU in this setting [I,A].
Irinotecan combinations
Irinotecan/5FU/LV combinations (FOLFIRI, IF) have been compared with CF in two randomized trials [53, 54] showing similar efficacy and better tolerance and can be considered adequate options for these patients (I,B).
Trastuzumab
The phase III TOGA [55] analyzed HER-2 status in 3807 patients. Twenty-two per cent were HER-2+ (594) and were randomized to receive chemotherapy alone (CX or CF) or chemo plus trastuzumab. In the trastuzumab arm, it was shown a statistically significant increase in the primary endpoint, which was OS as well as in PFS. A preplanned analysis of OS in the subgroup of patients with IHC 3+ or IHC 2+ with FISH+ showed an increase of 5 months OS in the trastuzumab arm (16.8 versus 11.8 months, HR 0.65). According to these results, we can consider that trastuzumab added to cisplatin + fluoropyrimidines can be considered the standard treatment in patients with gastric or EGJ advanced HER-2+ adenocarcinoma [IB].

Second-line chemotherapy

Less than 60 % of patients receive second- or third-line therapy for gastric cancer in clinical practice [55]. There is evidence that second-line chemotherapy achieves improvement in quality of life (QOL) and provides a median overall survival (OS) of 4–6 months [I,A]. Docetaxel, paclitaxel, irinotecan, and a targeted therapy against vascular endothelial grow factor receptor 2 (VEGFR2) with ramucirumab have demonstrated a significant benefit in OS in phase III trials [I,B]. Two recent meta-analysis have shown a significant reduction in the risk of death [56, 57] [I,A]. The reduction was HR = 0.55 for irinotecan, and HR = 0.71 for docetaxel [57].
Irinotecan The first study randomized 40 patients between irinotecan and best supportive care (BSC). Irinotecan showed a statistically significant survival benefit over BSC (median OS 4 versus 2.4 months, respectively) [I,B] [58].
A Korean trial randomized patients to BSC, to 3-weekly docetaxel or 2-weekly irinotecan. Chemotherapy showed a significant survival benefit (median OS 5.3 versus 3.8 months, HR = 0.65) over BSC alone [I,B] [59].
Docetaxel The UK COUGAR-02 trial randomized 168 patients to 3-weekly docetaxel versus BSC. Docetaxel improved median overall survival over BSC (5.2 months versus 3.6 months, respectively, HR = 0.67) [I,B]. Global quality of life (QOL) scores were similar between the two arms [60].
Paclitaxel A phase III trial with 219 Asian patients did not show a superiority of irinotecan against paclitaxel [I,B] (median OS 8.4 versus 9.5 months, respectively, HR = 1.14) [61].
Ramucirumab has shown a statistically significant efficacy as monotherapy (REGARD) or in combination with paclitaxel (RAINBOW). REGARD randomized 445 patients to ramucirumab versus placebo. Ramucirumab showed a significant OS benefit (5.2 months versus 3.8 months, HR = 0.77) over placebo [I,B] [62].
RAINBOW randomized 665 patients to ramucirumab plus paclitaxel or to paclitaxel plus placebo. Ramucirumab plus paclitaxel arm showed a significantly superior OS (9.6 months versus 7.3 months, HR = 0.80) over paclitaxel monotherapy [I,B] [63].

Future lines and targeted drugs

Since the approval of Trastuzumab in advanced HER2 positive gastric cancer, in 2010, there have been an increasing number of molecules and targets entering the preclinical and clinical research programs in GC.
New strategies underway to improve the results of trastuzumab in HER2-positive GC include the use of antibody–drug conjugates to deliver cytotoxic agents such trastuzumab emtansine (T-DM1) [64] or its combination with new monoclonal antibodies (MoAbs) that targets a different extracellular dimerization domain (Pertuzumab) [65]. Antiangiogenic therapy with Ramucirumab, a fully human immunoglobulin (Ig) G1 monoclonal antibody targeting VEGFR-2, has demonstrated improved survival both as monotherapy and in combination with paclitaxel [62, 63] in the second-line setting. By the contrary, 2 MoAbs targeting MET were unsuccessfully tested in this disease [66, 67].
Two recently communicated clinical trials have demonstrated some early signs of activity of both antiPD-1 and antiPD-L1 antibody immune checkpoint inhibitors in upper GI malignancies [68, 69].
Follow-up In the setting of operable gastric cancer, a regular follow-up may allow treatment of symptoms and early detection of recurrence, though there is no evidence that it improves survival outcomes [III, B]. If relapse/disease progression is suspected, then physical examination, blood tests, and radiological investigations or endoscopy should be carried out. We recommend anamnesis and physical examination every 3–6 months in the first 3 years following surgical intervention and then every 6–12 months during years 4 and 5 [V]. The benefit of following up the patients beyond year 5 is controversial [70].

Compliance with ethical standards

Conflict of interest

The authors declare that they have no conflict of interest.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Crew KD, Neugut AI. Epidemiology of upper gastrointestinal malignancies. Semin Oncol. 2004;31:450–64.CrossRefPubMed Crew KD, Neugut AI. Epidemiology of upper gastrointestinal malignancies. Semin Oncol. 2004;31:450–64.CrossRefPubMed
3.
Zurück zum Zitat Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.CrossRefPubMed Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.CrossRefPubMed
4.
Zurück zum Zitat Edge SB, Byrd DR, Compton CC, Frilz AG, Greene FL, Trotti A, editors. AJCC Cancer staging Manual. 7th ed. New York: Springer; 2010. Edge SB, Byrd DR, Compton CC, Frilz AG, Greene FL, Trotti A, editors. AJCC Cancer staging Manual. 7th ed. New York: Springer; 2010.
5.
Zurück zum Zitat Siewert JR, Stein HJ. Classification of adenocarcinoma of the oesophagogastric junction. Br J Surg. 1998;85:1457–9.CrossRefPubMed Siewert JR, Stein HJ. Classification of adenocarcinoma of the oesophagogastric junction. Br J Surg. 1998;85:1457–9.CrossRefPubMed
6.
Zurück zum Zitat Colquhoun A, Arnold M, Ferlay J, Goodman KJ, Forman D, Soerjomataram I. Global patterns of cardia and non-cardia gastric cancer incidence in 2012. Gut. 2015; 0:1–8. Colquhoun A, Arnold M, Ferlay J, Goodman KJ, Forman D, Soerjomataram I. Global patterns of cardia and non-cardia gastric cancer incidence in 2012. Gut. 2015; 0:1–8.
7.
Zurück zum Zitat Shah MA, Khanin R, Tang L, Janjigian YY, Klimstra DS, Gerdes H, et al. Molecular classification of gastric cancer: a new paradigm. Clin Cancer Res. 2011;17:2693–701.PubMedCentralCrossRefPubMed Shah MA, Khanin R, Tang L, Janjigian YY, Klimstra DS, Gerdes H, et al. Molecular classification of gastric cancer: a new paradigm. Clin Cancer Res. 2011;17:2693–701.PubMedCentralCrossRefPubMed
8.
Zurück zum Zitat Tan IB, Ivanova T, Lim KH, Ong CW, Deng N, Lee J, et al. Intrinsic subtypes of gastric cancer, based on gene expression pattern, predict survival and respond differently to chemotherapy. Gastroenterology. 2011;141:476–85 (485 e1-11). Tan IB, Ivanova T, Lim KH, Ong CW, Deng N, Lee J, et al. Intrinsic subtypes of gastric cancer, based on gene expression pattern, predict survival and respond differently to chemotherapy. Gastroenterology. 2011;141:476–85 (485 e1-11).
9.
Zurück zum Zitat Lei Z, Tan IB, Das K, Deng N, Zouridis H, Pattison S, et al. Identification of molecular subtypes of gastric cancer with different responses to PI3-kinase inhibitors and 5-fluorouracil. Gastroenterology. 2013;145:554–65.CrossRefPubMed Lei Z, Tan IB, Das K, Deng N, Zouridis H, Pattison S, et al. Identification of molecular subtypes of gastric cancer with different responses to PI3-kinase inhibitors and 5-fluorouracil. Gastroenterology. 2013;145:554–65.CrossRefPubMed
10.
Zurück zum Zitat Deng N, Goh LK, Wang H, Das K, Tao J, Tan IB, et al. A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets. Gut. 2012;61:673–84.PubMedCentralCrossRefPubMed Deng N, Goh LK, Wang H, Das K, Tao J, Tan IB, et al. A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets. Gut. 2012;61:673–84.PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.CrossRef Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.CrossRef
12.
Zurück zum Zitat Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449–56.CrossRefPubMed Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449–56.CrossRefPubMed
13.
Zurück zum Zitat Brettingham-Moore KH, Duong CP, Heriot AG, Thomas RJ, Phillips WA. Using gene expression profiling to predict response and prognosis in gastrointestinal cancers-the promise and the perils. Ann Surg Oncol. 2011;18:1484–91.CrossRefPubMed Brettingham-Moore KH, Duong CP, Heriot AG, Thomas RJ, Phillips WA. Using gene expression profiling to predict response and prognosis in gastrointestinal cancers-the promise and the perils. Ann Surg Oncol. 2011;18:1484–91.CrossRefPubMed
14.
Zurück zum Zitat Wang J, Yu JC, Kang WM, Ma ZQ. Prognostic significance of intraoperative chemotherapy and extensive lymphadenectomy in patients with node-negative gastric cancer. J Surg Oncol. 2012;105:400–4.CrossRefPubMed Wang J, Yu JC, Kang WM, Ma ZQ. Prognostic significance of intraoperative chemotherapy and extensive lymphadenectomy in patients with node-negative gastric cancer. J Surg Oncol. 2012;105:400–4.CrossRefPubMed
15.
Zurück zum Zitat Salem A, Hashem S, Mula-Hussain LY, Mohammed I, Nour A, Shelpai W, et al. Management strategies for locoregional recurrence in early-stage gastric cancer: retrospective analysis and comprehensive literature review. J Gastrointest Cancer. 2012;43:77–82.CrossRefPubMed Salem A, Hashem S, Mula-Hussain LY, Mohammed I, Nour A, Shelpai W, et al. Management strategies for locoregional recurrence in early-stage gastric cancer: retrospective analysis and comprehensive literature review. J Gastrointest Cancer. 2012;43:77–82.CrossRefPubMed
16.
Zurück zum Zitat Fujiwara M, Kodera Y, Misawa K, Kinoshita M, Kinoshita T, Miura S, et al. Longterm outcomes of early-stage gastric carcinoma patients treated with laparoscopy-assisted surgery. J Am Coll Surg. 2008;206:138–43.CrossRefPubMed Fujiwara M, Kodera Y, Misawa K, Kinoshita M, Kinoshita T, Miura S, et al. Longterm outcomes of early-stage gastric carcinoma patients treated with laparoscopy-assisted surgery. J Am Coll Surg. 2008;206:138–43.CrossRefPubMed
17.
Zurück zum Zitat Yamada Y, Arao T, Gotoda T, Taniguchi H, Oda I, Shirao K, et al. Identification of prognostic biomarkers in gastric cancer using endoscopic biopsy samples. Cancer Sci. 2008;99:2193–9.CrossRefPubMed Yamada Y, Arao T, Gotoda T, Taniguchi H, Oda I, Shirao K, et al. Identification of prognostic biomarkers in gastric cancer using endoscopic biopsy samples. Cancer Sci. 2008;99:2193–9.CrossRefPubMed
18.
Zurück zum Zitat Lo Nigro C, Monteverde M, Riba M, Lattanzio L, Tonissi F, Garrone O, et al. Expression profiling and long lasting responses to chemotherapy in metastatic gastric cancer. Int J Oncol. 2010;37:1219–28. Lo Nigro C, Monteverde M, Riba M, Lattanzio L, Tonissi F, Garrone O, et al. Expression profiling and long lasting responses to chemotherapy in metastatic gastric cancer. Int J Oncol. 2010;37:1219–28.
19.
Zurück zum Zitat Wang Z, Yan Z, Zhang B, Rao Z, Zhang Y, Liu J, et al. Identification of a 5-gene signature for clinical and prognostic prediction in gastric cancer patients upon microarray data. Med Oncol. 2013;30:678.CrossRefPubMed Wang Z, Yan Z, Zhang B, Rao Z, Zhang Y, Liu J, et al. Identification of a 5-gene signature for clinical and prognostic prediction in gastric cancer patients upon microarray data. Med Oncol. 2013;30:678.CrossRefPubMed
20.
Zurück zum Zitat Takeno A, Takemasa I, Seno S, Yamasaki M, Motoori M, Miyata H, et al. Gene expression profile prospectively predicts peritoneal relapse after curative surgery of gastric cancer. Ann Surg Oncol. 2010;17:1033–42.CrossRefPubMed Takeno A, Takemasa I, Seno S, Yamasaki M, Motoori M, Miyata H, et al. Gene expression profile prospectively predicts peritoneal relapse after curative surgery of gastric cancer. Ann Surg Oncol. 2010;17:1033–42.CrossRefPubMed
21.
Zurück zum Zitat Strong VE, D’Amico TA, Kleinberg L, Ajani J. Impact of the 7th Edition AJCC staging classification on the NCCN clinical practice guidelines in oncology for gastric and esophageal cancers. J Natl Compr Canc Netw. 2013;11:60–6.PubMed Strong VE, D’Amico TA, Kleinberg L, Ajani J. Impact of the 7th Edition AJCC staging classification on the NCCN clinical practice guidelines in oncology for gastric and esophageal cancers. J Natl Compr Canc Netw. 2013;11:60–6.PubMed
22.
Zurück zum Zitat Gomez-Martin C, Concha A, Corominas JM, García-Caballero T, García-García E, Iglesias M, et al. Consensus of the Spanish Society of Medical Oncology (SEOM) and Spanish Society of Pathology (SEAP) for HER2 testing in gastric carcinoma. Clin Trans Oncol. 2011;13:636–51.CrossRef Gomez-Martin C, Concha A, Corominas JM, García-Caballero T, García-García E, Iglesias M, et al. Consensus of the Spanish Society of Medical Oncology (SEOM) and Spanish Society of Pathology (SEAP) for HER2 testing in gastric carcinoma. Clin Trans Oncol. 2011;13:636–51.CrossRef
23.
Zurück zum Zitat Mocellin S, Pasquali S. Diagnostic accuracy of endoscopic ultrasonography (EUS) for the preoperative locoregional staging of primary gastric cancer. Cochrane Database Syst Rev.2015;2:CD009944. Mocellin S, Pasquali S. Diagnostic accuracy of endoscopic ultrasonography (EUS) for the preoperative locoregional staging of primary gastric cancer. Cochrane Database Syst Rev.2015;2:CD009944.
24.
Zurück zum Zitat De Andrade JP, Mezhir JJ. The critical role of peritoneal cytology in the staging of gastric cancer: an evidence-based review. J Surg Oncol. 2014;110:291–7.CrossRefPubMed De Andrade JP, Mezhir JJ. The critical role of peritoneal cytology in the staging of gastric cancer: an evidence-based review. J Surg Oncol. 2014;110:291–7.CrossRefPubMed
25.
Zurück zum Zitat Kaneko Y, Murray WK, Link E, Hicks RJ, Duong C. Improving patient selection for 18F-FDG PET scanning in the staging of gastric cancer. J Nucl Med. 2015;56:523–9.CrossRefPubMed Kaneko Y, Murray WK, Link E, Hicks RJ, Duong C. Improving patient selection for 18F-FDG PET scanning in the staging of gastric cancer. J Nucl Med. 2015;56:523–9.CrossRefPubMed
26.
Zurück zum Zitat Shimada H, Noie T, Ohashi M, Oba K, Takahashi Y. Clinical significance of serum tumor markers for gastric cancer: a systematic review of literature by the Task Force of the Japanese Gastric Cancer Association. Gastric Cancer. 2014;17:26–33.CrossRefPubMed Shimada H, Noie T, Ohashi M, Oba K, Takahashi Y. Clinical significance of serum tumor markers for gastric cancer: a systematic review of literature by the Task Force of the Japanese Gastric Cancer Association. Gastric Cancer. 2014;17:26–33.CrossRefPubMed
27.
Zurück zum Zitat Gotoda T, Iwasaki M, Kusano C, Seewald S, Oda I. Endoscopic resection of early gastric cancer treated by guideline and expanded National Cancer Centre criteria. Br J Surg. 2010;97:868–71.CrossRefPubMed Gotoda T, Iwasaki M, Kusano C, Seewald S, Oda I. Endoscopic resection of early gastric cancer treated by guideline and expanded National Cancer Centre criteria. Br J Surg. 2010;97:868–71.CrossRefPubMed
28.
29.
Zurück zum Zitat Songun I, Putter H, Kranenbarg EM, Sasako M, van de Velde CJ. Surgical treatment of gastric cancer: 15-year follow-up results of the randomised nationwide Dutch D1D2 trial. Lancet Oncol. 2010;11:439–49.CrossRefPubMed Songun I, Putter H, Kranenbarg EM, Sasako M, van de Velde CJ. Surgical treatment of gastric cancer: 15-year follow-up results of the randomised nationwide Dutch D1D2 trial. Lancet Oncol. 2010;11:439–49.CrossRefPubMed
30.
Zurück zum Zitat Jiang L, Yang KH, Guan QL, Zhao P, Chen Y, Tian JH. Survival and recurrence free benefits with different lymphadenectomy for resectable gastric cancer: a meta-analysis. J Surg Oncol. 2013;107:807–14.CrossRefPubMed Jiang L, Yang KH, Guan QL, Zhao P, Chen Y, Tian JH. Survival and recurrence free benefits with different lymphadenectomy for resectable gastric cancer: a meta-analysis. J Surg Oncol. 2013;107:807–14.CrossRefPubMed
31.
Zurück zum Zitat Schuhmacher C, Gretschel S, Lordick F, Reichardt P, Hohenberger W, Eisenberger CF, et al. Neoadjuvant chemotherapy compared with surgery alone for locally advanced cancer of the stomach and cardia: European Organisation for Research and Treatment of Cancer randomized trial 40954. J Clin Oncol. 2010;28:5210–8.PubMedCentralCrossRefPubMed Schuhmacher C, Gretschel S, Lordick F, Reichardt P, Hohenberger W, Eisenberger CF, et al. Neoadjuvant chemotherapy compared with surgery alone for locally advanced cancer of the stomach and cardia: European Organisation for Research and Treatment of Cancer randomized trial 40954. J Clin Oncol. 2010;28:5210–8.PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Sjoquist KM, Burmeister BH, Smithers BM, Zalcberg JR, Simes RJ, Barbour A, et al. Survival after neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal carcinoma: an updated meta-analysis. Lancet Oncol. 2011;12:681–92.CrossRefPubMed Sjoquist KM, Burmeister BH, Smithers BM, Zalcberg JR, Simes RJ, Barbour A, et al. Survival after neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal carcinoma: an updated meta-analysis. Lancet Oncol. 2011;12:681–92.CrossRefPubMed
33.
Zurück zum Zitat Stahl M, Walz MK, Stuschke M, Lehmann N, Meyer HJ, Riera-Knorrenschild J, 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;27:851–6.CrossRefPubMed Stahl M, Walz MK, Stuschke M, Lehmann N, Meyer HJ, Riera-Knorrenschild J, 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;27:851–6.CrossRefPubMed
34.
Zurück zum Zitat Shapiro J, van Lanschot JJ, Hulshof MC, van Hagen P, van Berge Henegouwen MI, Wijnhoven BP, et al. Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or junctional cancer (CROSS): long-term results of a randomised controlled trial. Lancet Oncol. 2015;16:1090–8.CrossRefPubMed Shapiro J, van Lanschot JJ, Hulshof MC, van Hagen P, van Berge Henegouwen MI, Wijnhoven BP, et al. Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or junctional cancer (CROSS): long-term results of a randomised controlled trial. Lancet Oncol. 2015;16:1090–8.CrossRefPubMed
35.
Zurück zum Zitat Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11–20.CrossRefPubMed Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11–20.CrossRefPubMed
36.
Zurück zum Zitat Ychou M, Boige V, Pignon JP, Conroy T, Bouché O, Lebreton G, et al. Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol. 2011;29:1715–21.CrossRefPubMed Ychou M, Boige V, Pignon JP, Conroy T, Bouché O, Lebreton G, et al. Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol. 2011;29:1715–21.CrossRefPubMed
37.
Zurück zum Zitat Ronellenfitsch U, Schwarzbach M, Hofheinz R, Kienle P, Kieser M, Slanger TE, et al. Preoperative chemo(radio)therapy versus primary surgery for gastroesophageal adenocarcinoma: systematic review with meta-analysis combining individual patient and aggregate data. Eur J Cancer. 2013;49:3149–58.CrossRefPubMed Ronellenfitsch U, Schwarzbach M, Hofheinz R, Kienle P, Kieser M, Slanger TE, et al. Preoperative chemo(radio)therapy versus primary surgery for gastroesophageal adenocarcinoma: systematic review with meta-analysis combining individual patient and aggregate data. Eur J Cancer. 2013;49:3149–58.CrossRefPubMed
38.
Zurück zum Zitat Cunningham D, Smyth E, Stenning S. Perioperative chemotherapy- bevacizumab for resectable gastro-esophageal adenocarcinoma: Results from the UK Medical Research Council randomised T03 trial (ISRCTN 46020948). Eur J Cancer. 2015;51 (supplS3:Abstract2201). Cunningham D, Smyth E, Stenning S. Perioperative chemotherapy- bevacizumab for resectable gastro-esophageal adenocarcinoma: Results from the UK Medical Research Council randomised T03 trial (ISRCTN 46020948). Eur J Cancer. 2015;51 (supplS3:Abstract2201).
39.
Zurück zum Zitat Macdonald JS, Smalley SR, Benedetti J, Hundahl SA, Estes NC, Stemmermann GN, et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med. 2001;345:725–30.CrossRefPubMed Macdonald JS, Smalley SR, Benedetti J, Hundahl SA, Estes NC, Stemmermann GN, et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med. 2001;345:725–30.CrossRefPubMed
40.
Zurück zum Zitat Smalley SR, Benedetti JK, Haller DG, Hundahl SA, Estes NC, Ajani JA, et al. Updated analysis of SWOG-directed intergroup study 0116: a phase III trial of adjuvant radiochemotherapy versus observation after curative gastric cancer resection. J Clin Oncol. 2012;30:2327–33.PubMedCentralCrossRefPubMed Smalley SR, Benedetti JK, Haller DG, Hundahl SA, Estes NC, Ajani JA, et al. Updated analysis of SWOG-directed intergroup study 0116: a phase III trial of adjuvant radiochemotherapy versus observation after curative gastric cancer resection. J Clin Oncol. 2012;30:2327–33.PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Group G, Paoletti X, Oba K, Michiels S, Ohashi Y, Pignon JP, et al. Benefit of adjuvant chemotherapy for resectable gastric cancer: a meta-analysis. JAMA. 2010;303:1729–37.CrossRef Group G, Paoletti X, Oba K, Michiels S, Ohashi Y, Pignon JP, et al. Benefit of adjuvant chemotherapy for resectable gastric cancer: a meta-analysis. JAMA. 2010;303:1729–37.CrossRef
42.
Zurück zum Zitat Sakuramoto S, Sasako M, Yamaguchi T, Kinoshita T, Fujii M, Nashimoto A, et al. Adjuvant chemotherapy for gastric cancer with S-1, an oral fluoropyrimidine. N Engl J Med. 2007;357:1810–20.CrossRefPubMed Sakuramoto S, Sasako M, Yamaguchi T, Kinoshita T, Fujii M, Nashimoto A, et al. Adjuvant chemotherapy for gastric cancer with S-1, an oral fluoropyrimidine. N Engl J Med. 2007;357:1810–20.CrossRefPubMed
43.
Zurück zum Zitat Sasako M, Sakuramoto S, Katai H, Kinoshita T, Furukawa H, Yamaguchi T, et al. Five-year outcomes of a randomized phase III trial comparing adjuvant chemotherapy with S-1 versus surgery alone in stage II or III gastric cancer. J Clin Oncol. 2011;29:4387–93.CrossRefPubMed Sasako M, Sakuramoto S, Katai H, Kinoshita T, Furukawa H, Yamaguchi T, et al. Five-year outcomes of a randomized phase III trial comparing adjuvant chemotherapy with S-1 versus surgery alone in stage II or III gastric cancer. J Clin Oncol. 2011;29:4387–93.CrossRefPubMed
44.
Zurück zum Zitat Noh SH, Park SR, Yang HK, Chung HC, Chung IJ, Kim SW, et al. Adjuvant capecitabine plus oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): 5-year follow-up of an open-label, randomised phase 3 trial. Lancet Oncol. 2014;15:1389–96.CrossRefPubMed Noh SH, Park SR, Yang HK, Chung HC, Chung IJ, Kim SW, et al. Adjuvant capecitabine plus oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): 5-year follow-up of an open-label, randomised phase 3 trial. Lancet Oncol. 2014;15:1389–96.CrossRefPubMed
45.
Zurück zum Zitat Lee J, Lim do H, Kim S, Park SH, Park JO, Park YS, 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;30:268–73.CrossRefPubMed Lee J, Lim do H, Kim S, Park SH, Park JO, Park YS, 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;30:268–73.CrossRefPubMed
46.
Zurück zum Zitat Park SH, Sohn TS, Lee J, Lim do H, Hong ME, Kim KM, et al. Phase III trial to compare adjuvant chemotherapy with capecitabine and cisplatin versus concurrent chemoradiotherapy in gastric cancer: final report of the adjuvant chemoradiotherapy in stomach tumors trial, including survival and subset analyses. J Clin Oncol. 2015;33:3130–6.CrossRefPubMed Park SH, Sohn TS, Lee J, Lim do H, Hong ME, Kim KM, et al. Phase III trial to compare adjuvant chemotherapy with capecitabine and cisplatin versus concurrent chemoradiotherapy in gastric cancer: final report of the adjuvant chemoradiotherapy in stomach tumors trial, including survival and subset analyses. J Clin Oncol. 2015;33:3130–6.CrossRefPubMed
47.
Zurück zum Zitat Wagner AD, Grothe W, Haerting J, Kleber G, Grothey A, Fleig WE. Chemotherapy in advanced gastric cancer: a systematic review and meta-analysis based on aggregate data. J Clin Oncol. 2006;24:2903–9.CrossRefPubMed Wagner AD, Grothe W, Haerting J, Kleber G, Grothey A, Fleig WE. Chemotherapy in advanced gastric cancer: a systematic review and meta-analysis based on aggregate data. J Clin Oncol. 2006;24:2903–9.CrossRefPubMed
48.
Zurück zum Zitat Van Cutsem E, Moiseyenko VM, Tjulandin S, Majlis A, Constenla M, Boni C, 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;24:4991–7.CrossRefPubMed Van Cutsem E, Moiseyenko VM, Tjulandin S, Majlis A, Constenla M, Boni C, 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;24:4991–7.CrossRefPubMed
49.
Zurück zum Zitat Cunningham D, Starling N, Rao S, Iveson T, Nicolson M, Coxon F, et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36–46.CrossRefPubMed Cunningham D, Starling N, Rao S, Iveson T, Nicolson M, Coxon F, et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36–46.CrossRefPubMed
50.
Zurück zum Zitat Al-Batran SE, Hartmann JT, Probst S, Schmalenberg H, Hollerbach S, Hofheinz R, 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;26:1435–42.CrossRefPubMed Al-Batran SE, Hartmann JT, Probst S, Schmalenberg H, Hollerbach S, Hofheinz R, 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;26:1435–42.CrossRefPubMed
51.
Zurück zum Zitat Kang YK, Kang WK, Shin DB, Chen J, Xiong J, Wang J, 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;20:666–73.CrossRefPubMed Kang YK, Kang WK, Shin DB, Chen J, Xiong J, Wang J, 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;20:666–73.CrossRefPubMed
52.
Zurück zum Zitat Ajani JA, Rodriguez W, Bodoky G, Moiseyenko V, Lichinitser M, Gorbunova V, 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;28:1547–53.CrossRefPubMed Ajani JA, Rodriguez W, Bodoky G, Moiseyenko V, Lichinitser M, Gorbunova V, 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;28:1547–53.CrossRefPubMed
53.
Zurück zum Zitat Dank M, Zaluski J, Barone C, Valvere V, Yalcin S, Peschel C, et al. Randomized phase III study comparing irinotecan combined with 5-fluorouracil and folinic acid to cisplatin combined with 5-fluorouracil in chemotherapy naive patients with advanced adenocarcinoma of the stomach or esophagogastric junction. Ann Oncol. 2008;19:1450–7.CrossRefPubMed Dank M, Zaluski J, Barone C, Valvere V, Yalcin S, Peschel C, et al. Randomized phase III study comparing irinotecan combined with 5-fluorouracil and folinic acid to cisplatin combined with 5-fluorouracil in chemotherapy naive patients with advanced adenocarcinoma of the stomach or esophagogastric junction. Ann Oncol. 2008;19:1450–7.CrossRefPubMed
54.
Zurück zum Zitat Guimbaud R, Louvet C, Ries P, Ychou M, Maillard E, André T, et al. Prospective, randomized, multicenter, phase III study of fluorouracil, leucovorin, and irinotecan versus epirubicin, cisplatin, and capecitabine in advanced gastric adenocarcinoma: a French intergroup (Federation Francophone de Cancerologie Digestive, Federation Nationale des Centres de Lutte Contre le Cancer, and Groupe Cooperateur Multidisciplinaire en Oncologie) study. J Clin Oncol. 2014;32:3520–6.CrossRefPubMed Guimbaud R, Louvet C, Ries P, Ychou M, Maillard E, André T, et al. Prospective, randomized, multicenter, phase III study of fluorouracil, leucovorin, and irinotecan versus epirubicin, cisplatin, and capecitabine in advanced gastric adenocarcinoma: a French intergroup (Federation Francophone de Cancerologie Digestive, Federation Nationale des Centres de Lutte Contre le Cancer, and Groupe Cooperateur Multidisciplinaire en Oncologie) study. J Clin Oncol. 2014;32:3520–6.CrossRefPubMed
55.
Zurück zum Zitat Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, 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;376:687–97.CrossRefPubMed Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, 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;376:687–97.CrossRefPubMed
56.
Zurück zum Zitat Iacovelli R, Pietrantonio F, Farcomeni A, et al. Chemotherapy or targeted therapy as second-line treatment of advanced gastric cancer. A systematic review and meta-analysis of published studies. PLoS One. 2014;9:e108940.PubMedCentralCrossRefPubMed Iacovelli R, Pietrantonio F, Farcomeni A, et al. Chemotherapy or targeted therapy as second-line treatment of advanced gastric cancer. A systematic review and meta-analysis of published studies. PLoS One. 2014;9:e108940.PubMedCentralCrossRefPubMed
57.
Zurück zum Zitat Kim HS, Kim HJ, Kim SY, et al. Second-line chemotherapy versus supportive cancer treatment in advanced gastric cancer: a meta-analysis. Ann Oncol. 2013;24:2850–4.CrossRefPubMed Kim HS, Kim HJ, Kim SY, et al. Second-line chemotherapy versus supportive cancer treatment in advanced gastric cancer: a meta-analysis. Ann Oncol. 2013;24:2850–4.CrossRefPubMed
58.
Zurück zum Zitat Thuss-Patience PC, Kretzschmar A, Bichev D, 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;47:2306–14.CrossRefPubMed Thuss-Patience PC, Kretzschmar A, Bichev D, 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;47:2306–14.CrossRefPubMed
59.
Zurück zum Zitat Kang JH, Lee SI, Lim do H et al. Salvage chemotherapy for pretreated gastric cancer: a randomized phase III trial comparing chemotherapy plus best supportive care with best supportive care alone. J Clin Oncol. 2012;30:1513–8. Kang JH, Lee SI, Lim do H et al. Salvage chemotherapy for pretreated gastric cancer: a randomized phase III trial comparing chemotherapy plus best supportive care with best supportive care alone. J Clin Oncol. 2012;30:1513–8.
60.
Zurück zum Zitat Ford HE, Marshall A, Bridgewater JA, 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.CrossRefPubMed Ford HE, Marshall A, Bridgewater JA, 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.CrossRefPubMed
61.
Zurück zum Zitat Hironaka S, Ueda S, Yasui H, 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;31:4438–44.CrossRefPubMed Hironaka S, Ueda S, Yasui H, 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;31:4438–44.CrossRefPubMed
62.
Zurück zum Zitat Fuchs CS, Tomasek J, Yong CJ, 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;383:31–9.CrossRefPubMed Fuchs CS, Tomasek J, Yong CJ, 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;383:31–9.CrossRefPubMed
63.
Zurück zum Zitat Wilke H, Muro K, Van Cutsem E, 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;15:1224–35.CrossRefPubMed Wilke H, Muro K, Van Cutsem E, 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;15:1224–35.CrossRefPubMed
64.
Zurück zum Zitat TDM1 vs taxane in Previously Treated Advanced HER2+ Gastric Cancer study. In NCT06419391. TDM1 vs taxane in Previously Treated Advanced HER2+ Gastric Cancer study. In NCT06419391.
65.
Zurück zum Zitat JACOB Study Pertuzumab With Trastuzumab and Chemotherapy in Patients With HER2+ Metastasic Gastric and Gastroesophageal Cancer In NCT01774786. JACOB Study Pertuzumab With Trastuzumab and Chemotherapy in Patients With HER2+ Metastasic Gastric and Gastroesophageal Cancer In NCT01774786.
66.
Zurück zum Zitat Cunningham D TN, Davidenko I. Phase III randomized double-blind multicenter, placebo cotrolled tiral of Rilotumumab plus epirrubicin, Cisplatin and Capecitibine (ECX) as first line therapy in patients with advanced MET–positive gastric or gastroesophageal Junction Cancer: RILOMET 1 Study. J Clin Oncol. 2015;33(suppl; abstr 4000). Cunningham D TN, Davidenko I. Phase III randomized double-blind multicenter, placebo cotrolled tiral of Rilotumumab plus epirrubicin, Cisplatin and Capecitibine (ECX) as first line therapy in patients with advanced MET–positive gastric or gastroesophageal Junction Cancer: RILOMET 1 Study. J Clin Oncol. 2015;33(suppl; abstr 4000).
67.
Zurück zum Zitat Shah MCJ, BEeIT. Randomized Phase II Study of FOLFOX +/- MET inhibitor, Ornatuzumab in advanced gastroesophageal adenocarcinoma (GEC). J Clin Oncol. 2015;33(suppl 3; abstr 2). Shah MCJ, BEeIT. Randomized Phase II Study of FOLFOX +/- MET inhibitor, Ornatuzumab in advanced gastroesophageal adenocarcinoma (GEC). J Clin Oncol. 2015;33(suppl 3; abstr 2).
68.
Zurück zum Zitat Segal NHAS, Brahmer JR. Preliminary Data from a multi arm expansion study of MEDI 4736, an anti PDL1 antibody. J CLin Oncol. 2014;32(15 suppl)134136–144. Segal NHAS, Brahmer JR. Preliminary Data from a multi arm expansion study of MEDI 4736, an anti PDL1 antibody. J CLin Oncol. 2014;32(15 suppl)134136–144.
69.
Zurück zum Zitat Muro K, Bang YJ, Shankaran V, et al. Relationship between PDL1 expression and clinical outcomes in patients with advanced gastric cancer treated with the anti PDL1 monoclonal antibody Pembrolizumab in KEYNOTE-012. J Clin Oncol. 33(Suppl 3; abstr 3). Muro K, Bang YJ, Shankaran V, et al. Relationship between PDL1 expression and clinical outcomes in patients with advanced gastric cancer treated with the anti PDL1 monoclonal antibody Pembrolizumab in KEYNOTE-012. J Clin Oncol. 33(Suppl 3; abstr 3).
70.
Zurück zum Zitat IGCC Working Group. The charter Scaligero on gastric cancer: follow up after gastrectomy for cancer. In: 10th international gastric cancer congress (IGCC) 2013. IGCC Working Group. The charter Scaligero on gastric cancer: follow up after gastrectomy for cancer. In: 10th international gastric cancer congress (IGCC) 2013.
Metadaten
Titel
Seom guidelines for the treatment of gastric cancer 2015
verfasst von
M. Martin-Richard
A. Custodio
C. García-Girón
C. Grávalos
C. Gomez
P. Jimenez-Fonseca
J. L. Manzano
C. Pericay
F. Rivera
A. Carrato
Publikationsdatum
01.12.2015
Verlag
Springer Milan
Erschienen in
Clinical and Translational Oncology / Ausgabe 12/2015
Print ISSN: 1699-048X
Elektronische ISSN: 1699-3055
DOI
https://doi.org/10.1007/s12094-015-1456-y

Weitere Artikel der Ausgabe 12/2015

Clinical and Translational Oncology 12/2015 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.