Skip to main content
Erschienen in: Gastric Cancer 4/2020

Open Access 01.07.2020 | Original Article

Increased assessment of HER2 in metastatic gastroesophageal cancer patients: a nationwide population-based cohort study

verfasst von: Willemieke P. M. Dijksterhuis, Rob H. A. Verhoeven, Sybren L. Meijer, Marije Slingerland, Nadia Haj Mohammad, Judith de Vos-Geelen, Laurens V. Beerepoot, Theo van Voorthuizen, Geert-Jan Creemers, Martijn G. H. van Oijen, Hanneke W. M. van Laarhoven

Erschienen in: Gastric Cancer | Ausgabe 4/2020

Abstract

Background

Addition of trastuzumab to first-line palliative chemotherapy in gastroesophageal cancer patients with HER2 overexpression has shown to improve survival. Real-world data on HER2 assessment and administration of trastuzumab are lacking. The aim of this study was to assess HER2 testing, trastuzumab administration, and overall survival (OS) in a nationwide cohort of metastatic gastroesophageal cancer patients.

Methods

Data of patients with synchronous metastatic gastroesophageal adenocarcinoma diagnosed in 2010–2016 that received palliative systemic treatment (n = 2846) were collected from the Netherlands Cancer Registry and Dutch Pathology Registry. The ToGA trial criteria were used to determine HER2 overexpression. Proportions of HER2 tested patients were analyzed between hospital volume categories using Chi-square tests, and over time using trend analysis. OS was tested using the Kaplan Meier method with log rank test.

Results

HER2 assessment increased annually, from 18% in 2010 to 88% in 2016 (P < 0.01). Median OS increased from 6.9 (2010–2013) to 7.9 months (2014–2016; P < 0.05). Between the hospitals, the proportion of tested patients varied between 29–100%, and was higher in high-volume hospitals (P < 0.01). Overall, 77% of the HER2 positive patients received trastuzumab. Median OS was higher in patients with positive (8.8 months) and negative (7.4 months) HER2 status, compared to non-tested patients (5.6 months; P < 0.05).

Conclusion

Increased determination of HER2 and administration of trastuzumab have changed daily practice management of metastatic gastroesophageal cancer patients receiving palliative systemic therapy, and possibly contributed to their improved survival. Further increase in awareness of HER2 testing and trastuzumab administration may improve quality of care and patient outcomes.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1007/​s10120-020-01039-7) contains supplementary material, which is available to authorized users.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Palliation by systemic therapy may improve quality as well as quantity of life in patients with metastatic gastroesophageal cancer [1-5]. In clinical trials, the addition of the targeted agent trastuzumab to cytotoxic therapy in metastatic gastroesophageal junction (GEJ) and gastric adenocarcinoma patients with overexpression of the human epidermal growth factor receptor 2 (HER2) has resulted in a median overall survival (OS) benefit of 2.8 months [6], and a positive impact on quality of life [7]. Trastuzumab has therefore become standard of care in HER2 positive tumors, and HER2 testing is strongly recommended in all patients with metastatic gastroesophageal adenocarcinoma eligible for HER2 targeted treatment [8-13].
HER2 testing and the administration of trastuzumab in gastroesophageal cancer might be underexposed within individual centers, because gastroesophageal cancer has a relatively low incidence in Western countries, and only 15–25% of the adenocarcinomas show HER2 overexpression [14-16]. In recent years, several studies have been published showing that gastroesophageal cancer patients treated in high-volume hospitals have better outcomes [17-25]. Patient volume can therefore be regarded as a proxy for quality of care, possibly due to multimodal expertise and a well-developed organization of care in high-volume hospitals [22, 26]. Moreover, although HER2 testing is routinely performed in breast cancer, HER2 expression in gastroesophageal cancer is more heterogenous as a reflection of the distinct biology of these tumors, and as a result, the interpretation of HER2 immunohistochemistry (IHC) patterns is more complicated [6, 27, 28].
Currently, data on HER2 testing, and the administration of trastuzumab in clinical practice are lacking. In this real-world study covering a nationwide cohort of synchronous metastatic gastroesophageal adenocarcinoma patients treated with systemic therapy, our aim was to explore the rate of HER2 testing, the administration of trastuzumab, interhospital variation, and survival in these patients.

Methods

Data collection

Patients with synchronous metastatic gastroesophageal adenocarcinoma (classified as C15 and C16 according to the International Classification of Diseases for Oncology [29]) treated with systemic therapy, and therefore eligible for HER2 targeted therapy, were selected from the nationwide Netherlands Cancer Registry (NCR). All patients diagnosed in 2010–2015 were identified, and a subset of patients diagnosed in 2016 because not all patients were registered in the NCR at the time of selection. Pathology reports of all confirmed cancer diagnoses in the Netherlands are archived in the nationwide network and registry of histo- and cytopathology (PALGA) [30]. Every pathology laboratory in the Netherlands is part of the PALGA network, and excerpts of all pathological reports are automatically transferred from the laboratories to the central databank of PALGA. Also, modifications in the excerpts or results of additional pathological tests, e.g., HER2 testing, are added to the central database automatically. Of included patients, information of HER2 testing was extracted from PALGA reports concerning histologic material with gastroesophageal origin.
Data on patient and tumor characteristics were extracted from the hospital’s medical records by trained data managers, and information on vital status from the Dutch population register (updated until 1 February 2019). OS was assessed from start of treatment until death or end of follow-up. Time to failure (TTF) was calculated from the start of treatment to the first progression that resulted in termination of first-line treatment, end of follow-up, or death within 90 days after the last hospital visit in case no progression was registered. Details on systemic treatment regimen and TTF were available in patients who were diagnosed in 2015 (n = 445) or in a subset of Dutch hospitals between 2010 and 2014 (n = 1107), due to logistic reasons. The subset of hospitals was selected as a representative sample of all Dutch hospitals in terms of patient volume, and hospital type and location, as described earlier [31].

Patient selection

The NCR provided 3164 patients diagnosed with gastroesophageal adenocarcinoma and synchronous metastases diagnosed between 2010 and 2016 and treated with systemic therapy. The linkage of the NCR and the PALGA database identified 3139 patients, with a total of 7545 available pathology excerpts (Fig. 1).

HER2 status

HER2 testing is usually initiated by the treating clinician in The Netherlands. A validated testing algorithm for HER2, based on the results of the ToGA trial [28], is suggested in international and national guidelines [8-10, 32-34]. The Dutch gastric cancer guideline recommends the use of validated HER2 antibodies for IHC and validated ISH tests, and a scoring system for the interpretation of these tests as described by Rüschoff et al. [27, 34] A gastroesophageal tumor can be considered HER2 positive when the result of the IHC staining pattern is 3+ , and negative when it is 0 or 1+ . In case of a equivocal IHC result (2+), additional testing using in situ hybridization (ISH) is indicated. The definition of HER2 ISH positivity is a HER2:chromosome 17 ratio of ≥ 2 [27]. Genomic testing techniques such as multiplex ligation-dependent probe amplification (MLPA) are used instead of ISH as well [12].
HER2 status was regarded as unknown if type and/or results of testing were not reported, because we could not verify if the HER2 criteria of the ToGA trial were used. In case of an equivocal IHC with an unknown ISH or MLPA result, HER2 status was also assumed unknown. If HER2 was tested multiple times, the last test result that was performed prior to or within 31 days after start of first-line systemic treatment was considered the definitive result, because this was expected to be decisive for the choice of systemic treatment. If HER2 testing was not mentioned in the reports, we assumed that it had not been performed.

Hospital volume

Per hospital the volume of all gastroesophageal cancer patients (both adenocarcinoma and squamous cell carcinoma) that received systemic therapy in 2015–2016, regardless of tumor stage and the intent of treatment, was calculated. With the aim to reflect current practice, the volume of the two most recent years, was used. Hospitals were categorized into quartiles according to these volumes to compare the proportion of HER2 tested patients.

Statistical analyses

Baseline characteristics and details on HER2 testing were displayed with counts and percentages, or medians and interquartile ranges (IQRs). Differences in the proportions of HER2 tested patients between the hospital volume categories were analyzed using Chi-square tests, and over time using the Cochran-Armitage test for trend. Factors possibly associated with HER2 testing were identified using logistic regression. Differences in survival were tested univariably with the log rank test using Kaplan Meier curves and through multivariable proportional hazards regression analyzes with adjustment for relevant patient and tumor characteristics. For survival analyzes, patients in whom HER2 was tested > 31 days after first-line systemic treatment were excluded to reduce immortal time bias. P values below 0.05 were considered statistically significant. Analyses were performed using SAS software (version 9.4, SAS institute, Cary, NC, USA).

Results

Patient characteristics

The majority of all 2846 included patients was male (76%), and median age was 64 (IQR, 56–71) years (Table 1). The primary tumor location was the esophagus in 41%, the non-cardia stomach in 40% and GEJ/cardia in 19%. More than half (54%) of the patients had an intestinal-type adenocarcinoma, followed by 27% with a diffuse, and 6% with an indeterminate type, based on the Lauren’s criteria [35]. In 13%, histological type was not specified. The majority of the tumors had a poor differentiation (53%).
Table 1
Baseline characteristics of included patients (n = 2846)
 
All patients (n = 2846) no. (%)
Non-tested (n = 1322) no. (%)
HER2 tested (n = 1524) no. (%)
P value
Male
2152 (75.6%)
1023 (77.4%)
1129 (74.1%)
0.041a
Age (years) median (IQR)
64.0 (56.0, 71.0)
65.0 (58.0, 72.0)
63.0 (55.0, 69.0)
 < 0.001b
  < 50
963 (33.8%)
383 (29.0%)
580 (38.1%)
 < 0.001a
 50–64
1079 (37.9%)
510 (38.6%)
569 (37.3%)
 
 65–79
735 (25.8%)
386 (29.2%)
349 (22.9%)
 
 ≥ 80
69 (2.4%)
43 (3.3%)
26 (1.7%)
 
Comorbidities
   
 < 0.001a
 0
739 (26.0%)
285 (21.6%)
454 (29.8%)
 
 1
555 (19.5%)
245 (18.5%)
310 (20.3%)
 
  ≥ 2
629 (22.1%)
278 (21.0%)
351 (23.0%)
 
 Unknown
923 (32.4%)
514 (38.9%)
409 (26.8%)
 
Tumor location
   
 < 0.001a
 Esophageal
1159 (40.7%)
589 (44.6%)
570 (37.4%)
 
 Gastroesophageal junction/cardia
545 (19.1%)
219 (16.6%)
326 (21.4%)
 
 Stomach (non-cardia)
1142 (40.1%)
514 (38.9%)
628 (41.2%)
 
Tumor histology
   
0.012a
 Adenocarcinoma NOS
362 (12.7%)
160 (12.1%)
202 (13.3%)
 
 Intestinal-type adenocarcinoma
1540 (54.1%)
744 (56.3%)
796 (52.2%)
 
 Diffuse type adenocarcinoma
772 (27.1%)
327 (24.7%)
445 (29.2%)
 
 Indeterminate type adenocarcinoma
172 (6.0%)
91 (6.9%)
81 (5.3%)
 
Tumor differentiation
   
 < 0.001a
 Well differentiated
37 (1.3%)
15 (1.1%)
22 (1.4%)
 
 Moderately differentiated
495 (17.4%)
221 (16.7%)
274 (18.0%)
 
 Poorly differentiated
1496 (52.6%)
648 (49.0%)
848 (55.6%)
 
 Unknown
818 (28.7%)
438 (33.1%)
380 (24.9%)
 
Metastatic sites
   
 < 0.001a
 1
1571 (55.2%)
757 (57.3%)
814 (53.4%)
 
  ≥ 2
1275 (44.8%)
565 (42.7%)
710 (46.6%)
 
Year of diagnosis
   
 < 0.001a
 2010
414 (14.5%)
341 (25.8%)
73 (4.8%)
 
 2011
386 (13.6%)
265 (20.0%)
121 (7.9%)
 
 2012
423 (14.9%)
240 (18.2%)
183 (12.0%)
 
 2013
410 (14.4%)
172 (13.0%)
238 (15.6%)
 
 2014
451 (15.8%)
156 (11.8%)
295 (19.4%)
 
 2015
445 (15.6%)
109 (8.2%)
336 (22.0%)
 
 2016
317 (11.1%)
39 (3.0%)
278 (18.2%)
 
Tumor histology and differentiation are based on the primary tumor. Diffuse type tumors were classified as poorly differentiated
IQR interquartile range, NOS not otherwise specified
aChi square test
bMann–Whitney U test

HER2 testing

HER2 status was determined in 54% of the patients (n = 1524; Table 1). The proportion of tested patients increased over time (P < 0.001), from 18% in 2010, to 88% in 2016 (Fig. 2). This trend was seen in esophageal (11–89%), GEJ (24–93%) and gastric tumors (22–83%; all P < 0.001). HER2 tested patients were significantly younger, more often female, and had more frequently GEJ/cardia or stomach compared to esophageal tumors, and diffuse type adenocarcinomas than non-tested patients (Table 1).
HER2 was positive in 19% of 1524 tested patients, and negative in 68% (Supplementary Table 1). In 204 (13%) patients, HER2 status was unknown because detailed HER2 test results were not described. The number of HER2 positive tumors increased from 14% in 2010–2012 to 20% in 2015–2016 (Fig. 2). Overall, HER2 positivity was found in 28% of esophageal, 16% of GEJ/cardia, and 12% of gastric adenocarcinomas (P < 0.001).

HER2 testing methods

Table 3 displays which diagnostic methods were used for the HER2 assessment, and all test results. IHC, ISH and MLPA were used in 88%, 49%, and 3% of the 1524 tested patients, respectively. Supplementary Table 2 displays which diagnostic methods were used for the HER2 assessment, and all test results of all performed tests. IHC, ISH and MLPA were used in 88%, 49%, and 3% of the 1524 tested patients, respectively, while testing methods were unknown in 13%. Of the patients in whom IHC was performed (n = 1328), scores of 0, 1+ , 2+ , 3+ were found in 38%, 23%, 24%, and 14%, respectively.
HER2 testing was performed more than once in 225 patients: in 194 patients, it was tested twice, in 30 patients three times, and in one patient four times. In 87% of the tested patients, HER2 was determined on solely the primary tumor, followed by metastasis only in 7%, and on both the primary tumor and metastasis in 6% of the patients. Testing methods were known in 1537/1764 tests, and in 398/1537 (26%) of these tests, ISH was used despite an IHC test result that would not necessarily require further testing (0, 1+ or 3+ ; Table 3).

Hospital variation

The subdivision of hospitals resulted in volume categories of < 13, 13–31, 32–76 and > 76 patients treated with systemic therapy in 2015 and 2016. The proportion of HER2 tested patients differed between these volumes in patients diagnosed in 2015–2016 (P < 0.001), with the highest proportions of tested patients being found in the high-volume centers (88%; Table 2). Interhospital variation in HER2 tested was 29–100% (Fig. 3).
Table 2
HER2 testing by hospital volume of systemic treatment in 2015–2016
Hospital volume
Hospitals no.
Patients no.
HER2 tested patients no. (%)
P value
 < 13 patients
17
72
49 (68.1%)
 < 0.001
13–31 patients
19
157
119 (75.8%)
 
32–76 patients
19
231
179 (77.5%)
 
 > 76 patients
19
302
267 (88.4%)
 
Hospital volume-based differences in proportion of HER2 tested patients with metastatic gastroesophageal cancer diagnosed in 2015 and 2016. Hospitals are categorized in quartiles based on the hospital volume of all gastroesophageal cancer patients treated with systemic therapy in 2015 and 2016

Factors contributing to probability of HER2 testing

Male sex, all but the highest hospital volumes, and death within 90 days after start of treatment were independently associated with lower probability of being tested for HER2 in patients diagnosed in 2015–2016, and GEJ/cardia tumors with a higher chance of testing to non-cardia gastric tumors (Table 3).
Table 3
Multivariable logistic regression analyses for chance of HER2 assessment for patients with metastatic gastroesophageal cancer treated with systemic therapy and diagnosed in 2015 and 2016 (n = 762)
  
Univariable
Multivariable
 
Patients no
OR
95%CI OR
P value
OR
95%CI OR
P value
Sex
       
 Female
166
Ref
  
Ref
  
 Male
596
0.41
0.24–0.69
0.001
0.43
0.24–0.77
0.005
Age (years)
       
  < 50
235
Ref
  
Ref
  
 50–64
292
0.67
0.42–1.06
0.087
0.84
0.51–1.40
0.510
 65–79
216
0.58
0.36–0.94
0.026
0.65
0.38–1.11
0.113
 ≥ 80
19
0.37
0.13–1.03
0.057
0.37
0.12–1.14
0.084
Performance status
       
 0 or 1
490
Ref
  
Ref
  
  ≥ 2
49
0.78
0.42–1.48
0.453
0.99
0.50–1.97
0.981
 Unknown
223
0.66
0.45–0.98
0.040
0.73
0.47–1.12
0.144
Number of comorbidities
       
 0
236
Ref
     
 1
208
0.61
0.37–1.00
0.048
0.62
0.36–1.06
0.081
  ≥ 2
256
0.54
0.34–0.87
0.011
0.66
0.39–1.11
0.119
 Unknown
62
0.68
0.33–1.41
0.295
0.50
0.23–1.10
0.087
Primary tumor location
       
 Esophagus
356
0.76
0.51–1.13
0.170
0.80
0.51–1.24
0.315
 GEJ/cardia
146
1.61
0.90–2.89
0.110
2.09
1.12–3.90
0.020
 Stomach
260
Ref
  
Ref
  
Year of diagnosis
       
 2015
445
Ref
  
Ref
  
 2016
317
2.31
1.55–3.45
 < 0.001
2.54
1.67–3.88
 < 0.001
Hospital volume
       
  < 13 patients
72
0.28
0.15–0.51
 < 0.001
0.26
0.14–0.51
 < 0.001
 13–31 patients
157
0.41
0.25–0.68
 < 0.001
0.37
0.22–0.64
< 0.001
 32–76 patients
231
0.45
0.28–0.72
 < 0.001
0.46
0.28–0.75
0.002
  > 76 patients
302
Ref
  
Ref
  
Deceased within 90 days after start systemic therapy
166
0.56
0.38–0.84
0.005
0.61
0.38–0.95
0.029
Multivariable logistic regression analyses for HER2 testing in patients diagnosed in 2015 and 2016
OR odds ratio, CI confidence interval, GEJ gastroesophageal junction

Survival

OS was 7.3 (IQR, 3.5, 12.6) months in all 2846 patients, and increased from 6.9 (IQR, 3.5, 12.0) months in patients diagnosed in 2010–2013 (n = 1633) to 7.9 (IQR, 3.6, 13.6) months in 2014–2016 (n = 1213). OS was 6.2 (IQR, 2.9, 10.7) months in non-tested patients (n = 1322) versus 8.3 (IQR, 4.2, 14.4) months in HER2 tested patients (n = 1524).
In 1355 patients that were tested within 1 month after start of systemic treatment, median OS in patients with unknown, negative and positive HER2 status was 7.6, 7.4, and 9.8 months, respectively (Fig. 4). OS was significantly higher in HER2 positive and negative patients, compared to non-tested patients (HER2 negative: adjusted hazard ratio (HR) 0.81, 95% confidence interval (CI) 0.65–0.99; HER2 positive: HR 0.65, 95%CI 0.50–0.86; Table 4). Diffuse type tumors, ≥ 2 metastatic locations, and performance status ≥ 2 were independently associated with worse survival. Hospital volume categorized into 4 quartiles was not independently associated with OS, while HRs in the two lowest quartiles were 1.26 and 1.19, respectively. When hospital volumes were subdivided in low (below median) versus high (above median), patients treated in low-volume hospitals had a significantly worse survival (HR 1.19, 95%CI 1.00–1.41; P = 0.044).
Table 4
Multivariable Cox regression analyses overall survival in patients diagnosed between 2015 and 2016 (n = 735a)
 
Patients no.
Median OS (months)
Univariable
Multivariable
HR
95%CI HR
P value
HR
95%CI HR
P value
Sex
        
 Female
161
6.1
Ref
  
Ref
  
 Male
574
7.6
0.94
0.79–1.13
0.520
0.98
0.80–1.19
0.804
Age (years)
        
  < 50
228
7.6
Ref
  
Ref
  
 50–64
282
7.3
1.02
0.85–1.22
0.833
1.05
0.87–1.27
0.641
 65–79
207
6.5
1.00
0.82–1.22
0.988
1.04
0.84–1.28
0.754
  ≥ 80
18
5.8
1.33
0.82–2.16
0.245
1.20
0.72–1.99
0.477
Performance status
        
 0 or 1
459
7.8
Ref
  
Ref
  
  ≥ 2
65
3.4
1.78
1.37–2.32
 < 0.001
1.75
1.34–2.30
 < 0.001
 Unknown
211
6.0
1.20
1.02–1.42
0.033
1.19
1.00–1.42
0.047
Number of comorbidities
        
 0
225
7.6
Ref
  
Ref
  
 1
205
7.2
0.93
0.76–1.13
0.474
0.91
0.74–1.11
0.351
 ≥ 2
246
6.2
1.18
0.98–1.42
0.088
1.15
0.94–1.41
0.165
 Unknown
59
8.9
0.89
0.66–1.19
0.427
0.83
0.61–1.13
0.231
Primary tumor location
        
 Esophagus
344
7.3
0.94
0.79–1.11
0.431
1.05
0.85–1.29
0.659
 GEJ/cardia
143
7.8
0.85
0.69–1.05
0.138
0.96
0.76–1.21
0.703
 Stomach
248
6.5
Ref
  
Ref
  
Tumor histology
        
 Adenocarcinoma NOS
178
7.3
Ref
  
Ref
  
 Intestinal-type adenocarcinoma
350
8.0
1.00
0.83–1.21
0.992
0.95
0.77–1.19
0.671
 Diffuse type adenocarcinoma
169
6.0
1.31
1.05–1.64
0.015
1.33
1.01–1.74
0.040
 Indeterminate type adenocarcinoma
38
5.9
1.36
0.95–1.94
0.096
1.26
0.86–1.84
0.243
Number of metastatic locations
        
 1
389
7.7
Ref
  
Ref
  
  ≥ 2
346
6.6
1.22
1.05–1.42
0.010
1.25
1.07–1.46
0.005
Year of diagnosis
        
 2015
429
6.7
Ref
  
Ref
  
 2016
306
7.7
0.88
0.76–1.03
0.107
0.95
0.79–1.14
0.580
Hospital volumeb
        
  < 13 patients
70
5.2
1.28
0.97–1.67
0.081
1.26
0.95–1.67
0.104
 13–31 patients
151
6.4
1.26
1.03–1.54
0.026
1.19
0.96–1.47
0.135
 32–76 patients
220
7.0
1.11
0.93–1.33
0.247
1.04
0.86–1.25
0.669
  > 76 patients
294
7.9
Ref
  
Ref
  
HER2 status
        
 HER2 not tested
148
5.6
Ref
  
Ref
  
 HER2 tested, HER2 negative
392
7.4
0.77
0.64–0.94
0.010
0.81
0.66–1.00
0.040
 HER2 tested, HER2 positive
118
8.8
0.62
0.48–0.80
 < 0.001
0.66
0.49–0.85
0.002
HER2 tested, HER2 unknown
77
7.9
0.81
0.61–1.07
0.136
0.81
0.60–1.06
0.165
Multivariable Cox regression analyses for overall survival in patients diagnosed in 2015 and 2016
OS overall survival, HR hazard ratio, CI confidence interval, GEJ gastroesophageal junction, NOS not otherwise specified
aPatients in whom HER2 was determined after 31 days of start of systemic treatment (N = 27) were excluded to prevent an immortal time bias
bHospitals are categorized in quartiles based on the hospital volume of all gastroesophageal cancer patients treated with systemic therapy in 2015 and 2016

Treatment with trastuzumab

Details of systemic treatment and TTF were known in 1552 patients diagnosed in 2010–2015. HER2 was determined in 53% of these patients, of whom 17% were HER2 positive and 69% negative, and in 14% HER2 was unknown. Of the 141 HER2 positive patients, 77% (n = 108) received a trastuzumab-containing regimen, which increased from 60% (n = 35) in 2010–2013 to 88% (n = 73) in 2014–2015. OS increased in this period from 6.9 (IQR, 3.2–11.7) to 7.2 (IQR, 3.4–12.8) months (P = 0.079). Ninety-seven of trastuzumab-treated patients received it in first-line, and 11 patients beyond first-line treatment. In trastuzumab-containing regimens, chemotherapy backbones were doublets in 59%, triplets in 20%, and monotherapy in 11%. Most frequently used backbones were capecitabine/5-FU with oxaliplatin (n = 33) or with cisplatin (n = 31).
In HER2 positive patients, median TTF of first-line trastuzumab-containing therapy was 6.5 (IQR, 3.0, 11.7) and OS 11.6 (IQR, 5.3, 21.6) months, while TTF of nontrastuzumab-containing first-line treatment was 5.4 (IQR, 3.1, 7.4) and OS 6.6 (IQR, 5.1, 11.0) months (n = 33). In HER2 negative patients, median TTF of first-line treatment was 5.2 (IQR, 2.2, 9.0) and OS 7.5 (IQR, 3.9, 13.1) months.

Discussion

Adequate HER2 testing is crucial for optimal decision-making on systemic treatment in metastatic gastroesophageal adenocarcinoma patients. In international guidelines it is therefore recommended to perform HER2 testing in all of these patients [8-10, 33]. In this nationwide cohort of 2846 patients with synchronous metastases and treated with palliative systemic therapy, HER2 testing increased over the study period from one in five to almost all patients. We found a large variety in the percentage of HER2 tested patients between the hospitals, and the volume of treated patients in a hospital independently associated with the probability of being tested for HER2.
Noteworthy, even in patients for whom the decision to be treated with systemic therapy had already been taken as is the case in our cohort still more than 10% of patients were not tested for HER2 in 2016. Even when we restrict our analysis to patients included in the ToGA study, i.e., with gastric or GEJ tumors, we still observed that 17% of gastric and 7% of GEJ adenocarcinoma patients diagnosed in 2016 were not tested. Male sex and treatment in lower hospital volumes were associated with a lower probability of HER2 assessment. Possible reasons for not testing could include contraindications for treatment with trastuzumab, and unawareness among physicians.
The HER2 overexpression rate of 19% is comparable with other studies [14-16]. In our study, this rate increased over time, probably because of the rise in tested esophageal adenocarcinomas, with a higher HER2 positivity rate compared to GEJ/cardia and stomach tumors. Overall, 23% of HER2 positive patients did not receive trastuzumab despite treatment with systemic therapy, which is remarkable as the additive side effects of trastuzumab are mild, while survival benefit is significant [6]. It cannot be excluded that financial reasons played a role [36]. For example, the reimbursement of trastuzumab could have been an issue if patients were not eligible for treatment with cisplatin, capecitabine or 5-FU, since the costs of trastuzumab are only covered when combined with this chemotherapy [37].
Furthermore, we found an interhospital variation in the proportion of HER2 tested patients of 29–100%, with a lower probability of undergoing HER2 assessment in low-volume compared to high-volume hospitals. A similar association with hospital volume was recently found in the probability of undergoing surgical treatment for gastric cancer [20]. Although we did not find a statistically significant association between hospital volume quartiles and, this was possibly a result of the limited number of patients in the lower hospital volumes. We did find this association when hospitals were categorized in two volume categories, which is in line with earlier published nationwide results [22]. This suggests HER2 assessment could increase if physicians of high-volume centers are involved in treatment decision-making, e.g., through regional multidisciplinary tumor boards.
Importantly, we found that in 26% of the HER2 assessments performed, ISH or MLPA was used despite a non-equivocal IHC result [28]. Reasons for additional ISH testing could include inadequate assessment of IHC staining due to HER2 heterogeneity or discordance between the primary tumor and metastasis [28, 38-40]. HER2 should therefore ideally be assessed on multiple specimens of the primary tumor, as well as on metastases, since HER2 targeted therapy is indicated if in one of the tumor specimens HER2 overexpression is observed [13].
HER2 positive patients treated with a trastuzumab-containing regimen had a longer survival compared to chemotherapy alone (11.6 and 6.6 months, respectively). However, survival in both groups was remarkably lower than in the ToGA trial (13.8 and 11.0 months, respectively) [6], probably due to restrictions in trial inclusion (e.g., performance status > 2), and because median age of our cohort was higher (64 versus 59 years in the ToGA trial). Nevertheless, the rise in trastuzumab administration over time could have contributed to the increased survival in our cohort from 6.9 (2010–2013) to 7.9 months (2014–2016).
Both HER2 positive and negative patients showed prolonged survival compared to non-tested patients. This supports the assumption of a selection of prognostically favorable patients that are tested for HER2, also endorsed by the higher number of tested patients without comorbidities compared to non-tested patients. Another explanation could be that non-tested patients are treated more frequently in low-volume hospitals. Moreover, the non-tested, but HER2 positive patients that did not receive trastuzumab could also have contributed to the lower survival in this group, since HER2 overexpression without targeted treatment is regarded a negative prognostic factor, although this is still subject of debate [41-43].
This is the first study in which real-world HER2 testing and outcomes of a nationwide gastroesophageal cancer cohort are described. However, the assumption that HER2 was not tested if it was not disclosed in pathology reports could have resulted in an underestimation of HER2 tested patients. Another limitation is the lack of information on reasons why HER2 testing was not performed or why trastuzumab was not administered. Lastly, as in any retrospective study, there were some missing data, which possibly hampered correction for confounding in multivariable analyses.
Our finding that still more than 10% of the patients treated with systemic therapy were not tested for HER2 is worrisome, not only because trastuzumab is currently the only targeted therapy in first-line palliative systemic treatment that has shown to improve survival rates, but also because other promising targets and biomarkers are on their way [44], such as programmed death-ligand 1 (PD-L1) [45, 46], Epstein-Barr Virus (EBV) [47, 48] and microsatellite instability (MSI) [49, 50] as biomarkers for checkpoint inhibition and selection in case of promising targeted therapies. Increased uptake of biomarker testing is therefore highly warranted in clinical practice.
In conclusion, daily practice management of metastatic gastroesophageal cancer has changed due to increased determination of HER2 status and administration of trastuzumab, which may have contributed to the improved survival in these patients over time. Advances in clinical practice could include a further increase in awareness of HER2 testing, especially in low-volume hospitals, and in trastuzumab administration.

Acknowledgements

The authors thank the registration team of the Netherlands Comprehensive Cancer Organisation (IKNL) for the collection of data for the Netherlands Cancer Registry. We also thank Baruch Kiestra for his help with the data extraction from the pathology reports.

Compliance with ethical standards

Conflict of interest

RHAV reports grants from BMS and Roche, outside the submitted work. NHM has served as a consultant for BMS, Lilly and MSD. JdV has received non-financial support from BTG, and Servier, and has served as a consultant for Shire and has received institutional research funding from Servier. MGHvO reports grants from Roche, during the conduct of the study, has received unrestricted research grants from BMS, Merck Serono, Nordic, Roche and Servier. HMWvL reports grants from Roche, during the conduct of the study, has served as a consultant for BMS, Celgene, Lilly, and Nordic and has received unrestricted research funding from Bayer, BMS, Celgene, Lilly, Merck Serono, MSD, Nordic, Philips, and Roche. The other authors have nothing to disclose.

Ethical approval

According to the Central Committee on Research involving Human Subjects, this type of study does not require approval from an ethics committee in the Netherlands. The study was approved by the Privacy Review Board of the NCR and the scientific committee of the Dutch Upper GI Cancer Group.
Consent to participate was not considered necessarily.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

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.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Ter VE, Mohammad NH, Van Valkenhoef G, Ngai LL, Mali RMA, Anderegg MC, et al. The efficacy and safety of first-line chemotherapy in advanced esophagogastric cancer: a network meta-analysis. J Natl Cancer Inst. 2016;108:1–13. Ter VE, Mohammad NH, Van Valkenhoef G, Ngai LL, Mali RMA, Anderegg MC, et al. The efficacy and safety of first-line chemotherapy in advanced esophagogastric cancer: a network meta-analysis. J Natl Cancer Inst. 2016;108:1–13.
2.
Zurück zum Zitat Al-Batran SE, Ajani JA. Impact of chemotherapy on quality of life in patients with metastatic esophagogastric cancer. Cancer. 2010;116:2511–8.PubMed Al-Batran SE, Ajani JA. Impact of chemotherapy on quality of life in patients with metastatic esophagogastric cancer. Cancer. 2010;116:2511–8.PubMed
3.
Zurück zum Zitat Janmaat VT, Steyerberg EW, van der Gaast A, Mathijssen RHJ, Bruno MJ, Peppelenbosch MP, et al. Palliative chemotherapy and targeted therapies for esophageal and gastroesophageal junction cancer. Cochrane Database Syst Rev. 2017;11:CD004063 Janmaat VT, Steyerberg EW, van der Gaast A, Mathijssen RHJ, Bruno MJ, Peppelenbosch MP, et al. Palliative chemotherapy and targeted therapies for esophageal and gastroesophageal junction cancer. Cochrane Database Syst Rev. 2017;11:CD004063
5.
Zurück zum Zitat Haj Mohammad N, ter Veer E, Ngai L, Mali R, van Oijen MGH, van Laarhoven HWM. Optimal first-line chemotherapeutic treatment in patients with locally advanced or metastatic esophagogastric carcinoma: triplet versus doublet chemotherapy: a systematic literature review and meta-analysis. Cancer Metastasis Rev. 2015;34:429–41.CrossRef Haj Mohammad N, ter Veer E, Ngai L, Mali R, van Oijen MGH, van Laarhoven HWM. Optimal first-line chemotherapeutic treatment in patients with locally advanced or metastatic esophagogastric carcinoma: triplet versus doublet chemotherapy: a systematic literature review and meta-analysis. Cancer Metastasis Rev. 2015;34:429–41.CrossRef
6.
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. https://doi.org/10.1016/S0140-6736(10)61121-X.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. https://​doi.​org/​10.​1016/​S0140-6736(10)61121-X.CrossRefPubMed
7.
Zurück zum Zitat Satoh T, Bang Y-J, Gotovkin EA, Hamamoto Y, Kang Y-K, Moiseyenko VM, et al. Quality of life in the trastuzumab for gastric cancer trial. Oncologist. 2014;19:712–9.CrossRef Satoh T, Bang Y-J, Gotovkin EA, Hamamoto Y, Kang Y-K, Moiseyenko VM, et al. Quality of life in the trastuzumab for gastric cancer trial. Oncologist. 2014;19:712–9.CrossRef
8.
Zurück zum Zitat Lordick F, Mariette C, Haustermans K, Obermannová R, Arnold D. Oesophageal cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2016;27:50–7.CrossRef Lordick F, Mariette C, Haustermans K, Obermannová R, Arnold D. Oesophageal cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2016;27:50–7.CrossRef
9.
Zurück zum Zitat Ajani JA, D’Amico TA, Almhanna K, Bentrem DJ, Chao J, Das P, et al. Gastric cancer, version 3.2016; clinical practice guidelines in oncology. JNCCN J Natl Compr Cancer Netw. 2016;14:1286–312.CrossRef Ajani JA, D’Amico TA, Almhanna K, Bentrem DJ, Chao J, Das P, et al. Gastric cancer, version 3.2016; clinical practice guidelines in oncology. JNCCN J Natl Compr Cancer Netw. 2016;14:1286–312.CrossRef
10.
Zurück zum Zitat Ajani JA, D’Amico TA, Almhanna K, Bentrem DJ, Chao J, Das P, et al. Esophageal and esophagogastric junction cancers, Version 1.2015. JNCCN J Natl Compr Cancer Netw. 2016;13:194–227.CrossRef Ajani JA, D’Amico TA, Almhanna K, Bentrem DJ, Chao J, Das P, et al. Esophageal and esophagogastric junction cancers, Version 1.2015. JNCCN J Natl Compr Cancer Netw. 2016;13:194–227.CrossRef
11.
Zurück zum Zitat Kodera Y, Sano T. Japanese gastric cancer treatment guidelines 2014 (ver 4). Gastric Cancer. 2017;20:1–19.CrossRef Kodera Y, Sano T. Japanese gastric cancer treatment guidelines 2014 (ver 4). Gastric Cancer. 2017;20:1–19.CrossRef
12.
Zurück zum Zitat Bartley AN, Washington MK, Ventura CB, Ismaila N, Colasacco C, Al Benson B, et al. HER2 testing and clinical decision making in gastroesophageal adenocarcinoma guideline from the College of American Pathologists, American Society for clinical Pathology, and American Society of Clinical Oncology. Am J Clin Pathol. 2016;146:647–69.CrossRef Bartley AN, Washington MK, Ventura CB, Ismaila N, Colasacco C, Al Benson B, et al. HER2 testing and clinical decision making in gastroesophageal adenocarcinoma guideline from the College of American Pathologists, American Society for clinical Pathology, and American Society of Clinical Oncology. Am J Clin Pathol. 2016;146:647–69.CrossRef
14.
Zurück zum Zitat Van Cutsem E, Bang YJ, Feng-yi F, Xu JM, Lee KW, Jiao SC, et al. HER2 screening data from ToGA: targeting HER2 in gastric and gastroesophageal junction cancer. Gastric Cancer Jpn. 2015;18:476–84.CrossRef Van Cutsem E, Bang YJ, Feng-yi F, Xu JM, Lee KW, Jiao SC, et al. HER2 screening data from ToGA: targeting HER2 in gastric and gastroesophageal junction cancer. Gastric Cancer Jpn. 2015;18:476–84.CrossRef
15.
Zurück zum Zitat Gravalos C, Jimeno A. HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol. 2008;19:1523–9.CrossRef Gravalos C, Jimeno A. HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol. 2008;19:1523–9.CrossRef
16.
Zurück zum Zitat Park D, Yun JW, Park JH, Oh SJ, Kim HJ, Cho YK, et al. HER-2/neu amplification is an independent prognostic factor in gastric cancer. Dig Dis Sci. 2006;51:1371–9.CrossRef Park D, Yun JW, Park JH, Oh SJ, Kim HJ, Cho YK, et al. HER-2/neu amplification is an independent prognostic factor in gastric cancer. Dig Dis Sci. 2006;51:1371–9.CrossRef
17.
Zurück zum Zitat Busweiler LAD, Dikken JL, Henneman D, van Berge Henegouwen MI, Ho VKY, Tollenaar RAEM, et al. The influence of a composite hospital volume on outcomes for gastric cancer surgery: a Dutch population-based study. J Surg Oncol. 2017;115:738–45.CrossRef Busweiler LAD, Dikken JL, Henneman D, van Berge Henegouwen MI, Ho VKY, Tollenaar RAEM, et al. The influence of a composite hospital volume on outcomes for gastric cancer surgery: a Dutch population-based study. J Surg Oncol. 2017;115:738–45.CrossRef
18.
Zurück zum Zitat Dikken JL, Van Sandick JW, Allum WH, Johansson J, Jensen LS, Putter H, et al. Differences in outcomes of oesophageal and gastric cancer surgery across Europe. Br J Surg. 2013;100:83–94.CrossRef Dikken JL, Van Sandick JW, Allum WH, Johansson J, Jensen LS, Putter H, et al. Differences in outcomes of oesophageal and gastric cancer surgery across Europe. Br J Surg. 2013;100:83–94.CrossRef
19.
Zurück zum Zitat Wouters MWJM, Gooiker GA, Van Sandick JW, Tollenaar RAEM. The volume-outcome relation in the surgical treatment of esophageal cancer: a systematic review and meta-analysis. Cancer. 2012;118:1754–63.CrossRef Wouters MWJM, Gooiker GA, Van Sandick JW, Tollenaar RAEM. The volume-outcome relation in the surgical treatment of esophageal cancer: a systematic review and meta-analysis. Cancer. 2012;118:1754–63.CrossRef
20.
Zurück zum Zitat Van Putten M, Verhoeven RHA, Van Sandick JW, Plukker JTM, Lemmens VEPP, Wijnhoven BPL, et al. Hospital of diagnosis and probability of having surgical treatment for resectable gastric cancer. Br J Surg. 2016;103:233–41.CrossRef Van Putten M, Verhoeven RHA, Van Sandick JW, Plukker JTM, Lemmens VEPP, Wijnhoven BPL, et al. Hospital of diagnosis and probability of having surgical treatment for resectable gastric cancer. Br J Surg. 2016;103:233–41.CrossRef
21.
Zurück zum Zitat van Putten M, Nelen SD, Lemmens VEPP, Stoot JHMB, Hartgrink HH, Gisbertz SS, et al. Overall survival before and after centralization of gastric cancer surgery in the Netherlands. Br J Surg. 2018;105:1807–15.CrossRef van Putten M, Nelen SD, Lemmens VEPP, Stoot JHMB, Hartgrink HH, Gisbertz SS, et al. Overall survival before and after centralization of gastric cancer surgery in the Netherlands. Br J Surg. 2018;105:1807–15.CrossRef
23.
Zurück zum Zitat Varagunam M, Hardwick R, Riley S, Chadwick G, Cromwell DA, Groene O. Changes in volume, clinical practice and outcome after reorganisation of oesophago-gastric cancer care in England: a longitudinal observational study. Eur J Surg Oncol Engl. 2018;44:524–31.CrossRef Varagunam M, Hardwick R, Riley S, Chadwick G, Cromwell DA, Groene O. Changes in volume, clinical practice and outcome after reorganisation of oesophago-gastric cancer care in England: a longitudinal observational study. Eur J Surg Oncol Engl. 2018;44:524–31.CrossRef
24.
Zurück zum Zitat Bachmann MO, Alderson D, Edwards D, Wotton S, Bedford C, Peters TJ, et al. Cohort study in South and West England of the influence of specialization on the management and outcome of patients with oesophageal and gastric cancers. Br J Surg. Engl. 2002;89:914–22.CrossRef Bachmann MO, Alderson D, Edwards D, Wotton S, Bedford C, Peters TJ, et al. Cohort study in South and West England of the influence of specialization on the management and outcome of patients with oesophageal and gastric cancers. Br J Surg. Engl. 2002;89:914–22.CrossRef
25.
Zurück zum Zitat Markar SR, Karthikesalingam A, Thrumurthy S, Low DE. Volume-outcome relationship in surgery for esophageal malignancy: systematic review and meta-analysis. J Gastrointest Surg. 2012;5:1055–63.CrossRef Markar SR, Karthikesalingam A, Thrumurthy S, Low DE. Volume-outcome relationship in surgery for esophageal malignancy: systematic review and meta-analysis. J Gastrointest Surg. 2012;5:1055–63.CrossRef
26.
Zurück zum Zitat Birkmeyer JD, Dimick JB. Understanding and reducing variation in surgical mortality. Annu Rev Med. 2008;60:405–15.CrossRef Birkmeyer JD, Dimick JB. Understanding and reducing variation in surgical mortality. Annu Rev Med. 2008;60:405–15.CrossRef
27.
Zurück zum Zitat Rüschoff J, Hanna W, Bilous M, Hofmann M, Osamura RY, Penault-Llorca F, et al. HER2 testing in gastric cancer: a practical approach. Mod Pathol. 2012;25:637–50.CrossRef Rüschoff J, Hanna W, Bilous M, Hofmann M, Osamura RY, Penault-Llorca F, et al. HER2 testing in gastric cancer: a practical approach. Mod Pathol. 2012;25:637–50.CrossRef
28.
Zurück zum Zitat Hofmann M, Stoss O, Shi D, Büttner R, Van De Vijver M, Kim W, et al. Assessment of a HER2 scoring system for gastric cancer: Results from a validation study. Histopathology. 2008;52:797–805.CrossRef Hofmann M, Stoss O, Shi D, Büttner R, Van De Vijver M, Kim W, et al. Assessment of a HER2 scoring system for gastric cancer: Results from a validation study. Histopathology. 2008;52:797–805.CrossRef
29.
Zurück zum Zitat Fritz A. International classification of diseases for oncology: ICD-O. 3rd ed. Geneva: World Heal. Organ; 2000. Fritz A. International classification of diseases for oncology: ICD-O. 3rd ed. Geneva: World Heal. Organ; 2000.
30.
Zurück zum Zitat Casparie M, Tiebosch TMG, Burger G, Blauwgeers H, van de Pol A, van Krieken JHJM, et al. Pathology databanking and biobanking in The Netherlands, a central role for PALGA, the nationwide histopathology and cytopathology data network and archive. Cell Oncol. 2007;29:19–24.PubMedPubMedCentral Casparie M, Tiebosch TMG, Burger G, Blauwgeers H, van de Pol A, van Krieken JHJM, et al. Pathology databanking and biobanking in The Netherlands, a central role for PALGA, the nationwide histopathology and cytopathology data network and archive. Cell Oncol. 2007;29:19–24.PubMedPubMedCentral
32.
Zurück zum Zitat Hage M, Smits MM, Imholz ALT, Louwen M, Koopman T. Pathologic diagnostics of HER2 positivity in gastroesophageal adenocarcinoma. Am J Clin Pathol. 2015;143:257–64.CrossRef Hage M, Smits MM, Imholz ALT, Louwen M, Koopman T. Pathologic diagnostics of HER2 positivity in gastroesophageal adenocarcinoma. Am J Clin Pathol. 2015;143:257–64.CrossRef
33.
Zurück zum Zitat Smyth EC, Verheij M, Allum W, Cunningham D, Cervantes A, Arnold D. Gastric cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Eur J Surg Oncol. 2016;27:v38–49. Smyth EC, Verheij M, Allum W, Cunningham D, Cervantes A, Arnold D. Gastric cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Eur J Surg Oncol. 2016;27:v38–49.
34.
36.
Zurück zum Zitat Lammers P, Criscitiello C, Curigliano G, Jacobs I. Barriers to the use of trastuzumab for HER2+ breast cancer and the potential impact of biosimilars: a physician survey in the united states and emerging markets. Pharmaceuticals. 2014;7:943–53.CrossRef Lammers P, Criscitiello C, Curigliano G, Jacobs I. Barriers to the use of trastuzumab for HER2+ breast cancer and the potential impact of biosimilars: a physician survey in the united states and emerging markets. Pharmaceuticals. 2014;7:943–53.CrossRef
38.
Zurück zum Zitat Fazlollahi L, Remotti HE, Iuga A, Yang HM, Lagana SM, Sepulveda AR. HER2 heterogeneity in gastroesophageal cancer detected by testing biopsy and resection specimens. Arch Pathol Lab Med. 2018;142:516–22.CrossRef Fazlollahi L, Remotti HE, Iuga A, Yang HM, Lagana SM, Sepulveda AR. HER2 heterogeneity in gastroesophageal cancer detected by testing biopsy and resection specimens. Arch Pathol Lab Med. 2018;142:516–22.CrossRef
39.
Zurück zum Zitat Park SR, Park YS, Ryu MH, Ryoo BY, Woo CG, Jung HY, et al. Extra-gain of HER2-positive cases through HER2 reassessment in primary and metastatic sites in advanced gastric cancer with initially HER2-negative primary tumours: Results of GASTric cancer HER2 reassessment study 1 (GASTHER1). Eur J Cancer. 2016;53:42–50. https://doi.org/10.1016/j.ejca.2015.09.018.CrossRefPubMed Park SR, Park YS, Ryu MH, Ryoo BY, Woo CG, Jung HY, et al. Extra-gain of HER2-positive cases through HER2 reassessment in primary and metastatic sites in advanced gastric cancer with initially HER2-negative primary tumours: Results of GASTric cancer HER2 reassessment study 1 (GASTHER1). Eur J Cancer. 2016;53:42–50. https://​doi.​org/​10.​1016/​j.​ejca.​2015.​09.​018.CrossRefPubMed
40.
Zurück zum Zitat Creemers A, Ter Veer E, De Waal L, Lodder P, Hooijer GKJ, Van Grieken NCT, et al. Discordance in HER2 status in gastro-esophageal adenocarcinomas: a systematic review and meta-analysis. Sci Rep. 2017;7:1–10.CrossRef Creemers A, Ter Veer E, De Waal L, Lodder P, Hooijer GKJ, Van Grieken NCT, et al. Discordance in HER2 status in gastro-esophageal adenocarcinomas: a systematic review and meta-analysis. Sci Rep. 2017;7:1–10.CrossRef
41.
Zurück zum Zitat Janjigian YY, Werner D, Pauligk C, Steinmetz K, Kelsen DP, Jäger E, et al. Prognosis of metastatic gastric and gastroesophageal junction cancer by HER2 status: a European and USA International collaborative analysis. Ann Oncol. 2012;23:2656–62.CrossRef Janjigian YY, Werner D, Pauligk C, Steinmetz K, Kelsen DP, Jäger E, et al. Prognosis of metastatic gastric and gastroesophageal junction cancer by HER2 status: a European and USA International collaborative analysis. Ann Oncol. 2012;23:2656–62.CrossRef
43.
Zurück zum Zitat Jørgensen JT, Hersom M. HER2 as a prognostic marker in gastric cancer - A systematic analysis of data from the literature. J Cancer. 2012;3:137–44.CrossRef Jørgensen JT, Hersom M. HER2 as a prognostic marker in gastric cancer - A systematic analysis of data from the literature. J Cancer. 2012;3:137–44.CrossRef
44.
Zurück zum Zitat Creemers A, Ebbing EA, Hooijer GKJ, Stap L, Jibodh-Mulder RA, Gisbertz SS, et al. The dynamics of HER2 status in esophageal adenocarcinoma. Oncotarget. 2018;9:26787–99.CrossRef Creemers A, Ebbing EA, Hooijer GKJ, Stap L, Jibodh-Mulder RA, Gisbertz SS, et al. The dynamics of HER2 status in esophageal adenocarcinoma. Oncotarget. 2018;9:26787–99.CrossRef
45.
Zurück zum Zitat Fuchs CS, Doi T, Jang RW, Muro K, Satoh T, Machado M, et al. Safety and efficacy of pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer: Phase 2 clinical KEYNOTE-059 trial. JAMA Oncol. 2018;4:1–8.CrossRef Fuchs CS, Doi T, Jang RW, Muro K, Satoh T, Machado M, et al. Safety and efficacy of pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer: Phase 2 clinical KEYNOTE-059 trial. JAMA Oncol. 2018;4:1–8.CrossRef
46.
Zurück zum Zitat Shitara K, Özgüroğlu M, Bang YJ, Di BM, Mandalà M, Ryu MH, et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet. 2018;1:123–33.CrossRef Shitara K, Özgüroğlu M, Bang YJ, Di BM, Mandalà M, Ryu MH, et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet. 2018;1:123–33.CrossRef
47.
49.
Zurück zum Zitat Pereira MA, Ramos MFKP, Faraj SF, Dias AR, Yagi OK, Zilberstein B, et al. Clinicopathological and prognostic features of Epstein-Barr virus infection, microsatellite instability, and PD-L1 expression in gastric cancer. J Surg Oncol. 2018;117:829–39.CrossRef Pereira MA, Ramos MFKP, Faraj SF, Dias AR, Yagi OK, Zilberstein B, et al. Clinicopathological and prognostic features of Epstein-Barr virus infection, microsatellite instability, and PD-L1 expression in gastric cancer. J Surg Oncol. 2018;117:829–39.CrossRef
Metadaten
Titel
Increased assessment of HER2 in metastatic gastroesophageal cancer patients: a nationwide population-based cohort study
verfasst von
Willemieke P. M. Dijksterhuis
Rob H. A. Verhoeven
Sybren L. Meijer
Marije Slingerland
Nadia Haj Mohammad
Judith de Vos-Geelen
Laurens V. Beerepoot
Theo van Voorthuizen
Geert-Jan Creemers
Martijn G. H. van Oijen
Hanneke W. M. van Laarhoven
Publikationsdatum
01.07.2020
Verlag
Springer Singapore
Erschienen in
Gastric Cancer / Ausgabe 4/2020
Print ISSN: 1436-3291
Elektronische ISSN: 1436-3305
DOI
https://doi.org/10.1007/s10120-020-01039-7

Weitere Artikel der Ausgabe 4/2020

Gastric Cancer 4/2020 Zur Ausgabe

Update Chirurgie

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

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

CME: 2 Punkte

Prof. Dr. med. Gregor Antoniadis Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

CME: 2 Punkte

Dr. med. Benjamin Meyknecht, PD Dr. med. Oliver Pieske Das Webinar S2e-Leitlinie „Distale Radiusfraktur“ beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

CME: 2 Punkte

Dr. med. Mihailo Andric
Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.