Introduction: The present study evaluated the potential benefit of adding cetuximab to neoadjuvant, adjuvant, or palliative standard therapy for pancreatic cancer. Methods: A systematic literature search was performed in MEDLINE, Web of Science, and Cochrane Central Register of Controlled Trials (CENTRAL). Only randomised controlled trials (RCTs) investigating the effect of adding cetuximab to standard chemotherapy in pancreatic cancer were included. Evaluated outcomes were overall survival, progression-free survival, objective response, and toxicity. For overall survival and progression-free survival, hazard ratios (HR) with 95% confidence intervals (CI) were chosen as effect measure. For objective response, odds ratios (OR) with 95% CI were used. Analysis was based on a random effects model. Results: After screening 568 publications, a total of 4 RCTs with 924 patients were included. In all trials, patients were adequately randomised with balanced intervention and control groups. There was no significant difference in overall survival (HR 1.04; 95% CI: 0.90–1.19; p = 0.60), progression-free survival (HR 1.06; 95% CI: 0.93–1.22; p = 0.36), or objective response (OR 0.99; 95% CI: 0.66 –1.49; p = 0.96) when adding cetuximab to a standard therapy. Toxicity was the same or higher in each of the included trials. According to GRADE, the certainty of the evidence is high. Therefore, adding cetuximab to pancreatic cancer therapy has no clinically relevant benefit. Conclusion: In the presence of no survival benefit, increased toxicity, and higher costs, a decreased cost-benefit ratio compared to the standard care must be suggested. Conducting further RCTs in unselected pancreatic cancer populations is unlikely to change this conclusion.

Pancreatic ductal adenocarcinoma is one of the most aggressive malignancies [1] and is typically diagnosed in an advanced state [2, 3], as it is characterised by early locoregional spread and distant metastasis. Gemcitabine used to be the standard first-line therapy for advanced pancreatic cancer for many years [3]. However, 1-year survival after gemcitabine therapy was reported at only 18–29% [4, 5]. Thus, alternate treatments for this disease are urgently needed. Novel chemotherapy combinations like FOLFIRINOX [6] or gemcitabine plus nab-paclitaxel have significantly improved overall survival for patients with metastatic disease. Unfortunately, except for erlotinib, targeted agents have proved unsuccessful in clinical pancreatic cancer trials to date [7]. Erlotinib with gemcitabine was superior to gemcitabine in a randomised phase III study, but the difference in overall survival was so small that the clinical relevance of this benefit is controversial [8]. Cetuximab, another targeted agent, has been tested in various clinical studies for pancreatic cancer [9-13]. Cetuximab is an immunoglobulin G1 monoclonal antibody that binds the epidermal growth factor receptor (EGFR) with high affinity, which competitively blocks ligand binding, inhibits tyrosine kinases activation, and results in receptor downregulation [14]. In 22–60% of human pancreatic carcinomas, EGFR is overexpressed [15, 16]. While some preclinical research shows positive results [17, 18], the effect of cetuximab in treating pancreatic cancer remains controversial [13]. Older systematic reviews evaluating the effect of cetuximab in pancreatic cancer also show inconsistent results [19, 20]. Further recent RCTs, which have not been considered in previous reviews, are now available [21, 22]. Therefore, a new systematic review is needed. The objective of this study was to systematically evaluate the potential benefits of treatment approaches including cetuximab for patients with pancreatic cancer.

This systematic review and meta-analysis was conducted according to an a priori defined protocol (PROSPERO CRD42017064450) and is reported according to the PRISMA guidelines [23]. The resources and facilities of the University of Heidelberg were used to conduct this study. There was no additional source of funding.

Systematic Literature Search

The Cochrane CENTRAL, MEDLINE (via PubMed), and Web of Science databases were searched [24] throughout July 2018. A search strategy based on a thesaurus (MeSH or Emtree) in combination with free text words was used. The search strategy for MEDLINE (via PubMed) was as follows:

((cetuximab[tw] OR (Erbitux[tw] OR “cetuximab”[Mesh]) AND (pancrea*[tw] OR “Pancreas”[Mesh]))

In addition, reference lists of relevant studies and related systematic reviews were screened manually. References citing trials eligible for inclusion were searched using the Science Citation Index via Web of Science. No language restriction was applied.

Study Selection

Randomised controlled trials (RCTs) investigating any treatment concept including cetuximab for the treatment of any stage of pancreatic cancer in a human adult population and reporting at least one clinically relevant endpoint (i.e., overall survival, progression-free survival, or objective response) were eligible for inclusion.

Following the recommendations of the Cochrane Collaboration [25], two independent reviewers screened the titles, abstracts, and full texts.

Data Extraction

Data extraction was also independently performed by two reviewers for quality assurance purposes [26]. A third reviewer resolved any discrepancies found by the first two reviewers.

The following items were extracted: title, author, year of publication, journal, language, trial duration, trial design, number of treatment groups, total number of patients, evaluable patients, withdrawals, loss to follow-up, funding source, and trial registration or published protocol.

Relevant outcome variables included overall survival, defined as the time period between beginning of randomisation and death for any reason; progression-free survival, defined as the time between randomisation and radiological progress of disease according to the RECIST criteria; objective response, defined as at least partial radiological remission of disease according to the RECIST criteria; and toxicity, as individually reported by the trials.

Critical Appraisal (Bias)

The methodological quality was assessed by the Cochrane Collaboration’s tool for assessing risk of bias [25]. The tool includes six standard domains of bias: selection bias (random sequence generation, allocation concealment), performance bias (blinding of participants and personnel), detection bias (outcome assessment), attrition bias (incomplete outcome data), reporting bias (selective reporting), and other bias. Each domain was judged to be at low, unclear, or high risk of bias according to the Cochrane Handbook for Systematic Reviews of Interventions [25]. Quantitative and qualitative results were merged and the quality of the evidence for each outcome was rated using the GRADE approach [27].

Statistical Analysis

Meta-analyses were performed with Review Manager (RevMan) version 5.3.5 (The Cochrane Collaboration, The Nordic Cochrane Centre, Copenhagen, Denmark).

To measure effects, hazard ratios (HR) with 95% confidence intervals (CI) were used for time-to-event outcomes (overall survival and progression-free survival), and odds ratios (OR) with 95% CI were used for the binary outcome (objective response). If a publication reported no HR or its corresponding standard error but contained appropriate information (e.g., Kaplan-Meier curves), the HR and its standard error were calculated using the methods provided by Tierney et al. [28] and Parmar et al. [29]. HRs were combined using the inverse-variance method, and ORs using the Mantel-Haenszel method. A random effects model was applied because relevant heterogeneity between the effects of the studies was assumed. Statistical heterogeneity was assessed by the I2 statistics [30]. Potential publication bias could not be evaluated due to the small number of included trials. For the same reason, no subgroup or sensitivity analyses were conducted.

A total of 568 articles were screened for eligibility. Of these, 41 were subject to full text analysis. Thirty-seven trials were excluded due to duplicate publication of the same cohort (n = 1), evaluation of a single treatment arm without a control group (n = 33), and a lack of information on clinical outcome (n = 3) (references available on request). Therefore, 4 RCTs with 924 patients were included in the qualitative and quantitative analysis [10, 21, 22, 31]. A PRISMA flow chart is shown in Figure 1.

Fig. 1.

PRISMA flow chart.

Fig. 1.

PRISMA flow chart.

Close modal

All RCTs evaluated cetuximab in a palliative setting. Regarding co-interventions, one trial investigated the combination of gemcitabine and cetuximab [10], one the combination of gemcitabine, cisplatin, and cetuximab [31], one the combination of gemcitabine, capecitabine, radiotherapy, and cetuximab [21], and one the combination of docetaxel, irinotecan, and cetuximab [22]. Two RCTs were conducted in Europe (Italy and the UK) [21, 31] and 2 originated from the USA [10, 22]. All RCTs had two trial arms with equally allocated treatment groups. The sample size varied across the trials (range: 13–743). The trials were published in journals with an impact factor of 3.25–13.28. An overview on the included RCTs is shown in Table 1.

Table 1.

List of included trials

List of included trials
List of included trials

Critical Appraisal (Bias)

Since all trials used an adequate measure of randomisation and allocation, none of them had a high risk of selection bias. Reporting of random sequence generation was precise (i.e., low risk of bias) in all 4 RCTs and for allocation concealment in 3 of the 4 RCTs. One RCT did not provide any information concerning this source of bias [21]. One trial was considered at a high risk of performance and detection bias, as neither participants nor personnel were blinded [10]. One trial reported appropriate blinding measures and was considered low risk [31]. Two trials did not report blinding measures and were at an unclear risk of bias [21, 22]. All trials were at a low risk of attrition bias or reporting bias, and no other biases were present in the 4 RCTs. Overall, 3 of the 4 trials were at a low risk of bias in all reported domains [21, 22, 31].

Quantitative Analysis

A quantitative analysis for overall survival, progression-free survival, and objective response was performed. Three of the 4 RCTs, with a total of 457 patients assigned to both the cetuximab and the non-cetuximab groups, reported overall survival rates. No significant difference in overall survival existed between the cetuximab and non-cetuximab groups (HR 1.04; 95% CI: 0.90–1.19; p = 0.60) (Fig. 2a).

Fig. 2.

Forest plots of overall survival (a), progression-free survival (b), and objective response (c).

Fig. 2.

Forest plots of overall survival (a), progression-free survival (b), and objective response (c).

Close modal

In 3 of the 4 RCTs reporting on progression-free survival, 457 patients were treated with cetuximab (with 457 patients in the control group). Progression-free survival was not different between the groups (HR 1.06; 95% CI: 0.93–1.22; p = 0.36) (Fig. 2b).

Objective response as defined by RECIST criteria was reported in all 4 RCTs, but in a smaller population of patients (cetuximab group: 418 patients, non-cetuximab group: 423 patients). Compared to the control group, patients treated with cetuximab had no significant benefit in objective response (OR 0.99; 95% CI: 0.66–1.49; p < 0.96) (Fig. 2c).

Altogether, cetuximab did not show any effect on the outcomes analysed. Statistical heterogeneity among the included trials could not be detected (I2 = 0% for all outcomes). Due to inconsistent reporting of toxicity in the trials, a meta-analysis was not possible. Table 2 shows a summary of reported toxicity, which was the same or higher in the cetuximab group.

Table 2.

Toxic effects reported in included trials

Toxic effects reported in included trials
Toxic effects reported in included trials

The objective of this systematic review was to summarise, evaluate, and critically appraise evidence for the potential benefit of cetuximab in the treatment of pancreatic cancer. Cetuximab suggested promising results in preclinical studies when added to gemcitabine in the treatment of human pancreatic carcinoma xenografts [17, 18]. In this systematic review, 4 RCTs adding cetuximab to standard palliative therapy were included. Cetuximab failed to demonstrate improved patient outcomes in terms of survival or objective response when compared to various chemotherapeutic regimens and/or other biological agents. According to the GRADE approach, the certainty of the evidence is high (Table 3). A quantitative analysis of toxicity was not possible; however, many severe side effects of cetuximab are known: reported grade 3 or 4 adverse events following the use of cetuximab include abdominal pain, neutropenia, thrombocytopenia, diarrhoea, nausea, allergy, and toxic skin reactions [32, 33]. Further, it is obvious that adding a drug to any standard care means higher costs. Therefore, in the presence of no survival benefit, the same or increased toxicity, and higher costs, a decreased cost-benefit ratio compared to the standard care must be suggested.

Table 3.

Summary of findings with quality of evidence (GRADE)

Summary of findings with quality of evidence (GRADE)
Summary of findings with quality of evidence (GRADE)

In the presence of existing synoptic evidence on this topic, it is important to compare those results with the present paper. Three systematic reviews investigating EGFR inhibitors in pancreatic cancer were found [19, 20, 34]. These reviews were published in 2010 and 2013, evaluating only one or two RCTs and not including the newest ones on this topic. Consequently, the present study contains the most comprehensive sample of trials using cetuximab in the treatment of patients with pancreatic cancer. The review of Liu et al. [34] investigated cetuximab in all kinds of advanced cancer and found a general benefit regarding survival; however, not for advanced pancreatic cancer. The review from Tian et al. [20], evaluating the use of EGFR inhibitors in the treatment of pancreatic cancer, shows no improvement in objective response and progression-free survival. The benefit of agents against EGFR on overall survival was shown, but this improvement in overall survival is likely because the authors did not specifically evaluate the effect of cetuximab alone, but also included other agents against EGFR, such as erlotinib. Similarly, Zagouri et al. [19] investigated different molecularly targeted therapies in metastatic pancreatic cancer and found the results promising to further investigate such therapies in pancreatic cancer disease.

Two limitations of the existing literature must be discussed: the heterogeneous control groups and the unselected patient population.

The standard of care in the included RCTs was highly varied. While Philip et al. [10] combined cetuximab with gemcitabine only, other RCTs used a combination of cetuximab with gemcitabine and cisplatin, or gemcitabine and capecitabine [21, 31]. One RCT even involved radiotherapy [21], and one did not use gemcitabine at all, combining cetuximab with docetaxel and irinotecan [22]. As a result, the control group for the meta-analyses is clinically heterogeneous. However, adding cetuximab to any treatment regime did not result in clinical benefit.

One critical point regarding cetuximab trials in pancreatic cancer patients is the fact that all studies were performed in unselected patients. It is well established that cetuximab is not effective in colorectal cancer patients with KRAS or NRAS mutations but very effective in those with RAS wild-type mutations [35]. Since more than 90% of patients with pancreatic cancer harbour mutations in the KRAS gene [36], it is not surprising that cetuximab is ineffective against this disease. Indeed, a randomised phase II trial investigating the combination of nimotuzumab, a different anti-EGFR monoclonal antibody, with gemcitabine showed a significant advantage in 1-year overall survival of 53.8 versus 15.8% for the combination group in KRAS wild-type patients (HR 0.32, p = 0.026), while the difference in KRAS-mutated patients was not statistically significant [37]. Furthermore, KRAS mutations are not the only molecular changes in pancreatic cancer; although pancreatic cancer does not harbour the most genetic alterations of all solid tumours, those that exist are spread over all relevant pathways with mutations in every patient [38]. This is one explanation why the conventional targeted therapy approach is not effective [39], but profiling of all druggable targets and a subsequent holistic analysis is required [40].

In conclusion, further trials investigating cetuximab in an unselected pancreatic cancer population must be viewed with a critical eye. The upper bound of the pooled effect for overall survival excludes a clinically relevant effect. A future RCT would therefore be unlikely to change the effect estimate or could even be unethical in the light of the data presented here. Further research in pancreatic cancer should focus on other treatment regimens than those including cetuximab or should investigate the benefit of cetuximab in the small minority of KRAS wild-type patients, respecting the entirety of all mutations in this dire tumour.

Adding cetuximab to pancreatic cancer therapy has no benefit. Conducting further RCTs in unselected patient populations is discouraged.

Ethical approval was not required as the present work is a meta-analysis.

The authors declare no competing interests according to the ICMJE guidelines that could have inappropriately influenced their work on this article.

The resources and facilities of the University of Heidelberg were used in conducting this study. This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Tobias Forster: planning of the study, screening of publications, formal analysis, biometric and statistical analysis, original draft, and writing. Felix J. Hüttner, Christoph Springfeld, Matthias Löhr, Thilo Hackert, and Markus K. Diener: review and editing. Eva Kalkum: literature search. Matthes Hackbusch: Biometric and statistical analysis. Pascal Probst: planning of the study, screening of publications, formal analysis, biometric and statistical analysis, writing, review, and editing. All authors read and approved the final paper.

All data can be obtained from the corresponding author on reasonable request.

1.
Löhr
JM
.
Pancreatic cancer should be treated as a medical emergency
.
BMJ
.
2014
Sep
;
349
sep04 12
:
g5261
.
[PubMed]
0959-8138
2.
Kleeff
J
,
Korc
M
,
Apte
M
,
La Vecchia
C
,
Johnson
CD
,
Biankin
AV
, et al.
Pancreatic cancer
.
Nat Rev Dis Primers
.
2016
Apr
;
2
(
1
):
16022
.
[PubMed]
2056-676X
3.
Neoptolemos
JP
,
Kleeff
J
,
Michl
P
,
Costello
E
,
Greenhalf
W
,
Palmer
DH
.
Therapeutic developments in pancreatic cancer: current and future perspectives
.
Nat Rev Gastroenterol Hepatol
.
2018
Jun
;
15
(
6
):
333
48
.
[PubMed]
1759-5045
4.
Tempero
M
,
Plunkett
W
,
Ruiz Van Haperen
V
,
Hainsworth
J
,
Hochster
H
,
Lenzi
R
, et al.
Randomized phase II comparison of dose-intense gemcitabine: thirty-minute infusion and fixed dose rate infusion in patients with pancreatic adenocarcinoma
.
J Clin Oncol
.
2003
Sep
;
21
(
18
):
3402
8
.
[PubMed]
0732-183X
5.
Burris
HA
 3rd
,
Moore
MJ
,
Andersen
J
,
Green
MR
,
Rothenberg
ML
,
Modiano
MR
, et al.
Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial
.
J Clin Oncol
.
1997
Jun
;
15
(
6
):
2403
13
.
[PubMed]
0732-183X
6.
Conroy
T
,
Desseigne
F
,
Ychou
M
,
Bouché
O
,
Guimbaud
R
,
Bécouarn
Y
, et al.;
Groupe Tumeurs Digestives of Unicancer
;
PRODIGE Intergroup
.
FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer
.
N Engl J Med
.
2011
May
;
364
(
19
):
1817
25
.
[PubMed]
0028-4793
7.
Mosquera
C
,
Maglic
D
,
Zervos
EE
.
Molecular targeted therapy for pancreatic adenocarcinoma: A review of completed and ongoing late phase clinical trials
.
Cancer Genet
.
2016
Dec
;
209
(
12
):
567
81
.
[PubMed]
2210-7762
8.
Moore
MJ
,
Goldstein
D
,
Hamm
J
,
Figer
A
,
Hecht
JR
,
Gallinger
S
, et al.;
National Cancer Institute of Canada Clinical Trials Group
.
Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group
.
J Clin Oncol
.
2007
May
;
25
(
15
):
1960
6
.
[PubMed]
0732-183X
9.
Lima
CS
,
Lin
EH
,
Kim
GP
, et al.
Phase II trial of ixabepilone (IXA) plus cetuximab (C) as first-line therapy for advanced pancreatic carcinoma (PC)
.
J Clin Oncol
.
2010
;
•••
:
28
.0732-183X
10.
Philip
PA
,
Benedetti
J
,
Corless
CL
,
Wong
R
,
O’Reilly
EM
,
Flynn
PJ
, et al.
Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: southwest Oncology Group-directed intergroup trial S0205
.
J Clin Oncol
.
2010
Aug
;
28
(
22
):
3605
10
.
[PubMed]
0732-183X
11.
Rembielak
A
,
Saleem
A
,
Valle
J
, et al.
Phase II Study of Cetuximab and Radiotherapy in Patients with Locally Advanced Pancreatic Cancer (PACER Study)
.
Clin Oncol
.
2009
;
21
:
254
5
.0305-7399
12.
Rembielak
AI
,
Jain
P
,
Jackson
AS
,
Green
MM
,
Santorelli
GR
,
Whitfield
GA
, et al.
Phase II Trial of Cetuximab and Conformal Radiotherapy Only in Locally Advanced Pancreatic Cancer with Concurrent Tissue Sampling Feasibility Study
.
Transl Oncol
.
2014
Feb
;
7
(
1
):
55
64
.
[PubMed]
1936-5233
13.
Merchan
JR
,
Ferrell
A
,
Macintyre
J
,
Ciombor
KK
,
Levi
J
,
Ribeiro
A
, et al.
Phase II study of gemcitabine, oxaliplatin, and cetuximab in advanced pancreatic cancer
.
Am J Clin Oncol
.
2012
Oct
;
35
(
5
):
446
50
.
[PubMed]
0277-3732
14.
Mendelsohn
J
.
Targeting the epidermal growth factor receptor for cancer therapy
.
J Clin Oncol
.
2002
Sep
;
20
(
18
Suppl
):
1S
13S
.
[PubMed]
0732-183X
15.
Korc
M
,
Chandrasekar
B
,
Yamanaka
Y
,
Friess
H
,
Buchier
M
,
Beger
HG
.
Overexpression of the epidermal growth factor receptor in human pancreatic cancer is associated with concomitant increases in the levels of epidermal growth factor and transforming growth factor alpha
.
J Clin Invest
.
1992
Oct
;
90
(
4
):
1352
60
.
[PubMed]
0021-9738
16.
Friess
H
,
Wang
L
,
Zhu
Z
,
Gerber
R
,
Schröder
M
,
Fukuda
A
, et al.
Growth factor receptors are differentially expressed in cancers of the papilla of vater and pancreas
.
Ann Surg
.
1999
Dec
;
230
(
6
):
767
74
.
[PubMed]
0003-4932
17.
Tai
CJ
,
Wang
H
,
Wang
CK
,
Tai
CJ
,
Huang
MT
,
Wu
CH
, et al.
Bevacizumab and cetuximab with conventional chemotherapy reduced pancreatic tumor weight in mouse pancreatic cancer xenografts
.
Clin Exp Med
.
2017
May
;
17
(
2
):
141
50
.
[PubMed]
1591-8890
18.
Larbouret
C
,
Robert
B
,
Bascoul-Mollevi
C
,
Penault-Llorca
F
,
Ho-Pun-Cheung
A
,
Morisseau
S
, et al.
Combined cetuximab and trastuzumab are superior to gemcitabine in the treatment of human pancreatic carcinoma xenografts
.
Ann Oncol
.
2010
Jan
;
21
(
1
):
98
103
.
[PubMed]
0923-7534
19.
Zagouri
F
,
Sergentanis
TN
,
Chrysikos
D
,
Zografos
CG
,
Papadimitriou
CA
,
Dimopoulos
MA
, et al.
Molecularly targeted therapies in metastatic pancreatic cancer: a systematic review
.
Pancreas
.
2013
Jul
;
42
(
5
):
760
73
.
[PubMed]
0885-3177
20.
Tian
W
,
Ding
W
,
Kim
S
,
Xu
X
,
Pan
M
,
Chen
S
.
Efficacy and safety profile of combining agents against epidermal growth factor receptor or vascular endothelium growth factor receptor with gemcitabine-based chemotherapy in patients with advanced pancreatic cancer: a meta-analysis
.
Pancreatology
.
2013
Jul-Aug
;
13
(
4
):
415
22
.
[PubMed]
1424-3903
21.
Khan
K
,
Cunningham
D
,
Peckitt
C
,
Barton
S
,
Tait
D
,
Hawkins
M
, et al.
miR-21 expression and clinical outcome in locally advanced pancreatic cancer: exploratory analysis of the pancreatic cancer Erbitux, radiotherapy and UFT (PERU) trial
.
Oncotarget
.
2016
Mar
;
7
(
11
):
12672
81
.
[PubMed]
1949-2553
22.
Burtness
B
,
Powell
M
,
Catalano
P
,
Berlin
J
,
Liles
DK
,
Chapman
AE
, et al.
Randomized Phase II Trial of Irinotecan/Docetaxel or Irinotecan/Docetaxel Plus Cetuximab for Metastatic Pancreatic Cancer: An Eastern Cooperative Oncology Group Study
.
Am J Clin Oncol
.
2016
Aug
;
39
(
4
):
340
5
.
[PubMed]
0277-3732
23.
Moher
D
,
Liberati
A
,
Tetzlaff
J
,
Altman
DG
;
PRISMA Group
.
Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement
.
Int J Surg
.
2010
;
8
(
5
):
336
41
.
[PubMed]
1743-9191
24.
Goossen
K
,
Tenckhoff
S
,
Probst
P
,
Grummich
K
,
Mihaljevic
AL
,
Büchler
MW
, et al.
Optimal literature search for systematic reviews in surgery
.
Langenbecks Arch Surg
.
2018
Feb
;
403
(
1
):
119
29
.
[PubMed]
1435-2443
25.
Higgins
JPT GS
: Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0, The Cochrane Collaboration, March
2011
26.
Buscemi
N
,
Hartling
L
,
Vandermeer
B
,
Tjosvold
L
,
Klassen
TP
.
Single data extraction generated more errors than double data extraction in systematic reviews
.
J Clin Epidemiol
.
2006
Jul
;
59
(
7
):
697
703
.
[PubMed]
0895-4356
27.
Guyatt
GH
,
Oxman
AD
,
Vist
GE
,
Kunz
R
,
Falck-Ytter
Y
,
Alonso-Coello
P
, et al.;
GRADE Working Group
.
GRADE: an emerging consensus on rating quality of evidence and strength of recommendations
.
BMJ
.
2008
Apr
;
336
(
7650
):
924
6
.
[PubMed]
0959-8138
28.
Tierney
JF
,
Stewart
LA
,
Ghersi
D
,
Burdett
S
,
Sydes
MR
.
Practical methods for incorporating summary time-to-event data into meta-analysis
.
Trials
.
2007
Jun
;
8
(
1
):
16
.
[PubMed]
1745-6215
29.
Parmar
MK
,
Torri
V
,
Stewart
L
.
Extracting summary statistics to perform meta-analyses of the published literature for survival endpoints
.
Stat Med
.
1998
Dec
;
17
(
24
):
2815
34
.
[PubMed]
0277-6715
30.
Higgins
JP
,
Thompson
SG
,
Deeks
JJ
,
Altman
DG
.
Measuring inconsistency in meta-analyses
.
BMJ
.
2003
Sep
;
327
(
7414
):
557
60
.
[PubMed]
0959-8138
31.
Cascinu
S
,
Berardi
R
,
Labianca
R
,
Siena
S
,
Falcone
A
,
Aitini
E
, et al.;
Italian Group for the Study of Digestive Tract Cancer (GISCAD)
.
Cetuximab plus gemcitabine and cisplatin compared with gemcitabine and cisplatin alone in patients with advanced pancreatic cancer: a randomised, multicentre, phase II trial
.
Lancet Oncol
.
2008
Jan
;
9
(
1
):
39
44
.
[PubMed]
1470-2045
32.
Kullmann
F
,
Hollerbach
S
,
Dollinger
MM
,
Harder
J
,
Fuchs
M
,
Messmann
H
, et al.
Cetuximab plus gemcitabine/oxaliplatin (GEMOXCET) in first-line metastatic pancreatic cancer: a multicentre phase II study
.
Br J Cancer
.
2009
Apr
;
100
(
7
):
1032
6
.
[PubMed]
0007-0920
33.
Xiong
HQ
,
Rosenberg
A
,
LoBuglio
A
,
Schmidt
W
,
Wolff
RA
,
Deutsch
J
, et al.
Cetuximab, a monoclonal antibody targeting the epidermal growth factor receptor, in combination with gemcitabine for advanced pancreatic cancer: a multicenter phase II Trial
.
J Clin Oncol
.
2004
Jul
;
22
(
13
):
2610
6
.
[PubMed]
0732-183X
34.
Liu
L
,
Cao
Y
,
Tan
A
,
Liao
C
,
Gao
F
.
Cetuximab-based therapy versus non-cetuximab therapy for advanced cancer: a meta-analysis of 17 randomized controlled trials
.
Cancer Chemother Pharmacol
.
2010
Apr
;
65
(
5
):
849
61
.
[PubMed]
0344-5704
35.
Sorich
MJ
,
Wiese
MD
,
Rowland
A
,
Kichenadasse
G
,
McKinnon
RA
,
Karapetis
CS
.
Extended RAS mutations and anti-EGFR monoclonal antibody survival benefit in metastatic colorectal cancer: a meta-analysis of randomized, controlled trials
.
Ann Oncol
.
2015
Jan
;
26
(
1
):
13
21
.
[PubMed]
0923-7534
36.
Dreyer
SB
,
Chang
DK
,
Bailey
P
,
Biankin
AV
.
Pancreatic Cancer Genomes: Implications for Clinical Management and Therapeutic Development
.
Clin Cancer Res
.
2017
Apr
;
23
(
7
):
1638
46
.
[PubMed]
1078-0432
37.
Schultheis
B
,
Reuter
D
,
Ebert
MP
,
Siveke
J
,
Kerkhoff
A
,
Berdel
WE
, et al.
Gemcitabine combined with the monoclonal antibody nimotuzumab is an active first-line regimen in KRAS wildtype patients with locally advanced or metastatic pancreatic cancer: a multicenter, randomized phase IIb study
.
Ann Oncol
.
2017
Oct
;
28
(
10
):
2429
35
.
[PubMed]
0923-7534
38.
Jones
S
,
Zhang
X
,
Parsons
DW
,
Lin
JC
,
Leary
RJ
,
Angenendt
P
, et al.
Core signaling pathways in human pancreatic cancers revealed by global genomic analyses
.
Science
.
2008
Sep
;
321
(
5897
):
1801
6
.
[PubMed]
0036-8075
39.
Löhr
JM
,
Kordes
M
,
Rutkowski
W
, et al.
: Overcoming diagnostic issues in precision treatment of pancreatic cancer Exp Rev Prec Med Drug Devel 3:189-195,
2018
40.
Malgerud
L
,
Lindberg
J
,
Wirta
V
,
Gustafsson-Liljefors
M
,
Karimi
M
,
Moro
CF
, et al.
Bioinformatory-assisted analysis of next-generation sequencing data for precision medicine in pancreatic cancer
.
Mol Oncol
.
2017
Oct
;
11
(
10
):
1413
29
.
[PubMed]
1574-7891
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.