Skip to main content
Erschienen in: BMC Cancer 1/2016

Open Access 01.12.2016 | Research article

The impact of KRAS mutations on prognosis in surgically resected colorectal cancer patients with liver and lung metastases: a retrospective analysis

Erschienen in: BMC Cancer | Ausgabe 1/2016

Abstract

Background

KRAS mutations are common in colorectal cancer (CRC). The role of KRAS mutation status as a prognostic factor remains controversial, and most large population-based cohorts usually consist of patients with non-metastatic CRC. We evaluated the impact of KRAS mutations on the time to recurrence (TTR) and overall survival (OS) in patients with metastatic CRC who underwent curative surgery with perioperative chemotherapy.

Methods

Patients who underwent curative resection for primary and synchronous metastases were retrospectively collected in a single institution during a 6 year period between January 2008 and June 2014. Patients with positive surgical margins, those with known BRAF mutation, or those with an unknown KRAS mutation status were excluded, and a total of 82 cases were identified. The pathological and clinical features were evaluated. Patients’ outcome with KRAS mutation status for TTR and OS were investigated by univariate and multivariate analysis.

Results

KRAS mutations were identified in 37.8 % of the patients and not associated with TTR or OS between KRAS wild type and KRAS mutation cohorts (log-rank p = 0.425 for TTR; log-rank p = 0.137 for OS). When patients were further subdivided into three groups according to mutation subtype (wild-type vs. KRAS codon 12 mutation vs. KRAS codon 13 mutation) or amino acid missense mutation type (G > A vs. G > T vs. G > C), there were no significant differences in TTR or OS. Mutational frequencies were significantly higher in patients with lung metastases compared with those with liver and ovary/bladder metastases (p = 0.039), however, KRAS mutation status was not associated with an increased risk of relapsed in the lung.

Conclusions

KRAS mutation was not associated with TTR or OS in patients with metastatic CRC who underwent curative surgery with perioperative chemotherapy.
Hinweise
Hae Su Kim and Jin Seok Heo are co-first author

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

HSK, JL, JH conceived and designated the study; JYL, ML and SHL helped to conceive the study and revised manuscript critically for important intellectually content; SK reviewed the pathologic specimens; WYL, YAP, YBC and SY critically revised the manuscript; STK, JOP and HYL helped acquisition and interpretation of data; YSC and WIK participated in statistical analysis and interpretation of data; HCK, YSP conceived the study, participated in the design of it and coordination. All authors read and approved the final manuscript.
Abkürzungen
BRAF
v-Raf murine sarcoma viral oncogene homolog B
CIs
Confidence intervals
CRC
Colorectal cancer
EGFR
Epidermal growth factor receptor
KRAS
V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog
OS
Overall survival
TTR
Time to recurrence

Background

Colorectal cancer (CRC) is the fourth leading cause of cancer-related death worldwide [1]. Although the development of molecular-targeted therapy has improved the survival of patients with metastatic CRC [2, 3], the majority of patients with stage IV CRC who undergo complete resection die from metastatic disease. Nevertheless, a good proportion of patients demonstrate good recurrence-free survival. CRC tumorigenesis is characterized by the accumulation of genetic alterations, and V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations are an early event in tumorigenesis [4]. KRAS mutations occur in approximately 30 to 40 % of patients with CRC, and 90 % of KRAS mutations occur in codon 12 or 13 [2, 5, 6]. KRAS mutations lead to constitutive activation of downstream pathways, including the Ras/Raf/MAP/MEK/ERK and/or PTEN/PI3K/Akt pathways [710]. KRAS mutations are established biomarkers for predicting the poor efficacy of anti-epidermal growth factor receptor (EGFR) monoclonal antibodies in patients with stage IV CRC [2, 5, 11], but the prognostic relevance of KRAS mutations remains controversial [1216]. Recent studies, in patients with resected stage II and/or III CRC, have highlighted the prognostic value of KRAS codon12 and 13 mutations, showing correlations between mutation subtype, cancer recurrence, and poor overall survival [1315].
Large population-based cohorts usually consist of patients with non-metastatic CRC [12, 14, 16, 17]. The prognostic impact of KRAS mutation in patients with synchronous metastatic CRC who undergo curative resection with perioperative chemotherapy is unknown. The current study investigated the impact of KRAS mutations on the time to recurrence (TTR) and overall survival (OS) in patients with stage IV CRC who underwent curative surgery with perioperative chemotherapy. In addition, the recurrence pattern according to KRAS mutation status after complete resection was evaluated.

Methods

Patients

In this retrospective study, patients who underwent curative resection for primary and synchronous metastases at our institution between January 2008 and June 2014 were identified from the hospital records. Patients who underwent separate colorectal resection and metastasectomy were excluded if the duration between the two procedures exceeded 2 months. Patients with positive surgical margins, those with known v-Raf murine sarcoma viral oncogene homolog B (BRAF) mutations, or those with an unknown KRAS mutation status were also excluded. All patients included in the study were administered 5-FU with/without oxaliplatin or irinotecan-based chemotherapy. Clinical and pathological data including sex, patient age, tumor location, resection site, staging at surgery (performed in accordance with the classification of the 6th Edition of the American Joint Committee on Cancer guidelines), BRAF mutation status, perioperative chemotherapy regimens, use of molecular targeting agents including cetuximab and bevacizumab, were collected. The study protocol was reviewed and approved by the SMC institutional review board.

Perioperative chemotherapy regimens

Oxaliplatin based chemotherapy was FOLFOX (oxaliplatin 85 mg/m2 on day 1, infused during 2 h; LV 200 mg/m2, infused during 2 h, followed by 5-FU as a 400 mg/m2 intravenous bolus then a 1200 mg/m2 infusion during 22 h on days 1 and 2) in 2 week treatment cycles or XELOX(oxaliplatin 130 mg/m2 on day 1 followed by oral capecitabine 1000 mg/m2 twice daily (day 1 to 14) in 3 week treatment cycles. Irinotecan based chemotherapy was FORFIRI (irinotecan 180 mg/m2 on day 1, infused during 2 h; LV 200 mg/m2, infused during 2 h, followed by 5-FU as a 400 mg/m2 intravenous bolus then a 1200 mg/m2 infusion during 22 h on days 1 and 2) in 2 week treatment cycles or XELIRI (irinotecan 250 mg/m2 on day 1 followed by oral capecitabine 1000 mg/m2 twice daily (day 1 to 14) in 3 week treatment cycles. If bevacizumab or cetuximab was used, patients received cetuximab (initial dose 400 mg/m2 infused during 2 h, and 250 mg/m2 weekly) or bevacizumab (5 mg/kg) followed by FOLFOX or FOLFIRI.

DNA extraction and mutation analysis

DNA was isolated from 10-μm formalin-fixed, paraffin-embedded tumor specimens using FFPE-DNA isolation kit (Qiagen, Hilden, Germany). A Qiagen the rascreen KRAS mutation kit was used to detect the seven most common KRAS codon 12 and 13 mutations. Specifically, the mutation was detected by real-time polymerase chain reaction based on amplification-refractory mutation system and Scorpion probes (Gly12Asp [GGT > GAT] G12D, Gly12Val [GGT > GAC] G12V, Gly12Cys [GGT > TGT] G12C, Gly12Ser [GGT > AGT] G12S, Gly12Ala [GGT > GCT] G12A, Gly12Arg [GGT > CGT] G12R, Gly13Asp [GGC > GAC] G13D).

Statistical analyses

Patients were subdivided into wild-type KRAS and mutant KRAS cohorts. The primary objective was to investigate the effect of KRAS mutation on the TTR. TTR was defined as the time from the date of operation to the date of local or metastatic recurrence. As of November 2014, overall survival data are not yet available for the mutant KRAS group. Data from recurrence-free patients were censored at the date of the last follow-up.
To compare baseline characteristics, categorical outcomes were analyzed using the chi-square test or Fisher’s exact test. Continuous variables are presented as medians and ranges. TTR and OS were calculated using the Kaplan-Meier method, and data was compared using the log-rank test. The Cox proportional hazard model was used to assess hazard ratios (HRs) of prognostic factor. All factors of statistical significance (p < 0.10) in univariate analysis were included in the multivariate analysis. Two-sided p values of <0.05 were considered as statistically significant. All statistical analyses were performed using the SPSS statistical software version 21 (IBM, Armonk, NY. USA).

Results

Patient characteristics

Between January 2008 and June 2014, 82 patients who were diagnosed with synchronous metastatic CRC and underwent curative resection of primary and metastatic lesions with perioperative chemotherapy were included in the analyses. Table 1 summarizes the patient characteristics according to KRAS mutation status. There was no significant difference in clinicopathologic features between the two groups. Baseline characteristics including age, sex, tumor location, tumor grade, T stage, N stage, synchronous metastasectomy site, and recurrence site were similar between the KRAS wild type and KRAS mutation cohorts. Regarding BRAF mutation status, all of the tested cases (76.8 %) were BRAF wild type.
Table 1
Baseline characteristics according to KRAS mutation status
Characteristics
No. of patients
KRAS
wild-type
mutant
p-value
(n = 82)
(n = 51)
(n = 31)
Age, year, Median (range)
55.8 (25–77)
58.8 (25–77)
55.5 (29–77)
0.565
≥65 years
17 (21 %)
12 (24 %)
5 (16 %)
0.423
Sex
   
0.867
 Male
44 (54 %)
27 (53 %)
17 (55 %)
 Female
38 (46 %)
24 (47 %)
14 (45 %)
Location
   
0.246
 Colon
54 (66 %)
36 (71 %)
18 (58 %)
 Rectum
28 (34 %)
15 (29 %)
13 (42 %)
Neoadjuvant Chemotherapy
21 (26 %)
11 (22 %)
10 (32 %)
0.282
Resection site
   
0.039
 Liver
57 (69 %)
39 (76 %)
18 (58 %)
 Lung
13 (16 %)
4 (8 %)
9 (29 %)
 Others (ovary, bladder)
12 (15 %)
8 (16 %)
4 (13 %)
Tumor grade
   
0.432
 Well
10 (12 %)
7 (14 %)
3 (10 %)
 Moderate/Poor
72 (78 %)
44 (86 %)
28 (90 %)
T stage
   
0.265
 T1
1 (1 %)
1 (2 %)
0 (0 %)
 T2
2 (2 %)
2 (4 %)
0 (0 %)
 T3
47 (57 %)
30 (59 %)
17 (55 %)
 T4
30 (37 %)
18 (35 %)
12 (39 %)
 Tx
2 (2 %)
0 (0 %)
2 (6 %)
N stage
   
0.824
 N0
12 (15 %)
8 (16 %)
4 (13 %)
 N1
31 (38 %)
18 (35 %)
13 (42 %)
 N2
39 (47 %)
25 (49 %)
14 (45 %)
1st Adjuvant Chemo-Regimen
   
0.923
 Oxaliplatin-based
70 (86 %)
44 (86 %)
26 (84 %)
 Irinotecan-based
10 (12 %)
6 (12 %)
4 (13 %)
 Only 5-FU
2 (2 %)
1 (2 %)
1 (3 %)
Use of Cetuximab at 1st post-operative chemotherapy
4 (5 %)
4 (8 %)
0 (0 %)
NA
Use of Becavizumab at 1st post-operativechemotherapy
13 (16 %)
6 (12 %)
7 (23 %)
0.194
Ever use of Cetuximab
16 (20 %)
16 (31 %)
0 (0 %)
NA
Ever use of Bevacizumab
23 (28 %)
10 (20 %)
13 (42 %)
0.029
Recurrence pattern (n = 57)
   
0.616
 Primary site
3 (5 %)
1 (2 %)
2 (8 %)
 Metastasectomy site
27 (47 %)
15 (46 %)
12 (50 %)
 New distant sites
27 (47 %)
17 (52 %)
10 (42 %)
Duration of follow up month, median (range)
25 (4–74)
25 (4–74)
34 (9–63)
0.763
Abbreviations: CI confidence interval, A.A amino acid

Subtype of KRAS mutations

Of 82 patients, KRAS mutations were detected in 31 (37.8 %) patients. Eighteen (58 %) patients harbored codon 12 mutations including 9 with c.35G > A (p.G12D, codon 12 GGT > GAT), 5 with c.35G > T (pG12V, codon 12 GGT > GTT), 2 with c.35G > C (p.G12A, codon 12 GGT > GCT), and 2 with c.34G > A (p.G12S, codon 12 GGT > AGT). For the 13 (42 %) patients with codon 13 mutations, all had the c.38G > A (p.G13D, codon 13 GGC > GAC) mutation. KRAS amino acid mutations were also analyzed. The G > A missense mutation was the most frequently observed mutation, followed by the G > T and G > C mutations.

The impact of KRAS mutations on TTR and OS

The median follow-up durations were 25 months (range, 4–74) and 34 months (range, 9–63) for patients with KRAS wild type and KRAS mutation status, respectively. During follow-up in surviving participants, there were 57 events for TTR analysis and 25 events for OS analysis. There were no significant differences in survival time distributions according to KRAS wild type and KRAS mutation status (log-rank p = 0.425 for TTR; log-rank p = 0.137 for OS, Fig. 1). In univariate and multivariate analyses, there were no significant differences in TTR or OS between KRAS wild type and KRAS mutation cohorts (Tables 2, 3 and 4). When patients were further subdivided into three groups according to mutation subtype (wild-type vs. KRAS codon 12 mutation vs. KRAS codon 13 mutation) or amino acid missense mutation type (G > A vs. G > T vs. G > C), there were no significant differences in TTR or OS.
Table 2
Univariate analysis for time to recurrence
Characteristics
Hazard ratio (95 % CI)
p-value
Location of primary tumor (rectum vs colon)
0.956 (0.548–1.669)
0.875
Age (≥65 vs <65)
0.856 (0.418–1.755)
0.671
Sex (female vs male)
0.678 (0.399–1.150)
0.150
Neoadjuvant chemotherapy (Yes vs No)
1.040 (0.563–1.923)
0.899
Tumor grade (moderate/poor vs well)
1.201 (0.508–2.843)
0.676
T stage (T4 vs T1-3)
1.041 (0.608–1.782)
0.885
N stage (N2 vs N0,1)
1.197 (0.703–2.037)
0.508
Resection site
  
 Liver
1
 
 Lung
0.694 (0.311–1.550)
0.373
 Others (ovary, uterus, bladder)
0.670 (0.299–1.502)
0.331
Use of Cetuximab at 1st post-operative chemotherapy (Yes vs No)
0.589 (0.143–2.425)
0.463
Use of Bevacizumab at 1st post-operative chemotherapy (Yes vs No)
0.582 (0.231–1.469)
0.252
KRAS (mutation vs wild)
1.245 (0.725–2.137)
1.245
KRAS subtype
  
 Wild
1
 
 12th
1.127 (0.599–2.123)
0.710
 13th
1.230 (0.561–2.697)
0.605
A.A Mutation type
  
 Wild (n = 51)
1
 
 Guanine to thymidine (n = 5)
0.737 (0.164–3.315)
0.691
 Guanine to cytosine (n = 2)
1.482 (0.766–2.864)
0.242
 Guanine to adenine (n = 24)
1.029 (0.553–1.931)
0.928
Abbreviations: CI confidence interval, A.A amino acid, HR hazard ratio
Table 3
Univariate analysis for overall survival
Characteristics
HR (95 % CI)
p-value
Location of primary tumor (rectum vs colon)
0.531 (0.212–1.333)
0.178
Age (≥65 vs <65)
7.492 (2.941–9.084)
<0.001
Sex (female vs male)
2.038 (0.908–4.578)
0.085
Neoadjuvant chemotherapy (Yes vs No)
1.114 (0.460–2.698)
0.811
Tumor grade (moderate/poor vs well)
1.332 (0.312–5.693)
0.698
T stage (T4 vs T1-3)
4.324 (1.857–10.068)
0.001
N stage (N2 vs N0,1)
1.906 (0.854–4.251)
0.115
Resection site
  
 Liver
1
 
 Lung
0.311 (0.041–2.335)
0.256
 Others (ovary, uterus, bladder)
1.036 (0.345–3.108)
0.950
Use of Cetuximab at 1st post-operative chemotherapy (Yes vs No)
3.777 (0.850–16.779)
0.081
Use of Bevacizumab at 1st post-operative chemotherapy (Yes vs No)
0.899 (0.267–3.027)
0.863
KRAS (mutation vs mutation)
0.500(0.198–1.267)
0.144
KRAS
  
 Wild
1
 
 12th
0.330 (0.076–1.428)
0.138
 13th
0.675 (0.227–2.010)
0.481
Abbreviations: CI confidence interval, A.A amino acid, HR hazard ratio
Factors of statistical significance (p < 0.10) in univariate analysis presented with boldface
Table 4
Multivariate analysis for overall survival
Characteristics
HR (95 % CI)
p-value
Age (≥65 vs <65)
9.749 (3.404–27.919)
<0.001
Sex (female vs male)
3.070 (1.260–7.478)
0.014
T stage (T4 vs T1-3)
3.511 (1.484–8.307)
0.004
Use of Cetuximab at 1st post-operative chemotherapy (Yes vs No)
1.185 (0.235–5.979)
0.837
Abbreviations: CI confidence interval A.A amino acid; HR, hazard ratio

The effect of KRAS mutation status on the recurrence site

Mutational frequencies were significantly higher in patients with lung metastases compared with those with liver and ovary/bladder metastases (KRAS mutant: lung 9/13 [69 %], liver 18/57 [31 %], ovary/bladder 4/12 [33 %]; p = 0.039). However, KRAS mutation status was not associated with an increased risk of relapse in the lung, and the majority of recurrence occurred at the previous metastasectomy sites (15/33 vs. 24/31 for KRAS wild type vs. KRAS mutation, respectively).

Discussion

The majority of studies evaluating the prognostic impact of KRAS mutational status in CRC have been conducted in patients with stage II/III disease. The QUASAR trial, which mainly evaluated patients with stage II CRC, revealed that KRAS mutations had a detrimental effect on recurrence and OS, despite adjuvant chemotherapy [17]. In contrast, the CALGB 89803 and PETACC-3 trials demonstrated that KRAS mutation status had no significant effect on recurrence or OS in patients with stage II/III colon cancer or CRC treated with adjuvant chemotherapy [12, 16]. However, conflicting findings were reported simultaneously in two large studies conducted by The Kirsten ras in-colorectal-cancer collaborative group, the RASCAL and RASCAL II trials, which were comprised of 2721 and 4268 patients, respectively [18, 19]. Although the first RASCAL study reported an association of KRAS mutations with an increased risk of recurrence and death for patients with all stages of CRC, recurrence in patients with Dukes’ D tumors was less than might be expected. The RASCAL II study concluded that there was a significant prognostic value in failure-free survival alone in patients with Dukes’ C cancer harboring a KRAS G12V mutation.
Few studies have evaluated the relationship between patients with stage IV disease at the time of diagnosis and KRAS mutations [2023]. Patients with metastatic CRC with limited metastases undergo curative primary resection with or without metastasectomy, anti-EGFR antibody therapy, and heterogeneous chemotherapy regimens, making it difficult to evaluate the precise prognostic value of KRAS status in this setting. To overcome this limitation, in this study, we included only patients who underwent curative resection of the primary and metastatic sites who received perioperative chemotherapy. To our knowledge, this study is the first to report TTR in such patients. In this homogenous cohort of Korean patients with metastatic CRC, we observed that KRAS mutation was not associated with TTR or OS, which is congruent with previous studies [2022]. Phipps et al., reported that KRAS mutations did not differ by stage at diagnosis, and that the prognostic value of KRAS mutations only became evident in patients with stage I-III disease [22]. Furthermore, Nash et al., reported that the prevalence of KRAS mutations did not vary with stage, but that KRAS mutations were strong independent predictors of survival for patients with stage I-III CRC [21].
We also investigated the association KRAS mutations with recurrence pattern in our cohort. KRAS mutations were significantly more common in lung metastases compared with liver and bladder/ovary metastases. These finding were concordant with those of Tie et al., who observed a significantly higher prevalence of KRAS mutations in patients with lung metastases compared with those with liver metastases [24]. In addition, in their study, KRAS mutations were associated with an increased risk of lung relapse in patients with stage II/III CRC who were enrolled on the VICTOR clinical trial [21]. However, in the present study, we did not observe recurrence-specific associations with KRAS mutation status. The differential impact of KRAS mutations on recurrence-specific sites according to disease stage requires evaluation in further studies.
Limitations of the present study included the relatively short follow-up, where the median OS was not reached in the KRAS mutation group. Nevertheless, sufficient TTR events occured enabling analysis of recurrence. In addition, the BRAF mutation status was not determined for 19 (33 %) patients, but BRAF mutations were only detected in a small proportion of patient and were not significantly different between KRAS wild type and KRAS mutated patients. In addition, the small sample size did not allow us to evaluate the impact of different KRAS mutation subtypes.
In conclusion, KRAS mutation was not associated with TTR or OS in curatively resected, metastatic CRC. Further validation of these finding is needed in metastatic CRC patients treated with curative resection in prospective controlled trials.

Conclusions

The present study, to our knowledge, is the first report on the effect of KRAS mutations on prognosis in surgically treated CRC patients with synchronous metastases. The most of previous studies evaluating the prognostic impact of KRAS in CRC have been conducted in patients with non-metastatic CRC, and the influence of KRAS mutations on outcome is conflicting. In our study, KRAS mutation was not associated with TTR or OS in metastatic CRC patients who undergo curative surgery and perioperative chemotherapy. KRAS mutation status was also not linked to recurrence pattern. Prospective studies will be necessary to evaluate the prognostic effect of KRAS mutation in metastatic CRC patients.
This research is strictly retrospective and involving the collection of existing data and records. The study protocol was reviewed and approved consent exemptions by the SMC institutional review board.

Acknowledgements

This work was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HI14C2750, and HI14C3418).
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. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

HSK, JL, JH conceived and designated the study; JYL, ML and SHL helped to conceive the study and revised manuscript critically for important intellectually content; SK reviewed the pathologic specimens; WYL, YAP, YBC and SY critically revised the manuscript; STK, JOP and HYL helped acquisition and interpretation of data; YSC and WIK participated in statistical analysis and interpretation of data; HCK, YSP conceived the study, participated in the design of it and coordination. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Siegel R, Ward E, Brawley O, Jemal A. Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin. 2011;61:212–36.CrossRefPubMed Siegel R, Ward E, Brawley O, Jemal A. Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin. 2011;61:212–36.CrossRefPubMed
2.
Zurück zum Zitat Karapetis CS, Khambata-Ford S, Jonker DJ, O’Callaghan CJ, Tu D, Tebbutt NC, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 2008;359:1757–65.CrossRefPubMed Karapetis CS, Khambata-Ford S, Jonker DJ, O’Callaghan CJ, Tu D, Tebbutt NC, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 2008;359:1757–65.CrossRefPubMed
3.
Zurück zum Zitat Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350:2335–42.CrossRefPubMed Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350:2335–42.CrossRefPubMed
4.
Zurück zum Zitat Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC, Leppert M, et al. Genetic alterations during colorectal-tumor development. N Engl J Med. 1988;319:525–32.CrossRefPubMed Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC, Leppert M, et al. Genetic alterations during colorectal-tumor development. N Engl J Med. 1988;319:525–32.CrossRefPubMed
5.
Zurück zum Zitat Van Cutsem E, Kohne CH, Hitre E, Zaluski J, Chang Chien CR, Makhson A, et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 2009;360:1408–17.CrossRefPubMed Van Cutsem E, Kohne CH, Hitre E, Zaluski J, Chang Chien CR, Makhson A, et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 2009;360:1408–17.CrossRefPubMed
6.
Zurück zum Zitat Nosho K, Irahara N, Shima K, Kure S, Kirkner GJ, Schernhammer ES, et al. Comprehensive biostatistical analysis of CpG island methylator phenotype in colorectal cancer using a large population-based sample. PLoS One. 2008;3:e3698.CrossRefPubMedPubMedCentral Nosho K, Irahara N, Shima K, Kure S, Kirkner GJ, Schernhammer ES, et al. Comprehensive biostatistical analysis of CpG island methylator phenotype in colorectal cancer using a large population-based sample. PLoS One. 2008;3:e3698.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Bos JL, Fearon ER, Hamilton SR, Verlaan-de Vries M, van Boom JH, van der Eb AJ, et al. Prevalence of ras gene mutations in human colorectal cancers. Nature. 1987;327:293–7.CrossRefPubMed Bos JL, Fearon ER, Hamilton SR, Verlaan-de Vries M, van Boom JH, van der Eb AJ, et al. Prevalence of ras gene mutations in human colorectal cancers. Nature. 1987;327:293–7.CrossRefPubMed
8.
Zurück zum Zitat Di Fiore F, Blanchard F, Charbonnier F, Le Pessot F, Lamy A, Galais MP, et al. Clinical relevance of KRAS mutation detection in metastatic colorectal cancer treated by Cetuximab plus chemotherapy. Br J Cancer. 2007;96:1166–9.CrossRefPubMedPubMedCentral Di Fiore F, Blanchard F, Charbonnier F, Le Pessot F, Lamy A, Galais MP, et al. Clinical relevance of KRAS mutation detection in metastatic colorectal cancer treated by Cetuximab plus chemotherapy. Br J Cancer. 2007;96:1166–9.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Benvenuti S, Sartore-Bianchi A, Di Nicolantonio F, Zanon C, Moroni M, Veronese S, et al. Oncogenic activation of the RAS/RAF signaling pathway impairs the response of metastatic colorectal cancers to anti-epidermal growth factor receptor antibody therapies. Cancer Res. 2007;67:2643–8.CrossRefPubMed Benvenuti S, Sartore-Bianchi A, Di Nicolantonio F, Zanon C, Moroni M, Veronese S, et al. Oncogenic activation of the RAS/RAF signaling pathway impairs the response of metastatic colorectal cancers to anti-epidermal growth factor receptor antibody therapies. Cancer Res. 2007;67:2643–8.CrossRefPubMed
10.
Zurück zum Zitat Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell. 2004;116:855–67.CrossRefPubMed Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell. 2004;116:855–67.CrossRefPubMed
11.
Zurück zum Zitat Bokemeyer C, Bondarenko I, Makhson A, Hartmann JT, Aparicio J, de Braud F, et al. Fluorouracil, leucovorin, and oxaliplatin with and without cetuximab in the first-line treatment of metastatic colorectal cancer. J Clin Oncol. 2009;27:663–71.CrossRefPubMed Bokemeyer C, Bondarenko I, Makhson A, Hartmann JT, Aparicio J, de Braud F, et al. Fluorouracil, leucovorin, and oxaliplatin with and without cetuximab in the first-line treatment of metastatic colorectal cancer. J Clin Oncol. 2009;27:663–71.CrossRefPubMed
12.
Zurück zum Zitat Roth AD, Tejpar S, Delorenzi M, Yan P, Fiocca R, Klingbiel D, et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60–00 trial. J Clin Oncol. 2010;28:466–74.CrossRefPubMed Roth AD, Tejpar S, Delorenzi M, Yan P, Fiocca R, Klingbiel D, et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60–00 trial. J Clin Oncol. 2010;28:466–74.CrossRefPubMed
13.
Zurück zum Zitat Imamura Y, Morikawa T, Liao X, Lochhead P, Kuchiba A, Yamauchi M, et al. Specific mutations in KRAS codons 12 and 13, and patient prognosis in 1075 BRAF wild-type colorectal cancers. Clin Cancer Res. 2012;18:4753–63.CrossRefPubMedPubMedCentral Imamura Y, Morikawa T, Liao X, Lochhead P, Kuchiba A, Yamauchi M, et al. Specific mutations in KRAS codons 12 and 13, and patient prognosis in 1075 BRAF wild-type colorectal cancers. Clin Cancer Res. 2012;18:4753–63.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Blons H, Emile JF, Le Malicot K, Julie C, Zaanan A, Tabernero J, et al. Prognostic value of KRAS mutations in stage III colon cancer: post hoc analysis of the PETACC8 phase III trial dataset. Ann Oncol. 2014;25:2378–85.CrossRefPubMed Blons H, Emile JF, Le Malicot K, Julie C, Zaanan A, Tabernero J, et al. Prognostic value of KRAS mutations in stage III colon cancer: post hoc analysis of the PETACC8 phase III trial dataset. Ann Oncol. 2014;25:2378–85.CrossRefPubMed
15.
Zurück zum Zitat Lee DW, Kim KJ, Han SW, Lee HJ, Rhee YY, Bae JM, et al. KRAS Mutation is Associated with Worse Prognosis in Stage III or High-risk Stage II Colon Cancer Patients Treated with Adjuvant FOLFOX. Ann Surg Oncol. 2015;22:187–94.CrossRefPubMed Lee DW, Kim KJ, Han SW, Lee HJ, Rhee YY, Bae JM, et al. KRAS Mutation is Associated with Worse Prognosis in Stage III or High-risk Stage II Colon Cancer Patients Treated with Adjuvant FOLFOX. Ann Surg Oncol. 2015;22:187–94.CrossRefPubMed
16.
Zurück zum Zitat Ogino S, Meyerhardt JA, Irahara N, Niedzwiecki D, Hollis D, Saltz LB, et al. KRAS mutation in stage III colon cancer and clinical outcome following intergroup trial CALGB 89803. Clin Cancer Res. 2009;15:7322–9.CrossRefPubMedPubMedCentral Ogino S, Meyerhardt JA, Irahara N, Niedzwiecki D, Hollis D, Saltz LB, et al. KRAS mutation in stage III colon cancer and clinical outcome following intergroup trial CALGB 89803. Clin Cancer Res. 2009;15:7322–9.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Hutchins G, Southward K, Handley K, Magill L, Beaumont C, Stahlschmidt J, et al. Value of mismatch repair, KRAS, and BRAF mutations in predicting recurrence and benefits from chemotherapy in colorectal cancer. J Clin Oncol. 2011;29:1261–70.CrossRefPubMed Hutchins G, Southward K, Handley K, Magill L, Beaumont C, Stahlschmidt J, et al. Value of mismatch repair, KRAS, and BRAF mutations in predicting recurrence and benefits from chemotherapy in colorectal cancer. J Clin Oncol. 2011;29:1261–70.CrossRefPubMed
18.
Zurück zum Zitat Andreyev HJ, Norman AR, Cunningham D, Oates JR, Clarke PA. Kirsten ras mutations in patients with colorectal cancer: the multicenter “RASCAL” study. J Natl Cancer Inst. 1998;90:675–84.CrossRefPubMed Andreyev HJ, Norman AR, Cunningham D, Oates JR, Clarke PA. Kirsten ras mutations in patients with colorectal cancer: the multicenter “RASCAL” study. J Natl Cancer Inst. 1998;90:675–84.CrossRefPubMed
19.
Zurück zum Zitat Andreyev HJ, Norman AR, Cunningham D, Oates J, Dix BR, Iacopetta BJ, et al. Kirsten ras mutations in patients with colorectal cancer: the ‘RASCAL II’ study. Br J Cancer. 2001;85:692–6.CrossRefPubMedPubMedCentral Andreyev HJ, Norman AR, Cunningham D, Oates J, Dix BR, Iacopetta BJ, et al. Kirsten ras mutations in patients with colorectal cancer: the ‘RASCAL II’ study. Br J Cancer. 2001;85:692–6.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Inoue Y, Saigusa S, Iwata T, Okugawa Y, Toiyama Y, Tanaka K, et al. The prognostic value of KRAS mutations in patients with colorectal cancer. Oncol Rep. 2012;28:1579–84.CrossRefPubMed Inoue Y, Saigusa S, Iwata T, Okugawa Y, Toiyama Y, Tanaka K, et al. The prognostic value of KRAS mutations in patients with colorectal cancer. Oncol Rep. 2012;28:1579–84.CrossRefPubMed
21.
Zurück zum Zitat Nash GM, Gimbel M, Cohen AM, Zeng ZS, Ndubuisi MI, Nathanson DR, et al. KRAS mutation and microsatellite instability: two genetic markers of early tumor development that influence the prognosis of colorectal cancer. Ann Surg Oncol. 2010;17:416–24.CrossRefPubMed Nash GM, Gimbel M, Cohen AM, Zeng ZS, Ndubuisi MI, Nathanson DR, et al. KRAS mutation and microsatellite instability: two genetic markers of early tumor development that influence the prognosis of colorectal cancer. Ann Surg Oncol. 2010;17:416–24.CrossRefPubMed
22.
Zurück zum Zitat Phipps AI, Buchanan DD, Makar KW, Win AK, Baron JA, Lindor NM, et al. KRAS-mutation status in relation to colorectal cancer survival: the joint impact of correlated tumour markers. Br J Cancer. 2013;108:1757–64.CrossRefPubMedPubMedCentral Phipps AI, Buchanan DD, Makar KW, Win AK, Baron JA, Lindor NM, et al. KRAS-mutation status in relation to colorectal cancer survival: the joint impact of correlated tumour markers. Br J Cancer. 2013;108:1757–64.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Perez-Ruiz E, Rueda A, Pereda T, Alcaide J, Bautista D, Rivas-Ruiz F, et al. Involvement of K-RAS mutations and amino acid substitutions in the survival of metastatic colorectal cancer patients. Tumour Biol. 2012;33:1829–35.CrossRefPubMed Perez-Ruiz E, Rueda A, Pereda T, Alcaide J, Bautista D, Rivas-Ruiz F, et al. Involvement of K-RAS mutations and amino acid substitutions in the survival of metastatic colorectal cancer patients. Tumour Biol. 2012;33:1829–35.CrossRefPubMed
24.
Zurück zum Zitat Tie J, Lipton L, Desai J, Gibbs P, Jorissen RN, Christie M, et al. KRAS mutation is associated with lung metastasis in patients with curatively resected colorectal cancer. Clin Cancer Res. 2011;17:1122–30.CrossRefPubMed Tie J, Lipton L, Desai J, Gibbs P, Jorissen RN, Christie M, et al. KRAS mutation is associated with lung metastasis in patients with curatively resected colorectal cancer. Clin Cancer Res. 2011;17:1122–30.CrossRefPubMed
Metadaten
Titel
The impact of KRAS mutations on prognosis in surgically resected colorectal cancer patients with liver and lung metastases: a retrospective analysis
Publikationsdatum
01.12.2016
Erschienen in
BMC Cancer / Ausgabe 1/2016
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-016-2141-4

Weitere Artikel der Ausgabe 1/2016

BMC Cancer 1/2016 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.