Prognostic value of KRAS/TP53/PIK3CA in non-small cell lung cancer.
Journal: 2019/March - Oncology Letters
ISSN: 1792-1074
Abstract:
The present study explored the association between KRAS proto-oncogene GTPase (KRAS), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA) and tumor protein p53 (TP53) mutations, and the clinical features and survival prognosis in 50 patients with non-small cell lung cancer (NSCLC). The most common concurrent single gene mutation was TP53, followed by KRAS and PIK3CA. Co-existing mutations were found in 17 patients. KRAS, PIK3CA and TP53 mutations were associated with carbohydrate antigen 19-9 expression, invasive growth, vacuolar signs and margin lobulation on chest CT. The incidence of distant metastasis (bone and adrenal) with KRAS and TP53 mutations was greater than that of local metastasis (pleura). Patients with the wild-type genes experienced longer progression-free survival (PFS) times than those with KRAS, TP53, KRAS/TP53 or PIK3CA/TP53 mutations. Patients with KRAS/TP53 or PIK3CA/TP53 mutations experienced shorter PFS times than those with a single KRAS or TP53 mutation. KRAS, PIK3CA and TP53 mutations were associated with distant metastases and a poor prognosis. Patients with NSCLC should receive routine KRAS, PIK3CA and TP53 gene sequencing to determine mutations for the analysis of clinical characteristics and prognosis.
Relations:
Content
Citations
(5)
References
(33)
Genes
(3)
Processes
(2)
Anatomy
(2)
Affiliates
(1)
Similar articles
Articles by the same authors
Discussion board
Oncology Letters. Feb/28/2019; 17(3): 3233-3240
Published online Feb/3/2019

Prognostic value of KRAS/TP53/PIK3CA in non-small cell lung cancer

Abstract

The present study explored the association between KRAS proto-oncogene GTPase (KRAS), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA) and tumor protein p53 (TP53) mutations, and the clinical features and survival prognosis in 50 patients with non-small cell lung cancer (NSCLC). The most common concurrent single gene mutation was TP53, followed by KRAS and PIK3CA. Co-existing mutations were found in 17 patients. KRAS, PIK3CA and TP53 mutations were associated with carbohydrate antigen 19-9 expression, invasive growth, vacuolar signs and margin lobulation on chest CT. The incidence of distant metastasis (bone and adrenal) with KRAS and TP53 mutations was greater than that of local metastasis (pleura). Patients with the wild-type genes experienced longer progression-free survival (PFS) times than those with KRAS, TP53, KRAS/TP53 or PIK3CA/TP53 mutations. Patients with KRAS/TP53 or PIK3CA/TP53 mutations experienced shorter PFS times than those with a single KRAS or TP53 mutation. KRAS, PIK3CA and TP53 mutations were associated with distant metastases and a poor prognosis. Patients with NSCLC should receive routine KRAS, PIK3CA and TP53 gene sequencing to determine mutations for the analysis of clinical characteristics and prognosis.

Introduction

Lung cancer has a high mortality rate of ~27% and is becoming more prevalent in younger populations (1). Despite progress in the diagnosis and treatment of lung cancer, the 5-year survival rate is only 16% (2). Individualized therapy is a promising treatment strategy for non-small cell lung cancer (3). Mutations in epidermal growth factor receptor (EGFR) drive the development of lung adenocarcinoma and have altered the traditional treatment approaches. Next-generation sequencing revealed that patients with wild-type EGFR or ALK could present concurrent oncogenic mutations in KRAS proto-oncogene GTPase (KRAS) (4), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA) (5) and tumor protein p53 (TP53) (6). These mutations may result in differential clinical features, treatment outcomes and survival prognoses. The association between KRAS, PIK3CA and TP53 mutations, clinical features, and the prognosis of patients with NSCLC is unclear. The present study retrospectively analyzed 89 cases of NSCLC patients with KRAS, PIK3CA and TP53 mutations to elucidate the association between gene mutation, clinical characteristics and survival prognosis as a basis for individualized treatment.

Patients and methods

Patient selection

A total of 122 patients accepted next-generation sequencing for advanced NSCLC at Shanghai Changhai Hospital (Shanghai, China) and were enrolled between January 2015 and December 2016. Missing information and loss to follow-up resulted in the exclusion of 33 patients. Blood samples and clinical data from 89 patients with identified genes were collected, including sex, age, smoking status, symptoms, laboratory test results, chest computed tomography (CT) results, tumor location, pathological type, Tumor-Node-Metastasis stage (7) and site of metastasis. Among the 89 samples, 50 exhibited KRAS, TP53 and PIK3CA mutations. The Ethics Committee of Shanghai Changhai Hospital approved the present study, and written informed consent was obtained from each participant.

Gene sequencing

Circulating Single-Molecule Amplification and Resequencing Technology (cSMART; Illumina CN500; Berry Genomics Co., Ltd., Beijing, China) was used to detect KRAS, PIK3CA and TP53 mutation in all patients with NSCLC. In brief, genomic DNA was extracted from the plasma of the patients using MagMAX Cell-Free DNA Isolation kit, (Thermo Fisher Scientific, Inc., Waltham, MA, USA; Article no. A29319) DNA was purified using a DNA purification kit (Berry Genomics Co., Ltd; Article no. R0037). The libraries were prepared from 10 ng plasma DNA by ligation of universal sequencing adaptors containing unique 6-bp barcodes. Modified DNA was denatured and single strands were circularized by Taq ligase. Bidirectional back-to-back primers, in either singleplex or multiplex format, were annealed close to the mutation loci. Inverse PCR was performed to replicate targeted genes. Amplified products were subjected to massive parallel sequencing on the MiSeq platform (Illumina, Inc., San Diego, CA, USA) to generate paired-end reads of 2×200 bp (8).

Treatment

All patients were administered with a first-line chemotherapy regimen of pemetrexed (500 mg/m2)/paclitaxel (135 mg/m2) and carboplatin (area under the curve=5). All patients provided written informed consent.

Survival analysis

Tumors were evaluated every 2 cycles during chemotherapy treatment or earlier when significant signs of progression, including aggravation of cough or hemoptysis, were present. Progression-free survival (PFS) was determined according to the Response Evaluation Criteria in Solid Tumors guidelines (version 1.1) (9). The PFS time was defined as the time from the beginning of chemotherapy to the presence of objective evidence of progression. The final follow-up date was June 30, 2017.

Statistical analysis

Survival curves were calculated using the Kaplan-Meier method from the beginning of chemotherapy to documented progression or mortality from any cause, differences in PFS were assessed using the log-rank test. Statistical analysis was performed with SPSS version 21 (IBM, Corp., Armonk, NY, USA). The χ2 test was used to compare the categorical variables. P<0.05 was considered to indicate a statistically significant difference.

Results

Patient characteristics

A total of 122 patients with NSCLC received cSMART sequencing and 33 patients were excluded due to missing information or loss to follow-up. A total of 89 patients were therefore enrolled in the present study, and the baseline demographic characteristics are shown in Table I. The study cohort consisted of 52 males and 37 females, with a median age of 61.0 years and a mean (± standard error) age of 59.4 (±12.2) years. Adenocarcinoma was histologically determined in 75 patients. There were 2 patients with adenosquamous carcinoma and 12 with squamous carcinoma. In total, 41 patients were smokers and 48 had never smoked.

Gene mutations

Oncogenic mutations were found in 50 patients, including KRAS (n=21, 23.6%), PIK3CA (n=8, 9.0%) and TP53 (n=40, 44.9%). Among the 21 patients with KRAS mutations, 18 had mutations in exon 2, 3 in exon 3 and 2 in exon 4. There were 8 patients with a PIK3CA mutation in exon 10. A total of 17/40 patients had TP53 mutations located in exon 5, 6 in exon 6, 10 in exon 7 and 19 in exon 8. Coexisting mutations were identified in 17 patients (19.1%), including KRAS/TP53 (n=10, 11.2%), PIK3CA/TP53 (n=4, 4.5%), KRAS/PIK3CA (n=1, 1.1%) and KRAS/PIK3CA/TP53 (n=2, 2.2%). There were 32 cases with EGFR mutations (36.0%), 3 cases with the EMAP-like 4-ALK receptor tyrosine kinase fusion oncogene (3.4%), and 3 cases of c-MET exon 14 skipping (3.4%). The KRAS/TP53/PIK3CA mutations and percentage distribution of the 50 patients are shown in Figs. 1 and 2.

Clinical characteristics

The clinical characteristics of the 89 patients in association with the gene mutations are shown in Table II. Patients with KRAS, TP53, PIK3CA and KRAS/TP53 mutations had a higher incidence of bone metastasis than those with the wild-type gene (61.9 vs. 25.6%, P=0.006; 62.5 vs. 25.6%, P=0.024; 62.5 vs. 25.6%, P=0.042; 70.0 vs. 25.6%, P=0.009). There was also a higher incidence of adrenal metastasis in the TP53 mutation vs. wild-type groups (12.5 vs. 5.1%, P=0.017). Patients with KRAS or KRAS/TP53 mutations had a lower incidence of pleural metastasis than those with the wild-type gene (14.3 vs. 43.6%, P=0.022; 0.0 vs. 43.6%, P=0.010). Infiltrative tumor growth was greater in patients with KRAS, TP53 and KRAS/TP53 mutations than in the wild-type group (71.4 vs. 51.3%, P=0.039; 67.5 vs. 51.3%, P=0.032; 90.0 vs. 51.3%, P=0.009).

KRAS/TP53 mutations were associated with elevated carbohydrate antigen 19-9 (CA19-9) expression, vacuolar signs and margin lobulation in chest CT imaging in patients. Differences in KRAS mutation were observed in margin lobulation and invasive growth in chest CT imaging, meanwhile, first symptoms, including cough and dyspnea, indicated a statistical significance between wild-type patients and those with PIK3CA mutation (P=0.034).

Survival analysis

The PFS times of the KRAS mutation and wild-type group were 8.9±2.3 months (95% CI, 4.3–13.5) and 15.3±1.6 months (95% CI, 12.1–18.4), respectively (P=0.045). Patients with a single TP53 mutation had a PFS time of 7.8±1.5 months (95% CI, 4.9–10.7), which was significantly shorter than that of the wild-type group (P<0.001). Patients with a KRAS/TP53 coexisting mutation had a shorter PFS time of 6.6±1.6 months (95% CI, 3.5–9.7) compared with the wild-type group (P<0.001). This result was similar among PIK3CA/TP53 patients (P=0.012). The difference in the PFS times was not statistically significant between the single KRAS and KRAS/TP53 mutations, the single TP53 and KRAS/TP53 mutations or the single TP53 and PIK3CA/TP53 mutations. (Table III; Fig. 3).

Discussion

NSCLC accounts for 70–80% of lung cancer cases and 60% of patients are diagnosed at stage III or IV (10). Oncogenes such as EGFR and ALK have shifted the treatment model of lung cancer from pathology-guided to molecular-guided precision medicine with targeted therapy (11). With the improvement in examination technology and the increase in available treatment methods, the genetic and clinical characteristics of NSCLC-related genes, including KRAS, PIK3CA and TP53, are highly informative.

The present study evaluated 89 cases of NSCLC patients with KRAS, PIK3CA and TP53 mutations. KRAS mutations were found in 21 cases within exon 2 (n=18), exon 3 (n=3) and exon 4 (n=2). The total mutation rate of KRAS was 23.6%, which was similar to the results of a study undertaken by Mao et al (12), but higher than the mutation rates of 4.4–5.3% reported by Luo et al (13) and Yi et al (14). The mutation rate of PIK3CA was 3% in a study undertaken by Scheffler et al (15), but Liang et al (16) reported a rate of 47.83%. The present study included 8 cases of PIK3CA exon 10 mutations and the total mutation rate was 9.0%. TP53 has the highest mutation rate of all NSCLC-related genes, reported as 39–46% (15,16). The present study identified 40 cases with TP53 mutations within exon 5 (n=17), exon 6 (n=6), exon 7 (n=10) and exon 8 (n=19). In present study the total mutation rate of TP53 was 44.9%, which is in accordance to previous researches (17,18).

Kris et al (19) found that 3% of patients with NSLCL exhibited a double gene mutation. The Cancer Genome Atlas determined that the mutation rate of KRAS/TP53 coexisting mutation could reach 20% (20). The present study identified 17 co-mutated samples with a rate of 19.1%, including 15 double-mutations of KRAS/TP53, PIK3CA/TP53 and KRAS/PIK3CA, and 2 cases of KRAS/PIK3CA/TP53 co-mutation. This difference may result from the sensitivity and sequencing depth of next-generation sequencing by cSMART. The varied sample size between studies may also contribute toward the discrepancies in gene mutation rates.

Clinical characteristics, including the baseline demographics, clinical manifestations, partial laboratory tests, partial pathological features and certain features of chest CT imaging, of patients with mutations were not significantly different from those of wild-type patients (P>0.05). This was consistent with the results of numerous previous studies (6,2125). By contrast, KRAS/TP53 were associated with elevated CA19-9 expression, vacuolar signs and margin lobulation in chest CT imaging. However, it is possible that the sample size of each subgroup resulted in the difference in certain clinical characteristics to some extent, and further study is required due to the limited sample size used in the present study.

Invasive growth of the tumor tissue in patients was associated with KRAS, TP53 and KRAS/TP53, which was consistent with the clinical features observed. The incidence of distant metastasis was higher than that of local metastasis in patients with KRAS and TP53 mutations. The possible mechanism of this is the activation of the EGFR downstream Rat sarcoma/Rapidly Accelerated Fibrosarcoma/mitogen-activated protein kinases signaling pathways by KRAS mutations to regulate cell differentiation and proliferation. Prolonged activation of the KRAS signal is hypothesized to cause tumor cell proliferation and progression (26). TP53 gene mutations result in an oncogenic transformation of the tumor suppressor gene due to a conformational change; therefore, the regulation of cell growth, apoptosis and DNA repair is disrupted, which allows tumor cells to proliferate, grow and metastasize (27,28).

The biological significance of these mutations remains uncertain, but to some extent specific driver genes have prognostic value. Mascaux et al (29) first reported a poor prognosis in NSCLC patients with KRAS mutations, and other studies have confirmed this hypothesis (30). Recent studies have found that TP53 gene mutations may generate the same results in patients with NSCLC (3133). PIK3CA encodes the type I phosphatidylinositol-3-kinase p110α catalytic subunit (34) and is important for the development of NSCLC. PIK3CA phosphorylates the EGFR bypass pathway, PI3K/AKT/mTOR, to activate downstream signaling that promotes the proliferation, survival, adhesion and differentiation of tumor cells (35). Liang et al (16) proposed that PIK3CA gene mutations are more likely to co-exist with other oncogenic mutations and that they may weakly induce independent carcinogenesis.

In the present study, patients with NSCLC who underwent first-line chemotherapy were divided into groups according to their genotype. For all patients who have EGFR mutation in Changhai hospital, targeted therapy is discussed and anti-EGFR tyrosine kinase inhibitors are recommended as the first-line treatment. The majority of these patients do receive targeted therapy. However, due to economic problems or for other reasons, certain patients cannot afford targeted therapy. For the baseline balance of the present study, the 89 patients who received first-line chemotherapy were chosen. Patients with a single KRAS or TP53 mutation experienced shorter PFS times than the wild-type patients, which was consistent with the results of the studies by Molina-Vila et al (36) and Meng et al (37). Shepherd et al (6) hypothesized that a double gene mutation, such as KRAS/TP53, in NSCLC patients may indicate a poor prognosis. Patients with KRAS/TP53 or PIK3CA/TP53 mutations experienced a shorter PFS time than those patients with the wild-type. The PFS time of the KRAS/TP53 group was shorter than that in the single KRAS and single TP53 groups, as was the time in the PIK3CA/TP53 group compared with the single TP53 group (P>0.05). We hypothesized that there could be a ‘gene superposition’ effect in NSCLC patients with a co-mutated gene, which leads to a shortened PFS compared with a single gene mutation. However, the trend observed in the present study was not statistically significant, which was in agreement with the results of a study by Jao et al (38). The mean PFS time of patients with KRAS/PIK3CA/TP53 gene co-mutations was 6.2 months, which was shorter than that of the double and single mutation groups. Only 2 patients had this co-mutation and therefore, a larger sample size is necessary for further study. Sampling error may also exist due to the next-generation sequencing technology and the limited sample size. The subgroups of gene mutations, as well as the chemotherapy regimen and doses, were not identical; therefore, further evidence should be obtained in a large clinical study.

In conclusion, the treatment strategy for NSCLC patients with KRAS, PIK3CA and TP53 mutations has not yet been defined. The present study determined the predictive value of KRAS, PIK3CA and TP53 mutations in patients with NSCLC. Additionally, the results of the present study suggested that patients with NSCLC should undergo routine KRAS, PIK3CA and TP53 sequencing to determine single or multiple gene mutations for the analysis of patient clinical characteristics and prognosis.

Figure 1.

Mutations of KRAS, PIK3CA and TP53 genes in 50 patients with non-small cell lung cancer. KRAS, KRAS proto-oncogene GTPase; PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α; TP53, tumor protein p53. Blue represent KRAS mutation; Red TP53 mutation and Green PIK3CA mutation.

Figure 2.

Percentage distributions of KRAS, PIK3CA and TP53 gene mutations in 50 patients with non-small cell lung cancer. KRAS, KRAS proto-oncogene GTPase; PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α; TP53, tumor protein p53.

Figure 3.

PFS Kaplan-Meier curves between different gene mutations group and wild-type group. (A) PFS in patients with non-small cell lung cancer was compared between KRAS mutant and all wild-type groups (8.9±2.3 vs. 15.3±1.6 months; P=0.045). PFS was compared between TP53 mutant and all wild-type groups (7.8±1.5 vs. 15.3±1.6 months; P<0.001), and between KRAS+TP53 mutant and all wild-type groups (6.6±1.6 vs. 15.3±1.6 months; P<0.001). Compared with the KRAS mutant and TP53 mutant, respectively, the PFS time of patients with the KRAS+TP53 mutant was shorter, but not statistically different. (B) PFS was also compared between patients with the PIK3CA+TP53 mutant and all wild-type groups (7.1±2.1 vs. 15.3±1.6 months; P=0.012). The PFS time of the patients with the PIK3CA+TP53 mutant was shorter than that of patients with the TP53 mutant, but was not statistically different. PFS, progression-free survival; mt, mutant; wt, wild-type; KRAS, KRAS proto-oncogene GTPase; PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α; TP53, tumor protein p53.

Table I.
Baseline demographic characteristics of the 89 patients with non-small cell lung cancer.
Characteristicsn (%)
Sex
Male52 (58.4)
Female37 (41.6)
Age, years
<6557 (64.0)
≥6532 (36.0)
Surgical history
Yes21 (23.6)
No68 (76.4)
Smoking status
Former/current41 (46.1)
Never48 (53.9)
First symptom
Yes60 (67.4)
No29 (32.6)
Tumor site
Left lung45 (50.6)
Right lung44 (49.4)
Histology
Adenocarcinoma75 (84.3)
Adenosquamous carcinoma2 (2.2)
Squamous cell carcinoma12 (13.5)
Invasive growth
Yes50 (56.2)
No39 (43.8)
TNM stage
I1 (1.1)
II3 (3.4)
III14 (15.7)
IV71 (79.8)
Metastasis
Yes71 (79.8)
No18 (20.2)
Metastatic site
Bone39 (43.8)
Brain20 (22.5)
Adrenal8 (9.0)
Liver9 (10.1)
Pleura27 (30.3)
Lymph nodes22 (24.7)
Table II.
Association between gene mutation and clinical features.
CharacteristicsAll wt, n (%)KRAS mt, n (%)χ2P-valuePIK3CA mt, n (%)χ2P-valueTP53 mt, n (%)χ2P-valueKRAS+TP53 mt, n (%)χ2P-valuePIL3CA+TP53 mt, n (%)χ2P-value
Sex0.3420.5590.2190.6400.0180.8941.5140.2190.1200.729
Male23 (59.0)14 (66.7)4 (50.0)23 (57.5)8 (80.0)2 (50.0)
Female16 (41.0)7 (33.3)4 (50.0)17 (42.5)2 (20.0)2 (50.0)
Age, years0.1150.6940.0030.9590.3070.5800.2450.6200.2020.653
<6524 (61.5)14 (66.7)5 (62.5)27 (67.5)7 (70.0)2 (50.0)
≥6515 (38.5)7 (33.3)3 (37.5)13 (32.5)3 (30.0)2 (50.0)
Tumor site0.0120.9140.7100.4000.1170.7320.1220.7270.0220.883
Left lung21 (53.8)11 (52.4)3 (37.5)20 (50.0)6 (60.0)2 (50.0)
Right lung18 (46.2)10 (47.6)5 (62.5)20 (50.0)4 (40.0)2 (50.0)
Smoking status0.3880.5330.0040.9470.1100.7413.1480.0760.0020.961
Former/current19 (48.7)12 (57.1)4 (50.0)18 (45.0)8 (80.0)2 (50.0)
Never20 (51.3)9 (42.9)4 (50.0)22 (55.0)2 (20.0)2 (50.0)
First symptom0.5870.4404.5190.034a1.6540.1981.1970.2742.3630.124
Yes24 (61.5)15 (71.4)8 (100.0)30 (75.0)8 (80.0)4 (100.0)
No15 (38.5)6 (28.6)0 (0.0)10 (25.0)2 (20.0)0 (0.0)
CA19-91.5160.2180.7260.3940.7480.3875.1080.024a1.3810.240
Normal30 (76.9)13 (61.9)3 (37.5)27 (67.5)6 (60.0)2 (50.0)
High9 (23.1)8 (38.1)5 (62.5)13 (32.5)4 (40.0)2 (50.0)
Invasive growth4.2500.039a2.6210.1054.5750.032a6.8830.009a1.4390.230
No19 (48.7)6 (28.6)2 (25.0)13 (32.5)1 (10.0)1 (25.0)
Yes20 (51.3)15 (71.4)6 (75.0)27 (67.5)9 (90.0)3 (75.0)
Margin lobulation4.2900.038a0.0010.9702.4950.1144.2730.039a1.3360.248
Yes29 (74.4)10 (47.6)6 (75.0)23 (57.5)4 (40.0)4 (100.0)
No10 (25.6)11 (52.4)2 (25.0)17 (42.5)6 (60.0)0 (0.0)
Pleural traction0.9230.3370.7100.4000.0140.9050.0470.8284.2100.040a
Yes18 (46.2)7 (33.3)5 (62.5)19 (47.5)5 (50.0)4 (100.0)
No21 (53.8)14 (66.7)3 (37.5)21 (52.5)5 (50.0)0 (0.0)
Vacuolar signs1.1870.2760.7770.3780.3360.5623.9210.048a0.4480.503
Yes5 (12.8)5 (23.8)2 (25.0)7 (17.5)4 (40.0)1 (25.0)
No34 (87.2)16 (76.2)6 (75.0)33 (82.5)6 (60.0)3 (75.0)
Site of metastasis
Bone10 (25.6)13 (61.9)7.5940.006a5 (62.5)4.1500.042a25 (62.5)5.0810.024a7 (70.0)6.9120.009a2 (50.0)1.0700.301
Brain6 (15.4)7 (33.3)2.5910.1073 (37.5)2.0970.14810 (25.0)1.6100.2042 (20.0)0.1240.7251 (25.0)0.2460.620
Adrenal2 (5.1)4 (19.0)2.9390.0862 (25.0)3.3670.0675 (12.5)5.6480.017a2 (20.0)2.3480.1251 (25.0)2.2070.137
Liver4 (10.3)3 (14.3)0.2150.6431 (12.5)0.0350.8515 (12.5)2.8060.0942 (20.0)0.7030.4020 (0.0)0.4520.501
Pleura17 (43.6)3 (14.3)5.2750.022a2 (25.0)0.9530.3298 (20.0)0.0320.8590 (0.0)6.6750.010a0 (0.0)2.8840.089
Lymph nodes6 (15.4)6 (28.6)1.4840.2233 (37.5)2.0970.14814 (35.0)1.6100.2044 (40.0)2.9690.0851 (25.0)0.2460.620

aP<0.05. KRAS, KRAS proto-oncogene GTPase; PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α; TP53, tumor protein p53.

Table III.

Survival prognosis in non-small cell lung cancer patients with KRAS, PIK3CA and TP53 gene mutations.

KRAS mt vs. all wtTP53 mt vs. all wtKRAS+TP53 mt vs. all wtKRAS+TP53 mt vs. KRAS mtKRAS+TP53 mt vs. TP53 mtPIK3CA+TP53 mt vs. all wtPIK3CA+TP53 mt vs. TP53 mt







VariablesKRAS mtAll wtTP53 mtAll wtKRAS+ TP53 mtAll wtKRAS+ TP53 mtKRAS mtKRAS+ TP53 mtTP53 mtPIK3CA+ TP53 mtAll wtPIK3CA+ TP53 mtTP53 mt
PFS time, months8.9±2.315.3±1.67.8±1.515.3±1.66.6±1.615.3±1.66.6±1.68.9±1.36.6±1.67.8±1.57.1±1.515.3±1.67.13±117.8±1.5
95% CI4.3–13.512.1–18.44.9–10.712.1–18.43.5–9.712.1–18.43.5–9.74.3–13.53.5–9.74.9–10.73.1–11.212.1–18.43.1–11.24.9–10.7
P-value0.045<0.001<0.0010.3980.8730.0120.986

KRAS, KRAS proto-oncogene GTPase; PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α; TP53, tumor protein p53; wt, wild-type; mt, mutant; CI, confidence interval.

Acknowledgements

The authors would like to thank the staff of the Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital (Shanghai, China) for providing assistance in data management and statistical analysis.

Funding

The present study was supported by the Shanghai Scientific Research Projects (grant no. 15411960400).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

JZ, YH, RC and CB conceived and designed the study. JL analyzed the statistics. CB provided a part of patients' clinical data and monitored the whole study; JZ and RC wrote the original draft, YH and CB reviewed and edited the draft. All authors have read and approved the final version of this manuscript.

Ethics approval and consent to participate

The study was approved by the Ethics Committee of Changhai Hospital affiliated to Second Military Medical University (Shanghai, China).

Patient consent for publication

Written nformed consent and permission for publication was obtained for all patients in the present study.

Competing interests

The authors declare that they have no competing interests.

References

  • 1. SiegelRMaJZouZJemalACancer statistics, 2014CA Cancer J Clin649292014[PubMed][Google Scholar]
  • 2. ZengHZhengRGuoYZhangSZouXWangNZhangLTangJChenJWeiKCancer survival in China, 2003–2005: A population-based studyInt J Cancer136192119302015[PubMed][Google Scholar]
  • 3. MoreiraALThorntonRHPersonalized medicine for non-small-cell lung cancer: Implications of recent advances in tissue acquisition for molecular and histologic testingClin Lung Cancer133343392012[PubMed][Google Scholar]
  • 4. MartinPLeighlNBTsaoMSShepherdFAKRAS mutations as prognostic and predictive markers in non-small cell lung cancerJ Thorac Oncol85305422013[PubMed][Google Scholar]
  • 5. PapadimitrakopoulouVDevelopment of PI3K/AKT/mTOR pathway inhibitors and their application in personalized therapy for non-small-cell lung cancerJ Thorac Oncol7131513262012[PubMed][Google Scholar]
  • 6. ShepherdFALacasBLe TeuffGHainautPJännePAPignonJPLe ChevalierTSeymourLDouillardJYGrazianoSPooled analysis of the prognostic and predictive effects of TP53 comutation status combined with KRAS or EGFR mutation in early-stage resected non-small-cell lung cancer in four trials of adjuvant chemotherapyJ Clin Oncol35201820272017[PubMed][Google Scholar]
  • 7. DetterbeckFCBoffaDJKimAWTanoueLTThe eighth edition lung cancer stage classificationChest1511932032017[PubMed][Google Scholar]
  • 8. LvWWeiXGuoRLiuQZhengYChangJBaiTLiHZhangJSongZNoninvasive prenatal testing for Wilson disease by use of circulating single-molecule amplification and resequencing technology (cSMART)Clin Chem611721812015[PubMed][Google Scholar]
  • 9. WatanabeHOkadaMKajiYSatouchiMSatoYYamabeYOnayaHEndoMSoneMAraiYNew response evaluation criteria in solid tumours-revised RECIST guideline (version 1.1)Gan To Kagaku Ryoho36249525012009(In Japanese)[PubMed][Google Scholar]
  • 10. TsimSO'DowdCAMilroyRDavidsonSStaging of non-small cell lung cancer (NSCLC): A reviewRespir Med104176717742010[PubMed][Google Scholar]
  • 11. CooperWAO'TooleSBoyerMHorvathLMaharAWhat's new in non-small cell lung cancer for pathologists: The importance of accurate subtyping, EGFR mutations and ALK rearrangementsPathology431031152011[PubMed][Google Scholar]
  • 12. MaoCQiuLXLiaoRYDuFBDingHYangWCLiJChenQKRAS mutations and resistance to EGFR-TKIs treatment in patients with non-small cell lung cancer: A meta-analysis of 22 studiesLung Cancer692722782010[PubMed][Google Scholar]
  • 13. LuoWWangHXuWJAnalysis of KRAS mutation in patients with non- small cell lung cancerGuangdong Med J35202520282014(In Chinese)[Google Scholar]
  • 14. YiSQZhuangYZhuWDAnalysis of KRAS gene mutations in non-small cell lung cancerZhonghua Lin Chuan Yi Shi Za Zhi (Electronic Edition)7911191152013(In Chinese)[Google Scholar]
  • 15. SchefflerMBosMGardiziMKönigKMichelsSFassunkeJHeydtCKünstlingerHIhleMUeckerothFPIK3CA mutations in non-small cell lung cancer (NSCLC): Genetic heterogeneity, prognostic impact and incidence of prior malignanciesOncotarget6131513262015[PubMed][Google Scholar]
  • 16. LiangNXLiuYXLiuLLiSQCo-mutation of PIK3CA and other oncogenes in patients with non-small cell lung cancerMed J PUMCH61861902015(In Chinese)[Google Scholar]
  • 17. HuangCLTakiTAdachiMKonishiTHigashiyamaMKinoshitaMHadamaTMiyakeMMutations of p53 and K-ras genes as prognostic factors for non-small cell lung cancerInt J Oncol125535691998[PubMed][Google Scholar]
  • 18. KatoSHanSYLiuWOtsukaKShibataHKanamaruRIshiokaCUnderstanding the function-structure and function-mutation relationships of p53 tumor suppressor protein by high-resolution missense mutation analysisProc Natl Acad Sci USA100842484292003[PubMed][Google Scholar]
  • 19. KrisMGJohnsonBEBerryLDKwiatkowskiDJIafrateAJWistubaIIVarella-GarciaMFranklinWAAronsonSLSuPFUsing multiplexed assays of oncogenic drivers in lung cancers to select targeted drugsJama311199820062014[PubMed][Google Scholar]
  • 20. Cancer Genome Atlas Research Network: Comprehensive molecular profiling of lung adenocarcinomaNature5115435502014[PubMed][Google Scholar]
  • 21. GaoJChenJQZhangLLiangZYRelationship between EGFR and KRAS mutations and prognosis in Chinese patients with non-small cell lung cancer: A mutation analysis with real-time polymerase chain reaction using scorpion amplification refractory mutation systemZhonghua Bing Li Xue Za Zhi416526562012(In Chinese)[PubMed][Google Scholar]
  • 22. KimHRAhnJRLeeJGBangDHHaSJHongYKKimSMNamKCRhaSYSooRAThe impact of cigarette smoking on the frequency of and qualitative differences in KRAS mutations in Korean patients with lung adenocarcinomaYonsei Med J548658742013[PubMed][Google Scholar]
  • 23. LiYLiYYangTWeiSWangJWangMWangYZhouQLiuHChenJClinical significance of EML4-ALK fusion gene and association with EGFR and KRAS gene mutations in 208 Chinese patients with non-small cell lung cancerPLoS One8e520932013[PubMed][Google Scholar]
  • 24. ZhangQWangJLiXZhangHNongJQinNZhangXWuYYangXLvJZhangSClinical Analysis of 107 NSCLC Patients Harboring KRAS MutationZhongguo Fei Ai Za Zhi192572622016(In Chinese)[PubMed][Google Scholar]
  • 25. HuWLiuYChenJConcurrent gene alterations with EGFR mutation and treatment efficacy of EGFR-TKIs in Chinese patients with non-small cell lung cancerOncotarget8250462017[PubMed][Google Scholar]
  • 26. KorpantyGJGrahamDMVincentMDLeighlNBBiomarkers that currently affect clinical practice in lung cancer: EGFR, ALK, MET, ROS-1, and KRASFront Oncol42042014[PubMed][Google Scholar]
  • 27. DebenCVan den BosscheJVan Der SteenNLardonFWoutersAde BeeckKOHermansCJacobsJPeetersMVan CampGDeep sequencing of the TP53 gene reveals a potential risk allele for non-small cell lung cancer and supports the negative prognostic value of TP53 variantsTumour Biol3910104283176943272017[PubMed][Google Scholar]
  • 28. VanderlaanPARangachariDMockusSMSpotlowVReddiHVMalcolmJHubermanMSJosephLJKobayashiSSCostaDBMutations in TP53, PIK3CA, PTEN and other genes in EGFR mutated lung cancers: Correlation with clinical outcomesLung Cancer10617212017[PubMed][Google Scholar]
  • 29. MascauxCIanninoNMartinBPaesmansMBerghmansTDusartMHallerALothairePMeertAPNoelSThe role of RAS oncogene in survival of patients with lung cancer: A systematic review of the literature with meta-analysisBr J Cancer921311392005[PubMed][Google Scholar]
  • 30. LinEYRupaniRGitlitzBJMarkers in lung cancerSpringerNew York, NY2013[PubMed]
  • 31. ScocciantiCVesinAMartelGOlivierMBrambillaETimsitJFTavecchioLBrambillaCFieldJKHainautP;European Early Lung Cancer Consortium: Prognostic value of TP53, KRAS and EGFR mutations in nonsmall cell lung cancer: The EUELC cohortEur Respir J401771842012[PubMed][Google Scholar]
  • 32. MaXRousseauVSunHLantuejoulSFilipitsMPirkerRPopperHMendiboureJVataireALLe ChevalierTSignificance of TP53 mutations as predictive markers of adjuvant cisplatin-based chemotherapy in completely resected non-small-cell lung cancerMol Oncol85555642014[PubMed][Google Scholar]
  • 33. LeeSYJeonHSHwangboYJeongJYParkJYLeeEJJinGShinKMYooSSLeeJThe influence of TP53 mutations on the prognosis of patients with early stage non-small cell lung cancer may depend on the intratumor heterogeneity of the mutationsMol Carcinog54931012015[PubMed][Google Scholar]
  • 34. McgowanMHovenASLund-IversenMSolbergSHellandÅHirschFRBrustugunOTPIK3CA mutations as prognostic factor in squamous cell lung carcinomaLung Cancer10352572017[PubMed][Google Scholar]
  • 35. KangSBaderAGVogtPKPhosphatidylinositol 3-kinase mutations identified in human cancer are oncogenicProc Natl Acad Sci USA1028028072005[PubMed][Google Scholar]
  • 36. Molina-VilaMABertran-AlamilloJGascóAMayo-de-las-CasasCSánchez-RoncoMPujantell-PastorLBonannoLFavarettoAGCardonaAFVergnenègreANondisruptive p53 mutations are associated with shorter survival in patients with advanced non-small cell lung cancerClin Cancer Res20464746592014[PubMed][Google Scholar]
  • 37. MengDYuanMLiXChenLYangJZhaoXMaWXinJPrognostic value of K-RAS mutations in patients with non-small cell lung cancer: A systematic review with meta-analysisLung Cancer811102013[PubMed][Google Scholar]
  • 38. JaoKTomasiniPKamel-ReidSTsaoMSPrognostic effect of single versus multiple somatic mutations in non-small cell lung cancer (NSCLC)J Clin Oncol3375212015[Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.