Skip to main content
Journal of Clinical Oncology logoLink to Journal of Clinical Oncology
. 2009 Jan 21;27(6):857–871. doi: 10.1200/JCO.2008.17.6297

Genetic Variations in the PI3K/PTEN/AKT/mTOR Pathway Are Associated With Clinical Outcomes in Esophageal Cancer Patients Treated With Chemoradiotherapy

Michelle AT Hildebrandt 1, Hushan Yang 1, Mien-Chie Hung 1, Julie G Izzo 1, Maosheng Huang 1, Jie Lin 1, Jaffer A Ajani 1, Xifeng Wu 1,
PMCID: PMC2738430  PMID: 19164214

Abstract

Purpose

The phosphoinositide-3-kinase (PI3K), phosphatase and tensin homolog (PTEN), v-akt murine thymoma viral oncogene homolog (AKT), and mammalian target of rapamycin (mTOR) signaling pathway has been implicated in resistance to several chemotherapeutic agents. In this retrospective study, we determined whether common genetic variations in this pathway are associated with clinical outcomes in esophageal cancer patients with adenocarcinoma or squamous cell carcinoma who have undergone chemoradiotherapy and surgery.

Patients and Methods

Sixteen tagging single nucleotide polymorphisms (SNPs) in PIK3CA, PTEN, AKT1, AKT2, and FRAP1 (encoding mTOR) were genotyped in these patients and analyzed for associations with response to therapy, survival, and recurrence.

Results

We observed an increased recurrence risk with genetic variations in AKT1 and AKT2 (hazard ratio [HR], 2.21; 95% CI, 1.06 to 4.60; and HR, 3.30; 95% CI, 1.64 to 6.66, respectively). This effect was magnified with an increasing number of AKT adverse genotypes. In contrast, a predictable protective effect by PTEN genetic variants on recurrence was evident. Survival tree analysis identified higher-order interactions that resulted in variation in recurrence-free survival from 12 to 42 months, depending on the combination of SNPs. Genetic variations in AKT1, AKT2, and FRAP1 were associated with survival. Patients homozygous for either of the FRAP1 SNPs assayed had a more than three-fold increased risk of death. Two genes—AKT2 and FRAP1—were associated with a poor treatment response, while a better response was associated with heterozygosity for AKT1:rs3803304 (odds ratio, 0.50; 95% CI, 0.25 to 0.99).

Conclusion

These results suggest that common genetic variations in this pathway modulate clinical outcomes in patients who undergo chemoradiotherapy. With further validation, these results may be used to build a model of individualized therapy for the selection of the optimal chemotherapeutic regimen.

INTRODUCTION

An estimated 16,400 new cases of esophageal cancer (EC) will be diagnosed in 2008.1 Surgery is one of the standard treatments for patients with resectable tumors but frequently preoperative chemoradiotherapy is used to treat both adenocarcinoma and squamous cell carcinoma ECs.25 The most commonly utilized chemotherapy agents belong to fluoropyrimidines, taxanes, and platinum compounds. Unfortunately, even with the multimodal approach, current treatments result in a poor overall 5-year survival rate of 25% to 28%.69

Heterogeneity in response to chemoradiotherapy may be due to several factors, including age, sex, ethnicity, and drug-drug interactions. In addition, genetic variations in pharmacokinetic, pharmacodynamic, and drug action pathways have been shown to be important in determining sensitivity or resistance to treatment.10 Therefore, one strategy to increase the effectiveness of chemoradiotherapy is to gain a better understanding of the influence a patient's genetic background has on response to treatment. Our group has previously reported that genetic variations in several drug action pathways were associated with variation in clinical outcomes in EC.11 In this study, we expand those results by analyzing an important signaling pathway comprised of phosphoinositide-3-kinase (PI3K), phosphatase and tensin homolog (PTEN), v-akt murine thymoma viral oncogene homolog (AKT), and mammalian target of rapamycin (mTOR).

Signaling through the PI3K/PTEN/AKT/mTOR pathway is responsible for balancing cell survival and apoptosis.12,13 The signal is initiated by growth factors and hormones that bind receptor tyrosine kinases such as epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptor (VEGFR), and platelet-derived growth factor receptor (PDGFR).14 These receptors then activate PI3Ks resulting in a kinase cascade through AKT and mTOR, generating cell survival, growth, and angiogenesis signals.15 PTEN negatively regulates this pathway by dephosphorylating phosphatidylinositol trisphosphate (PIP3) and negating the signal generated by PI3K.16 This pathway has been shown to be commonly activated in cancer, including EC, and in the progression of Barrett's neoplasm to EC.12,17,18 Furthermore, studies in several cancer types have demonstrated that this pathway has a key role in the development of resistance to platinum compounds, taxanes, and fluoropyrimidines.1924 To our knowledge, no studies have addressed how genetic variations in this pathway influence outcomes in patients with EC treated with these chemotherapeutic agents.

In this study, we determined whether common genetic variations in AKT1, AKT2, PIK3CA (catalytic subunit of PI3K), PTEN, and FRAP1 (mTOR) were associated with clinical outcomes in patients who received chemoradiotherapy. Tagged single nucleotide polymorphisms (SNPs) were selected for each gene and genotyped in patients with EC. This pathway-based tagging approach allowed us to query genetic variations in the major effectors of this pathway and identify associations with clinical outcomes.

PATIENTS AND METHODS

Patient Population

This study included 210 patients with resectable adenocarcinoma (174 cases) or squamous cell carcinoma (36 cases) who were recruited between 1985 and 2003 at The University of Texas M. D. Anderson Cancer Center (Houston, TX).11 All patients had undergone chemoradiotherapy followed by surgery, or induction chemotherapy followed by chemoradiotherapy and surgery.

Clinical Data Collection

Patients with EC were staged as described previously.11 After chemoradiotherapy, patients underwent restaging and surgery. Pathologic response to treatment was measured by previously described methodology.25,26 Response was defined as no residual carcinoma in the primary tumor site. A poor response was any response less than a complete response. Study end points were pathologic response to therapy, recurrence, and survival. This study was approved by the M. D. Anderson Cancer Center institutional review board.

SNP Selection and Genotyping

Genomic DNA was extracted from paraffin slides using the PicoPure DNA extraction kit (Arcturus Bioscience, Mountain View, CA). Tagging SNPs were selected within and 5-kb flanking each gene using the tagger algorithm28 with a cutoff of 0.80 for r2 and a minor allele frequency between 0.10 and 0.35 based on data from Centre d'Etude du Polymorphisme Humain samples genotyped by the HapMap Project (www.hapmap.org).27 A total of sixteen SNPs were selected to represent genetic variation of 95 SNPs in the pathway (Appendix Table A1, online only). TaqMan genotyping assays, including quality control measures, were performed as previously described11 using the 7900HT sequence detection system (Applied Biosystems, Foster City, CA).

Statistical Analysis

Hazard ratios (HRs) for recurrence and survival end points were estimated by applying the Cox proportional hazards model while adjusting for age, sex, smoking status, alcohol consumption, radiation dosage, chemoradiotherapy sequence, clinical stage, chemotherapy regimens, histologic tumor type, tumor location, pathologic stage, and histologic viability. The Kaplan-Meier survival function and log-rank tests were used to assess differences in recurrence-free and overall survival times. For pathologic response to therapy, unconditional multivariate logistic regression analysis was done to estimate adjusted odds ratios (ORs) along with the corresponding 95% CIs for each SNP. We also evaluated the combined effects by the number of unfavorable genotypes identified from the main effects analysis of single SNPs. The statistical analyses described above were completed using the STATA software (version 8, STATA, College Station, TX). Survival tree analyses were used to identify higher-order gene-gene interactions. Survival tree analysis was performed using the STREE program (http://masal.med.yale.edu/stree/) which uses recursive-partitioning to identify subgroups of individuals at higher risk. All statistical analyses were two sided, and P < .05 was considered statistically significant.

RESULTS

Patient Characteristics

Of the 210 patients with EC enrolled in this study, DNA was available for 207 and of those, 186 were white (90%; Appendix Table A2, online only). Because of the small number of patients from other ethnic groups, we focused our study on white patients only. Fifty-nine percent of patients were ever smokers, and 33.1% used alcohol on a daily basis. Approximately half of patients (51%) presented with stage IIA disease. More than 97% of the patients (182) were treated with either a fluoropyrimidine, platinum agent, or taxane. Of these 182 patients, 177 received a fluoropyrimidine (95%), 132 received a platinum agent (71%), and 94 received a taxane (51%). The median follow-up time was 18.8 months, with 97 deaths and 59 recurrences. The overall median survival time was 34.5 months.

Associations Between SNPs and Recurrence Risk

Variant genotypes were analyzed for association with recurrence risk in patients after chemoradiotherapy. Three SNPs—AKT1:rs2498804, AKT2:rs892119, and PTEN:rs12357281—were associated with variation in recurrence risk (Table 1).

Table 1.

PI3K/PTEN/AKT/mTOR Pathway Genotypes and Recurrence

SNP and Genotype Any of the Three
Fluoropyrimidine + Any
No Recurrence/Recurrence (No.) HR* 95% CI P No Recurrence/Recurrence (No.) HR* 95% CI P
AKT1:rs3803304
    CC 60/26 1 (reference) 57/26 1 (reference)
    CG 54/25 1.44 0.75 to 2.78 .276 53/24 1.38 0.71 to 2.71 .341
    GG 7/5 1.87 0.59 to 5.91 .289 7/5 1.72 0.54 to 5.54 .361
    CG + GG 1.51 0.81 to 2.81 .200 1.43 0.76 to 2.72 .269
AKT1:rs2498804
    GG 50/20 1 (reference) 47/20 1 (reference)
    GT 63/30 2.04 0.95 to 4.37 .068 62/29 1.99 0.92 to 4.29 .080
    TT 8/7 3.21 1.07 to 9.61 .037 8/7 3.06 1.01 to 9.28 .049
    GT + TT 2.21 1.06 to 4.60 .034 2.14 1.02 to 4.50 .045
AKT1:rs2494738
    AA 105/49 1 (reference) 101/48 1 (reference)
    AG 18/7 0.94 0.36 to 2.44 .900 18/7 0.96 0.37 to 2.49 .936
AKT1:rs1130214
    GG 60/28 1 (reference) 57/27 1 (reference)
    GT 56/28 1.91 0.94 to 3.87 .073 55/28 1.91 0.94 to 3.87 .072
    TT 7/2 1.83 0.34 to 9.38 .482 7/2 1.82 0.34 to 9.80 .488
    GT + TT 1.90 0.94 to 3.85 .073 1.91 0.94 to 3.86 .072
AKT2:rs892119
    AA 95/33 1 (reference) 94/32 1 (reference)
    AG 25/23 3.48 1.66 to 7.28 .001 23/23 3.72 1.76 to 7.84 .001
    GG 3/2 2.42 0.49 to 12.01 .280 2/2 2.57 0.51 to 12.88 .251
    AG + GG 3.30 1.64 to 6.66 .001 3.52 1.73 to 7.17 .001
AKT2:rs8100018
    CC 61/24 1 (reference) 59/23 1 (reference)
    CG 47/30 1.26 0.64 to 2.46 .505 45/30 1.32 0.67 to 2.60 .430
    GG 13/3 0.41 0.10 to 1.58 .193 13/3 0.44 0.11 to 1.74 .242
    CG + GG 1.02 0.54 to 1.94 .944 1.09 0.57 to 2.09 .799
FRAP1:rs11121704
    CC 57/31 1 (reference) 54/30 1 (reference)
    CT 55/23 0.90 0.43 to 1.88 .783 54/23 0.94 0.48 to 1.96 .866
    TT 11/3 1.79 0.45 to 7.09 .409 11/3 1.79 0.45 to 7.06 .404
    CT + TT 0.97 0.48 to 1.99 .938 1.01 0.50 to 2.07 .974
FRAP1:rs2295080
    GG 47/21 1 (reference) 44/20 1 (reference)
    GT 56/29 1.81 0.81 to 4.05 .149 55/29 1.86 0.83 to 4.16 .132
    TT 14/4 3.04 0.82 to 11.27 .096 14/4 3.00 0.82 to 11.04 .098
    GT + TT 1.92 0.87 to 4.21 .105 2.00 0.61 to 6.48 .250
PIK3CA:rs7651265
    AA 95/40 1 (reference) 91/40 1 (reference)
    AG 23/15 1.54 0.74 to 3.19 .248 23/14 1.47 0.70 to 3.08 .312
    GG 2/0 2/0
    AG + GG 1.43 0.69 to 2.95 .332 1.36 0.65 to 2.85 .409
PIK3CA:rs7640662
    CC 95/45 1 (reference) 93/44 1 (reference)
    CG 27/11 1.02 0.46 to 2.25 .970 25/11 1.06 0.48 to 2.34 .881
    GG 2/1 1.26 0.14 to 11.76 .838 2/1 1.22 0.13 to 11.45 .864
    CG + GG 1.04 0.48 to 2.21 .927 1.08 0.51 to 2.29 .849
PIK3CA:rs7621329
    CC 80/32 1 (reference) 77/35 1 (reference)
    CT 31/16 0.83 0.40 to 1.71 .620 30/15 0.84 0.41 to 1.74 .643
    TT 6/3 1.76 0.42 to 7.38 .442 6/3 1.70 0.41 to 7.12 .467
    CT + TT 0.92 0.47 to 1.83 .822 0.93 0.47 to 1.84 .838
PIK3CA:rs6443624
    AA 76/32 1 (reference) 73/32 1 (reference)
    AC 36/22 1.40 0.69 to 2.85 .348 35/21 1.39 0.68 to 2.82 .365
    CC 10/4 0.95 0.28 to 3.16 .929 10/4 0.94 0.29 to 3.13 .926
    AC + CC 1.30 0.66 to 2.57 .444 1.29 0.65 to 2.54 .461
SNP and Genotype Platinum Compound + Any
Taxane + Any
No Recurrence/Recurrence (No.) HR* 95% CI P No Recurrence/Recurrence (No.) HR* 95% CI P
AKT1:rs3803304
    CC 43/20 1 (reference) 28/12 1 (reference)
    CG 37/21 2.21 0.95 to 5.12 .065 31/13 2.24 0.75 to 6.72 .149
    GG 5/3 1.62 0.37 to 7.15 .526 4/3 4.75 0.54 to 42.06 .162
    CG + GG 2.08 0.94 to 4.60 .070 2.43 0.83 to 7.14 .107
AKT1:rs2498804
    GG 32/16 1 (reference) 26/7 1 (reference)
    GT 48/23 2.60 1.00 to 6.75 .049 31/17 11.95 1.89 to 75.41 .008
    TT 5/5 3.55 0.97 to 13.02 .056 5/5 21.96 2.68 to 179.88 .004
    GT + TT 2.79 1.13 to 6.89 .026 14.10 2.40 to 83.02 .003
AKT1:rs2494738
    AA 71/39 1 (reference) 57/24 1 (reference)
    AG 15/5 0.95 0.31 to 2.86 .921 8/4 1.33 0.23 to 7.63 .751
AKT1:rs1130214
    GG 45/23 1 (reference) 31/15 1 (reference)
    GT 37/21 2.08 0.87 to 4.98 .099 30/12 2.39 0.84 to 6.79 .102
    TT 4/1 1.34 0.11 to 15.84 .817 4/2 10.94 1.55 to 76.98 .016
    GT + TT 2.06 0.86 to 4.94 .104 2.72 0.97 to 7.62 .056
AKT2:rs892119
    AA 68/25 1 (reference) 49/16 1 (reference)
    AG 16/18 7.36 2.79 to 19.42 .000056 14/12 5.77 1.70 to 19.52 .005
    GG 2/2 2.98 0.54 to 16.51 .211 2/1 2.56 0.16 to 40.00 .503
    AG + GG 6.20 2.45 to 15.69 .00012 5.23 1.62 to 16.88 .006
AKT2:rs8100018
    CC 41/18 1 (reference) 35/14 1 (reference)
    CG 36/27 1.27 0.58 to 2.80 .554 23/12 1.35 0.44 to 4.17 .603
    GG 7/0 7/3 0.33 0.05 to 2.29 .262
    CG + GG 0.91 0.42 to 1.99 .822 0.97 0.34 to 2.81 .956
FRAP1:rs11121704
    CC 43/26 1 (reference) 30/15 1 (reference)
    CT 36/17 1.17 0.50 to 2.72 .718 30/12 0.47 0.12 to 1.90 .289
    TT 7/2 2.56 0.40 to 16.30 .320 5/2 2.33 0.26 to 20.89 .450
    CT + TT 1.29 0.58 to 2.86 .526 0.52 0.13 to 2.04 .346
FRAP1:rs2295080
    GG 35/18 1 (reference) 23/10 1 (reference)
    GT 35/22 2.35 0.88 to 6.23 .087 36/15 1.31 0.28 to 6.19 .730
    TT 10/2 2.35 0.37 to 15.06 .367 5/3 12.35 1.19 to 128.38 .035
    GT + TT 2.35 0.91 to 6.07 .079 1.43 0.30 to 6.82 .650
PIK3CA:rs7651265
    AA 68/30 1 (reference) 51/20 1 (reference)
    AG 14/13 2.13 0.81 to 5.58 .123 12/7 3.13 0.79 to 12.35 .103
    GG 2/0 0/0
    AG + GG 1.75 0.70 to 4.38 .232
PIK3CA:rs7640662
    CC 65/34 1 (reference) 51/24 1 (reference)
    CG 20/10 1.24 0.50 to 3.08 .639 13/4 1.15 0.29 to 4.51 .844
    GG 2/1 1.38 0.14 to 13.53 .784 1/1 1.96 0.17 to 22.84 .589
    CG + GG 1.26 0.53 to 2.99 .604 1.27 0.37 to 4.41 .705
PIK3CA:rs7621329
    CC 54/25 1 (reference) 45/17 1 (reference)
    CT 22/14 0.90 0.35 to 2.31 .822 16/8 0.50 0.15 to 1.71 .270
    TT 5/2 1.08 0.18 to 6.66 .930 3/2 11.38 1.60 to 80.88 .015
    CT + TT 0.92 0.37 to 2.28 .862 0.90 0.30 to 2.71 .846
PIK3CA:rs6443624
    AA 54/24 1 (reference) 40/17 1 (reference)
    AC 23/18 1.81 0.72 to 4.52 .205 20/9 0.77 0.24 to 2.46 .661
    CC 8/3 1.26 0.30 to 5.37 .753 4/3 7.13 1.20 to 42.36 .031
    AC + CC 1.69 0.69 to 4.13 .247 1.20 0.41 to 3.53 .737
SNP and Genotype Any of the Three
Fluoropyrimidine + Any
No Recurrence/Recurrence (No.) HR* 95% CI P No Recurrence/Recurrence (No.) HR* 95% CI P
PIK3CA:rs2699887
    AA 76/37 1 (reference) 74/36 1 (reference)
    AG 37/17 1.20 0.58 to 2.49 .618 35/17 1.29 0.62 to 2.71 .498
    GG 6/3 0.73 0.18 to 2.93 .654 6/3 0.81 0.20 to 3.22 .762
    AG + GG 1.09 0.55 to 2.17 .795 1.18 0.59 to 2.37 .639
PTEN:rs2299939
    AA 82/38 1 (reference) 79/37 1 (reference)
    AC 34/16 1.06 0.53 to 2.15 .864 34/16 1.05 0.52 to 2.13 .883
    CC 5/3 0.45 0.09 to 2.21 .328 5/3 0.46 0.10 to 2.22 .335
    AC + CC 0.91 0.47 to 1.76 .784 0.91 0.47 to 1.75 .776
PTEN:rs12569998
    GG 95/44 1 (reference) 93/44 1 (reference)
    GT 26/12 0.80 0.37 to 1.73 .570 24/11 0.81 0.37 to 1.76 .598
    TT 1/1 1.40 0.12 to 15.73 .785 1/1 1.51 0.13 to 17.19 .742
    GT + TT 0.83 0.39 to 1.75 .617 0.84 0.40 to 1.79 .650
PTEN:rs12357281
    CC 95/49 1 (reference) 92/48 1 (reference)
    CG 22/7 0.34 0.13 to 0.89 .027 21/7 0.33 0.13 to 0.87 .025
    GG 1/0 1/0
    CG + GG 0.34 0.13 to 0.88 .027 0.33 0.13 to 0.87 .025
AKT1:rs2498804 and AKT2:rs892119 unfavorable genotype analysis
    No. of unfavorable genotypes
        0 36/13 1 (reference) 35/13 1 (reference)
        1 69/27 2.87 1.15 to 7.19 .024 67/26 2.80 1.12 to 7.03 .028
        2 15/17 6.52 2.34 to 18.18 < .001 14/17 6.36 2.28 to 17.72 < .001
SNP and Genotype Platinum Compound + Any
Taxane + Any
No Recurrence/Recurrence (No.) HR* 95% CI P No Recurrence/Recurrence (No.) HR* 95% CI P
PIK3CA:rs2699887
    AA 53/27 1 (reference) 41/19 1 (reference)
    AG 25/15 1.35 0.58 to 3.10 .486 18/6 1.95 0.57 to 6.66 .286
    GG 5/2 0.36 0.04 to 3.09 .353 4/3 1.15 0.25 to 5.36 .856
    AG + GG 1.16 0.51 to 2.64 .726 1.59 0.56 to 4.49 .383
PTEN:rs2299939
    AA 59/28 1 (reference) 42/20 1 (reference)
    AC 21/14 0.94 0.39 to 2.26 .895 20/8 1.38 0.41 to 4.63 .605
    CC 4/3 0.36 0.06 to 2.21 .267 1/1 0.37 0.01 to 11.18 .566
    AC + CC 0.78 0.35 to 1.75 .544 1.17 0.37 to 3.68 .784
PTEN:rs12569998
    GG 67/34 1 (reference) 53/20 1 (reference)
    GT 17/10 1.09 0.40 to 2.93 .868 10/8 1.06 0.29 to 3.93 .927
    TT 1/0 1/1 1.08 0.05 to 22.39 .962
    GT + TT 1.06 0.39 to 2.82 .915 1.06 0.32 to 3.59 .920
PTEN:rs12357281
    CC 72/38 1 (reference) 44/26 1 (reference)
    CG 11/6 0.36 0.11 to 1.21 .098 17/2 0.05 0.006 to 0.461 .008
    GG 0/0 1/0
    CG + GG 0.05 0.006 to 0.458 .008
AKT1:rs2498804 and AKT2:rs892119 unfavorable genotype analysis
    No. of unfavorable genotypes
        0 24/12 1 (reference) 18/4 1 (reference)
        1 49/17 3.54 1.10 to 11.46 .040 36/15 9.09 1.29 to 64.26 .027
        2 11/15 10.73 3.21 to 35.82 < .001 8/10 83.37 9.56 to 726.78 < .001

Abbreviations: PI3K, phosphoinositide-3-kinase; PTEN, phosphatase and tensin homolog; AKT, v-akt murine thymoma viral oncogene homolog; mTOR, mammalian target of rapamycin; SNP, single nucleotide polymorphism; HR, hazard ratio.

*

Adjusted for age, sex, smoking status, alcohol consumption, radiation dosage, chemoradiotherapy sequence, clinical stage, chemotherapy regimens, histologic tumor type, tumor location, pathologic stage, and histologic viability.

AKT1 and AKT2 genetic variations.

The AKT1 and AKT2 SNPs resulted in increased risk, with adjusted HRs of 2.21 (95% CI, 1.06 to 4.60) and 3.30 (95% CI, 1.64 to 6.66), respectively. These two SNPs were also associated with recurrence when the results were stratified by treatment (Table 1). Furthermore, AKT2:rs892119 resulted in dramatically different median recurrence-free survival times of 42 months for patients with wild-type genotype compared with 12 months for those with one or two variant alleles (Fig 1).

Fig. 1.

Fig. 1.

Kaplan-Meier curves of recurrence-free survival times in patients with esophageal cancer (EC) with AKT2:rs892119 treated with (A) any of the three drugs, (B) fluoropyrimidine, (C) platinum compound, and (D) taxane. The numbers in parentheses are the numbers of patients with EC with recurrence/total patients with the respective genotype. MST, median survival time in months.

Because AKT1 and AKT2 SNPs were consistently associated with recurrence risk, we performed an unfavorable genotype analysis to determine the effect of having one or both of these SNPs. One unfavorable genotype resulted in a nearly three-fold increased recurrence risk (95% CI, 1.15 to 7.19; Table 1). This risk increased to more than six-fold (HR, 6.52; 95% CI, 2.34 to 18.18) in patients with two unfavorable genotypes. The same result was also observed when results were stratified by treatment, with HRs for two unfavorable genotypes of 6.36 (95% CI, 2.28 to 17.72), 10.73 (95% CI, 3.21 to 35.82), and 83.4 (95% CI, 9.56 to 726.78) for fluoropyrimidine, platinum compound, and taxane groups, respectively. These results demonstrate that AKT1 and AKT2 genetic variation has an additive effect on recurrence risk and recurrence-free survival rates.

PTEN genetic variation.

In contrast, and as would be predicted by PTEN′s negative regulation of signaling through the pathway, PTEN:rs12357281 was associated with a decreased recurrence risk. Patients carrying at least one variant allele had a significant reduction in risk (HR, 0.34; 95% CI, 0.13 to 0.88). This same pattern was also observed for fluoropyrimidine and taxane groups, with HRs of 0.33 (95% CI, 0.13 to 0.87) and 0.05 (95% CI, 0.0006 to 0.458), respectively (Table 1).

Higher-order gene-gene interactions.

Overall, seven SNPs were found to be associated with recurrence risk in patients treated with a taxane as part of their overall treatment regimen. Survival tree analysis was used to identify interactions within these SNPs. AKT2:rs892119, PIK3CA:rs6443624, and PTEN:rs12357281 demonstrated gene-gene interactions, resulting in four terminal nodes with different recurrence-free survival times (Fig 2A). The initial split on the survival tree was due to AKT2:rs892119 (node 4), indicating that this SNP is the primary factor contributing to variation in recurrence risk in this population. The reference group for the analysis (node 1) was composed of individuals with wild-type AKT2:rs892119 and wild-type PIK3CA:rs6443624 genotypes. Patients in this node had the longest recurrence-free survival time of 48 months. This duration was comparable to patients in node 2 who carried the wild-type alleles for AKT2:892119, but variants of both PIK3CA:rs6443624 and PTEN:rs12357281. These results suggest that the protective effect conferred by PTEN is able to counteract the negative consequences of PIK3CA genetic variation and shift the recurrence-free survival time from 12 to 42 months (Fig 2B).

Fig. 2.

Fig. 2.

Gene-gene interactions in the phosphoinositide-3-kinase (PI3K), phosphatase and tensin homolog (PTEN), v-akt murine thymoma viral oncogene homolog (AKT), and mammalian target of rapamycin (mTOR) pathway that modified recurrence risk and recurrence-free survival in patients with esophageal cancer (EC) treated with a taxane. (A) Survival tree analysis showing the interactions between three SNPs. (B) Kaplan-Meier curves of recurrence-free survival times in patients in the four terminal nodes, as identified by a survival tree analysis. MST, median survival time; HR, hazard ratio.

Associations Between SNPs and Survival

The PI3K/PTEN/AKT/mTOR pathway did not appear to be a large contributor to variation in survival times. Of the 16 SNPs assayed, only four were found to be significantly associated with survival in any of the treatment groups: AKT1:rs1130214, AKT2:rs892119, FRAP1:rs11121704, and FRAP1:rs2295080 (Table 2).

Table 2.

PI3K/PTEN/AKT/mTOR Pathway Genotypes and Survival

SNP and Genotype Any of the Three
Fluoropyrimidine + Any
No. Alive/Dead HR* 95% CI P No. Alive/Dead HR* 95% CI P
AKT1:rs3803304
    CC 45/41 1 (reference) 43/40 1 (reference)
    CG 38/41 1.12 0.70 to 1.80 .645 37/40 1.14 0.7 to 1.84 .602
    GG 8/4 1.09 0.36 to 3.33 .876 8/4 1.18 0.39 to 3.59 .775
    CG + GG 1.12 0.71 to 1.76 .641 1.14 0.72 to 1.82 .579
AKT1:rs2498804
    GG 34/36 1 (reference) 32/35 1 (reference)
    GT 47/46 1.14 0.69 to 1.90 .601 46/45 1.18 0.71 to 1.98 .518
    TT 9/6 1.50 0.57 to 3.97 .409 9/6 1.64 0.62 to 4.34 .321
    GT + TT 1.18 0.72 to 1.93 .513 1.23 0.74 to 2.03 .424
AKT1:rs2494738
    AA 75/79 1 (reference) 72/77 1 (reference)
    AG 17/8 0.84 0.38 to 1.85 .668 17/8 0.89 0.41 to 1.95 .772
AKT1:rs1130214
    GG 46/42 1 (reference) 43/41 1 (reference)
    GT 40/44 1.26 0.76 to 2.09 .372 40/43 1.23 0.74 to 2.04 .426
    TT 6/3 1.98 0.52 to 7.51 .315 6/3 2.13 0.56 to 8.15 .270
    GT + TT 1.28 0.77 to 2.11 .337 1.25 0.76 to 2.06 .386
AKT2:rs892119
    AA 69/59 1 (reference) 69/57 1 (reference)
    AG 22/26 1.31 0.74 to 2.31 .354 20/26 1.47 0.83 to 2.61 .189
    GG 1/4 1.08 0.32 to 3.70 .898 0/4
    AG + GG 1.27 0.75 to 2.16 .378 1.40 0.81 to 2.4 .229
AKT2:rs8100018
    CC 46/39 1 (reference) 44/38 1 (reference)
    CG 35/42 1.06 0.63 to 1.77 .825 34/41 1.03 0.61 to 1.74 .900
    GG 10/6 0.65 0.26 to 1.66 .370 10/6 0.63 0.25 to 1.61 .335
    CG + GG 0.96 0.60 to 1.56 .884 0.94 0.58 to 1.53 .800
FRAP1:rs11121704
    CC 43/45 1 (reference) 41/43 1 (reference)
    CT 44/34 0.84 0.50 to 1.43 .529 43/34 0.87 0.51 to 1.48 .599
    TT 5/9 3.53 1.48 to 8.39 .004 5/9 3.82 1.58 to 9.23 .003
    CT + TT 1.04 0.63 to 1.71 .878 1.08 0.65 to 1.78 .777
FRAP1:rs2295080
    GG 35/33 1 (reference) 33/31 1 (reference)
    GT 47/38 0.96 0.53 to 1.75 .906 46/38 1.00 0.54 to 1.84 .999
    TT 6/12 4.19 1.83 to 9.61 .001 6/12 4.66 1.99 to 10.94 .0004
    GT + TT 1.25 0.72 to 2.17 .433 1.30 0.74 to 2.31 .354
PIK3CA:rs7651265
    AA 70/65 1 (reference) 67/64 1 (reference)
    AG 18/20 1.18 0.67 to 2.10 .567 18/19 1.17 0.65 to 2.09 .602
    GG 2/0 2/0
    AG + GG 1.02 0.59 to 1.78 .939 1.04 0.59 to 1.82 .902
PIK3CA:rs7640662
    CC 73/67 1 (reference) 72/65 1 (reference)
    CG 18/20 0.89 0.50 to 1.58 .685 16/20 0.99 0.55 to 1.77 .969
    GG 1/2 2.13 0.45 to 10.11 .341 1/2 2.17 0.46 to 10.25 .330
    CG + GG 0.95 0.55 to 1.66 .869 1.06 0.61 to 1.84 .844
PIK3CA:rs7621329
    CC 59/56 1 (reference) 57/55 1 (reference)
    CT 23/24 0.95 0.55 to 1.65 .857 22/23 0.97 0.56 to 1.67 .911
    TT 6/3 0.72 0.20 to 2.62 .617 6/3 0.85 0.23 to 3.1 .810
    CT + TT 0.91 0.55 to 1.52 .725 0.95 0.57 to 1.59 .854
PIK3CA:rs6443624
    AA 56/52 1 (reference) 54/51 1 (reference)
    AC 26/32 1.47 0.85 to 2.54 .163 25/31 1.53 0.88 to 2.65 .128
    CC 9/5 0.84 0.30 to 2.35 .738 9/5 0.92 0.33 to 2.57 .867
    AC + CC 1.31 0.79 to 2.18 .290 1.39 0.83 to 2.32 .213
SNP and Genotype Platinum Compound + Any
Taxane + Any
No. Alive/Dead HR* 95% CI P No. Alive/Dead HR* 95% CI P
AKT1:rs3803304
    CC 31/32 1 (reference) 25/15 1 (reference)
    CG 24/34 1.51 0.85 to 2.67 .159 24/20 1.88 0.81 to 4.37 .143
    GG 6/2 0.70 0.15 to 3.25 .653 4/3 2.17 0.35 to 13.3 .403
    CG + GG 1.40 0.80 to 2.43 .239 1.90 0.83 to 4.36 .129
AKT1:rs2498804
    GG 21/27 1 (reference) 21/12 1 (reference)
    GT 33/38 1.28 0.69 to 2.37 .434 26/22 1.42 0.51 to 3.92 .503
    TT 6/4 1.21 0.37 to 3.99 .757 5/5 2.48 0.57 to 10.77 .224
    GT + TT 1.27 0.69 to 2.33 .436 1.54 0.57 to 4.15 .395
AKT1:rs2494738
    AA 49/61 1 (reference) 45/36 1 (reference)
    AG 12/8 0.84 0.37 to 1.91 .683 9/3 0.68 0.17 to 2.67 .576
AKT1:rs1130214
    GG 33/35 1 (reference) 28/18 1 (reference)
    GT 25/33 1.19 0.66 to 2.15 .562 23/19 1.69 0.73 to 3.94 .222
    TT 3/2 1.85 0.37 to 9.26 .455 3/3 8.92 1.56 to 51.17 .014
    GT + TT 1.21 0.67 to 2.18 .521 1.82 0.79 to 4.19 .157
AKT2:rs892119
    AA 45/48 1 (reference) 41/24 1 (reference)
    AG 16/18 1.38 0.71 to 2.68 .343 12/14 3.27 1.27 to 8.40 .014
    GG 0/4 1/2 6.25 0.89 to 43.88 .065
    AG + GG 1.39 0.75 to 2.59 .292 3.54 1.43 to 8.78 .006
AKT2:rs8100018
    CC 30/29 1 (reference) 26/23 1 (reference)
    CG 24/39 1.16 0.65 to 2.09 .617 22/13 0.76 0.32 to 1.83 .547
    GG 6/1 0.17 0.02 to 1.30 .088 6/4 0.23 0.05 to 1.04 .056
    CG + GG 1.01 0.56 to 1.79 .985 0.55 0.25 to 1.21 .138
FRAP1:rs11121704
    CC 31/38 1 (reference) 25/20 1 (reference)
    CT 26/27 0.97 0.53 to 1.76 .920 28/14 0.58 0.23 to 1.45 .244
    TT 4/5 2.77 0.85 to 9.00 .091 1/6 7.03 1.81 to 27.35 .005
    CT + TT 1.12 0.64 to 1.97 .692 0.85 0.37 to 1.97 .710
FRAP1:rs2295080
    GG 25/28 1 (reference) 20/13 1 (reference)
    GT 27/30 0.99 0.51 to 1.91 .971 33/18 0.32 0.10 to 1.01 .053
    TT 5/7 2.66 0.91 to 7.75 .073 1/7 8.28 2.02 to 33.92 .003
    GT + TT 1.15 0.61 to 2.16 .663 0.73 0.26 to 2.05 .554
PIK3CA:rs7651265
    AA 48/50 1 (reference) 41/30 1 (reference)
    AG 10/17 1.73 0.85 to 3.49 .130 11/8 0.78 0.25 to 2.38 .661
    GG 2/0
    AG + GG 1.43 0.72 to 2.82 .305
PIK3CA:rs7640662
    CC 46/53 1 (reference) 43/32 1 (reference)
    CG 14/16 0.79 0.41 to 1.53 .483 10/7 1.03 0.37 to 2.86 .952
    GG 1/2 2.18 0.45 to 10.64 .335 1/1 0.87 0.09 to 8.24 .905
    CG + GG 0.87 0.46 to 1.63 .665 1.00 0.39 to 2.60 .992
PIK3CA:rs7621329
    CC 38/41 1 (reference) 35/27 1 (reference)
    CT 15/21 1.19 0.61 to 2.35 .612 15/9 0.59 0.21 to 1.69 .328
    TT 5/2 0.74 0.16 to 3.57 .712 3/2 1.38 0.24 to 7.87 .716
    CT + TT 1.11 0.59 to 2.07 .752 0.70 0.27 to 1.81 .459
PIK3CA:rs6443624
    AA 38/40 1 (reference) 32/25 1 (reference)
    AC 15/26 1.55 0.82 to 2.93 .173 17/12 1.58 0.62 to 4.01 .336
    CC 7/4 1.28 0.41 to 4.02 .675 4/3 1.22 0.21 to 7.05 .828
    AC + CC 1.50 0.82 to 2.75 .186 1.52 0.62 to 3.71 .360
SNP and Genotype Any of the Three
Fluoropyrimidine + Any
No. Alive/Dead HR* 95% CI P No. Alive/Dead HR* 95% CI P
PIK3CA:rs2699887
    AA 57/56 1 (reference) 56/54 1 (reference)
    AG 26/28 1.15 0.68 to 1.96 .601 24/28 1.28 0.75 to 2.2 .362
    GG 5/4 0.48 0.15 to 1.50 .206 5/4 0.50 0.16 to 1.56 .230
    AG + GG 0.99 0.60 to 1.65 .984 1.10 0.65 to 1.84 .728
PTEN:rs2299939
    AA 62/58 1 (reference) 60/56 1 (reference)
    AC 26/24 1.45 0.84 to 2.51 .186 26/24 1.46 0.84 to 2.52 .181
    CC 3/5 1.35 0.44 to 4.09 .598 3/5 1.40 0.45 to 4.3 .559
    AC + CC 1.43 0.86 to 2.40 .172 1.45 0.86 to 2.43 .163
PTEN:rs12569998
    GG 68/71 1 (reference) 66/71 1 (reference)
    GT 21/17 0.63 0.34 to 1.18 .148 20/15 0.62 0.33 to 1.16 .137
    TT 1/1 3.52 0.40 to 30.89 .257 1/1 4.64 0.51 to 41.97 .172
    GT + TT 0.66 0.36 to 1.22 .188 0.65 0.35 to 1.21 .178
PTEN:rs12357281
    CC 72/72 1 (reference) 70/70 1 (reference)
    CG 15/14 0.78 0.40 to 1.52 .468 14/14 0.77 0.39 to 1.52 .458
    GG 1/0 0/0
    CG + GG 0.77 0.39 to 1.50 .444
SNP and Genotype Platinum Compound + Any
Taxane + Any
No. Alive/Dead HR* 95% CI P No. Alive/Dead HR* 95% CI P
PIK3CA:rs2699887
    AA 35/45 1 (reference) 36/24 1 (reference)
    AG 19/21 0.77 0.41 to 1.42 .397 12/12 2.32 0.84 to 6.38 .104
    GG 4/3 0.30 0.07 to 1.27 .101 4/3 0.44 0.11 to 1.80 .253
    AG + GG 0.68 0.37 to 1.23 .203 1.15 0.49 to 2.67 .749
PTEN:rs2299939
    AA 42/45 1 (reference) 36/26 1 (reference)
    AC 16/19 1.47 0.78 to 2.79 .234 16/12 1.43 0.58 to 3.52 .432
    CC 2/5 1.99 0.56 to 7.08 .286 1/1 0.67 0.04 to 10.91 .778
    AC + CC 1.55 0.85 to 2.80 .149 1.33 0.56 to 3.15 .515
PTEN:rs12569998
    GG 45/56 1 (reference) 42/31 1 (reference)
    GT 14/13 0.62 0.28 to 1.37 .234 10/8 0.35 0.12 to 1.02 .055
    TT 0/1 1/1 3.11 0.21 to 46.74 .412
    GT + TT 0.70 0.33 to 1.52 .373 0.42 0.15 to 1.18 .099
PTEN:rs12357281
    CC 50/60 1 (reference) 40/30 1 (reference)
    CG 9/8 0.73 0.30 to 1.76 .478 11/8 1.16 0.42 to 3.16 .777
    GG 0/0 1/0
    CG + GG 1.11 0.41 to 3.02 .837

Abbreviations: PI3K, phosphoinositide-3-kinase; PTEN, phosphatase and tensin homolog; AKT, v-akt murine thymoma viral oncogene homolog; mTOR, mammalian target of rapamycin; SNP, single nucleotide polymorphism; HR, hazard ratio.

*

Adjusted for age, sex, smoking status, alcohol consumption, radiation dosage, chemoradiotherapy sequence, clinical stage, chemotherapy regimens, histologic tumor type, tumor location, pathologic stage, and histologic viability.

FRAP1 genetic variations.

Both of the FRAP1 SNPs genotyped were significant with HRs of 3.53 (95% CI, 1.48 to 8.39) and 4.19 (95% CI, 1.83 to 9.61) for homozygous variants of FRAP1:rs11121704 and FRAP1:rs2295080, respectively (Table 2). These same SNPs were also associated with increased risk of death in the fluoropyrimidine and taxane treatment groups.

AKT1 and AKT2 genetic variations.

AKT1 and AKT2 SNPs were associated with survival in taxane-treated patients only. AKT1:rs1130214 resulted in a nearly nine-fold (HR, 8.92; 95% CI, 1.56 to 51.17) increased risk of death in these patients, while AKT2:rs892119 was associated with a 3.5-fold increase (95% CI, 1.43 to 8.78). The relationships between AKT1:rs1130214 and AKT2:rs892119 with survival were not observed in any of the other treatment groups, suggesting that the effect of these genetic variants may be restricted to taxane-based therapy. However, the small sample size of the taxane group may be a factor contributing to these results.

Associations Between SNPs and Response to Therapy

Response to therapy was analyzed for associations with genetic variations in the pathway with three SNPs showing significance: AKT2:rs892119, AKT1:rs3803304, and FRAP1:rs1121704 (Table 3).

Table 3.

PI3K/PTEN/AKT/mTOR Pathway Genotypes and Response to Therapy

SNP and Genotype Any of the Three
Fluoropyrimidine + Any
Response/No Response OR* 95% CI P Response/No Response OR* 95% CI P
AKT1:rs3803304
    CC 25/61 1 (reference) 25/58 1 (reference)
    CG 35/44 0.50 0.25 to 0.99 .047 34/43 0.54 0.27 to 1.08 .083
    GG 4/8 0.93 0.25 to 3.53 .920 4/8 0.96 0.25 to 3.63 .953
    CG + GG 0.54 0.28 to 1.05 .071 0.59 0.30 to 1.15 .118
AKT1:rs2498804
    GG 23/47 1 (reference) 23/44 1 (reference)
    GT 36/57 0.75 0.37 to 1.52 .419 35/56 0.83 0.40 to 1.70 .605
    TT 5/10 1.15 0.33 to 4.01 .821 5/10 1.21 0.35 to 4.20 .767
    GT + TT 0.79 0.40 to 1.58 .510 0.87 0.43 to 1.75 .703
AKT1:rs2494738
    AA 59/95 1 (reference) 58/91 1 (reference)
    AG 6/19 2.01 0.73 to 5.57 .177 6/19 2.07 0.75 to 5.75 .163
AKT1:rs1130214
    GG 31/57 1 (reference) 30/54 1 (reference)
    GT 31/53 1.12 0.56 to 2.25 .754 31/52 1.06 0.52 to 2.15 .879
    TT 4/5 0.74 0.17 to 3.2 .693 4/5 0.71 0.16 to 3.11 .652
    GT + TT 1.08 0.54 to 2.13 .833 1.02 0.51 to 2.04 .961
AKT2:rs892119
    AA 53/75 1 (reference) 53/73 1 (reference)
    AG 13/35 2.54 1.14 to 5.65 .023 12/34 2.68 1.18 to 6.06 .018
    GG 0/5 0/4
    AG + GG 2.81 1.27 to 6.21 .010 2.98 1.33 to 6.68 .008
AKT2:rs8100018
    CC 33/52 1 (reference) 32/50 1 (reference)
    CG 26/51 1.30 0.66 to 2.58 .446 26/49 1.20 0.60 to 2.39 .612
    GG 6/10 0.83 0.24 to 2.84 .769 6/10 0.83 0.24 to 2.84 .768
    CG + GG 1.22 0.63 to 2.36 .549 1.13 0.58 to 2.20 .713
FRAP1:rs11121704
    CC 37/51 1 (reference) 36/48 1 (reference)
    CT 25/53 1.35 0.68 to 2.67 .395 25/52 1.32 0.66 to 2.63 .437
    TT 4/10 1.92 0.53 to 6.87 .318 4/10 1.88 0.52 to 6.77 .336
    CT + TT 1.43 0.74 to 2.74 .283 1.40 0.72 to 2.70 .320
FRAP1:rs2295080
    GG 29/39 1 (reference) 28/36 1 (reference)
    GT 26/59 1.60 0.77 to 3.32 .210 26/58 1.58 0.76 to 3.31 .224
    TT 8/10 0.99 0.33 to 2.95 .983 8/10 0.99 0.33 to 2.97 .985
    GT + TT 1.44 0.72 to 2.86 .301 1.43 0.71 to 2.86 .316
PIK3CA:rs7651265
    AA 53/82 1 (reference) 52/79 1 (reference)
    AG 11/27 1.45 0.61 to 3.42 .399 11/26 1.40 0.59 to 3.33 .443
    GG 0/2 0/2
    AG + GG 1.61 0.69 to 3.75 .273 1.56 0.66 to 3.65 .307
PIK3CA:rs7640662
    CC 50/90 1 (reference) 50/87 1 (reference)
    CG 14/24 0.83 0.37 to 1.84 .639 13/23 0.85 0.37 to 1.95 .704
    GG 2/1 0.34 0.03 to 4.00 .390 2/1 0.35 0.03 to 4.21 .410
    CG + GG 0.76 0.35 to 1.65 .493 0.79 0.35 to 1.74 .552
PIK3CA:rs7621329
    CC 44/71 1 (reference) 43/69 1 (reference)
    CT 15/32 1.27 0.57 to 2.84 .562 15/30 1.16 0.51 to 2.61 .726
    TT 3/6 1.64 0.36 to 7.37 .519 3/6 1.68 0.37 to 7.64 .501
    CT + TT 1.34 0.64 to 2.79 .442 1.25 0.59 to 2.63 .560
PIK3CA:rs6443624
    AA 44/64 1 (reference) 43/62 1 (reference)
    AC 18/40 1.57 0.74 to 3.35 .239 18/38 1.46 0.68 to 3.13 .328
    CC 4/10 2.06 0.57 to 7.48 .270 4/10 2.02 0.55 to 7.33 .288
    AC + CC 1.67 0.82 to 3.36 .155 1.56 0.77 to 3.19 .217
SNP and Genotype Platinum Compound + Any
Taxane + Any
Response/No Response OR* 95% CI P Response/No Response OR* 95% CI P
AKT1:rs3803304
    CC 18/45 1 (reference) 12/28 1 (reference)
    CG 26/32 0.45 0.19 to 1.04 .060 18/26 0.56 0.20 to 1.55 .265
    GG 2/6 1.44 0.24 to 8.72 .694 3/4 0.67 0.11 to 4.05 .664
    CG + GG 0.51 0.23 to 1.16 .108 0.58 0.22 to 1.54 .271
AKT1:rs2498804
    GG 15/33 1 (reference) 12/21 1 (reference)
    GT 27/44 0.67 0.28 to 1.61 .373 18/30 0.83 0.29 to 2.38 .736
    TT 3/7 1.29 0.26 to 6.38 .752 4/6 0.90 0.18 to 4.49 .896
    GT + TT 0.73 0.31 to 1.72 .470 0.85 0.31 to 2.34 .746
AKT1:rs2494738
    AA 41/69 1 (reference) 31/50 1 (reference)
    AG 5/15 1.79 0.56 to 5.72 .323 3/9 2.54 0.52 to 12.35 .248
AKT1:rs1130214
    GG 23/45 1 (reference) 19/27 1 (reference)
    GT 23/35 0.89 0.38 to 2.07 .790 14/28 1.39 0.51 to 3.85 .521
    TT 1/4 2.07 0.20 to 21.71 .545 2/4 1.15 0.17 to 7.85 .886
    GT + TT 0.94 0.41 to 2.17 .888 1.36 0.51 to 3.62 .540
AKT2:rs892119
    AA 37/56 1 (reference) 28/37 1 (reference)
    AG 10/24 2.18 0.85 to 5.60 .105 7/19 3.68 1.05 to 12.89 .042
    GG 0/4 0/3
    AG + GG 2.46 0.97 to 6.23 .059 4.12 1.18 to 14.37 .026
AKT2:rs8100018
    CC 23/36 1 (reference) 20/29 1 (reference)
    CG 20/43 1.35 0.60 to 3.06 .471 11/24 1.22 0.43 to 3.44 .703
    GG 3/4 0.85 0.15 to 4.67 .849 4/6 0.47 0.08 to 2.70 .396
    CG + GG 1.29 0.58 to 2.85 .536 1.06 0.39 to 2.84 .914
FRAP1:rs11121704
    CC 27/42 1 (reference) 21/24 1 (reference)
    CT 18/35 1.12 0.49 to 2.56 .779 12/30 2.73 0.97 to 7.66 .057
    TT 2/7 2.81 0.48 to 16.40 .250 2/5 2.94 0.47 to 18.52 .250
    CT + TT 1.29 0.59 to 2.82 .521 2.76 1.04 to 7.37 .042
FRAP1:rs2295080
    GG 22/31 1 (reference) 15/18 1 (reference)
    GT 17/40 1.69 0.69 to 4.10 .248 16/35 2.11 0.74 to 6.03 .165
    TT 5/7 1.15 0.29 to 4.49 .843 3/5 1.64 0.30 to 9.04 .569
    GT + TT 1.55 0.68 to 3.58 .300 2.02 0.74 to 5.51 .172
PIK3CA:rs7651265
    AA 38/60 1 (reference) 29/42 1 (reference)
    AG 8/19 1.73 0.59 to 5.05 .317 4/15 2.10 0.55 to 8.02 .279
    GG 0/2 0/0
    AG + GG 1.96 0.68 to 5.63 .211
PIK3CA:rs7640662
    CC 32/67 1 (reference) 28/47 1 (reference)
    CG 13/17 0.53 0.21 to 1.35 .184 6/11 0.85 0.25 to 2.93 .794
    GG 2/1 0.23 0.02 to 2.87 .255 1/1 0.97 0.04 to 24.89 .987
    CG + GG 0.49 0.20 to 1.19 .116 0.86 0.27 to 2.79 .804
PIK3CA:rs7621329
    CC 29/50 1 (reference) 25/37 1 (reference)
    CT 12/24 1.19 0.45 to 3.15 .725 7/17 1.51 0.47 to 4.86 .490
    TT 2/5 2.02 0.32 to 12.63 .452 2/3 0.94 0.14 to 6.45 .948
    CT + TT 1.33 0.55 to 3.23 .530 1.35 0.47 to 3.85 .573
PIK3CA:rs6443624
    AA 30/48 1 (reference) 24/33 1 (reference)
    AC 14/27 1.28 0.51 to 3.21 .597 9/20 1.84 0.62 to 5.43 .271
    CC 3/8 2.50 0.56 to 11.11 .227 2/5 1.36 0.22 to 8.58 .744
    AC + CC 1.50 0.64 to 3.51 .353 1.72 0.64 to 4.61 .280
SNP and Genotype Any of the Three
Fluoropyrimidine + Any
Response/No Response OR* 95% CI P Response/No Response OR* 95% CI P
PIK3CA:rs2699887
    AA 41/72 1 (reference) 41/69 1 (reference)
    AG 20/34 0.94 0.45 to 1.97 .880 19/33 0.98 0.47 to 2.08 .965
    GG 3/6 1.06 0.22 to 5.06 .939 3/6 1.14 0.24 to 5.46 .874
    AG + GG 0.96 0.48 to 1.93 .910 1.00 0.49 to 2.05 .991
PTEN:rs2299939
    AA 48/72 1 (reference) 48/68 1 (reference)
    AC 16/34 1.28 0.61 to 2.68 .507 16/34 1.33 0.64 to 2.79 .446
    CC 1/7 4.63 0.51 to 42.07 .174 1/7 4.92 0.54 to 45.00 .158
    AC + CC 1.47 0.72 to 2.98 .289 1.53 0.75 to 3.11 .243
PTEN:rs12569998
    GG 54/85 1 (reference) 53/84 1 (reference)
    GT 10/28 1.73 0.73 to 4.15 .216 10/25 1.50 0.62 to 3.64 .369
    TT 1/1 0.64 0.04 to 11.45 .761 1/1 0.59 0.03 to 10.76 .725
    GT + TT 1.63 0.70 to 3.79 .259 1.41 0.60 to 3.34 .430
PTEN:rs12357281
    CC 51/93 1 (reference) 51/89 1 (reference)
    CG 9/20 1.24 0.50 to 3.03 .642 8/20 1.44 0.57 to 3.66 .440
    GG 1/0 1/0
    CG + GG 1.11 0.46 to 2.66 .813 1.26 0.51 to 3.10 .614
SNP and Genotype Platinum Compound + Any
Taxane + Any
Response/No Response OR* 95% CI P Response/No Response OR* 95% CI P
PIK3CA:rs2699887
    AA 26/54 1 (reference) 25/35 1 (reference)
    AG 17/23 0.54 0.22 to 1.31 .171 7/17 1.73 0.53 to 5.63 .362
    GG 3/4 0.47 0.08 to 2.88 .417 2/5 1.94 0.28 to 13.46 .502
    AG + GG 0.53 0.23 to 1.23 .139 1.78 0.61 to 5.22 .295
PTEN:rs2299939
    AA 36/51 1 (reference) 25/37 1 (reference)
    AC 9/26 1.84 0.73 to 4.65 .200 9/19 1.27 0.44 to 3.65 .663
    CC 1/6 4.12 0.43 to 39.61 .220 0/2
    AC + CC 2.05 0.85 to 4.98 .112 1.50 0.53 to 4.22 .440
PTEN:rs12569998
    GG 39/62 1 (reference) 30/43 1 (reference)
    GT 6/21 2.22 0.74 to 6.67 .155 4/14 2.29 0.56 to 9.34 .246
    TT 1/0 1/1 0.59 0.03 to 10.84 .723
    GT + TT 1.89 0.66 to 5.39 .237 1.88 0.51 to 6.87 .342
PTEN:rs12357281
    CC 37/73 1 (reference) 26/44 1 (reference)
    CG 7/10 0.71 0.23 to 2.17 .549 5/14 1.82 0.53 to 6.20 .340
    GG 0/0 1/0
    CG + GG 1.46 0.46 to 4.67 .522

Abbreviations: SNP, single nucleotide polymorphism; OR, odds ratio; PI3K, phosphoinositide-3-kinase; PTEN, phosphatase and tensin homolog; AKT, v-akt murine thymoma viral oncogene homolog; mTOR, mammalian target of rapamycin.

*

Adjusted for age, sex, smoking status, alcohol consumption, radiation dosage, chemoradiotherapy sequence, clinical stage, chemotherapy regimens, histologic tumor type, tumor location, pathologic stage, and histologic viability.

AKT1 genetic variation.

Patients heterozygous for AKT1:rs3803304 experienced a better response to chemoradiotherapy than those with a wild-type genotype (OR, 0.50; 95% CI, 0.25 to 0.99).

AKT2 genetic variation.

Interestingly, the same AKT2 SNP (rs892119) that had been significantly associated with increased risk of both recurrence and death was also found to be associated with a poorer response (OR, 2.81; 95% CI, 1.27 to 6.21). This effect was also observed in the fluoropyrimidine (OR, 2.98; 95% CI, 1.33 to 6.68), and taxane (OR, 4.12; 95% CI, 1.18 to14.37) treatment groups.

FRAP1 genetic variation.

The FRAP1:rs11121704 SNP, which was associated with poor survival, also contributed to a poor pathologic response (OR, 2.76; 95% CI, 1.04 to 7.37) in patients treated with a taxane with at least one variant allele.

DISCUSSION

The PI3K/PTEN/AKT/mTOR pathway plays an important role in balancing cell growth and death. This pathway is often activated in several cancer types—including EC—and has been shown to be important in the development of resistance to several commonly used classes of chemotherapeutic agents. In this study, we determined whether genetic variations in the genes for PI3K, PTEN, AKT1, AKT2, and mTOR were associated with variation in recurrence, survival, and pathologic response. To our knowledge, ours is the first study to apply a tagging SNP approach to determine the role of this pathway in clinical outcomes for any cancer type.

Significant associations were observed between several SNPs and clinical outcomes. In individual SNP analyses, we identified seven SNPs associated with recurrence risk and recurrence-free survival rates. Patients treated with any of the three chemotherapeutic agents had a dramatic increase in their recurrence risk with two unfavorable AKT1 or AKT2 genotypes. This increased risk was also observed in fluoropyrimidine-, platinum-, and taxane-treated patients (6-, 10-, and > 80-fold increases, respectively). Although these results require further validation, they indicate that variations in these genes play a role in modulating EC recurrence. The importance of AKT2:rs892119 in determining recurrence risk was further supported in a survival tree analysis of patients treated with taxanes. This SNP was the basis of the initial split in the tree, suggesting that genetic variation tagged by this SNP is a major risk factor for developing a recurrence.

Interestingly, AKT2:rs892119 had consistent effect on variation in all three clinical outcomes studied and was not drug specific. AKT2:rs892119 is intronic and represents genetic variation across five SNPs genotyped in the Centre d'Etude du Polymorphisme Humain population. It is possible that AKT2:rs892119 is the functional SNP through alterations in normal splicing patterns or transcription of AKT2. However, it is likely that this SNP is not the functional variant but a surrogate marker for the underling genetic variation within that region on the genome. Additional studies will be required to identify the causative sequence variation and the mechanism(s) responsible for our observations. Nevertheless, our results suggest that the functional SNP tagged by AKT2:rs892119 results in activation of AKT2 and increased signaling through this pathway. This is of particularly intriguing because AKT plays a major role in regulating cell survival and growth.13 AKT activation due to overexpression or gene amplification has been shown to be involved in resistance to several chemotherapeutic agents for cancers such as lung, uterine, and ovarian cancer.1922 However, to our knowledge, no studies have shown associations between common genetic variations in AKT and clinical outcomes in any cancer type.

We selected tagging SNPs for five genes in the PI3K/PTEN/AKT/mTOR pathway. These genes were chosen because they represent the core functional components of the pathway, but this pathway is complex, with several other genes warranting investigation on the basis of the results of this study. Two phosphoinositide-dependent kinases—PDK1 and PDK2—are responsible for phosphorylating AKT, resulting in AKT activation.29 Directly downstream of AKT in the pathway are the tuberous sclerosis complex (TSC) tumor suppressor genes—TSC1 and TSC2. AKT phosphorylation of TSC2 inhibits the function of this complex, allowing for the activation of mTOR.30 Genetic variation in these four genes—PDK1, PDK2, TSC1, and TSC2—may contribute to additional variation in clinical outcome, especially in combination with genetically altered AKT.

PTEN acts as a negative regulator of PI3K/AKT/mTOR signaling by reversing PIP3 activation. Loss of PTEN function results in unrestrained signaling through this pathway and ultimately increased cell growth and proliferation.31 This is a common feature of cancer and has been observed in several cancer types, including brain, breast, and prostate.3234 However, PTEN mutations occur infrequently in EC,35 and decreased protein expression is found in approximately 40% of EC tumors.36 Tachibana et al37 reported that patients with positive PTEN expression in the nucleus had higher overall survival rates than did those without. In our study, we found that a SNP located in an intron of PTEN was associated with decreased recurrence risk—possibly due to increased expression of PTEN protein in esophageal tissue. Because PTEN:rs12357281 is a tagging SNP, it is likely not the functional SNP. Although, as with all tagging SNPs, there is a possibility that it is the functional variant. However, even without the identification of the functional SNP, our results imply that common variation in PTEN is an important modulator of recurrence risk in patients with EC. Furthermore, the gene-gene interactions identified in our survival tree analysis highlight the complexity of the effects of genetic variation on recurrence risk and recurrence-free survival and support the importance of AKT2:rs892119 and PTEN:rs12357281 in modulating these outcomes.

In contrast to PTEN that acts as a brake for this pathway, increased mTOR (FRAP1) activity results in increased growth signals through phosphorylation of 4EBP and p70S6K.38 mTOR is activated in many cancers, including EC.18 In our study, patients who were homozygous for either of the FRAP1 SNPs had an increased risk of death. In addition, FRAP1:rs11121704 homozygosity was associated with a poor response to taxane. These observations are consistent with those of increased mTOR signaling, resulting in poorer clinical outcomes for patients with genetically polymorphic FRAP1.

We observed that genetic variations in the PI3K/PTEN/AKT/mTOR pathway appeared to have more effect on clinical outcomes in patients treated with taxanes than in patients treated with either a fluoropyrimidine or platinum-containing agent. This may be partly due to the small sample size of the taxane treatment group, but this observation was particularly evident for associations with recurrence risk, with at least one SNP from every gene studied found to be significant in patients treated with taxane. The pathway's involvement in clinical outcomes in platinum-treated patients was limited to associations with recurrence risk for AKT1 and AKT2 SNPs. Similarly, few significant associations were found between clinical outcome and genetic variation in fluoropyrimidine-treated patients. The observations in the fluoropyrimidine and platinum agent groups were replicated in the larger group of patients treated with any of the three drugs. In contrast, several associations were observed only in the taxane treatment group. Although sample size may be an issue, these drugs have different mechanisms of action, and these differences may account for the differences in association between this pathway and clinical outcome.

In conclusion, we found significant associations between common genetic variants in the PI3K/PTEN/AKT/mTOR pathway and clinical outcomes in patients with EC. Although we limited our analyses to patients receiving chemoradiotherapy, we are not able to conclude that these markers are predictive of drug response since we are unable to exclude that they may be prognostic factors. It would be interesting to analyze these SNPs in a control group undergoing surgery alone to assess their prognostic impact, but we did not have enough patients in this category in this study. Nevertheless, if validated as predictive markers for chemotherapy, these results, with the integration of clinical, epidemiological, and genetic data, could become the basis for individualizing therapy For example, markers predictive of a good drug response could be useful for preselecting patients to a specific chemotherapeutic. In contrast, patients with a poor marker signature who are predicted to receive no benefits from chemotherapy may receive only surgery. The ultimate goal is to allow for the selection of the optimal therapy that would provide the most benefit and least toxicity for patients with EC.

Supplementary Material

[Data Supplement]

Appendix

The Appendix is included in the full-text version of this article, available online at www.jco.org. It is not included in the PDF version (via Adobe® Reader®).

Table A1.

Tagging SNP Characteristics

SNP Gene SNP Type SNP Location Alleles MAF No. of SNPs Tagged
rs3803304 AKT1 Intron chr14:104310191 C/G 0.225 1
rs2498804 AKT1 3′-flanking region chr14:104304140 G/T 0.333 5
rs2494738 AKT1 Intron chr14:104317731 A/G 0.117 1
rs1130214 AKT1 Intron/5′-UTR chr14:104330779 G/T 0.275 1
rs892119 AKT2 Intron chr19:45451912 A/G 0.142 5
rs8100018 AKT2 Intron chr19:45443863 C/G 0.258 8
rs11121704 FRAP1 Intron chr1:11216546 C/T 0.293 38
rs2295080 FRAP1 5′-flanking region chr1:11245215 G/T 0.308 2
rs7651265 PIK3CA Intron chr3:180375723 A/G 0.125 1
rs7640662 PIK3CA Intron chr3:180384695 C/G 0.150 1
rs7621329 PIK3CA Intron chr3:180357568 C/T 0.183 19
rs6443624 PIK3CA Intron chr3:180380368 A/C 0.242 1
rs2699887 PIK3CA Intron chr3:180349102 A/G 0.233 2
rs2299939 PTEN Intron chr10:89647130 A/C 0.133 3
rs12569998 PTEN Intron chr10:89664137 G/T 0.183 5
rs12357281 PTEN Intron chr10:89690651 C/G 0.110 2

Abbreviations: SNP, single nucleotide polymorphism; MAF, minor allele frequency.

Table A2.

Patient Characteristics

Characteristic No. of Patients %
Total 186 Sex
    Male 161 86.6
    Female 25 13.4
Age, years
    Mean 60.8
    SD 9.32
    Range 32-79
Smoking status
    Never 77 41.4
    Ever 109 58.6
Alcohol use, > 4 ounces per day
    No 121 66.9
    Yes 60 33.1
Histological type
    Adenocarcinoma 158 84.9
    Squamous cell carcinoma 28 15.1
Clinical stage
    IIA 94 50.5
    IIB 14 7.5
    III 68 36.6
    IVA 10 5.4
Chemoradiation
    Any of the three, cisplatin, FU, or taxol 182 97.8
    FU plus any 177 95.2
    Cisplatin plus any 132 71.0
    Taxol plus any 94 50.5

Abbreviations: SD, standard deviation; FU, fluorouracil.

Footnotes

Supported in part by Grants no. R01 CA111922 and R25 CA57730 and grants from the Smith, Dallas, Park, and Cantu Families and the Rivercreek Foundation.

Authors' disclosures of potential conflicts of interest and author contributions are found at the end of this article.

AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

The author(s) indicated no potential conflicts of interest.

AUTHOR CONTRIBUTIONS

Conception and design: Michelle Hildebrandt, Hushan Yang, Xifeng Wu

Provision of study materials or patients: Jaffer A. Ajani, Xifeng Wu

Collection and assembly of data: Michelle Hildebrandt, Jie Lin

Data analysis and interpretation: Michelle Hildebrandt, Mien-Chie Hung, Julie G. Izzo, Maosheng Huang, Jie Lin, Xifeng Wu

Manuscript writing: Michelle Hildebrandt, Jaffer A. Ajani, Xifeng Wu

Final approval of manuscript: Michelle Hildebrandt, Hushan Yang, Mien-Chie Hung, Julie G. Izzo, Maosheng Huang, Jie Lin, Jaffer A. Ajani, Xifeng Wu

References

  • 1.American Cancer Society. Cancer Facts and Figures 2008. Atlanta, Ga: American Cancer Society; 2008. [Google Scholar]
  • 2.Brenner B, Ilson DH, Minsky BD. Treatment of localized esophageal cancer. Semin Oncol. 2004;31:554–565. doi: 10.1053/j.seminoncol.2004.04.015. [DOI] [PubMed] [Google Scholar]
  • 3.Iyer R, Wilkinson N, Demmy T, et al. Controversies in the multimodality management of locally advanced esophageal cancer: Evidence-based review of surgery alone and combined-modality therapy. Ann Surg Oncol. 2004;11:665–673. doi: 10.1245/ASO.2004.10.026. [DOI] [PubMed] [Google Scholar]
  • 4.Urba S. Esophageal cancer: Preoperative or definitive chemoradiation. Ann Oncol. 2004;15(suppl 4):iv93–iv96. doi: 10.1093/annonc/mdh910. [DOI] [PubMed] [Google Scholar]
  • 5.Tepper JE, Krasna MJ, Niedzwiecki D. Superiority of trimodality therapy to surgery alone in esophageal cancer: Results of CALGB 9781. J Clin Oncol. 2006;24(suppl):181s. doi: 10.1200/JCO.2007.12.9593. abstr 4012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Burmeister BH, Smithers BM, Gebski V, et al. Surgery alone versus chemoradiotherapy followed by surgery for resectable cancer of the oesophagus: A randomised controlled phase III trial. Lancet Oncol. 2005;6:659–668. doi: 10.1016/S1470-2045(05)70288-6. [DOI] [PubMed] [Google Scholar]
  • 7.Gebski V, Burmeister B, Smithers BM, et al. Survival benefits from neoadjuvant chemoradiotherapy or chemotherapy in oesophageal carcinoma: A meta-analysis. Lancet Oncol. 2007;8:226–234. doi: 10.1016/S1470-2045(07)70039-6. [DOI] [PubMed] [Google Scholar]
  • 8.Walsh TN, Noonan N, Hollywood D, et al. A comparison of multimodal therapy and surgery for esophageal adenocarcinoma. N Engl J Med. 1996;335:462–467. doi: 10.1056/NEJM199608153350702. [DOI] [PubMed] [Google Scholar]
  • 9.Urba SG, Orringer MB, Turrisi A, et al. Randomized trial of preoperative chemoradiation versus surgery alone in patients with locoregional esophageal carcinoma. J Clin Oncol. 2001;19:305–313. doi: 10.1200/JCO.2001.19.2.305. [DOI] [PubMed] [Google Scholar]
  • 10.Evans WE, Relling MV. Pharmacogenomics: Translating functional genomics into rational therapeutics. Science. 1999;286:487–491. doi: 10.1126/science.286.5439.487. [DOI] [PubMed] [Google Scholar]
  • 11.Wu X, Gu J, Wu TT, et al. Genetic variations in radiation and chemotherapy drug action pathways predict clinical outcomes in esophageal cancer. J Clin Oncol. 2006;24:3789–3798. doi: 10.1200/JCO.2005.03.6640. [DOI] [PubMed] [Google Scholar]
  • 12.Nicholson KM, Anderson NG. The protein kinase B/Akt signaling pathway in human malignancy. Cell Signal. 2002;14:381–395. doi: 10.1016/s0898-6568(01)00271-6. [DOI] [PubMed] [Google Scholar]
  • 13.Bellacosa A, Kumar CC, Di Cristofano A, et al. Activation of AKT kinases in cancer: Implications for therapeutic targeting. Adv Cancer Res. 2005;94:29–86. doi: 10.1016/S0065-230X(05)94002-5. [DOI] [PubMed] [Google Scholar]
  • 14.Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell. 2000;103:211–225. doi: 10.1016/s0092-8674(00)00114-8. [DOI] [PubMed] [Google Scholar]
  • 15.Manning BD, Cantley LC. AKT/PKB signaling: Navigating downstream. Cell. 2007;129:1261–1274. doi: 10.1016/j.cell.2007.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Stambolic V, Suzuki A, de la Pompa JL, et al. Negative regulation of PKB/Akt-dependent cell survival by the tumor suppressor PTEN. Cell. 1998;95:29–39. doi: 10.1016/s0092-8674(00)81780-8. [DOI] [PubMed] [Google Scholar]
  • 17.Sagatys E, Garrett CR, Boulware D, et al. Activation of the serine/threonine protein kinase Akt during the progression of Barrett neoplasia. Hum Pathol. 2007;38:1526–1531. doi: 10.1016/j.humpath.2007.03.003. [DOI] [PubMed] [Google Scholar]
  • 18.Hou G, Xue L, Lu Z, et al. An activated mTOR/p70S6K signaling pathway in esophageal squamous cell carcinoma cell lines and inhibition of the pathway by rapamycin and siRNA against mTOR. Cancer Lett. 2007;253:236–248. doi: 10.1016/j.canlet.2007.01.026. [DOI] [PubMed] [Google Scholar]
  • 19.Lee S, Choi EJ, Jin C, et al. Activation of PI3K/Akt pathway by PTEN reduction and PIK3CA mRNA amplification contributes to cisplatin resistance in an ovarian cancer cell line. Gynecol Oncol. 2005;97:26–34. doi: 10.1016/j.ygyno.2004.11.051. [DOI] [PubMed] [Google Scholar]
  • 20.Liu LZ, Zhou XD, Qian G, et al. AKT1 amplification regulates cisplatin resistance in human lung cancer cells through the mammalian target of rapamycin/p70S6K1 pathway. Cancer Res. 2007;67:6325–6332. doi: 10.1158/0008-5472.CAN-06-4261. [DOI] [PubMed] [Google Scholar]
  • 21.Kim SH, Juhnn YS, Song YS. Akt involvement in paclitaxel chemoresistance of human ovarian cancer cells. Ann N Y Acad Sci. 2007;1095:82–89. doi: 10.1196/annals.1397.012. [DOI] [PubMed] [Google Scholar]
  • 22.Gagnon V, Van Themsche C, Turner S, et al. Akt and XIAP regulate the sensitivity of human uterine cancer cells to cisplatin, doxorubicin and taxol. Apoptosis. 2008;13:259–271. doi: 10.1007/s10495-007-0165-6. [DOI] [PubMed] [Google Scholar]
  • 23.Yang X, Fraser M, Moll UM, et al. Akt-mediated cisplatin resistance in ovarian cancer: Modulation of p53 action on caspase-dependent mitochondrial death pathway. Cancer Res. 2006;66:3126–3136. doi: 10.1158/0008-5472.CAN-05-0425. [DOI] [PubMed] [Google Scholar]
  • 24.Murakami D, Tsujitani S, Osaki T, et al. Expression of phosphorylated Akt (pAkt) in gastric carcinoma predicts prognosis and efficacy of chemotherapy. Gastric Cancer. 2007;10:45–51. doi: 10.1007/s10120-006-0410-7. [DOI] [PubMed] [Google Scholar]
  • 25.Chirieac LR, Swisher SG, Ajani JA, et al. Post-therapy pathologic stage predicts survival in patients with esophageal carcinoma receiving preoperative chemoradiation. Cancer. 2005;103:1347–1355. doi: 10.1002/cncr.20916. [DOI] [PubMed] [Google Scholar]
  • 26.Wu TT, Chirieac LR, Abraham SC, et al. Excellent interobserver agreement on grading the extent of residual carcinoma after preoperative chemoradiation in esophageal and esophagogastric junction carcinoma: A reliable predictor for patient outcome. Am J Surg Pathol. 2007;31:58–64. doi: 10.1097/01.pas.0000213312.36306.cc. [DOI] [PubMed] [Google Scholar]
  • 27.Consortium TIH. A haplotype map of the human genome. Nature. 2005;437:1299–1320. doi: 10.1038/nature04226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.de Bakker PI, Yelensky R, Pe'er I, et al. Efficiency and power in genetic association studies. Nat Genet. 2005;37:1217–1223. doi: 10.1038/ng1669. [DOI] [PubMed] [Google Scholar]
  • 29.Alessi DR, James SR, Downes CP, et al. Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Balpha. Curr Biol. 1997;7:261–269. doi: 10.1016/s0960-9822(06)00122-9. [DOI] [PubMed] [Google Scholar]
  • 30.Tee AR, Fingar DC, Manning BD, et al. Tuberous sclerosis complex-1 and -2 gene products function together to inhibit mammalian target of rapamycin (mTOR)-mediated downstream signaling. Proc Natl Acad Sci U S A. 2002;99:13571–13576. doi: 10.1073/pnas.202476899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Sansal I, Sellers WR. The biology and clinical relevance of the PTEN tumor suppressor pathway. J Clin Oncol. 2004;22:2954–2963. doi: 10.1200/JCO.2004.02.141. [DOI] [PubMed] [Google Scholar]
  • 32.Ali IU, Schriml LM, Dean M. Mutational spectra of PTEN/MMAC1 gene: A tumor suppressor with lipid phosphatase activity. J Natl Cancer Inst. 1999;91:1922–1932. doi: 10.1093/jnci/91.22.1922. [DOI] [PubMed] [Google Scholar]
  • 33.Suzuki H, Freije D, Nusskern DR, et al. Interfocal heterogeneity of PTEN/MMAC1 gene alterations in multiple metastatic prostate cancer tissues. Cancer Res. 1998;58:204–209. [PubMed] [Google Scholar]
  • 34.Saal LH, Gruvberger-Saal SK, Persson C, et al. Recurrent gross mutations of the PTEN tumor suppressor gene in breast cancers with deficient DSB repair. Nat Genet. 2008;40:102–107. doi: 10.1038/ng.2007.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hu YC, Lam KY, Tang JC, et al. Mutational analysis of the PTEN/MMAC1 gene in primary oesophageal squamous cell carcinomas. Mol Pathol. 1999;52:353–356. doi: 10.1136/mp.52.6.353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Chang MS, Lee HS, Lee BL, et al. Differential protein expression between esophageal squamous cell carcinoma and dysplasia, and prognostic significance of protein markers. Pathol Res Pract. 2005;201:417–425. doi: 10.1016/j.prp.2005.04.005. [DOI] [PubMed] [Google Scholar]
  • 37.Tachibana M, Shibakita M, Ohno S, et al. Expression and prognostic significance of PTEN product protein in patients with esophageal squamous cell carcinoma. Cancer. 2002;94:1955–1960. doi: 10.1002/cncr.0678. [DOI] [PubMed] [Google Scholar]
  • 38.Guertin DA, Sabatini DM. Defining the role of mTOR in cancer. Cancer Cell. 2007;12:9–22. doi: 10.1016/j.ccr.2007.05.008. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

[Data Supplement]

Articles from Journal of Clinical Oncology are provided here courtesy of American Society of Clinical Oncology

RESOURCES