Gene expression profiles of tumor regression grade in locally advanced rectal cancer after neoadjuvant chemoradiotherapy

  • Authors:
    • Susumu Saigusa
    • Koji Tanaka
    • Yuji Toiyama
    • Kohei Matsushita
    • Mikio Kawamura
    • Yoshinaga Okugawa
    • Junichiro Hiro
    • Yasuhiro Inoue
    • Keiichi Uchida
    • Yasuhiko Mohri
    • Masato Kusunoki
  • View Affiliations

  • Published online on: June 14, 2012     https://doi.org/10.3892/or.2012.1863
  • Pages: 855-861
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tumor regression grading (TRG) reportedly has prognostic value in rectal cancer patients after pre-operative chemoradiotherapy (CRT). The aim of this retrospective study was to differentiate gene expression profiles based on TRG in residual cancer cells after CRT. We evaluated pathological response using the criteria of four TRG systems: the Japanese Society for the Cancer of Colon and Rectum (JSCCR), Mandard, Dworak and Rödel. Total RNA was obtained using microdissection from 52 locally advanced rectal cancer specimens from patients who underwent pre-operative CRT to examine the expression levels of 20 genes [PCNA, MKI67, CDKN1A (p21Cip1), CDK2, CHEK1, PDRG1, LGR5, PROM1 (CD133), CD44, SOX2, POU5F1 (OCT4), LKB1, VEGF, EGFR, HGF, MET, HIF1, GLUT1, BAX and BCL2] using real-time quantitative RT-PCR. Gene expression was compared across the four TRG systems. LGR5 gene expression levels in CRT non-responders were significantly higher than in responders in all four grading systems. Patients with elevated PDRG1 and GLUT1 gene expression had poor pathological response in three TRG systems (JSCCR, Dworak and Rödel). MKI67 gene expression in non-responders was significantly higher than in responders in two grading systems (JSCCR and Rödel). While, BAX gene expression in responders was significantly higher than in non-responders in the Mandard TRG system. The results of this study suggest that TRG may reflect characteristics, such as proliferative activity, stemness potency and resistance to hypoxia, of residual cancer cells following pre-operative CRT.

Introduction

Pre-operative CRT followed by surgery is the standard treatment for patients with locally advanced rectal cancer. However, disease recurrence remains the major cause of mortality in these patients (1,2). Identifying predictors for disease recurrence or poor prognosis would aid in the successful treatment of these patients. Tumor regression grade (TRG) following pre-operative CRT is determined by quantifying the proportion of residual cancer cells to the stroma of the entire tumor bed. In rectal cancer, several studies have found that TRG or pathologic response is a predictor of clinical outcome, including disease recurrence and survival (38). Hence, it is possible that gene expression correlated with TRG might reflect the characteristics, including resistance to CRT, of residual cancer cells and might be associated with prognosis in patients with rectal cancer after pre-operative CRT. DNA repair pathways (9), cell cycle pathways (10), hypoxia (11,12), anti-apoptosis (13) and cancer stem cells (14,15) have been implicated in the mechanisms of CRT resistance. Twenty genes were selected for a comparison in expression levels between CRT responders and non-responders: PCNA and MKI67 (Ki67) as proliferative markers; CDKN1A (p21Cip1) and CDK2 as cell cycle associated markers after irradiation; CHEK1 and PDRG1 (p53 and DNA downregulated gene) as DNA damage associated makers; LGR5, PROM1 known as CD133, CD44, SOX2, POU5F1 known as OCT4 and LKB1 known as SKT11, as stem cell associated markers; VEGFA, HGF and MET as growth factors; HIF1 and GLUT1 as hypoxia associated markers; BAX and BCL2 as apoptosis associated markers in this transcriptional analysis. The aim of this retrospective study was to examine the expression of certain genes in TRGs and determine if the two are associated with clinical outcome in patients with locally advanced rectal cancer after pre-operative CRT.

Materials and methods

Patients and specimens

From 2001 to 2008, 64 patients with rectal cancer received pre-operative CRT followed by surgery at Mie University Hospital. The following criteria were used for induction of pre-operative CRT. Patients must i) be no more than 80 years of age; ii) be in clinical stage II/III based on the International Union Against Cancer TNM classification, with no evidence of distant metastases; iii) exhibit no invasion of the external sphincter muscle or elevator muscle of the anus; and iv) show no evidence of deep venous thrombosis. A total of 52 cases that received a curative operation and excluded pathological complete response were available for this study. The study design was approved by the hospital ethics review board. All patients signed informed consent forms allowing use of their tissues in this study.

5-Fluorouracil-based chemoradiotherapy regimen

Patients with rectal cancer were treated with short-course (a dose of 20 Gy in four fractions) or long-course (a dose of 45 Gy in 25 fractions) radiotherapy using a four-field box technique with concurrent chemotherapy to take advantage of 5-fluorouracil (5-FU) radio-sensitization. Patients underwent concurrent pharmacokinetic modulation chemotherapy (5-FU given by intravenous infusion, 600 mg/m2 for 24 h and tegafur-uracil (UFT) given orally, 400 mg/m2) orally for 5 days (16). Forty-one patients received short-course radiotherapy with chemotherapy over 1 week. The remaining eleven patients received long-course radiotherapy with chemotherapy for 4 weeks. The time interval between pre-operative CRT and surgery was 2–3 weeks for short-course irradiation patients, and 4–6 weeks for long-course irradiation patients. All patients underwent standard surgery, including total mesorectal excision, and received 5-FU-based adjuvant chemotherapy after surgery for 6 months to 1 year.

Clinical and pathological response to CRT

The clinical response after pre-operative CRT was evaluated by barium enema, endoscopy, computed tomography and magnetic resonance imaging, and was then graded as a complete response, a partial response, no change or progressive disease. Histological sections were sliced at a thickness of 5 μm and stained with hematoxylin and eosin. The TRG method for sampling and examining the tumor site from colorectal excision specimens removed following neoadjuvant therapy was found in Bateman et al(17). The median number of sections per case examined was 4.5 (range, 2–7). TRG was evaluated using criteria from four sources: Japanese Society for Cancer of the Colon and Rectum (JSCCR) (18), Mandard et al(19), Dworak et al(20) and Rödel et al(3). Each TRG was evaluated by two investigators (K. Tanaka and Y. Okugawa) in a blinded fashion without knowledge of the clinical and pathological information.

Microdissection and RNA extraction from formalin-fixed paraffin-embedded specimens

Microdissection of formalin-fixed paraffin-embedded (FFPE) was performed as previously described (21). Microdissected specimens were digested with proteinase K in lysis buffer containing Tris-HCl, ethylenediaminetetraacetic acid and sodium dodecyl sulfate, as previously published with minor modifications (22). RNA was purified by phenol/chloroform extraction and precipitated using ethanol. The concentration and quality of RNA was measured with UV absorbance at 260 and 280 nm (A260/280 ratio).

cDNA synthesis

The fragmented mRNA from FFPE tissue materials was reverse transcribed using random hexamer priming instead of oligo(dt)-based priming. cDNA was synthesized with random hexamers and Superscript III Reverse Transcriptase (Invitrogen, Carlsbad, CA) according to the manufacturer’s instructions.

Quantitative real-time polymerase chain reaction

Real-time quantitative RT-PCR analysis was performed using a fluorescence-based real-time detection method (TaqMan) and an ABI PRISM 7700 Sequence Detection System (Applied Biosystems, Inc., Foster City, CA). Although SYBR-Green-based detection is less specific than TaqMan-based detection, we used SYBR-Green due to time and cost constraints. Primers were strictly selected or designed to span introns to avoid amplification from contaminating genomic DNA. Target sequences were kept as small as possible (~100 bp) to ensure the detection of fragmented and partially degraded RNA. To confirm primer specificity, a single band of expected amplicon size for each target gene was verified using gel electrophoresis on a 2% agarose gel and visualized with ethidium bromide. Primers for PCNA, MKI67, CDKN1A (p21Cip1), CDK2, CHEK1, PDRG1, LGR5, PROM1 (CD133), CD44, SOX2, POU5F1 (OCT4), LKB1, VEGF, EGFR, HGF, MET, HIF1, GLUT1, BAX, BCL2, GAPDH and ACTB (β-actin) were designed with Primer3 software (Biology Workbench Version 3.2, San Diego Supercomputer Center, University of California, San Diego). Primer sequences are shown in Table I. PCR was performed in a final volume of 25 μl with a SYBR-Green PCR Master Mix using 1 μl cDNA and 400 nM of each primer for the respective genes. Cycling conditions were 50°C for 2 min and 95°C for 10 min followed by 40 cycles at 95°C for 15 sec and 60°C for 1 min.

Table I

Primer sequences of target genes.

Table I

Primer sequences of target genes.

Gene symbolForwardReverse
PCNA GAAGCACCAAACCAGGAGAA TATCGGCATATACGTGCAAA
MKI67 TGAGCCTGTACGGCTAAAACA TTGACTTCCTTCCATTCTGAAG
CDKN1A GACTCTCAGGGTCGAAAACG GGATTAGGGCTTCCTCTTGG
CDK2 CATTCCTCTTCCCCTCATCA TTTAAGGTCTCGGTGGAGGA
CHEK1 GACTGGGACTTGGTGCAAAC CACTGCGACTGATTCAG
PDRG1 CCTCACCCTGAGACAAAGGA GGCGGTTGACCTTCACTTTA
LGR5 GATGTTGCTCAGGGTGGACT GGGAGCAGCTGACTGATGTT
PROM1 GCTTTGCAATCTCCCTGTTG TTGATCCGGGTTCTTACCTG
CD44 CGGACACCATGGACAAGTTT CACGTGGAATACACCTGCAA
SOX2 CAAGATGCACAACTCGGAGA GCTTAGCCTCGTCGATGAAC
POU5F1 CTGGAGAAGGAGAAGCTGGA CAAATTGCTCGAGTTCTTTCTG
LKB1 CTCTTACGGCAAGGTGAAGG TTGTGCCGTAACCTCCTCAG
VEGFA CAGAAGGAGGAGGGCAGAA CTCGATTGGATGGCAGTAGC
EGFR CCTATGTGCAGAGGAATTATGATCTTT CCACTGTGTTGAGGGCAATG
HGF ATTTGGCCATGTTTTGACC AGCTGCGTCCTTTACCAATG
MET AGGTGTGGGAAAAACCTGA ATTCAGCTGTTGCAGGGAAG
HIF1 CCGCTGGAGACACAATCATA CTTCCTCAAGTTGCTGGTCA
GLUT1 CCTGCAGTTTGGCTACAACA GTGGACCCATGTCTGGTTG
BAX CTTTGCCAGCAAACTGGTG CAGCCCATGATGGTTCTGA
BCL2 TCGCCCTGTGGATGACTGA CAGAGACAGCCAGGAGAAATCAA
GAPDH GGAAGGTGAAGGTCGGAGTC AATGAAGGGGTCATTCATGG
ACTB ACAGAGCCTCGCCTTTGC GCGGCGATATCATCATCC

[i] PCNA, proliferation cell nuclear antigen; MKI67, Ki67; CDKN1A, p21Cip1; CDK2, cyclin-dependent kinase 2; PDRG1, p53 and DNA-damage regulated 1; LGR5, leucine-rich repeat-containing G protein-coupled receptor 5 known as GPR49; CHEK1, checkpoint kinase 1; PROM1, CD133; POU5F1, OCT4; LKB1, serine/threonine kinase 11 known as STK11; VEGFA, vascular endothelial growth factor; EGFR, epidermal growth factor receptor; HGF, hepatocyte growth factor; MET, HGF receptor; HIF1, hypoxia inducible factor 1α; GLUT1, glucose transporter 1; ACTB, β-actine.

Relative mRNA levels of target genes

The parameter Ct (threshold cycle) is defined as the fractional cycle number at which the fluorescence generated by cleavage of the probe passes a fixed threshold above baseline. The Ct is inversely proportional to the amount of cDNA, i.e., a higher Ct value means that more PCR cycles are required to reach a certain level of detection. Relative mRNA levels were determined using a standard curve. Standard curves and line equations were generated using 5-fold serially diluted solutions of cDNA from the LoVo colon cancer cell line or from qPCR Human Reference Total RNA (Clontech, Mountain View, CA). All standard curves were linear in the analyzed range with an acceptable correlation coefficient (R2). Target gene expression was calculated using the standard curve. Quantitative normalization of cDNA in each sample was performed using expression of the ACTB and GAPDH gene as internal controls. Finally, mRNA levels of the target gene were presented as ratios between the genes of interest and the internal reference gene. Real-time PCR assays were performed twice for each sample and mean values were used for calculations of mRNA levels.

Statistical analyses

All statistical analyses were performed using Stat View 5.0 for Windows (SAS Institute Inc., Cary, NC). Values of each target gene are expressed as median values (inter-quartile range) in tables. Associations between continuous variables and categorical variables were evaluated using the Mann-Whitney U test for two groups. Recurrence-free survival (RFS) and overall survival (OS) time were calculated from the date of surgery to the date of disease recurrence and death, respectively. RFS and OS probability were calculated using the Kaplan-Meier product limit method; intergroup differences were determined using a log-rank test. Two-sided P-values of <0.05 were considered statistically significant.

Results

Patient and tumor characteristics

The median age of the patients was 64.5 years (range, 38–77 years) and the median follow-up period was 52 months (range, 3–129 months). The male to female ratio was 2.9:1. The median age was 64.5 years (range, 37–77 years) and the male to female ratio was 3.7:1. The post-CRT pathological T stages were pT1 (n=5), pT2 (n=12), pT3 (n=33) and pT4 (n=2). Seventeen patients (33%) had lymph node metastases. Forty-four tumors (84.6%) showed well or moderately differentiated adenocarcinoma histology. Three patients (5.7%) had local recurrence alone. A total of 12 patients (23.1%) had distant recurrence. Patterns of distant recurrence were seen as liver and lung metastases in 2 patients, lung metastasis alone in 6 patients and peritoneal metastasis in 2 patients (Table II).

Table II

Patient and tumor characteristics.

Table II

Patient and tumor characteristics.

Variablesn=52 (%)
Age, median 64.5 years (range, 38–77)
Gender
 Male41 (79.0)
 Female11 (21.0)
Pre-T classification
 1/210 (19.2)
 3/442 (80.8)
Pre-N classification
 Negative17 (32.7)
 Positive35 (67.3)
Pre-stage
 I/II17 (32.7)
 III35 (67.3)
Down staging
 No29 (55.8)
 Yes23 (44.2)
Post-T classification
 1/217 (32.7)
 3/435 (67.3)
Post-N classification
 Negative35 (67.3)
 Positive17 (32.7)
Lymphatic invasion
 Negative13 (25.0)
 Positive39 (75.0)
Vascular invasion
 Negative21 (40.4)
 Positive31 (59.6)
Histology
 Well/moderately44 (84.6)
 Poorly/signet/mucinous8 (15.4)
Post-operative stage
 I/II33 (63.5)
 III19 (36.5)
Radiation
 Short, 20 Gy/4 fractions41 (78.8)
 Long, 45 Gy/25 fractions11 (21.2)
Recurrence
 None37 (71.2)
 Local alone3 (5.7)
 Distant with/without local failure12 (23.1)
Evaluation of pathological response by TRG

Table III shows the pathological response after CRT using each TRG. Responders were categorized as patients with JSCCR TRG 2, Mandard TRG 2, Dworak and Rödel TRG 3. The others were non-responders. We excluded the cases with pathological complete response and no regression in this study (JSCCR TRG 0 and 3, Mandard TRG 1 and 5, Dworak TRG 0 and 4, Rödel TRG 0 and 4).

Table III

Evaluation of pathological response by TRGs.

Table III

Evaluation of pathological response by TRGs.

Criterian=52 (%)
JSCCR
 TRG 0-
 TRG 1a13 (25.0)
 TRG 1b30 (57.6)
 TRG 29 (17.4)
 TRG 3-
Mandard
 TRG 1-
 TRG 215 (28.8)
 TRG 323 (44.2)
 TRG 414 (27.0)
 TRG 5-
Dworak
 TRG 0-
 TRG 114 (27.0)
 TRG 232 (61.5)
 TRG 36 (11.5)
 TRG 4-
Rödel
 TRG 0-
 TRG 110 (19.2)
 TRG 222 (42.3)
 TRG 320 (38.5)
 TRG 4-

[i] TRG, tumor regression grade. Cases with pathological complete response and no regression (JSCCR TRG 0 and 3, Mandard TRG 1 and 5, Dworak TRG 0 and 4, Rödel TRG 0 and 4) were excluded in this study.

Gene expression profiles of residual cancer according to TRG

Total RNA was isolated from 52 specimens and transcriptional analysis was performed for 20 genes. Table IV shows the gene expression levels of residual cancer according to TRG. LGR5 gene expression in non-responders was significantly higher than in responders among all TRG systems. Patients with elevated PDRG1 and GLUT1 gene expression had poor pathological response using the JSCCR, Dworak and Rödel TRG criteria. MKI67 gene expression in non-responders was significantly higher than in responders in the JSCCR and Rödel TRG systems. While, BAX gene expression in responders was significantly higher than in non-responders in Mandard TRG system. Patients with poor pathological response based on the Rödel criteria had significantly higher gene expression of MKI67, PDRG1, LGR5, LKB1, EGFR, MET and GLUT1.

Table IV

Gene expression profiles of residual cancer according to TRG systems.

Table IV

Gene expression profiles of residual cancer according to TRG systems.

JSCCRMandardDworakRödel




Gene symbol (range)Non-responder (n=43)Responder (n=9)P-valueNon-responder (n=37)Responder (n=15)P-valueNon-responder (n=46)Responder (n=6)P-valueNon-responder (n=32)Responder (n=20)P-value
PCNA (0–0.433)0.02480.00570.21220.02480.01040.29750.02210.00630.13960.04140.00910.0585
MKI67 (0–46.647)2.16350.00020.0282a2.43040.20720.05542.02240.00030.25212.71350.30990.0207a
CDKN1A (0–11.562)1.42911.99660.33931.46041.40380.75421.44481.54620.99991.65391.24980.3766
CDK2 (0–32.813)0.46280.28930.73330.51660.28930.46440.44050.07330.41150.52790.24840.2332
CHEK (0–0.318)0.02050.00370.66460.02380.00220.14750.01920.00550.79740.02580.00440.5879
PDRG1 (0–0.308)0.04850.00910.0077a0.04380.00960.08410.04090.00480.0296a0.10680.01180.0135a
LGR5 (0–54.643)7.74400.18100.0018a10.66602.01200.0037a7.23450.09050.0039a9.70807.49850.0105a
PROM1 (0–0.523)0.10700.02800.45330.10700.06600.69370.11100.01950.25190.10400.08200.5662
CD44 (0–1.274)0.15000.10800.91300.15000.10800.93540.15100.05250.45490.17400.10500.1025
SOX2 (0–71.914)2.25903.21500.32732.25900.25410.14042.40001.69050.22902.83651.75950.0923
POU5F1 (0–600.343)6.72205.75400.25066.72205.75400.26239.63853.21300.07586.006010.99500.5473
LKB1 (0–0.442)0.02330.00270.07120.02330.01630.27930.02260.00950.26230.03190.00570.0110a
VEGFA (0–0.092)0.02890.03310.75550.02780.03160.93780.03090.03160.67330.02780.03160.7618
EGFR (0–0.256)0.01280.01830.48350.01420.01040.28860.01290.01890.75180.01480.00930.0377a
HGF (0–1.2323)0.13340.03160.72260.17920.00780.27410.144050.01580.40050.20080.030550.0621
MET (0–3.477)0.260.0660.12390.260.0660.18510.2570.0590.08010.5260.0630.0310a
HIF1 (0–0.390)0.07750.15530.58360.07750.13880.35810.07760.11300.90880.07760.07220.8361
GLUT1 (0–8.189)0.70590.30170.0151a0.65980.51400.22950.64080.19040.0274a0.72590.39810.0352a
BAX (0–15.062)0.71281.07880.06960.68551.05480.0339a0.71711.06680.11180.77510.70630.8655
BCL2 (0–0.617)0.03360.11390.68010.03660.07450.92040.03160.16960.29860.03930.06070.9835

{ label (or @symbol) needed for fn[@id='tfn3-or-28-03-0855'] } Values of each target gene are expressed as the median value. JSCCR, Japanese Society for Cancer of the Colon and Rectum. Mann-Whitney U test.

a P<0.05.

Recurrence-free and overall survival based on expression levels of PDRG1, LGR5 and GLUT1

We investigated whether the gene expression levels correlated with TRG were also associated with patient prognosis after pre-operative CRT followed by curative surgery because it has been reported as a predicter of clinical outcome. Four genes were selected (MKI67, PDRG1, LGR5 and GLUT1) that were significantly correlated with more than two TRG systems. The median value of MKI67, PDRG1, LGR5 and GLUT1 gene expression was 0.00000895, 0.0308, 6.295 and 0.566, respectively. We categorized the case with more than the median value as the high gene expression group, and the remaining as the low group. Fig. 1 shows the survival curve for RFS according to each gene expression level using Kaplan-Meier analysis. Patients with LGR expression levels above the cut-off values showed a significantly poorer RFS than did patients with expression levels below the cut-off values (P=0.0262). Patients in high MKI67 expression group had poorer RFS than that in low group without significant difference (P=0.0549). Patients with high GLUT1 gene expression had significantly poorer OS than those with low one (GLUT1 P=0.0093). MKI67, PDRG1 and LGR5 gene expression were not associated with OS (MKI67, P=0.6018; PDRG1, P=0.5493; LGR5, P=0.6487) (data not shown).

Discussion

Rectal cancer is one of the most common cancers in Japan and the Western world. The introduction of pre-operative CRT and total mesorectal excision for the management of locally advanced rectal cancer significantly decreased local recurrence rates and improved sphincter preservation and patient survival (2326). However, tumor recurrence remains the major cause of mortality in these patients, and the mechanism of tumor recurrence after pre-operative CRT remains unclear. In this study, the expression levels of 20 genes were correlated with TRG to determine if gene expression levels can be associated with tumor recurrence in locally advanced rectal cancer after pre-operative CRT.

Tumor down-staging, post-operative stage, N, T classification and TRG have been identified as important prognostic factors in rectal cancer patients following pre-operative CRT (38,2729). We observed that advanced post-operative stage, the presence of lymph node metastasis and poor pathological response based on JSCCR and Rödel TRGs were significantly associated with recurrence-free survival in this study of 52 patients (log-rank test; post-operative stage, P=0.0137; presence of lymph node metastasis, P=0.0244; JSCCR, P=0.0464; Rödel, P=0.0338) (data not shown). Our data were similar to previous studies although down-staging and T classification were not associated with clinical outcome. Transcriptional analyses of gene expression according to TRG were then performed. To the best of our knowledge, there are few reports on the correlation between TRG and gene expression of residual cancer cells after pre-operative CRT. The expression levels of eight genes were correlated with TRGs. LGR5 gene expression levels in non-responders was significantly higher than those in responders in all four TRG systems. LGR5 is a potential marker for stem cells in the small intestine and colon (30). Overexpression of LGR5 has also been implicated in colorectal carcinogenesis (31). Elevated PDRG1 and GLUT1 gene expression level was significantly correlated with poor pathological response in three of the four TRG systems (JSCCR, Dworak and Rödel). PDRG1 is regulated by DNA damage due to ultraviolet radiation and is implicated in tumor cell growth (32,33). GLUT1 serves as a hypoxic indicator and is a hypoxic marker in colorectal cancer (34). Immunohistochemistry of GLUT1 expression in pre-treatment rectal cancer biopsy samples suggests that GLUT1 might be a useful predictive marker of response to CRT in rectal cancer (35). MKI67, LKB1, EGFR, MET and BAX expression levels were significantly correlated with one to two TRG systems. MKI67 is a proliferative marker (36) and LKB1 has been implicated in the maintenance of hematopoietic stem cells and tumorigenesis through the suppression of apoptosis (3739). BCL2/BAX act as anti- or pro-apoptotic regulator. We observed that high BAX, but not BCL2, expression was correlated with better pathological response based on Mandard TRG system. Reduction of BAX function in human colorectal cancer cells has been associated with resistance to chemotherapy (40) and BAX expression was correlated with outcome to neoadjuvant CRT (41) using imuunohistochemistry in pre-CRT samples. Our result suggested that post-CRT BAX expression also might reveal the correlation of resistance to CRT. We examined whether gene expression of TRGs were associated with prognosis in rectal cancer after pre-operative CRT followed by surgery. We observed that patients with high LGR5 expression had significant correlation to RFS. Elevated GLUT1 expression was significantly associated with poor OS.

Biomarkers for tumor recurrence and prognosis after pre-operative CRT followed by curative surgery remain to be established. Our results may help identify prognostic predictors and clarify the strategy of adjuvant therapy for tumor recurrence after pre-operative CRT. However, it is not clear if the gene expressions correlated with TRGs are inherent or acquired after CRT due to the inability to compare expression levels between pre- and post-CRT samples. Therefore, these results did not directly show the predictive value of response to pre-operative CRT. We plan to examine the gene expression levels between pre-CRT biopsy and post-CRT samples.

In conclusion, TRG may reflect certain characteristics, such as proliferative activity, stemness potency and resistance to hypoxia, of residual cancer cells after pre-operative CRT. Moreover, the gene expression levels correlated with TRG were associated with poor recurrence-free survival in patients with locally advanced rectal cancer after neoadjuvant CRT. However, data in this study should be interpreted with some caution. The major limitation was the small number of patients, especially for patients with recurrence, and the retrospective nature of the study. A larger study population will allow us to validate these conclusions.

Acknowledgements

The authors would like to thank Yuka Kato and Motoko Ueda for providing excellent technical assistance.

References

1 

Van den Brink M, Stiggelbout AM, van den Hout WB, et al: Clinical nature and prognosis of locally recurrent rectal cancer after total mesorectal excision with or without preoperative radiotherapy. J Clin Oncol. 22:3958–3964. 2004.PubMed/NCBI

2 

Collette L, Bosset JF, den Dulk M, et al: Patients with curative resection of cT3-4 rectal cancer after preoperative radiotherapy or radiochemotherapy: does anybody benefit from adjuvant fluorouracil-based chemotherapy? A trial of the European Organisation for Research and Treatment of Cancer Radiation Oncology Group. J Clin Oncol. 25:4379–4386. 2007.

3 

Rodel C, Martus P, Papadoupolos T, et al: Prognostic significance of tumor regression after preoperative chemoradiotherapy for rectal cancer. J Clin Oncol. 23:8688–8696. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Vironen J, Juhola M, Kairaluoma M, et al: Tumour regression grading in the evaluation of tumour response after different preoperative radiotherapy treatments for rectal carcinoma. Int J Colorectal Dis. 20:440–445. 2005. View Article : Google Scholar

5 

Vecchio FM, Valentini V, Minsky BD, et al: The relationship of pathologic tumor regression grade (TRG) and outcomes after preoperative therapy in rectal cancer. Int J Radiat Oncol Biol Phys. 62:752–760. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Bujko K, Michalski W, Kepka L, et al: Association between pathologic response in metastatic lymph nodes after preoperative chemoradiotherapy and risk of distant metastases in rectal cancer: an analysis of outcomes in a randomized trial. Int J Radiat Oncol Biol Phys. 67:369–377. 2007. View Article : Google Scholar

7 

Dhadda AS, Dickinson P, Zaitoun AM, et al: Prognostic importance of Mandard tumour regression grade following pre-operative chemo/radiotherapy for locally advanced rectal cancer. Eur J Cancer. 47:1138–1145. 2011. View Article : Google Scholar

8 

Topova L, Hellmich G, Puffer E, et al: Prognostic value of tumor response to neoadjuvant therapy in rectal carcinoma. Dis Colon Rectum. 54:401–411. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Jorgensen TJ: Enhancing radiosensitivity: targeting the DNA repair pathways. Cancer Biol Ther. 8:665–670. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Maity A, McKenna WG and Muschel RJ: The molecular basis for cell cycle delays following ionizing radiation: a review. Radiother Oncol. 31:1–13. 1994. View Article : Google Scholar : PubMed/NCBI

11 

Coleman CN: Hypoxia in tumors: a paradigm for the approach to biochemical and physiologic heterogeneity. J Natl Cancer Inst. 80:310–317. 1988. View Article : Google Scholar : PubMed/NCBI

12 

Overgaard J: Hypoxic radiosensitization: adored and ignored. J Clin Oncol. 25:4066–4074. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Deorukhkar A and Krishnan S: Targeting inflammatory pathways for tumor radiosensitization. Biochem Pharmacol. 80:1904–1914. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Baumann M, Krause M and Hill R: Exploring the role of cancer stem cells in radioresistance. Nat Rev Cancer. 8:545–554. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Zeki SS, Graham TA and Wright NA: Stem cells and their implications for colorectal cancer. Nat Rev Gastroenterol Hepatol. 8:90–100. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Yoshikawa R, Kusunoki M, Yanagi H, et al: Dual antitumor effects of 5-fluorouracil on the cell cycle in colorectal carcinoma cells: a novel target mechanism concept for pharmacokinetic modulating chemotherapy. Cancer Res. 61:1029–1037. 2001.

17 

Bateman AC, Jaynes E and Bateman AR: Rectal cancer staging post neoadjuvant therapy - how should the changes be assessed? Histopathology. 54:713–721. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Japanese Society for Cancer of the Colon and Rectum. General rules for clinical and pathological studies on cancer of the colon, rectum and anus. Kanehara & Co; Tokyo: 2006

19 

Mandard AM, Dalibard F, Mandard JC, et al: Pathologic assessment of tumor regression after preoperative chemoradiotherapy of esophageal carcinoma. Clinicopathologic correlations. Cancer. 73:2680–2686. 1994. View Article : Google Scholar

20 

Dworak O, Keilholz L and Hoffmann A: Pathological features of rectal cancer after preoperative radiochemotherapy. Int J Colorectal Dis. 12:19–23. 1997. View Article : Google Scholar : PubMed/NCBI

21 

Saigusa S, Tanaka K, Toiyama Y, et al: Correlation of CD133, OCT4, and SOX2 in rectal cancer and their association with distant recurrence after chemoradiotherapy. Ann Surg Oncol. 16:3488–3498. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Bijwaard KE, Aguilera NS, Monczak Y, et al: Quantitative real-time reverse transcription-PCR assay for cyclin D1 expression: utility in the diagnosis of mantle cell lymphoma. Clin Chem. 47:195–201. 2001.PubMed/NCBI

23 

Sauer R, Becker H, Hohenberger W, et al: Preoperative versus postoperative chemoradiotherapy for rectal cancer. N Engl J Med. 351:1731–1740. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Gerard JP, Chapet O, Nemoz C, et al: Improved sphincter preservation in low rectal cancer with high-dose preoperative radiotherapy: the lyon R96–02 randomized trial. J Clin Oncol. 22:2404–2409. 2004.PubMed/NCBI

25 

Guillem JG, Chessin DB, Cohen AM, et al: Long-term oncologic outcome following preoperative combined modality therapy and total mesorectal excision of locally advanced rectal cancer. Ann Surg. 241:829–838. 2005. View Article : Google Scholar

26 

Bosset JF, Collette L, Calais G, et al: Chemotherapy with preoperative radiotherapy in rectal cancer. N Engl J Med. 355:1114–1123. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Kaminsky-Forrett MC, Conroy T, Luporsi E, et al: Prognostic implications of downstaging following preoperative radiation therapy for operable T3-T4 rectal cancer. Int J Radiat Oncol Biol Phys. 42:935–941. 1998. View Article : Google Scholar : PubMed/NCBI

28 

Chapet O, Romestaing P, Mornex F, et al: Preoperative radiotherapy for rectal adenocarcinoma: which are strong prognostic factors? Int J Radiat Oncol Biol Phys. 61:1371–1377. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Kim TH, Chang HJ, Kim DY, et al: Pathologic nodal classification is the most discriminating prognostic factor for disease-free survival in rectal cancer patients treated with preoperative chemoradiotherapy and curative resection. Int J Radiat Oncol Biol Phys. 77:1158–1165. 2010. View Article : Google Scholar

30 

Barker N, van Es JH, Kuipers J, et al: Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 449:1003–1007. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Uchida H, Yamazaki K, Fukuma M, et al: Overexpression of leucine-rich repeat-containing G protein-coupled receptor 5 in colorectal cancer. Cancer Sci. 101:1731–1737. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Luo X, Huang Y and Sheikh MS: Cloning and characterization of a novel gene PDRG that is differentially regulated by p53 and ultraviolet radiation. Oncogene. 22:7247–7257. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Jiang L, Luo X, Shi J, et al: PDRG1, a novel tumor marker for multiple malignancies that is selectively regulated by genotoxic stress. Cancer Biol Ther. 11:567–573. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Chung FY, Huang MY, Yeh CS, et al: GLUT1 gene is a potential hypoxic marker in colorectal cancer patients. BMC Cancer. 9:2412009. View Article : Google Scholar : PubMed/NCBI

35 

Brophy S, Sheehan KM, McNamara DA, et al: GLUT-1 expression and response to chemoradiotherapy in rectal cancer. Int J Cancer. 125:2778–2782. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Hofstadter F, Knuchel R and Ruschoff J: Cell proliferation assessment in oncology. Virchows Arch. 427:323–341. 1995. View Article : Google Scholar

37 

Herrmann JL, Byekova Y, Elmets CA, et al: Liver kinase B1 (LKB1) in the pathogenesis of epithelial cancers. Cancer Lett. 306:1–9. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Gurumurthy S, Xie SZ, Alagesan B, et al: The Lkb1 metabolic sensor maintains haematopoietic stem cell survival. Nature. 468:659–663. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Gan B, Hu J, Jiang S, et al: Lkb1 regulates quiescence and metabolic homeostasis of haematopoietic stem cells. Nature. 468:701–704. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Yamaguchi H, Bhalla K and Wang HG: Bax plays a pivotal role in thapsigargin-induced apoptosis of human colon cancer HCT116 cells by controlling Smac/Diablo and Omi/HtrA2 release from mitochondria. Cancer Res. 63:1483–1489. 2003.PubMed/NCBI

41 

Chang HJ, Jung KH, Kim DY, et al: Bax, a predictive marker for therapeutic response to preoperative chemoradiotherapy in patients with rectal carcinoma. Hum Pathol. 36:364–371. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September 2012
Volume 28 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Saigusa S, Tanaka K, Toiyama Y, Matsushita K, Kawamura M, Okugawa Y, Hiro J, Inoue Y, Uchida K, Mohri Y, Mohri Y, et al: Gene expression profiles of tumor regression grade in locally advanced rectal cancer after neoadjuvant chemoradiotherapy. Oncol Rep 28: 855-861, 2012
APA
Saigusa, S., Tanaka, K., Toiyama, Y., Matsushita, K., Kawamura, M., Okugawa, Y. ... Kusunoki, M. (2012). Gene expression profiles of tumor regression grade in locally advanced rectal cancer after neoadjuvant chemoradiotherapy. Oncology Reports, 28, 855-861. https://doi.org/10.3892/or.2012.1863
MLA
Saigusa, S., Tanaka, K., Toiyama, Y., Matsushita, K., Kawamura, M., Okugawa, Y., Hiro, J., Inoue, Y., Uchida, K., Mohri, Y., Kusunoki, M."Gene expression profiles of tumor regression grade in locally advanced rectal cancer after neoadjuvant chemoradiotherapy". Oncology Reports 28.3 (2012): 855-861.
Chicago
Saigusa, S., Tanaka, K., Toiyama, Y., Matsushita, K., Kawamura, M., Okugawa, Y., Hiro, J., Inoue, Y., Uchida, K., Mohri, Y., Kusunoki, M."Gene expression profiles of tumor regression grade in locally advanced rectal cancer after neoadjuvant chemoradiotherapy". Oncology Reports 28, no. 3 (2012): 855-861. https://doi.org/10.3892/or.2012.1863