Upregulation of stromal cell-derived factor 1α expression is associated with the resistance to neoadjuvant chemoradiotherapy of locally advanced rectal cancer: Angiogenic markers of neoadjuvant chemoradiation

  • Authors:
    • Han Jo Kim
    • Sang Byung Bae
    • Dongjun Jeong
    • Eun Seog Kim
    • Chang-Nam Kim
    • Dong-Guk Park
    • Tae Sung Ahn
    • Sung Woo Cho
    • Eung Jin Shin
    • Moon Soo Lee
    • Moo Jun Baek
  • View Affiliations

  • Published online on: September 19, 2014     https://doi.org/10.3892/or.2014.3504
  • Pages: 2493-2500
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The ability to achieve pathologic downstaging after neoadjuvant chemoradiotherapy (NCRT) is correlated with improved survival in locally advanced rectal cancer (LARC). However, there is no effective predictive markers. In this study, the expression of angiogenic markers was evaluated in pre-treatment biopsies and corresponding post-treatment resection specimens, and were correlated to histopathological tumour characteristics and response. Fifty-five patients with stage II/III rectal cancer treated with 5-fluorouracil (5-FU)-based NCRT were studied. All patients were administered NCRT followed by surgical resection. Immunohistochemical staining for angiogenic markers [hypoxia-inducible factor 1α (HIF‑1α), vascular endothelial growth factor (VEGF), stromal cell‑derived factor 1α (SDF-1α) and placental growth factor (PlGF)] was performed on specimens obtained before NCRT and after surgery. Expression of VEGF, PlGF and HIF-1α protein was downregulated after NCRT in the rectal cancer tissues (P<0.001, P=0.001 and P=0.044, respectively). However, SDF-1α was upregulated after NCRT (P<0.001). Moreover, upregulated expression of SDF-1α (P=0.016) and positive PlGF staining (P=0.001) after NCRT were significantly associated with resistance to NCRT. On multivariate analysis, positive PlGF staining after NCRT was found to be independently associated with resistance to NCRT (P=0.013). Our data suggest that SDF-1α and PlGF should be evaluated as new targets for NCRT in LARC.

Introduction

Rectal cancer (RC) is a major health issue and is one of the leading causes of cancer-related death worldwide (1). Neoadjuvant chemoradiotherapy (NCRT) followed by surgical resection is the current standard treatment for locally advanced rectal cancer (LARC). It offers improved local control, reduced toxicity and higher rates of sphincter preservation without compromising oncological outcome compared with post-operative treatment (2,3). A pathologic complete response (pCR) is one of the best predictive markers of a favourable prognosis. However, approximately 15–30% of patients experience a pCR, whereas the majority of patients have some degree of residual disease after NCRT (4). Thus, if patients with tumours that are responsive to NCRT could be identified at the time of diagnosis, then NCRT could be administered in a more individualised manner.

Recent studies have attempted to identify predictive biomarkers such as Ki-67, p53, p21, p27, bax, BCL-2, vascular endothelial growth factor (VEGF), epidermal growth factor receptor (EGFR), survivin and thymidylate synthase (5,6). However, clinical use of these biomarkers requires further evaluation in prospective clinical trials (7).

Angiogenesis is necessary for tumour growth and malignant progression, with VEGF being a key angiogenic factor. High VEGF expression was found to be associated with poor survival in colorectal cancers (8). In particular, bevacizumab, a humanised monoclonal antibody inhibiting VEGF-A, in combination with standard chemotherapy regimens was beneficial both in terms of response rate and survival as first-and second-line treatment of patients affected by metastatic colorectal cancer. In patients affected by LARC who underwent radical surgery and adjuvant chemoradiation, tumour VEGF overexpression was found to be associated with a statistically higher risk of local recurrence and metastasis (9). Not only VEGF, but placental growth factor (PlGF) are potentially useful predictive factors in rectal cancer (10).

Hypoxia is one of the key stimuli for the release of angiogenic markers (AMs) necessary for angiogenesis and tumour growth. Hypoxic tumours not only have a more aggressive nature (11,12), but studies in cervical and head and neck cancer demonstrated that tumour hypoxia decreases the response to radiation treatment (13). The effect of tumour hypoxia on the response to radiation therapy is relevant to the management of rectal cancer.

Hypoxia-inducible factor 1α (HIF-1α) is a protein involved in the cellular response to hypoxia centrally. It activates a variety of downstream genes involved in anaerobic metabolism and angiogenesis (1416). These downstream gene protein products, which include VEGF, stromal cell-derived factor 1α (SDF-1α) and PlGF, promote cell survival under hypoxic conditions. Among patients with colorectal cancer, expression of these 4 AMs has been shown to correlate with rates of lymph node or liver metastasis, disease-free survival, and overall survival (1720). However, the effects of pre-operative treatment on expression of AMs in rectal cancer remain unclear. The 4 AMs chosen for this study include HIF-1α, VEGF, SDF-1α and PlGF.

The aims of this exploratory study were to a) characterise expression of AMs in LARC before NCRT, b) investigate the change in AM expression after NCRT and c) evaluate the relationship between AM expression and tumour response.

Patients and methods

Patients and neoadjuvant chemoradiotherapy regimen

Between March 2005 and August 2009 at Soonchunhyang University Hospital, a total of 55 patients with non-metastatic, locally advanced (radiological T3–T4 or N+ and/or clinically bulky) and biopsy-proven primary rectal cancer received NCRT. The whole pelvic field received 25 fractions of 180 cGy/day over 5 weeks, for a total of 4,500–5,040 cGy, using a four-field box technique. Chemotherapy was administered intravenously and consisted of 5-fluorouracil (5-FU; 425 mg/m2/day) and leucovorin (20 mg/m2/day) during the first and fifth weeks of radiotherapy. Surgical resection at 6–8 weeks was performed following the completion of NCRT. All data were collected and recorded prospectively, and the clinical and pathological features were reviewed retrospectively. The patients were classified according to the 6th edition of the American Joint Committee on Cancer staging system (21). Surgical specimens were evaluated for histopathologic staging as well as for pathologic response to NCRT. The detailed characteristics of the patients are listed in Table I. Our study was approved by the Clinical Ethics Review Committee of the Soonchunhyang University Hospital, Cheonan, Republic of Korea. Clinical consent was obtained from all patients.

Table I

Association between AMs and clinicopathological parameters.

Table I

Association between AMs and clinicopathological parameters.

Clinicopathological parametersHIF-1α expressionVEGF expressionPlGF expressionSDF-1α expression




NegativePositiveP-valueNegativePositiveP-valueNegativePositiveP-valueNegativePositiveP-value
Total patients, n (%)29 (52.7)26 (47.3)24 (43.6)31 (56.4)19 (34.5)36 (65.5)16 (29.1)39 (70.9)
Age (years)0.1530.8770.9570.466
 <65211415201223926
 ≥65812911713713
Gender0.3850.8760.7331.000
 Male2419192414291330
 Female57575739
Pre-treatment tumour stage0.4180.5051.0000.712
 32222182615291232
 474654747
Pre-treatment nodal stage0.2200.0780.5700.388
 083835656
 1913715616517
 21210913814616
Post-treatment tumour stage0.9380.9260.2620.648
 042333324
 110010101
 226355317
 32018172110281226
 420111111
Post-treatment nodal stage0.1380.2810.4890.277
 02315182015231424
 14857210012
 223142323

[i] AMs, angiogenic markers; HIF-1α, hypoxia-inducible factor 1α; VEGF, vascular endothelial growth factor; PlGF, placenta growth factor; SDF-1α, stromal cell-derived factor 1α.

Tissue microarray (TMA) construction

Areas representative of cancer were marked on haematoxylin and eosin-stained slides and TMAs were constructed. TMAs were created from formalin-fixed by 10% neutral buffered formalin, paraffin-embedded tissues using a 2-mm-diameter punch (Unitma; Unitech Science, Seoul, Korea). TMA blocks were assembled by obtaining duplicate cores from one patient block and re-embedding the two cores in an arrayed recipient block (Unitma; Unitech Science). A TMA block contains 60 cores from 30 samples.

Tumour response

Clinical stage was performed by an independent review conducted by a radiologist, and pathologic stage was reviewed by two independent pathologists. Downstaging was defined as staging reduction from pre-treatment stage (cStage) to pathologic stage (ypStage) (i.e. cIII to ypII, ypI or yp0; cII to ypI or yp0). Pathologic response (tumour regression) to NCRT was semiquantitatively determined by the amount of viable tumour versus the amount of fibrosis, ranging from no evidence of any NCRT effect to a complete response with no viable tumour identified, as described by Dworak et al (22). The following were characteristics of each grade: grade 0, no regression; grade 1, minor regression (dominant tumour mass with obvious fibrosis in 25% or less of the tumour mass); grade 2, moderate regression (dominant tumour mass with obvious fibrosis in 26–50% of the tumour mass); grade 3, good regression (dominant fibrosis outgrowing the tumour mass; i.e. >50% tumour regression); and grade 4, total regression (no viable tumour cells, only fibrotic mass). Patients with tumour regression grade (TRG) of 3 or 4 were considered as the responder group in our study.

Immunohistochemical (IHC) staining

The TMAs were sectioned at 4-μm intervals, deparaffinised three times in xylene for 30 min and rehydrated with graded alcohols (100% ethyl alcohol for 5 min, 95% ethyl alcohol for 3 min and 75% ethyl alcohol for 3 min) and then heated in antigen retrieval solution (sodium citrate, pH 6.0) in a microwave for 20 min. Sections were incubated in H2O2 for 10 min at room temperature. Furthermore, the sections were incubated with 150 ml of the primary antibodies, VEGF (1:200; Millipore, USA), PlGF (1:200; R&D system, USA), HIF-1α (1:50; Proteintech, USA) and SDF-1α (1:100; Novus Biologicals, USA) at 4°C overnight. Subsequently, the sections were washed in PBS buffer three times for 3 min, treated with 150 ml secondary antibody for 1 h at room temperature and stained with DAB solution (Dako, USA). The sections were then washed in PBS buffer for 10 min. Finally, the sections were counterstained with hematoxylin for 3 min at room temperature, washed in distilled water 3 times for 3 min and mounted with coverslips.

IHC analysis

The VEGF, PlGF, HIF-1α and SDF-1α stained tissue cores were examined by 2 independent pathologists and a consensus score was determined for each specimen. A positive reaction for both antibodies was scored into 4 grades, according to the intensity of the staining: 0, 1+, 2+ and 3+. The percentages of positive cells were also scored into 4 categories: 0 (0%), 1 (1–33%), 2 (34–66%), and 3 (67–100%). The final score, calculated as the product of the intensity score multiplied by the percentage score, was classified as follows: 0 for negative; 1–3 for weak; 4–6 for moderate; and 7–9 for strong.

Statistical analysis

The correlations between expression levels of hypoxia-related proteins and pathologic response to NCRT were evaluated by the χ2 or Fisher’s exact test. The univariate and multivariate analyses between response to NCRT and clinical or histopathologic parameters were performed by binary logistic regression model. All P-values quoted were two-sided, and P<0.05 was considered to indicate a statistically significant difference. All the analyses were performed using SPSS v. 17.0 (SPSS, Inc., Chicago, IL, USA).

Results

Association between AM expression and clinicopathological variables

The mean age of the 55 patients with LARC was 56 years (range, 18–82 years). In regards to gender, 43 (78.2%) were male, and 12 (21.8%) were female. Regarding the stage of disease, 11 (20.0%) were at stage II, and 44 (80.0%) were at stage III. Concerning the T stage, 44 (80.0%) were T3 and 11 (20.0%) were T4. The number of negative lymph node metastases was 11 (20.0%); N1 was 22 (40.0%), and N2 was 22 (40.0%). A pCR was obtained in 9.1% cases (5 patients). Patient characteristics are summarised in Table I. As shown in Table I, expression levels of AMs were not statistically correlated to the clincopathological variables.

Change in AM expression in LARC before NCRT and after surgery

The positive expression rate of HIF-1α, VEGF, PlGF and SDF-1α was 47.3% (26/55), 56.4% (31/55), 65.5% (36/55) and 70.9% (39/55) before NCRT, respectively. Weak, moderate and strong staining intensity of AMs is illustrated in Fig. 1. The expression rate of HIF-1α, VEGF, SDF-1α and PlGF was increased by 1.8% (1/55), 3.6% (2/55), 30.9% (17/55) and 7.3% (4/55) after NCRT, respectively. Expression of VEGF, PlGF and HIF-1α protein was downregulated after NCRT in the rectal cancer tissues (P<0.001, P=0.001 and P=0.044, respectively). However, SDF-1α was upregulated after NCRT (P<0.001; Table II, Fig. 2).

Table II

Results of AM immunoreactivity before NCRT and after surgery.

Table II

Results of AM immunoreactivity before NCRT and after surgery.

HIF-1αVEGFSDF-1αPlGF




Staining scoreBefore NCRTAfter surgeryBefore NCRTAfter surgeryBefore NCRTAfter surgeryBefore NCRTAfter surgery
Negative2948243916101930
Weak177161217121919
Moderate80112151984
Strong104271492

[i] NCRT, neoadjuvant chemoradiotherapy; AMs, angiogenic markers.

Relationship between tumour response to NCRT and clinicopathological variables

Upregulated expression of SDF-1α (P<0.016) and positive PlGF staining (P=0.001) after NCRT were significantly associated with resistance to NCRT. However, other clinicopathologic variables showed no correlation with tumour response (Table III). In multivariate analyses, positive PlGF staining after NCRT was found to be associated with resistance to NCRT [P=0.013; OR=0.197, 95% confidence interval (CI), 0.055–0.705]. Only low pre-treatment tumour lymph node staging was associated with pCR (P=0.002; Table IV).

Table III

Association between tumour response and clinicopathological parameters.

Table III

Association between tumour response and clinicopathological parameters.

Clinicopathological parametersTumour responseP-value

RNR
Age (years)0.877
 <652015
 ≥65119
Gender0.416
 Male2320
 Female84
Pre-treatment tumour stage0.180
 32717
 447
Pre-treatment nodal stage0.517
 047
 1157
 21210
Pre-treatment VEGF staining0.773
 Negative1311
 Positive1813
Pre-treatment PlGF staining0.190
 Negative136
 Positive1818
Pre-treatment SDF-1α staining0.235
 Negative115
 Positive2019
Pre-treatment HIF-1α staining0.368
 Negative1811
 Positive1313
Post-treatment VEGF staining0.074
 Negative1920
 Positive124
Post-treatment PlGF staining0.001
 Negative237
 Positive817
Post-treatment SDF-1α staining0.159
 Negative82
 Positive2322
Post-treatment HIF-1α staining0.686
 Negative2820
 Positive34
Change of staining status
 VEGF0.400
  Decreased1011
  Same2012
  Increased11
 PlGF0.568
  Decreased1211
  Same199
  Increased04
 SDF-1α0.016
  Decreased30
  Same2213
  Increased611
 HIF-1α0.343
  Decreased1111
  Same1913
  Increased10

[i] R, responder; NR, non-responder.

Table IV

Association between pCR and clinicopathological parameters.

Table IV

Association between pCR and clinicopathological parameters.

Clinicopathological parameterspCRP-value

(−)(+)
Age (years)1.000
 <65323
 ≥65182
Gender0.298
 Male403
 Female102
Pre-treatment tumour stage0.571
 3395
 4110
Pre-treatment nodal stage0.002
 074
 1211
 2220
Pre-treatment VEGF staining0.643
 Negative213
 Positive292
Pre-treatment PlGF staining0.327
 Negative163
 Positive342
Pre-treatment SDF-1α staining0.622
 Negative142
 Positive363
Pre-treatment HIF-1α staining0.355
 Negative254
 Positive251
Post-treatment VEGF staining1.000
 Negative354
 Positive151
Post-treatment PlGF staining0.056
 Negative255
 Positive250
Post-treatment SDF-1α staining0.220
 Negative82
 Positive423
Post-treatment HIF-1α staining1.000
 Negative435
 Positive70
Change of staining status
 VEGF0.817
  Decreased192
  Same293
  Increased20
 PlGF0.835
  Decreased212
  Same253
  Increased40
 SDF-1α0.053
  Decreased21
  Same314
  Increased170
 HIF-1α0.418
  Decreased211
  Same284
  Increased10

[i] pCR, pathologic complete response.

Relationship with AM expression

Before NCRT, an association was identified between HIF-1α expression and SDF-1α (P=0.034). HIF-1α was not correlated with VEGF and PlGF. However, SDF-1α had an association with PlGF (P=0.005). After surgery, HIF-1α expression was not correlated with SDF-1α (P=0.621), and SDF-1α tended to be associated with PlGF (P=0.052).

Discussion

Recently, studies have attempted to identify predictive biomarkers, yet various studies only compared pre-treatment and post-treatment changes in biomarker expression (5). In this study, we investigated the predictive relevance of AM expression both in pre-treatment biopsies and in corresponding surgical specimens of 55 patients with LARC treated with standadised 5-FU-based NCRT. Comparing pre-treatment biopsies and surgical specimens, we observed a downregulation of VEGF, PlGF and HIF-1α. However, SDF-1α was upregulated after NCRT. In addition, upregulated SDF-1α after NCRT was significantly associated with resistance to NCRT. Our findings suggest that SDF-1α is one of the important targets for resistance to NCRT and this finding is significant.

SDF-1α, also known as chemokine ligand 12 (CXCL12), and its receptor CXCR4, play important roles in the onset and progression of primary or metastatic cancer from various organs (2326). In colorectal cancer (CRC), elevated SDF-1α expression is associated with metastasis and poor prognosis (27,28). In our investigation, upregulation of SDF-1α in surgical specimens was related to resistance to NCRT. Thus, SDF-1α appears to be a predictive marker to chemoradiation treatment. In an in vitro study using a CRC cell line, the results indicate that CXCR4 antagonistic therapy might prevent tumour cell dissemination and metastasis in CRC patients, consequently improving survival (29). Therefore, the targeting of SDF-1α represents an attractive adjuvant treatment to eradicate cancer cells and induce anti-angiogenic effects in highly hypoxic tumours. Further study evaluating the distinctive value of SDF-1α expression in LARC patients receiving NCRT is warranted. However, we did not observe a relationship between expression of AMs before NCRT and tumour reponse. Therefore, it is not possible to choose the ‘right’ patients who may require additional therapeutics (such as anti-angiogenesis), except NCRT, by analhysis of the specimen before treatment. These findings are difficult for clinical application.

We also found that positive expression of PlGF after NCRT was correlated with resistance to NCRT in multivariate analyses. PlGF is a cytokine in the VEGF family of growth factors, with 53% homology to VEGF (30). It primarily regulates the angiogenic switch under pathologic states (31). PlGF recruits smooth muscle precursors that envelop developing vessels in tumours and together with VEGF produces more stable and mature vessels. PlGF may also facilitate metastasis by increasing the motility and invasion of malignant cells (32). Tumour overexpression of PlGF and VEGF together is associated with increased tumour angiogenesis and cancer growth (33,34). However, in general, there was no correlation between elevated VEGF expression and survival (35,36). Our results suggest that PlGF, than VEGF, is also an important target for resistance to NCRT. It would be worthwhile to determine whether or not PlGF is a predictive biomarker for patients with LARC receiving NCRT.

As shown in this study, an association was identified between HIF-1α and SDF-1α (P=0.034). HIF-1α was not correlated with VEGF and PlGF. However, SDF-1α had an association with PlGF (P=0.005). Although HIF-1α expression is known to drive expression of downstream proteins, differences in individual protein half-lives may not allow for a direct relationship between HIF-1α and other proteins (37). Downstream proteins may have been influenced by other signaling pathways independent of HIF-1α, making their expression levels somewhat variable in relation to HIF-1α. The limited sample size and the heterogeneity of intratumoural oxygenation may also be responsible for these findings.

In summary, SDF-1α and PlGF are relevant for resistance to NCRT. By comparison of pre-therapeutic and post-therapeutic intratumoural SDF-1α and PlGF, our results suggest that therapeutic strategies to downregulate expression of SDF-1α and PlGF during pre-operative treatment or to inhibit SDF-1α/PlGF mediated signaling pathways may further increase the individual tumour response and, as a consequence, improve patient prognosis. Based on our results, patients with increased expression of SDF-1α or positive expression of PlGF after NCRT might benefit from additional anti-SDF-1α/PlGF therapeutics.

Acknowledgements

The authors would like to thank Kim Hyung-Joo and Park So-Young for providing excellent technical assistance.

References

1 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2012. CA Cancer J Clin. 62:10–29. 2012. View Article : Google Scholar

2 

Sauer R, Becker H, Hohenberger W, Rodel C, Wittekind C, Fietkau R, Martus P, Tschmelitsch J, Hager E, Hess CF, Karstens JH, Liersch T, Schmidberger H and Raab R; German Rectal Cancer Study Group. Preoperative versus postoperative chemoradiotherapy for rectal cancer. N Engl J Med. 351:1731–1740. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Roh MS, Colangelo LH, O’Connell MJ, Yothers G, Deutsch M, Allegra CJ, Kahlenberg MS, Baez-Diaz L, Ursiny CS, Petrelli NJ and Wolmark N: Preoperative multimodality therapy improves disease-free survival in patients with carcinoma of the rectum: NSABP R-03. J Clin Oncol. 27:5124–5130. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Maas M, Nelemans PJ, Valentini V, Das P, Rödel C, Kuo LJ, Calvo FA, García-Aguilar J, Glynne-Jones R, Haustermans K, Mohiuddin M, Pucciarelli S, Small W Jr, Suárez J, Theodoropoulos G, Biondo S, Beets-Tan RG and Beets GL: Long-term outcome in patients with a pathological complete response after chemoradiation for rectal cancer: a pooled analysis of individual patient data. Lancet Oncol. 11:835–844. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Sprenger T, Rödel F, Beissbarth T, Conradi LC, Rothe H, Homayounfar K, Wolff HA, Ghadimi BM, Yildirim M, Becker H, Rödel C and Liersch T: Failure of downregulation of survivin following neoadjuvant radiochemotherapy in rectal cancer is associated with distant metastases and shortened survival. Clin Cancer Res. 17:1623–1631. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Edden Y, Wexner SD and Berho M: The use of molecular markers as a method to predict the response to neoadjuvant therapy for advanced stage rectal adenocarcinoma. Colorectal Dis. 14:555–561. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Trakarnsanga A, Ithimakin S and Weiser MR: Treatment of locally advanced rectal cancer: controversies and questions. World J Gastroenterol. 18:5521–5532. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Chin KF, Greenman J, Gardiner E, Kumar H, Topping K and Monson J: Pre-operative serum vascular endothelial growth factor can select patients for adjuvant treatment after curative resection in colorectal cancer. Br J Cancer. 83:1425–1431. 2000. View Article : Google Scholar

9 

Cascinu S, Graziano F, Catalano V, Staccioli MP, Rossi MC, Baldelli AM, Barni S, Brenna A, Secondino S, Muretto P and Catalano G: An analysis of p53, BAX and vascular endothelial growth factor expression in node-positive rectal cancer. Relationships with tumour recurrence and event-free survival of patients treated with adjuvant chemoradiation. Br J Cancer. 86:744–749. 2002. View Article : Google Scholar

10 

Willett CG, Duda DG, di Tomaso E, Boucher Y, Ancukiewicz M, Sahani DV, Lahdenranta J, Chung DC, Fischman AJ, Lauwers GY, Shellito P, Czito BG, Wong TZ, Paulson E, Poleski M, Vujaskovic Z, Bentley R, Chen HX, Clark JW and Jain RK: Efficacy, safety, and biomarkers of neoadjuvant bevacizumab, radiation therapy, and fluorouracil in rectal cancer: a multidisciplinary phase II study. J Clin Oncol. 27:3020–3026. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Brizel DM, Scully SP, Harrelson JM, Layfield LJ, Bean JM, Prosnitz LR and Dewhirst MW: Tumor oxygenation predicts for the likelihood of distant metastases in human soft tissue sarcoma. Cancer Res. 56:941–943. 1996.PubMed/NCBI

12 

Nordsmark M, Alsner J, Keller J, Nielsen OS, Jensen OM, Horsman MR and Overgaard J: Hypoxia in human soft tissue sarcomas: adverse impact on survival and no association with p53 mutations. Br J Cancer. 84:1070–1075. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Koukourakis MI, Bentzen SM, Giatromanolaki A, Wilson GD, Daley FM, Saunders MI, Dische S, Sivridis E and Harris AL: Endogenous markers of two separate hypoxia response pathways (hypoxia inducible factor 2 alpha and carbonic anhydrase 9) are associated with radiotherapy failure in head and neck cancer patients recruited in the CHART randomized trial. J Clin Oncol. 24:727–735. 2006. View Article : Google Scholar

14 

Sowter HM, Ratcliffe PJ, Watson P, Greenberg AH and Harris AL: HIF-1-dependent regulation of hypoxic induction of the cell death factors BNIP3 and NIX in human tumors. Cancer Res. 61:6669–6673. 2001.PubMed/NCBI

15 

Lal A, Peters H, St Croix B, Haroon ZA, Dewhirst MW, Strausberg RL, Kaanders JH, van der Kogel AJ and Riggins GJ: Transcriptional response to hypoxia in human tumors. J Natl Cancer Inst. 93:1337–1343. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Zhong H, De Marzo AM, Laughner E, Lim M, Hilton DA, Zagzag D, Buechler P, Isaacs WB, Semenza GL and Simons JW: Overexpression of hypoxia-inducible factor 1 alpha in common human cancers and their metastases. Cancer Res. 59:5830–5835. 1999.PubMed/NCBI

17 

Ishigami SI, Arii S, Furutani M, Niwano M, Harada T, Mizumoto M, Mori A, Onodera H and Imamura M: Predictive value of vascular endothelial growth factor (VEGF) in metastasis and prognosis of human colorectal cancer. Br J Cancer. 78:1379–1384. 1998. View Article : Google Scholar

18 

Rajaganeshan R, Prasad R, Guillou PJ, Poston G, Scott N and Jayne DG: The role of hypoxia in recurrence following resection of Dukes’ B colorectal cancer. Int J Colorectal Dis. 23:1049–1055. 2008.PubMed/NCBI

19 

Wei SC, Liang JT, Tsao PN, Hsieh FJ, Yu SC and Wong JM: Preoperative serum placenta growth factor level is a prognostic biomarker in colorectal cancer. Dis Colon Rectum. 52:1630–1636. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Saigusa S, Toiyama Y, Tanaka K, Yokoe T, Okugawa Y, Fujikawa H, Matsusita K, Kawamura M, Inoue Y, Miki C and Kusunoki M: Cancer-associated fibroblasts correlate with poor prognosis in rectal cancer after chemoradiotherapy. Int J Oncol. 38:655–663. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Greene FL, Page DL, Fleming ID, Fritz AG, Balch CM and Haller DG: AJCC Cancer Staging Handbook - TNM Classification of Malignant Tumors. 6th edition. Springer-Verlag; New York: 2002

22 

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

23 

Müller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, Barrera JL, Mohar A, Verastegui E and Zlotnik A: Involvement of chemokine receptors in breast cancer metastasis. Nature. 410:50–56. 2001.PubMed/NCBI

24 

Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL and Weinberg RA: Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 121:335–348. 2005. View Article : Google Scholar

25 

Schrader AJ, Lechner O, Templin M, Dittmar KE, Machtens S, Mengel M, Probst-Kepper M, Franzke A, Wollensak T, Gatzlaff P, Atzpodien J, Buer J and Lauber J: CXCR4/CXCL12 expression and signalling in kidney cancer. Br J Cancer. 86:1250–1256. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Taichman RS, Cooper C, Keller ET, Pienta KJ, Taichman NS and McCauley LK: Use of the stromal cell-derived factor-1/CXCR4 pathway in prostate cancer metastasis to bone. Cancer Res. 62:1832–1837. 2002.PubMed/NCBI

27 

Matsusue R, Kubo H, Hisamori S, Okoshi K, Takagi H, Hida K, Nakano K, Itami A, Kawada K, Nagayama S and Sakai Y: Hepatic stellate cells promote liver metastasis of colon cancer cells by the action of SDF-1/CXCR4 axis. Ann Surg Oncol. 16:2645–2653. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Yoshitake N, Fukui H, Yamagishi H, Sekikawa A, Fujii S, Tomita S, Ichikawa K, Imura J, Hiraishi H and Fujimori T: Expression of SDF-1 alpha and nuclear CXCR4 predicts lymph node metastasis in colorectal cancer. Br J Cancer. 98:1682–1689. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Heckmann D, Laufs S, Maier P, Zucknick M, Giordano FA, Veldwijk MR, Eckstein V, Wenz F, Zeller WJ, Fruehauf S and Allgayer H: A Lentiviral CXCR4 overexpression and knockdown model in colorectal cancer cell lines reveals plerixafor-dependent suppression of SDF-1α-induced migration and invasion. Onkologie. 34:502–508. 2011.PubMed/NCBI

30 

Wei SC, Tsao PN, Yu SC, Shun CT, Tsai-Wu JJ, Wu CH, Su YN, Hsieh FJ and Wong JM: Placenta growth factor expression is correlated with survival of patients with colorectal cancer. Gut. 54:666–672. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Carmeliet P, Moons L, Luttun A, Vincenti V, Compernolle V, De Mol M, Wu Y, Bono F, Devy L, Beck H, Scholz D, Acker T, DiPalma T, Dewerchin M, Noel A, Stalmans I, Barra A, Blacher S, VandenDriessche T, Ponten A, Eriksson U, Plate KH, Foidart JM, Schaper W, Charnock-Jones DS, Hicklin DJ, Herbert JM, Collen D and Persico MG: Synergism between vascular endothelial growth factor and placental growth factor contributes to angiogenesis and plasma extravasation in pathological conditions. Nat Med. 7:575–583. 2001. View Article : Google Scholar

32 

Fischer C, Jonckx B, Mazzone M, Zacchigna S, Loges S, Pattarini L, Chorianopoulos E, Liesenborghs L, Koch M, De Mol M, Autiero M, Wyns S, Plaisance S, Moons L, van Rooijen N, Giacca M, Stassen JM, Dewerchin M, Collen D and Carmeliet P: Anti-PlGF inhibits growth of VEGF(R)-inhibitor-resistant tumors without affecting healthy vessels. Cell. 131:463–475. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Landriscina M, Cassano A, Ratto C, Longo R, Ippoliti M, Palazzotti B, Crucitti F and Barone C: Quantitative analysis of basic fibroblast growth factor and vascular endothelial growth factor in human colorectal cancer. Br J Cancer. 78:765–770. 1998. View Article : Google Scholar : PubMed/NCBI

34 

Adini A, Kornaga T, Firoozbakht F and Benjamin LE: Placental growth factor is a survival factor for tumor endothelial cells and macrophages. Cancer Res. 62:2749–2752. 2002.PubMed/NCBI

35 

Lee JC, Chow NH, Wang ST and Huang SM: Prognostic value of vascular endothelial growth factor expression in colorectal cancer patients. Eur J Cancer. 36:748–753. 2000. View Article : Google Scholar : PubMed/NCBI

36 

Khorana AA, Ryan CK, Cox C, Eberly S and Sahasrabudhe DM: Vascular endothelial growth factor, CD68, and epidermal growth factor receptor expression and survival in patients with stage II and stage III colon carcinoma: a role for the host response in prognosis. Cancer. 97:960–968. 2003. View Article : Google Scholar

37 

Lee-Kong SA, Ruby JA, Chessin DB, Pucciarelli S, Shia J, Riedel ER, Nitti D and Guillem JG: Hypoxia-related proteins in patients with rectal cancer undergoing neoadjuvant combined modality therapy. Dis Colon Rectum. 55:990–995. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2014
Volume 32 Issue 6

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
Kim HJ, Bae SB, Jeong D, Kim ES, Kim C, Park D, Ahn TS, Cho SW, Shin EJ, Lee MS, Lee MS, et al: Upregulation of stromal cell-derived factor 1α expression is associated with the resistance to neoadjuvant chemoradiotherapy of locally advanced rectal cancer: Angiogenic markers of neoadjuvant chemoradiation. Oncol Rep 32: 2493-2500, 2014
APA
Kim, H.J., Bae, S.B., Jeong, D., Kim, E.S., Kim, C., Park, D. ... Baek, M.J. (2014). Upregulation of stromal cell-derived factor 1α expression is associated with the resistance to neoadjuvant chemoradiotherapy of locally advanced rectal cancer: Angiogenic markers of neoadjuvant chemoradiation. Oncology Reports, 32, 2493-2500. https://doi.org/10.3892/or.2014.3504
MLA
Kim, H. J., Bae, S. B., Jeong, D., Kim, E. S., Kim, C., Park, D., Ahn, T. S., Cho, S. W., Shin, E. J., Lee, M. S., Baek, M. J."Upregulation of stromal cell-derived factor 1α expression is associated with the resistance to neoadjuvant chemoradiotherapy of locally advanced rectal cancer: Angiogenic markers of neoadjuvant chemoradiation". Oncology Reports 32.6 (2014): 2493-2500.
Chicago
Kim, H. J., Bae, S. B., Jeong, D., Kim, E. S., Kim, C., Park, D., Ahn, T. S., Cho, S. W., Shin, E. J., Lee, M. S., Baek, M. J."Upregulation of stromal cell-derived factor 1α expression is associated with the resistance to neoadjuvant chemoradiotherapy of locally advanced rectal cancer: Angiogenic markers of neoadjuvant chemoradiation". Oncology Reports 32, no. 6 (2014): 2493-2500. https://doi.org/10.3892/or.2014.3504