Open Access

Regulatory T‑cell density and cytotoxic T lymphocyte density are associated with complete response to neoadjuvant paclitaxel and carboplatin chemoradiotherapy in gastric cancer

  • Authors:
    • Di Huang
    • Yongjiang Yang
    • Shuai Zhang
    • Zhuobin Su
    • Tao Peng
    • Xiaoyuan Wang
    • Yifeng Zhao
    • Shuguang Li
  • View Affiliations

  • Published online on: September 3, 2018     https://doi.org/10.3892/etm.2018.6684
  • Pages: 3813-3820
  • Copyright: © Huang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Regulatory T-cell density and cytotoxic T lymphocyte density are crucial in regulating antitumor immune responses. Tumor infiltration has marked therapeutic effects in gastric carcinoma, and there is evidence that chemoradiotherapy (CRT) exhibits an immune‑mediated component. In the present study, the density of CD4+ and CD8+ cells were evaluated in post‑CRT surgical samples from 68 patients with gastric cancer using immunohistochemistry. The associations between T‑cell density, cytotoxic T lymphocyte density and clinical survival rate were also analyzed. Cytotoxic T lymphocyte density was associated with gastric carcinoma regression grade and regulatory T‑cell density was also associated with gastric carcinoma regression grade. Of the patients who had a pathologic complete response, 84 and 76% were found to have a high CD3+ and CD4+ cell density, which was significantly different to patients who had a low CD3+ and CD4+ cell density. High cytotoxic T lymphocyte density was also associated with improved survival rates of patients with gastric cancer. In conclusion, these outcomes indicated that regulatory T‑cell density and cytotoxic T lymphocyte density in the tumor microenvironment were associated with the response to neoadjuvant CRT and may represent a therapeutic target for gastric cancer.

Introduction

Gastric cancer is one of the most common human malignancies and is the second most common cause of cancer-associated mortality in the world (13). A high risk of metastasis of gastric cancer cells and the resistance of cancer cells to apoptosis in patients with gastric cancer has been reported in the previous reports (46). Patients with gastric cancer present with higher morbidity and mortality rates, compared with other types of carcinoma of the digestive system (79). Therefore, investigating efficient antitumor treatments are required in the field of cancer research and treatments.

Currently, neoadjuvant chemoradiotherapy (CRT) is widely used for the treatment of locally advanced gastric cancer to inhibit tumor growth, making it more amenable to resection, and to decrease the risk of tumor cell metastasis (1013). Paclitaxel chemotherapy is an effective drug for advanced gastric cancer with acceptable toxic side-effects, and has been considered as an effective anticancer agent due to the smooth transition to the subsequent regimen (14). Carboplatin radiotherapy in combination with 2-(8-hydroxy-6-methoxy-1-oxo-1H-2-benzopyran-3-yl) propionic acid can inhibit gastric tumor growth, which may be an effective drug in the treatment of gastric cancer (15). However, the combined therapeutic effects of paclitaxel and carboplatin CRT in the CRT response in gastric cancer remain to be elucidated.

The presence of tumor-infiltrating lymphocytes (TILs) is associated with improved clinical outcome in gastric cancer, and TILs can suppress further invasion and/or metastasis of gastric carcinoma (16). A previous study indicated that the density of tumor-infiltrating T cells is a notable prognostic indicator for gastric cancer following treatment with CRT (17). TILs in gastric adenocarcinoma also present with marked compartmentalization with high numbers of lymphocytes in the stroma and low intraepithelial lymphocyte counts, which underline the importance of inflammation for tumor therapy (18). Another study showed that immunoscoring involves the assessment of T-cell density in the central tumor and invasive margin based on expression of pairs of T-cell markers (CD3 and CD8, and CD3 and CD45), which can be used to evaluate the prognostic and histopathologic tumor-node-metastasis stage for patients with cancer (19). Therefore, evaluating the association between therapeutic effects of antitumor agents and the pathologic complete response (pCR) to neoadjuvant CRT is crucial for the treatment of patients with gastric cancer.

In the present study, the T regulatory-Treg-lymphocyte density and cytotoxic T lymphocyte density were analyzed in surgically resected samples from 68 patients with locally advanced gastric cancer treated with CRT (paclitaxel and carboplatin). The results showed that regulatory T-cell density and cytotoxic T lymphocyte density were associated with pCR to neoadjuvant CRT (paclitaxel and carboplatin) in the gastric cancer response to CRT.

Materials and methods

Patients, treatment and follow-up

A total of 68 patients with gastric cancer were recruited at The First Affiliated Hospital of Hebei North University (Hebei, China) between February 2010 and May 2015. Clinical gastric tumor stage was determined by computed tomography and magnetic resonance imaging prior to treatment with CRT. Neoadjuvant radiotherapy, comprising paclitaxel and carboplatin, was administered for 4 weeks. Surgical resection (20), involving curative (R0) resection and D2 lymphadenectomy, of the gastric tumor was performed following the completion of neoadjuvant treatment. The follow-up period in the present study was a total of 60 months, every 5 months. The survival rate of patients with gastric cancer was recorded and cancer-specific survival (CSS) was analyzed as the time between the date of surgery and the date the patient succumbed to mortality from gastric cancer. Recurrence-free survival (RFS) was evaluated as the time between the date of surgery and the date of first gastric cancer recurrence.

CRT administration

The patients received paclitaxel (80 mg/m2 per day intravenously on days 1, 8, and 15; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) in combination with carboplatin (75 mg/m2 per day intravenously on days 1, 8, and 15, Sigma-Aldrich; Merck KGaA) in 4-week treatment cycles.

Histopathologic assessment

Tumor regression grading was analyzed as part of routine pathological review using the Dworak system (21) and defined as grade 4 corresponding to pCR and grade 0 corresponding to no regression.

Distinguishing regulatory T cells and cytotoxic T lymphocytes

Regulatory T cells were distinguished using typical markers CD4+CD25+Foxp3+, as described previously (22). Cytotoxic T lymphocytes were distinguished using typical markers CD3+CD4CD8+, as described previously (23).

Immunohistochemistry

The fresh tissue specimens were deparaffinized in xylene and dehydrated through a graduated alcohol series. The paraffin-embedded tissue sections (4-µm) were prepared and epitope retrieval was performed using Tris-EDTA buffer solution (pH 9.0, Sigma-Aldrich; Merck KGaA; cat. no. SRE0063) for 60 min at 65°C. The paraffin sections were subjected with hydrogen peroxide (3%) for 15 min at 37°C and then blocked with 5% BSA (Sigma-Aldrich; Merck KGaA) for 2 h at 37°C. The sections were incubated with anti-CD3 (1:1,000, cat. no. ab16669; Abcam, Cambridge, UK), anti-CD4 (1:1,000, cat. no. ab133616; Abcam), and anti-CD8 (1:1,000, cat. no. ab4055; Abcam), respectively, for 12 h at 4°C. All sections were washed three times with PBS for 15 min at 37°C and then incubated with goat anti-rabbit IgG H&L secondary antibodies (Alexa Fluor® 488, 1:1,000, cat. no. ab150077; Abcam) for 2 h at 37°C. The tumor sections were observed in three-random views under a fluorescence microscope (YS100; Nikon Corporation, Tokyo, Japan). Densitometric quantification of the immunohistochemistry data was performed by using Quantity-One 1.0 software (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Assessment of T-cell density

Images of the gastric tumor sections (4-µm) were captured using a high-resolution digital scanner (Aperio Scanscope XT; Leica Biosystems, North Ryde, NSW, Australia) at ×40 magnification. The T-cell density (cells per mm2 tissue) was analyzed using software (Aperio Imagescope version 11; Leica Biosystems).

Statistical analysis

Data are presented as the mean and standard error of the mean and statistical analyses were performed using SAS version 9.3 (SAS Institute, Inc., Cary, NC, USA). Differences in T-cell density in each group were analyzed using general linear models. Associations of clinical variables and pCR were analyzed using logistic regression. The Kaplan-Meier method with and the log-rank test was used to analyze time-to-event outcomes (CSS and RFS). The prognostic value of CD3+, CD4+ and CD8+ for cancer-specific survival rate was analyzed using the Kaplan-Meier method and multivariate Cox proportional hazard model analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

Patient characteristics and treatment response

A total of 68 patients with gastric cancer were identified and tumor samples from these 68 patients were included in the tissue microarrays. The clinical characteristics of the 68 patients with gastric cancer are shown in Table I. There were 48 (70.6%) patients with clinical T3 stage, 12 (17.6%) patients with clinical T2 stage 2, four (5.9%) patients with clinical T4 stage and four (5.9%) patients with metastatic disease (Table II).

Table I.

Characteristics of patients with gastric cancer.

Table I.

Characteristics of patients with gastric cancer.

Characteristicn
Patients68
Male34
Female34
Age in years, mean (SD)63 (12.3)a
Weeks between end of CRT and surgery, median (IQR)6 (5,7)b
Pretreatment clinical T stage, n (%)
  T212 (17.6)
  T348 (70.6)
  T44 (5.9)

[i] CRD, chemoradiotherapy.

Table II.

Clinical and pathological response of patients with gastric cancer to neoadjuvant chemoradiotherapy paclitaxel and carboplatin.

Table II.

Clinical and pathological response of patients with gastric cancer to neoadjuvant chemoradiotherapy paclitaxel and carboplatin.

Stagen (%)
T212 (17.6)
T348 (70.6)
T44 (5.9)
Metastatic4 (5.9)
Regulatory T-cell density in the gastric tumor microenvironment following CRT

The present study analyzed the regulatory T-cell density in the gastric tumor microenvironment following CRT. The density of CD3+ and CD4+ T cells were decreased in the T4 stage gastric tumor tissue (Fig. 1A). The T3 stage gastric tumor tissue showed a similar distribution of CD3+ and CD4+ T cells (Fig. 1B). However, the density of CD3+ and CD4+ T cells were increased in the T2 gastric tumor tissue (Fig. 1C).

Cytotoxic T lymphocyte in the gastric tumor microenvironment following CRT

The present study analyzed the cytotoxic T lymphocyte density in the gastric tumor microenvironment following CRT. The T4 stage gastric tumor tissue exhibited a lower density of CD8+ cells compared with prior to CRT (Fig. 2A). The density of CD8+ cells in T3 stage gastric tumor tissue did not differ significantly between pre-CRT and post CRT (Fig. 2B). The density of CD8+ cells were increased in T2 stage gastric tumor tissue (Fig. 2C).

Regulatory T-cell density is associated with the response to CRT

Analysis of the regulatory T-cell density revealed an inverse correlation between CD3+, CD4+ cell density in gastric cancer regression (r=−0.204 and −0.194, respectively, Fig. 3A). Among the patients who had pCR, 31% had a low density of CD3+ cells, which was lower than 56% of those with pCR with a high CD3+ cell density (Fig. 3B). The results also demonstrated that, of patients with a high CD4+ cell density, 62% had a pCR, which was significantly higher than that in patients with a low CD4+ cell density (Fig. 3C).

Cytotoxic T lymphocyte density is associated with the response to CRT

The association between cytotoxic T lymphocytes and pCR was also analyzed in the present study. As shown in Fig. 3A, cytotoxic T lymphocyte CD8+ cells had an inverse correlation with gastric cancer regression (r=−0.312, Fig. 4A). It was observed that 65% patients with a high CD8+ cell density showed pCR, whereas 32% patients with a low CD8+ cell density showed pCR (Fig. 4B).

Regulatory T-cell and cytotoxic T lymphocyte density as a prognostic marker in neoadjuvantly treated gastric cancer

A total of 56 patients (82%) survived the 60-month follow-up from the date of surgery, 12 (21%) patients succumbed to mortality during the follow-up period, 28 patients (50%) survived without cancer recurrence, 12 patients (21%) showed local recurrence, and four patients (8%) showed colon metastases (Table III). It was shown that CD3+ and CD4+ cell density were associated with the survival rate of patients with gastric cancer (Fig. 5A-D). The probabilities of 5-year CSS and RFS were 79, vs. 64% and 72, vs. 56% in the CD3+ high, vs. low groups, respectively (Fig. 5A and B). The probabilities of 5-year CSS and RFS were 84, vs. 64% and 82, vs. 62% in the CD4+ high, vs. low groups, respectively (Fig. 5C and D). It was also shown that CD8+ cell density was associated with the survival rates of patients with gastric cancer (Fig. 5E and F). The probabilities of 5-year CSS and RFS were 75%, vs. 60% and 81, vs. 62% in the CD8+ high, vs. low groups, respectively (Fig. 5E and F). The densities of CD3+, CD4+ and CD8+ cells were associated with CSS of patients with gastric cancer (Table IV). In addition, tumor size and concomitant diseases markedly affected the CSS of patients with gastric cancer.

Table III.

Clinical outcomes of patients with gastric cancer following neoadjuvant chemoradiotherapy with paclitaxel and carboplatin.

Table III.

Clinical outcomes of patients with gastric cancer following neoadjuvant chemoradiotherapy with paclitaxel and carboplatin.

Outcomen (%)
Mortality12 (21)
Survival without cancer recurrence28 (50)
Local recurrence12 (21)
Colon metastases4 (8)

Table IV.

Univariate and multivariable analyses of prognostic factors for CSS following curative gastric cancer resection.

Table IV.

Univariate and multivariable analyses of prognostic factors for CSS following curative gastric cancer resection.

CSS

UnivariateMultivariate


Prognostic factorLog-rankP-valueHR95% CIP-value
CD3+ cell density
  Interaction
  No interaction1.1930.026a1.7041.104–2.8240.038a
CD4+ cell density
  Interaction
  No interaction1.8260.0580.8860.914–2.3200.026a
CD8+ cell density
  Interaction
  No interaction2.5200.037a2.1581.104–2.8240.025a
Concomitant disease
  Interaction
  No interaction4.7260.016a1.8621.426–3.0260.018a
Tumor size (mm)
  <503.7460.014a1.6381.242–2.8580.0163a
  ≥50

a P<0.05 was considered to indicate a statistically significant difference. CSS, cancer-specific survival; HR, hazard ratio; CI, confidence interval.

Discussion

Although previous reports have shown that CRT treatment can inhibit gastric cancer growth in the process of cancer progression (2426), the associations between regulatory T-cell density, cytotoxic T lymphocyte density and pCR to paclitaxel and carboplatin have not reported in gastric cancer. In the present study, the densities of regulatory T-cell density and cytotoxic T lymphocytes were analyzed in gastric cancer tissues from a total of 68 patients. An association was found between regulatory T-cell density and cytotoxic T lymphocyte density in the local tumor microenvironment following neoadjuvant CRT of paclitaxel and carboplatin in locally gastric cancer. Owing to the evidence of an immune-mediated component to chemoradiotherapy, it was hypothesized that regulatory T-cells inhibited the response of gastric carcinoma to neoadjuvant CRT and cytotoxic T lymphocytes promoted the response of gastric carcinoma to neoadjuvant CRT.

Currently, paclitaxel is one of the most widely used clinical anticancer drugs in chemotherapy (27). The combined administration of paclitaxel has been reported to markedly enhance tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis in resistant cancer cells in vitro and in vivo (28), and Zhu et al indicated that CD4+Foxp3+ regulatory T-cell impairment by paclitaxel is independent of Toll-like receptor 4 (29). In the present study, the results revealed that the regulatory T-cell density of CD3+ and CD4+ T cells were decreased in T4 stage gastric tumor and increased in T2 gastric tumor following CRT. However, T3 stage gastric tumor showed similar distribution of CD3+ and CD4+ T cells between pre-CRT and post-CRT gastric cancer tissues. The findings also indicated that regulatory T-cell CD3+, CD4+ cell density was inversely correlated with gastric cancer regression. However, further analysis is required in larger gastric cancer populations.

Carboplatin is active in advanced gastric and esophageal cancer (30). A report found that carboplatin and paclitaxel added to radiation and fluoropyrimidine analogs is a well-tolerated regimen in the adjuvant setting, which is a safe and feasible regimen in this relatively high-risk group of patients with gastric cancer and is of interest for future development (31). The density of tumor-infiltrating T cells (CD8+ T cells), is an indicator of improved survival rate in patients with gastric cancer, and the CD8 T-cell density predicts the likelihood of tumor regression following CRT (32). In the present study, it was found that cytotoxic T lymphocyte CD38+ CD8+ density was decreased in T4 stage gastric tumor tissue and increased in T2 stage gastric tumor tissue. Cytotoxic T lymphocyte CD3+ CD8+ density was also inversely correlated with gastric cancer regression.

In conclusion, the results of the present study indicated that regulatory T-cell and cytotoxic T lymphocyte density may be considered as prognostic markers in neoadjuvantly treated gastric cancer. Notably, regulatory T-cell and cytotoxic T lymphocyte density were associated with pCR and the improved long-term outcome of patients with gastric cancer. These results suggest that regulatory T-cells in the local microenvironment may inhibit the response of gastric cancer to neoadjuvant CRT and cytotoxic T lymphocytes may promote the response of gastric cancer to CRT.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

DH performed the experiments. YY, SZ, ZS, TP, XW and YZ prepared and analysed experimental data. SL designed the experiments.

Ethics approval and consent to participate

This study was approved by the Ethics Committee of the First Affiliated Hospital of Hebei North University. All patients provided written informed consent prior to undergoing any protocol-specific screening procedures or treatments.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Visa L, Jimenez-Fonseca P, Martinez EA, Hernández R, Custodio A, Garrido M, Viudez A, Buxo E, Echavarria I, Cano JM, et al: Efficacy and safety of chemotherapy in older versus non-older patients with advanced gastric cancer: A real-world data, non-inferiority analysis. J Geriatr Oncol. 9:254–264. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Suzuki H and Mori H: World trends for H. pylori eradication therapy and gastric cancer prevention strategy by H. pylori test-and-treat. J Gastroenterol. 53:354–361. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Carmona-Bayonas A, Jimenez-Fonseca P, Custodio A, Sánchez Cánovas M, Hernández R, Pericay C, Echavarria I, Lacalle A, Visa L, Palomo Rodríguez A, et al: Anthracycline-based triplets do not improve the efficacy of platinum-fluoropyrimidine doublets in first-line treatment of advanced gastric cancer: Real-world data from the AGAMEMON National Cancer Registry. Gastric Cancer. 21:96–105. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Sun N, Sun Q, Liu Q, Zhang T, Zhu Q, Wang W, Cao M and Zang QI: α-fetoprotein-producing gastric carcinoma: A case report of a rare subtype and literature review. Oncol Lett. 11:3101–3104. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Tian Q, Zeng J, Tao X, Zhang Z, Zhou X and Wang Y: Clinical pathology of metastatic gastric carcinoma to the breast: A report of two cases and a review of literature. Oncol Lett. 11:3081–3084. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Petrioli R, Roviello G, Zanotti L, Roviello F, Polom K, Bottini A, Marano L, Francini E, Marrelli D and Generali D: Epirubicin-based compared with docetaxel-based chemotherapy for advanced gastric carcinoma: A systematic review and meta-analysis. Crit Rev Oncol Hematol. 102:82–88. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Shan B, Shan L, Morris D, Golani S and Saxena A: Systematic review on quality of life outcomes after gastrectomy for gastric carcinoma. J Gastrointest Oncol. 6:544–560. 2015.PubMed/NCBI

8 

Bollschweiler E, Berlth F, Baltin C, Mönig S and Hölscher AH: Treatment of early gastric cancer in the Western World. World J Gastroenterol. 20:5672–5678. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Yoshikawa T, Aoyama T, Hayashi T, Kuwabara H, Mikayama Y, Ogata T, Cho H and Tsuburaya A: Neoadjuvant chemotherapy for gastric cancer-evidence from the world and future strategy in Japan. Gan To Kagaku Ryoho. 39:866–870. 2012.(In Japanese). PubMed/NCBI

10 

Kocáková I, Vetchá H, Soumarová R and Vyzula R: Role of adjuvant chemoradiotherapy in the treatment of gastric carcinoma. Cas Lek Cesk. 142 Suppl 1:S26–S28. 2003.(In Czech).

11 

Sher DJ: Neoadjuvant chemoradiotherapy for stage III Non-small cell lung cancer. Front Oncol. 7:2812017. View Article : Google Scholar : PubMed/NCBI

12 

van der Sluis FJ, van Westreenen HL, van Etten B, van Leeuwen BL and de Bock GH: Pretreatment identification of patients likely to have pathologic complete response after neoadjuvant chemoradiotherapy for rectal cancer. Int J Colorectal Dis. 33:149–157. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Sun Y, Zhang Y, Wu X, Lin H, Lu X, Huang Y, Xu Z, Huang S, Wang X and Chi P: Prognostic significance of neoadjuvant rectal score in locally advanced rectal cancer after neoadjuvant chemoradiotherapy and construction of a prediction model. J Surg Oncol. 117:737–744. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Rino Y, Yukawa N, Wada N, Suzuki M, Murakami H, Yamada T, Nakayama H, Yamamoto N, Sato T, Yamada R, et al: Phase II study of S-1 monotherapy as a First-line, combination therapy of S-1 plus cisplatin as a Second-line, and weekly paclitaxel monotherapy as a Third-line therapy in patients with advanced gastric carcinoma: Phase II study of S-1, S-1 plus cisplatin, and weekly paclitaxel in patients with advanced gastric carcinoma. Clin Med Oncol. 2:375–383. 2008.PubMed/NCBI

15 

Chen JL, Zhu JS, Hong J, Chen MX, Lu JL, Chen WX, Shen B, Zhu ZM and Chen NW: Effect of 2-(8-hydroxy-6-methoxy-1-oxo-1H-2-benzopyran-3-yl) propionic acid in combination with carboplatin on gastric carcinoma growth in vivo. World J Gastroenterol. 13:509–514. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Fukuda K, Tsujitani S, Maeta Y, Yamaguchi K, Ikeguchi M and Kaibara N: The expression of RCAS1 and tumor infiltrating lymphocytes in patients with T3 gastric carcinoma. Gastric Cancer. 5:220–227. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Koyama S: Differential expression of intracellular apoptotic signaling molecules in tumor and tumor-infiltrating lymphocytes during development of invasion and/or metastasis of gastric carcinoma. Dig Dis Sci. 48:2290–2300. 2003. View Article : Google Scholar : PubMed/NCBI

18 

Haas M, Buttner M, Rau TT, Fietkau R, Grabenbauer GG and Distel LV: Inflammation in gastric adenocarcinoma of the cardia: How do EBV infection, Her2 amplification and cancer progression influence tumor-infiltrating lymphocytes? Virchows Arch. 458:403–411. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Mlecnik B, Tosolini M, Kirilovsky A, Berger A, Bindea G, Meatchi T, Bruneval P, Trajanoski Z, Fridman WH, Pagès F and Galon J: Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction. J Clin Oncol. 29:610–618. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Waddell T, Verheij M, Allum W, Cunningham D, Cervantes A and Arnold D: Gastric cancer: ESMO-ESSO-ESTRO clinical practice guidelines for diagnosis, treatment and follow-up. Eur J Surg Oncol. 40:584–591. 2014. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Szurek E, Cebula A, Wojciech L, Pietrzak M, Rempala G, Kisielow P and Ignatowicz L: Differences in expression level of helios and Neuropilin-1 do not distinguish thymus-derived from extrathymically-induced CD4+Foxp3+ regulatory T cells. PLoS One. 10:e01411612015. View Article : Google Scholar : PubMed/NCBI

23 

Brandt D, Sergon M, Abraham S, Mäbert K and Hedrich CM: TCR+CD3+CD4CD8 effector T cells in psoriasis. Clin Immunol. 181:51–59. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Wang X, Shen Y, Zhu H, Zhao Y, Li Z, Qiu M, Li Q, Gou H, Yang Y, Cao D, et al: A phase II trial of concurrent 3D-CRT/IMRT and oxaliplatin, 5-fluorouracil and leucovorin (FOLFOX) in gastric cancer patients with R0 gastrectomy and D2 lymph node dissection. Gastric Cancer. 19:245–254. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Wang X, Li G, Zhang Y, Bai S, Xu F, Wei Y and Gong Y: Single-arc volumetric-modulated arc therapy (sVMAT) as adjuvant treatment for gastric cancer: Dosimetric comparisons with three-dimensional conformal radiotherapy (3D-CRT) and intensity-modulated radiotherapy (IMRT). Med Dosim. 38:395–400. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Papadimitriou K, Vassiliou V, Kountourakis P, Polyviou P, Andreopoulos D and Papamichael D: Adjuvant chemoradiotherapy (CRT) for high-risk gastric cancer (GC) patients: Single-center experience using infusional 5-fluorouracil (5FU) and radiotherapy (RT). Med Oncol. 29:2716–2717. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Plosker GL and Hurst M: Paclitaxel: A pharmacoeconomic review of its use in non-small cell lung cancer. Pharmacoeconomics. 19:1111–1134. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Li L, Wen XZ, Bu ZD, Cheng XJ, Xing XF, Wang XH, Zhang LH, Guo T, Du H, Hu Y, et al: Paclitaxel enhances tumoricidal potential of TRAIL via inhibition of MAPK in resistant gastric cancer cells. Oncol Rep. 35:3009–3017. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Zhu Y, Liu N, Xiong SD, Zheng YJ and Chu YW: CD4+Foxp3+ regulatory T-cell impairment by paclitaxel is independent of toll-like receptor 4. Scand J Immunol. 73:301–308. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Wang X, Pang L and Feng J: A phase II study of etoposide, doxorubicin, and carboplatin in the treatment of advanced gastric cancer. Am J Clin Oncol. 25:71–75. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Mobayed M, Heilbrun LK, Shields AF, Washington T, Venkatramanamoorthy R, Philip PA and El-Rayes BF: Safety and feasibility of carboplatin and paclitaxel followed by fluoropyrimidine analogs and radiation as adjuvant therapy for gastric cancer. Case Rep Oncol. 2:220–228. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Lu X, Yang L, Yao D, Wu X, Li J, Liu X, Deng L, Huang C, Wang Y, Li D and Liu J: Tumor antigen-specific CD8+ T cells are negatively regulated by PD-1 and Tim-3 in human gastric cancer. Cell Immunol. 313:43–51. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2018
Volume 16 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang D, Yang Y, Zhang S, Su Z, Peng T, Wang X, Zhao Y and Li S: Regulatory T‑cell density and cytotoxic T lymphocyte density are associated with complete response to neoadjuvant paclitaxel and carboplatin chemoradiotherapy in gastric cancer. Exp Ther Med 16: 3813-3820, 2018
APA
Huang, D., Yang, Y., Zhang, S., Su, Z., Peng, T., Wang, X. ... Li, S. (2018). Regulatory T‑cell density and cytotoxic T lymphocyte density are associated with complete response to neoadjuvant paclitaxel and carboplatin chemoradiotherapy in gastric cancer. Experimental and Therapeutic Medicine, 16, 3813-3820. https://doi.org/10.3892/etm.2018.6684
MLA
Huang, D., Yang, Y., Zhang, S., Su, Z., Peng, T., Wang, X., Zhao, Y., Li, S."Regulatory T‑cell density and cytotoxic T lymphocyte density are associated with complete response to neoadjuvant paclitaxel and carboplatin chemoradiotherapy in gastric cancer". Experimental and Therapeutic Medicine 16.5 (2018): 3813-3820.
Chicago
Huang, D., Yang, Y., Zhang, S., Su, Z., Peng, T., Wang, X., Zhao, Y., Li, S."Regulatory T‑cell density and cytotoxic T lymphocyte density are associated with complete response to neoadjuvant paclitaxel and carboplatin chemoradiotherapy in gastric cancer". Experimental and Therapeutic Medicine 16, no. 5 (2018): 3813-3820. https://doi.org/10.3892/etm.2018.6684