Open Access

Effect of gastrointestinal microbiome and its diversity on the expression of tumor‑infiltrating lymphocytes in breast cancer

  • Authors:
    • Jiajie Shi
    • Cuizhi Geng
    • Meixiang Sang
    • Wei Gao
    • Sainan Li
    • Shan Yang
    • Zheng Li
  • View Affiliations

  • Published online on: March 22, 2019     https://doi.org/10.3892/ol.2019.10187
  • Pages: 5050-5056
  • Copyright: © Shi 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

The diversity of the gastrointestinal microbiome is closely associated with human health. In the present study, the gastrointestinal microbiome and tumor‑infiltrating lymphocytes (TILs) were compared in patients with breast cancer (BC). A total of 80 patients with BC were divided into three groups based on the expression of TILs, as follows: High expression of TILs (TIL‑H), medium expression of TILs (TIL‑M) and low expression of TILs (TIL‑L). DNA of the gastrointestinal microbiome was determined by Illumina sequencing and taxonomy of 16S ribosomal RNA genes. A χ2 test and UniFrac analysis of β‑diversity were applied to assess the association between clinical characteristics and diversity of the gastrointestinal microbiome. The β‑diversity distribution was statistically significant (weighted UniFrac, P<0.01; unweighted UniFrac, P<0.01) when comparing the TIL‑L and TIL‑H groups and when comparing the three groups (TIL‑H vs. TIL‑M vs. TIL‑L). At the genus level, higher abundances of Mycobacterium, Rhodococcus, Catenibacterium, Bulleidia, Anaerofilum, Sneathia, Devosia and TG5, but lower abundances of Methanosphaera and Anaerobiospirillum (P<0.05) were identified in the TIL‑L group compared with the TIL‑H group. At the species level, the stercoris, barnesiae, coprophilus, flavefaciens and C21_c20 species exhibited a higher abundance in the TIL‑L group, whereas producta and komagatae exhibited a greater abundance in the TIL‑H group (P<0.05). Collectively, the diversity of the gastrointestinal microbiome was associated with the expression of TILs in patients with BC.

Introduction

Breast cancer (BC) is the most common malignant tumor and the primary cause of cancer-associated mortality in women worldwide (1). Although numerous treatments, including chemotherapy, radiotherapy, endocrine therapy and targeted therapy, are currently available for BC, the response of patients greatly varies partly due to their own antitumor immunity (2). Accumulating evidence suggests that adaptive immunity mediated by T and B lymphocytes provides the critical foundation for effective and sustained antitumor responses. Tumor-infiltrating lymphocytes (TILs) are likely to be the most relevant indicator of tumor immunity in solid tumors, with prognostic value (3). In BC, extensive tumor infiltration by cytotoxic CD8+ T cells is markedly associated with patient survival (4,5) and response to therapy (6). Furthermore, the baseline expression of TILs can predict the pathological complete response (pCR) result following neoadjuvant chemotherapy in patients with BC (7), which is an important prognostic indicator.

Previous studies revealed that the composition of the gastrointestinal microbiome is a major environmental factor that varies among individuals, which may affect systemic immunity (8,9). The gastrointestinal microbiome has been demonstrated to initiate the differentiation of T cells, the expansion of specific molecular subsets (10,11) and the activation state of innate antigen-presenting cells (APCs), which may eventually affect priming of the systemic immune response (12,13). In addition, the gastrointestinal microbiome may improve the outcomes of cancer treatment by impairing inflammatory activation in response to different therapeutic protocols (14). Numerous studies have confirmed the association between the gastrointestinal microbiome and tumors, particularly in colon carcinoma (1517). However, for extraintestinal tumors, to the best of our knowledge, such an association has not been established. The present study aimed to assess whether the diversity of the gastrointestinal microbiome was associated with different expression patterns of TILs in patients with BC.

Materials and methods

Patients

Between March 2017 and October 2017, a total of 90 biopsy-confirmed female patients with BC were enrolled in the present study at the Breast Center of The Fourth Hospital of Hebei Medical University (Shijiazhuang, China). All patients were first treated by chemotherapy, followed by surgical treatment, as appropriate. Available clinicopathological data included age, staging, menstrual state, estrogen receptor (ER) and/or progesterone receptor (PgR) status, human epidermal growth factor receptor 2 (HER2) status, TIL classification, and pCR cases. ER and PgR were assessed as positive if ≥1% of tumor cells exhibited nuclear staining (18). HER2-positive status was defined as a score of 3+ based on immunohistochemistry assay or HER2 gene amplification using fluorescent in situ hybridization, as described previously (19). The Miller-Payne grading system was used to evaluate the pathological response in surgical specimens (20), and pCR was defined as the absence of residual invasive tumor cells in the breast and axillary lymph nodes (ypT0/is + ypN0) in surgical specimens. All procedures were supervised and approved by the Human Tissue Research Committee of The Fourth Hospital of Hebei Medical University, and informed consent was provided by all participants.

Assessment of TILs using a three-grade scale

Core needle biopsy was performed in the examination room. Briefly, between three and five lump tissues were obtained from different directions to obtain a suitable number of tissue samples. Subsequently, the tissue strip was placed in 4% neutral formalin solution and sent to the Pathology Department. Evaluation of TILs on the core needle biopsy specimens was performed by two experienced pathologists who were familiar with the evaluation criteria recommended by the International TILs Working Group in 2014 (21). The whole slide was screened using a low-power field, while an area with many lymphocytes was identified as a ‘hotspot’. TILs were then evaluated by light microscopy in a medium-power field (magnification, ×100). The region of interest was restricted within the tumor borders as described by Salgado et al (21). TIL score was defined as the proportion of the area infiltrated by lymphocytes within the tumor itself plus the adjacent stroma, and the scores were classified as low (<10), intermediate (10–50) and high (>50%), accordingly (22).

16S ribosomal DNA (rDNA) amplification

Fresh fecal samples were collected from the 90 patients with BC and stored at −80°C. DNA was extracted from all samples using a ProbeGene® Soil genomic DNA extraction kit (ProbeGene, Jiangsu, China) according to the manufacturer's protocol, and purified DNA was stored at −80°C prior to further analysis.

The 16S rDNA V3-V4 region of the ribosomal RNA gene was amplified by polymerase chain reaction using primers 341F (5′-CCTACGGGNGGCWGCAG-3′) and 806R (5′-GGACTACHVGGGTATCTAAT-3′), where the barcode was an eight-base sequence unique to each sample. Polymerase chain reaction was performed in a 50-µl reaction system consisting of 5 µl 10X KOD buffer, 5 µl 2.5 mM deoxyribonucleotide triphosphates, 1.5 µl of each primer (5 µM), 1 µl KOD polymerase (Toyobo (Shanghai) Co., Ltd., Shanghai, China) and 100 ng template DNA. Briefly, following a denaturation step at 95°C for 2 min, the amplifications were carried out with 27 cycles at a melting temperature of 98°C for 10 sec, an annealing temperature of 62°C for 30 sec, and an extension temperature of 68°C for 30 sec, followed by a final extension step at 68°C for 10 min. Each experiment was conducted in triplicate. Amplicons were subjected to electrophoresis on 2% agarose gels, purified using the AxyPrep DNA Gel Extraction kit (Axygen; Corning Inc., Corning, NY, US), according to the manufacturer's protocol, and semi-quantified using the QuantiFluor dsDNA system (Promega Corporation, Madison, WI, US). Purified amplicons were pooled in equimolar concentrations and underwent paired-end sequencing (2×250) on the Illumina HiSeq2500 platform (Illumina, Inc., San Diego, CA, USA) according to the manufacturer's protocols.

Statistical analysis

Statistical analysis was performed using SPSS 23.0 software (SPSS, Inc., Chicago, IL, USA). Clinical characteristics, including age, menopausal state, staging, level of HER2 and ER/PgR expression, were analyzed using a χ2 test. P<0.05 was considered to indicate a statistically significant difference.

Raw data from Illumina sequencing contain adapters or low-quality reads that may affect subsequent data assembly and analysis. Therefore, to obtain high-quality clean reads, raw reads were filtered according to the following criteria: i) Reads containing >10% unknown nucleotides were removed; and ii) reads containing <80% high-quality bases (Q-value, >20) were excluded.

Paired-end clean reads were merged as raw tags using FLASH v1.2.11 (23) with a minimum overlap of 10 bp and a mismatch error rate of 2%. Noisy sequences of raw tags were filtered by QIIME v1.9.1 (24) pipeline under specific filtering conditions (25). Clean tags were searched against the reference database (http://drive5.com/uchime/uchime_download.html) to perform reference-based chimera detection using the UCHIME algorithm (http://www.drive5.com/usearch/manual/uchime_algo.html). All chimeric tags were removed, and effective tags were finally obtained for further analysis.

The effective tags were clustered into operational taxonomic units (OTUs) of ≥97% similarity using the UPARSE (26) pipeline. The tag sequence with highest abundance was selected as a representative sequence within each cluster. Venn analysis was performed among groups to identify unique and common OTUs. The representative sequences were classified into organisms by a naive Bayesian model using RDP classifier v2.2 (27) based on the Greengenes (28) database (https://www.arb-silva.de/). Weighted and unweighted UniFrac distance matrices were generated using QIIME for β-diversity analysis. Between-group comparison of β-diversity was performed using Welch's t-test and Wilcoxon rank test in R. β-diversity comparison among groups was computed using Tukey's HSD test and Kruskal-Wallis H test in R. Analysis of similarity (ANOSIM) was used to test whether the differences among groups were significantly greater than those within groups. Biomarker features in each group were screened by Metastats software (v.20090414) (29).

Results

Clinical characteristics

A total of 80 patients were included in the present study (10 cases were unavailable since mass was removed in another hospital or the biopsy section could not be found) and divided into three groups as follows: High expression of TILs (TIL-H; n=21), medium expression of TILs (TIL-M; n=34) and low expression of TILs (TIL-L; n=25). Associations between TIL distribution and clinical characteristics, including age, menstrual status, staging, HER2 expression and ER/PgR expression, were assessed (Table I). Only the expression status of HER2 was positively associated with TIL distribution (P=0.037). A total of 58 patients who underwent surgery following chemotherapy (20 from TIL-H group, 22 from TIL-M group and 16 from TIL-L group) were evaluated for chemotherapy efficiency (Table II).

Table I.

Clinical characteristics associated with TILs.

Table I.

Clinical characteristics associated with TILs.

No. of cases

CharacteristicsTIL-lowTIL-mediumTIL-highχ2P-value
Age (years) 0.5410.736
  <457147
  45–5961212
  ≥60786
Staging 2.7010.259
  I000
  II757
  III112012
  IV396
Menopausal state 0.2690.874
  Yes111915
  No101510
HER2 6.5970.037
  Positive6713
  Negative152612
ER/PgR 3.2510.197
  Positive182515
  Negative3810

[i] ER, estrogen receptor; HER2, human epidermal growth factor receptor 2; PgR, progesterone receptor; TIL, tumor-infiltrating lymphocytes.

Table II.

Comparison of pCR of patients in the TIL-H group and patients in the other two groups.

Table II.

Comparison of pCR of patients in the TIL-H group and patients in the other two groups.

PatientsTIL-low and TIL-mediumTIL-Hχ2P-value
pCR     2     43.0150.082
Non-pCR3616

[i] H, high; pCR, pathological complete response; TIL, tumor-infiltrating lymphocytes.

Analysis of species differences

There were 3,174, 2,996 and 2,877 different OTUs in the TIL-H, TIL-M and TIL-L groups, respectively. The number of common and unique OTUs is shown in Fig. 1A, and Fig. 1B shows the top 10 species and their abundances. Tables III and IV illustrated that the gastrointestinal microbiome, when compared among the three groups (TIL-L vs. TIL-M vs. TIL-H) or compared between the TIL-L and TIL-H groups, exhibited significantly different β-diversities in weighted and unweighted UniFrac analyses, which suggested that low expression of TILs was associated with lower β-diversity (P<0.01). Furthermore, ANOSIM revealed a greater intergroup difference between the TIL-L and TIL-H groups compared with the intragroup difference, which indicated that the grouping was correct (P=0.042; Fig. 2).

Table III.

Weighted UniFrac distance difference analysis.

Table III.

Weighted UniFrac distance difference analysis.

GroupsTest methodP-value
TIL-L vs. TIL-Mt-test0.0004a
TIL-L vs. TIL-MWilcoxon0.0001a
TIL-L vs. TIL-Ht-test 1.0147×10−6a
TIL-L vs. TIL-HWilcoxon 1.8884×10−7a
TIL-M vs. TIL-Ht-test0.1227
TIL-M vs. TIL-HWilcoxon0.1245
TIL-L vs. TIL-M vs. TIL-HKruskal-Wallis 7.2565×10−7a
TIL-L vs. TIL-M vs. TIL-HTukey honest significant difference 7.0541×10−7a

a P≤0.01. H, high; L, low; M, medium; TIL, tumor-infiltrating lymphocytes.

Table IV.

Unweighted UniFrac distance difference analysis.

Table IV.

Unweighted UniFrac distance difference analysis.

GroupsTest methodP-value
TIL-L vs. TIL-Mt-test0.4007
TIL-L vs. TIL-MWilcoxon0.1997
TIL-L vs. TIL-Ht-test 1.2397×10−12a
TIL-L vs. TIL-HWilcoxon 7.5180×10−12a
TIL-M vs. TIL-Ht-test 4.9195×10−12a
TIL-M vs. TIL-HWilcoxon 2.9383×10−12a
TIL-L vs. TIL-M vs. TIL-HKruskal-Wallis 7.0570×10−14a
TIL-L vs. TIL-M vs. TIL-HTukey honest significant difference 1.9653×10−15a

a P≤0.01. H, high; L, low; M, medium; TIL, tumor-infiltrating lymphocytes.

Relative abundance of microbiota in TIL-H and TIL-L groups

The different distributions of microbiota in TIL-H and TIL-L groups were assessed using Metastats software. Table V demonstrated that At the genus level, patients in the TIL-L group had higher abundances of Mycobacterium, Rhodococcus, Catenibacterium, Bulleidia, Anaerofilum, Sneathia, Devosia and TG5, but lower abundances of Methanosphaera and Anaerobiospirillum compared with the TIL-H group (P<0.05). At the species level, the abundances of stercoris, barnesiae, coprophilus, flavefaciens and C21_c20 were greater in the TIL-L group, while the abundances of producta and komagatae were greater in the TIL-H group (P<0.05).

Table V.

Bacterial species exhibiting significant alterations in relative abundance between TIL-H and TIL-L groups.

Table V.

Bacterial species exhibiting significant alterations in relative abundance between TIL-H and TIL-L groups.

PhylumClassOrderFamilyGenusSpeciesMean, TIL-L (%)Mean, TIL-H (%)P-valueFDR
ActinobacteriaActinobacteria Actinomycetales Mycobacteriaceae Mycobacterium 1.35×10−50 7.97×10−60.001752485
ActinobacteriaActinobacteria ActinomycetalesNocardiaceae Rhodococcus 7.45×10−600.0008724550.01998002
ActinobacteriaCoriobacteriia Coriobacteriales Coriobacteriaceae Collinsella Stercoris 7.11×10−500.0009990010.01998002
BacteroidetesBacteroidiaBacteroidalesBacteroidaceae Bacteroides Barnesiae 9.69×10−500.0009990010.01998002
BacteroidetesBacteroidiaBacteroidalesBacteroidaceae Bacteroides Coprophilus0.012465402 5.90×10−50.0029970030.038784745
Euryarchaeota Methanobacteria Methanobacteriales Methanobacteriaceae Methanosphaera 0 5.40×10−60.0013880660.025447882
Firmicutes Erysipelotrichi Erysipelotrichales Erysipelotrichaceae Catenibacterium 7.09×10−500.0009990010.01998002
FirmicutesClostridiaClostridiales LachnospiraceaeBlautia Producta 2.71×10−50.0003546210.0009990010.01998002
Firmicutes Erysipelotrichi Erysipelotrichales Erysipelotrichaceae Bulleidia 6.88×10−600.0019083640.032295398
FirmicutesClostridiaClostridiales Ruminococcaceae Anaerofilum 1.43×10−5 2.46×10−60.0025181820.037610265
FirmicutesClostridiaClostridiales Ruminococcaceae Ruminococcus Flavefaciens 1.04×10−5 9.19×10−70.0027352920.037610265
FusobacteriaFusobacteriia Fusobacteriales Leptotrichiaceae Sneathia 2.31×10−500.0009990010.01998002
Proteobacteria AlphaproteobacteriaRhizobiales HyphomicrobiaceaeDevosia 1.05×10−50 3.81×10−50.004192394
Proteobacteria Deltaproteobacteria Desulfovibrionales Desulfovibrionaceae DesulfovibrioC21_c20 9.42×10−600.000182350.013372361
Proteobacteria GammaproteobacteriaAeromonadales Succinivibrionaceae Anaerobiospirillum 0 1.25×10−50.0009990010.01998002
Proteobacteria AlphaproteobacteriaRhizobiales Methylobacteriaceae Methylobacterium Komagatae0 4.99×10−60.002620060.037610265
SynergistetesSynergistiaSynergistales DethiosulfovibrionaceaeTG5 7.16×10−6 8.63×10−70.0004999140.01998002

[i] The difference of microbial community abundance between the two groups was tested using the Metastats software. The P-value was obtained, then the Q-value was corrected by FDR, and the significance of the difference was evaluated by screening the P-value or Q-value. P≤0.05 indicated a statistically significant difference. FDR, false discovery rate; H, high; L, low; TIL, tumor-infiltrating lymphocytes.

Discussion

Recently, increasing attention has been paid to the effects of the gastrointestinal microbiome on human diseases. Diversity of the gastrointestinal microbiome is closely associated with human health, including immunity, digestion, obesity (30), diabetes (31,32), heart disease (33,34), acquired immune deficiency syndrome (35) and cancer (36,37).

The present study demonstrated that the gastrointestinal microbiome was distinctly diverse and compositionally different among different TIL expression groups of patients with BC. Higher TIL expression was associated with a greater diversity of the gastrointestinal microbiome in the present study. A previous study only suggested that patients with BC possess statistically different microbiota composition compared with controls (38), and the gastrointestinal microbiome is associated with the clinical or biological characteristics of patients with BC (38,39). However, the present study revealed that microbiome diversity was associated with TIL distribution. Additionally, differentially expressed microbiota species among different TIL groups were identified. Among the gastrointestinal microbiome, barnesiae and coprophilus belong to the genus Bacteroides that can modulate estrogen metabolism and function as a risk factor for BC (4043); in this study, higher abundance of barnesiae was associated with the low expression of TILs., indicating barnesiae could be a risk factor for BC. The mechanism underlying barnesiae-modulated estrogen metabolism in response to immunity modification in BC remains unclear. The state of TIL expression in situ exhibits a strong association with the outcomes and treatment efficiency of patients with BC, and the gastrointestinal microbiome can regulate immune activation, following treatment with chemotherapeutic agents (14,4446). The results of the present study revealed that greater quantity and abundance of the gastrointestinal microbiome were positively associated with the expression of TILs, demonstrating an internal link between the microbiome and immunity in the pathogenesis of BC. Therefore, the treatment efficiency should be assessed in a cohort consisting of large-scale samples.

Patients with triple-negative BC and HER2-positive BC may benefit from neoadjuvant chemotherapy (47), and the data of the present study revealed that HER2 expression was positively associated with high TIL expression. Furthermore, patients with high TIL expression exhibited good outcomes following chemotherapy. All these findings implied an inherent link among microbiome, immunity and treatment efficiency in patients with BC.

The small sample size is the main limitation of the present study. Additionally, further studies regarding the mechanism need to be performed in the future. In these, a BC mouse model will be established to verify the conclusions of the present study by altering the gastrointestinal microbiome.

In conclusion, expression levels of TILs were associated with the diversity of the gastrointestinal microbiome in patients with BC. The results of the present study suggested that the gastrointestinal microbiome may affect the prognosis of patients with BC by interacting with TIL expression.

Acknowledgements

The authors would like to thank Professor Zhanjun Guo (Rheumatic Immunology Department), Dr Xi Zhang and Dr Meng Cheng (Breast Oncology Department) from The Fourth Hospital of Hebei Medical University (Shijiazhuang, China) for their help in reviewing this manuscript.

Funding

The present study was supported by grants from Hebei Science and Technology Planning (grant no. 16967788D).

Availability of data and materials

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

Authors' contributions

CG designed the experiment, provided financial support, revised the manuscript and gave final approval of the version to be published. MS was responsible for the interpretation of data. JS and WG performed the experiments, acquired the data and wrote the paper. SL, SY and ZL made substantive contributions to the work, including data collecting and manuscript revising.

Ethics approval and consent to participate

Ethical approval for this project was obtained from the Ethics Committee of The Fourth Hospital of Hebei Medical University. Informed consent was provided by all participants. All procedures involving human participants were performed in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards.

Patient consent for publication

The patients' data were anonymized, and hospital numbers and associated data may be provided only for scientific purposes. All patients provided written informed consent for their data to be published.

Competing interests

The authors declare that they have no competing interests.

References

1 

Akram M, Iqbal M, Daniyal M and Khan AU: Awareness and current knowledge of breast cancer. Biol Res. 50:332017. View Article : Google Scholar : PubMed/NCBI

2 

de la Cruz-Merino L, Chiesa M, Caballero R, Rojo F, Palazón N, Carrasco FH and Sánchez-Margalet V: Breast cancer immunology and immunotherapy: Current status and future perspectives. Int Rev Cell Mol Biol. 331:12017. View Article : Google Scholar : PubMed/NCBI

3 

Gooden MJ, de Bock GH, Leffers N, Daemen T and Nijman HW: The prognostic influence of tumour-infiltrating lymphocytes in cancer: A systematic review with meta-analysis. Br J Cancer. 105:93–103. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Mahmoud SM, Paish EC, Powe DG, Macmillan RD, Grainge MJ, Lee AH, Ellis IO and Green AR: Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Clin Oncol. 29:1949–1955. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Liu S, Foulkes WD, Leung S, Gao D, Lau S, Kos Z and Nielsen TO: Prognostic significance of FOXP3+ tumor infiltrating lymphocytes in breast cancer depends on estrogen receptor and human epidermal growth factor receptor-2 expression status and concurrent cytotoxic T-cell infiltration. Breast Cancer Res. 16:4322014. View Article : Google Scholar : PubMed/NCBI

6 

Seo AN, Lee HJ, Kim EJ, Kim HJ, Jang MH, Lee HE, Kim YJ, Kim JH and Park SY: Tumour-infiltrating CD8+ lymphocytes as an independent predictive factor for pathological complete response to primary systemic therapy in breast cancer. Br J Cancer. 109:2705–2713. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Denkert C, Loibl S, Noske A, Roller M, Muller BM, Komor M, Budczies J, Darb-Esfahani S, Kronenwett R, Hanusch C, et al: Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol. 28:105–113. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Spranger S, Sivan A, Corrales L and Gajewski TF: Tumor and host factors controlling antitumor immunity and efficacy of cancer immunotherapy. Adv Immunol. 130:75–93. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Qin Q, Miao J, Wang S, Yu Q, Li M, He F and Wang G: Association between intestinal flora and immunity in middle-aged and aged people by PCR-DGGE. Wei Sheng Yan Jiu. 46:40–45. 2017.(In Chinese). PubMed/NCBI

10 

Hooper LV, Littman DR and Macpherson AJ: Interactions between the microbiota and the immune system. Science. 336:1268–1273. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Ivanov II and Honda K: Intestinal commensal microbes as immune modulators. Cell Host Microbe. 12:496–508. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Abt MC, Osborne LC, Monticelli LA, Doering TA, Alenghat T, Sonnenberg GF, Paley MA, Antenus M, Williams KL, Erikson J, et al: Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity. 37:158–170. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Ganal SC, Sanos SL, Kallfass C, Oberle K, Johner C, Kirschning C, Lienenklaus S, Weiss S, Staeheli P, Aichele P and Diefenbach A: Priming of natural killer cells by nonmucosal mononuclear phagocytes requires instructive signals from commensal microbiota. Immunity. 37:171–186. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S, et al: Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 342:967–970. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Dingemanse C, Belzer C, van Hijum SA, Günthel M, Salvatori D, den Dunnen JT, Kuijper EJ, Devilee P, de Vos WM, van Ommen GB and Robanus-Maandag EC: Akkermansia muciniphila and Helicobacter typhlonius modulate intestinal tumor development in mice. Carcinogenesis. 36:1388–1396. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Omar Al-Hassi H, Ng O and Brookes M: Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut. 67:3952018. View Article : Google Scholar : PubMed/NCBI

17 

Peled JU, Devlin SM, Staffas A, Lumish M, Khanin R, Littmann ER, Ling L, Kosuri S, Maloy M, Slingerland JB, et al: Intestinal microbiota and relapse after hematopoietic-cell transplantation. J Clin Oncol. 35:1650–1659. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Hammond ME, Hayes DF, Dowsett M, Allred DC, Hagerty KL, Badve S, Fitzgibbons PL, Francis G, Goldstein NS, Hayes M, et al: American society of clinical oncology/college of American pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Clin Oncol. 28:2784–2795. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Wolff AC, Hammond MEH, Allison KH, Harvey BE, Mangu PB, Bartlett JMS, Bilous M, Ellis IO, Fitzgibbons P, Hanna W, et al: Human epidermal growth factor receptor 2 testing in breast cancer: American society of clinical oncology/college of American pathologists clinical practice guideline focused update. J Clin Oncol. 36:2105–2122. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Ogston KN, Miller ID, Payne S, Hutcheon AW, Sarkar TK, Smith I, Schofield A and Heys SD: A new histological grading system to assess response of breast cancers to primary chemotherapy: prognostic significance and survival. Breast. 12:320–327. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, Wienert S, Van den Eynden G, Baehner FL, Penault-Llorca F, et al: The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol. 26:259–271. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Hida AI and Ohi Y: Evaluation of tumor-infiltrating lymphocytes in breast cancer; proposal of a simpler method. Ann Oncol. 26:23512015. View Article : Google Scholar : PubMed/NCBI

23 

Magoč T and Salzberg SL: FLASH: Fast length adjustment of short reads to improve genome assemblies. Bioinformatics. 27:2957–2963. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, Fierer N, Peña AG, Goodrich JK, Gordon JI, et al: QIIME allows analysis of high-throughput community sequencing data. Nat Methods. 7:335–336. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Bokulich NA, Subramanian S, Faith JJ, Gevers D, Gordon JI, Knight R, Mills DA and Caporaso JG: Quality-filtering vastly improves diversity estimates from Illumina amplicon sequencing. Nat Methods. 10:57–59. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Edgar and Robert C: UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat Methods. 10:996–998. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Wang Q, Garrity GM, Tiedje JM and Cole JR: Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol. 73:5261–5267. 2007. View Article : Google Scholar : PubMed/NCBI

28 

DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, Huber T, Dalevi D, Hu P and Andersen GL: Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl Environ Microbiol. 72:5069–5072. 2006. View Article : Google Scholar : PubMed/NCBI

29 

White JR, Nagarajan N and Pop M: Statistical methods for detecting differentially abundant features in clinical metagenomic samples. PLoS Comput Biol. 5:e10003522009. View Article : Google Scholar : PubMed/NCBI

30 

Cotillard A, Kennedy SP, Kong LC, Prifti E, Pons N, Le Chatelier E, Almeida M, Quinquis B, Levenez F, Galleron N, et al: Dietary intervention impact on gut microbial gene richness. Nature. 500:585–588. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Larsen N, Vogensen FK, van den Berg FW, Nielsen DS, Andreasen AS, Pedersen BK, Al-Soud WA, Sørensen SJ, Hansen LH and Jakobsen M: Gut microbiota in human adults with type 2 differs from non-diabetic adults. PLoS One. 5:e908552000.

32 

Markle JG, Frank DN, Mortin-Toth S, Robertson CE, Feazel LM, Rolle-Kampczyk U, von Bergen M, McCoy KD, Macpherson AJ and Danska JS: Sex differences in the gut microbiome drive hormone-dependent regulation of autoim munity. Science. 339:1084–1088. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, et al: Intestinal microbiota metabolism of L carnitine, a nutrient in red meat promotes atherosclerosis. Nat Med. 19:576–585. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Wang T, Cai G, Qiu Y, Fei N, Zhang M, Pang X, Jia W, Cai S and Zhao L: Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 6:320–329. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Vujkovic-Cvijin I, Dunham RM, Iwai S, Maher MC, Albright RG, Broadhurst MJ, Hernandez RD, Lederman MM, Huang Y, Somsouk M, et al: Dysbiosis of the gut microbiota is associated with HIV disease progression and tryptophan catabolism. Sci Transl Med. 5:193ra912013. View Article : Google Scholar : PubMed/NCBI

36 

Fox JG, Feng Y, Theve EJ, Raczynski AR, Fiala JLA, Doernte AL, Williams M, McFaline JL, Essigmann JM, Schauer DB, et al: Gut microbes define liver cancer risk in mice exposed to chemical and viral trans genic hepatocarcinogens. Gut. 59:88–97. 2009. View Article : Google Scholar

37 

Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung YM, et al: Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 472:57–63. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Goedert JJ, Jones G, Hua X, Xu X, Yu G, Flores R, Falk RT, Gail MH, Shi J, Ravel J and Feigelson HS: Investigation of the association between the fecal microbiota and breast cancer in postmenopausal women: a population-based case-control pilot study. J Natl Cancer Inst. 107:djv1472015. View Article : Google Scholar : PubMed/NCBI

39 

Luu TH, Michel C, Bard JM, Dravet F, Nazih H and Bobin-Dubigeon C: Intestinal proportion of Blautia sp. is associated with clinical stage and histoprognostic grade in patients with early-stage breast cancer. Nutr Cancer. 69:267–275. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Fuhrman BJ, Feigelson HS, Flores R, Gail MH, Xu X, Ravel J and Goedert JJ: Associations of the fecal microbiome with urinary estrogens and estrogen metabolites in postmenopausal women. J Clin Endocrinol Metab. 99:4632–4640. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Flores R, Shi J, Fuhrman B, Xu X, Veenstra TD, Gail MH, Gajer P, Ravel J and Goedert JJ: Fecal microbial determinants of fecal and systemic estrogens and estrogen metabolites: A cross-sectional study. J Transl Med. 10:2532012. View Article : Google Scholar : PubMed/NCBI

42 

Estrogen excretion patterns and plasma levels in vegetarian and omnivorous women. Nutr Rev. 41:180–183. 1983.PubMed/NCBI

43 

Goldin BR, Adlercreutz H, Gorbach SL, Warram JH, Dwyer JT, Swenson L and Woods MN: Estrogen excretion patterns and plasma levels in vegetarian and omnivorous women. N Engl J Med. 307:1542–1547. 1982. View Article : Google Scholar : PubMed/NCBI

44 

Savas P, Salgado R, Denkert C, Sotiriou C, Darcy PK, Smyth MJ and Loi S: Clinical relevance of host immunity in breast cancer: from TILs to the clinic. Nat Rev Clin Oncol. 13:228–241. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillère R, Hannani D, Enot DP, Pfirschke C, Engblom C, Pittet MJ, et al: The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 342:971–976. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pagès C, Tosolini M, Camus M, Berger A, Wind P, et al: Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 313:1960–1964. 2006. View Article : Google Scholar : PubMed/NCBI

47 

Rastogi P, Anderson SJ, Bear HD, Geyer CE, Kahlenberg MS, Robidoux A, Margolese RG, Hoehn JL, Vogel VG, Dakhil SR, et al: Preoperative chemotherapy: Updates of national surgical adjuvant breast and bowel project protocols B-18 and B-27. J Clin Oncol. 26:778–785. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2019
Volume 17 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shi J, Geng C, Sang M, Gao W, Li S, Yang S and Li Z: Effect of gastrointestinal microbiome and its diversity on the expression of tumor‑infiltrating lymphocytes in breast cancer. Oncol Lett 17: 5050-5056, 2019
APA
Shi, J., Geng, C., Sang, M., Gao, W., Li, S., Yang, S., & Li, Z. (2019). Effect of gastrointestinal microbiome and its diversity on the expression of tumor‑infiltrating lymphocytes in breast cancer. Oncology Letters, 17, 5050-5056. https://doi.org/10.3892/ol.2019.10187
MLA
Shi, J., Geng, C., Sang, M., Gao, W., Li, S., Yang, S., Li, Z."Effect of gastrointestinal microbiome and its diversity on the expression of tumor‑infiltrating lymphocytes in breast cancer". Oncology Letters 17.6 (2019): 5050-5056.
Chicago
Shi, J., Geng, C., Sang, M., Gao, W., Li, S., Yang, S., Li, Z."Effect of gastrointestinal microbiome and its diversity on the expression of tumor‑infiltrating lymphocytes in breast cancer". Oncology Letters 17, no. 6 (2019): 5050-5056. https://doi.org/10.3892/ol.2019.10187