Cargando…

Metagenomic Analyses Reveal Distinct Gut Microbiota Signature for Predicting the Neoadjuvant Chemotherapy Responsiveness in Breast Cancer Patients

BACKGROUND: Growing evidence supports the modulatory role of human gut microbiome on neoadjuvant chemotherapy (NAC) efficacy. However, the relationships among the gut microbiome, tumor-infiltrating lymphocytes (TILs), and NAC response for breast cancer (BC) patients remain unclear. We thus proposed...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Yuanyuan, Dong, Bingbin, Wu, Wei, Wang, Jiawei, Jin, Hao, Chen, Kangmei, Huang, Kangling, Huang, Songyin, Yao, Yandan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9010823/
https://www.ncbi.nlm.nih.gov/pubmed/35433455
http://dx.doi.org/10.3389/fonc.2022.865121
_version_ 1784687567363375104
author Li, Yuanyuan
Dong, Bingbin
Wu, Wei
Wang, Jiawei
Jin, Hao
Chen, Kangmei
Huang, Kangling
Huang, Songyin
Yao, Yandan
author_facet Li, Yuanyuan
Dong, Bingbin
Wu, Wei
Wang, Jiawei
Jin, Hao
Chen, Kangmei
Huang, Kangling
Huang, Songyin
Yao, Yandan
author_sort Li, Yuanyuan
collection PubMed
description BACKGROUND: Growing evidence supports the modulatory role of human gut microbiome on neoadjuvant chemotherapy (NAC) efficacy. However, the relationships among the gut microbiome, tumor-infiltrating lymphocytes (TILs), and NAC response for breast cancer (BC) patients remain unclear. We thus proposed this preliminary study to investigate the relationship between gut microbiome and BC patients’ responses to NAC treatment as well as underlying mechanisms. METHODS: Prior to receiving NAC, the fecal metagenome collected from 23 patients with invasive BC was analyzed. Patients were subsequently assigned to the NAC non-effectual group and the NAC effectual group based on their response to NAC. The peripheral T lymphocyte subset counts were examined by flow cytometry methods. CellMinor analysis was employed to explore the relationship between CD4 mRNA expression and the reaction of tumor cells to NAC drugs. RESULTS: The gut microbiomes of the NAC non-effectual group showed characteristics of low diversity with low abundances, distinct metagenomic composition with decreased butyrate-producing and indolepropionic acid-producing bacteria, and increased potential pathobionts compared with the NAC effectual group. The combination of Coprococcus, Dorea, and uncultured Ruminococcus sp. serves as signature bacteria for distinguishing NAC non-effectual group patients from the NAC effectual group. The absolute numbers of CD4(+) and CD8(+) TIL infiltration in tumors in the NAC non-effectual group were significantly lower than those in the effectual group. Similar findings were reported for the CD4(+) T lymphocytes in the peripheral blood (p’s < 0.05). NAC effectual-related signature bacteria were proportional to these patients’ CD4(+) T lymphocyte counts in peripheral blood and tumors (p’s < 0.05). CellMinor analysis showed that the CD4 mRNA expression level dramatically climbed with increased sensitivity of tumor cells to NAC drugs such as cyclophosphamide, cisplatin, and carboplatin (p’s < 0.05). CONCLUSIONS: The composition of the gut microbial community differs between BC patients for whom NAC is effective to those that are treatment resistant. The modulation of the gut microbiota on host CD4(+) T lymphocytes may be one critical mechanism underlying chemosensitivity and NAC pathologic response. Taken together, gut microbiota may serve as a potential biomarker for NAC response, which sheds light on novel intervention targets in the treatment of NAC non-effectual BC patients.
format Online
Article
Text
id pubmed-9010823
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-90108232022-04-16 Metagenomic Analyses Reveal Distinct Gut Microbiota Signature for Predicting the Neoadjuvant Chemotherapy Responsiveness in Breast Cancer Patients Li, Yuanyuan Dong, Bingbin Wu, Wei Wang, Jiawei Jin, Hao Chen, Kangmei Huang, Kangling Huang, Songyin Yao, Yandan Front Oncol Oncology BACKGROUND: Growing evidence supports the modulatory role of human gut microbiome on neoadjuvant chemotherapy (NAC) efficacy. However, the relationships among the gut microbiome, tumor-infiltrating lymphocytes (TILs), and NAC response for breast cancer (BC) patients remain unclear. We thus proposed this preliminary study to investigate the relationship between gut microbiome and BC patients’ responses to NAC treatment as well as underlying mechanisms. METHODS: Prior to receiving NAC, the fecal metagenome collected from 23 patients with invasive BC was analyzed. Patients were subsequently assigned to the NAC non-effectual group and the NAC effectual group based on their response to NAC. The peripheral T lymphocyte subset counts were examined by flow cytometry methods. CellMinor analysis was employed to explore the relationship between CD4 mRNA expression and the reaction of tumor cells to NAC drugs. RESULTS: The gut microbiomes of the NAC non-effectual group showed characteristics of low diversity with low abundances, distinct metagenomic composition with decreased butyrate-producing and indolepropionic acid-producing bacteria, and increased potential pathobionts compared with the NAC effectual group. The combination of Coprococcus, Dorea, and uncultured Ruminococcus sp. serves as signature bacteria for distinguishing NAC non-effectual group patients from the NAC effectual group. The absolute numbers of CD4(+) and CD8(+) TIL infiltration in tumors in the NAC non-effectual group were significantly lower than those in the effectual group. Similar findings were reported for the CD4(+) T lymphocytes in the peripheral blood (p’s < 0.05). NAC effectual-related signature bacteria were proportional to these patients’ CD4(+) T lymphocyte counts in peripheral blood and tumors (p’s < 0.05). CellMinor analysis showed that the CD4 mRNA expression level dramatically climbed with increased sensitivity of tumor cells to NAC drugs such as cyclophosphamide, cisplatin, and carboplatin (p’s < 0.05). CONCLUSIONS: The composition of the gut microbial community differs between BC patients for whom NAC is effective to those that are treatment resistant. The modulation of the gut microbiota on host CD4(+) T lymphocytes may be one critical mechanism underlying chemosensitivity and NAC pathologic response. Taken together, gut microbiota may serve as a potential biomarker for NAC response, which sheds light on novel intervention targets in the treatment of NAC non-effectual BC patients. Frontiers Media S.A. 2022-04-01 /pmc/articles/PMC9010823/ /pubmed/35433455 http://dx.doi.org/10.3389/fonc.2022.865121 Text en Copyright © 2022 Li, Dong, Wu, Wang, Jin, Chen, Huang, Huang and Yao https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Oncology
Li, Yuanyuan
Dong, Bingbin
Wu, Wei
Wang, Jiawei
Jin, Hao
Chen, Kangmei
Huang, Kangling
Huang, Songyin
Yao, Yandan
Metagenomic Analyses Reveal Distinct Gut Microbiota Signature for Predicting the Neoadjuvant Chemotherapy Responsiveness in Breast Cancer Patients
title Metagenomic Analyses Reveal Distinct Gut Microbiota Signature for Predicting the Neoadjuvant Chemotherapy Responsiveness in Breast Cancer Patients
title_full Metagenomic Analyses Reveal Distinct Gut Microbiota Signature for Predicting the Neoadjuvant Chemotherapy Responsiveness in Breast Cancer Patients
title_fullStr Metagenomic Analyses Reveal Distinct Gut Microbiota Signature for Predicting the Neoadjuvant Chemotherapy Responsiveness in Breast Cancer Patients
title_full_unstemmed Metagenomic Analyses Reveal Distinct Gut Microbiota Signature for Predicting the Neoadjuvant Chemotherapy Responsiveness in Breast Cancer Patients
title_short Metagenomic Analyses Reveal Distinct Gut Microbiota Signature for Predicting the Neoadjuvant Chemotherapy Responsiveness in Breast Cancer Patients
title_sort metagenomic analyses reveal distinct gut microbiota signature for predicting the neoadjuvant chemotherapy responsiveness in breast cancer patients
topic Oncology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9010823/
https://www.ncbi.nlm.nih.gov/pubmed/35433455
http://dx.doi.org/10.3389/fonc.2022.865121
work_keys_str_mv AT liyuanyuan metagenomicanalysesrevealdistinctgutmicrobiotasignatureforpredictingtheneoadjuvantchemotherapyresponsivenessinbreastcancerpatients
AT dongbingbin metagenomicanalysesrevealdistinctgutmicrobiotasignatureforpredictingtheneoadjuvantchemotherapyresponsivenessinbreastcancerpatients
AT wuwei metagenomicanalysesrevealdistinctgutmicrobiotasignatureforpredictingtheneoadjuvantchemotherapyresponsivenessinbreastcancerpatients
AT wangjiawei metagenomicanalysesrevealdistinctgutmicrobiotasignatureforpredictingtheneoadjuvantchemotherapyresponsivenessinbreastcancerpatients
AT jinhao metagenomicanalysesrevealdistinctgutmicrobiotasignatureforpredictingtheneoadjuvantchemotherapyresponsivenessinbreastcancerpatients
AT chenkangmei metagenomicanalysesrevealdistinctgutmicrobiotasignatureforpredictingtheneoadjuvantchemotherapyresponsivenessinbreastcancerpatients
AT huangkangling metagenomicanalysesrevealdistinctgutmicrobiotasignatureforpredictingtheneoadjuvantchemotherapyresponsivenessinbreastcancerpatients
AT huangsongyin metagenomicanalysesrevealdistinctgutmicrobiotasignatureforpredictingtheneoadjuvantchemotherapyresponsivenessinbreastcancerpatients
AT yaoyandan metagenomicanalysesrevealdistinctgutmicrobiotasignatureforpredictingtheneoadjuvantchemotherapyresponsivenessinbreastcancerpatients