We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Short CommunicationOpen Accesscc iconby iconnc iconnd icon

The effect of low-dose chemotherapy on the tumor microenvironment and its antitumor activity combined with anti-PD-1 antibody

    Fangyu Lin

    Department of Oncology, Fujian Medical University Union Hospital, 29 Xin-Quan Road, Fuzhou, 350001, People's Republic of China

    ,
    Hao Chen

    Department of Oncology, Fujian Medical University Union Hospital, 29 Xin-Quan Road, Fuzhou, 350001, People's Republic of China

    ,
    Tao Jiang

    Department of Oncology, Fujian Medical University Union Hospital, 29 Xin-Quan Road, Fuzhou, 350001, People's Republic of China

    ,
    Jianwei Zheng

    Department of Oncology, Fujian Medical University Union Hospital, 29 Xin-Quan Road, Fuzhou, 350001, People's Republic of China

    ,
    Qin Liu

    Department of Oncology, Fujian Medical University Union Hospital, 29 Xin-Quan Road, Fuzhou, 350001, People's Republic of China

    ,
    Baoyu Yang

    Department of Oncology, Fujian Medical University Union Hospital, 29 Xin-Quan Road, Fuzhou, 350001, People's Republic of China

    ,
    Xinli Wang

    Department of Oncology, Fujian Medical University Union Hospital, 29 Xin-Quan Road, Fuzhou, 350001, People's Republic of China

    &
    Xiaoyan Lin

    *Author for correspondence: Tel.: +86 0591 8337 3933;

    E-mail Address: xiaoyanlin@yahoo.com

    Department of Oncology, Fujian Medical University Union Hospital, 29 Xin-Quan Road, Fuzhou, 350001, People's Republic of China

    Published Online:https://doi.org/10.2217/imt-2021-0018

    Aim: This study aimed to explore the effects of low-dose chemotherapy in the tumor microenvironment (TME) on a gastric cancer xenograft and its antitumor activity combined with the anti-PD-1 antibody. Materials & methods: Mice with gastric cancer were divided into four groups. The body weight and tumor volume of the mice were recorded. The TME was analyzed using flow cytometry. Results: Low-dose paclitaxel increased the PD-L1 expression level and the number of CD8+ T cells, but not the CD4+ T and myeloid-derived suppressor cells or PD-1+ CD8+ T cells in the TME. Low-dose 5-fluorouracil reduced the number of myeloid-derived suppressor cells and PD-1+ CD8+ T cells, but the PD-L1 expression level and the number of CD4+ T and CD8+ T cells did not change in the TME. The anti-PD-1 antibody inhibited tumor growth, but the combination therapy did not show superior antitumor activity. Conclusion: Low-dose chemotherapy altered the TME but failed to improve the responses to the anti-PD-1 antibody.

    Plain language summary

    The anti-PD-1 antibody shows potential as an anticancer therapy for tumors, including gastric cancer. However, the antitumor effect of the anti-PD-1 antibody alone is unsatisfactory. The tumor microenvironment (TME) is an environment in which a tumor develops and survives. The TME comprises heterogeneous molecules and cell types, including immune cells, endothelial cells and fibroblasts, besides cancer cells. This study aimed to explore the effects of low-dose chemotherapy on the TME and its antitumor effect when combined with anti-PD-1 antibody. The TME was analyzed using the flow cytometry method. Although low-dose paclitaxel and low-dose 5-fluorouracil changed the TME, both failed to enhance the antitumor activity when combined with the anti-PD-1 antibody.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Sitarz R, Skierucha M, Mielko J, Offerhaus GJA, Maciejewski R, Polkowski WP. Gastric cancer: epidemiology, prevention, classification, and treatment. Cancer Manag. Res. 10, 239–248 (2018).
    • 2. Curiel TJ, Wei S, Dong H et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat. Med. 9(5), 562–567 (2003). •• B7-H1 on monocyte-derived myeloid dendritic cells regulates T-cell immunity.
    • 3. Patsoukis N, Sari D, Boussiotis VA. PD-1 inhibits T cell proliferation by upregulating p27 and p15 and suppressing Cdc25A. Cell Cycle 11(23), 4305–4309 (2012).
    • 4. Kareva I, Waxman DJ, Lakka Klement G. Metronomic chemotherapy: an attractive alternative to maximum tolerated dose therapy that can activate anti-tumor immunity and minimize therapeutic resistance. Cancer Lett. 358(2), 100–106 (2015). •• The paper presents evidence supporting a mechanistic explanation for the improved activity of metronomic chemotherapy.
    • 5. Cardillo I, Spugnini EP, Galluzzo P et al. Functional and pharmacodynamic evaluation of metronomic cyclophosphamide and docetaxel regimen in castration-resistant prostate cancer. Future Oncol. 9(9), 1375–1388 (2013).
    • 6. Rozados VR, Mainetti LE, Rico MJ, Zacarías Fluck MF, Matar P, Scharovsky OG. The immune response and the therapeutic effect of metronomic chemotherapy with cyclophosphamide. Oncol. Res. 18(11–12), 601–605 (2009). •• Modulation of the immune response would be involved in the antitumor effect of metronomic chemotherapy with cyclophosphamide.
    • 7. Shevchenko I, Karakhanova S, Soltek S et al. Low-dose gemcitabine depletes regulatory T cells and improves survival in the orthotopic Panc02 model of pancreatic cancer. Int. J. Cancer 133(1), 98–107 (2013).
    • 8. Kang YK, Boku N, Satoh T et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 390(10111), 2461–2471 (2017). • Patients with advanced gastric or gastroesophageal junction cancer might benefit from nivolumab.
    • 9. Bang YJ, Ruiz EY, Van Cutsem E et al. Phase III, randomised trial of avelumab versus physician's choice of chemotherapy as third-line treatment of patients with advanced gastric or gastro-oesophageal junction cancer: primary analysis of JAVELIN Gastric 300. Ann. Oncol. 29(10), 2052–2060 (2018).
    • 10. Bang YJ, Kang YK, Catenacci DV et al. Pembrolizumab alone or in combination with chemotherapy as first-line therapy for patients with advanced gastric or gastroesophageal junction adenocarcinoma: results from the phase II nonrandomized KEYNOTE-059 study. Gastric Cancer 22(4), 828–837 (2019).
    • 11. Boku N, Ryu MH, Kato K et al. Safety and efficacy of nivolumab in combination with S-1/capecitabine plus oxaliplatin in patients with previously untreated, unresectable, advanced, or recurrent gastric/gastroesophageal junction cancer: interim results of a randomized, phase II trial (ATTRACTION-4). Ann. Oncol. 30(2), 250–258 (2019). • Nivolumab combined with chemotherapy was well tolerated and demonstrated encouraging efficacy for unresectable advanced or recurrent HER2-negative gastric/gastroesophogeal junction cancer.
    • 12. Shitara K, Van Cutsem E, Bang YJ et al. Efficacy and safety of pembrolizumab or pembrolizumab plus chemotherapy vs chemotherapy alone for patients with first-line, advanced gastric cancer: The KEYNOTE-062 phase 3 randomized clinical trial. JAMA Oncol. 6(10), 1571–1580 (2020). •• Pembrolizumab or pembrolizumab plus chemotherapy was not superior to chemotherapy for the overall survival and progression-free survival end points tested among patients with untreated, advanced gastric/gastroesophogeal junction cancer.
    • 13. Catenacci DV, Wainberg Z, Fuchs CS et al. LBA-009KEYNOTE-059 cohort 3: safety and efficacy of pembrolizumab monotherapy for first-line treatment of patients (pts) with PD-L1-positive advanced gastric/gastroesophageal (G/GEJ) cancer. Ann. Oncol. 28(Suppl. 3), mdx302.008 (2017).
    • 14. Teng MW, Ngiow SF, Ribas A, Smyth MJ. Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res. 75(11), 2139–2145 (2015). •• Classification of tumors into four groups on the basis of their PD-L1 status and the presence or absence of T-cell infiltration might help define optimal therapy for patients with advanced cancers.
    • 15. Hegde PS, Karanikas V, Evers S. The where, the when, and the how of immune monitoring for cancer immunotherapies in the era of checkpoint inhibition. Clin. Cancer Res. 22(8), 1865–1874 (2016).
    • 16. Longley DB, Harkin DP, Johnston PG. 5-Fluorouracil: mechanisms of action and clinical strategies. Nat. Rev. Cancer 3(5), 330–338 (2003).
    • 17. Backus HH, Dukers DF, Van Groeningen CJ et al. 5-Fluorouracil induced Fas upregulation associated with apoptosis in liver metastases of colorectal cancer patients. Ann. Oncol. 12(2), 209–216 (2001).
    • 18. Zhu L, Chen L. Progress in research on paclitaxel and tumor immunotherapy. Cell. Mol. Biol. Lett. 24, 40 (2019).
    • 19. Husain Z, Huang Y, Seth P, Sukhatme VP. Tumor-derived lactate modifies antitumor immune response: effect on myeloid-derived suppressor cells and NK cells. J. Immunol. 191(3), 1486–1495 (2013).
    • 20. Talmadge JE, Gabrilovich DI. History of myeloid-derived suppressor cells. Nat. Rev. Cancer 13(10), 739 (2013).
    • 21. Sevko A, Kremer V, Falk C et al. Application of paclitaxel in low non-cytotoxic doses supports vaccination with melanoma antigens in normal mice. J. Immunotoxicol. 9(3), 275–281 (2012).
    • 22. Hartgrink HH, Jansen EP, Van Grieken NC, Van De Velde CJ. Gastric cancer. Lancet 374(9688), 477–490 (2009).
    • 23. Yang L, Debusk LM, Fukuda K et al. Expansion of myeloid immune suppressor Gr+ CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell 6(4), 409–421 (2004).
    • 24. Yu P-C, Long D, Liao C-C, Zhang S. Association between density of tumor-infiltrating lymphocytes and prognoses of patients with gastric cancer. Medicine 97(27), e11387 (2018).
    • 25. Badoual C, Hans S, Merillon N et al. PD-1-expressing tumor-infiltrating T cells are a favorable prognostic biomarker in HPV-associated head and neck cancer. Cancer Res. 73(1), 128–138 (2013).
    • 26. Li J, Shayan G, Avery L et al. Tumor-infiltrating Tim-3+ T cells proliferate avidly except when PD-1 is co-expressed: evidence for intracellular cross talk. Oncoimmunology 5(10), e1200778 (2016).
    • 27. Li J, Jie H-B, Lei Y et al. PD-1/SHP-2 inhibits Tc1/Th1 phenotypic responses and the activation of T cells in the tumor microenvironment. Cancer Res. 75(3), 508–518 (2015).
    • 28. Shen T, Zhou L, Shen H et al. Prognostic value of programmed cell death protein 1 expression on CD8+ T lymphocytes in pancreatic cancer. Sci. Rep. 7(1), 7848 (2017).
    • 29. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 9(3), 162–174 (2009). • This paper discusses the origin, mechanisms of expansion and suppressive functions of myeloid-derived suppressor cells.