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
Research Article

Low-dose metronomic gemcitabine pretreatments overcome the resistance of breast cancer to immune checkpoint therapy

    Xichen Zheng‡

    *Author for correspondence: Tel.: +86 188 9651 6375;

    E-mail Address: xczheng@fudan.edu.cn

    Institute of Pediatrics, Children’s Hospital of Fudan University, Shanghai, 200000, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Jiajie Kuai‡

    Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory & Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory & Immune Medicine, Anhui Medical University, Hefei, 230000, China

    Cyrus Tang Hematology Center of Soochow University, Suzhou, 215000, China

    ‡Authors contributed equally

    Search for more papers by this author

    &
    Guanghui Shen

    Institute of Pediatrics, Children’s Hospital of Fudan University, Shanghai, 200000, China

    Published Online:https://doi.org/10.2217/imt-2022-0254

    Aims: Immunotherapy has revolutionized cancer management. However, response to immunotherapy is heterogeneous. Thus, strategies to improve antitumor immune responses in resistant tumors, such as breast cancer, are urgently needed. Methods: Established murine tumors were treated with anti-CTLA4 or anti-PD-1 alone or combined with metronomic gemcitabine (met-GEM). Tumor vascular function, immune cell tumor infiltration and gene transcription were determined. Results: Low-dose met-GEM (2 mg/kg) treatments improved tumor vessel perfusion and increased tumor-infiltrating T cells. Notably, low-dose met-GEM pretreatments converted resistant tumors to respond to immunotherapy. Moreover, combined therapy reduced tumor vessel density, improved tumor vessel perfusion, increased T-cell tumor infiltration and upregulated the expression of some anticancer genes. Conclusion: Low-dose met-GEM pretreatment reconditioned the tumor immune microenvironment and improved immunotherapy efficacy in murine breast cancer.

    Plain language summary

    Breast cancer is the most commonly diagnosed cancer in women globally. However, only a small subset of breast cancer patients benefits from treatment with immunotherapy. Thus, strategies aiming to enhance the antitumor effects of immunotherapies in breast cancer are an urgent requirement. We found that low-dose metronomic gemcitabine treatments improved tumor vessel function and increased tumor-infiltrating T cells, and did not deplete myeloid-derived suppressor cells, but did not affect tumor growth in the murine breast cancer model. Notably, however, low-dose metronomic gemcitabine pretreatments sensitized breast cancers to immunotherapy. Our findings provide important insights into the optimal strategies for combining immunotherapy with improved tumor vessel perfusion.

    Tweetable abstract

    Low-dose metronomic gemcitabine (met-GEM) improves tumor vessel perfusion and increases tumor-infiltrating T cells. Pretreatment with low-dose met-GEM sensitizes resistant breast cancers to immunotherapy.

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

    References

    • 1. Sung H, Ferlay J, Siegel RL et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71(3), 209–249 (2021).
    • 2. Woolston C. Breast cancer. Nature 527(7578), S101 (2015).
    • 3. Huang Y, Kim BYS, Chan CK, Hahn SM, Weissman IL, Jiang W. Improving immune-vascular crosstalk for cancer immunotherapy. Nat. Rev. Immunol 18(3), 195–203 (2018). • Summarizes the reciprocal regulation between tumor vascular normalization and immune reprogramming forms in tumors.
    • 4. Tumeh PC, Harview CL, Yearley JH et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515(7528), 568–571 (2014).
    • 5. Hassel JC. Ipilimumab plus nivolumab for advanced melanoma. Lancet Oncol. 17(11), 1471–1472 (2016).
    • 6. Hanna A, Balko JM. Breast cancer resistance mechanisms: challenges to immunotherapy. Breast Cancer Res. Treat. 190(1), 5–17 (2021).
    • 7. Cristescu R, Mogg R, Ayers M et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 362(6411), eaar3593 (2018).
    • 8. Wu SY, Xu Y, Chen L et al. Combined angiogenesis and PD-1 inhibition for immunomodulatory TNBC: concept exploration and biomarker analysis in the FUTURE-C-Plus trial. Mol. Cancer 21(1), 84 (2022).
    • 9. Adams S, Schmid P, Rugo HS et al. Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: cohort A of the phase II KEYNOTE-086 study. Ann. Oncol. 30(3), 397–404 (2019). •• The effiency of anti-PD-1 treatment is limited in breast cancer patients.
    • 10. Spitzer MH, Carmi Y, Reticker-Flynn NE et al. Systemic immunity is required for effective cancer immunotherapy. Cell 168(3), 487–502.e15 (2017).
    • 11. Mathieu J, Zhang Z, Zhou W et al. HIF induces human embryonic stem cell markers in cancer cells. Cancer Res. 71(13), 4640–4652 (2011).
    • 12. Folkman J. Tumor angiogenesis: therapeutic implications. N. Engl. J. Med. 285(21), 1182–1186 (1971).
    • 13. Viallard C, Larrivee B. Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis 20(4), 409–426 (2017).
    • 14. Bergers G, Song S. The role of pericytes in blood-vessel formation and maintenance. Neuro Oncol 7(4), 452–464 (2005).
    • 15. Morikawa S, Baluk P, Kaidoh T, Haskell A, Jain RK, Mcdonald DM. Abnormalities in pericytes on blood vessels and endothelial sprouts in tumors. Am. J. Pathol. 160(3), 985–1000 (2002).
    • 16. Carmeliet P, Jain RK. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat. Rev. Drug Discov. 10(6), 417–427 (2011).
    • 17. Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nat. Med. 7(9), 987–989 (2001).
    • 18. Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307(5706), 58–62 (2005). • Tumor vessel normalization may serve as a therapeutic window for combinational therapy.
    • 19. Jain RK. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell 26(5), 605–622 (2014).
    • 20. Zheng X, Fang Z, Liu X et al. Increased vessel perfusion predicts the efficacy of immune checkpoint blockade. J. Clin. Invest. 128(5), 2104–2115 (2018).
    • 21. Zhang N, Yin R, Zhou P et al. DLL1 orchestrates CD8+ T cells to induce long-term vascular normalization and tumor regression. Proc. Natl Acad. Sci. USA 118(22), e2020057118 (2021).
    • 22. Zheng X, Zhang N, Qian L et al. CTLA4 blockade promotes vessel normalization in breast tumors via the accumulation of eosinophils. Int. J. Cancer 146(6), 1730–1740 (2020).
    • 23. Li Q, Wang Y, Jia W et al. Low-dose anti-angiogenic therapy sensitizes breast cancer to PD-1 blockade. Clin. Cancer Res. 26(7), 1712–1724 (2020).
    • 24. Maiti R. Metronomic chemotherapy. J. Pharmacol. Pharmacother. 5(3), 186–192 (2014).
    • 25. Van Moorsel CJ, Peters GJ, Pinedo HM. Gemcitabine: future prospects of single-agent and combination studies. Oncologist 2(3), 127–134 (1997).
    • 26. Hanahan D, Bergers G, Bergsland E. Less is more, regularly: metronomic dosing of cytotoxic drugs can target tumor angiogenesis in mice. J. Clin. Invest. 105(8), 1045–1047 (2000).
    • 27. Kerbel RS, Kamen BA. The anti-angiogenic basis of metronomic chemotherapy. Nat. Rev. Cancer 4(6), 423–436 (2004).
    • 28. Vives M, Ginesta MM, Gracova K et al. Metronomic chemotherapy following the maximum tolerated dose is an effective anti-tumour therapy affecting angiogenesis, tumour dissemination and cancer stem cells. Int. J. Cancer 133(10), 2464–2472 (2013).
    • 29. Pasquier E, Kavallaris M, Andre N. Metronomic chemotherapy: new rationale for new directions. Nat. Rev. Clin. Oncol. 7(8), 455–465 (2010).
    • 30. Yi SY, Ruan J, Zhao L, Ke Y, Li XN. Metronomic gemcitabine targeted tumor vascular microenvironment decreases the population of CD133(+) cells in hepatocarcinoma xenografts. Cancer Biomark. 14(6), 427–433 (2014).
    • 31. Cham KK, Baker JH, Takhar KS et al. Metronomic gemcitabine suppresses tumour growth, improves perfusion, and reduces hypoxia in human pancreatic ductal adenocarcinoma. Br. J. Cancer 103(1), 52–60 (2010). •• Metronomic gemcitabine could improve tumor vessel perfusion and reduces hypoxia in a preclinical model.
    • 32. Mpekris F, Baish JW, Stylianopoulos T, Jain RK. Role of vascular normalization in benefit from metronomic chemotherapy. Proc. Natl Acad. Sci. USA 114(8), 1994–1999 (2017).
    • 33. Yapp DT, Wong MQ, Kyle AH et al. The differential effects of metronomic gemcitabine and antiangiogenic treatment in patient-derived xenografts of pancreatic cancer: treatment effects on metabolism, vascular function, cell proliferation, and tumor growth. Angiogenesis 19(2), 229–244 (2016).
    • 34. Zhang X, Wang D, Li Z et al. Low-dose gemcitabine treatment enhances immunogenicity and natural killer cell-driven tumor immunity in lung cancer. Front. Immunol. 11, 331 (2020).
    • 35. Deshmukh SK, Tyagi N, Khan MA et al. Gemcitabine treatment promotes immunosuppressive microenvironment in pancreatic tumors by supporting the infiltration, growth, and polarization of macrophages. Sci. Rep. 8(1), 12000 (2018).
    • 36. Fridman WH, Pages F, Sautes-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat. Rev. Cancer 12(4), 298–306 (2012).
    • 37. Fife BT, Bluestone JA. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol. Rev. 224, 166–182 (2008).
    • 38. Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am. J. Clin. Oncol. 39(1), 98–106 (2016).
    • 39. Schaaf MB, Garg AD, Agostinis P. Defining the role of the tumor vasculature in antitumor immunity and immunotherapy. Cell Death Dis. 9(2), 115 (2018).
    • 40. Kammertoens T, Friese C, Arina A et al. Tumour ischaemia by interferon-gamma resembles physiological blood vessel regression. Nature 545(7652), 98–102 (2017).
    • 41. Romagnani P, Lasagni L, Annunziato F, Serio M, Romagnani S. CXC chemokines: the regulatory link between inflammation and angiogenesis. Trends Immunol. 25(4), 201–209 (2004).
    • 42. Lai KC, Liu CJ, Lin TJ et al. Blocking TNF-alpha inhibits angiogenesis and growth of IFIT2-depleted metastatic oral squamous cell carcinoma cells. Cancer Lett. 370(2), 207–215 (2016).
    • 43. De Weerdt O, Van De Donk NW, Veth G, Bloem AC, Hagenbeek A, Lokhorst HM. Continuous low-dose cyclophosphamide-prednisone is effective and well tolerated in patients with advanced multiple myeloma. Neth. J. Med. 59(2), 50–56 (2001).
    • 44. Glode LM, Barqawi A, Crighton F, Crawford ED, Kerbel R. Metronomic therapy with cyclophosphamide and dexamethasone for prostate carcinoma. Cancer 98(8), 1643–1648 (2003).
    • 45. Lord R, Nair S, Schache A et al. Low dose metronomic oral cyclophosphamide for hormone resistant prostate cancer: a phase II study. J. Urol. 177(6), 2136–2140 (2007).
    • 46. Brizzi MP, Berruti A, Ferrero A et al. Continuous 5-fluorouracil infusion plus long acting octreotide in advanced well-differentiated neuroendocrine carcinomas. A phase II trial of the Piemonte oncology network. BMC Cancer 9, 388 (2009).
    • 47. Chen YP, Shen JY, Deng ZJ et al. Low-dose metronomic chemotherapy improves tumor control in nasopharyngeal carcinoma. Cancer Commun. (Lond.) 42(10), 909–912 (2022).
    • 48. Jurado JM, Sanchez A, Pajares B, Perez E, Alonso L, Alba E. Combined oral cyclophosphamide and bevacizumab in heavily pre-treated ovarian cancer. Clin. Transl. Oncol. 10(9), 583–586 (2008).
    • 49. Garcia AA, Hirte H, Fleming G et al. Phase II clinical trial of bevacizumab and low-dose metronomic oral cyclophosphamide in recurrent ovarian cancer: a trial of the California, Chicago, and Princess Margaret Hospital phase II consortia. J. Clin. Oncol. 26(1), 76–82 (2008).
    • 50. Buckstein R, Kerbel RS, Shaked Y et al. High-dose celecoxib and metronomic ‘low-dose’ cyclophosphamide is an effective and safe therapy in patients with relapsed and refractory aggressive histology non-Hodgkin’s lymphoma. Clin. Cancer Res. 12(17), 5190–5198 (2006).
    • 51. Reardon DA, Desjardins A, Vredenburgh JJ et al. Metronomic chemotherapy with daily, oral etoposide plus bevacizumab for recurrent malignant glioma: a phase II study. Br. J. Cancer 101(12), 1986–1994 (2009).
    • 52. Masuda N, Higaki K, Takano T et al. A phase II study of metronomic paclitaxel/cyclophosphamide/capecitabine followed by 5-fluorouracil/epirubicin/cyclophosphamide as preoperative chemotherapy for triple-negative or low hormone receptor expressing/HER2-negative primary breast cancer. Cancer Chemother. Pharmacol. 74(2), 229–238 (2014).
    • 53. Dellapasqua S, Mazza M, Rosa D et al. Pegylated liposomal doxorubicin in combination with low-dose metronomic cyclophosphamide as preoperative treatment for patients with locally advanced breast cancer. Breast 20(4), 319–323 (2011).
    • 54. Bottini A, Generali D, Brizzi MP et al. Randomized phase II trial of letrozole and letrozole plus low-dose metronomic oral cyclophosphamide as primary systemic treatment in elderly breast cancer patients. J. Clin. Oncol. 24(22), 3623–3628 (2006).
    • 55. Cardoso F, Paluch-Shimon S, Senkus E et al. 5th ESO–ESMO international consensus guidelines for advanced breast cancer (ABC 5). Ann. Oncol. 31(12), 1623–1649 (2020).
    • 56. Thomssen C, Luftner D, Untch M et al. International Consensus Conference for Advanced Breast Cancer, Lisbon 2019: ABC5 Consensus – assessment by a German Group of experts. Breast Care (Basel) 15(1), 82–95 (2020).
    • 57. Orlando L, Cardillo A, Ghisini R et al. Trastuzumab in combination with metronomic cyclophosphamide and methotrexate in patients with HER-2 positive metastatic breast cancer. BMC Cancer 6, 225 (2006).
    • 58. Orecchioni S, Talarico G, Labanca V, Calleri A, Mancuso P, Bertolini F. Vinorelbine, cyclophosphamide and 5-FU effects on the circulating and intratumoural landscape of immune cells improve anti-PD-L1 efficacy in preclinical models of breast cancer and lymphoma. Br. J. Cancer 118(10), 1329–1336 (2018).
    • 59. Chen Q, Xia R, Zheng W et al. Metronomic paclitaxel improves the efficacy of PD-1 monoclonal antibodies in breast cancer by transforming the tumor immune microenvironment. Am. J. Transl. Res. 12(2), 519–530 (2020).
    • 60. Emblem KE, Mouridsen K, Bjornerud A et al. Vessel architectural imaging identifies cancer patient responders to anti-angiogenic therapy. Nat. Med. 19(9), 1178–1183 (2013).
    • 61. Browder T, Butterfield CE, Kraling BM et al. Antiangiogenic scheduling of chemotherapy improves efficacy against experimental drug-resistant cancer. Cancer Res. 60(7), 1878–1886 (2000).
    • 62. Klement G, Baruchel S, Rak J et al. Continuous low-dose therapy with vinblastine and VEGF receptor-2 antibody induces sustained tumor regression without overt toxicity. J. Clin. Invest. 105(8), R15–R24 (2000).
    • 63. Bertolini F, Paul S, Mancuso P et al. Maximum tolerable dose and low-dose metronomic chemotherapy have opposite effects on the mobilization and viability of circulating endothelial progenitor cells. Cancer Res. 63(15), 4342–4346 (2003).
    • 64. Shaked Y, Emmenegger U, Man S et al. Optimal biologic dose of metronomic chemotherapy regimens is associated with maximum antiangiogenic activity. Blood 106(9), 3058–3061 (2005).
    • 65. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 100(1), 57–70 (2000).
    • 66. Carmeliet P. Angiogenesis in life, disease and medicine. Nature 438(7070), 932–936 (2005).
    • 67. Klement GL, Yip TT, Cassiola F et al. Platelets actively sequester angiogenesis regulators. Blood 113(12), 2835–2842 (2009).
    • 68. Jakobsson L, Bentley K, Gerhardt H. VEGFRs and Notch: a dynamic collaboration in vascular patterning. Biochem. Soc. Trans. 37(Pt 6), 1233–1236 (2009).
    • 69. Bocci G, Francia G, Man S, Lawler J, Kerbel RS. Thrombospondin 1, a mediator of the antiangiogenic effects of low-dose metronomic chemotherapy. Proc. Natl Acad. Sci. USA 100(22), 12917–12922 (2003).
    • 70. Rak J, Mitsuhashi Y, Sheehan C et al. Oncogenes and tumor angiogenesis: differential modes of vascular endothelial growth factor up-regulation in ras-transformed epithelial cells and fibroblasts. Cancer Res. 60(2), 490–498 (2000).
    • 71. Ikeda H, Old LJ, Schreiber RD. The roles of IFN gamma in protection against tumor development and cancer immunoediting. Cytokine Growth Factor Rev. 13(2), 95–109 (2002).
    • 72. Liang SC, Latchman YE, Buhlmann JE et al. Regulation of PD-1, PD-L1, and PD-L2 expression during normal and autoimmune responses. Eur. J. Immunol. 33(10), 2706–2716 (2003).
    • 73. Abiko K, Matsumura N, Hamanishi J et al. IFN-gamma from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer. Br. J. Cancer 112(9), 1501–1509 (2015).
    • 74. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat. Immunol. 3(11), 991–998 (2002).
    • 75. Emens LA, Middleton G. The interplay of immunotherapy and chemotherapy: harnessing potential synergies. Cancer Immunol. Res. 3(5), 436–443 (2015).
    • 76. De Cicco P, Ercolano G, Ianaro A. The New Era of Cancer Immunotherapy: Targeting Myeloid-Derived Suppressor Cells to Overcome Immune Evasion. Front Immunol.. 11, 1680 (2020).
    • 77. Gabrilovich DI. Myeloid-derived suppressor cells. Cancer Immunol. Res. 5(1), 3–8 (2017).
    • 78. Tesi RJ. MDSC; the most important cell you have never heard of. Trends Pharmacol. Sci. 40(1), 4–7 (2019).
    • 79. Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin. Cancer Res. 11(18), 6713–6721 (2005).
    • 80. Le HK, Graham L, Cha E, Morales JK, Manjili MH, Bear HD. Gemcitabine directly inhibits myeloid derived suppressor cells in BALB/c mice bearing 4T1 mammary carcinoma and augments expansion of T cells from tumor-bearing mice. Int. Immunopharmacol. 9(7-8), 900–909 (2009).
    • 81. Bucher F, Lee J, Shin S et al. Interleukin-5 suppresses vascular endothelial growth factor-induced angiogenesis through STAT5 signaling. Cytokine 110, 397–403 (2018).
    • 82. Campbell HD, Tucker WQ, Hort Y et al. Molecular cloning, nucleotide sequence, and expression of the gene encoding human eosinophil differentiation factor (interleukin 5). Proc. Natl Acad. Sci. USA 84(19), 6629–6633 (1987).
    • 83. Renninger ML, Seymour RE, Whiteley LO, Sundberg JP, Hogenesch H. Anti-IL5 decreases the number of eosinophils but not the severity of dermatitis in Sharpin-deficient mice. Exp. Dermatol. 19(3), 252–258 (2010).
    • 84. Mantovani A, Allavena P, Marchesi F, Garlanda C. Macrophages as tools and targets in cancer therapy. Nat. Rev. Drug Discov. 21(11), 799–820 (2022).
    • 85. Yang Y, Guo Z, Chen W et al. M2 Macrophage-Derived Exosomes Promote Angiogenesis and Growth of Pancreatic Ductal Adenocarcinoma by Targeting E2F2. Mol. Ther. 29(3), 1226–1238 (2021).
    • 86. Chen P, Bonaldo P. Role of macrophage polarization in tumor angiogenesis and vessel normalization: implications for new anticancer therapies. Int. Rev. Cell Mol. Biol. 301, 1–35 (2013).