Combination of anti-angiogenic therapy Apatinib and immune therapy potentiate tumor microenvironment

Chunyi Gao, Tianhui Hu

Article ID: 1376
Vol 5, Issue 2.1, 2021

VIEWS - 42 (Abstract) 32 (PDF)

Abstract


Tumor immune therapy, especially anti-programmed cell death ligand-1/programmed cell death-1 (PD-L1/PD-1) treatment, is currently the focus of substantial attention. However, despite its enormous successes, the overall response rate of cancer immunotherapy remains suboptimal. There is an increased interest in combining PD-L1/PD-1 treatment with anti-angiogenic drug Apatinib to enhance antitumor effect. Presently available data seem to suggest that Apatinib may exert immune suppressive effects to make the PD-L1/PD-1 treatment works. Here, we review the extensive tumor microenvironment immune modulatory effects from antiangiogenic agents Apatinib in order to supporting VEGFR2 targettherapies in clinical trials are existing.


Keywords


Apatinib; Tumor Environment; PD-1/PD-L1; Tumor Immune Therapy

Full Text:

PDF


References


1. Hoeksema MA, Gijbels MJ, Velden S, et al. Targeting macrophage Histone deacetylase 3 stabilizes atherosclerotic lesions. EMBO Molecular Medicine 2014; 6(9): 1124–1132.

2. Zhao L, Zhang W, Li C, et al. Research progress on antitumor mechanism of Apatinib. Cancer Research on Prevention and Treatment 2021; 48(1): 7–11.

3. Li Y, Zhao H, Ren X. Relationship of VEGF/VEGFR with immune and cancer cells: Staggering or forward? Cancer Biology & Medicine 2016; 13(2): 206–214.

4. Ragunathrao VAB, Anwar M, Akhter MZ, et al. Sphingosine-1-phosphate receptor 1 activity promotes tumor growth by amplifying VEGF-VEGFR2 angiogenic signaling. Cell Reports 2019; 29(11): 3472–3487.

5. Maroufin F, Rashidi MR, Vahedian V, et al. Therapeutic potentials of Apatinib in cancer treatment: Possible mechanisms and clinical relevance. Life Sciences 2020; 241: 117106.

6. Mi Y, Liang Y, Huang H, et al. Apatinib (YN968D1) reverses multidrug resistance by inhibiting the efflux function of multiple ATP-binding cassette transporters. Cancer Research 2010; 70(20): 7981–7991.

7. Xu J, Zhang Y, Jia R, et al. Anti-PD-1 antibody SHR-1210 combined with Apatinib for advanced hepatocellular carcinoma, gastric, or esophagogastric junction cancer: An open-label, dose escalation and expansion study. Clinical Cancer Research 2019; 25(2): 515–523.

8. Zheng Y, Yang X, Yan C, et al. Effect of Apatinib plus neoadjuvant chemotherapy followed by resection on pathologic response in patients with locally advanced gastric adenocarcinoma: A single-arm, open-label, phase II trial. European Journal of Cancer 2020; 130: 12–19.

9. Liu C, Jia Q, Wei H, et al. Apatinib in patients with advanced chordoma: A single-arm, single-centre, phase 2 study. Lancet Oncology 2020; 21(9): 1244–1252.

10. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science 2018; 359(6382): 1350–1355.

11. Lyon AR, Yousaf N, Battisti NML, et al. Immune checkpoint inhibitors and cardiovascular toxicity. Lancet Oncology 2018; 19(9): e447–e458.

12. Wei SC, Aang NAS, Sharma R, et al. Combination anti-CTLA-4 plus anti-PD-1 checkpoint blockade utilizes cellular mechanisms partially distinct from monotherapies. Proceedings of the National Academy of Sciences 2019; 116(45): 22699–22709.

13. Wang DY, Salem JE, Cohen JV, et al. Fatal toxic effects associated with immune checkpoint inhibitors: A systematic review and meta-analysis. JAMA Oncology 2018; 4(12): 1721–1728.

14. Wei F, Wu Y, Tang L, et al. BPIFB1 (LPLUNC1) inhibits migration and invasion of nasopharyngeal carcinoma by interacting with VTN and VIM. British Journal of Cancer 2018; 118(2): 233–247.

15. Vitale I, Manic G, Coussens LM, et al. Macrophages and metabolism in the tumor microenvironment. Cell Metabolism 2019; 30(1): 36–50.

16. Shemesh CS, Hsu JC, Hosseini I, et al. Personalized cancer vaccines: Clinical landscape, challenges and opportunities. Molecular Therapy 2021; 29(2): 555–570.

17. McDermott DF, Huseni MA, Atkins MB, et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nature Medicine 2018; 24(6): 749–757.

18. Wallin JJ, Bendell JC, Funke R, et al. Atezolizumab in combination with bevacizumab enhances anti-gen-specific T-cell migration in metastatic renal cell Carcinoma. Nature Communications 2016; 7: 12624.

19. Allen E, Jabouille A, Rivera LB, et al. Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Science Translational Medicine 2017; 9(385): eaak9679.

20. Tada Y, Togashi Y, Kotani D, et al. Targeting VEGFR2 with Ramucirumab strongly impacts effector/activated regulatory T cells and CD8+ T cells in the tumor microenvironment. Journal for Immunotherapy of Cancer 2018; 6(1): 106.

21. Almeida PED, Mak J, Hernandez G, et al. Anti-VEGF treatment enhances CD8+ T-cell antitumor activity by amplifying hypoxia. Cancer Immunol Research 2020; 8(6): 806–818.

22. Zhao S, Ren S, Jiang T, et al. Low-dose Apatinib optimizes tumor microenvironment and potentiates antitumor effect of PD-1/PD-L1 blockade in lung cancer. Cancer Immunol Research 2019; 7(4): 630–643.

23. Xu Y, Zhang X, Wang Y, et al. A VEGFR2-MICA bispecific antibody activates tumor-infiltrating lymphocytes and exhibits potent anti-tumor efficacy in mice. Cancer Immunology, Immunotherapy 2019; 68(9): 1429–1441.

24. Li Q, Wang Y, Jia W, et al. Low-dose Anti-angio-genic therapy sensitizes breast cancer to PD-1 blockade. Clinical Cancer Research 2020; 26(7): 1712–1724.

25. Huang Y, Yuan J, Righi E, et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proceedings of the National Academy of Sciences of the United States of America 2012; 109(43): 17561–17566.

26. Horikawa N, Abiko K, Matsumura N, et al. Expression of vascular endothelial growth factor in ovarian cancer inhibits tumor immunity through the accumulation of myeloid-derived suppressor cells. Clinical Cancer Research 2017; 23(2): 587–599.

27. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science 2003; 299(5609): 1057–1061.

28. Suzuki H, Onishi H, Wada J, et al. VEGFR2 is selectively expressed by FOXP3high CD4+ Treg. European Journal of Immunology 2010; 40(1): 197–203.

29. Terme M, Pernot S, Marcheteau E, et al. VEGFA-VEGFR pathway blockade inhibits tumor-induced regulatory T-cell proliferation in colorectal cancer. Cancer Research 2013; 73(2): 539–549.

30. Yasuda S, Sho M, Yamato I, et al. Simultaneous blockade of programmed death 1 and vascular endothelial growth factor receptor 2 (VEGFR2) induces synergistic anti-tumour effect in vivo. Clinical & Experimental Immunology 2013; 172(3): 500–506.

31. Shigeta K, Datta M, Hato T, et al. Dual programmed death receptor-1 and vascular endothelial growth factor receptor-2 blockade promotes vascular vormalization and enhances antitumor immune responses in hepatocellular carcinoma. Hepatology 2020; 71(4): 1247–1261.

32. Gao C, Jiang Z, Wang G. Mechanism of macrophage polarization in breast cancer associated microenvironment. Chinese Clinical Oncology 2019; 24(3): 274–280.

33. Hato T, Zhu AX, Duda DG. Rationally combining anti-VEGF therapy with checkpoint inhibitors in hepatocellular carcinoma. Immunotherapy 2016; 8(3): 299–313.

34. Alfaro C, Suarez N, Gonzalez A, et al. Influence of bevacizumab, sunitinib and sorafenib as single agents or in combination on the inhibitory effects of VEGF on human dendritic cell differentiation from monocytes. British Journal of Cancer 2009; 100(7): 1111–1119.

35. Dikov MM, Ohm JE, Ray N, et al. Differential roles of vascular endothelial growth factor receptors 1and 2 in dendritic cell differentiation. Journal of Immunology 2005; 174(1): 215–222.

36. Mimura K, Kono K, Takahashi A, et al. Vascular endothelial growth factor inhibits the function of human mature dendritic cells mediated by VEGF receptor-2. Cancer Immunol Immunother 2007; 56(6): 761–770.

37. Cavalheiro ML, Lorena P, Patricia S, et al. Vascular endothelial growth factor-A enhances indoleamine 2, 3-dioxygenase expression by dendritic cells and subsequently impacts lymphocyte proliferation. Memorias Do Instituto Oswaldo Cruz 2014; 109(1): 7079.

38. Ilkovitch D, Lopez DM. The liver is a site for tumor-induced myeloid-derived suppressor cell accumulation and immunosuppression. Cancer Research 2009; 69(13): 5514–5521.

39. Hoechst B, Voigtlaender T, Ormandy L, et al. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology 2009; 50(3): 799–807.

40. Solito S, Falisi E, Diaz-Montero CM, et al. A human promyelocytic-like population is responsible for the immune suppression mediated by myeloid-derived suppressor cells. Blood 2011; 118(8): 2254–2265.

41. Wu F, Zhang S, Xiong A, et al. A Phase II clinical trial of Apatinib in pretreated advanced non-squamous non-small-cell lung cancer. Clinical Lung Cancer 2018; 19(6): e831–e842.

42. Xu J, Shen J, Gu S, et al. Camrelizumab in combination with Apatinib in patients with advanced hepatocellular carcinoma (RESCUE): A nonrandomized, open-label, phase II trial. Clinical Cancer Research 2021; 27(4): 1003–1011.

43. Li J, Qin S, Xu J, et al. Randomized, double-blind, placebo-controlled phase III trial of Apatinib in patients with chemotherapy-refractory advanced or metastatic adenocarcinoma of the stomach or gastroesophageal junction. Journal of Clinical Oncology 2016; 34(13): 1448–1454.

44. Liu J, Liu Q, Li Y, et al. Efficacy and safety of camrelizumab combined with Apatinib in advanced triple-negative breast cancer: An open-label phase II trial. Journal for Immuno Therapy of Cancer 2020; 8(1): e000696.

45. Takahashi Y, Kitadai Y, Bucana CD, et al. Expression of vascular endothelial growth factor and its receptor, KDR, correlates with vascularity, metastasis, and proliferation of human colon cancer. Cancer Research 1995; 55(18): 3964–3968.

46. Park JS, Kim IK, Han S, et al. Normalization of tumor vessels by Tie2 activation and Ang2 inhibition enhances drug delivery and produces a favorable tumor microenvironment. Cancer Cell 2016; 30(6): 953–967.

47. Kim Y, Nam HJ, Lee J, et al. Methylation-dependent regulation of HIF-1α stability restricts retinal and tumour angiogenesis. Nature Communications 2016; 7: 10347.

48. Mulligan JK, Rosenzweig SA, Young MRI. Tumor secretion of VEGF induces endothelial cells to suppress T cell functions through the production of PGE2. Journal of Immunotherapy 2010; 33(2): 126–135.

49. Jayaraman P, Parikh F, Lopez-Rivera E, et al. Tumor-expressed inducible nitric oxide synthase controls induction of functional myeloid-derived suppressor cells through modulation of vascular endothelial growth factor release. Journal of Immunology 2012; 188(11): 5365–5376.

50. Hansen W, Hutzler M, Abel S, et al. Neuropilin 1 deficiency on CD4+ Foxp3+ regulatory T cells impairs mouse melanoma growth. The Journal of Experimental Medicine 2012; 209(11): 2001–2016.




DOI: https://doi.org/10.24294/ti.v5.i2.1.1376

Refbacks

  • There are currently no refbacks.


Copyright (c) 2021 Chunyi Gao, Tianhui Hu

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

This site is licensed under a Creative Commons Attribution 4.0 International License.