Skip to main content

EDITORIAL article

Front. Oncol., 18 April 2023
Sec. Cancer Immunity and Immunotherapy
This article is part of the Research Topic Targeting Tumor Vasculature to Enhance Cancer Immunotherapy View all 9 articles

Editorial: Targeting tumor vasculature to enhance cancer immunotherapy

  • 1State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
  • 2Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and Shenzhen International Cancer Centre, Shenzhen University School of Medicine, Shenzhen, China
  • 3Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
  • 4Department of Advanced Medical Science, Kobe University Graduate School of Science, Technology and Innovation, Kobe, Japan

Background

Cancer immunotherapy is an innovative treatment for tumors at present. In 2013, Science announced tumor immunotherapy as the technological breakthrough of the year (1). In almost 10 years of clinical trials, although immunotherapy represented by PD-1 monoclonal antibody drugs has shown obvious efficacy in patients with different types of cancer, their objective response rate (ORR) has only been about 20%, which eventually leads to disease progression (Chen et al.). Thus, new approaches that improve the clinical benefits of tumor immunotherapy are urgently needed.

The sufficient infiltration of immune effector cells and immunomodulator-related molecules used in tumor immunotherapy through tumor vascularization is a prerequisite for tumor immunotherapy response (24). However, the vasculature of tumors is highly abnormal and dysfunctional. Consequently, immune effector cells have an impaired ability to penetrate into solid tumors and often exhibit compromised functions. Tumor vascular normalization, overcoming tumor endothelial cell anergy, and the blockade of immunosuppressive molecules are current efforts in the targeting of the vasculature of tumors with the aim of improving the efficacy of cancer immunotherapy.

Tumor vascular normalization

Given that the abnormal tumor vasculature is highly permeable, leaky, and tortuous with low perivascular coverage, which impairs blood flow and limits the immune cells and antibodies (5), strategies that normalize these aberrant blood vessels may therefore improve intertumoral immune cell infiltration and facilitate their antitumor activities. Multiple therapeutic strategies have been developed to normalize the tumor vasculature by tightening the endothelial cell junctions and improving pericyte coverage (6). Appropriate low-dose antiangiogenic therapy against VEGF/VEGFR was found to induce tumor vascular normalization, resulting in improved delivery of drugs and oxygen to targeted cancer cells (7). Accordingly, Fan et al. demonstrated that low-dose anlotinib can induce tumor vascular normalization and improves anti-PD-1 therapy. Zheng et al. further highlighted the recent advances of antiangiogenic immunotherapies in preclinical and clinical settings to solidify the concept that vascular normalization triggered by vasculature-targeting strategies potentiates cancer immunotherapy. Studies of murine and human tumors have identified the onset of normalization, typically 1–2 days after commencement of therapy, followed by an eventual “closure” of the normalization window (8). This opening window opportunity for designing controlled stepwise cancer cell death and immunological augmentation have been reviewed in detail by Swamy. However, these features of vascular normalization by low-dose antiangiogenic therapy strategies were eventually lost and replaced by pronounced vascular regression (9). Genetic approaches to normalization [such as promoting endothelial cell quiescence (e.g., PHD2 knockdown (10)) and enhancing vascular function (e.g., RGS5 knockdown (11))] give rise to a more prolonged normalization phenotype, in the absence of dramatic vessel regression. Li et al. established a hypoxia and angiogenesis prognostic model (HAPM) that has good predictive efficiency for PD-1 expression and T-cell exclusion, suggesting that this model may be utilized to forecast the genes for vascular normalization and the benefits of immunotherapy. In addition, Akter et al. discussed the therapeutic prospects of targeting heme and mitochondrial respiration in normalizing tumor vasculature, which provides a new theoretical basis for future research on the combination of vascular normalization and immunotherapy.

Overcoming endothelial cell anergy

Abnormal tumor vasculature can also form a condition of inflammatory signals resulting in diminished leukocyte–vessel wall interactions and, therefore, decreased inflammatory infiltration, a process referred to as “endothelial anergy” (12). This interaction is mediated by cell adhesion molecules on both leukocytes and endothelium, such as intercellular adhesion molecule-1 (ICAM-1, CD54), vascular cell adhesion molecule-1 (VCAM-1, CD106), and E-selectin (CD62E) (13, 14). Rodriguez et al. provided insight into the mechanisms regulating peripheral node addressin (PNAd) biosynthesis in tumor endothelial cells and provided another platform to enhance its expression to support a continual influx of immune cells, sustaining antitumor immunity.

Blockade of immunosuppressive molecules

Beside the endothelial cell anergy, abnormal tumor vasculature can express a range of inhibitory molecules, thereby creating a barrier for immune cells to infiltrate into the tumor tissue. Galectin 1 (15), the FAS ligand (FASL) (16), PD-L1 (17), and indoleamine 2, 3-dioxygenase(IDO) (18) were found to be selectively expressed in the vasculature of various malignancies, resulting in limited infiltration by activated T cells. Ileiwat et al. reviewed the mechanistic immunosuppressive role of the tumor vasculature and potential nanoparticle-mediated therapeutic strategies.

Conclusion

In solid tumors, blood vessels are abnormal and dysfunctional, and thus immune effector cell infiltration is impaired. Although targeting the tumor vasculature has been shown to improve the efficacy of cancer immunotherapies, recent studies suggest that enhanced immune stimulation also, in turn, improves tumor vascular normalization (19, 20). A more comprehensive understanding of the crosstalk between the immune system and tumor vasculature can provide new strategies for treating human cancers.

Author contributions

TX and JY drafted the manuscript. LF and TS revised the manuscript. All authors approved the submission.

Funding

This work was supported by grants from the National Natural Science Foundation of China (82172795).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Couzin-Frankel J. Breakthrough of the year 2013. Cancer Immunother Sci (2013) 342:1432–3. doi: 10.1126/science.342.6165.1432

CrossRef Full Text | Google Scholar

2. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature (2014) 515:568–71. doi: 10.1038/nature13954

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Tang H, Wang Y, Chlewicki LK, Zhang Y, Guo J, Liang W, et al. Facilitating T cell infiltration in tumor microenvironment overcomes resistance to PD-L1 blockade. Cancer Cell (2016) 29:285–96. doi: 10.1016/j.ccell.2016.02.004

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Joyce JA, Fearon DT. T Cell exclusion, immune privilege, and the tumor microenvironment. Science (2015) 348:74–80. doi: 10.1126/science.aaa6204

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Liu K, Zhang X, Xu W, Chen J, Yu J, Gamble JR, et al. Targeting the vasculature in hepatocellular carcinoma treatment: starving versus normalizing blood supply. Clin Transl Gastroenterol (2017) 8:e98. doi: 10.1038/ctg.2017.28

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Martin JD, Seano G, Jain RK. Normalizing function of tumor vessels: progress, opportunities, and challenges. Annu Rev Physiol (2019) 81:505–34. doi: 10.1146/annurev-physiol-020518-114700

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science (2005) 307:58–62. doi: 10.1126/science.1104819

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Goel S, Duda DG, Xu L, Munn LL, Boucher Y, Fukumura D, et al. Normalization of the vasculature for treatment of cancer and other diseases. Physiol Rev (2011) 91:1071–121. doi: 10.1152/physrev.00038.2010

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Fukumura D, Duda DG, Munn LL, Jain RK. Tumor microvasculature and microenvironment: novel insights through intravital imaging in pre-clinical models. Microcirculation (2010) 17:206–25. doi: 10.1111/j.1549-8719.2010.00029.x

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Mazzone M, Dettori D, de Oliveira RL, Loges S, Schmidt T, Jonckx B, et al. Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits metastasis via endothelial normalization. Cell (2009) 136:839–51. doi: 10.1016/j.cell.2009.01.020

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Hamzah J, Jugold M, Kiessling F, Rigby P, Manzur M, Marti HH, et al. Vascular normalization in Rgs5-deficient tumours promotes immune destruction. Nature (2008) 453:410–4. doi: 10.1038/nature06868

PubMed Abstract | CrossRef Full Text | Google Scholar

12. De Sanctis F, Ugel S, Facciponte J, Facciabene A. The dark side of tumor-associated endothelial cells. Semin Immunol (2018) 35:35–47. doi: 10.1016/j.smim.2018.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Weishaupt C, Steinert M, Brunner G, Schulze HJ, Fuhlbrigge RC, Goerge T, et al. Activation of human vascular endothelium in melanoma metastases induces ICAM-1 and e-selectin expression and results in increased infiltration with effector lymphocytes. Exp Dermatol (2019) 28:1258–69. doi: 10.1111/exd.14023

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Griffioen AW, Damen CA, Blijham GH, Groenewegen G. Tumor angiogenesis is accompanied by a decreased inflammatory response of tumor-associated endothelium. Blood (1996) 88:667–73. doi: 10.1182/blood.V88.2.667.bloodjournal882667

PubMed Abstract | CrossRef Full Text | Google Scholar

15. He J, Baum LG. Endothelial cell expression of galectin-1 induced by prostate cancer cells inhibits T-cell transendothelial migration. Lab Invest (2006) 86:578–90. doi: 10.1038/labinvest.3700420

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Motz GT, Santoro SP, Wang LP, Garrabrant T, Lastra RR, Hagemann IS, et al. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat Med (2014) 20:607–15. doi: 10.1038/nm.3541

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Rodig N, Ryan T, Allen JA, Pang H, Grabie N, Chernova T, et al. Endothelial expression of PD-L1 and PD-L2 down-regulates CD8+ T cell activation and cytolysis. Eur J Immunol (2003) 33:3117–26. doi: 10.1002/eji.200324270

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Riesenberg R, Weiler C, Spring O, Eder M, Buchner A, Popp T, et al. Expression of indoleamine 2,3-dioxygenase in tumor endothelial cells correlates with long-term survival of patients with renal cell carcinoma. Clin Cancer Res (2007) 13:6993–7002. doi: 10.1158/1078-0432.CCR-07-0942

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Tian L, Goldstein A, Wang H, Ching Lo H, Sun Kim I, Welte T, et al. Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming. Nature (2017) 544:250–4. doi: 10.1038/nature21724

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Zheng X, Fang Z, Liu X, Deng S, Zhou P, Wang X, et al. Increased vessel perfusion predicts the efficacy of immune checkpoint blockade. J Clin Invest (2018) 128:2104–15. doi: 10.1172/JCI96582

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: cancer immunotherapy, tumor vascularization, vascular normalization, endothelial cell anergy, immunosuppressive molecules

Citation: Yang J, Fu L, Shirakawa T and Xiang T (2023) Editorial: Targeting tumor vasculature to enhance cancer immunotherapy. Front. Oncol. 13:1199811. doi: 10.3389/fonc.2023.1199811

Received: 04 April 2023; Accepted: 10 April 2023;
Published: 18 April 2023.

Edited and Reviewed by:

Catherine Sautes-Fridman, INSERM U1138 Centre de Recherche des Cordeliers (CRC), France

Copyright © 2023 Yang, Fu, Shirakawa and Xiang. 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.

*Correspondence: Tong Xiang, xiangtong@sysucc.org.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.