REFERENCES
1. Yang M, Zhang C, Wang R, Wu X, Li H, Yoon J. Cancer immunotherapy elicited by immunogenic cell death based on smart nanomaterials. Small Methods. 2023;7:2201381.
2. Petiti J, Arpinati L, Menga A, Carrà G. The influence of fatty acid metabolism on T cell function in lung cancer. FEBS J. 2025;292:3596-615.
3. Feng Y, Tang Q, Wang B, et al. Targeting the tumor microenvironment with biomaterials for enhanced immunotherapeutic efficacy. J Nanobiotechnol. 2024;22:3005.
4. Sadeghi M, Moslehi A, Kheiry H, et al. The sensitivity of acute myeloid leukemia cells to cytarabine is increased by suppressing the expression of Heme oxygenase-1 and hypoxia-inducible factor 1-alpha. Cancer Cell Int. 2024;24:3393.
5. Song CW, Kim H, Kim M, et al. Role of HIF-1α in the responses of tumors to radiotherapy and chemotherapy. Cancer Res Treat. 2025;57:1-10.
6. Qi H, Ma X, Ma Y, Jia L, Liu K, Wang H. Mechanisms of HIF1A-mediated immune evasion in gastric cancer and the impact on therapy resistance. Cell Biol Toxicol. 2024;40:9917.
7. Xun Z, Zhou H, Shen M, et al. Identification of hypoxia-ALCAMhigh macrophage- exhausted T cell axis in tumor microenvironment remodeling for immunotherapy resistance. Adv Sci. 2024;11:e2309885.
8. Lu Y, Sun Q, Guan Q, et al. The XOR-IDH3α axis controls macrophage polarization in hepatocellular carcinoma. J Hepatol. 2023;79:1172-84.
9. Shi S, Ou X, Liu C, Wen H, Ke J. Research progress of HIF-1a on immunotherapy outcomes in immune vascular microenvironment. Front Immunol. 2025;16:1549276.
10. Rodríguez F, Caruana P, De la Fuente N, et al. Nano-based approved pharmaceuticals for cancer treatment: present and future challenges. Biomolecules. 2022;12:784.
11. Meng X, Shen Y, Zhao H, Lu X, Wang Z, Zhao Y. Redox-manipulating nanocarriers for anticancer drug delivery: a systematic review. J Nanobiotechnol. 2024;22:2859.
12. Zhang J, Zhang A, Guo Y, et al. Nanoparticle-mediated cuproptosis and photodynamic synergistic strategy: a novel horizon for cancer therapy. Cancer Med. 2025;14:e70599.
13. Suvac A, Ashton J, Bristow RG. Tumour hypoxia in driving genomic instability and tumour evolution. Nat Rev Cancer. 2025;25:167-88.
14. Cheng X, Li H, Ge X, et al. Tumor-microenvironment- responsive size-shrinkable drug-delivery nanosystems for deepened penetration into tumors. Front Mol Biosci. 2020;7:576420.
15. Bhattarai D, Xu X, Lee K. Hypoxia-inducible factor-1 (HIF-1) inhibitors from the last decade (2007 to 2016): a “structure–activity relationship” perspective. Med Res Rev. 2018;38:1404-42.
16. Akanji MA, Rotimi D, Adeyemi OS. Hypoxia-inducible factors as an alternative source of treatment strategy for cancer. Oxid Med Cell Longev. 2019;2019:8547846.
17. Yan Y, Li H, Yao H, Cheng X. Nanodelivery systems delivering hypoxia-inducible factor-1 alpha short interfering RNA and antisense oligonucleotide for cancer treatment. Front Nanotechnol. 2022;4:932976.
18. Hatanaka M, Shimba S, Sakaue M, et al. Hypoxia-inducible factor-3 alpha functions as an accelerator of 3T3-L1 adipose differentiation. Biol Pharm Bull. 2009;32:1166-72.
19. Mandl M, Lieberum M, Depping R. A HIF-1α-driven feed-forward loop augments HIF signalling in Hep3B cells by upregulation of ARNT. Cell Death Dis. 2016;7:e2284.
20. Ju C, Colgan SP, Eltzschig HK. Hypoxia-inducible factors as molecular targets for liver diseases. J Mol Med. 2016;94:613-27.
21. Lee JW, Bae SH, Jeong JW, Kim SH, Kim KW. Hypoxia-inducible factor (HIF-1)α: its protein stability and biological functions. Exp Mol Med. 2004;36:1-12.
22. Bharadwaj LA, Davies GF, Xavier IJ, Ovsenek N. l-carnosine and verapamil inhibit hypoxia-induced expression of hypoxia inducible factor (HIF-1 α) in H9c2 cardiomyoblasts. Pharmacol Res. 2002;45:175-81.
23. Appelhoff RJ, Tian Y, Raval RR, et al. Differential function of the prolyl hydroxylases PHD1, PHD2, and PHD3 in the regulation of hypoxia-inducible factor. J Biol Chem. 2004;279:38458-65.
24. Berra E. HIF prolyl-hydroxylase 2 is the key oxygen sensor setting low steady-state levels of HIF-1alpha in normoxia. EMBO J. 2003;22:4082-90.
25. Chua YL, Dufour E, Dassa EP, et al. Stabilization of hypoxia-inducible factor-1α protein in hypoxia occurs independently of mitochondrial reactive oxygen species production. J Biol Chem. 2010;285:31277-84.
26. Pereira T, Zheng X, Poellinger L. Degradation of the hypoxia-inducible factor 1α, where does it happen? Cell Cycle. 2006;5:2720-2.
27. Peng X, Gao H, Xu R, Wang H, Mei J, Liu C. The interplay between HIF-1α and noncoding RNAs in cancer. J Exp Clin Cancer Res. 2020;39:1535.
28. Kanno H, Matsumoto S, Yoshizumi T, et al. Role of SOCS and VHL proteins in neuronal differentiation and development. IJMS. 2023;24:3880.
29. Choudhry H, Harris AL. Advances in hypoxia-inducible factor biology. Cell Metab. 2018;27:281-98.
30. Smolarz B, Łukasiewicz H, Samulak D, et al. Hypoxia-induced factor-1α and its role in endometrial cancer. Anticancer Res. 2024;44:3697-712.
31. Seymour L, Nuru N, Johnson KR, et al. Roles of post-translational modifications of transcription factors involved in breast cancer hypoxia. Molecules. 2025;30:645.
32. Zhao Z, Mu H, Li Y, Liu Y, Zou J, Zhu Y. Clinicopathological and prognostic value of hypoxia-inducible factor-1α in breast cancer: a meta-analysis including 5177 patients. Clin Transl Oncol. 2020;22:1892-906.
33. Zhang C, Yang K, Yang G. Design strategies for enhancing antitumor efficacy through tumor microenvironment exploitation using albumin-based nanosystems: a review. Int J Biol Macromol. 2024;258:129070.
34. Yang M, Li J, Gu P, Fan X. The application of nanoparticles in cancer immunotherapy: targeting tumor microenvironment. Bioact Mater. 2021;6:1973-87.
35. Liu J, Jiang Y, Chen L, Qian Z, Zhang Y. Associations between HIFs and tumor immune checkpoints: mechanism and therapy. Discov Onc. 2024;15:836.
36. Famta P, Shah S, Vambhurkar G, et al. Amelioration of breast cancer therapies through normalization of tumor vessels and microenvironment: paradigm shift to improve drug perfusion and nanocarrier permeation. Drug Deliv Transl Res. 2025;15:389-406.
37. Saha P, Ettel P, Weichhart T. Leveraging macrophage metabolism for anticancer therapy: opportunities and pitfalls. Trends Pharmacol Sci. 2024;45:335-49.
38. Yuan Q, Jia L, Yang J, Li W. The role of macrophages in liver metastasis: mechanisms and therapeutic prospects. Front Immunol. 2025;16:1542197.
39. Zhang C, Yin X, Hao L, et al. Integrin-targeted, activatable nanophototherapeutics for immune modulation: enhancing photoimmunotherapy efficacy in prostate cancer through macrophage reprogramming. Aggregate. 2025;6:e70001.
40. Vitale I, Manic G, Coussens LM, Kroemer G, Galluzzi L. Macrophages and metabolism in the tumor microenvironment. Cell Metabol. 2019;30:36-50.
41. He Z, Zhang S. Tumor-associated macrophages and their functional transformation in the hypoxic tumor microenvironment. Front Immunol. 2021;12:741305.
42. Murdoch C, Giannoudis A, Lewis CE. Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood. 2004;104:2224-34.
43. Pyonteck SM, Akkari L, Schuhmacher AJ, et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med. 2013;19:1264-72.
44. Tripathi C, Tewari BN, Kanchan RK, et al. Macrophages are recruited to hypoxic tumor areas and acquire a pro-angiogenic M2-polarized phenotype via hypoxic cancer cell derived cytokines Oncostatin M and Eotaxin. Oncotarget. 2014;5:5350-68.
45. Ke X, Chen C, Song Y, et al. Hypoxia modifies the polarization of macrophages and their inflammatory microenvironment, and inhibits malignant behavior in cancer cells. Oncol Lett. 2019;18:5871-8.
46. Pillai SR, Damaghi M, Marunaka Y, Spugnini EP, Fais S, Gillies RJ. Causes, consequences, and therapy of tumors acidosis. Cancer Metastasis Rev. 2019;38:205-22.
47. Colegio OR, Chu N, Szabo AL, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. 2014;513:559-63.
48. Zhao Y, Wang D, Xu T, et al. Bladder cancer cells re-educate TAMs through lactate shuttling in the microfluidic cancer microenvironment. Oncotarget. 2015;6:39196-210.
49. Paolini L, Adam C, Beauvillain C, et al. Lactic acidosis together with GM-CSF and M-CSF induces human macrophages toward an inflammatory protumor phenotype. Cancer Immunol Res. 2020;8:383-95.
50. Zhao Y, Zhao B, Wang X, et al. Macrophage transcriptome modification induced by hypoxia and lactate. Exp Ther Med. 2019;18:4811-9.
51. Zhang L, Li S. Lactic acid promotes macrophage polarization through MCT-HIF1α signaling in gastric cancer. Exp Cell Res. 2020;388:111846.
52. Bohn T, Rapp S, Luther N, et al. Tumor immunoevasion via acidosis-dependent induction of regulatory tumor-associated macrophages. Nat Immunol. 2018;19:1319-29.
53. Chen P, Zuo H, Xiong H, et al. Gpr132 sensing of lactate mediates tumor–macrophage interplay to promote breast cancer metastasis. Proc Natl Acad Sci U S A. 2017;114:580-5.
54. Zhang D, Tang Z, Huang H, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019;574:575-80.
55. Wu JY, Huang TW, Hsieh YT, et al. Cancer-derived succinate promotes macrophage polarization and cancer metastasis via succinate receptor. Mol Cell. 2020;77:213-27.e5.
56. Cheng S, Wu C, Tsai Y, et al. CXCR4 antagonist-loaded nanoparticles reprogram the tumor microenvironment and enhance immunotherapy in hepatocellular carcinoma. J Control Release. 2025;379:967-81.
57. Jiang M, Li X, Zhang J, et al. Dual inhibition of endoplasmic reticulum stress and oxidation stress manipulates the polarization of macrophages under hypoxia to sensitize immunotherapy. ACS Nano. 2021;15:14522-34.
58. Fu Y, Zhang Y, Zhang Y, et al. Nanoreactors with cascade catalytic activity reprogram the tumor microenvironment for enhanced immunotherapy by synchronously regulating Treg and macrophage cells. ACS Appl Mater Interfaces. 2024;16:49053-68.
59. Wang Y, Fan Y, Zhang X, et al. In situ production and precise release of bioactive GM-CSF and siRNA by engineered bacteria for macrophage reprogramming in cancer immunotherapy. Biomaterials. 2025;317:123037.
60. Huang R, Kang T, Chen S. The role of tumor-associated macrophages in tumor immune evasion. J Cancer Res Clin Oncol. 2024;150:5777.
61. Medvedeva GF, Kuzmina DO, Nuzhina J, Shtil AA, Dukhinova MS. How macrophages become transcriptionally dysregulated: a hidden impact of antitumor therapy. Int J Mol Sci. 2021;22:2662.
62. Chen Y, Song Y, Du W, Gong L, Chang H, Zou Z. Tumor-associated macrophages: an accomplice in solid tumor progression. J Biomed Sci. 2019;26:78.
63. Li M, He L, Zhu J, Zhang P, Liang S. Targeting tumor-associated macrophages for cancer treatment. Cell Biosci. 2022;12:823.
64. Xu B, Sun H, Song X, Liu Q, Jin W. Mapping the tumor microenvironment in TNBC and deep exploration for M1 macrophages-associated prognostic genes. Front Immunol. 2022;13:923481.
65. Xu Y, Wang X, Liu L, Wang J, Wu J, Sun C. Role of macrophages in tumor progression and therapy (Review). Int J Oncol. 2022;60:57.
66. Xu Y, Zeng H, Jin K, et al. Immunosuppressive tumor-associated macrophages expressing interlukin-10 conferred poor prognosis and therapeutic vulnerability in patients with muscle-invasive bladder cancer. J Immunother Cancer. 2022;10:e003416.
67. Xue VW, Chung JY, Córdoba CAG, et al. Transforming growth factor-β: a multifunctional regulator of cancer immunity. Cancers. 2020;12:3099.
68. Viola A, Munari F, Sánchez-Rodríguez R, Scolaro T, Castegna A. The metabolic signature of macrophage responses. Front Immunol. 2019;10:1462.
69. Xiao L, Wang Q, Peng H. Tumor-associated macrophages: new insights on their metabolic regulation and their influence in cancer immunotherapy. Front Immunol. 2023;14:1157291.
70. Bied M, Ho WW, Ginhoux F, Blériot C. Roles of macrophages in tumor development: a spatiotemporal perspective. Cell Mol Immunol. 2023;20:983-92.
71. Wu Q, Zhou W, Yin S, et al. Blocking triggering receptor expressed on myeloid cells-1-positive tumor-associated macrophages induced by hypoxia reverses immunosuppression and anti-programmed cell death ligand 1 resistance in liver cancer. Hepatology. 2019;70:198-214.
72. Lubitz GS, Brody JD. Not just neighbours: positive feedback between tumour-associated macrophages and exhausted T cells. Nat Rev Immunol. 2022;22:3.
73. Kersten K, Hu KH, Combes AJ, et al. Spatiotemporal co-dependency between macrophages and exhausted CD8+ T cells in cancer. Cancer Cell. 2022;40:624-38.e9.
74. Nixon BG, Kuo F, Ji L, et al. Tumor-associated macrophages expressing the transcription factor IRF8 promote T cell exhaustion in cancer. Immunity. 2022;55:2044-58.e5.
75. Nagaraj S, Gupta K, Pisarev V, et al. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat Med. 2007;13:828-35.
76. Li J, Wang L, Chen X, et al. CD39/CD73 upregulation on myeloid-derived suppressor cells via TGF-β-mTOR-HIF-1 signaling in patients with non-small cell lung cancer. OncoImmunology. 2017;6:e1320011.
77. Hoskin D, Mader J, Furlong S, Conrad D, Blay J. Inhibition of T cell and natural killer cell function by adenosine and its contribution to immune evasion by tumor cells (Review). Int J Oncol. 2008;32:527-35.
78. Xu S, Wang C, Yang L, et al. Targeting immune checkpoints on tumor-associated macrophages in tumor immunotherapy. Front Immunol. 2023;14:1199631.
79. Patsoukis N, Brown J, Petkova V, Liu F, Li L, Boussiotis VA. Selective effects of PD-1 on Akt and Ras pathways regulate molecular components of the cell cycle and inhibit T cell proliferation. Sci Signal. 2012;5:ra46.
80. Barkal AA, Brewer RE, Markovic M, et al. CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy. Nature. 2019;572:392-6.
81. Zeng S, Wang J, Kang H, Li H, Peng X, Yoon J. Photon-driven dye induction pyroptosis: an emerging anti-tumor immunotherapy paradigm. Angew Chem Int Ed Engl. 2025;64:e202417899.
82. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15:486-99.
83. Reina-Campos M, Scharping NE, Goldrath AW. CD8+ T cell metabolism in infection and cancer. Nat Rev Immunol. 2021;21:718-38.
84. Wu H, Zhao X, Hochrein SM, et al. Mitochondrial dysfunction promotes the transition of precursor to terminally exhausted T cells through HIF-1α-mediated glycolytic reprogramming. Nat Commun. 2023;14:6858.
85. Bensaad K, Favaro E, Lewis CA, et al. Fatty acid uptake and lipid storage induced by HIF-1α contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep. 2014;9:349-65.
86. Patsoukis N, Bardhan K, Chatterjee P, et al. PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat Commun. 2015;6:6692.
87. Bengsch B, Johnson AL, Kurachi M, et al. Bioenergetic insufficiencies due to metabolic alterations regulated by the inhibitory receptor PD-1 are an early driver of CD8+ T cell exhaustion. Immunity. 2016;45:358-73.
88. Wouters BG, Koritzinsky M. Hypoxia signalling through mTOR and the unfolded protein response in cancer. Nat Rev Cancer. 2008;8:851-64.
89. He J, Zhou Y, Sun L. Emerging mechanisms of the unfolded protein response in therapeutic resistance: from chemotherapy to Immunotherapy. Cell Commun Signal. 2024;22:89.
90. Ma S, Zhao Y, Lee WC, et al. Hypoxia induces HIF1α-dependent epigenetic vulnerability in triple negative breast cancer to confer immune effector dysfunction and resistance to anti-PD-1 immunotherapy. Nat Commun. 2022;13:4118.
91. Mao C, Deng F, Zhu W, et al. In situ editing of tumour cell membranes induces aggregation and capture of PD-L1 membrane proteins for enhanced cancer immunotherapy. Nat Commun. 2024;15:9723.
92. Xue F, Ren X, Kong C, et al. Polymeric PD1/PDL1 bispecific antibody enhances immune checkpoint blockade therapy. Mater Today Bio. 2024;28:101239.
93. Bigos KJ, Quiles CG, Lunj S, et al. Tumour response to hypoxia: understanding the hypoxic tumour microenvironment to improve treatment outcome in solid tumours. Front Oncol. 2024;14:1331355.
94. Jiang X, Wang J, Deng X, et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol Cancer. 2019;18:10.
95. Noman MZ, Desantis G, Janji B, et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 2014;211:781-90.
96. Suresh S, Chen B, Zhu J, et al. eIF5B drives integrated stress response-dependent translation of PD-L1 in lung cancer. Nat Cancer. 2020;1:533-45.
97. Ding XC, Wang LL, Zhang XD, et al. The relationship between expression of PD-L1 and HIF-1α in glioma cells under hypoxia. J Hematol Oncol. 2021;14:92.
98. You L, Wu W, Wang X, et al. The role of hypoxia-inducible factor 1 in tumor immune evasion. Med Res Rev. 2021;41:1622-43.
99. Alves CC, Donadi EA, Giuliatti S. Structural characterization of the interaction of hypoxia inducible factor-1 with its hypoxia responsive element at the -964G > a variation site of the HLA-G promoter region. Int J Mol Sci. 2021;22:13046.
100. Wang S, Wang J, Xia Y, et al. Harnessing the potential of HLA-G in cancer therapy: advances, challenges, and prospects. J Transl Med. 2024;22:130.
101. Labiano S, Palazón A, Bolaños E, et al. Hypoxia-induced soluble CD137 in malignant cells blocks CD137L-costimulation as an immune escape mechanism. Oncoimmunology. 2016;5:e1062967.
102. Hakimi AA, Attalla K, DiNatale RG, et al. A pan-cancer analysis of PBAF complex mutations and their association with immunotherapy response. Nat Commun. 2020;11:4168.
103. Wu F, Sun G, Nai Y, Shi X, Ma Y, Cao H. NUP43 promotes PD-L1/nPD-L1/PD-L1 feedback loop via TM4SF1/JAK/STAT3 pathway in colorectal cancer progression and metastatsis. Cell Death Discov. 2024;10:241.
104. Lee D, Cho M, Kim E, Seo Y, Cha JH. PD-L1: from cancer immunotherapy to therapeutic implications in multiple disorders. Mol Ther. 2024;32:4235-55.
105. Shi S, Ou X, Liu C, Li R, Zheng Q, Hu L. NF-κB signaling and the tumor microenvironment in osteosarcoma: implications for immune evasion and therapeutic resistance. Front Immunol. 2025;16:1518664.
106. Wen Q, Han T, Wang Z, Jiang S. Role and mechanism of programmed death-ligand 1 in hypoxia-induced liver cancer immune escape. Oncol Lett. 2020;19:2595-601.
107. Mortezaee K, Majidpoor J. Transforming growth factor-β signalling in tumour resistance to the anti-PD-(L)1 therapy: updated. J Cell Mol Med. 2023;27:311-21.
108. Ho JJD, Balukoff NC, Cervantes G, Malcolm PD, Krieger JR, Lee S. Oxygen-sensitive remodeling of central carbon metabolism by archaic eIF5B. Cell Rep. 2018;22:17-26.
109. Yu A, Fu L, Jing L, et al. Methionine-driven YTHDF1 expression facilitates bladder cancer progression by attenuating RIG-I-modulated immune responses and enhancing the eIF5B-PD-L1 axis. Cell Death Differ. 2025;32:776-91.
110. Palazón A, Martínez-Forero I, Teijeira A, et al. The HIF-1α hypoxia response in tumor-infiltrating T lymphocytes induces functional CD137 (4-1BB) for immunotherapy. Cancer Discov. 2012;2:608-23.
111. Walter Jackson Iii, Yang Y, Salman S, et al. Pharmacologic HIF stabilization activates costimulatory receptor expression to increase antitumor efficacy of adoptive T cell therapy. Sci Adv. 2024;10:eadq2366.
112. Jiang Z, Fang Z, Hong D, Wang X. Cancer immunotherapy with “vascular-immune” crosstalk as entry point: associated mechanisms, therapeutic drugs and nano-delivery systems. Int J Nanomedicine. 2024;19:7383-98.
113. Mattila P, Majuri ML, Mattila PS, Renkonen R. TNF alpha-induced expression of endothelial adhesion molecules, ICAM-1 and VCAM-1, is linked to protein kinase C activation. Scand J Immunol. 1992;36:159-65.
114. Xia P, Gamble JR, Rye KA, et al. Tumor necrosis factor-alpha induces adhesion molecule expression through the sphingosine kinase pathway. Proc Natl Acad Sci U S A. 1998;95:14196-201.
115. Liu B, Gao J, Lyu BC, et al. Expressions of TGF-β2, bFGF and ICAM-1 in lens epithelial cells of complicated cataract with silicone oil tamponade. Int J Ophthalmol. 2017;10:1034-9.
116. Motz GT, Santoro SP, Wang LP, et al. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat Med. 2014;20:607-15.
117. Norling LV, Sampaio AL, Cooper D, Perretti M. Inhibitory control of endothelial galectin-1 on in vitro and in vivo lymphocyte trafficking. FASEB J. 2008;22:682-90.
118. Buckanovich RJ, Facciabene A, Kim S, et al. Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy. Nat Med. 2008;14:28-36.
119. Tan J, Fan W, Liu T, et al. TREM2+ macrophages suppress CD8+ T-cell infiltration after transarterial chemoembolisation in hepatocellular carcinoma. J Hepatol. 2023;79:126-40.
120. Engin AB, Engin A. Indoleamine 2,3-dioxygenase activity-induced acceleration of tumor growth, and protein kinases-related novel therapeutics regimens. In: Engin AB, Engin A, editors. Protein kinase-mediated decisions between life and death. Cham: Springer International Publishing; 2021. pp. 339-56.
121. Herbert A, Ng H, Jessup W, et al. Hypoxia regulates the production and activity of glucose transporter-1 and indoleamine 2,3-dioxygenase in monocyte-derived endothelial-like cells: possible relevance to infantile haemangioma pathogenesis. Br J Dermatol. 2011;164:308-15.
122. Lee WS, Yang H, Chon HJ, Kim C. Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity. Exp Mol Med. 2020;52:1475-85.
123. Groth C, Hu X, Weber R, et al. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br J Cancer. 2019;120:16-25.
124. Khan O, Giles JR, McDonald S, et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature. 2019;571:211-8.
125. Kim CG, Jang M, Kim Y, et al. VEGF-A drives TOX-dependent T cell exhaustion in anti-PD-1-resistant microsatellite stable colorectal cancers. Sci Immunol. 2019;4:eaay0555.
126. Khan KA, Kerbel RS. Improving immunotherapy outcomes with anti-angiogenic treatments and vice versa. Nat Rev Clin Oncol. 2018;15:310-24.
127. Goumans MJ, Liu Z, ten Dijke P. TGF-beta signaling in vascular biology and dysfunction. Cell Res. 2009;19:116-27.
128. Tian M, Neil JR, Schiemann WP. Transforming growth factor-β and the hallmarks of cancer. Cell Signal. 2011;23:951-62.
129. Falco S. The discovery of placenta growth factor and its biological activity. Exp Mol Med. 2012;44:1-9.
130. Fukumura D, Kloepper J, Amoozgar Z, Duda DG, Jain RK. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat Rev Clin Oncol. 2018;15:325-40.
131. Huang Y, Kim BYS, Chan CK, Hahn SM, Weissman IL, Jiang W. Improving immune-vascular crosstalk for cancer immunotherapy. Nat Rev Immunol. 2018;18:195-203.
132. Jiang M, Qin B, Luo L, et al. A clinically acceptable strategy for sensitizing anti-PD-1 treatment by hypoxia relief. J Control Release. 2021;335:408-19.
133. Chen A, Yang F, Kuang J, et al. A versatile nanoplatform for broad-spectrum immunotherapy by reversing the tumor microenvironment. ACS Appl Mater Interfaces. 2021;13:45335-45.
134. Zhen X, Li Y, Yuan W, et al. Biointerface-engineered hybrid nanovesicles for targeted reprogramming of tumor microenvironment. Adv Mater. 2024;36:e2401495.
135. Wu J, Wang X, Chen L, et al. Oxygen microcapsules improve immune checkpoint blockade by ameliorating hypoxia condition in pancreatic ductal adenocarcinoma. Bioact Mater. 2023;20:259-70.
136. Feng X, Chen Z, Liu Z, Fu X, Song H, Zhang Q. Self-delivery photodynamic-hypoxia alleviating nanomedicine synergizes with anti-PD-L1 for cancer immunotherapy. Int J Pharm. 2023;639:122970.
137. Fang T, Cao X, Wang L, Chen M, Deng Y, Chen G. Bioresponsive and immunotherapeutic nanomaterials to remodel tumor microenvironment for enhanced immune checkpoint blockade. Bioact Mater. 2024;32:530-42.
138. Zhu L, Li J, Guo Z, Kwok HF, Zhao Q. Synergistic combination of targeted nano-nuclear-reactors and anti-PD-L1 nanobodies evokes persistent T cell immune activation for cancer immunotherapy. J Nanobiotechnology. 2022;20:521.
139. Zhang X, Bai J, Sun S, et al. Chiral nanoassembly remodels tumor microenvironment through non-oxygen-dependent depletion lactate for effective photodynamic immunotherapy. Biomaterials. 2025;319:123203.
140. He M, Zhang M, Xu T, et al. Enhancing photodynamic immunotherapy by reprograming the immunosuppressive tumor microenvironment with hypoxia relief. J Control Release. 2024;368:233-50.
141. Chang CC, Dinh TK, Lee YA, et al. Nanoparticle delivery of MnO2 and antiangiogenic therapy to overcome hypoxia-driven tumor escape and suppress hepatocellular carcinoma. ACS Appl Mater Interfaces. 2020;12:44407-19.
142. Bao Y, Li G, Li S, et al. Multifunctional tumor-targeting carbon dots for tumor microenvironment activated ferroptosis and immunotherapy in cancer treatment. ACS Appl Mater Interfaces. 2023;15:56834-45.
143. Sun K, Yu J, Hu J, et al. Salicylic acid-based hypoxia-responsive chemodynamic nanomedicines boost antitumor immunotherapy by modulating immunosuppressive tumor microenvironment. Acta Biomater. 2022;148:230-43.
144. Gong J, Cheng D, Liu C, et al. Hybrid cell membrane-coated nanoparticles for synergizing sonodynamic therapy and immunotherapy against triple-negative breast cancer. Adv Healthc Mater. 2025;14:e2404184.
145. Wang Y, Yu J, Luo Z, et al. Engineering endogenous tumor-associated macrophage-targeted biomimetic nano-RBC to reprogram tumor immunosuppressive microenvironment for enhanced chemo-immunotherapy. Adv Mater. 2021;33:e2103497.
146. Cao Y, Qiao B, Chen Q, et al. Tumor microenvironment remodeling via targeted depletion of M2-like tumor-associated macrophages for cancer immunotherapy. Acta Biomater. 2023;160:239-51.
147. Liu Y, Qiu N, Shen L, et al. Nanocarrier-mediated immunogenic chemotherapy for triple negative breast cancer. J Control Release. 2020;323:431-41.
148. Jiang T, Wang B, Wang T, Zhang L, Chen X, Zhao X. TAM-hijacked immunoreaction rescued by hypoxia-pathway-intervened strategy for enhanced metastatic cancer immunotherapy. Small. 2024;20:e2305728.
149. Zhao Q, He X, Qin X, et al. Enhanced therapeutic efficacy of combining losartan and chemo-immunotherapy for triple negative breast cancer. Front Immunol. 2022;13:938439.
150. Zhang D, Li Q, Chen X, et al. An injectable hydrogel to modulate T cells for cancer immunotherapy. Small. 2022;18:e2202663.
151. Wang D, Zhang L, Yang WH, et al. Arginine-loaded nano-calcium-phosphate-stabilized lipiodol pickering emulsions potentiates transarterial embolization-immunotherapy. Adv Sci. 2025;12:e2410484.
152. Chen N, Li Z, Liu H, et al. Enhancing PD-1 blockade in NSCLC: reprogramming tumor immune microenvironment with albumin-bound statins targeting lipid rafts and mitochondrial respiration. Bioact Mater. 2025;49:140-53.
153. Zhou Z, Liu Y, Song W, et al. Metabolic reprogramming mediated PD-L1 depression and hypoxia reversion to reactivate tumor therapy. J Control Release. 2022;352:793-812.
154. Zhao Z, Dong S, Liu Y, et al. Tumor microenvironment-activable manganese-boosted catalytic immunotherapy combined with PD-1 checkpoint blockade. ACS Nano. 2022;16:20400-18.
155. Xiong W, Qi L, Jiang N, et al. Metformin liposome-mediated PD-L1 downregulation for amplifying the photodynamic immunotherapy efficacy. ACS Appl Mater Interfaces. 2021;13:8026-41.
156. An J, Guo R, Liu M, Hu H, Zhang H. Multi-modal Ca2+ nanogenerator via reversing T cell exhaustion for enhanced chemo-immunotherapy. J Control Release. 2024;372:715-27.
157. Yi L, Jiang X, Zhou Z, et al. A hybrid nanoadjuvant simultaneously depresses PD-L1/TGF-β1 and activates cGAS-STING pathway to overcome radio-immunotherapy resistance. Adv Mater. 2024;36:e2304328.
158. Wang T, Chen S, Sun J, Li K. Functional co-delivery nanoliposomes based on improving hypoxia for increasing photoimmunotherapy efficacy of cold tumors. Int J Pharm. 2024;663:124581.
159. Wang S, Zhou Z, Hu R, et al. Metabolic intervention liposome boosted lung cancer radio-immunotherapy via hypoxia amelioration and PD-L1 restraint. Adv Sci. 2023;10:e2207608.
160. Jiang X, Yi L, Li C, et al. Mitochondrial disruption nanosystem simultaneously depressed programmed death ligand-1 and transforming growth factor-β to overcome photodynamic immunotherapy resistance. ACS Nano. 2024;18:3331-48.
161. Chen Y, Zuo M, Jana D, et al. Priming of cancer-immunity cycle by alleviating hypoxia-induced ferroptosis resistance and immunosuppression. Biomaterials. 2025;315:122911.
162. Wu Q, Liu M, Zhang H, et al. WO3-x@ferrocene-folic acid composites induce cancer cell death and activate immunity via PTT/CDT. Small. 2025;21:e2500104.
163. Zhang X, He C, He X, et al. HIF-1 inhibitor-based one-stone-two-birds strategy for enhanced cancer chemodynamic-immunotherapy. J Control Release. 2023;356:649-62.
164. Wang Z, Zhu M, Dong R, et al. TH-302-loaded nanodrug reshapes the hypoxic tumour microenvironment and enhances PD-1 blockade efficacy in gastric cancer. J Nanobiotechnology. 2023;21:440.
165. Ji Y, Qu S, Shi G, et al. Triggered cascade-activation nanoplatform to alleviate hypoxia for effective tumor immunotherapy guided by NIR-II imaging. ACS Nano. 2024;18:31421-34.
166. Taleb M, Atabakhshi-Kashi M, Wang Y, et al. Bifunctional therapeutic peptide assembled nanoparticles exerting improved activities of tumor vessel normalization and immune checkpoint inhibition. Adv Healthc Mater. 2021;10:e2100051.
167. Lan J, Zeng R, Li Z, et al. Biomimetic nanomodulators with synergism of photothermal therapy and vessel normalization for boosting potent anticancer immunity. Adv Mater. 2024;36:e2408511.
168. Li J, Wei R, Yao W, et al. iRGD-mediated liposomal nanoplatforms for improving hepatocellular carcinoma targeted combination immunotherapy and monitoring tumor response via IVIM-MRI. J Mater Chem B. 2024;12:9963-78.
169. Zheng X, Shi Y, Tang D, et al. Near-infrared-II nanoparticles for vascular normalization combined with immune checkpoint blockade via photodynamic immunotherapy inhibit uveal melanoma growth and metastasis. Adv Sci. 2023;10:e2206932.
170. Zhang D, Jiang C, Zheng X, et al. Normalization of tumor vessels by lenvatinib-based metallo-nanodrugs alleviates hypoxia and enhances calreticulin-mediated immune responses in orthotopic HCC and organoids. Small. 2023;19:e2207786.
171. Feng X, Zeng L, Wu L, et al. Self-delivery nanodrug to manipulate tumor microenvironment for boosting photodynamic cancer immunotherapy. Biomed Pharmacother. 2024;178:117220.
172. Zhou Y, Ren X, Hou Z, Wang N, Jiang Y, Luan Y. Engineering a photosensitizer nanoplatform for amplified photodynamic immunotherapy via tumor microenvironment modulation. Nanoscale Horiz. 2021;6:120-31.
173. Li F, Shao X, Liu D, et al. Vascular disruptive hydrogel platform for enhanced chemotherapy and anti-angiogenesis through alleviation of immune surveillance. Pharmaceutics. 2022;14:1809.
174. Zhou Z, Chen J, Liu Y, et al. Cascade two-stage tumor re-oxygenation and immune re-sensitization mediated by self-assembled albumin-sorafenib nanoparticles for enhanced photodynamic immunotherapy. Acta Pharm Sin B. 2022;12:4204-23.