1 |
HANAHAN D, COUSSENS L M. Accessories to the crime: functions of cells recruited to the tumor microenvironment[J]. Cancer Cell, 2012, 21(3): 309-322.
|
2 |
ANDERSON N M, SIMON M C. The tumor microenvironment[J]. Curr Biol, 2020, 30(16): R921-R925.
|
3 |
FU T, DAI L J, WU S Y, et al. Spatial architecture of the immune microenvironment orchestrates tumor immunity and therapeutic response[J]. J Hematol Oncol, 2021, 14(1): 98.
|
4 |
DUAN Q Q, ZHANG H L, ZHENG J N, et al. Turning cold into hot: firing up the tumor microenvironment[J]. Trends Cancer, 2020, 6(7): 605-618.
|
5 |
SUN Y. Tumor microenvironment and cancer therapy resistance[J]. Cancer Lett, 2016, 380(1): 205-215.
|
6 |
MANTOVANI A, ALLAVENA P, SICA A, et al. Cancer-related inflammation[J]. Nature, 2008, 454(7203): 436-444.
|
7 |
SHARMA P, HU-LIESKOVAN S, WARGO J A, et al. Primary, adaptive, and acquired resistance to cancer immunotherapy[J]. Cell, 2017, 168(4): 707-723.
|
8 |
POTT S, LIEB J D. Single-cell ATAC-seq: strength in numbers[J]. Genome Biol, 2015, 16(1): 172.
|
9 |
SOLINAS G, GERMANO G, MANTOVANI A, et al. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation[J]. J Leukoc Biol, 2009, 86(5): 1065-1073.
|
10 |
CHIM L K, WILLIAMS I L, BASHOR C J, et al. Tumor-associated macrophages induce inflammation and drug resistance in a mechanically tunable engineered model of osteosarcoma[J]. Biomaterials, 2023, 296: 122076.
|
11 |
BOUTILIER A J, ELSAWA S F. Macrophage polarization states in the tumor microenvironment[J]. Int J Mol Sci, 2021, 22(13): 6995.
|
12 |
GINHOUX F, GUILLIAMS M. Tissue-resident macrophage ontogeny and homeostasis[J]. Immunity, 2016, 44(3): 439-449.
|
13 |
MILLS C D, KINCAID K, ALT J M, et al. M-1/M-2 macrophages and the Th1/Th2 paradigm[J]. J Immunol, 2000, 164(12): 6166-6173.
|
14 |
WANG L X, ZHANG S X, WU H J, et al. M2b macrophage polarization and its roles in diseases[J]. J Leukoc Biol, 2019, 106(2): 345-358.
|
15 |
JEANNIN P, PAOLINI L, ADAM C, et al. The roles of CSFs on the functional polarization of tumor-associated macrophages[J]. FEBS J, 2018, 285(4): 680-699.
|
16 |
CHRISTOFIDES A, STRAUSS L, YEO A, et al. The complex role of tumor-infiltrating macrophages[J]. Nat Immunol, 2022, 23(8): 1148-1156.
|
17 |
MOSSER D M, EDWARDS J P. Exploring the full spectrum of macrophage activation[J]. Nat Rev Immunol, 2008, 8(12): 958-969.
|
18 |
KHALAF K, HANA D, CHOU J T T, et al. Aspects of the tumor microenvironment involved in immune resistance and drug resistance[J]. Front Immunol, 2021, 12: 656364.
|
19 |
MA J, SHAYITI F, MA J, et al. Tumor-associated macrophage-derived CCL5 promotes chemotherapy resistance and metastasis in prostatic cancer[J]. Cell Biol Int, 2021, 45(10): 2054-2062.
|
20 |
WANG H C, HAUNG L Y, WANG C J, et al. Tumor-associated macrophages promote resistance of hepatocellular carcinoma cells against sorafenib by activating CXCR2 signaling[J]. J Biomed Sci, 2022, 29(1): 99.
|
21 |
LI D B, JI H F, NIU X J, et al. Tumor-associated macrophages secrete CC-chemokine ligand 2 and induce tamoxifen resistance by activating PI3K/Akt/mTOR in breast cancer[J]. Cancer Sci, 2020, 111(1): 47-58.
|
22 |
LI H, LUO F, JIANG X Y, et al. CircITGB6 promotes ovarian cancer cisplatin resistance by resetting tumor-associated macrophage polarization toward the M2 phenotype[J]. J Immunother Cancer, 2022, 10(3): e004029.
|
23 |
ZHANG H, LIU L, LIU J B, et al. Roles of tumor-associated macrophages in anti-PD-1/PD-L1 immunotherapy for solid cancers[J]. Mol Cancer, 2023, 22(1): 58.
|
24 |
TSUKAMOTO M, IMAI K, ISHIMOTO T, et al. PD-L1 expression enhancement by infiltrating macrophage-derived tumor necrosis factor-α leads to poor pancreatic cancer prognosis[J]. Cancer Sci, 2019, 110(1): 310-320.
|
25 |
CHEN Y J, LI G N, LI X J, et al. Targeting IRG1 reverses the immunosuppressive function of tumor-associated macrophages and enhances cancer immunotherapy[J]. Sci Adv, 2023, 9(17): eadg0654.
|
26 |
YUAN S Y, CHEN W J, YANG J, et al. Tumor-associated macrophage-derived exosomes promote EGFR-TKI resistance in non-small cell lung cancer by regulating the AKT, ERK1/2 and STAT3 signaling pathways[J]. Oncol Lett, 2022, 24(4): 356.
|
27 |
LIU C, ZHAO Z L, GUO S K, et al. Exosomal miR-27a-3p derived from tumor-associated macrophage suppresses propranolol sensitivity in infantile hemangioma[J]. Cell Immunol, 2021, 370: 104442.
|
28 |
CHEN C, ZHANG L, RUAN Z Y. GATA3 encapsulated by tumor-associated macrophage-derived extracellular vesicles promotes immune escape and chemotherapy resistance of ovarian cancer cells by upregulating the CD24/siglec-10 axis[J]. Mol Pharm, 2023, 20(2): 971-986.
|
29 |
XIA Y Q, RAO L, YAO H M, et al. Engineering macrophages for cancer immunotherapy and drug delivery[J]. Adv Mater, 2020, 32(40): e2002054.
|
30 |
RODRIGUEZ-GARCIA A, LYNN R C, POUSSIN M, et al. CAR-T cell-mediated depletion of immunosuppressive tumor-associated macrophages promotes endogenous antitumor immunity and augments adoptive immunotherapy[J]. Nat Commun, 2021, 12(1): 877.
|
31 |
GUNASSEKARAN G R, POONGKAVITHAI VADEVOO S M, BAEK M C, et al. M1 macrophage exosomes engineered to foster M1 polarization and target the IL-4 receptor inhibit tumor growth by reprogramming tumor-associated macrophages into M1-like macrophages[J]. Biomaterials, 2021, 278: 121137.
|
32 |
LI C X, XU X F, WEI S H, et al. Tumor-associated macrophages: potential therapeutic strategies and future prospects in cancer[J]. J Immunother Cancer, 2021, 9(1): e001341.
|
33 |
CASSETTA L, POLLARD J W. A timeline of tumour-associated macrophage biology[J]. Nat Rev Cancer, 2023, 23(4): 238-257.
|
34 |
SIKIC B I, LAKHANI N, PATNAIK A, et al. First-in-human, first-in-class phase Ⅰ trial of the anti-CD47 antibody Hu5F9-G4 in patients with advanced cancers[J]. J Clin Oncol, 2019, 37(12): 946-953.
|
35 |
ADVANI R, FLINN I, POPPLEWELL L, et al. CD47 blockade by Hu5F9-G4 and rituximab in non-Hodgkin′s lymphoma[J]. N Engl J Med, 2018, 379(18): 1711-1721.
|
36 |
VEGLIA F, SANSEVIERO E, GABRILOVICH D I. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity[J]. Nat Rev Immunol, 2021, 21(8): 485-498.
|
37 |
RODRÍGUEZ P C, OCHOA A C. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives[J]. Immunol Rev, 2008, 222: 180-191.
|
38 |
CIMEN BOZKUS C, ELZEY B D, CRIST S A, et al. Expression of cationic amino acid transporter 2 is required for myeloid-derived suppressor cell-mediated control of T cell immunity[J]. J Immunol, 2015, 195(11): 5237-5250.
|
39 |
BAUMANN T, DUNKEL A, SCHMID C, et al. Regulatory myeloid cells paralyze T cells through cell-cell transfer of the metabolite methylglyoxal[J]. Nat Immunol, 2020, 21(5): 555-566.
|
40 |
ANTONIOS J P, SOTO H, EVERSON R G, et al. Immunosuppressive tumor-infiltrating myeloid cells mediate adaptive immune resistance via a PD-1/PD-L1 mechanism in glioblastoma[J]. Neuro Oncol, 2017, 19(6): 796-807.
|
41 |
PICO DE COAÑA Y, POSCHKE I, GENTILCORE G, et al. Ipilimumab treatment results in an early decrease in the frequency of circulating granulocytic myeloid-derived suppressor cells as well as their Arginase1 production[J]. Cancer Immunol Res, 2013, 1(3): 158-162.
|
42 |
DOLCETTI L, PERANZONI E, UGEL S, et al. Hierarchy of immunosuppressive strength among myeloid-derived suppressor cell subsets is determined by GM-CSF[J]. Eur J Immunol, 2010, 40(1): 22-35.
|
43 |
MILLRUD C R, BERGENFELZ C, LEANDERSSON K. On the origin of myeloid-derived suppressor cells[J]. Oncotarget, 2017, 8(2): 3649-3665.
|
44 |
NEFEDOVA Y, NAGARAJ S, ROSENBAUER A, et al. Regulation of dendritic cell differentiation and antitumor immune response in cancer by pharmacologic-selective inhibition of the Janus-activated kinase 2/signal transducers and activators of transcription 3 pathway[J]. Cancer Res, 2005, 65(20): 9525-9535.
|
45 |
NI X L, HU G H, CAI X. The success and the challenge of all-trans retinoic acid in the treatment of cancer[J]. Crit Rev Food Sci Nutr, 2019, 59(sup1): S71-S80.
|
46 |
FUJITA M, KOHANBASH G, FELLOWS-MAYLE W, et al. COX-2 blockade suppresses gliomagenesis by inhibiting myeloid-derived suppressor cells[J]. Cancer Res, 2011, 71(7): 2664-2674.
|
47 |
AL-KHAMI A A, ZHENG L Q, DEL VALLE L, et al. Exogenous lipid uptake induces metabolic and functional reprogramming of tumor-associated myeloid-derived suppressor cells[J]. Oncoimmunology, 2017, 6(10): e1344804.
|
48 |
JIAN S L, CHEN W W, SU Y C, et al. Glycolysis regulates the expansion of myeloid-derived suppressor cells in tumor-bearing hosts through prevention of ROS-mediated apoptosis[J]. Cell Death Dis, 2017, 8(5): e2779.
|
49 |
ZITVOGEL L, APETOH L, GHIRINGHELLI F, et al. Immunological aspects of cancer chemotherapy[J]. Nat Rev Immunol, 2008, 8(1): 59-73.
|
50 |
ERIKSSON E, WENTHE J, IRENAEUS S, et al. Gemcitabine reduces MDSCs, Tregs and TGFβ-1 while restoring the Teff/Treg ratio in patients with pancreatic cancer[J]. J Transl Med, 2016, 14(1): 282.
|
51 |
SAKAGUCHI S, SAKAGUCHI N, ASANO M, et al. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor α-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases[J]. J Immunol, 1995, 155(3): 1151-1164.
|
52 |
EXPOSITO F, REDRADO M, HOURY M, et al. PTEN loss confers resistance to anti-PD-1 therapy in non-small cell lung cancer by increasing tumor infiltration of regulatory T cells[J]. Cancer Res, 2023, 83(15): 2513-2526.
|
53 |
MARSHALL L A, MARUBAYASHI S, JORAPUR A, et al. Tumors establish resistance to immunotherapy by regulating Treg recruitment via CCR4[J]. J Immunother Cancer, 2020, 8(2): e000764.
|
54 |
GAO Y N, YOU M J, FU J L, et al. Intratumoral stem-like CCR4+ regulatory T cells orchestrate the immunosuppressive microenvironment in HCC associated with hepatitis B[J]. J Hepatol, 2022, 76(1): 148-159.
|
55 |
D'ALISE A M, LEONI G, LUCIA M D, et al. Maximizing cancer therapy via complementary mechanisms of immune activation: PD-1 blockade, neoantigen vaccination, and Tregs depletion[J]. J Immunother Cancer, 2021, 9(11): e003480.
|
56 |
FONG W, LI Q, JI F F, et al. Lactobacillus gallinarum-derived metabolites boost anti-PD1 efficacy in colorectal cancer by inhibiting regulatory T cells through modulating IDO1/Kyn/AHR axis[J]. Gut, 2023, 72(12): 2272-2285.
|
57 |
IMBERT C, MONTFORT A, FRAISSE M, et al. Resistance of melanoma to immune checkpoint inhibitors is overcome by targeting the sphingosine kinase-1[J]. Nat Commun, 2020, 11(1): 437.
|
58 |
LI Z T, DENG Y Y, SUN H H, et al. Redox modulation with a perfluorocarbon nanoparticle to reverse Treg-mediated immunosuppression and enhance anti-tumor immunity[J]. J Control Release, 2023, 358: 579-590.
|
59 |
PIPER M, VAN COURT B, MUELLER A, et al. Targeting Treg-expressed STAT3 enhances NK-mediated surveillance of metastasis and improves therapeutic response in pancreatic adenocarcinoma[J]. Clin Cancer Res, 2022, 28(5): 1013-1026.
|
60 |
REVENKO A, CARNEVALLI L S, SINCLAIR C, et al. Direct targeting of FOXP3 in Tregs with AZD8701, a novel antisense oligonucleotide to relieve immunosuppression in cancer[J]. J Immunother Cancer, 2022, 10(4): e003892.
|
61 |
AMOOZGAR Z, KLOEPPER J, REN J, et al. Targeting Treg cells with GITR activation alleviates resistance to immunotherapy in murine glioblastomas[J]. Nat Commun, 2021, 12(1): 2582.
|
62 |
TANG F, LI J H, QI L, et al. A pan-cancer single-cell panorama of human natural killer cells[J]. Cell, 2023, 186(19): 4235-4251.e20.
|
63 |
LI L, MOHANTY V, DOU J Z, et al. Loss of metabolic fitness drives tumor resistance after CAR-NK cell therapy and can be overcome by cytokine engineering[J]. Sci Adv, 2023, 9(30): eadd6997.
|
64 |
MYERS J A, MILLER J S. Exploring the NK cell platform for cancer immunotherapy[J]. Nat Rev Clin Oncol, 2021, 18(2): 85-100.
|
65 |
FANTINI M, ARLEN P M, TSANG K Y. Potentiation of natural killer cells to overcome cancer resistance to NK cell-based therapy and to enhance antibody-based immunotherapy[J]. Front Immunol, 2023, 14: 1275904.
|
66 |
LUO H Y, ZHOU Y H, ZHANG J, et al. NK cell-derived exosomes enhance the anti-tumor effects against ovarian cancer by delivering cisplatin and reactivating NK cell functions[J]. Front Immunol, 2022, 13: 1087689.
|
67 |
NAKAMURA T, SATO T, ENDO R, et al. STING agonist loaded lipid nanoparticles overcome anti-PD-1 resistance in melanoma lung metastasis via NK cell activation[J]. J Immunother Cancer, 2021, 9(7): e002852.
|
68 |
VALERI A, GARCÍA-ORTIZ A, CASTELLANO E, et al. Overcoming tumor resistance mechanisms in CAR-NK cell therapy[J]. Front Immunol, 2022, 13: 953849.
|
69 |
ZUO H, YANG M J, JI Q, et al. Targeting neutrophil extracellular traps: a novel antitumor strategy[J]. J Immunol Res, 2023, 2023: 5599660.
|
70 |
MOUSSET A, LECORGNE E, BOURGET I, et al. Neutrophil extracellular traps formed during chemotherapy confer treatment resistance via TGF-β activation[J]. Cancer Cell, 2023, 41(4): 757-775.e10.
|
71 |
FLORES-BORJA F, BOSMA A, NG D, et al. CD19+CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation[J]. Sci Transl Med, 2013, 5(173): 173ra23.
|
72 |
ZHOU X, SU Y X, LAO X M, et al. CD19+IL-10+ regulatory B cells affect survival of tongue squamous cell carcinoma patients and induce resting CD4+ T cells to CD4+Foxp3+ regulatory T cells[J]. Oral Oncol, 2016, 53: 27-35.
|
73 |
LI S R, MIRLEKAR B, JOHNSON B M, et al. STING-induced regulatory B cells compromise NK function in cancer immunity[J]. Nature, 2022, 610(7931): 373-380.
|
74 |
BARTOSIŃSKA J, PURKOT J, KARCZMARCZYK A, et al. Differential function of a novel population of the CD19+CD24hiCD38hi Bregs in psoriasis and multiple myeloma[J]. Cells, 2021, 10(2): 411.
|