• Journal of Innovative Optical Health Sciences
  • Vol. 16, Issue 3, 2330007 (2023)
Trisha I. Valerio1, Coline L. Furrer1, Negar Sadeghipour1,2, Sophia-Joy X. Patrock1..., Sayre A. Tillery1, Ashley R. Hoover1, Kaili Liu1 and Wei R. Chen1,*|Show fewer author(s)
Author Affiliations
  • 1Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, USA
  • 2School of Electrical and Computer Engineering, University of Oklahoma, Norman, Oklahoma 73019, USA
  • show less
    DOI: 10.1142/S1793545823300070 Cite this Article
    Trisha I. Valerio, Coline L. Furrer, Negar Sadeghipour, Sophia-Joy X. Patrock, Sayre A. Tillery, Ashley R. Hoover, Kaili Liu, Wei R. Chen. Immune modulations of the tumor microenvironment in response to phototherapy[J]. Journal of Innovative Optical Health Sciences, 2023, 16(3): 2330007 Copy Citation Text show less
    References

    [2] The ever-increasing importance of cancer as a leading cause of premature death worldwide. Cancer, 127, 3029-3030(2021). https://doi.org/10.1002/cncr.33587

    [3] On the origin of cancer metastasis. Crit. Rev. Oncogenesis, 18, 43-73(2013). https://doi.org/10.1615/critrevoncog.v18.i1-2.40

    [4] The lingering mysteries of metastatic recurrence in breast cancer. Br. J. Cancer, 124, 13-26(2021). https://doi.org/10.1038/s41416-020-01161-4

    [5] Drug resistance in cancer: An overview. Cancers, 6, 1769-1792(2014). https://doi.org/10.3390/cancers6031769

    [6] The different mechanisms of cancer drug resistance: A brief review. Adv. Pharm. Bull., 7, 339-348(2017). https://doi.org/10.15171/apb.2017.041

    [7] Assessment of the evolution of cancer treatment therapies. Cancers, 3, 3279-3330(2011). https://doi.org/10.3390/cancers3033279

    [8] Combination therapy in combating cancer. Oncotarget, 8, 38022-38043(2017). https://doi.org/10.18632/oncotarget.16723

    [9] The most common side effects experienced by patients were receiving first cycle of chemotherapy. Iran. J. Public Health, 47, 1218-1219(2018).

    [10] Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science (New York, N.Y.), 313, 1960-1964(2006). https://doi.org/10.1126/science.1129139

    [11] The tumor microenvironment at a glance. J. Cell Sci., 125, 5591-5596(2012). https://doi.org/10.1242/jcs.116392

    [12] Tumor microenvironment abnormalities: Causes, consequences, and strategies to normalize. J. Cell. Biochem., 101, 937-949(2007). https://doi.org/10.1002/jcb.21187

    [13] Association between tumor-stroma ratio and prognosis in solid tumor patients: A systematic review and meta-analysis. Oncotarget, 7, 68954-68965(2016). https://doi.org/10.18632/oncotarget.12135

    [14] Strategies for delivering nanoparticles across tumor blood vessels. Adv. Funct. Mater., 31, 2007363(2021). https://doi.org/10.1002/adfm.202007363

    [15] A review of the development of tumor vasculature and its effects on the tumor microenvironment. Hypoxia (Auckland, N.Z.), 5, 21-32(2017). https://doi.org/10.2147/HP.S133231

    [16] An overview of the tumor microenvironment, from cells to complex networks (Review). Exp. Ther. Med., 21, 96(2021). https://doi.org/10.3892/etm.2020.9528

    [17] Cancer-associated fibroblasts: Overview, progress, challenges, and directions. Cancer Gene Ther., 28, 984-999(2021). https://doi.org/10.1038/s41417-021-00318-4

    [18] Hypoxia and the tumor microenvironment. Technol. Cancer Res. Treat., 20, 15330338211036304(2021). https://doi.org/10.1177/15330338211036304

    [19] Cancer stem cells and the tumor microenvironment in gastric cancer. Front. Oncol., 11, 803974(2022). https://doi.org/10.3389/fonc.2021.803974

    [20] Tumor endothelial cells. Cold Spring Harbor Perspect. Med., 2, a006536(2012). https://doi.org/10.1101/cshperspect.a006536

    [21] Adipocytes in the tumour microenvironment. Adv. Exp. Med. Biol., 1234, 1-13(2020). https://doi.org/10.1007/978-3-030-37184-5_1

    [22] The emerging roles of pericytes in modulating tumor microenvironment. Front. Cell Dev. Biol., 9, 676342(2021). https://doi.org/10.3389/fcell.2021.676342

    [23] The tumor microenvironment and its role in promoting tumor growth. Oncogene, 27, 5904-5912(2008). https://doi.org/10.1038/onc.2008.271

    [24] The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct. Target. Ther., 5, 166(2020). https://doi.org/10.1038/s41392-020-00280-x

    [25] The tumor microenvironment. Curr. Biol., 30, R921-R925(2020). https://doi.org/10.1016/j.cub.2020.06.081

    [26] Nano-ablative immunotherapy for cancer treatment. Nanophotonics, 10, 3247-3266(2021). https://doi.org/10.1515/nanoph-2021-0171

    [27] Single-cell-based computer simulation of the oxygen-dependent tumour response to irradiation. Phys. Med. Biol., 52, 4775-4789(2007). https://doi.org/10.1088/0031-9155/52/16/005

    [28] Modeling and computer simulations of tumor growth and tumor response to radiotherapy. Radiat. Res., 162, 71-83(2004). https://doi.org/10.1667/rr3193

    [29] The clinical role of the TME in solid cancer. Br. J. Cancer, 120, 45-53(2019). https://doi.org/10.1038/s41416-018-0327-z

    [30] How cancers escape immune destruction and mechanisms of action for the new significantly active immune therapies: Helping nonimmunologists decipher recent advances. Oncol., 21, 233-243(2016). https://doi.org/10.1634/theoncologist.2015-0282

    [31] Decreased number of mast cells infiltrating into needle biopsy specimens leads to a better prognosis of prostate cancer. Br. J. Cancer, 97, 952-956(2007). https://doi.org/10.1038/sj.bjc.6603962

    [32] Increase of mast cells and tumor angiogenesis in oral squamous cell carcinoma. J. Oral Pathol. Med., 32, 195-199(2003). https://doi.org/10.1034/j.1600-0714.2003.00128.x

    [33] Tumor vascularity and tryptase-positive mast cells correlate with a poor prognosis in melanoma. Eur. J. Clin. Invest., 33, 420-425(2003). https://doi.org/10.1046/j.1365-2362.2003.01152.x

    [34] A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer, 20, 174-186(2020). https://doi.org/10.1038/s41568-019-0238-1

    [35] Immune escape mechanisms as a guide for cancer immunotherapy. Clin. Cancer Res., 21, 687-692(2015). https://doi.org/10.1158/1078-0432.CCR-14-1860

    [36] Mechanisms of tumor escape in the context of the T-cell-inflamed and the non-T-cell-inflamed tumor microenvironment. Int. Immunol., 28, 383-391(2016). https://doi.org/10.1093/intimm/dxw014

    [37] Progress in cancer survival, mortality, and incidence in seven high-income countries 1995–2014 (ICBP SURVMARK-2): A population-based study. Lancet Oncol., 20, 1493-1505(2019). https://doi.org/10.1016/S1470-2045(19)30456-5

    [38] Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun. Signal., 18, 59(2020). https://doi.org/10.1186/s12964-020-0530-4

    [39] Multiscale computational modeling of cancer growth using features derived from microCT images. Sci. Rep., 11, 18524(2021). https://doi.org/10.1038/s41598-021-97966-1

    [40] Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol., 3, 991-998(2002). https://doi.org/10.1038/ni1102-991

    [41] Therapy-induced modulation of the tumor microenvironment: New opportunities for cancer therapies. Front. Oncol., 10, 582884(2020). https://doi.org/10.3389/fonc. 2020.582884

    [42] The role of dendritic cells in tumor microenvironments and their uses as therapeutic targets. BMB Rep., 54, 31-43(2021). https://doi.org/10.5483/BMBRep.2021.54.1.224

    [43] Dendritic cell biology and its role in tumor immunotherapy. J. Hematol. Oncol., 13, 107(2020). https://doi.org/10.1186/s13045-020-00939-6

    [44] Origin and functions of tissue macrophages. Immunity, 41, 21-35(2014). https://doi.org/10.1016/j.immuni.2014.06.013

    [45] Diverse macrophages polarization in tumor microenvironment. Arch. Pharmacal Res., 39, 1588-1596(2016). https://doi.org/10.1007/s12272-016-0820-y

    [46] Role of M1-polarized tumor-associated macrophages in the prognosis of advanced ovarian cancer patients. Sci. Rep., 10, 6096(2020). https://doi.org/10.1038/s41598-020-63276-1

    [47] Tumor-infiltrating lymphocytes in the immunotherapy era. Cell. Mol. Immunol., 18, 842-859(2021). https://doi.org/10.1038/s41423-020-00565-9

    [48] Tumor infiltrating lymphocytes: The regulator of melanoma evolution. Oncol. Lett., 17, 4155-4161(2019). https://doi.org/10.3892/ol.2019.9940

    [49] Cytotoxic CD8+ T cells in cancer and cancer immunotherapy. Br. J. Cancer, 124, 359-367(2021). https://doi.org/10.1038/s41416-020-01048-4

    [50] T-cell priming by dendritic cells in lymph nodes occurs in three distinct phases. Nature, 427, 154-159(2004). https://doi.org/10.1038/nature02238

    [51] Dendritic cells and CD8 T cell immunity in tumor microenvironment. Front. Immunol., 9, 3059(2018). https://doi.org/10.3389/fimmu.2018.03059

    [52] CD8+ cytotoxic T lymphocytes in cancer immunotherapy: A review. J. Cell. Physiol., 234, 8509-8521(2019). https://doi.org/10.1002/jcp.27782

    [53] The outstanding antitumor capacity of CD4+ T helper lymphocytes. Biochim. et Biophys. Acta Rev. Cancer, 1874, 188439(2020). https://doi.org/10.1016/j.bbcan.2020.188439

    [54] The cytokine milieu in the interplay of pathogenic Th1/Th17 cells and regulatory T cells in autoimmune disease. Cell. Mol. Immunol., 7, 182-189(2010). https://doi.org/10.1038/cmi.2010.22

    [55] CD4 T-cell subsets and tumor immunity: The helpful and the not-so-helpful. Cancer Immunol. Res., 2, 91-98(2014). https://doi.org/10.1158/2326-6066.CIR-13-0216

    [56] Modulation of CD4 T cell response according to tumor cytokine microenvironment. Cancers, 13, 373(2021). https://doi.org/10.3390/cancers13030373

    [57] Unique ectopic lymph node-like structures present in human primary colorectal carcinoma are identified by immune gene array profiling. Am. J. Pathol., 179, 37-45(2011). https://doi.org/10.1016/j.ajpath.2011.03.007

    [58] CD4+ follicular helper T cell infiltration predicts breast cancer survival. J. Clin. Invest., 123, 2873-2892(2013). https://doi.org/10.1172/JCI67428

    [59] Regulatory T (Treg) cells in cancer: Can Treg cells be a new therapeutic target?. Cancer Sci., 110, 2080-2089(2019). https://doi.org/10.1111/cas.14069

    [60] T regulatory cells and priming the suppressive tumor microenvironment. Front. Immunol., 10, 2453(2019). https://doi.org/10.3389/fimmu.2019.02453

    [61] Regulatory T cells exhibit distinct features in human breast cancer. Immunity, 45, 1122-1134(2016). https://doi.org/10.1016/j.immuni.2016.10.032

    [62] Tumor infiltrating B-cells signal functional humoral immune responses in breast cancer. JCI Insight, 5, e129641(2019). https://doi.org/10.1172/jci.insight.129641

    [63] Prognostic impact of tumour-infiltrating B cells and plasma cells in colorectal cancer. Int. J. Cancer, 139, 1129-1139(2016). https://doi.org/10.1002/ijc.30138

    [64] Immune landscape of viral- and carcinogen-driven head and neck cancer. Immunity, 52, 183-199.e9(2020). https://doi.org/10.1016/j.immuni.2019.11.014

    [65] Depleting tumor infiltrating B cells to boost antitumor immunity with tumor immune-microenvironment reshaped hybrid nanocage. ACS Nano, 16, 4263-4277(2022). https://doi.org/10.1021/acsnano.1c10283

    [66] Natural killer cells in antitumour adoptive cell immunotherapy. Nat. Rev. Cancer, 22, 557-575(2022). https://doi.org/10.1038/s41568-022-00491-0

    [67] Tailoring natural killer cell immunotherapy to the tumour microenvironment. Semin. Immunol., 31, 30-36(2017). https://doi.org/10.1016/j.smim.2017.09.001

    [68] A review of cancer immunotherapy: From the past, to the present, to the future. Curr. Oncol., 27, S87-S97(2020). https://doi.org/10.3747/co.27.5223

    [69] Immunotherapy: Enhancing the efficacy of this promising therapeutic in multiple cancers. Clin. Sci., 133, 181-193(2019). https://doi.org/10.1042/CS20181003

    [70] Cancer immunotherapy. Cancer Biother. Radiopharm., 26, 1-64(2011). https://doi.org/10.1089/cbr.2010.0902

    [71] Current approaches of photothermal therapy in treating cancer metastasis with nanotherapeutics. Theranostics, 6, 762-772(2016). https://doi.org/10.7150/thno.14988

    [72] Update on laser photochemotherapy: An alternative for cancer treatment. Anti-Cancer Agents Med. Chem., 11, 772-779(2011). https://doi.org/10.2174/187152011797378742

    [73] Photodynamic therapy of cancer: An update. CA: Cancer J. Clin., 61, 250-281(2011). https://doi.org/10.3322/caac.20114

    [74] Photosensitizers for photodynamic therapy. Adv. Healthcare Mater., 8, e1900132(2019). https://doi.org/10.1002/adhm.201900132

    [75] Photodynamic therapy and anti-tumor immunity. Lasers Surg. Med., 38, 509-515(2006). https://doi.org/10.1002/lsm.20362

    [76] Nanoscale covalent organic polymers as a biodegradable nanomedicine for chemotherapy-enhanced photodynamic therapy of cancer. Nano Res., 11, 3244-3257(2018). https://doi.org/10.1007/s12274-017-1858-y

    [77] Advances in nanomaterial-mediated photothermal cancer therapies: Toward clinical applications. Biomedicines, 9, 305(2021). https://doi.org/10.3390/biomedicines9030305

    [78] Recent progress in cancer thermal therapy using gold nanoparticles. J. Phys. Chem. C, 120, 4691-4716(2016).

    [79] Nanomaterials as photothermal therapeutic agents. Prog. Mater. Sci., 99, 1-26(2019). https://doi.org/10.1016/j.pmatsci.2018.07.005

    [80] Photothermal therapy-induced immunogenic cell death based on natural melanin nanoparticles against breast cancer. Chem. Commun., 56, 1389-1392(2020). https://doi.org/10.1039/c9cc08447a

    [81] Nanomaterial-based tumor photothermal immunotherapy. Int. J. Nanomed., 15, 9159-9180(2020). https://doi.org/10.2147/IJN.S249252

    [82] Molecularly targeted photothermal ablation improves tumor specificity and immune modulation in a rat model of hepatocellular carcinoma. Commun. Biol., 3, 783(2020). https://doi.org/10.1038/s42003-020-01522-y

    [83] Nanomaterial applications in photothermal therapy for cancer. Materials (Basel, Switzerland), 12, 779(2019). https://doi.org/10.3390/ma12050779

    [84] Advances in nanomaterials for use in photothermal and photodynamic therapeutics (Review). Mol. Med. Rep., 20, 5-15(2019). https://doi.org/10.3892/mmr.2019.10218

    [85] Thermal dose as a universal tool to evaluate nanoparticle-induced photothermal therapy. Int. J. Pharm., 587, 119657(2020). https://doi.org/10.1016/j.ijpharm.2020.119657

    [86] Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat. Rev. Clin. Oncol., 17, 657-674(2020). https://doi.org/10.1038/s41571-020-0410-2

    [87] Neutrophil infiltration and whole-cell vaccine elicited by N-dihydrogalactochitosan combined with NIR phototherapy to enhance antitumor immune response and T cell immune memory. Theranostics, 10, 1814-1832(2020). https://doi.org/10.7150/thno.38515

    [88] Single-cell RNA sequencing reveals localized tumour ablation and intratumoural immunostimulant delivery potentiate T cell mediated tumour killing. Clin. Transl. Med., 12, e937(2022). https://doi.org/10.1002/ctm2.937

    [89] Antigen presentation and interferon signatures in B cells driven by localized ablative cancer immunotherapy correlate with extended survival. Theranostics, 12, 639-656(2022). https://doi.org/10.7150/thno.65773

    [90] Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol., 16, 71(2018). https://doi.org/10.1186/s12951-018-0392-8

    [91] Smart nanoparticles for drug delivery application: Development of versatile nanocarrier platforms in biotechnology and nanomedicine. J. Nanomater., 2019, 3702518, 26(2019). https://doi.org/10.1155/2019/3702518

    [92] Tumor-microenvironment-responsive nanomedicine for enhanced cancer immunotherapy. Adv. Sci., 9, e2103836(2022). https://doi.org/10.1002/advs.202103836

    [93] Nanomedicine-based drug delivery towards tumor biological and immunological microenvironment. Acta Pharm. Sin. B, 10, 2110-2124(2020). https://doi.org/10.1016/j.apsb.2020.05.008

    [94] Modulating the tumor microenvironment to enhance tumor nanomedicine delivery. Front. Pharmacol., 8, 952(2017). https://doi.org/10.3389/fphar.2017.00952

    [95] Mechanism of laser immunotherapy: Role of immunoadjuvant and selective photothermal laser-tissue interaction. Int. Workshop Photon. Imag. Biol. Med., 4536, 82-89(2002). https://doi.org/10.1117/12.462529

    [96] Nanoparticle-based photothermal and photodynamic immunotherapy for tumor treatment. Int. J. Cancer, 143, 3050-3060(2018). https://doi.org/10.1002/ijc.31717

    [97] A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol., 20, 651-668(2020). https://doi.org/10.1038/s41577-020-0306-5

    [98] Targeted cancer therapies. Am. Fam. Physician, 103, 155-163(2021).

    [99] Multifunctional receptor-targeting antibodies for cancer therapy. Lancet Oncol., 16, e543-e554(2015). https://doi.org/10.1016/S1470-2045(15)00039-X

    [100] Therapeutic cancer vaccines. Nat. Rev. Cancer, 21, 360-378(2021). https://doi.org/10.1038/s41568-021-00346-0

    [101] Cancer vaccines. Hematol./Oncol. Clin. North America, 33, 199-214(2019). https://doi.org/10.1016/j.hoc.2018.12.001

    [102] Adoptive cell transfer as personalized immunotherapy for human cancer. Science (New York, N.Y.), 348, 62-68(2015). https://doi.org/10.1126/science.aaa4967

    [103] Oncolytic virus therapy in cancer: A current review. World J. Virol., 10, 229-255(2021). https://doi.org/10.5501/wjv.v10.i5.229

    [104] Immune checkpoint inhibitors in cancer therapy. Curr. Oncol., 29, 3044-3060(2022). https://doi.org/10.3390/curroncol29050247

    [105] Cytokines in clinical cancer immunotherapy. Br. J. Cancer, 120, 6-15(2019). https://doi.org/10.1038/s41416-018-0328-y

    [106] Use of adjuvants for immunotherapy. Hum. Vaccines Immunother., 13, 1774-1777(2017). https://doi.org/10.1080/21645515.2017.1321725

    [107] InCVAX–a novel strategy for treatment of late-stage, metastatic cancers through photoimmunotherapy induced tumor-specific immunity. Cancer Lett., 359, 169-177(2015). https://doi.org/10.1016/j.canlet.2015.01.029

    [108] Pembrolizumab for persistent, recurrent, or metastatic cervical cancer. New Engl. J. Med., 385, 1856-1867(2021). https://doi.org/10.1056/NEJMoa2112435

    [109] Pembrolizumab (Keytruda). Hum. Vaccines Immunother., 12, 2777-2789(2016). https://doi.org/10.1080/21645515.2016.1199310

    [110] Nivolumab as programmed death-1 (PD-1) inhibitor for targeted immunotherapy in tumor. J. Cancer, 8, 410-416(2017). https://doi.org/10.7150/jca.17144

    [111] Nivolumab. Drugs Today, 50, 791-802(2014). https://doi.org/10.1358/dot.2014.50.12.2235103

    [112] Tremelimumab: A review of development to date in solid tumors. Immunotherapy, 5, 215-229(2013). https://doi.org/10.2217/imt.13.9

    [113] Tremelimumab (CP-675,206), a cytotoxic T lymphocyte associated antigen 4 blocking monoclonal antibody in clinical development for patients with cancer. Oncol, 12, 873-883(2007). https://doi.org/10.1634/theoncologist.12-7-873

    [114] Ipilimumab: A novel immunostimulatory monoclonal antibody for the treatment of cancer. Pharmacol. Res., 65, 9-22(2012). https://doi.org/10.1016/j.phrs.2011.09.002

    [115] The efficacy and safety of combined immune checkpoint inhibitors (nivolumab plus ipilimumab): a systematic review and meta-analysis. World Journal of Surgical Oncology, 18, 150(2020). https://doi.org/10.1186/s12957-020-01933-5

    [116] Product review: Avelumab, an anti-PD-L1 antibody. Hum. Vaccin. Immunother., 15, 891-908(2019). https://doi.org/10.1080/21645515.2018.1551671

    [117] Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma. New Engl. J. Med., 383, 1218-1230(2020). https://doi.org/10.1056/NEJMoa2002788

    [118] Durvalumab in cancer medicine: A comprehensive review. Expert Opin. Biol. Ther., 19, 927-935(2019). https://doi.org/10.1080/14712598.2019.1635115

    [119] Durvalumab for the treatment of non-small cell lung cancer. Expert Rev. Respir. Med., 12, 627-639(2018). https://doi.org/10.1080/17476348.2018.1494575

    [120] Cancer immunotherapy using checkpoint blockade. Science (New York, N.Y.), 359, 1350-1355(2018). https://doi.org/10.1126/science.aar4060

    [121] Immune checkpoint inhibitor toxicities. Mayo Clin. Proc., 94, 1321-1329(2019). https://doi.org/10.1016/j.mayocp.2019.03.012

    [122] Setting the scene — a future ‘epidemic’ of immune-related adverse events in association with checkpoint inhibitor therapy. Rheumatology (Oxford, England), 58, vii1-vii6(2019). https://doi.org/10.1093/rheumatology/kez402

    [123] Mechanisms of resistance to immune checkpoint inhibitors. Br. J. Cancer, 118, 9-16(2018). https://doi.org/10.1038/bjc.2017.434

    [124] Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell, 184, 5309-5337(2021). https://doi.org/10.1016/j.cell.2021.09.020

    [125] Acquired resistance to immune checkpoint inhibitors. Cancer Cell, 37, 443-455(2020). https://doi.org/10.1016/j.ccell.2020.03.017

    [126] The determinants of tumour immunogenicity. Nat. Rev. Cancer, 12, 307-313(2012). https://doi.org/10.1038/nrc3246

    [127] Antigen presentation and tumor immunogenicity in cancer immunotherapy response prediction. eLife, 8, e49020(2019). https://doi.org/10.7554/eLife.49020

    [128] Prediction of prognosis, immunogenicity and efficacy of immunotherapy based on glutamine metabolism in lung adenocarcinoma. Front. Immunol., 13, 960738(2022). https://doi.org/10.3389/fimmu.2022.960738

    [129] Distinguishable prognostic signatures and tumor immunogenicity between pancreatic head cancer and pancreatic body/tail cancer. Front. Oncol., 12, 890715(2022). https://doi.org/10.3389/fonc.2022.890715

    [130] Mechanisms of immunogenic cell death and immune checkpoint blockade therapy. Kaohsiung J. Med. Sci., 37, 448-458(2021). https://doi.org/10.1002/kjm2.12375

    [131] Next generation of immune checkpoint therapy in cancer: New developments and challenges. J. Hematol. Oncol., 11, 39(2018). https://doi.org/10.1186/s13045-018-0582-8

    [132] Mild photothermal therapy potentiates anti-PD-L1 treatment for immunologically cold tumors via an all-in-one and all-in-control strategy. Nat. Commun., 10, 4871(2019). https://doi.org/10.1038/s41467-019-12771-9

    [133] Depleting tumor-associated Tregs via nanoparticle-mediated hyperthermia to enhance anti-CTLA-4 immunotherapy. Nanomed., 15, 77-92(2020). https://doi.org/10.2217/nnm-2019-0190

    [134] CAR-T cell therapy: Current limitations and potential strategies. Blood Cancer J., 11, 69(2021). https://doi.org/10.1038/s41408-021-00459-7

    [135] Risks and benefits of chimeric antigen receptor T-cell (CAR-T) Therapy in cancer: A systematic review and meta-analysis. Transfus. Med. Rev., 33, 98-110(2019). https://doi.org/10.1016/j.tmrv.2019.01.005

    [136] Advances in universal CAR-T cell therapy. Front. Immunol., 12, 744823(2021). https://doi.org/10.3389/fimmu.2021.744823

    [137] Photothermal therapy promotes tumor infiltration and antitumor activity of CAR T cells. Adv. Mater. (Deerfield Beach, Fla.), 31, e1900192(2019). https://doi.org/10.1002/adma.201900192

    [138] Myeloid-derived suppressor cell membrane-coated magnetic nanoparticles for cancer theranostics by inducing macrophage polarization and synergizing immunogenic cell death. Adv. Funct. Mater., 28, 1801389(2018). https://doi.org/10.1002/adfm.201801389

    [139] Tumor microenvironment-triggered nanosystems as dual-relief tumor hypoxia immunomodulators for enhanced phototherapy. Theranostics, 10, 9132-9152(2020). https://doi.org/10.7150/thno.46076

    [140] Enhanced systemic antilymphoma immune response by photothermal therapy with CpG deoxynucleotide-coated nanoparticles. Blood Adv., 6, 4581-4592(2022). https://doi.org/10.1182/bloodadvances.2022008040

    [141] Local phototherapy synergizes with immunoadjuvant for treatment of pancreatic cancer through induced immunogenic tumor vaccine. Clin. Cancer Res., 24, 5335-5346(2018). https://doi.org/10.1158/1078-0432.CCR-18-1126

    [142] Nanoscale reduced graphene oxide-mediated photothermal therapy together with IDO inhibition and PD-L1 blockade synergistically promote antitumor immunity. ACS Appl. Mater. Interfaces, 11, 1876-1885(2019). https://doi.org/10.1021/acsami.8b18751

    [143] Phototheranostic metal-phenolic networks with antiexosomal PD-L1 enhanced ferroptosis for synergistic immunotherapy. J. Am. Chem. Soc., 144, 787-797(2022). https://doi.org/10.1021/jacs.1c09753

    [144] Phototherapy facilitates tumor recruitment and activation of natural killer T cells for potent cancer immunotherapy. Nano Lett., 21, 6304-6313(2021). https://doi.org/10.1021/acs.nanolett.1c02238

    [145] Light-triggered multifunctional nanoplatform for efficient cancer photo-immunotherapy. J. Nanobiotechnol., 20, 181(2022). https://doi.org/10.1186/s12951-022-01388-8

    [146] New insights into tumor-infiltrating B lymphocytes in breast cancer: Clinical impacts and regulatory mechanisms. Front. Immunol., 9, 470(2018). https://doi.org/10.3389/fimmu.2018.00470

    [147] B lymphocytes and cancer: A love-hate relationship. Trends Cancer, 2, 747-757(2016). https://doi.org/10.1016/j.trecan.2016.10.010

    [148] The regulatory role of B cells in autoimmunity, infections and cancer: Perspectives beyond IL10 production. FEBS Lett., 589, 3362-3369(2015). https://doi.org/10.1016/j.febslet.2015.08.048

    [149] Presence of B cells in tertiary lymphoid structures is associated with a protective immunity in patients with lung cancer. Am. J. Respir. Crit. Care Med., 189, 832-844(2014). https://doi.org/10.1164/rccm.201309-1611OC

    [150] B lymphocyte inhibition of anti-tumor response depends on expansion of Treg but is independent of B-cell IL-10 secretion. Cancer Immunol. Immunother., 62, 87-99(2013). https://doi.org/10.1007/s00262-012-1313-6

    [151] Adoptive transfer of tumor reactive B cells confers host T-cell immunity and tumor regression. Clin. Cancer Res., 17, 4987-4995(2011). https://doi.org/10.1158/1078-0432.CCR-11-0207

    [152] B cell-regulated immune responses in tumor models and cancer patients. Oncoimmunology, 2, e25443(2013). https://doi.org/10.4161/onci.25443

    Trisha I. Valerio, Coline L. Furrer, Negar Sadeghipour, Sophia-Joy X. Patrock, Sayre A. Tillery, Ashley R. Hoover, Kaili Liu, Wei R. Chen. Immune modulations of the tumor microenvironment in response to phototherapy[J]. Journal of Innovative Optical Health Sciences, 2023, 16(3): 2330007
    Download Citation