Hot and cold tumors: Immunological features and the therapeutic strategies
Lianjie Wang
Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Lianjie Wang, Hui Geng, Yujie Liu contributed equally to this work.
Search for more papers by this authorHui Geng
Department of Internal Medicine, Shanghai International Medical Center, Shanghai, China
Lianjie Wang, Hui Geng, Yujie Liu contributed equally to this work.
Search for more papers by this authorYujie Liu
Department of Nephrology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Lianjie Wang, Hui Geng, Yujie Liu contributed equally to this work.
Search for more papers by this authorLei Liu
Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Search for more papers by this authorYanhua Chen
Department of the Tumor Research Center, Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Search for more papers by this authorFanchen Wu
Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Search for more papers by this authorZhiyi Liu
Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Search for more papers by this authorCorresponding Author
Shiliang Ling
Department of Medical Oncology, Ningbo Hospital of Traditional Chinese Medicine, Zhejiang Province, Ningbo, China
Correspondence
Lihong Zhou, Department of Medical Oncology and Cancer Institute of Integrative, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
Email: [email protected]
Yan Wang, Department of Medical Oncology and Cancer Institute of Integrative, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
Email: [email protected]
Shiliang Ling, Department of Medical Oncology, Ningbo Hospital of Traditional Chinese Medicine, Ningbo 315010, China.
Email: [email protected]
Search for more papers by this authorCorresponding Author
Yan Wang
Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Correspondence
Lihong Zhou, Department of Medical Oncology and Cancer Institute of Integrative, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
Email: [email protected]
Yan Wang, Department of Medical Oncology and Cancer Institute of Integrative, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
Email: [email protected]
Shiliang Ling, Department of Medical Oncology, Ningbo Hospital of Traditional Chinese Medicine, Ningbo 315010, China.
Email: [email protected]
Search for more papers by this authorCorresponding Author
Lihong Zhou
Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Correspondence
Lihong Zhou, Department of Medical Oncology and Cancer Institute of Integrative, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
Email: [email protected]
Yan Wang, Department of Medical Oncology and Cancer Institute of Integrative, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
Email: [email protected]
Shiliang Ling, Department of Medical Oncology, Ningbo Hospital of Traditional Chinese Medicine, Ningbo 315010, China.
Email: [email protected]
Search for more papers by this authorLianjie Wang
Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Lianjie Wang, Hui Geng, Yujie Liu contributed equally to this work.
Search for more papers by this authorHui Geng
Department of Internal Medicine, Shanghai International Medical Center, Shanghai, China
Lianjie Wang, Hui Geng, Yujie Liu contributed equally to this work.
Search for more papers by this authorYujie Liu
Department of Nephrology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Lianjie Wang, Hui Geng, Yujie Liu contributed equally to this work.
Search for more papers by this authorLei Liu
Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Search for more papers by this authorYanhua Chen
Department of the Tumor Research Center, Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Search for more papers by this authorFanchen Wu
Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Search for more papers by this authorZhiyi Liu
Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Search for more papers by this authorCorresponding Author
Shiliang Ling
Department of Medical Oncology, Ningbo Hospital of Traditional Chinese Medicine, Zhejiang Province, Ningbo, China
Correspondence
Lihong Zhou, Department of Medical Oncology and Cancer Institute of Integrative, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
Email: [email protected]
Yan Wang, Department of Medical Oncology and Cancer Institute of Integrative, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
Email: [email protected]
Shiliang Ling, Department of Medical Oncology, Ningbo Hospital of Traditional Chinese Medicine, Ningbo 315010, China.
Email: [email protected]
Search for more papers by this authorCorresponding Author
Yan Wang
Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Correspondence
Lihong Zhou, Department of Medical Oncology and Cancer Institute of Integrative, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
Email: [email protected]
Yan Wang, Department of Medical Oncology and Cancer Institute of Integrative, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
Email: [email protected]
Shiliang Ling, Department of Medical Oncology, Ningbo Hospital of Traditional Chinese Medicine, Ningbo 315010, China.
Email: [email protected]
Search for more papers by this authorCorresponding Author
Lihong Zhou
Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
Correspondence
Lihong Zhou, Department of Medical Oncology and Cancer Institute of Integrative, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
Email: [email protected]
Yan Wang, Department of Medical Oncology and Cancer Institute of Integrative, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
Email: [email protected]
Shiliang Ling, Department of Medical Oncology, Ningbo Hospital of Traditional Chinese Medicine, Ningbo 315010, China.
Email: [email protected]
Search for more papers by this authorAbstract
The “hotness” or “coldness” of the tumors are determined by the information of the cancer cells themselves, tumor immune characteristics, tumor microenvironment, and signaling mechanisms, which are key factors affecting cancer patients’ clinical efficacy. The switch mechanism of “hotness” and “coldness” and its corresponding pathological characteristics and treatment strategies are the frontier and hot spot of tumor treatment. How to distinguish the “hotness” or “coldness” effectively and clarify the causes, microenvironment state, and characteristics are very important for the tumor response and efficacy treatments. Starting from the concept of hot and cold tumor, this review systematically summarized the molecular characteristics, influencing factors, and therapeutic strategies of “hot and cold tumors,” and analyzed the immunophenotypes, the tumor microenvironment, the signaling pathways, and the molecular markers that contribute to “hot and cold tumors” in details. Different therapeutic strategies for “cold and hot tumors” based on clinical efficacy were analyzed with drug targets and proteins for “cold and hot tumors.” Furthermore, this review combines the therapeutic strategies of different “hot and cold tumors” with traditional medicine and modern medicine, to provide a basis and guidance for clinical decision-making of cancer treatment.
CONFLICT OF INTEREST STATEMENT
The authors declare that there are no conflicts of interest regarding the publication of this paper.
Open Research
DATA AVAILABILITY STATEMENT
Not applicable.
Not applicable.
REFERENCES
- 1Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: gLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021; 71(3): 209-249.
- 2Tsimberidou AM, Fountzilas E, Nikanjam M, Kurzrock R. Review of precision cancer medicine: evolution of the treatment paradigm. Cancer Treat Rev. 2020; 86:102019.
- 3Mirnezami R, Nicholson J, Darzi A. Preparing for precision medicine. N Engl J Med. 2012; 366(6): 489-491.
- 4Garraway LA, Verweij J, Ballman KV. Precision oncology: an overview. J Clin Oncol. 2013; 31(15): 1803-1805.
- 5Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med. 2015; 372(9): 793-795.
- 6Rubin MA. Health: make precision medicine work for cancer care. Nature. 2015; 520(7547): 290-291.
- 7Janiaud P, Serghiou S, Ioannidis JPA. New clinical trial designs in the era of precision medicine: an overview of definitions, strengths, weaknesses, and current use in oncology. Cancer Treat Rev. 2019; 73: 20-30.
- 8Duan J, Cui L, Zhao X, et al. Use of immunotherapy with programmed cell death 1 vs programmed cell death ligand 1 inhibitors in patients with cancer: a systematic review and meta-analysis. JAMA Oncol. 2020; 6(3): 375-384.
- 9Lin H, Cheng J, Mu W, Zhou J, Zhu L. Advances in universal CAR-T cell therapy. Front Immunol. 2021; 12:744823.
- 10Ma S, Li X, Wang X, et al. Current progress in CAR-T cell therapy for solid tumors. Int J Biol Sci. 2019; 15(12): 2548-2560.
- 11Newick K, O'Brien S, Moon E, Albelda SM. CAR T cell therapy for solid tumors. Annu Rev Med. 2017; 68: 139-152.
- 12Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001; 357(9255): 539-545.
- 13Coley WB II. Contribution to the knowledge of sarcoma. Ann Surg. 1891; 14(3): 199-220.
- 14Camus M, Tosolini M, Mlecnik B, et al. Coordination of intratumoral immune reaction and human colorectal cancer recurrence. Cancer Res. 2009; 69(6): 2685-2693.
- 15Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017; 541(7637): 321-330.
- 16Galon J, Mlecnik B, Bindea G, et al. Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. J Pathol. 2014; 232(2): 199-209.
- 17Pages F, Mlecnik B, Marliot F, et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet. 2018; 391(10135): 2128-2139.
- 18Miller KD, Nogueira L, Mariotto AB, et al. Cancer treatment and survivorship statistics, 2019. CA Cancer J Clin. 2019; 69(5): 363-385.
- 19Ioannides CG, Whiteside TL. T cell recognition of human tumors: implications for molecular immunotherapy of cancer. Clin Immunol Immunopathol. 1993; 66(2): 91-106.
- 20Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov. 2019; 18(3): 197-218.
- 21Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015; 348(6230): 56-61.
- 22Galon J, Costes A, Sanchez-Cabo F, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006; 313(5795): 1960-1964.
- 23Horjus Talabur Horje CS, Smids C, Meijer JW, et al. High endothelial venules associated with T cell subsets in the inflamed gut of newly diagnosed inflammatory bowel disease patients. Clin Exp Immunol. 2017; 188(1): 163-173.
- 24Muniz LR, Pacer ME, Lira SA, Furtado GC. A critical role for dendritic cells in the formation of lymphatic vessels within tertiary lymphoid structures. J Immunol. 2011; 187(2): 828-834.
- 25Ley K. M1 means kill; M2 means heal. J Immunol. 2017; 199(7): 2191-2193.
- 26Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 2013; 14(10): 1014-1022.
- 27Kwok G, Yau TC, Chiu JW, Tse E, Kwong YL. Pembrolizumab (Keytruda). Hum Vaccin Immunother. 2016; 12(11): 2777-2789.
- 28Conforti F, Pala L, Bagnardi V, et al. Cancer immunotherapy efficacy and patients' sex: a systematic review and meta-analysis. Lancet Oncol. 2018; 19(6): 737-746.
- 29Finkelmeier F, Waidmann O, Trojan J. Nivolumab for the treatment of hepatocellular carcinoma. Expert Rev Anticancer Ther. 2018; 18(12): 1169-1175.
- 30Smith KM, Desai J. Nivolumab for the treatment of colorectal cancer. Expert Rev Anticancer Ther. 2018; 18(7): 611-618.
- 31Fulchiero E, Jimeno A. Nivolumab. Drugs Today (Barc). 2014; 50(12): 791-802.
- 32Koppolu V, Rekha Vasigala VK. Checkpoint immunotherapy by nivolumab for treatment of metastatic melanoma. J Cancer Res Ther. 2018; 14(6): 1167-1175.
- 33Cercek A, Lumish M, Sinopoli J, et al. PD-1 blockade in mismatch repair-deficient, locally advanced rectal cancer. N Engl J Med. 2022; 386(25): 2363-2376.
- 34Markham A. Dostarlimab: first approval. Drugs. 2021; 81(10): 1213-1219.
- 35Zhang L, Hao B, Geng Z, Geng Q. Toripalimab: the first domestic anti-tumor PD-1 antibody in China. Front Immunol. 2021; 12:730666.
- 36Keam SJ. Toripalimab: first global approval. Drugs. 2019; 79(5): 573-578.
- 37Lee A, Keam SJ. Tislelizumab: first approval. Drugs. 2020; 80(6): 617-624.
- 38Liu SY, Wu YL. Tislelizumab: an investigational anti-PD-1 antibody for the treatment of advanced non-small cell lung cancer (NSCLC). Expert Opin Investig Drugs. 2020; 29(12): 1355-1364.
- 39Markham A, Keam SJ. Camrelizumab: first global approval. Drugs. 2019; 79(12): 1355-1361.
- 40Luo H, Lu J, Bai Y, et al. Effect of camrelizumab vs placebo added to chemotherapy on survival and progression-free survival in patients with advanced or metastatic esophageal squamous cell carcinoma: the ESCORT-1st randomized clinical trial. JAMA. 2021; 326(10): 916-925.
- 41Hoy SM. Sintilimab: first global approval. Drugs. 2019; 79(3): 341-346.
- 42Zhang L, Mai W, Jiang W, Geng Q. Sintilimab: a promising anti-tumor PD-1 antibody. Front Oncol. 2020; 10:594558.
- 43Dhillon S. Penpulimab: first approval. Drugs. 2021; 81(18): 2159-2166.
- 44Song Y, Zhou K, Jin C, et al. Penpulimab for relapsed or refractory classical hodgkin lymphoma: a multicenter, single-arm, pivotal phase I/II trial (AK105-201). Front Oncol. 2022; 12:925236.
- 45Markham A. Zimberelimab: first approval. Drugs. 2021; 81(17): 2063-2068.
- 46Lin N, Zhang M, Bai H, et al. Efficacy and safety of GLS-010 (zimberelimab) in patients with relapsed or refractory classical Hodgkin lymphoma: a multicenter, single-arm, phase II study. Eur J Cancer. 2022; 164: 117-126.
- 47Lee A. Serplulimab: first approval. Drugs. 2022; 82(10): 1137-1141.
- 48Zhu Y, Liu K, Qin Q, Zhu H. Serplulimab plus chemotherapy as first-line treatment for extensive-stage small-cell lung cancer: a cost-effectiveness analysis. Front Immunol. 2022; 13:1044678.
- 49Qin S, Li J, Zhong H, et al. Serplulimab, a novel anti-PD-1 antibody, in patients with microsatellite instability-high solid tumours: an open-label, single-arm, multicentre, phase II trial. Br J Cancer. 2022; 127(12): 2241-2248.
- 50Cao J, Wang B, Zhang J, Tao Z, Wang L, Hu X. Phase 1b clinical trial of pucotenlimab (HX008), a novel anti-PD-1 monoclonal antibody, combined with gemcitabine and cisplatin in the first-line treatment of metastatic triple-negative breast cancer. Front Oncol. 2022; 12:837963.
- 51Dhillon S. Pucotenlimab: first approval. Drugs. 2022; 82(15): 1557-1564.
- 52Markham A. Atezolizumab: first global approval. Drugs. 2016; 76(12): 1227-1232.
- 53Reddy SM, Carroll E, Nanda R. Atezolizumab for the treatment of breast cancer. Expert Rev Anticancer Ther. 2020; 20(3): 151-158.
- 54Syed YY. Durvalumab: first global approval. Drugs. 2017; 77(12): 1369-1376.
- 55Alvarez-Argote J, Dasanu CA. Durvalumab in cancer medicine: a comprehensive review. Expert Opin Biol Ther. 2019; 19(9): 927-935.
- 56Li J, Deng Y, Zhang W, et al. Subcutaneous envafolimab monotherapy in patients with advanced defective mismatch repair/microsatellite instability high solid tumors. J Hematol Oncol. 2021; 14(1): 95.
- 57Markham A. Envafolimab: first approval. Drugs. 2022; 82(2): 235-240.
- 58Dhillon S, Duggan S. Sugemalimab: first approval. Drugs. 2022; 82(5): 593-599.
- 59Zhou C, Wang Z, Sun Y, et al. Sugemalimab versus placebo, in combination with platinum-based chemotherapy, as first-line treatment of metastatic non-small-cell lung cancer (GEMSTONE-302): interim and final analyses of a double-blind, randomised, phase 3 clinical trial. Lancet Oncol. 2022; 23(2): 220-233.
- 60Zhou Q, Chen M, Jiang O, et al. Sugemalimab versus placebo after concurrent or sequential chemoradiotherapy in patients with locally advanced, unresectable, stage III non-small-cell lung cancer in China (GEMSTONE-301): interim results of a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2022; 23(2): 209-219.
- 61Wang J, Zhou C, Yao W, et al. Adebrelimab or placebo plus carboplatin and etoposide as first-line treatment for extensive-stage small-cell lung cancer (CAPSTONE-1): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2022; 23(6): 739-747.
- 62Yan W, Zhong WZ, Liu YH, et al. Adebrelimab (SHR-1316) in combination with chemotherapy as perioperative treatment in patients with resectable stage II to III NSCLCs: an open-label, multicenter, phase 1b trial. J Thorac Oncol. 2023; 18(2): 194-203.
- 63Cameron F, Whiteside G, Perry C. Ipilimumab: first global approval. Drugs. 2011; 71(8): 1093-1104.
- 64Korman AJ, Garrett-Thomson SC, Lonberg N. The foundations of immune checkpoint blockade and the ipilimumab approval decennial. Nat Rev Drug Discov. 2022; 21(7): 509-528.
- 65Keam SJ. Cadonilimab: first approval. Drugs. 2022; 82(12): 1333-1339.
- 66Pang X, Huang Z, Zhong T, et al. Cadonilimab, a tetravalent PD-1/CTLA-4 bispecific antibody with trans-binding and enhanced target binding avidity. MAbs. 2023; 15(1):2180794.
- 67Boger C, Behrens HM, Mathiak M, Kruger S, Kalthoff H, Rocken C. PD-L1 is an independent prognostic predictor in gastric cancer of Western patients. Oncotarget. 2016; 7(17): 24269-24283.
- 68Yang B, Liu T, Qu Y, et al. Progresses and perspectives of anti-PD-1/PD-L1 antibody therapy in head and neck cancers. Front Oncol. 2018; 8: 563.
- 69Jiang Y, Chen M, Nie H, Yuan Y. PD-1 and PD-L1 in cancer immunotherapy: clinical implications and future considerations. Hum Vaccin Immunother. 2019; 15(5): 1111-1122.
- 70Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012; 12(4): 252-264.
- 71Ai L, Chen J, Yan H, et al. Research status and outlook of PD-1/PD-L1 inhibitors for cancer therapy. Drug Des Devel Ther. 2020; 14: 3625-3649.
- 72Rongshan C, Liyan X. Research progress of predictive markers for efficacy of immune checkpoint inhibitors for esophageal squamous cell carcinoma. Cancer Res Prevent Treat. 2022; 49(12): 1302-1306.
- 73Kulangara K, Zhang N, Corigliano E, et al. Clinical utility of the combined positive score for programmed death ligand-1 expression and the approval of pembrolizumab for treatment of gastric cancer. Arch Pathol Lab Med. 2019; 143(3): 330-337.
- 74Kulangara K, Hanks DA, Waldroup S, et al. Development of the combined positive score (CPS) for the evaluation of PD-L1 in solid tumors with the immunohistochemistry assay PD-L1 IHC 22C3 pharmDx. J Clin Oncol. 2017; 35(15): e14589-e14589. _suppl.
- 75Yue-jun W, Na-na W, Miao-miao Y, et al. Pathological study on expression and distribution of PD-L1 protein in gastric cancer. Chinese J Clin Exp Pathol. 2019; 35(09): 1053-1059.
- 76Hogner A, Moehler M. Immunotherapy in gastric cancer. Curr Oncol. 2022; 29(3): 1559-1574.
- 77Chang WJ, Du Y, Zhao X, Ma LY, Cao GW. Inflammation-related factors predicting prognosis of gastric cancer. World J Gastroenterol. 2014; 20(16): 4586-4596.
- 78Johansson M, Denardo DG, Coussens LM. Polarized immune responses differentially regulate cancer development. Immunol Rev. 2008; 222: 145-154.
- 79Mossanen JC, Tacke F. Role of lymphocytes in liver cancer. Oncoimmunology. 2013; 2(11):e26468.
- 80Wenya C, Shaoling R, Zhenguo Z. Research progress of tumor infiltrating lymphocytes in tumor immunity. Guangxi J Tradit Chinese Med. 2020; 43(05): 66-68.
- 81Hendry S, Salgado R, Gevaert T, et al. Assessing tumor-infiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the international immunooncology biomarkers working group: part 1: assessing the host immune response, TILs in invasive breast carcinoma and ductal carcinoma in situ, metastatic tumor deposits and areas for further research. Adv Anat Pathol. 2017; 24(5): 235-251.
- 82Denkert C, Loibl S, Noske A, et al. Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol. 2010; 28(1): 105-113.
- 83Ying L, Yan F, Meng Q, et al. Understanding immune phenotypes in human gastric disease tissues by multiplexed immunohistochemistry. J Transl Med. 2017; 15(1): 206.
- 84Shirasawa M, Yoshida T, Shimoda Y, et al. Differential immune-related microenvironment determines programmed cell death protein-1/programmed death-ligand 1 blockade efficacy in patients with advanced NSCLC. J Thorac Oncol. 2021; 16(12): 2078-2090.
- 85Williams DS, Mouradov D, Jorissen RN, et al. Lymphocytic response to tumour and deficient DNA mismatch repair identify subtypes of stage II/III colorectal cancer associated with patient outcomes. Gut. 2019; 68(3): 465-474.
- 86Ruddle NH. Basics of inducible lymphoid organs. Curr Top Microbiol Immunol. 2020; 426: 1-19.
- 87Munoz-Erazo L, Rhodes JL, Marion VC, Kemp RA. Tertiary lymphoid structures in cancer - considerations for patient prognosis. Cell Mol Immunol. 2020; 17(6): 570-575.
- 88Da-chuan Z, Wen-ting H, Lei P, Yang H, Jing-ting J, Qing L. Tertiary lymphoid structure in gastric cancer and its clinical significance. Chinese J Clin Exp Pathol. 2019; 35(02): 139-143.
- 89Helmink BA, Reddy SM, Gao J, et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature. 2020; 577(7791): 549-555.
- 90Dieu-Nosjean MC, Antoine M, Danel C, et al. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol. 2008; 26(27): 4410-4417.
- 91Goc J, Germain C, Vo-Bourgais TK, et al. Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res. 2014; 74(3): 705-715.
- 92Silina K, Soltermann A, Attar FM, et al. Germinal centers determine the prognostic relevance of tertiary lymphoid structures and are impaired by corticosteroids in lung squamous cell carcinoma. Cancer Res. 2018; 78(5): 1308-1320.
- 93Ukita M, Hamanishi J, Yoshitomi H, et al. CXCL13-producing CD4+ T cells accumulate in the early phase of tertiary lymphoid structures in ovarian cancer. JCI Insight. 2022; 7(12):e157215.
- 94Salem D, Chelvanambi M, Storkus WJ, Fecek RJ. Cutaneous melanoma: mutational status and potential links to tertiary lymphoid structure formation. Front Immunol. 2021; 12:629519.
- 95Cabrita R, Lauss M, Sanna A, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020; 577(7791): 561-565.
- 96Posch F, Silina K, Leibl S, et al. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer. Oncoimmunology. 2018; 7(2):e1378844.
- 97Ruffin AT, Cillo AR, Tabib T, et al. B cell signatures and tertiary lymphoid structures contribute to outcome in head and neck squamous cell carcinoma. Nat Commun. 2021; 12(1): 3349.
- 98Li K, Guo Q, Zhang X, et al. Oral cancer-associated tertiary lymphoid structures: gene expression profile and prognostic value. Clin Exp Immunol. 2020; 199(2): 172-181.
- 99Finkin S, Yuan D, Stein I, et al. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat Immunol. 2015; 16(12): 1235-1244.
- 100Calderaro J, Petitprez F, Becht E, et al. Intra-tumoral tertiary lymphoid structures are associated with a low risk of early recurrence of hepatocellular carcinoma. J Hepatol. 2019; 70(1): 58-65.
- 101Prabhakaran S, Rizk VT, Ma Z, et al. Evaluation of invasive breast cancer samples using a 12-chemokine gene expression score: correlation with clinical outcomes. Breast Cancer Res. 2017; 19(1): 71.
- 102Gadgil R, Barthelemy J, Lewis T, Leffak M. Replication stalling and DNA microsatellite instability. Biophys Chem. 2017; 225: 38-48.
- 103Shinozuka K, Tang H, Jones RB, Li D, Nieto Y. Impact of polymorphic variations of gemcitabine metabolism, DNA damage repair, and drug-resistance genes on the effect of high-dose chemotherapy for relapsed or refractory lymphoid malignancies. Biol Blood Marrow Transplant. 2016; 22(5): 843-849.
- 104Liu X, Meltzer SJ. Gastric cancer in the era of precision medicine. Cell Mol Gastroenterol Hepatol. 2017; 3(3): 348-358.
- 105Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015; 372(26): 2509-2520.
- 106Verma R, Agarwal AK, Sakhuja P, Sharma PC. Microsatellite instability in mismatch repair and tumor suppressor genes and their expression profiling provide important targets for the development of biomarkers in gastric cancer. Gene. 2019; 710: 48-58.
- 107Kloor M, Staffa L, Ahadova A, von Knebel Doeberitz M. Clinical significance of microsatellite instability in colorectal cancer. Langenbecks Arch Surg. 2014; 399(1): 23-31.
- 108Li K, Luo H, Huang L, Luo H, Zhu X. Microsatellite instability: a review of what the oncologist should know. Cancer Cell Int. 2020; 20: 16.
- 109Chang L, Chang M, Chang HM, Chang F. Microsatellite instability: a predictive biomarker for cancer immunotherapy. Appl Immunohistochem Mol Morphol. 2018; 26(2): e15-e21.
- 110Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017; 357(6349): 409-413.
- 111Overman MJ, Lonardi S, Wong KYM, et al. Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J Clin Oncol. 2018; 36(8): 773-779.
- 112Overman MJ, McDermott R, Leach JL, et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study. Lancet Oncol. 2017; 18(9): 1182-1191.
- 113Diaz LA, Marabelle A, Delord J-P, et al. Pembrolizumab therapy for microsatellite instability high (MSI-H) colorectal cancer (CRC) and non-CRC. J Clin Oncol. 2017; 35(15_suppl): 3071-3071.
- 114O'Malley DM, Bariani GM, Cassier PA, et al. Pembrolizumab in patients with microsatellite instability-high advanced endometrial cancer: results from the KEYNOTE-158 study. J Clin Oncol. 2022; 40(7): 752-761.
- 115Marabelle A, Le DT, Ascierto PA, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. 2020; 38(1): 1-10.
- 116Zhang X, Shi M, Chen T, Zhang B. Characterization of the immune cell infiltration landscape in head and neck squamous cell carcinoma to aid immunotherapy. Mol Ther Nucleic Acids. 2020; 22: 298-309.
- 117Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature. 2013; 500(7463): 415-421.
- 118Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014; 371(23): 2189-2199.
- 119Chan TA, Yarchoan M, Jaffee E, et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol. 2019; 30(1): 44-56.
- 120Subbiah V, Solit DB, Chan TA, Kurzrock R. The FDA approval of pembrolizumab for adult and pediatric patients with tumor mutational burden (TMB) >/= 10: a decision centered on empowering patients and their physicians. Ann Oncol. 2020; 31(9): 1115-1118.
- 121Goodman A, Kato S, Bazhenova L, et al. Comprehensive genomic profiling to identify tumor mutational burden (TMB) as an independent predictor of response to immunotherapy in diverse cancers. J Clin Oncol. 2017; 35(15_suppl): e14508-e14508.
- 122Lee M, Samstein RM, Valero C, Chan TA, Morris LGT. Tumor mutational burden as a predictive biomarker for checkpoint inhibitor immunotherapy. Hum Vaccin Immunother. 2020; 16(1): 112-115.
- 123Marabelle A, Fakih M, Lopez J, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020; 21(10): 1353-1365.
- 124Donehower LA, Soussi T, Korkut A, et al. Integrated analysis of TP53 gene and pathway alterations in the cancer genome atlas. Cell Rep. 2019; 28(11): 3010.
- 125Kastenhuber ER, Lowe SW. Putting p53 in context. Cell. 2017; 170(6): 1062-1078.
- 126Li L, Li M, Wang X. Cancer type-dependent correlations between TP53 mutations and antitumor immunity. DNA Repair (Amst). 2020; 88:102785.
- 127Goodman AM, Kato S, Bazhenova L, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017; 16(11): 2598-2608.
- 128Maeda K, Kang SM, Sawada T, et al. Expression of intercellular adhesion molecule-1 and prognosis in colorectal cancer. Oncol Rep. 2002; 9(3): 511-544.
- 129Fujihara T, Yashiro M, Inoue T, et al. Decrease in ICAM-1 expression on gastric cancer cells is correlated with lymph node metastasis. Gastric Cancer. 1999; 2(4): 221-225.
- 130Ogawa Y, Hirakawa K, Nakata B, et al. Expression of intercellular adhesion molecule-1 in invasive breast cancer reflects low growth potential, negative lymph node involvement, and good prognosis. Clin Cancer Res. 1998; 4(1): 31-36.
- 131Koyama S, Ebihara T, Fukao K. Expression of intercellular adhesion molecule 1 (ICAM-1) during the development of invasion and/or metastasis of gastric carcinoma. J Cancer Res Clin Oncol. 1992; 118(8): 609-614.
- 132Zhang P, Huang Y. Genomic alterations in KMT2 family predict outcome of immune checkpoint therapy in multiple cancers. J Hematol Oncol. 2021; 14(1): 39.
- 133Wang F, Zhao Q, Wang YN, et al. Evaluation of POLE and POLD1 mutations as biomarkers for immunotherapy outcomes across multiple cancer types. JAMA Oncol. 2019; 5(10): 1504-1506.
- 134Reck M, Rodriguez-Abreu D, Robinson AG, et al. Updated analysis of KEYNOTE-024: pembrolizumab versus platinum-based chemotherapy for advanced non-small-cell lung cancer with PD-L1 tumor proportion score of 50% or greater. J Clin Oncol. 2019; 37(7): 537-546.
- 135Sezer A, Kilickap S, Gumus M, et al. Cemiplimab monotherapy for first-line treatment of advanced non-small-cell lung cancer with PD-L1 of at least 50%: a multicentre, open-label, global, phase 3, randomised, controlled trial. Lancet. 2021; 397(10274): 592-604.
- 136Qz S-YML, Yan H-H, Bin G, et al. Sintilimab versus pembrolizumab in monotherapy or combination with chemotherapy as first-line therapy for advanced non–small cell lung cancer: Results from phase 2, randomized clinical trial (CTONG1901).
- 137Sheng X, Chen H, Hu B, et al. Safety, efficacy, and biomarker analysis of toripalimab in patients with previously treated advanced urothelial carcinoma: results from a multicenter phase II trial POLARIS-03. Clin Cancer Res. 2022; 28(3): 489-497.
- 138Zhao M, Liu X, Yuan C, et al. 16P Camrelizumab monotherapy or plus apatinib for PD-L1-positive advanced pulmonary sarcomatoid carcinoma: a single-arm, open-label, multicenter, phase II study. Ann Oncol. 2022; 33: S36-S37.
- 139Li J, Xu Y, Zang A, et al. A phase 2 study of tislelizumab monotherapy in patients with previously treated, locally advanced unresectable ormetastatic microsatellite instability-high/mismatch repair deficient solid tumors. J Clin Oncol. 2021; 39(15): 2569-2569. _suppl.
- 140Spigel D, de Marinis F, Giaccone G, et al. IMpower110: interim overall survival (OS) analysis of a phase III study of atezolizumab (atezo) vs platinum-based chemotherapy (chemo) as first-line (1L) treatment (tx) in PD-L1–selected NSCLC. Ann Oncol. 2019; 30: v915.
- 141Joseph AS, Brahmer JR, Sarkis BA, et al. Safety and clinical activity of first-line durvalumab in advanced NSCLC: updated results from a Phase 1/2 study. J Clin Oncol. 2017; 35(15_suppl):e20504.
- 142Taieb J, Andre T, El Hajbi F, et al. Avelumab versus standard second line treatment chemotherapy in metastatic colorectal cancer patients with microsatellite instability: the SAMCO-PRODIGE 54 randomised phase II trial. Dig Liver Dis. 2021; 53(3): 318-323.
- 143Wu X, Ji J, Lou H, et al. Efficacy and safety of cadonilimab, an anti-PD-1/CTLA4 bi-specific antibody, in previously treated recurrent or metastatic (R/M) cervical cancer: a multicenter, open-label, single-arm, phase II trial (075). Gynecol Oncol. 2022; 166: S47-S48.
- 144Kim JM, Chen DS. Immune escape to PD-L1/PD-1 blockade: seven steps to success (or failure). Ann Oncol. 2016; 27(8): 1492-1504.
- 145Smyth MJ, Ngiow SF, Ribas A, Teng MW. Combination cancer immunotherapies tailored to the tumour microenvironment. Nat Rev Clin Oncol. 2016; 13(3): 143-158.
- 146Kitao H, Iimori M, Kataoka Y, et al. DNA replication stress and cancer chemotherapy. Cancer Sci. 2018; 109(2): 264-271.
- 147Gotwals P, Cameron S, Cipolletta D, et al. Prospects for combining targeted and conventional cancer therapy with immunotherapy. Nat Rev Cancer. 2017; 17(5): 286-301.
- 148Gallego Perez-Larraya J, Garcia-Moure M, Labiano S, et al. Oncolytic DNX-2401 virus for pediatric diffuse intrinsic pontine glioma. N Engl J Med. 2022; 386(26): 2471-2481.
- 149Mathew M, Enzler T, Shu CA, Rizvi NA. Combining chemotherapy with PD-1 blockade in NSCLC. Pharmacol Ther. 2018; 186: 130-137.
- 150Liang H, Lin G, Wang W, et al. Feasibility and safety of PD-1/L1 inhibitors for non-small cell lung cancer in front-line treatment: a Bayesian network meta-analysis. Transl Lung Cancer Res. 2020; 9(2): 188-203.
- 151Zhou Y, Chen C, Zhang X, et al. Immune-checkpoint inhibitor plus chemotherapy versus conventional chemotherapy for first-line treatment in advanced non-small cell lung carcinoma: a systematic review and meta-analysis. J Immunother Cancer. 2018; 6(1): 155.
- 152Langer CJ, Gadgeel SM, Borghaei H, et al. Carboplatin and pemetrexed with or without pembrolizumab for advanced, non-squamous non-small-cell lung cancer: a randomised, phase 2 cohort of the open-label KEYNOTE-021 study. Lancet Oncol. 2016; 17(11): 1497-1508.
- 153Gandhi L, Rodriguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018; 378(22): 2078-2092.
- 154Ho TTB, Nasti A, Seki A, et al. Combination of gemcitabine and anti-PD-1 antibody enhances the anticancer effect of M1 macrophages and the Th1 response in a murine model of pancreatic cancer liver metastasis. J Immunother Cancer. 2020; 8(2):e001367.
- 155Davern M, Donlon NE, Sheppard A, et al. Chemotherapy regimens induce inhibitory immune checkpoint protein expression on stem-like and senescent-like oesophageal adenocarcinoma cells. Transl Oncol. 2021; 14(6):101062.
- 156Di Tacchio M, Macas J, Weissenberger J, et al. Tumor vessel normalization, immunostimulatory reprogramming, and improved survival in glioblastoma with combined inhibition of PD-1, angiopoietin-2, and VEGF. Cancer Immunol Res. 2019; 7(12): 1910-1927.
- 157Gao F, Yang C. Anti-VEGF/VEGFR2 monoclonal antibodies and their combinations with PD-1/PD-L1 inhibitors in clinic. Curr Cancer Drug Targets. 2020; 20(1): 3-18.
- 158Amin A, Plimack ER, Ernstoff MS, et al. Safety and efficacy of nivolumab in combination with sunitinib or pazopanib in advanced or metastatic renal cell carcinoma: the CheckMate 016 study. J Immunother Cancer. 2018; 6(1): 109.
- 159Zhao 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 Res. 2019; 7(4): 630-643.
- 160McDermott 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. Nat Med. 2018; 24(6): 749-757.
- 161Reck M, Mok TSK, Nishio M, et al. Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (IMpower150): key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial. Lancet Respir Med. 2019; 7(5): 387-401.
- 162Sigismund S, Avanzato D, Lanzetti L. Emerging functions of the EGFR in cancer. Mol Oncol. 2018; 12(1): 3-20.
- 163Liu Z, Han C, Dong C, et al. Hypofractionated EGFR tyrosine kinase inhibitor limits tumor relapse through triggering innate and adaptive immunity. Sci Immunol. 2019; 4(38):eaav6473.
- 164Sugiyama E, Togashi Y, Takeuchi Y, et al. Blockade of EGFR improves responsiveness to PD-1 blockade in EGFR-mutated non-small cell lung cancer. Sci Immunol. 2020; 5(43):eaav3937.
- 165Chen DS, Hurwitz H. Combinations of bevacizumab with cancer immunotherapy. Cancer J. 2018; 24(4): 193-204.
- 166Yi C, Chen L, Lin Z, et al. Lenvatinib targets FGF receptor 4 to enhance antitumor immune response of anti-programmed cell death-1 in HCC. Hepatology. 2021; 74(5): 2544-2560.
- 167Ikeda K, Kudo M, Kawazoe S, et al. Phase 2 study of lenvatinib in patients with advanced hepatocellular carcinoma. J Gastroenterol. 2017; 52(4): 512-519.
- 168Makker V, Taylor MH, Aghajanian C, et al. Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer. J Clin Oncol. 2020; 38(26): 2981-2992.
- 169Finn RS, Ikeda M, Zhu AX, et al. Phase ib study of lenvatinib plus pembrolizumab in patients with unresectable hepatocellular carcinoma. J Clin Oncol. 2020; 38(26): 2960-2970.
- 170Arance A, de la Cruz-Merino L, Petrella TM, et al. Phase II LEAP-004 study of lenvatinib plus pembrolizumab for melanoma with confirmed progression on a programmed cell death protein-1 or programmed death ligand 1 inhibitor given as monotherapy or in combination. J Clin Oncol. 2022:JCO2200221.
- 171Wu K, Yi M, Qin S, Chu Q, Zheng X, Wu K. The efficacy and safety of combination of PD-1 and CTLA-4 inhibitors: a meta-analysis. Exp Hematol Oncol. 2019; 8: 26.
- 172Kim SY, Halmos B. Choosing the best first-line therapy: nSCLC with no actionable oncogenic driver. Lung Cancer Manag. 2020; 9(3):LMT36.
- 173Puri S, Shafique M. Combination checkpoint inhibitors for treatment of non-small-cell lung cancer: an update on dual anti-CTLA-4 and anti-PD-1/PD-L1 therapies. Drugs Context. 2020; 9: 2019-2019-2.
- 174Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015; 27(4): 450-461.
- 175Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol. 2016; 39(1): 98-106.
- 176Harjunpaa H, Guillerey C. TIGIT as an emerging immune checkpoint. Clin Exp Immunol. 2020; 200(2): 108-119.
- 177Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci USA. 2010; 107(9): 4275-4280.
- 178Hodi FS, Chesney J, Pavlick AC, et al. Combined nivolumab and ipilimumab versus ipilimumab alone in patients with advanced melanoma: 2-year overall survival outcomes in a multicentre, randomised, controlled, phase 2 trial. Lancet Oncol. 2016; 17(11): 1558-1568.
- 179Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2019; 381(16): 1535-1546.
- 180Hannier S, Tournier M, Bismuth G, Triebel F. CD3/TCR complex-associated lymphocyte activation gene-3 molecules inhibit CD3/TCR signaling. J Immunol. 1998; 161(8): 4058-4065.
- 181Ferris RL, Blumenschein G Jr, Fayette J, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016; 375(19): 1856-1867.
- 182Huang RY, Eppolito C, Lele S, Shrikant P, Matsuzaki J, Odunsi K. LAG3 and PD1 co-inhibitory molecules collaborate to limit CD8+ T cell signaling and dampen antitumor immunity in a murine ovarian cancer model. Oncotarget. 2015; 6(29): 27359-27377.
- 183Goding SR, Wilson KA, Xie Y, et al. Restoring immune function of tumor-specific CD4+ T cells during recurrence of melanoma. J Immunol. 2013; 190(9): 4899-4909.
- 184Sun F, Guo ZS, Gregory AD, Shapiro SD, Xiao G, Qu Z. Dual but not single PD-1 or TIM-3 blockade enhances oncolytic virotherapy in refractory lung cancer. J Immunother Cancer. 2020; 8(1):e000294.
- 185Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med. 2010; 207(10): 2187-2194.
- 186Lim JY, Gerber SA, Murphy SP, Lord EM. Type I interferons induced by radiation therapy mediate recruitment and effector function of CD8(+) T cells. Cancer Immunol Immunother. 2014; 63(3): 259-271.
- 187Wang X, Schoenhals JE, Li A, et al. Suppression of type I IFN signaling in tumors mediates resistance to anti-PD-1 treatment that can be overcome by radiotherapy. Cancer Res. 2017; 77(4): 839-850.
- 188Deng L, Liang H, Burnette B, et al. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. 2014; 124(2): 687-695.
- 189Hwang WL, Pike LRG, Royce TJ, Mahal BA, Loeffler JS. Safety of combining radiotherapy with immune-checkpoint inhibition. Nat Rev Clin Oncol. 2018; 15(8): 477-494.
- 190Lhuillier C, Rudqvist NP, Elemento O, Formenti SC, Demaria S. Radiation therapy and anti-tumor immunity: exposing immunogenic mutations to the immune system. Genome Med. 2019; 11(1): 40.
- 191Howells CC, Stinauer MA, Diot Q, et al. Normal liver tissue density dose response in patients treated with stereotactic body radiation therapy for liver metastases. Int J Radiat Oncol Biol Phys. 2012; 84(3): e441-446.
- 192Timmerman RD, Herman J, Cho LC. Emergence of stereotactic body radiation therapy and its impact on current and future clinical practice. J Clin Oncol. 2014; 32(26): 2847-2854.
- 193Huang Q, Li F, Liu X, et al. Caspase 3-mediated stimulation of tumor cell repopulation during cancer radiotherapy. Nat Med. 2011; 17(7): 860-866.
- 194Altorki NK, McGraw TE, Borczuk AC, et al. Neoadjuvant durvalumab with or without stereotactic body radiotherapy in patients with early-stage non-small-cell lung cancer: a single-centre, randomised phase 2 trial. Lancet Oncol. 2021; 22(6): 824-835.
- 195Ahmed KA, Stallworth DG, Kim Y, et al. Clinical outcomes of melanoma brain metastases treated with stereotactic radiation and anti-PD-1 therapy. Ann Oncol. 2016; 27(3): 434-441.
- 196Garg AD, Agostinis P. Cell death and immunity in cancer: from danger signals to mimicry of pathogen defense responses. Immunol Rev. 2017; 280(1): 126-148.
- 197Cai J, Lin Y, Zhang H, et al. Selective replication of oncolytic virus M1 results in a bystander killing effect that is potentiated by Smac mimetics. Proc Natl Acad Sci USA. 2017; 114(26): 6812-6817.
- 198VanSeggelen H, Tantalo DG, Afsahi A, Hammill JA, Bramson JL. Chimeric antigen receptor-engineered T cells as oncolytic virus carriers. Mol Ther Oncolytics. 2015; 2:15014.
- 199Kaufman HL, Kohlhapp FJ, Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov. 2015; 14(9): 642-662.
- 200Coffin R. Interview with Robert Coffin, inventor of T-VEC: the first oncolytic immunotherapy approved for the treatment of cancer. Immunotherapy. 2016; 8(2): 103-106.
- 201Ribas A, Dummer R, Puzanov I, et al. Oncolytic virotherapy promotes intratumoral t cell infiltration and improves anti-PD-1 immunotherapy. Cell. 2017; 170(6): 1109-1119 e10.
- 202Guo ZS, Lu B, Guo Z, et al. Vaccinia virus-mediated cancer immunotherapy: cancer vaccines and oncolytics. J Immunother Cancer. 2019; 7(1): 6.
- 203Kowalsky SJ, Liu Z, Feist M, et al. Superagonist IL-15-armed oncolytic virus elicits potent antitumor immunity and therapy that are enhanced with PD-1 blockade. Mol Ther. 2018; 26(10): 2476-2486.
- 204Wang N, Wang J, Zhang Z, et al. A novel vaccinia virus enhances anti-tumor efficacy and promotes a long-term anti-tumor response in a murine model of colorectal cancer. Mol Ther Oncolytics. 2021; 20: 71-81.
- 205Liu Z, Ravindranathan R, Kalinski P, Guo ZS, Bartlett DL. Rational combination of oncolytic vaccinia virus and PD-L1 blockade works synergistically to enhance therapeutic efficacy. Nat Commun. 2017; 8:14754.
- 206Lee YS, Lee WS, Kim CW, et al. Oncolytic vaccinia virus reinvigorates peritoneal immunity and cooperates with immune checkpoint inhibitor to suppress peritoneal carcinomatosis in colon cancer. J Immunother Cancer. 2020; 8(2):e000857.
- 207Bommareddy PK, Aspromonte S, Zloza A, Rabkin SD, Kaufman HL. MEK inhibition enhances oncolytic virus immunotherapy through increased tumor cell killing and T cell activation. Sci Transl Med. 2018; 10(471):eaau0417.
- 208Melero I, Gato M, Shekarian T, et al. Repurposing infectious disease vaccines for intratumoral immunotherapy. J Immunother Cancer. 2020; 8(1):e000443.
- 209Shekarian T, Sivado E, Jallas AC, et al. Repurposing rotavirus vaccines for intratumoral immunotherapy can overcome resistance to immune checkpoint blockade. Sci Transl Med. 2019; 11(515):eaat5025.
- 210Shemesh CS, Hsu JC, Hosseini I, et al. Personalized cancer vaccines: clinical landscape, challenges, and opportunities. Mol Ther. 2021; 29(2): 555-570.
- 211Ott PA, Hu Z, Keskin DB, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017; 547(7662): 217-221.
- 212Kong X, Lu P, Liu C, et al. A combination of PD1/PDL1 inhibitors: the prospect of overcoming the weakness of tumor immunotherapy (Review). Mol Med Rep. 2021; 23(5): 362.
- 213Liu R, Zhou C, Wang D, et al. Enhancement of DNA vaccine potency by sandwiching antigen-coding gene between secondary lymphoid tissue chemokine (SLC) and IgG Fc fragment genes. Cancer Biol Ther. 2006; 5(4): 427-434.
- 214Tondini E, Arakelian T, Oosterhuis K, et al. A poly-neoantigen DNA vaccine synergizes with PD-1 blockade to induce T cell-mediated tumor control. Oncoimmunology. 2019; 8(11):1652539.
- 215Xu G, Feng D, Yao Y, et al. Listeria-based hepatocellular carcinoma vaccine facilitates anti-PD-1 therapy by regulating macrophage polarization. Oncogene. 2020; 39(7): 1429-1444.
- 216Weber JS, Kudchadkar RR, Yu B, et al. Safety, efficacy, and biomarkers of nivolumab with vaccine in ipilimumab-refractory or -naive melanoma. J Clin Oncol. 2013; 31(34): 4311-4318.
- 217Gibney GT, Kudchadkar RR, DeConti RC, et al. Safety, correlative markers, and clinical results of adjuvant nivolumab in combination with vaccine in resected high-risk metastatic melanoma. Clin Cancer Res. 2015; 21(4): 712-720.
- 218Crosby EJ, Acharya CR, Haddad AF, et al. Stimulation of oncogene-specific tumor-infiltrating t cells through combined vaccine and alphaPD-1 enable sustained antitumor responses against established HER2 breast cancer. Clin Cancer Res. 2020; 26(17): 4670-4681.
- 219Xia GQ, Lei TR, Yu TB, Zhou PH. Nanocarrier-based activation of necroptotic cell death potentiates cancer immunotherapy. Nanoscale. 2021; 13(2): 1220-1230.
- 220Chawla SP, Van Tine BA, Pollack SM, et al. Phase II randomized study of CMB305 and atezolizumab compared with atezolizumab alone in soft-tissue sarcomas expressing NY-ESO-1. J Clin Oncol. 2022; 40(12): 1291-1300.
- 221Ai L, Xu A, Xu J. Roles of PD-1/PD-L1 pathway: signaling, cancer, and beyond. Adv Exp Med Biol. 2020; 1248: 33-59.
- 222Xu W, Jones M, Liu B, et al. Efficacy and mechanism-of-action of a novel superagonist interleukin-15: interleukin-15 receptor alphaSu/Fc fusion complex in syngeneic murine models of multiple myeloma. Cancer Res. 2013; 73(10): 3075-3086.
- 223Wong HC, Jeng EK, Rhode PR. The IL-15-based superagonist ALT-803 promotes the antigen-independent conversion of memory CD8(+) T cells into innate-like effector cells with antitumor activity. Oncoimmunology. 2013; 2(11):e26442.
- 224Guo Y, Luan L, Patil NK, Sherwood ER. Immunobiology of the IL-15/IL-15Ralpha complex as an antitumor and antiviral agent. Cytokine Growth Factor Rev. 2017; 38: 10-21.
- 225Redman JM, Steinberg SM, Gulley JL. Quick efficacy seeking trial (QuEST1): a novel combination immunotherapy study designed for rapid clinical signal assessment metastatic castration-resistant prostate cancer. J Immunother Cancer. 2018; 6(1): 91.
- 226Wrangle JM, Velcheti V, Patel MR, et al. ALT-803, an IL-15 superagonist, in combination with nivolumab in patients with metastatic non-small cell lung cancer: a non-randomised, open-label, phase 1b trial. Lancet Oncol. 2018; 19(5): 694-704.
- 227Zheng X, Wu Y, Bi J, et al. The use of supercytokines, immunocytokines, engager cytokines, and other synthetic cytokines in immunotherapy. Cell Mol Immunol. 2022; 19(2): 192-209.
- 228Schliemann C, Palumbo A, Zuberbuhler K, et al. Complete eradication of human B-cell lymphoma xenografts using rituximab in combination with the immunocytokine L19-IL2. Blood. 2009; 113(10): 2275-2283.
- 229Wagner K, Schulz P, Scholz A, Wiedenmann B, Menrad A. The targeted immunocytokine L19-IL2 efficiently inhibits the growth of orthotopic pancreatic cancer. Clin Cancer Res. 2008; 14(15): 4951-4960.
- 230Lanyu C, Kaiwen H. Research progress of TCM improving tumor immunosuppressive microenvironment based on programmed death 1/programmed death ligand 1 signaling pathway. Chinese J Inform Tradit Chinese Med. 2020; 27(02): 135-137.
- 231Weihan T, Zhao J, Qian W, Fengming Y, Ran Y, Chuan Z. A study on properties and actions of Chinese medicinals of PD-1/PD-L1 inhibitor. New Chinese Med. 2020; 52(12): 206-208.
- 232Hongzhi C, Weilin L, Zhan G, Huiqing Z. Etiology and pathogeny analysis and treatment exploration in traditional Chinese medicine of immune-related adverse reactions of PD-1/1 inhibitors. World Chinese Med. 2021; 16(09): 1386-1390+1399.
- 233Nanyan W, Cuihua J, Meng G, Jian Z, Zhiqi Y, Ke P. Paeoniflorin inhibits programmed cell death-1-ligand 1 expression in HepG2 cells by regulating JAK/STAT3 signal pathway. J China Pharm Univ. 2019; 50(02): 213-221.
- 234Liu Y, Liu X, Zhang N, et al. Berberine diminishes cancer cell PD-L1 expression and facilitates antitumor immunity via inhibiting the deubiquitination activity of CSN5. Acta Pharm Sin B. 2020; 10(12): 2299-2312.
- 235Qing A, Li-wei H, Hong-yan W, et al. Mechanismof cordycepin promotes tumor immunity by regulating PD-1 receptor of CD4 T lymphocytes+. J Nanjing Univ Tradit Chinese Med. 2018; 34(05): 495-498.
- 236Xu H, Van der Jeught K, Zhou Z, et al. Atractylenolide I enhances responsiveness to immune checkpoint blockade therapy by activating tumor antigen presentation. J Clin Invest. 2021; 131(10):e146832.
- 237Yazhen Z, Xianlei L, Jinhua L, Shengyou L. Study on Guilu Erxianjiao in inhibiting tumor growth of H22 tumor-bearing mice by regulating PD-1 expression. China Modern Doctor. 2018; 56(36): 1-5.
- 238Lv J, Jia Y, Li J, et al. Gegen Qinlian decoction enhances the effect of PD-1 blockade in colorectal cancer with microsatellite stability by remodelling the gut microbiota and the tumour microenvironment. Cell Death Dis. 2019; 10(6): 415.
- 239Vetizou M, Trinchieri G. Anti-PD1 in the wonder-gut-land. Cell Res. 2018; 28(3): 263-264.
- 240Andrews MC, Vasanthakumar A. Gut microbiota - a double-edged sword in cancer immunotherapy. Trends Cancer. 2023; 9(1): 3-5.
- 241Yi M, Jiao D, Qin S, Chu Q, Li A, Wu K. Manipulating gut microbiota composition to enhance the therapeutic effect of cancer immunotherapy. Integr Cancer Ther. 2019; 18:1534735419876351.
- 242Montalban-Arques A, Katkeviciute E, Busenhart P, et al. Commensal Clostridiales strains mediate effective anti-cancer immune response against solid tumors. Cell Host Microbe. 2021; 29(10): 1573-1588 e7.
- 243Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015; 350(6264): 1084-1089.
- 244Xiao-yu Z, Jie L. Discussion on traditional Chinese medicine promoting the transformation of ‘cold' tumor to ‘hot' based on yin-yang theory. China J Tradit Chinese Med Pharmacy. 2022; 37(08): 4356-4359.
- 245Li X, Shao C, Shi Y, Han W. Lessons learned from the blockade of immune checkpoints in cancer immunotherapy. J Hematol Oncol. 2018; 11(1): 31.
- 246Jiansheng L, Fuchun S. Explaination to apoptosis of tumor cells by the theory of Yin and Yang of Chinese medicine. Liaoning J Tradit Chinese Med. 2010; 37(06): 1034-1035.
- 247Shumin Y, Zhenye X, Haibin D, Xiaozhen Z. Mechanism and advantages of traditional Chinese medicine in remodeling tumor microenvironment. Acad J Shanghai Univ Tradit Chinese Med. 2019; 33(05): 1-7.
- 248Qing S, Qing J, Qi L. Research on the clinical and mechanism of traditional Chinese medicine regulating tumor immunity. China J Tradit Chinese Med Pharmacy. 2018; 33(10): 4542-4545.
- 249Chen Q, Sun T, Jiang C. Recent advancements in nanomedicine for ‘cold’ tumor immunotherapy. Nanomicro Lett. 2021; 13(1): 92.
- 250Liu YT, Sun ZJ. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics. 2021; 11(11): 5365-5386.
- 251Giustarini G, Pavesi A, Adriani G. Nanoparticle-based therapies for turning cold tumors hot: how to treat an immunosuppressive tumor microenvironment. Front Bioeng Biotechnol. 2021; 9:689245.
- 252Jeon MJ, Lee H, Lee J, et al. Development of potent immune modulators targeting stimulator of interferon genes receptor. J Med Chem. 2022; 65(7): 5407-5432.
- 253Xiong W, Gao X, Zhang T, et al. USP8 inhibition reshapes an inflamed tumor microenvironment that potentiates the immunotherapy. Nat Commun. 2022; 13(1): 1700.
- 254Sun R, Limkin EJ, Vakalopoulou M, et al. A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective multicohort study. Lancet Oncol. 2018; 19(9): 1180-1191.
- 255Lu S, Stein JE, Rimm DL, et al. Comparison of biomarker modalities for predicting response to PD-1/PD-L1 checkpoint blockade: a systematic review and meta-analysis. JAMA Oncol. 2019; 5(8): 1195-1204.
- 256Fu Y, Jung AW, Torne RV, et al. Pan-cancer computational histopathology reveals mutations, tumor composition and prognosis. Nat Cancer. 2020; 1(8): 800-810.
- 257Wu TD, Madireddi S, de Almeida PE, et al. Peripheral T cell expansion predicts tumour infiltration and clinical response. Nature. 2020; 579(7798): 274-278.
- 258Pelly VS, Moeini A, Roelofsen LM, et al. Anti-inflammatory drugs remodel the tumor immune environment to enhance immune checkpoint blockade efficacy. Cancer Discov. 2021; 11(10): 2602-2619.