REFERENCES

1. Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med 2010;363:1938-48.

2. Curigliano G, Goldhirsch A. The triple-negative subtype: new ideas for the poorest prognosis breast cancer. J Natl Cancer Inst Monogr 2011;2011:108-10.

3. Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 2012;490:61-70.

4. Penault-Llorca F, Viale G. Pathological and molecular diagnosis of triple-negative breast cancer: a clinical perspective. Ann Oncol 2012;23 Suppl 6:vi19-22.

5. Papadimitriou M, Mountzios G, Papadimitriou CA. The role of PARP inhibition in triple-negative breast cancer: unraveling the wide spectrum of synthetic lethality. Cancer Treat Rev 2018;67:34-44.

6. Dent R, Trudeau M, Pritchard KI, et al. Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res 2007;13:4429-34.

7. Li X, Yang J, Peng L, et al. Triple-negative breast cancer has worse overall syrvival and cause-specific survival than non-triple-negative breast cancer. Breast Cancer Res Treat 2017;161:279-87.

8. Narang P, Chen M, Sharma AA, Anderson KS, Wilson MA. The neoepitope landscape of breast cancer: implications for immunotherapy. BMC Cancer 2019;19:200.

9. Kim I, Sanchez K, Mcarthur HL, Page D. Immunotherapy in triple-negative breast cancer: present and future. Curr Breast Cancer Rep 2019;11:259-71.

10. Lehmann BD, Bauer JA, Chen X, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest 2011;121:2750-67.

11. Burstein MD, Tsimelzon A, Poage GM, et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin Cancer Res 2015;21:1688-98.

12. Jézéquel P, Loussouarn D, Guérin-Charbonnel C, et al. Gene-expression molecular subtyping of triple-negative breast cancer tumours: importance of immune response. Breast Cancer Res 2015;17:43.

13. Ahn SG, Kim SJ, Kim C, Jeong J. Molecular classification of triple-negative breast cancer. J Breast Cancer 2016;19:223-30.

14. Gonzalez-Angulo AM, Timms KM, Liu S, et al. Incidence and outcome of BRCA mutations in unselected patients with triple receptor-negative breast cancer. Clin Cancer Res 2011;17:1082-9.

15. Hartman AR, Kaldate RR, Sailer LM, et al. Prevalence of BRCA mutations in an unselected population of triple-negative breast cancer. Cancer 2012;118:2787-95.

16. Sharma P, Klemp JR, Kimler BF, et al. Germline BRCA mutation evaluation in a prospective triple-negative breast cancer registry: implications for hereditary breast and/or ovarian cancer syndrome testing. Breast Cancer Res Treat 2014;145:707-14.

17. Couch FJ, Hart SN, Sharma P, et al. Inherited mutations in 17 breast cancer susceptibility genes among a large triple-negative breast cancer cohort unselected for family history of breast cancer. J Clin Oncol 2015;33:304-11.

18. Pellegrino B, Musolino A, Llop-Guevara A, et al. Homologous recombination repair deficiency and the immune response in breast cancer: a literature review. Transl Oncol 2020;13:410-22.

19. Wen WX, Leong CO. Association of BRCA1- and BRCA2-deficiency with mutation burden, expression of PD-L1/PD-1, immune infiltrates, and T cell-inflamed signature in breast cancer. PLoS One 2019;14:e0215381.

20. Nik-Zainal S, Davies H, Staaf J, et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature 2016;534:47-54.

21. Budczies J, Bockmayr M, Denkert C, et al. Classical pathology and mutational load of breast cancer - integration of two worlds. J Pathol Clin Res 2015;1:225-38.

22. Luen S, Virassamy B, Savas P, Salgado R, Loi S. The genomic landscape of breast cancer and its interaction with host immunity. Breast 2016;29:241-50.

23. Samstein RM, Lee CH, Shoushtari AN, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 2019;51:202-6.

24. Goodman 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;16:2598-608.

25. Loi S, Michiels S, Salgado R, et al. Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial. Ann Oncol 2014;25:1544-50.

26. Salgado R, Denkert C, Demaria S, et al. International TILs Working Group 2014. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol 2015;26:259-71.

27. Denkert C, von Minckwitz G, Darb-esfahani S, et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol 2018;19:40-50.

28. Stanton SE, Adams S, Disis ML. Variation in the incidence and magnitude of tumor-infiltrating lymphocytes in breast cancer subtypes: a systematic review. JAMA Oncol 2016;2:1354-60.

29. Beckers RK, Selinger CI, Vilain R, et al. Programmed death ligand 1 expression in triple-negative breast cancer is associated with tumour-infiltrating lymphocytes and improved outcome. Histopathology 2016;69:25-34.

30. Mittendorf EA, Philips AV, Meric-Bernstam F, et al. PD-L1 expression in triple-negative breast cancer. Cancer Immunol Res 2014;2:361-70.

31. U.S. Food & Drug. FDA approves atezolizumab for PD-L1 positive unresectable locally advanced or metastatic triple-negative breast cancer. Available from: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-atezolizumab-pd-l1-positive-unresectable-locally-advanced-or-metastatic-triple-negative [Last accessed on 6 Aug 2021].

32. Vivier E, Artis D, Colonna M, et al. Innate lymphoid cells: 10 Years On. Cell 2018;174:1054-66.

33. Demaria O, Cornen S, Daëron M, Morel Y, Medzhitov R, Vivier E. Harnessing innate immunity in cancer therapy. Nature 2019;574:45-56.

34. Bonilla FA, Oettgen HC. Adaptive immunity. J Allergy Clin Immunol 2010;125:S33-40.

35. Emens LA. Breast cancer immunobiology driving immunotherapy: vaccines and immune checkpoint blockade. Expert Rev Anticancer Ther 2012;12:1597-611.

36. Emens LA. Breast cancer immunotherapy: facts and hopes. Clin Cancer Res 2018;24:511-20.

37. Smyth MJ, Dunn GP, Schreiber RD. Cancer immunosurveillance and immunoediting: the roles of immunity in suppressing tumor development and shaping tumor immunogenicity. Adv Immunol 2006;90:1-50.

38. DeNardo DG, Coussens LM. Inflammation and breast cancer. Balancing immune response: crosstalk between adaptive and innate immune cells during breast cancer progression. Breast Cancer Res 2007;9:212.

39. Coussens LM, Pollard JW. Leukocytes in mammary development and cancer. Cold Spring Harb Perspect Biol 2011;3:a003285.

40. DeNardo DG, Barreto JB, Andreu P, et al. CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell 2009;16:91-102.

41. Ribas A. Adaptive immune resistance: how cancer protects from immune attack. Cancer Discov 2015;5:915-9.

42. Allard B, Beavis PA, Darcy PK, Stagg J. Immunosuppressive activities of adenosine in cancer. Curr Opin Pharmacol 2016;29:7-16.

43. Ho PC, Kaech SM. Reenergizing T cell anti-tumor immunity by harnessing immunometabolic checkpoints and machineries. Curr Opin Immunol 2017;46:38-44.

44. Zhang H, Chen J. Current status and future directions of cancer immunotherapy. J Cancer 2018;9:1773-81.

45. Munhoz RR, Postow MA. Recent advances in understanding antitumor immunity. F1000Res 2016;5:2545.

46. Dustin ML. Cancer immunotherapy: killers on sterols. Nature 2016;531:583-4.

47. Park J, Kwon M, Shin EC. Immune checkpoint inhibitors for cancer treatment. Arch Pharm Res 2016;39:1577-87.

48. Khalil DN, Smith EL, Brentjens RJ, Wolchok JD. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol 2016;13:273-90.

49. of cancer immunotherapy from the bench to the bedside. Adv Cancer Res 2019;143:1-62.

50. Mir MA. . Chapter 1. Introduction to costimulation and costimulatory molecules. In: Mir MA, Editor. Developing costimulatory molecules for immunotherapy of diseases. Cambridge, Massachusetts: Academic Press; 2015. p. 1-43.

51. Esfahani K, Roudaia L, Buhlaiga N, Del Rincon SV, Papneja N, Miller WH Jr. A review of cancer immunotherapy: from the past, to the present, to the future. Curr Oncol 2020;27:S87-97.

52. Adams S, Gatti-Mays ME, Kalinsky K, et al. Current landscape of immunotherapy in breast cancer: a review. JAMA Oncol 2019;5:1205-14.

53. Mina LA, Lim S, Bahadur SW, Firoz AT. Immunotherapy for the treatment of breast cancer: emerging new data. Breast Cancer (Dove Med Press) 2019;11:321-8.

54. Nanda R, Chow LQ, Dees EC, et al. Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 study. J Clin Oncol 2016;34:2460-7.

55. Dirix LY, Takacs I, Jerusalem G, et al. Avelumab, an anti-PD-L1 antibody, in patients with locally advanced or metastatic breast cancer: a phase 1b JAVELIN solid tumor study. Breast Cancer Res Treat 2018;167:671-86.

56. Emens LA, Cruz C, Eder JP, et al. Long-term clinical outcomes and biomarker analyses of atezolizumab therapy for patients with metastatic triple-negative breast cancer: a phase 1 study. JAMA Oncol 2019;5:74-82.

57. Adams S, Loi S, Toppmeyer DL, et al. KEYNOTE-086 cohort B: pembrolizumab monotherapy for PD-L1-positive, previously untreated, metastatic triple-negative breast cancer (mTNBC). Cancer Res 2018;78(4 Suppl):abstract nr PD6-10.

58. Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol 2017;17:97-111.

59. Garg AD, More S, Rufo N, et al. Trial watch: immunogenic cell death induction by anticancer chemotherapeutics. Oncoimmunology 2017;6:e1386829.

60. Alegre ML, Frauwirth KA, Thompson CB. T-cell regulation by CD28 and CTLA-4. Nat Rev Immunol 2001;1:220-8.

61. der Merwe PA, Dushek O. Mechanisms for T cell receptor triggering. Nat Rev Immunol 2011;11:47-55.

62. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science 2018;359:1350-5.

63. Chambers CA, Kuhns MS, Egen JG, Allison JP. CTLA-4-mediated inhibition in regulation of T cell responses: mechanisms and manipulation in tumor immunotherapy. Annu Rev Immunol 2001;19:565-94.

64. Egen JG, Kuhns MS, Allison JP. CTLA-4: new insights into its biological function and use in tumor immunotherapy. Nat Immunol 2002;3:611-8.

65. Parry RV, Chemnitz JM, Frauwirth KA, et al. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol Cell Biol 2005;25:9543-53.

66. Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol 2016;39:98-106.

67. Linsley PS, Bradshaw J, Greene J, Peach R, Bennett KL, Mittler RS. Intracellular trafficking of CTLA-4 and focal localization towards sites of TCR engagement. Immunity 1996;4:535-43.

68. Krummel MF, Allison JP. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med 1995;182:459-65.

69. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996;271:1734-6.

70. Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711-23.

71. European Medicines Agency. Yervoy. Available from: https://www.ema.europa.eu/en/medicines/human/EPAR/yervoy [Last accessed on 6 Aug 2021].

72. Kleef R, Moss R, Szasz AM, Bohdjalian A, Bojar H, Bakacs T. Complete clinical remission of stage IV triple-negative breast cancer lung metastasis administering low-dose immune checkpoint blockade in combination with hyperthermia and interleukin-2. Integr Cancer Ther 2018;17:1297-303.

73. McArthur HL, Diab A, Page DB, et al. A pilot study of preoperative single-dose ipilimumab and/or cryoablation in women with early-stage breast cancer with comprehensive immune profiling. Clin Cancer Res 2016;22:5729-37.

74. McArthur H, Comen E, Bryce Y, et al. A randomized phase 2 study of peri-operative ipilimumab, nivolumab and cryoablation versus standard care in women with residual, early stage/resectable, triple negative breast cancer after standard-of-care neoadjuvant chemotherapy. Cancer Res 2020;80(4 Suppl):abstract nr OT1-01.

75. Australian New Zealand Clinical Trials Registry. Available from: https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=367255 [Last accessed on 6 Aug 2021].

76. Barroso-sousa R, Trippa L, Lange P, et al. Nimbus: a phase II study of nivolumab plus ipilimumab in metastatic hypermutated HER2-negative breast cancer. J Clin Oncol 2019;37:TPS1115.

77. Hanson DC, Canniff PC, Primiano MJ, et al. Preclinical in vitro characterization of anti-CTLA4 therapeutic antibody CP-675,206. Abstract 3802. Cancer Res 2004;64(7 Supplement):877.

78. Canniff PC, Donovan CB, Burkwit JJ, et al. CP-675,206 anti-CTLA4 antibody clinical candidate enhances IL-2 production in cancer patient T cells in vitro regardless of tumor type or stage of disease. Cancer Res 2004;64(7 Supplement):164.

79. Comin-Anduix B, Escuin-Ordinas H, Ibarrondo FJ. Tremelimumab: research and clinical development. Onco Targets Ther 2016;9:1767-76.

80. He M, Chai Y, Qi J, et al. Remarkably similar CTLA-4 binding properties of therapeutic ipilimumab and tremelimumab antibodies. Oncotarget 2017;8:67129-39.

81. Ribas A, Camacho LH, Lopez-Berestein G, et al. Antitumor activity in melanoma and anti-self responses in a phase I trial with the anti-cytotoxic T lymphocyte-associated antigen 4 monoclonal antibody CP-675,206. J Clin Oncol 2005;23:8968-77.

82. Camacho LH, Antonia S, Sosman J, et al. Phase I/II trial of tremelimumab in patients with metastatic melanoma. J Clin Oncol 2009;27:1075-81.

83. Kirkwood JM, Lorigan P, Hersey P, et al. Phase II trial of tremelimumab (CP-675,206) in patients with advanced refractory or relapsed melanoma. Clin Cancer Res 2010;16:1042-8.

84. Ribas A, Kefford R, Marshall MA, et al. Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J Clin Oncol 2013;31:616-22.

85. Santa-Maria CA, Kato T, Park JH, et al. A pilot study of durvalumab and tremelimumab and immunogenomic dynamics in metastatic breast cancer. Oncotarget 2018;9:18985-96.

86. Reits EA, Hodge JW, Herberts CA, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med 2006;203:1259-71.

87. Sharabi AB, Nirschl CJ, Kochel CM, et al. Stereotactic radiation therapy augments antigen-specific PD-1-mediated antitumor immune responses via cross-presentation of tumor antigen. Cancer Immunol Res 2015;3:345-55.

88. Lugade AA, Moran JP, Gerber SA, Rose RC, Frelinger JG, Lord EM. Local radiation therapy of B16 melanoma tumors increases the generation of tumor antigen-specific effector cells that traffic to the tumor. J Immunol 2005;174:7516-23.

89. Chakraborty M, Abrams SI, Camphausen K, et al. Irradiation of tumor cells up-regulates Fas and enhances CTL lytic activity and CTL adoptive immunotherapy. J Immunol 2003;170:6338-47.

90. Matsumura S, Wang B, Kawashima N, et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J Immunol 2008;181:3099-107.

91. Jiang DM, Fyles A, Nguyen LT, et al. Phase I study of local radiation and tremelimumab in patients with inoperable locally recurrent or metastatic breast cancer. Oncotarget 2019;10:2947-58.

92. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 1992;11:3887-95.

93. Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 1999;11:141-51.

94. Seidel JA, Otsuka A, Kabashima K. Anti-PD-1 and anti-CTLA-4 therapies in cancer: mechanisms of action, efficacy, and limitations. Front Oncol 2018;8:86.

95. Agata Y, Kawasaki A, Nishimura H, et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol 1996;8:765-72.

96. Kinter AL, Godbout EJ, McNally JP, et al. The common gamma-chain cytokines IL-2, IL-7, IL-15, and IL-21 induce the expression of programmed death-1 and its ligands. J Immunol 2008;181:6738-46.

97. Thibult ML, Mamessier E, Gertner-Dardenne J, et al. PD-1 is a novel regulator of human B-cell activation. Int Immunol 2013;25:129-37.

98. Lim TS, Chew V, Sieow JL, et al. PD-1 expression on dendritic cells suppresses CD8+ T cell function and antitumor immunity. Oncoimmunology 2016;5:e1085146.

99. Dong H, Zhu G, Tamada K, Chen L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med 1999;5:1365-9.

100. Sugiura D, Maruhashi T, Okazaki IM, et al. Restriction of PD-1 function by cis-PD-L1/CD80 interactions is required for optimal T cell responses. Science 2019;364:558-66.

101. Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol 2004;173:945-54.

102. Sheppard KA, Fitz LJ, Lee JM, et al. PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3zeta signalosome and downstream signaling to PKCtheta. FEBS Lett 2004;574:37-41.

103. Patsoukis N, Brown J, Petkova V, Liu F, Li L, Boussiotis VA. Selective effects of PD-1 on Akt and Ras pathways regulate molecular components of the cell cycle and inhibit T cell proliferation. Sci Signal 2012;5:ra46.

104. Hui E, Cheung J, Zhu J, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 2017;355:1428-33.

105. Carter L, Fouser LA, Jussif J, et al. PD-1:PD-L inhibitory pathway affects both CD4+ and CD8+ T cells and is overcome by IL-2. Eur J Immunol 2002;32:634-43.

106. Nurieva R, Thomas S, Nguyen T, et al. T-cell tolerance or function is determined by combinatorial costimulatory signals. EMBO J 2006;25:2623-33.

107. Freeman GJ, Long AJ, Iwai Y, et al. Engagement of the Pd-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 2000;192:1027-34.

108. Lee JY, Lee HT, Shin W, et al. Structural basis of checkpoint blockade by monoclonal antibodies in cancer immunotherapy. Nat Commun 2016;7:13354.

109. Callahan MK, Postow MA, Wolchok JD. Targeting T cell co-receptors for cancer therapy. Immunity 2016;44:1069-78.

110. Highlights of prescribing information. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/125554s078lbl.pdf [Last accessed on 6 Aug 2021].

111. Khoury Κ, Isaacs C, Gatti-Mays ME, et al. Nivolumab or capecitabine or combination therapy as adjuvant therapy for triple negative breast cancer (TNBC) with residual disease following neoadjuvant chemotherapy: the OXEL study. Cancer Res 2019;79(4 Supplement):abstract OT3-04.

112. Masuda N, Lee SJ, Ohtani S, et al. Adjuvant capecitabine for breast cancer after preoperative chemotherapy. N Engl J Med 2017;376:2147-59.

113. Clinical Trial. Pre-operative phase II trial for breast cancer with nivolumab in combination with novel IO (BELLINI Trial). Available from: https://clinicaltrials.gov/ct2/show/NCT03815890 [Last accessed on 6 Aug 2021].

114. Waterhouse D, Gutierrez M, Bekaii-Saab T, et al. Nab-paclitaxel (nab-P) plus nivolumab (Nivo) in human epidermal growth factor receptor 2 (HER2)-negative recurrent metastatic breast cancer (MBC). Cancer Res 2016;76(4 Supplement):abstract OT1-01.

115. Garrido-Castro AC, Barry WT, Traina TA, et al. A randomized phase II trial of carboplatin with or without nivolumab in first- or second-line metastatic TNBC. J Clin Oncol 2018;36:TPS1118.

116. Ozaki Y, Mukohara T, Tsurutani J, et al. A multicenter phase II study evaluating the efficacy of nivolumab plus paclitaxel plus bevacizumab triple-combination therapy as a first-line treatment in patients with HER2-negative metastatic breast cancer: WJOG9917B NEWBEAT trial. Cancer Res 2020;80(4 Suppl):abstract PD1-03.

117. Ozaki Y, Kitano S, Matsumoto K, et al. Biomarker study of patients with HER2-negative metastatic breast cancer receiving combination therapy with nivolumab, bevacizumab and paclitaxel as first-line treatment (WJOG9917BTR). Cancer Res 2019;79(4 Supplement):abstract OT1-12.

118. Kok M, Voorwerk L, Horlings H, et al. Adaptive phase II randomized trial of nivolumab after induction treatment in triple negative breast cancer (TONIC trial): final response data stage I and first translational data. J Clin Oncol 2018;36:1012.

119. Voorwerk L, Slagter M, Horlings HM, et al. Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the TONIC trial. Nat Med 2019;25:920-8.

120. Demaria S, Ng B, Devitt ML, et al. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys 2004;58:862-70.

121. Biasi AR, Villena-Vargas J, Adusumilli PS. Cisplatin-induced antitumor immunomodulation: a review of preclinical and clinical evidence. Clin Cancer Res 2014;20:5384-91.

122. Scurr M, Pembroke T, Bloom A, et al. Low-dose cyclophosphamide induces antitumor T-cell responses, which associate with survival in metastatic colorectal cancer. Clin Cancer Res 2017;23:6771-80.

123. Seymour L, Bogaerts J, Perrone A, et al. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol 2017;18:e143-52.

124. Hutloff A, Dittrich AM, Beier KC, et al. ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature 1999;397:263-6.

125. Yoshinaga SK, Whoriskey JS, Khare SD, et al. T-cell co-stimulation through B7RP-1 and ICOS. Nature 1999;402:827-32.

126. Dong C, Juedes AE, Temann UA, et al. ICOS co-stimulatory receptor is essential for T-cell activation and function. Nature 2001;409:97-101.

127. Rudd CE, Schneider H. Unifying concepts in CD28, ICOS and CTLA4 co-receptor signalling. Nat Rev Immunol 2003;3:544-56.

128. Wei SC, Levine JH, Cogdill AP, et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell 2017;170:1120-33.e17.

129. Amatore F, Gorvel L, Olive D. Inducible co-stimulator (ICOS) as a potential therapeutic target for anti-cancer therapy. Expert Opin Ther Targets 2018;22:343-51.

130. Fu T, He Q, Sharma P. The ICOS/ICOSL pathway is required for optimal antitumor responses mediated by anti-CTLA-4 therapy. Cancer Res 2011;71:5445-54.

131. Fan X, Quezada SA, Sepulveda MA, Sharma P, Allison JP. Engagement of the ICOS pathway markedly enhances efficacy of CTLA-4 blockade in cancer immunotherapy. J Exp Med 2014;211:715-25.

132. Clinical Trial. Phase 1/2 multicenter trial of ICOS agonist monoclonal antibody (mAb) JTX-2011 alone and in combination with nivolumab, ipilimumab, or pembrolizumab in adult subjects with advanced and/or refractory solid tumor malignancies. Available from: https://clinicaltrials.gov/ct2/show/NCT02904226 [Last accessed on 6 Aug 2021].

133. Yap TA, Burris HA, Kummar S, et al. ICONIC: biologic and clinical activity of first in class ICOS agonist antibody JTX-2011 +/- nivolumab (nivo) in patients (pts) with advanced cancers. J Clin Oncol 2018;36:3000.

134. Barroso-sousa R, Guo H, Barry WT, et al. A phase II study of nivolumab in combination with cabozantinib for metastatic triple-negative breast cancer (mTNBC). JCO 2018;36:TPS1119.

135. Yakes FM, Chen J, Tan J, et al. Cabozantinib (XL184), a novel MET and VEGFR2 inhibitor, simultaneously suppresses metastasis, angiogenesis, and tumor growth. Mol Cancer Ther 2011;10:2298-308.

136. Grüllich C. Cabozantinib: multi-kinase inhibitor of MET, AXL, RET, and VEGFR2. Recent Results Cancer Res 2018;211:67-75.

137. Markowitz JN, Fancher KM. Cabozantinib: a multitargeted oral tyrosine kinase inhibitor. Pharmacotherapy 2018;38:357-69.

138. Desai A, Small EJ. Treatment of advanced renal cell carcinoma patients with cabozantinib, an oral multityrosine kinase inhibitor of MET, AXL and VEGF receptors. Future Oncol 2019;15:2337-48.

139. Fogli S, Porta C, Del Re M, et al. Optimizing treatment of renal cell carcinoma with VEGFR-TKIs: a comparison of clinical pharmacology and drug-drug interactions of anti-angiogenic drugs. Cancer Treat Rev 2020;84:101966.

140. Liao W, Lin JX, Leonard WJ. IL-2 family cytokines: new insights into the complex roles of IL-2 as a broad regulator of T helper cell differentiation. Curr Opin Immunol 2011;23:598-604.

141. Boyman O, Sprent J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol 2012;12:180-90.

142. Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 1999;17:2105-16.

143. Rosenberg SA, Yang JC, Topalian SL, et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. JAMA 1994;271:907-13.

144. McDermott DF, Regan MM, Clark JI, et al. Randomized phase III trial of high-dose interleukin-2 versus subcutaneous interleukin-2 and interferon in patients with metastatic renal cell carcinoma. J Clin Oncol 2005;23:133-41.

145. Payne R, Glenn L, Hoen H, et al. Durable responses and reversible toxicity of high-dose interleukin-2 treatment of melanoma and renal cancer in a community hospital biotherapy program. J Immunother Cancer 2014;2:13.

146. Charych D, Khalili S, Dixit V, et al. Modeling the receptor pharmacology, pharmacokinetics, and pharmacodynamics of NKTR-214, a kinetically-controlled interleukin-2 (IL2) receptor agonist for cancer immunotherapy. PLoS One 2017;12:e0179431.

147. NEKTAR. Bempegaldesleukin (NKTR-214). Available from: https://www.nektar.com/pipeline/rd-pipeline/nktr-214 [Last accessed on 6 Aug 2021].

148. Tolaney S, Baldini C, Spira A, et al. . Clinical activity of BEMPEG plus NIVO observed in metastatic TNBC: preliminary results from the TNBC cohort of the Ph1/2 PIVOT-02 study. CRI-CIMT-EATI-AACR, 5th International Cancer Immunotherapy Conference. Paris: Abstracts Book; 2019. p. 2.

149. Mosser DM, Zhang X. Interleukin-10: new perspectives on an old cytokine. Immunol Rev 2008;226:205-18.

150. Saraiva M, O'Garra A. The regulation of IL-10 production by immune cells. Nat Rev Immunol 2010;10:170-81.

151. Bedke T, Muscate F, Soukou S, Gagliani N, Huber S. Title: IL-10-producing T cells and their dual functions. Semin Immunol 2019;44:101335.

152. Zheng LM, Ojcius DM, Garaud F, et al. Interleukin-10 inhibits tumor metastasis through an NK cell-dependent mechanism. J Exp Med 1996;184:579-84.

153. Groux H, Cottrez F, Rouleau M, Mauze S, Antonenko S, et al. A transgenic model to analyze the immunoregulatory role of IL-10 secreted by antigen-presenting cells. J Immunol 1999;162:1723-9.

154. Fujii S, Shimizu K, Shimizu T, Lotze MT. Interleukin-10 promotes the maintenance of antitumor CD8(+) T-cell effector function in situ. Blood 2001;98:2143-51.

155. Emmerich J, Mumm JB, Chan IH, et al. IL-10 directly activates and expands tumor-resident CD8(+) T cells without de novo infiltration from secondary lymphoid organs. Cancer Res 2012;72:3570-81.

156. Asadullah K, Sterry W, Stephanek K, et al. IL-10 is a key cytokine in psoriasis. Proof of principle by IL-10 therapy: a new therapeutic approach. J Clin Invest 1998;101:783-94.

157. Duncan SA, Dixit S, Sahu R, et al. Prolonged release and functionality of interleukin-10 encapsulated within PLA-PEG nanoparticles. Nanomaterials (Basel) 2019;9:1074.

158. Naing A, Papadopoulos KP, Autio KA, et al. Safety, Antitumor activity, and immune activation of pegylated recombinant human interleukin-10 (AM0010) in patients with advanced solid tumors. J Clin Oncol 2016;34:3562-9.

159. Naing A, Infante JR, Papadopoulos KP, et al. PEGylated IL-10 (Pegilodecakin) induces systemic immune activation, CD8+ T cell invigoration and polyclonal T cell expansion in cancer patients. Cancer Cell 2018;34:775-791.e3.

160. Naing A, Wong DJ, Infante JR, et al. Pegilodecakin combined with pembrolizumab or nivolumab for patients with advanced solid tumours (IVY): a multicentre, multicohort, open-label, phase 1b trial. Lancet Oncol 2019;20:1.

161. Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015;348(6230):56-61.

162. Lee HT, Lee SH, Heo YS. Molecular interactions of antibody drugs targeting PD-1, PD-L1, and CTLA-4 in immuno-oncology. Molecules 2019;24:1190.

163. Highlights of prescribing information. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/125514s040lbl.pdf [Last accessed on 6 Aug 2021].

164. European Medicines Agency. Keytruda. Available from: https://www.ema.europa.eu/en/medicines/human/EPAR/keytruda [Last accessed on 6 Aug 2021].

165. Schmid P, Salgado R, Park YH, et al. Pembrolizumab plus chemotherapy as neoadjuvant treatment of high-risk, early-stage triple-negative breast cancer: results from the phase 1b open-label, multicohort KEYNOTE-173 study. Ann Oncol 2020;31:569-81.

166. von Minckwitz G, Schneeweiss A, Loibl S, et al. Neoadjuvant carboplatin in patients with triple-negative and HER2-positive early breast cancer (GeparSixto; GBG 66): a randomised phase 2 trial. Lancet Oncol 2014;15:747-56.

167. Sikov WM, Berry DA, Perou CM, et al. Impact of the addition of carboplatin and/or bevacizumab to neoadjuvant once-per-week paclitaxel followed by dose-dense doxorubicin and cyclophosphamide on pathologic complete response rates in stage II to III triple-negative breast cancer: CALGB 40603 (Alliance). J Clin Oncol 2015;33:13-21.

168. Cortazar P, Zhang L, Untch M, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet 2014;384:164-72.

169. Nanda R, Liu MC, Yau C, et al. Effect of pembrolizumab plus neoadjuvant chemotherapy on pathologic complete response in women with early-stage breast cancer: an analysis of the ongoing phase 2 adaptively randomized I-SPY2 trial. JAMA Oncol 2020;6:676-84.

170. Schmid P, Cortes J, Pusztai L, et al. KEYNOTE-522 Investigators. Pembrolizumab for early triple-negative breast cancer. N Engl J Med 2020;382:810-21.

171. Clinical Trial. Neoadjuvant phase II study of pembrolizumab and carboplatin plus docetaxel in triple negative breast cancer. Available from: https://clinicaltrials.gov/ct2/show/NCT03639948 [Last accessed on 6 Aug 2021].

172. Adams S, Schmid P, Rugo HS, et al. Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: cohort A of the phase II KEYNOTE-086 study. Ann Oncol 2019;30:397-404.

173. Adams S, Loi S, Toppmeyer D, et al. Pembrolizumab monotherapy for previously untreated, PD-L1-positive, metastatic triple-negative breast cancer: cohort B of the phase II KEYNOTE-086 study. Ann Oncol 2019;30:405-11.

174. Cortés J, Lipatov O, Im S, et al. KEYNOTE-119: Phase III study of pembrolizumab (pembro) versus single-agent chemotherapy (chemo) for metastatic triple negative breast cancer (mTNBC). Ann Oncol 2019;30:v859-60.

175. Apetoh L, Ghiringhelli F, Tesniere A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med 2007;13:1050-9.

176. Surace L, Scheifinger NA, Gupta A, van den Broek M. Radiotherapy supports tumor-specific immunity by acute inflammation. Oncoimmunology 2016;5:e1060391.

177. Ho AY, Barker CA, Arnold BB, et al. A phase 2 clinical trial assessing the efficacy and safety of pembrolizumab and radiotherapy in patients with metastatic triple-negative breast cancer. Cancer 2020;126:850-60.

178. Vanpouille-Box C, Alard A, Aryankalayil MJ, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun 2017;8:15618.

179. Weiss GJ, Waypa J, Blaydorn L, et al. A phase Ib study of pembrolizumab plus chemotherapy in patients with advanced cancer (PembroPlus). Br J Cancer 2017;117:33-40.

180. Kwa MJ, Iwano A, Esteva FJ, et al. Phase II trial of pembrolizumab in combination with nab-paclitaxel in patients with metastatic HER2-negative breast cancer. JCO 2017;35:TPS1124.

181. Vincent BG, Moore DT, Moore SG, et al. LCCC 1525: Combination immunotherapy with cyclophosphamide plus pembrolizumab in patients with advanced triple negative breast cancer (TNBC). JCO 2017;35:TPS1125.

182. Ge Y, Domschke C, Stoiber N, et al. Metronomic cyclophosphamide treatment in metastasized breast cancer patients: immunological effects and clinical outcome. Cancer Immunol Immunother 2012;61:353-62.

183. Madondo MT, Quinn M, Plebanski M. Low dose cyclophosphamide: Mechanisms of T cell modulation. Cancer Treat Rev 2016;42:3-9.

184. Togashi Y, Shitara K, Nishikawa H. Regulatory T cells in cancer immunosuppression - implications for anticancer therapy. Nat Rev Clin Oncol 2019;16:356-71.

185. Hughes E, Scurr M, Campbell E, Jones E, Godkin A, Gallimore A. T-cell modulation by cyclophosphamide for tumour therapy. Immunology 2018;154:62-8.

186. Engel JB, Honig A, Kapp M, et al. Mechanisms of tumor immune escape in triple-negative breast cancers (TNBC) with and without mutated BRCA 1. Arch Gynecol Obstet 2014;289:141-7.

187. Taylor NA, Vick SC, Iglesia MD, et al. Treg depletion potentiates checkpoint inhibition in claudin-low breast cancer. J Clin Invest 2017;127:3472-83.

188. Gandhi L, Rodríguez-Abreu D, Gadgeel S, et al. KEYNOTE-189 Investigators. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med 2018;378:2078-92.

189. Paz-Ares L, Luft A, Vicente D, et al. KEYNOTE-407 Investigators. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med 2018;379:2040-51.

190. Socinski MA, Jotte RM, Cappuzzo F, et al. IMpower150 Study Group. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med 2018;378:2288-301.

191. West H, Mccleod M, Hussein M, et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2019;20:924-37.

192. Jotte R, Cappuzzo F, Vynnychenko I, et al. Atezolizumab in combination with carboplatin and nab-paclitaxel in advanced squamous NSCLC (IMpower131): results from a randomized phase III trial. J Thorac Oncol 2020;15:1351-60.

193. Vidula N, Goga A, Hwang J, et al. A randomized phase II study of pembrolizumab, an anti-PD (programmed cell death) 1 antibody, in combination with carboplatin compared to carboplatin alone in breast cancer patients with chest wall disease, with immunologic and genomic correlative studies. J Clin Oncol 2018;36:TPS1113.

194. Page DB, Chun B, Pucilowska J, et al. Pembrolizumab (pembro) with paclitaxel (taxol) or capecitabine (cape) as early treatment of metastatic triple-negative breast cancer (mTNBC). J Clin Oncol 2019;37:1015.

195. Shah AN, Flaum L, Helenowski I, et al. Phase II study of pembrolizumab and capecitabine for triple negative and hormone receptor-positive, HER2-negative endocrine-refractory metastatic breast cancer. J Immunother Cancer 2020;8:e000173.

196. Fostira F, Papadimitriou M, Papadimitriou C. Current practices on genetic testing in ovarian cancer. Ann Transl Med 2020;8:1703.

197. U.S. Food & Drug. Available from: www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm592357 [Last accessed on 6 Aug 2021].

198. U.S. Food & Drug. FDA approves talazoparib for gBRCAm HER2-negative locally advanced or metastatic breast cancer. Available from: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-talazoparib-gbrcam-her2-negative-locally-advanced-or-metastatic-breast-cancer [Last accessed on 6 Aug 2021].

199. Vinayak S, Tolaney SM, Schwartzberg L, et al. Open-label clinical trial of niraparib combined with pembrolizumab for treatment of advanced or metastatic triple-negative breast cancer. JAMA Oncol 2019;5:1132-40.

200. Varnum BC, Young C, Elliott G, et al. Axl receptor tyrosine kinase stimulated by the vitamin K-dependent protein encoded by growth-arrest-specific gene 6. Nature 1995;373:623-6.

201. Shieh YS, Lai CY, Kao YR, et al. Expression of axl in lung adenocarcinoma and correlation with tumor progression. Neoplasia 2005;7:1058-64.

202. Gay CM, Balaji K, Byers LA. Giving AXL the axe: targeting AXL in human malignancy. Br J Cancer 2017;116:415-23.

203. Zhu C, Wei Y, Wei X. AXL receptor tyrosine kinase as a promising anti-cancer approach: functions, molecular mechanisms and clinical applications. Mol Cancer 2019;18:153.

204. Wiseman CL, Kharazi A. Phase I study with SV-BR-1 breast cancer cell line vaccine and GMCSF: clinical experience in 14 patients. TOBCANJ 2010;2:4-11.

205. National Cancer Institute. AXL inhibitor BGB324. Available from: https://www.cancer.gov/publications/dictionaries/cancer-drug/def/bemcentinib [Last accessed on 6 Aug 2021].

206. Wnuk-Lipinska K, Davidsen K, Blø M, et al. BGB324, a selective small molecule inhibitor of receptor tyrosine kinase AXL, abrogates tumor intrinsic and microenvironmental immune suppression and enhances immune checkpoint inhibitor efficacy in lung and mammary adenocarcinoma models. Cancer Res 2017;77(13 Suppl):abstract nr 626.

207. Yule M, Wnuk-Lipinska K, Davidsen K, et al. A phase II multi-center study of BGB324 in combination with pembrolizumab in patients with previously treated, locally advanced and unresectable or metastatic triple negative breast cancer (TNBC) or triple negative inflammatory breast cancer (TN-IBC). Cancer Res 2018;78(4 Suppl):abstract OT1-01.

208. Li B, Allendorf DJ, Hansen R, et al. Yeast beta-glucan amplifies phagocyte killing of iC3b-opsonized tumor cells via complement receptor 3-Syk-phosphatidylinositol 3-kinase pathway. J Immunol 2006;177:1661-9.

209. Liu J, Gunn L, Hansen R, Yan J. Combined yeast-derived beta-glucan with anti-tumor monoclonal antibody for cancer immunotherapy. Exp Mol Pathol 2009;86:208-14.

210. Bose N, Chan AS, Guerrero F, et al. Binding of soluble yeast β-glucan to human neutrophils and monocytes is complement-dependent. Front Immunol 2013;4:230.

211. O'day S, Borges VF, Chmielowski B, et al. An open label, multicenter phase II study combining imprime PGG (PGG) with pembrolizumab (P) in previously treated metastatic triple-negative breast cancer (mTNBC). JCO 2019;37:2550-2550.

212. Abdou Y, Williams LE, Kalinski P, Opyrchal M. Chemokine modulation to enhance the effectiveness of pembrolizumab in patients with metastatic triple negative breast cancer. Cancer Res 2019;79(4 Suppl):abstract OT2-06.

213. Theodoraki MN, Yerneni S, Sarkar SN, et al. Helicase-driven activation of NFκB-COX2 pathway mediates the immunosuppressive component of dsRNA-driven inflammation in the human tumor microenvironment. Cancer Res 2018;78:4292-302.

214. Mitchell WM. Efficacy of rintatolimod in the treatment of chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME). Expert Rev Clin Pharmacol 2016;9:755-70.

215. Taylor KM, Morgan HE, Johnson A, Hadley LJ, Nicholson RI. Structure-function analysis of LIV-1, the breast cancer-associated protein that belongs to a new subfamily of zinc transporters. Biochem J 2003;375:51-9.

216. Taylor KM, Hiscox S, Nicholson RI. Zinc transporter LIV-1: a link between cellular development and cancer progression. Trends Endocrinol Metab 2004;15:461-3.

217. Sussman D, Smith LM, Anderson ME, et al. SGN-LIV1A: a novel antibody-drug conjugate targeting LIV-1 for the treatment of metastatic breast cancer. Mol Cancer Ther 2014;13:2991-3000.

218. Cao AT, Higgins S, Stevens N, Gardai SJ, Sussman D. Additional mechanisms of action of ladiratuzumab vedotin contribute to increased immune cell activation within the tumor. Cancer Res 2018;78(13 Suppl):abstract nr 2742.

219. Nagayama A, Vidula N, Ellisen L, Bardia A. Novel antibody-drug conjugates for triple negative breast cancer. Ther Adv Med Oncol 2020;12:1758835920915980.

220. Han HS, Alemany CA, Brown-glaberman UA, et al. SGNLVA-002: Single-arm, open label phase Ib/II study of ladiratuzumab vedotin (LV) in combination with pembrolizumab for first-line treatment of patients with unresectable locally advanced or metastatic triple-negative breast cancer. JCO 2019;37:TPS1110.

221. Lacher MD, Bauer G, Fury B, et al. SV-BR-1-GM, a Clinically effective GM-CSF-secreting breast cancer cell line, expresses an immune signature and directly activates CD4+ T Lymphocytes. Front Immunol 2018;9:776.

222. National Cancer Institute. Allogeneic GM-CSF-secreting breast cancer vaccine SV-BR-1-GM. Available from: https://www.cancer.gov/publications/dictionaries/cancer-drug/def/allogeneic-gm-csf-secreting-breast-cancer-vaccine-sv-br-1-gm [Last accessed on 6 Aug 2021].

223. Williams W, Dakhil SR, Holmes JP, Bhattacharya S, Calfa C, et al. . Efficacy and safety of a modified whole tumor cell targeted immunotherapy in patients with advanced breast cancer alone and in combination with immune checkpoint inhibitors. Cancer Res 2020;80:(4 Supplement):abstract P3-09-08.

224. National Cancer Institute. Spartalizumab. Available from: https://www.cancer.gov/publications/dictionaries/cancer-drug/def/spartalizumab [Last accessed on 6 Aug 2021].

225. Naing A, Gainor JF, Gelderblom H, et al. A first-in-human phase 1 dose escalation study of spartalizumab (PDR001), an anti-PD-1 antibody, in patients with advanced solid tumors. J Immunother Cancer 2020;8:e000530.

226. Minami H, Doi T, Toyoda M, et al. Phase I study of the antiprogrammed cell death-1 Ab spartalizumab (PDR001) in Japanese patients with advanced malignancies. Cancer Sci 2021;112:725-33.

227. Hong DS, Schoffski P, Calvo A, et al. Phase I/II study of LAG525 ± spartalizumab (PDR001) in patients (pts) with advanced malignancies. JCO 2018;36:3012-3012.

228. NCBI. Available from: https://www.ncbi.nlm.nih.gov/medgen/891402 [Last accessed on 6 Aug 2021].

229. Baumeister SH, Freeman GJ, Dranoff G, Sharpe AH. Coinhibitory Pathways in Immunotherapy for Cancer. Annu Rev Immunol 2016;34:539-73.

230. Andrews LP, Marciscano AE, Drake CG, Vignali DA. LAG3 (CD223) as a cancer immunotherapy target. Immunol Rev 2017;276:80-96.

231. Workman CJ, Vignali DA. The CD4-related molecule, LAG-3 (CD223), regulates the expansion of activated T cells. Eur J Immunol 2003;33:970-9.

232. Workman CJ, Cauley LS, Kim IJ, Blackman MA, Woodland DL, Vignali DA. Lymphocyte activation gene-3 (CD223) regulates the size of the expanding T cell population following antigen activation in vivo. J Immunol 2004;172:5450-5.

233. Calvo A, Joensuu H, Sebastian M, et al. Phase Ib/II study of lacnotuzumab (MCS110) combined with spartalizumab (PDR001) in patients (pts) with advanced tumors. JCO 2018;36:3014-3014.

234. National Cancer Institute. Lacnotuzumab (Code C80044). Available from: https://ncit.nci.nih.gov/ncitbrowser/ConceptReport.jsp?dictionary=NCI%20Thesaurus&code=C80044 [Last accessed on 6 Aug 2021].

235. Kumar S, Ramesh A, Kulkarni A. Targeting macrophages: a novel avenue for cancer drug discovery. Expert Opin Drug Discov 2020;15:561-74.

236. Pognan F, Couttet P, Demin I, et al. Colony-stimulating factor-1 antibody lacnotuzumab in a phase 1 healthy volunteer study and mechanistic investigation of safety outcomes. J Pharmacol Exp Ther 2019;369:428-42.

237. Achkova D, Maher J. Role of the colony-stimulating factor (CSF)/CSF-1 receptor axis in cancer. Biochem Soc Trans 2016;44:333-41.

238. Qiu SQ, Waaijer SJH, Zwager MC, de Vries EGE, van der Vegt B, Schröder CP. Tumor-associated macrophages in breast cancer: Innocent bystander or important player? Cancer Treat Rev 2018;70:178-89.

239. United States securities and exchange commission. Available from: https://sec.report/Document/0001193125-19-003027 [Last accessed on 6 Aug 2021].

240. Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol 2015;15:760-70.

241. Corrales L, Glickman LH, McWhirter SM, et al. Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity. Cell Rep 2015;11:1018-30.

242. Meric-bernstam F, Sandhu SK, Hamid O, et al. Phase Ib study of MIW815 (ADU-S100) in combination with spartalizumab (PDR001) in patients (pts) with advanced/metastatic solid tumors or lymphomas. J Clin Oncol 2019;37:2507.

243. National Cancer Institute. avelumab. Available from: https://www.cancer.gov/publications/dictionaries/cancer-drug/def/avelumab [Last accessed on 6 Aug 2021].

244. Liu K, Tan S, Chai Y, et al. Structural basis of anti-PD-L1 monoclonal antibody avelumab for tumor therapy. Cell Res 2017;27:151-3.

245. Picardo SL, Doi J, Hansen AR. Structure and optimization of checkpoint inhibitors. Cancers (Basel) 2019;12:38.

246. Juliá EP, Amante A, Pampena MB, Mordoh J, Levy EM. Avelumab, an IgG1 anti-PD-L1 immune checkpoint inhibitor, triggers NK cell-mediated cytotoxicity and cytokine production against triple negative breast cancer cells. Front Immunol 2018;9:2140.

247. U.S. Food and Drug. FDA approves first treatment for rare form of skin cancer. Available from: https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-rare-form-skin-cancer [Last accessed on 6 Aug 2021].

248. U.S. Food and Drug. FDA grants accelerated approval to avelumab for urothelial carcinoma. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-avelumab-urothelial-carcinoma [Last accessed on 6 Aug 2021].

249. U.S. Food and Drug. FDA approves avelumab plus axitinib for renal cell carcinoma. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-avelumab-plus-axitinib-renal-cell-carcinoma [Last accessed on 6 Aug 2021].

250. U.S. Food and Drug. FDA approves avelumab for urothelial carcinoma maintenance treatment. Available from: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-avelumab-urothelial-carcinoma-maintenance-treatment [Last accessed on 6 Aug 2021].

251. Bavencio, INN-avelumab. Available from: https://www.ema.europa.eu/en/documents/product-information/bavencio-epar-product-information_en.pdf [Last accessed on 6 Aug 2021].

252. Clinical Trial. A proof of concept window trial of the immunological effects of avelumab and aspirin in triple-negative breast cancer. Available from: https://clinicaltrials.gov/ct2/show/NCT04188119 [Last accessed on 6 Aug 2021].

253. Karavitis J, Hix LM, Shi YH, Schultz RF, Khazaie K, Zhang M. Regulation of COX2 expression in mouse mammary tumor cells controls bone metastasis and PGE2-induction of regulatory T cell migration. PLoS One 2012;7:e46342.

254. Wong JL, Obermajer N, Odunsi K, Edwards RP, Kalinski P. Synergistic COX2 Induction by IFNγ and TNFα self-limits type-1 immunity in the human tumor microenvironment. Cancer Immunol Res 2016;4:303-11.

255. Miao J, Lu X, Hu Y, et al. Prostaglandin E2 and PD-1 mediated inhibition of antitumor CTL responses in the human tumor microenvironment. Oncotarget 2017;8:89802-10.

256. Conte PF, Dieci MV, Bisagni G, et al. Phase III randomized study of adjuvant treatment with the ANTI-PD-L1 antibody avelumab for high-risk triple negative breast cancer patients: the A-BRAVE trial. JCO 2020;38:TPS598-TPS598.

257. Yap TA, Konstantinopoulos P, Telli ML, et al. JAVELIN PARP medley, a phase 1b/2 study of avelumab plus talazoparib: results from advanced breast cancer cohorts. Cancer Res 2020;80(4 Suppl):abstract nr P1-19.

258. Mavratzas A, Seitz J, Smetanay K, Schneeweiss A, Jäger D, Fremd C. Atezolizumab for use in PD-L1-positive unresectable, locally advanced or metastatic triple-negative breast cancer. Future Oncol 2020;16:4439-53.

259. Herbst RS, Soria JC, Kowanetz M, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014;515:563-7.

260. Matsumoto K, Fukuyama S, Eguchi-Tsuda M, et al. B7-DC induced by IL-13 works as a feedback regulator in the effector phase of allergic asthma. Biochem Biophys Res Commun 2008;365:170-5.

261. Geyer CE, Loibl S, Rastogi P, et al. NSABP B-59/GBG 96-GeparDouze: a randomized double-blind phase III clinical trial of neoadjuvant chemotherapy (NAC) with atezolizumab or placebo in patients (pts) with triple-negative breast cancer (TNBC) followed by adjuvant atezolizumab or placebo. J Clin Oncol 2018;36:15_suppl.

262. Loibl S, Jackisch C, Rastogi P, et al. GeparDouze/NSABP B-59: A randomized double-blind phase III clinical trial of neoadjuvant chemotherapy with atezolizumab or placebo in patients with triple negative breast cancer (TNBC) followed by adjuvant atezolizumab or placebo. Ann Oncol 2019;30:iii38.

263. Gianni L, Huang C-S, Egle D, et al. . GS3-04: pathologic complete response (pCR) to neoadjuvant treatment with or without atezolizumab in triple negative, early high-risk and locally advanced breast cancer. NeoTRIPaPDL1 michelangelo randomized study. Abstract presented at: 2019 San Antonio Breast Cancer Symposium; Dec 10-14, 2019; San Antonio, TX.

264. Mittendorf EA, Zhang H, Barrios CH, et al. Neoadjuvant atezolizumab in combination with sequential nab-paclitaxel and anthracycline-based chemotherapy versus placebo and chemotherapy in patients with early-stage triple-negative breast cancer (IMpassion031): a randomised, double-blind, phase 3 trial. Lancet 2020;396:1090-100.

265. Clinical Trial. A randomized, double-blind, phase III clinical trial of neoadjuvant chemotherapy with atezolizumab or placebo in patients with triple-negative breast cancer followed by adjuvant continuation of atezolizumab or placebo. Available from: https://clinicaltrials.gov/ct2/show/NCT03281954 [Last accessed on 6 Aug 2021].

266. Mcarthur HL, Ignatiadis M, Guillaume S, et al. ALEXANDRA/IMpassion030: A phase III study of standard adjuvant chemotherapy with or without atezolizumab in early-stage triple-negative breast cancer. JCO 2019;37:TPS598.

267. Clinical Trial. A phase I, open-label, dose-escalation study of the safety and pharmacokinetics of atezolizumab (MPDL3280A) administered intravenously as a single agent to patients with locally advanced or metastatic solid tumors or hematologic malignancies. Available from: https://clinicaltrials.gov/ct2/show/NCT01375842 [Last accessed on 6 Aug 2021].

268. Adams S, Diamond JR, Hamilton E, et al. Atezolizumab plus nab-paclitaxel in the treatment of metastatic triple-negative breast cancer with 2-year survival follow-up: a phase 1b clinical trial. JAMA Oncol 2019;5:334-42.

269. Schmid P, Adams S, Rugo HS, et al. IMpassion130 Trial Investigators. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med 2018;379:2108-21.

270. Schmid P, Rugo HS, Adams S, et al. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2020;21:44-59.

271. Iwata H, Inoue K, Kaneko K, et al. Subgroup analysis of Japanese patients in a Phase 3 study of atezolizumab in advanced triple-negative breast cancer (IMpassion130). Jpn J Clin Oncol 2019;49:1083-91.

272. Adams S, Diéras V, Barrios CH, et al. Patient-reported outcomes from the phase III IMpassion130 trial of atezolizumab plus nab-paclitaxel in metastatic triple-negative breast cancer. Ann Oncol 2020;31:582-9.

273. Weng X, Huang X, Li H, et al. First-line treatment with atezolizumab plus nab-paclitaxel for advanced triple-negative breast cancer: a cost-effectiveness analysis. Am J Clin Oncol 2020;43:340-8.

274. Ruiz-borrego M, Martin Jimenez M, Ruiz A, et al. Phase III evaluating the addition of fulvestrant (F) to anastrozol (A) as adjuvant therapy in postmenopausal women with hormone receptor positive HER2 negative (HR+/HER2-) early breast cancer (EBC): results from the GEICAM/2006-10 study. Ann Oncol 2017;28:v43.

275. Businesswire. Genentech provides update on phase III study of tecentriq in combination with paclitaxel for people with metastatic triple-negative breast cancer. Available from: https://www.businesswire.com/news/home/20200806005915/en/Genentech-Provides-Update-on-Phase-III-Study-of-Tecentriq-in-Combination-With-Paclitaxel-for-People-With-Metastatic-Triple-Negative-Breast-Cancer [Last accessed on 6 Aug 2021].

276. Dent R, Andre F, Goncalves A, et al. IMpassion132: a double-blind randomized phase 3 trial evaluating chemotherapy (CT) ± atezolizumab (atezo) for early progressing locally advanced/metastatic triple-negative breast cancer (mTNBC). J Clin Oncol 2018;36:TPS1115.

277. Ibrahim R, Stewart R, Shalabi A. PD-L1 blockade for cancer treatment: MEDI4736. Semin Oncol 2015;42:474-83.

278. Stewart R, Morrow M, Hammond SA, et al. Identification and characterization of MEDI4736, an antagonistic anti-PD-L1 monoclonal antibody. Cancer Immunol Res 2015;3:1052-62.

279. Syed YY. Durvalumab: first global approval. Drugs 2017;77:1369-76.

280. IMFINZI, INN-durvalumab. Available from: https://www.ema.europa.eu/en/documents/product-information/imfinzi-epar-product-information_en.pdf [Last accessed on 6 Aug 2021].

281. U.S. Food & Drug. FDA approves durvalumab for extensive-stage small cell lung cancer. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-durvalumab-extensive-stage-small-cell-lung-cancer [Last accessed on 6 Aug 2021].

282. Pusztai L, Reisenbichler E, Bai Y, et al. Durvalumab (MEDI4736) concurrent with nab-paclitaxel and dose dense doxorubicin cyclophosphamide (ddAC) as neoadjuvant therapy for triple negative breast cancer (TNBC). Cancer Res 2020;80(4 Suppl):abstract nr PD1-01.

283. Loibl S, Untch M, Burchardi N, et al. A randomised phase II study investigating durvalumab in addition to an anthracycline taxane-based neoadjuvant therapy in early triple-negative breast cancer: clinical results and biomarker analysis of GeparNuevo study. Ann Oncol 2019;30:1279-88.

284. Clinical Trial. A randomized phase II study evaluating pathologic response rates following pre-operative non-anthracycline chemotherapy, durvalumab (MEDI4736) +/- RAdiation therapy (RT) in triple negative breast cancer (TNBC): the PANDoRA Study. Available from: https://clinicaltrials.gov/ct2/show/NCT03872505 [Last accessed on 6 Aug 2021].

285. Clinical Trial. PHOENIX DDR/Anti-PD-L1 trial: a pre-surgical window of opportunity and post-surgical adjuvant biomarker study of DNA damage response inhibition and/or anti-PD-L1 immunotherapy in patients with neoadjuvant chemotherapy resistant residual triple negative breast cancer. Available from: https://clinicaltrials.gov/ct2/show/NCT03740893 [Last accessed on 6 Aug 2021].

286. National Cancer Institute. Ceralasertib. Available from: https://www.cancer.gov/publications/dictionaries/cancer-drug/def/ceralasertib?redirect=true [Last accessed on 6 Aug 2021].

287. Clinical Trial. A phase IB/II study of durvalumab (MEDI4736) combined with dose-dense EC in a neoadjuvant setting for patients with locally advanced luminal B HER2(-) or triple negative breast cancers. Available from: https://clinicaltrials.gov/ct2/show/NCT03356860 [Last accessed on 6 Aug 2021].

288. Clinical Trial. PERSONALIZED MEDICINE GROUP / UCBG UC-0105/1304: SAFIR02_breast - evaluation of the efficacy of high throughput genome analysis as a therapeutic decision tool for patients with metastatic breast cancer. Available from: https://clinicaltrials.gov/ct2/show/NCT02299999 [Last accessed on 6 Aug 2021].

289. Bachelot T, Filleron T, Bieche I, et al. Durvalumab compared to maintenance chemotherapy in metastatic breast cancer: the randomized phase II SAFIR02-BREAST IMMUNO trial. Nat Med 2021;27:250-5.

290. Durvalumab and paclitaxel combination for treatment of metastatic triple negative breast cancer is safe with very promising efficacy. Available from: https://www.annalsofoncology.org/article/S0923-7534(19)59484-4/pdf [Last accessed on 6 Aug 2021].

291. Zimmer AS, Nichols E, Cimino-Mathews A, et al. A phase I study of the PD-L1 inhibitor, durvalumab, in combination with a PARP inhibitor, olaparib, and a VEGFR1-3 inhibitor, cediranib, in recurrent women's cancers with biomarker analyses. J Immunother Cancer 2019;7:197.

292. National Cancer Institute. Cediranib maleate. Available from: https://www.cancer.gov/publications/dictionaries/cancer-drug/def/cediranib-maleate [Last accessed on 6 Aug 2021].

293. Bindra RS, Schaffer PJ, Meng A, et al. Down-regulation of Rad51 and decreased homologous recombination in hypoxic cancer cells. Mol Cell Biol 2004;24:8504-18.

294. Kandalaft LE, Motz GT, Busch J, Coukos G. . Angiogenesis and the tumor vasculature as antitumor immune modulators: the role of vascular endothelial growth factor and endothelin. Cancer Immunol Immunother 2011;344:129-48.

295. Domchek SM, Postel-vinay S, Im S, et al. Olaparib and durvalumab in patients with germline BRCA-mutated metastatic breast cancer (MEDIOLA): an open-label, multicentre, phase 1/2, basket study. Lancet Oncol 2020;21:1155-64.

Journal of Cancer Metastasis and Treatment
ISSN 2454-2857 (Online) 2394-4722 (Print)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/