REFERENCES
1. Armenta DA, Dixon SJ. Investigating nonapoptotic cell death using chemical biology approaches. Cell Chem Biol 2020;27:376-86.
2. Galluzzi L, Vitale I, Aaronson SA, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ 2018;25:486-541.
3. Schweichel JU, Merker HJ. The morphology of various types of cell death in prenatal tissues. Teratology 1973;7:253-66.
4. Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H, Vandenabeele P. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol 2014;15:135-47.
5. Döhner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017;129:424-47.
6. Döhner H, Wei AH, Appelbaum FR, et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood 2022;140:1345-77.
8. Dombret H, Seymour JF, Butrym A, et al. International phase 3 study of azacitidine vs. conventional care regimens in older patients with newly diagnosed AML with > 30% blasts. Blood 2015;126:291-9.
9. Kantarjian HM, Thomas XG, Dmoszynska A, et al. Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol 2012;30:2670-7.
10. DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med 2020;383:617-29.
11. Garciaz S, Saillard C, Hicheri Y, Hospital MA, Vey N. Venetoclax in acute myeloid leukemia: molecular basis, evidences for preclinical and clinical efficacy and strategies to target resistance. Cancers 2021;13:5608.
12. Maiti A, Rausch CR, Cortes JE, et al. Outcomes of relapsed or refractory acute myeloid leukemia after frontline hypomethylating agent and venetoclax regimens. Haematologica 2021;106:894-8.
13. Maiti A, Carter BZ, Andreeff M, Konopleva MY. Beyond BCL-2 inhibition in acute myloid leukemia: other approaches to leverage the apoptotic pathway. Clin Lymphoma Myeloma Leuk 2022;22:652-8.
14. Lachowiez CA, Atluri H, DiNardo CD. Advancing the standard: venetoclax combined with intensive induction and consolidation therapy for acute myeloid leukemia. Ther Adv Hematol 2022;13:20406207221093964.
15. Czabotar PE, Lessene G, Strasser A, Adams JM. Control of apoptosis by the BCL-2 protein family: implications for physiology and therapy. Nat Rev Mol Cell Biol 2014;15:49-63.
16. Tait SW, Green DR. Mitochondria and cell death: outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol 2010;11:621-32.
17. Cheng EH, Sheiko TV, Fisher JK, Craigen WJ, Korsmeyer SJ. VDAC2 inhibits BAK activation and mitochondrial apoptosis. Science 2003;301:513-7.
18. Chen HC, Kanai M, Inoue-Yamauchi A, et al. An interconnected hierarchical model of cell death regulation by the BCL-2 family. Nat Cell Biol 2015;17:1270-81.
19. Hardwick JM, Soane L. Multiple functions of BCL-2 family proteins. Cold Spring Harb Perspect Biol 2013;5:a008722-a008722.
20. Letai A, Bassik MC, Walensky LD, Sorcinelli MD, Weiler S, Korsmeyer SJ. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell 2002;2:183-92.
21. Carneiro BA, El-Deiry WS. Targeting apoptosis in cancer therapy. Nat Rev Clin Oncol 2020;17:395-417.
22. Meier P, Vousden KH. Lucifer’s labyrinth--ten years of path finding in cell death. Mol Cell 2007;28:746-54.
23. Deveraux QL, Takahashi R, Salvesen GS, Reed JC. X-linked IAP is a direct inhibitor of cell-death proteases. Nature 1997;388:300-4.
24. Bai L, Smith DC, Wang S. Small-molecule SMAC mimetics as new cancer therapeutics. Pharmacol Ther 2014;144:82-95.
25. Vercammen D, Beyaert R, Denecker G, et al. Inhibition of caspases increases the sensitivity of L929 cells to necrosis mediated by tumor necrosis factor. J Exp Med 1998;187:1477-85.
26. Degterev A, Huang Z, Boyce M, et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol 2005;1:112-9.
28. Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 2012;149:1060-72.
29. Hirschhorn T, Stockwell BR. The development of the concept of ferroptosis. Free Radic Biol Med 2019;133:130-43.
30. Skouta R, Dixon SJ, Wang J, et al. Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models. J Am Chem Soc 2014;136:4551-6.
31. Zilka O, Shah R, Li B, et al. On the mechanism of cytoprotection by ferrostatin-1 and liproxstatin-1 and the role of lipid peroxidation in ferroptotic cell death. ACS Cent Sci 2017;3:232-43.
32. Stockwell BR, Friedmann Angeli JP, Bayir H, et al. Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell 2017;171:273-85.
33. Gao M, Monian P, Quadri N, Ramasamy R, Jiang X. Glutaminolysis and transferrin regulate ferroptosis. Mol Cell 2015;59:298-308.
34. Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, Stockwell BR. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA 2016;113:E4966-75.
35. Doll S, Proneth B, Tyurina YY, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol 2017;13:91-8.
36. Kagan VE, Mao G, Qu F, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol 2017;13:81-90.
37. Bersuker K, Hendricks JM, Li Z, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 2019;575:688-92.
38. Doll S, Freitas FP, Shah R, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 2019;575:693-8.
39. Kraft VAN, Bezjian CT, Pfeiffer S, et al. GTP cyclohydrolase 1/tetrahydrobiopterin counteract ferroptosis through lipid remodeling. ACS Cent Sci 2020;6:41-53.
40. Soula M, Weber RA, Zilka O, et al. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat Chem Biol 2020;16:1351-60.
41. Kang R, Kroemer G, Tang D. The tumor suppressor protein p53 and the ferroptosis network. Free Radic Biol Med 2019;133:162-8.
42. Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res 2019;29:347-64.
43. Amaravadi RK, Kimmelman AC, Debnath J. Targeting autophagy in cancer: recent advances and future directions. Cancer Discov 2019;9:1167-81.
44. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol 2009;7:99-109.
45. Vitale I, Galluzzi L, Castedo M, Kroemer G. Mitotic catastrophe: a mechanism for avoiding genomic instability. Nat Rev Mol Cell Biol 2011;12:385-92.
46. Vakifahmetoglu H, Olsson M, Zhivotovsky B. Death through a tragedy: mitotic catastrophe. Cell Death Differ 2008;15:1153-62.
47. Gao M, Monian P, Pan Q, Zhang W, Xiang J, Jiang X. Ferroptosis is an autophagic cell death process. Cell Res 2016;26:1021-32.
48. Zhou Y, Shen Y, Chen C, et al. The crosstalk between autophagy and ferroptosis: what can we learn to target drug resistance in cancer? Cancer Biol Med 2019;16:630-46.
49. Lee BL, Stowe IB, Gupta A, et al. Caspase-11 auto-proteolysis is crucial for noncanonical inflammasome activation. J Exp Med 2018;215:2279-88.
50. Canli Ö, Alankuş YB, Grootjans S, et al. Glutathione peroxidase 4 prevents necroptosis in mouse erythroid precursors. Blood 2016;127:139-48.
51. Seiler A, Schneider M, Förster H, et al. Glutathione peroxidase 4 senses and translates oxidative stress into 12/15-lipoxygenase dependent- and AIF-mediated cell death. Cell Metab 2008;8:237-48.
52. Hughes SA, Lin M, Weir A, et al. Caspase-8-driven apoptotic and pyroptotic crosstalk causes cell death and IL-1β release in X-linked inhibitor of apoptosis (XIAP) deficiency. EMBO J 2023;42:e110468.
53. Garciaz S, Guirguis AA, Müller S, et al. Pharmacologic reduction of mitochondrial iron triggers a noncanonical BAX/BAK-dependent cell death. Cancer Discov 2022;12:774-91.
54. Mai TT, Hamaï A, Hienzsch A, et al. Salinomycin kills cancer stem cells by sequestering iron in lysosomes. Nat Chem 2017;9:1025-33.
55. Saxena K, DiNardo C, Daver N, Konopleva M. Harnessing apoptosis in AML. Clin Lymphoma Myeloma Leuk 2020;20 Suppl 1:S61-4.
56. Lowe SW, Bodis S, McClatchey A, et al. P53 status and the efficacy of cancer therapy in vivo. Science 1994;266:807-10.
57. Hu N, Guo Y, Xue H, et al. Abstract 3077: Preclinical characterization of BGB-11417, a potent and selective Bcl-2 inhibitor with superior antitumor activities in haematological tumor models. Cancer Res 2020;80:3077-3077.
58. Tiran AL, Claperon A, Davidson J, et al. Abstract 1276: identification of S65487/VOB560 as a potent and selective intravenous 2nd-generation BCL-2 inhibitor active in wild-type and clinical mutants resistant to Venetoclax. Cancer Res 2021;81:1276.
59. Ailawadhi S, Chanan-khan AAA, Chen Z, et al. First-in-human study of lisaftoclax (APG-2575), a novel BCL-2 inhibitor (BCL-2i), in patients (pts) with relapsed/refractory (R/R) CLL and other hematologic malignancies (HMs). J Clin Oncol 2021;39:7502.
60. Walker AR, Bergua Burgues JM, Montesinos P, et al. Phase 1 study of LP-108 as monotherapy and in combination with azacitidine in patients with relapsed or refractory myelodysplastic syndromes (MDS), chronic myelomonocytic leukemia (CMML), or acute myeloid leukemia (AML). J Clin Oncol 2022;40:TPS7071-TPS7071.
61. Kotschy A, Szlavik Z, Murray J, et al. The MCL1 inhibitor S63845 is tolerable and effective in diverse cancer models. Nature 2016;538:477-82.
62. Tron AE, Belmonte MA, Adam A, et al. Discovery of Mcl-1-specific inhibitor AZD5991 and preclinical activity in multiple myeloma and acute myeloid leukemia. Nat Commun 2018;9:5341.
63. Caenepeel S, Brown SP, Belmontes B, et al. AMG 176, a selective MCL1 inhibitor, is effective in hematologic cancer models alone and in combination with established therapies. Cancer Discov 2018;8:1582-97.
64. Caenepeel S, Karen R, Belmontes B, et al. Abstract 6218: discovery and preclinical evaluation of AMG 397, a potent, selective and orally bioavailable MCL1 inhibitor. Cancer Res 2020;80:6218-6218.
65. Wei AH, Roberts AW, Spencer A, et al. Targeting MCL-1 in hematologic malignancies: rationale and progress. Blood Rev 2020;44:100672.
66. Zeidner JF, Karp JE. Clinical activity of alvocidib (flavopiridol) in acute myeloid leukemia. Leuk Res 2015;39:1312-8.
67. Zeidner JF, Foster MC, Blackford AL, et al. Randomized multicenter phase II study of flavopiridol (alvocidib), cytarabine, and mitoxantrone (FLAM) versus cytarabine/daunorubicin (7 + 3) in newly diagnosed acute myeloid leukemia. Haematologica 2015;100:1172-9.
68. Bogenberger J, Whatcott C, Hansen N, et al. Combined venetoclax and alvocidib in acute myeloid leukemia. Oncotarget 2017;8:107206-22.
69. Zeidner JF, Lee DJ, Frattini M, et al. Phase I study of alvocidib followed by 7 + 3 (cytarabine + daunorubicin) in newly diagnosed acute myeloid leukemia. Clin Cancer Res 2021;27:60-9.
70. Tibes R, Bogenberger JM. Transcriptional silencing of MCL-1 through cyclin-dependent kinase inhibition in acute myeloid leukemia. Front Oncol 2019;9:1205.
71. Baker A, Gregory GP, Verbrugge I, et al. The CDK9 inhibitor dinaciclib exerts potent apoptotic and antitumor effects in preclinical models of MLL-rearranged acute myeloid leukemia. Cancer Res 2016;76:1158-69.
72. Konopleva M, Brander DM, Patel K, et al. A phase 1 dose-escalation study of the oral CDK inhibitor voruciclib in patients with relapsed/refractory B-cell malignancies or acute myeloid leukemia (AML): preliminary results of the completed dose escalation stage in AML. Blood 2021;138:3423-3423.
73. Dey J, Deckwerth TL, Kerwin WS, et al. Voruciclib, a clinical stage oral CDK9 inhibitor, represses MCL-1 and sensitizes high-risk Diffuse Large B-cell Lymphoma to BCL2 inhibition. Sci Rep 2017;7:18007.
74. Cidado J, Boiko S, Proia T, et al. AZD4573 is a highly selective CDK9 inhibitor that suppresses MCL-1 and induces apoptosis in hematologic cancer cells. Clin Cancer Res 2020;26:922-34.
75. Frame S, Saladino C, MacKay C, et al. Fadraciclib (CYC065), a novel CDK inhibitor, targets key pro-survival and oncogenic pathways in cancer. PLoS One 2020;15:e0234103.
76. Chantkran W, Hsieh YC, Zheleva D, Frame S, Wheadon H, Copland M. Interrogation of novel CDK2/9 inhibitor fadraciclib (CYC065) as a potential therapeutic approach for AML. Cell Death Discov 2021;7:137.
77. Borthakur GM, Kadia TM, Al Azzawi H, Zheleva D, Blake D, Chiao JH. Combining CDK2/9 inhibitor CYC065 with venetoclax, a BCL2 inhibitor, to treat patients with relapsed or refractory AML or MDS. Blood 2019;134:1379-1379.
78. Goh KC, Novotny-diermayr V, Hart S, et al. Abstract 2542: TG02, a novel multi-kinase inhibitor with potent anti-leukemic activity. Cancer Res 2010;70:2542-2542.
79. Kivioja JL, Thanasopoulou A, Kumar A, et al. Dasatinib and navitoclax act synergistically to target NUP98-NSD1+/FLT3-ITD+ acute myeloid leukemia. Leukemia 2019;33:1360-72.
80. Kontro M, Kumar A, Majumder MM, et al. HOX gene expression predicts response to BCL-2 inhibition in acute myeloid leukemia. Leukemia 2017;31:301-9.
81. Pullarkat VA, Lacayo NJ, Jabbour E, et al. Venetoclax and navitoclax in combination with chemotherapy in patients with relapsed or refractory acute lymphoblastic leukemia and lymphoblastic lymphoma. Cancer Discov 2021;11:1440-53.
82. Harvey EP, Hauseman ZJ, Cohen DT, et al. Identification of a covalent molecular inhibitor of anti-apoptotic BFL-1 by disulfide tethering. Cell Chem Biol 2020;27:647-56.e6.
83. Liu N, Wang D, Lian C, et al. Selective covalent targeting of anti-apoptotic BFL-1 by a sulfonium-tethered peptide. Chembiochem 2021;22:340-4.
84. Reyna DE, Garner TP, Lopez A, et al. Direct activation of BAX by BTSA1 overcomes apoptosis resistance in acute myeloid leukemia. Cancer Cell 2017;32:490-505.e10.
85. Liang T, Zhou Y, Elhassan RM, Hou X, Yang X, Fang H. HDAC-Bax multiple ligands enhance Bax-dependent apoptosis in HeLa cells. J Med Chem 2020;63:12083-99.
86. Luo X, O'Neill KL, Huang K. The third model of Bax/Bak activation: a Bcl-2 family feud finally resolved? F1000Res 2020;9:935.
87. van Delft MF, Chappaz S, Khakham Y, et al. A small molecule interacts with VDAC2 to block mouse BAK-driven apoptosis. Nat Chem Biol 2019;15:1057-66.
88. Yuan Z, Dewson G, Czabotar PE, Birkinshaw RW. VDAC2 and the BCL-2 family of proteins. Biochem Soc Trans 2021;49:2787-95.
89. Wang BT, Kothambawala T, Wang L, et al. Multimeric anti-DR5 IgM agonist antibody IGM-8444 is a potent inducer of cancer cell apoptosis and synergizes with chemotherapy and BCL-2 inhibitor ABT-199. Mol Cancer Ther 2021;20:2483-94.
90. Phillips DC, Buchanan FG, Cheng D, et al. Hexavalent TRAIL fusion protein eftozanermin alfa optimally clusters apoptosis-inducing TRAIL receptors to induce on-target antitumor activity in solid tumors. Cancer Res 2021;81:3402-14.
91. de Jonge MJ, Carneiro BA, Devriese L, et al. First-in-human study of abbv-621, a TRAIL receptor agonist fusion protein, in patients (Pts) with relapsed/refractory (RR) acute myeloid leukemia (AML) and diffuse large B-cell lymphoma (DLBCL). Blood 2019;134:3924.
92. Humphreys L, Espona-Fiedler M, Longley DB. FLIP as a therapeutic target in cancer. FEBS J 2018;285:4104-23.
93. Longley DB, Higgins C, Fox J, et al. Abstract 5220: development of first-in-class small molecule inhibitors of FLIP which activate caspase-8, the nodal regulator of apoptosis, necroptosis and pyroptosis. Cancer Res 2020;80:5220.
94. Higgins CA, Fox J, Roberts J, et al. Abstract 1342: development and preclinical evaluation of unique first-in-class small molecule inhibitors of the anti-apoptotic protein FLIP. Cancer Res 2021;81:1342.
95. Hashimoto M, Saito Y, Nakagawa R, et al. Combined inhibition of XIAP and BCL2 drives maximal therapeutic efficacy in genetically diverse aggressive acute myeloid leukemia. Nat Cancer 2021;2:340-56.
96. Zhou J, Lu X, Tan TZ, Chng WJ. X-linked inhibitor of apoptosis inhibition sensitizes acute myeloid leukemia cell response to TRAIL and chemotherapy through potentiated induction of proapoptotic machinery. Mol Oncol 2018;12:33-47.
97. Carter BZ, Gronda M, Wang Z, et al. Small-molecule XIAP inhibitors derepress downstream effector caspases and induce apoptosis of acute myeloid leukemia cells. Blood 2005;105:4043-50.
98. Erba HP, Sayar H, Juckett M, et al. Safety and pharmacokinetics of the antisense oligonucleotide (ASO) LY2181308 as a single-agent or in combination with idarubicin and cytarabine in patients with refractory or relapsed acute myeloid leukemia (AML). Invest New Drugs 2013;31:1023-34.
99. Schimmer AD, Estey EH, Borthakur G, et al. Phase I/II trial of AEG35156 X-linked inhibitor of apoptosis protein antisense oligonucleotide combined with idarubicin and cytarabine in patients with relapsed or primary refractory acute myeloid leukemia. J Clin Oncol 2009;27:4741-6.
100. Schimmer AD, Herr W, Hänel M, et al. Addition of AEG35156 XIAP antisense oligonucleotide in reinduction chemotherapy does not improve remission rates in patients with primary refractory acute myeloid leukemia in a randomized phase II study. Clin Lymphoma Myeloma Leuk 2011;11:433-8.
101. Boddu P, Carter BZ, Verstovsek S, Pemmaraju N. SMAC mimetics as potential cancer therapeutics in myeloid malignancies. Br J Haematol 2019;185:219-31.
102. Frey NV, Luger S, Mangan J, et al. A phase I study using single agent birinapant in patients with relapsed myelodysplastic syndrome and acute myelogenous leukemia. Blood 2014;124:3758-3758.
103. Donnellan WB, Diez-campelo M, Heuser M, et al. A phase 2 study of azacitidine (5-AZA) with or without birinapant in subjects with higher risk myelodysplastic syndrome (MDS) or chronic myelomonocytic leukemia (CMML). J Clin Oncol 2016;34:7060.
104. Morrish E, Copeland A, Moujalled DM, et al. Clinical MDR1 inhibitors enhance Smac-mimetic bioavailability to kill murine LSCs and improve survival in AML models. Blood Adv 2020;4:5062-77.
105. Mita MM, LoRusso PM, Papadopoulos KP, et al. A phase I study of ASTX660, an antagonist of inhibitors of apoptosis proteins, in adults with advanced cancers or lymphoma. Clin Cancer Res 2020;26:2819-26.
106. Ward GA, Lewis EJ, Ahn JS, et al. ASTX660, a novel non-peptidomimetic antagonist of cIAP1/2 and XIAP, potently induces TNFα-dependent apoptosis in cancer cell lines and inhibits tumor growth. Mol Cancer Ther 2018;17:1381-91.
107. Pemmaraju N, Carter BZ, Bose P, et al. Final results of a phase 2 clinical trial of LCL161, an oral SMAC mimetic for patients with myelofibrosis. Blood Adv 2021;5:3163-73.
108. Brumatti G, Ma C, Lalaoui N, et al. The caspase-8 inhibitor emricasan combines with the SMAC mimetic birinapant to induce necroptosis and treat acute myeloid leukemia. Sci Transl Med 2016;8:339ra69.
109. Safferthal C, Rohde K, Fulda S. Therapeutic targeting of necroptosis by Smac mimetic bypasses apoptosis resistance in acute myeloid leukemia cells. Oncogene 2017;36:1487-502.
110. Su Z, Yang Z, Xie L, DeWitt JP, Chen Y. Cancer therapy in the necroptosis era. Cell Death Differ 2016;23:748-56.
111. Hillert LK, Bettermann-Bethge K, Nimmagadda SC, Fischer T, Naumann M, Lavrik IN. Targeting RIPK1 in AML cells carrying FLT3-ITD. Int J Cancer 2019;145:1558-69.
112. Li J, Liao D, Wang F, et al. RIPK1 inhibition enhances the therapeutic efficacy of chidamide in FLT3-ITD positive AML, both in vitro and in vivo. Leuk Lymphoma 2022;63:1167-79.
113. Xin J, You D, Breslin P, et al. Sensitizing acute myeloid leukemia cells to induced differentiation by inhibiting the RIP1/RIP3 pathway. Leukemia 2017;31:1154-65.
114. Huang X, Xiao F, Li Y, Qian W, Ding W, Ye X. Bypassing drug resistance by triggering necroptosis: recent advances in mechanisms and its therapeutic exploitation in leukemia. J Exp Clin Cancer Res 2018;37:310.
115. Müller MD, Holst PJ, Nielsen KN. A systematic review of expression and immunogenicity of human endogenous retroviral proteins in cancer and discussion of therapeutic approaches. Int J Mol Sci 2022;23:1330.
116. Fresquet V, Garcia-Barchino MJ, Larrayoz M, et al. Endogenous retroelement activation by epigenetic therapy reverses the warburg effect and elicits mitochondrial-mediated cancer cell death. Cancer Discov 2021;11:1268-85.
117. Zhang J, Liu Y, Li Q, Xu A, Hu Y, Sun C. Ferroptosis in hematological malignancies and its potential network with abnormal tumor metabolism. Biomed Pharmacother 2022;148:112747.
118. Pardieu B, Pasanisi J, Ling F, et al. Cystine uptake inhibition potentiates front-line therapies in acute myeloid leukemia. Leukemia 2022;36:1585-95.
119. Yu Y, Xie Y, Cao L, et al. The ferroptosis inducer erastin enhances sensitivity of acute myeloid leukemia cells to chemotherapeutic agents. Mol Cell Oncol 2015;2:e1054549.
120. Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 2014;156:317-31.
121. Yusuf RZ, Saez B, Sharda A, et al. Aldehyde dehydrogenase 3a2 protects AML cells from oxidative death and the synthetic lethality of ferroptosis inducers. Blood 2020;136:1303-16.
122. Woo JH, Shimoni Y, Yang WS, et al. Elucidating compound mechanism of action by network perturbation analysis. Cell 2015;162:441-51.
123. Guo J, Xu B, Han Q, et al. Ferroptosis: a novel anti-tumor action for cisplatin. Cancer Res Treat 2018;50:445-60.
124. Birsen R, Larrue C, Decroocq J, et al. APR-246 induces early cell death by ferroptosis in acute myeloid leukemia. Haematologica 2022;107:403-16.
125. Hong Y, Ren T, Wang X, et al. APR-246 triggers ferritinophagy and ferroptosis of diffuse large B-cell lymphoma cells with distinct TP53 mutations. Leukemia 2022;36:2269-80.
126. Du J, Wang T, Li Y, et al. DHA inhibits proliferation and induces ferroptosis of leukemia cells through autophagy dependent degradation of ferritin. Free Radic Biol Med 2019;131:356-69.
127. Smith KH, Budhraja A, Lynch J, et al. The heme-regulated inhibitor pathway modulates susceptibility of poor prognosis B-lineage acute leukemia to BH3-mimetics. Mol Cancer Res 2021;19:636-50.
128. Zhang X, Ai Z, Zhang Z, et al. Dihydroartemisinin triggers ferroptosis in multidrug-resistant leukemia cells. DNA Cell Biol 2022;41:705-15.
129. Beatty A, Singh T, Tyurina YY, et al. Ferroptotic cell death triggered by conjugated linolenic acids is mediated by ACSL1. Nat Commun 2021;12:2244.
130. Dierge E, Debock E, Guilbaud C, et al. Peroxidation of n-3 and n-6 polyunsaturated fatty acids in the acidic tumor environment leads to ferroptosis-mediated anticancer effects. Cell Metab 2021;33:1701-1715.e5.
131. Grignano E, Birsen R, Chapuis N, Bouscary D. From iron chelation to overload as a therapeutic strategy to induce ferroptosis in leukemic cells. Front Oncol 2020;10:586530.
132. Müller S, Sindikubwabo F, Cañeque T, et al. CD44 regulates epigenetic plasticity by mediating iron endocytosis. Nat Chem 2020;12:929-38.
133. Rafiq S, McKenna SL, Muller S, Tschan MP, Humbert M. Lysosomes in acute myeloid leukemia: potential therapeutic targets? Leukemia 2021;35:2759-70.
134. Seo W, Silwal P, Song IC, Jo EK. The dual role of autophagy in acute myeloid leukemia. J Hematol Oncol 2022;15:51.
135. Rudat S, Pfaus A, Cheng YY, et al. RET-mediated autophagy suppression as targetable co-dependence in acute myeloid leukemia. Leukemia 2018;32:2189-202.
136. Heydt Q, Larrue C, Saland E, et al. Oncogenic FLT3-ITD supports autophagy via ATF4 in acute myeloid leukemia. Oncogene 2018;37:787-97.
137. Putyrski M, Vakhrusheva O, Bonn F, et al. Disrupting the LC3 interaction region (LIR) binding of selective autophagy receptors sensitizes AML cell lines to cytarabine. Front Cell Dev Biol 2020;8:208.
138. Dykstra KM, Fay HRS, Massey AC, et al. Inhibiting autophagy targets human leukemic stem cells and hypoxic AML blasts by disrupting mitochondrial homeostasis. Blood Adv 2021;5:2087-100.
139. Horne GA, Stobo J, Kelly C, et al. A randomised phase II trial of hydroxychloroquine and imatinib versus imatinib alone for patients with chronic myeloid leukaemia in major cytogenetic response with residual disease. Leukemia 2020;34:1775-86.
140. Dupont M, Huart M, Lauvinerie C, et al. Autophagy targeting and hematological mobilization in FLT3-ITD acute myeloid leukemia decrease repopulating capacity and relapse by inducing apoptosis of committed leukemic cells. Cancers 2022;14:453.
141. Ronan B, Flamand O, Vescovi L, et al. A highly potent and selective Vps34 inhibitor alters vesicle trafficking and autophagy. Nat Chem Biol 2014;10:1013-9.
142. Ghosh J, Kapur R. Role of mTORC1-S6K1 signaling pathway in regulation of hematopoietic stem cell and acute myeloid leukemia. Exp Hematol 2017;50:13-21.
143. Kasner MT, Mick R, Jeschke GR, et al. Sirolimus enhances remission induction in patients with high risk acute myeloid leukemia and mTORC1 target inhibition. Invest New Drugs 2018;36:657-66.
144. Park S, Chapuis N, Saint Marcoux F, et al. A phase Ib GOELAMS study of the mTOR inhibitor RAD001 in association with chemotherapy for AML patients in first relapse. Leukemia 2013;27:1479-86.
145. Burnett AK, Das Gupta E, Knapper S, et al. Addition of the mammalian target of rapamycin inhibitor, everolimus, to consolidation therapy in acute myeloid leukemia: experience from the UK NCRI AML17 trial. Haematologica 2018;103:1654-61.
146. Liesveld JL, Baran A, Azadniv M, et al. A phase II study of sequential decitabine and rapamycin in acute myelogenous leukemia. Leuk Res 2022;112:106749.
147. Johnson DC, Taabazuing CY, Okondo MC, et al. DPP8/DPP9 inhibitor-induced pyroptosis for treatment of acute myeloid leukemia. Nat Med 2018;24:1151-6.
148. Basiorka AA, McGraw KL, Eksioglu EA, et al. The NLRP3 inflammasome functions as a driver of the myelodysplastic syndrome phenotype. Blood 2016;128:2960-75.
149. Sallman DA, List A. The central role of inflammatory signaling in the pathogenesis of myelodysplastic syndromes. Blood 2019;133:1039-48.
150. McLemore AF, Hou HA, Meyer BS, et al. Somatic gene mutations expose cytoplasmic DNA to co-opt the cGAS/STING/NLRP3 axis in myelodysplastic syndromes. JCI Insight 2022:7.
151. Chakraborty S, Shapiro LC, de Oliveira S, Rivera-Pena B, Verma A, Shastri A. Therapeutic targeting of the inflammasome in myeloid malignancies. Blood Cancer J 2021;11:152.
152. Zahr A, Borthakur G. Emerging cell cycle inhibitors for acute myeloid leukemia. Expert Opin Emerg Drugs 2017;22:137-48.
153. Sinha D, Duijf PHG, Khanna KK. Mitotic slippage: an old tale with a new twist. Cell Cycle 2019;18:7-15.
154. Conti G, Dias MH, Bernards R. Fighting Drug Resistance through the Targeting of Drug-Tolerant Persister Cells. Cancers 2021;13:1118.
155. Rodriguez R, Schreiber SL, Conrad M. Persister cancer cells: iron addiction and vulnerability to ferroptosis. Mol Cell 2022;82:728-40.
156. Poillet-Perez L, Sarry JE, Joffre C. Autophagy is a major metabolic regulator involved in cancer therapy resistance. Cell Rep 2021;36:109528.
157. Thijssen R, Diepstraten ST, Moujalled D, et al. Intact TP-53 function is essential for sustaining durable responses to BH3-mimetic drugs in leukemias. Blood 2021;137:2721-35.
158. Nechiporuk T, Kurtz SE, Nikolova O, et al. The TP53 apoptotic network is a primary mediator of resistance to BCL2 inhibition in AML cells. Cancer Discov 2019;9:910-25.
159. Kim K, Maiti A, Loghavi S, et al. Outcomes of TP53-mutant acute myeloid leukemia with decitabine and venetoclax. Cancer 2021;127:3772-81.
160. Antoszczak M, Müller S, Cañeque T, et al. Iron-sensitive prodrugs that trigger active ferroptosis in drug-tolerant pancreatic cancer cells. J Am Chem Soc 2022;144:11536-45.
161. Cui Z, Fu Y, Yang Z, et al. Comprehensive analysis of a ferroptosis pattern and associated prognostic signature in acute myeloid leukemia. Front Pharmacol 2022;13:866325.
162. Shao R, Wang H, Liu W, et al. Establishment of a prognostic ferroptosis-related gene profile in acute myeloid leukaemia. J Cell Mol Med 2021;25:10950-60.
163. Song Y, Tian S, Zhang P, Zhang N, Shen Y, Deng J. Construction and validation of a novel ferroptosis-related prognostic model for acute myeloid leukemia. Front Genet 2021;12:708699.
164. Zhang X, Zhang X, Liu K, et al. HIVEP3 cooperates with ferroptosis gene signatures to confer adverse prognosis in acute myeloid leukemia. Cancer Med 2022;11:5050-65.
165. Montalban-Bravo G, Class CA, Ganan-Gomez I, et al. Transcriptomic analysis implicates necroptosis in disease progression and prognosis in myelodysplastic syndromes. Leukemia 2020;34:872-81.
166. Bhatt S, Pioso MS, Olesinski EA, et al. Reduced mitochondrial apoptotic priming drives resistance to BH3 mimetics in acute myeloid leukemia. Cancer Cell 2020;38:872-890.e6.
167. Collignon A, Hospital MA, Montersino C, et al. A chemogenomic approach to identify personalized therapy for patients with relapse or refractory acute myeloid leukemia: results of a prospective feasibility study. Blood Cancer J 2020;10:64.
168. Kantarjian H, Kadia T, DiNardo C, et al. Acute myeloid leukemia: current progress and future directions. Blood Cancer J 2021;11:41.