Checkpoint inhibitors as potential therapeutics in acute myeloid leukemia
Abstract
Despite recent progress in treatment methods, acute myeloid leukemia (AML) continues to pose significant clinical
challenges and is associated with generally unfavorable prognoses. Patients considered fit for intensive therapy are
usually treated with cytarabine-anthracycline-based induction chemotherapy. Allogeneic hematopoietic stem cell
transplantation (allo-HSCT) is recommended for patients with adverse-risk AML and most patients with intermediaterisk
AML. The standard therapy for patients who are deemed too poorly for intensive treatment is the combination
of azacitidine and venetoclax. AML cells can escape the immune system through various mechanisms, including
reduced expression of MHC complex molecules, ligand shedding, manipulation of chemical signaling, and enhanced
inhibitory ligand expression. Increased expression of ligands for T-cell-regulation checkpoints is present in AML cells
and correlates with worse outcomes. Therefore, this article reviews the current research progress in immune checkpoint
inhibitors in AML.
Keywords: checkpoint inhibitorstherapyacute myeloid leukemiasurvival
References
- Wierzbowska A. Ostra białaczka szpikowa. In: Krzakowski M. ed. Zalecenia postępowania diagnostyczno-terapeutycznego w nowotworach złośliwych. Onkologia w Praktyce Klinicznej, Gdańsk. ; 2023.
- Strzałka P, Czemerska M, Krawiec K, et al. Characterization and prognostic factors of secondary to MDS/MPN and therapy-related AML: a single-center study. Acta Haematologica Polonica. 2023; 54(3): 176–186.
- Boscaro E, Urbino I, Catania FM, et al. Modern Risk Stratification of Acute Myeloid Leukemia in 2023: Integrating Established and Emerging Prognostic Factors. Cancers (Basel). 2023; 15(13).
- 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(12): 1345–1377.
- Brzozowski K, Pluta A, Włodarczyk M, et al. DAC and CLAM are equally effective as early second induction in newly diagnosed AML patients with persistent leukemia in early bone marrow evaluation – a retrospective analysis of Polish Adult Leukemia Group. Acta Haematologica Polonica. 2024; 55(5): 245–251.
- Holowiecki J, Grosicki S, Kyrcz-Krzemien S, et al. Cladribine in Combination with Standard Daunorubicine and Cytarabine (DAC) as a Remission Induction Treatment Improves the Overall Survival in Untreated Adults with AML Aged < 60 y Contrary to Combination Including Fludarabine (DAF): A Multicenter, Randomized, Phase III PALG AML 1/2004 DAC/DAF/DA Study in 673 Patients-A Final Update. Blood. 2009; 114(22): 2055–2055.
- Pluta A, Robak T, Wrzesien-Kus A, et al. Addition of cladribine to the standard induction treatment improves outcomes in a subset of elderly acute myeloid leukemia patients. Results of a randomized Polish Adult Leukemia Group (PALG) phase II trial. Am J Hematol. 2017; 92(4): 359–366.
- Budziszewska BK, Salomon-Perzyński A, Pruszczyk K, et al. Cladribine Combined with Low-Dose Cytarabine as Frontline Treatment for Unfit Elderly Acute Myeloid Leukemia Patients: Results from a Prospective Multicenter Study of Polish Adult Leukemia Group (PALG). Cancers (Basel). 2021; 13(16).
- Ali S, Dunmore HM, Karres D, et al. The EMA Review of Mylotarg (Gemtuzumab Ozogamicin) for the Treatment of Acute Myeloid Leukemia. Oncologist. 2019; 24(5): e171–e179.
- Shimony S, Stahl M, Stone RM. Acute myeloid leukemia: 2023 update on diagnosis, risk-stratification, and management. Am J Hematol. 2023; 98(3): 502–526.
- Rzetelska Z, Szczepaniak A, Gil L. Managing post-transplant relapse in FLT3-mutated AML with gilteritinib. Acta Haematologica Polonica. 2024; 55(5): 285–286.
- Numan Y, Abdel Rahman Z, Grenet J, et al. Gilteritinib clinical activity in relapsed/refractory FLT3 mutated acute myeloid leukemia previously treated with FLT3 inhibitors. Am J Hematol. 2022; 97(3): 322–328.
- DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N Engl J Med. 2020; 383(7): 617–629.
- Stankiewicz J, Demidowicz E, Bartoszewicz N, et al. Relapsed childhood acute myeloid leukemia: prognostic factors and outcomes: experience from a single oncology center. Acta Haematologica Polonica. 2023.
- DeWolf S, Tallman MS. How I treat relapsed or refractory AML. Blood. 2020; 136(9): 1023–1032.
- Weigert N, Rowe JM, Lazarus HM, et al. Consolidation in AML: Abundant opinion and much unknown. Blood Rev. 2022; 51: 100873.
- Molica M, Breccia M, Foa R, et al. Maintenance therapy in AML: The past, the present and the future. Am J Hematol. 2019; 94(11): 1254–1265.
- Mądry K, Budziszewska B, Lis K, et al. Treatment recommendations developed by MDS experts of the Polish Adult Leukemia Group (PALG) for management of myelodysplastic syndromes (MDSs) and other MDS-related conditions in Poland for 2021. Acta Haematologica Polonica. 2022; 53(2): 75–93.
- Khaldoyanidi S, Nagorsen D, Stein A, et al. Immune Biology of Acute Myeloid Leukemia: Implications for Immunotherapy. J Clin Oncol. 2021; 39(5): 419–432.
- Abaza Y, Zeidan AM. Immune Checkpoint Inhibition in Acute Myeloid Leukemia and Myelodysplastic Syndromes. Cells. 2022; 11(14).
- Bolkun L, Tynecka M, Walewska A, et al. The Association between Immune Checkpoint Proteins and Therapy Outcomes in Acute Myeloid Leukaemia Patients. Cancers (Basel). 2023; 15(18).
- Perna F, Espinoza-Gutarra MR, Bombaci G, et al. Immune-Based Therapeutic Interventions for Acute Myeloid Leukemia. Cancer Treat Res. 2022; 183: 225–254.
- Takahashi T, Tagami T, Yamazaki S, et al. Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med. 2000; 192(2): 303–310.
- Laurent S, Palmisano GL, Martelli AM, et al. Molecular characterization and applications of recombinant scFv antibodies to CD152 co-stimulatory molecule. Tissue Antigens. 2000; 55(3): 229–238.
- Zhong RK, Loken M, Lane TA, et al. CTLA-4 blockade by a human MAb enhances the capacity of AML-derived DC to induce T-cell responses against AML cells in an autologous culture system. Cytotherapy. 2006; 8(1): 3–12.
- Blazar BR, Taylor PA, Panoskaltsis-Mortari A, et al. Opposing roles of CD28:B7 and CTLA-4:B7 pathways in regulating in vivo alloresponses in murine recipients of MHC disparate T cells. J Immunol. 1999; 162(11): 6368–6377.
- Saad P, Kasi A. Ipilimumab. In: StatPearls. Treasure Island (FL): StatPearls Publishing; April. ; 10: 2023.
- Davids MS, Kim HT, Bachireddy P, et al. Leukemia and Lymphoma Society Blood Cancer Research Partnership. Ipilimumab for Patients with Relapse after Allogeneic Transplantation. N Engl J Med. 2016; 375(2): 143–153.
- Zeidan AM, Knaus HA, Robinson TM, et al. A Multi-center Phase I Trial of Ipilimumab in Patients with Myelodysplastic Syndromes following Hypomethylating Agent Failure. Clin Cancer Res. 2018; 24(15): 3519–3527.
- Bashey A, Medina B, Corringham S, et al. CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation. Blood. 2009; 113(7): 1581–1588.
- Murdock HM, Ho VT, Garcia JS, et al. Adding venetoclax to fludarabine/busulfan RIC transplant for high-risk MDS and AML is feasible, safe, and active. Blood Adv. 2021; 5(24): 5536–5545.
- 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(10): 6738–6746.
- Boussiotis VA. Molecular and Biochemical Aspects of the PD-1 Checkpoint Pathway. N Engl J Med. 2016; 375(18): 1767–1778.
- Latchman Y, Wood CR, Chernova T, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001; 2(3): 261–268.
- Ohaegbulam KC, Assal A, Lazar-Molnar E, et al. Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends Mol Med. 2015; 21(1): 24–33.
- Wang X, Teng F, Kong Li, et al. PD-L1 expression in human cancers and its association with clinical outcomes. Onco Targets Ther. 2016; 9: 5023–5039.
- Daver N, Basu S, Garcia-Manero G, et al. Defining the Immune Checkpoint Landscape in Patients (pts) with Acute Myeloid Leukemia (AML). Blood. 2016; 128(22): 2900–2900.
- Williams P, Basu S, Garcia-Manero G, et al. The distribution of T-cell subsets and the expression of immune checkpoint receptors and ligands in patients with newly diagnosed and relapsed acute myeloid leukemia. Cancer. 2019; 125(9): 1470–1481.
- Zhang L, Gajewski TF, Kline J. PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model. Blood. 2009; 114(8): 1545–1552.
- Zhou Q, Munger ME, Highfill SL, et al. Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia. Blood. 2010; 116(14): 2484–2493.
- Bołkun Ł, Starosz A, Krętowska-Grunwald A, et al. Effects of Combinatory In Vitro Treatment with Immune Checkpoint Inhibitors and Cytarabine on the Anti-Cancer Immune Microenvironment in De Novo AML Patients. Cancers (Basel). 2024; 16(2).
- Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol. 2018; 62: 29–39.
- Daver N, Basu S, Garcia-Manero G, et al. Phase IB/II Study of Nivolumab in Combination with Azacytidine (AZA) in Patients (pts) with Relapsed Acute Myeloid Leukemia (AML). Blood. 2016; 128(22): 763–763.
- Daver N, Garcia-Manero G, Basu S, et al. Efficacy, Safety, and Biomarkers of Response to Azacitidine and Nivolumab in Relapsed/Refractory Acute Myeloid Leukemia: A Nonrandomized, Open-Label, Phase II Study. Cancer Discov. 2019; 9(3): 370–383.
- Daver N, Garcia-Manero G, Konopleva M, et al. Azacitidine (AZA) with Nivolumab (Nivo), and AZA with Nivo + Ipilimumab (Ipi) in Relapsed/Refractory Acute Myeloid Leukemia: A Non-Randomized, Prospective, Phase 2 Study. Blood. 2019; 134(Supplement_1): 830–830.
- Ravandi F, Assi R, Daver N, et al. Idarubicin, cytarabine, and nivolumab in patients with newly diagnosed acute myeloid leukaemia or high-risk myelodysplastic syndrome: a single-arm, phase 2 study. Lancet Haematol. 2019; 6(9): e480–e488.
- Liu H, Sharon E, Karrison T, et al. Randomized Phase II Study to Assess the Role of Nivolumab As Single Agent to Eliminate Minimal Residual Disease and Maintain Remission in Acute Myelogenous Leukemia (AML) Patients after Chemotherapy (NCI9706 protocol; REMAIN Trial). Blood. 2022; 140(Supplement 1): 1716–1719.
- Gojo I, Stuart R, Webster J, et al. Multi-Center Phase 2 Study of Pembroluzimab (Pembro) and Azacitidine (AZA) in Patients with Relapsed/Refractory Acute Myeloid Leukemia (AML) and in Newly Diagnosed (≥65 Years) AML Patients. Blood. 2019; 134(Supplement_1): 832–832.
- Goswami M, Gui G, Dillon LW, et al. Pembrolizumab and decitabine for refractory or relapsed acute myeloid leukemia. J Immunother Cancer. 2022; 10(1).
- Zeidner JF, Vincent BG, Ivanova A, et al. Phase II Trial of Pembrolizumab after High-Dose Cytarabine in Relapsed/Refractory Acute Myeloid Leukemia. Blood Cancer Discov. 2021; 2(6): 616–629.
- Tschernia NP, Kumar V, Moore DT, et al. Safety and Efficacy of Pembrolizumab Prior to Allogeneic Stem Cell Transplantation for Acute Myelogenous Leukemia. Transplant Cell Ther. 2021; 27(12): 1021.e1–1021.e5.
- Zeidan A, Cavenagh J, Voso M, et al. Efficacy and Safety of Azacitidine (AZA) in Combination with the Anti-PD-L1 Durvalumab (durva) for the Front-Line Treatment of Older Patients (pts) with Acute Myeloid Leukemia (AML) Who Are Unfit for Intensive Chemotherapy (IC) and Pts with Higher-Risk Myelodysplastic Syndromes (HR-MDS): Results from a Large, International, Randomized Phase 2 Study. Blood. 2019; 134(Supplement_1): 829–829.
- Zeidan AM, Boss I, Beach CL, et al. A randomized phase 2 trial of azacitidine with or without durvalumab as first-line therapy for older patients with AML. Blood Adv. 2022; 6(7): 2219–2229.
- Zeidan AM, Boss I, Beach CL, et al. A randomized phase 2 trial of azacitidine with or without durvalumab as first-line therapy for older patients with AML. Blood Adv. 2022; 6(7): 2219–2229.
- Zheng H, Mineishi S, Claxton D, et al. A phase I clinical trial of avelumab in combination with decitabine as first line treatment of unfit patients with acute myeloid leukemia. Am J Hematol. 2021; 96(2): E46–E50.
- Saxena K, Herbrich SM, Pemmaraju N, et al. A phase 1b/2 study of azacitidine with PD-L1 antibody avelumab in relapsed/refractory acute myeloid leukemia. Cancer. 2021; 127(20): 3761–3771.
- Altman J, Bhatnagar B, Abedin S, et al. Gilteritinib Can be Safely Combined with Atezolizumab for the Treatment of Relapsed or Refractory FLT3-Mutated AML: Results of a Phase 1 Study. Blood. 2021; 138(Supplement 1): 2343–2343.
- Prebet T, Goldberg AD, Jurcic JG, et al. A phase 1b study of atezolizumab in combination with guadecitabine for the treatment of acute myeloid leukemia. Leuk Lymphoma. 2022; 63(9): 2180–2188.
- Das M, Zhu C, Kuchroo VK. Tim-3 and its role in regulating anti-tumor immunity. Immunol Rev. 2017; 276(1): 97–111.
- Li C, Yu X, Zhu Y, et al. TIM-3 Is Highly Expressed On Blast Cells In Patients With Acute Myeloid Leukemia. Blood. 2013; 122(21): 4931–4931.
- Tan J, Yu Z, Huang J, et al. Increased PD-1+Tim-3+ exhausted T cells in bone marrow may influence the clinical outcome of patients with AML. Biomark Res. 2020; 8: 6.
- Kikushige Y, Shima T, Takayanagi Si, et al. TIM-3 is a promising target to selectively kill acute myeloid leukemia stem cells. Cell Stem Cell. 2010; 7(6): 708–717.
- Zeidan AM, Komrokji RS, Brunner AM. TIM-3 pathway dysregulation and targeting in cancer. Expert Rev Anticancer Ther. 2021; 21(5): 523–534.
- Xu S, Zhang Na, Rinne ML, et al. Sabatolimab (MBG453) model-informed drug development for dose selection in patients with myelodysplastic syndrome/acute myeloid leukemia and solid tumors. CPT Pharmacometrics Syst Pharmacol. 2023; 12(11): 1653–1665.
- Brunner A, Borate U, Esteve J, et al. AML-190: Anti-TIM-3 Antibody MBG453 in Combination with Hypomethylating Agents (HMAs) in Patients with High-Risk Myelodysplastic Syndrome (HR-MDS) and Acute Myeloid Leukemia: A Phase 1 Study. Clinical Lymphoma Myeloma and Leukemia. 2020; 20: S188–S189.
- Brunner A, Traer E, Vey N, et al. Allogeneic Hematopoietic Cell Transplantation Outcomes of Patients with R/R AML or Higher-Risk MDS Treated with the TIM-3 Inhibitor MBG453 (Sabatolimab) and Hypomethylating Agents. Blood. 2021; 138(Supplement 1): 3677–3677.
- Zeidan A, Westermann J, Kovacsovics T, et al. AML-484 First Results of a Phase II Study (STIMULUS-AML1) Investigating Sabatolimab + Azacitidine + Venetoclax in Patients With Newly Diagnosed Acute Myeloid Leukemia (ND AML). Clinical Lymphoma Myeloma and Leukemia. 2022; 22: S255.
- Zeiser R, Devillier R, Mico' M, et al. TIM-3 Inhibitor Sabatolimab for Patients with Acute Myeloid Leukemia (AML) with Measurable Residual Disease (MRD) Detected after Allogeneic Stem Cell Transplantation (AlloSCT): Preliminary Findings from the Phase Ib/II Stimulus-AML2 Study. Blood. 2023; 142(Supplement 1): 59–59.
- Ming Q, Celias DP, Wu C, et al. LAG3 ectodomain structure reveals functional interfaces for ligand and antibody recognition. Nat Immunol. 2022; 23(7): 1031–1041.
- Workman CJ, Dugger KJ, Vignali DAA. Cutting edge: molecular analysis of the negative regulatory function of lymphocyte activation gene-3. J Immunol. 2002; 169(10): 5392–5395.
- Kouo T, Huang L, Pucsek AB, et al. Galectin-3 Shapes Antitumor Immune Responses by Suppressing CD8+ T Cells via LAG-3 and Inhibiting Expansion of Plasmacytoid Dendritic Cells. Cancer Immunol Res. 2015; 3(4): 412–423.
- Wang J, Sanmamed MF, Datar I, et al. Fibrinogen-like Protein 1 Is a Major Immune Inhibitory Ligand of LAG-3. Cell. 2019; 176(1-2): 334–347.e12.
- Chen Y, Tan J, Huang S, et al. Higher frequency of the CTLA-4 LAG-3 T-cell subset in patients with newly diagnosed acute myeloid leukemia. Asia Pac J Clin Oncol. 2020; 16(2): e12–e18.
- Chen C, Liang C, Wang S, et al. Expression patterns of immune checkpoints in acute myeloid leukemia. J Hematol Oncol. 2020; 13(1): 28.
- Abdelhakim H, Cortez L, Li M, et al. LAG3 Inhibition Decreases AML-Induced Immunosuppression and Improves T Cell-Mediated Killing. Blood. 2019; 134(Supplement_1): 3605–3605.
- Flieswasser T, Van den Eynde A, Van Audenaerde J, et al. The CD70-CD27 axis in oncology: the new kids on the block. J Exp Clin Cancer Res. 2022; 41(1): 12.
- Riether C, Schürch CM, Bührer ED, et al. CD70/CD27 signaling promotes blast stemness and is a viable therapeutic target in acute myeloid leukemia. J Exp Med. 2017; 214(2): 359–380.
- Pabst T, Vey N, Adès L, et al. Results from a phase I/II trial of cusatuzumab combined with azacitidine in patients with newly diagnosed acute myeloid leukemia who are ineligible for intensive chemotherapy. Haematologica. 2023; 108(7): 1793–1802.
- van Duijn A, Van der Burg SH, Scheeren FA. CD47/SIRPα axis: bridging innate and adaptive immunity. J Immunother Cancer. 2022; 10(7).
- Ostendorf BN, Flenner E, Flörcken A, et al. Phenotypic characterization of aberrant stem and progenitor cell populations in myelodysplastic syndromes. PLoS One. 2018; 13(5): e0197823.
- Majeti R, Chao MP, Alizadeh AA, et al. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 2009; 138(2): 286–299.
- Chao MP, Jaiswal S, Weissman-Tsukamoto R, et al. Calreticulin is the dominant pro-phagocytic signal on multiple human cancers and is counterbalanced by CD47. Sci Transl Med. 2010; 2(63): 63ra94.
- Liu J, Wang L, Zhao F, et al. Pre-Clinical Development of a Humanized Anti-CD47 Antibody with Anti-Cancer Therapeutic Potential. PLoS One. 2015; 10(9): e0137345.
- Gilead Sciences. Gilead Statement on Discontinuation of Phase 3 ENHANCE-3 Study in AML. 2024; available at:. https://www.gilead.com/company/company-statements/2024/gilead-statement-on-discontinuation-of-phase-3-enhance-3-study-in-aml..
- Garcia-Manero G, Przespolewski A, Abaza Y, et al. Evorpacept (ALX148), a CD47-Blocking Myeloid Checkpoint Inhibitor, in Combination with Azacitidine and Venetoclax in Patients with Acute Myeloid Leukemia (ASPEN-05): Results from Phase 1a Dose Escalation Part. Blood. 2022; 140(Supplement 1): 9046–9047.
- Qi J, Li J, Jiang B, et al. A Phase I/IIa Study of Lemzoparlimab, a Monoclonal Antibody Targeting CD47, in Patients with Relapsed and/or Refractory Acute Myeloid Leukemia (AML) and Myelodysplastic Syndrome (MDS): Initial Phase I Results. Blood. 2020; 136(Supplement 1): 30–31.