Skip to main content
Biology of acute leukemia
- Dores et al. Acute leukemia incidence and patient survival among children and adults in the United States, 2001-2007. Blood 2012;119:34
- The Cancer Genome Atlas Research Network. Genomic and Epigenomic Landscapes of Adult De Novo Acute Myeloid Leukemia. NEJM 2013;368:2059 (At least one “driver” mutation found in almost every case; average number of mutations per case around 13)
- Lindsley and Ebert. The biology and clinical impact of genetic lesions in myeloid malignancies. Blood 2013;122:3741
- Bodini et al. The hidden genomic landscape of acute myeloid leukemia: subclonal structure revealed by undetected mutations. Blood 2015;125:600
- Thomas and Majeti. Biology and relevance of human acute myeloid leukemia stem cells. Blood 2017;129:1577
- Ediriwickrema et al. Single-cell genomics in AML: extending the frontiers of AML research. Blood 2023;141:345
- Lane et al. The leukemic stem cell niche: current concepts and therapeutic opportunities. Blood 2009;114:1150
- Ferrando and López-Otín. Clonal evolution in leukemia. Nat Med 2017;23:1135
- Stauber et al. Preleukemic and leukemic evolution at the stem cell level. Blod 2021;137:1013
- Chen et al. Myelodysplastic syndrome progression to acute myeloid leukemia at the stem cell level. Nat Med 2019;25:103
- Ho et al. Evolution of acute myelogenous leukemia stem cell properties after treatment and progression. Blood 2016;128:1671
- Parkin et al. Clonal evolution and devolution after chemotherapy in adult acute myelogenous leukemia. Blood 2013;121:369 (Relapse is due to incomplete eradication of AML “founder clones”)
- Peterson et al. Acute myeloid leukemia with the 8q22;21q22 translocation: secondary mutational events and alternative t(8;21) transcripts. Blood 2007;110:799
- Wang et al. The Leukemogenicity of AML1-ETO Is Dependent on Site-Specific Lysine Acetylation. Science 2011;333:765 (“Lysine acetyltransferases represent a potential therapeutic target in AML”)
- Mardis et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. NEJM 2009; 361:1058
- Patel et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. NEJM 2012; 366:1079
- Sachs and Lotem. Control of programmed cell death in normal and leukemic cells: new implications for therapy. Blood 1993;82:15
- Pedersen-Bjergaard et al. Genetic pathways in therapy-related myelodysplasia and acute myelogenous leukemia. Blood 2002;99:1909
- Link et al. Identification of a Novel TP53 Cancer Susceptibility Mutation Through Whole-Genome Sequencing of a Patient With Therapy-Related AML. JAMA 2011;305:1568 (Inherited mutation predisposing to therapy-related AML)
- Wong et al. Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukemia. Nature 2014;518:552 (TP53 mutations present in a subset of hematopoietic stem cells prior to therapy predispose to t-AML)
- Wolfraim et al. Loss of Smad3 in Acute T-Cell Lymphoblastic Leukemia. NEJM 2004;351:552
- Mullighan et al. CREBBP mutations in relapsed acute lymphoblastic leukemia. Nature 2011;471:235
- Roberts et al. Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia. NEJM 2014;371:1005
- Godley and Le Beau. The histone code and treatments for acute myeloid leukemia. NEJM 2012;366:960
- Eppert et al. Stem cell gene expression programs influence clinical outcome in human leukemia. Nat Med 2011;17:1086 (Expression of “stem cell” genes by blasts associated with worse prognosis in AML)
- Abdel-Wahab and Levine. Mutations in epigenetic modifiers in the pathogenesis and therapy of acute myeloid leukemia. Blood 2013;121:3563
- Li et al. Distinct evolution and dynamics of epigenetic and genetic heterogeneity in acute myeloid leukemia. Nat Med 2016;22:792(With editorial)
- Delhommeau et al. Mutation in TET2 in myeloid cancers. NEJM 2009;360:2289 (Mutation an early event in AML, MDS, MPD. See also the accompanying editorial)
- Dey et al. Loss of the tumor suppressor BAP1 causes myeloid transformation. Science 2012;337:1541(With editorial)
- Wang et al. Targeted Inhibition of Mutant IDH2 in Leukemia Cells Induces Cellular Differentiation. Science 2013;340:622(with editorial)
- Chan et al. Isocitrate dehydrogenase 1 and 2 mutations induce BCL-2 dependence in acute myeloid leukemia. Nat Med 2015;21:178(potential “synthetic lethal” treatment approach; with editorial)
- Swaminathan et al. BACH2 mediates negative selection and p53-dependent tumor suppression at the pre-B cell receptor checkpoint. Nat Med 2013;19:1014(Identification of an important safeguard against leukemogenesis in B cells)
- Chantepie et al. Hematogones: a new prognostic factor for acute myeloblastic leukemia. Blood 2011;117:1315
- Gurbuxani et al. Recent insights into the mechanisms of myeloid leukemogenesis in Down syndrome. Blood 2004;103:399
- Hole et al. Do reactive oxygen species play a role in myeloid leukemias? Blood 2011;117:5816
- Dawson et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 2011;478:529
- Placke et al. Requirement for CDK6 in MLL-rearranged acute myeloid leukemia. Blood 2014;124:13
- Chen et al. DOT1L inhibits SIRT1-mediated epigenetic silencing to maintain leukemic gene expression in MLL-rearranged leukemia. Nat Med 2015;21:335
- Liang et al. Therapeutic Targeting of MLL Degradation Pathways in MLL-Rearranged Leukemia. Cell 2017;168:59(Stabilizing wild-type MLL protein inhibits leukemia cell proliferation; see also NEJM commentaryon this research)
- Walter et al. Clonal architecture of secondary acute myeloid leukemia. NEJM 2012;366:1090(85% of marrow cells clonal; founding clone with hundreds of mutations and subclones with many more)
- Makishima et al. Mutations in the spliceosome machinery, a novel and ubiquitous pathway in leukemogenesis. Blood 2012;119:3203
- Damm et al. Mutations affecting mRNA splicing define distinct clinical phenotypes and correlate with patient outcome in myelodysplastic syndromes. Blood 2012;119:3211
- Senapati et al. Venetoclax abrogates the prognostic impact of splicing factor gene mutations in newly diagnosed acute myeloid leukemia. Blood 2023;142:1647
- Mansour et al. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element. Science 2014;346:1373(Mutation enhances binding of MYB transcription factor, enhancing TAL1 expression in T-ALL; with editorial)
- Esposito et al. Synthetic lethal targeting of oncogenic transcription factors in acute leukemia by PARP inhibitors. Nat Med 2015;21:1481(With editorial)
- Fucikova et al. Calreticulin exposure by malignant blasts correlates with robust anticancer immunity and improved clinical outcome in AML patients. Blood 2016;128:3113
- Schneider et al. SAMHD1 is a biomarker for cytarabine response and a therapeutic target in acute myeloid leukemia. Nat Med 2017;23;250
- Herold et al. Targeting SAMHD1 with the Vpx protein to improve cytarabine therapy for hematological malignancies. Nat Med 2017;23:256
- Göllner et al. Loss of the histone methyltransferase EZH2 induces resistance to multiple drugs in acute myeloid leukemia. Nat Med 2017;234: 23:69
- Guryanova et al. DNMT3A mutations promote anthracycline resistance in acute myeloid leukemia via impaired nucleosome remodeling. Nat Med 2016;22:1488
- Vu et al. The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat Med 2017;23:1369(With editorial)
- Colom Díaz et al. Hematopoietic stem cell aging and leukemia transformation. Blood 2023;142:542
Inherited myeloid malignancy
- Brown et al. Secondary leukemia in patients with germline transcription factor mutations (RUNX1, GATA2, CEBPA). Blood 2020;136:24
- Godley and Shimamura. Genetic predisposition to hematologic malignancies: management and surveillance. Blood 2017;130:424
- Yang et al. Identification and prioritization of myeloid malignancy germline variants in a large cohort of adult patients with AML. Blood 2022;139:1208 (13.6% of adult AML patients have potentially pathogeneic germline variants)
- Kohlmann and Schiffman. Discussing and managing hematologic germ line variants. Blood 2016;128:2497
- Drazer et al. How I diagnose and manage individuals at risk for inherited myeloid malignancies. Blood 2016;128:1800
- Churpek et al. Genomic analysis of germ line and somatic variants in familial myelodysplasia/acute myeloid leukemia. Blood 2015;126:2484
- Shinawi et al. Syndromic thrombocytopenia and predisposition to acute myelogenous leukemia caused by constitutional microdeletions on chromosome 21q. Blood 2008;112:1042
- Homan et al. Hereditary platelet disorders associated with germ line variants in RUNX1, ETV6, and ANKRD26. Blood 2023;141:1533
- Simon et al. High frequency of germline RUNX1 mutations in patients with RUNX1-mutated AML. Blood 2020;135:1882
- Cunningham et al. Natural history study of patients with familial platelet disorder with associated myeloid malignancy. Blood 2023;142:2146 (RUNX1)
- Wahlster et al. ANKRD26-related thrombocytopenia 2 with a baseline increase in blasts: implications for clinical surveillance. Blood 2025;146:254 (Increased marrow blasts without progression to myeloid malignancy)
- Crispino and Horwitz. GATA factor mutations in hematologic disease. Blood 2017;129:2103
- Spinner et al. GATA2 deficiency: a protean disorder of hematopoiesis, lymphatics, and immunity. Blood 2014;123:809
- Calvo and Hickstein. The spectrum of GATA2 deficiency syndrome. Blood 2023;142:1524
- Vinh et al. Autosomal dominant and sporadic monocytopenia with susceptibility to mycobacteria, fungi, papillomaviruses, and myelodysplasia. Blood 2010;115:1519 (GATA-2 deficiency)
- Dickinson et al. Exome sequencing identifies GATA-2 mutation as the cause of dendritic cell, monocyte, B and NK lymphoid deficiency. Blood 2011;118:2656 (mycobacterial infection, pulmonary alveolar proteinosis, MDS and AML)
- Hsu et al. Mutations in GATA2 are associated with the autosomal dominant and sporadic monocytopenia and mycobacterial infection (MonoMAC) syndrome. Blood 2011;118:2653
- Ganapathi et al. GATA2 deficiency-associated bone marrow disorder differs from idiopathic aplastic anemia. Blood 2015;125:56
- Schratz et al. Cancer spectrum and outcomes in the Mendelian short telomere syndromes. Blood 2020;135:1946
- Duployez et al. Prognostic impact of DDX41 germline mutations in intensively treated acute myeloid leukemia patients: an ALFA-FILO study. Blood 2022;140:756 (Male predominance, older, low WBC, better response to intensive chemo; with commentary)
- Makishima et al. Germ line DDX41 mutations define a unique subtype of myeloid neoplasms. Blood 2023;141:534 (Lifelong risk of AML 50%; explains 80% of germ line predisposition to AML)
- Makishima et al. DDX41-associated susceptibility to myeloid neoplasms. Blood 2023;141:1544
- Kovilakam et al. Prevalence and significance of DDX41 gene variants in the general population. Blood 2023;142:1185
- Kraft and Godley. Identifying potential germline variants from sequencing hematopoietic malignancies. Blood 2020;136:2498
- Bick et al Inherited causes of clonal haematopoiesis in 97,691 whole genomes. Nature 2020;586:763
- Feurstein et al. Germ line predisposition variants occur in myelodysplastic syndrome patients of all ages. Blood 2022;140:2533 (7% of MDS patients had potentially pathogenic germline variants)
- Reilly and Shimamura. Predisposition to myeloid malignancies in Shwachman-Diamond syndrome: biological insights and clinical advances. Blood 2023;141:1513
- Hakkarainen et al. The clinical picture of ERCC6L2 disease: from bone marrow failure to acute leukemia. Blood 2023;141:2853
- Zerella et al. Germ line ERG haploinsufficiency defines a new syndrome with cytopenia and hematological malignancy predisposition. Blood 2024;144:1765
AML: General
- Arber et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016;127:2391
- Döhner 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
- Arber et al. International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: integrating morphologic, clinical, and genomic data. Blood 2022;140:1200
- DiNardo et al. Acute myeloid leukaemia. Lancet 2023;401:2073
- Döhner et al. Acute myeloid leukemia. NEJM 2015;373:1136
- Walter et al. Significance of FAB subclassification of “acute myeloid leukemia, NOS” in the 2008 WHO classification: analysis of 5848 newly diagnosed patients. Blood 2013;121:2424(Morphology does not provide independent prognostic information in AML if molecular features are known)
- Kern et al. Early blast clearance by remission induction therapy is a major independent prognostic factor for both achievement of complete remission and long-term outcome in acute myeloid leukemia: data from the German AML Cooperative Group (AMLCG) 1992 Trial. Blood 2003;101:64
- Elliott et al. Early peripheral blood blast clearance during induction chemotherapy for acute myeloid leukemia predicts superior relapse-free survival. Blood 2007;110:4172
- Buccisano et al. Prognostic and therapeutic implications of minimal residual disease detection in acute myeloid leukemia. Blood 2012;119:332
- Jourdan et al. Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia. Blood 2013;121:2213(MRD best predictor of relapse)
- Ivey et al. Assessment of Minimal Residual Disease in Standard-Risk AML. NEJM 2016;374:422(With editorial)
- Schuurhuis et al. Minimal/measurable residual disease in AML: a consensus document from the European LeukemiaNet MRD Working Party. Blood 2018;131:1275
- Jongen-Lavrencic et al. Molecular Minimal Residual Disease in Acute Myeloid Leukemia. NEJM 2018;378:1189(MRD found during CR, but not persistence of mutations associated with clonal hematopoiesis, predicted relapse; with editorial)
- Dillon et al. DNA Sequencing to Detect Residual Disease in Adults With Acute Myeloid Leukemia Prior to Hematopoietic Cell Transplant. JAMA 2023:329:745 (Marked increase in relapse rate and decreased survival if MRD detected prior to HSCT)
- Hasserjian et al. Clonal hematopoiesis and measurable residual disease assessment in acute myeloid leukemia. Blood 2020;135:1729
- Wang et al. In adults with t(8;21)AML, posttransplant RUNX1/RUNX1T1-based MRD monitoring, rather than c-KIT mutations, allows further risk stratification. Blood 2014;124:1880
- Derolf et al. Improved patient survival for acute myeloid leukemia: a population-based study of 9729 patients diagnosed in Sweden between 1973 and 2005. Blood 2009;113: 3666(5 yr “relative survival” rates 65% for age <18, 58% for ages 19-41, 36% for ages 41-60, 15% for ages 61-70, 5% for ages 71-80, 1% for age >80)
- Kayser et al. The impact of therapy-related acute myeloid leukemia (AML) on outcome in 2853 adult patients with newly diagnosed AML. Blood 2011;117:2137
- Voso et al. What’s new in the pathogenesis and treatment of therapy-related myeloid neoplasms. Blood 2021;138:749
- Morton et al. Evolving risk of therapy-related acute myeloid leukemia following cancer chemotherapy among adults in the United States, 1975-2008. Blood 2013;121:2996
- Nardi et al. Acute Myeloid Leukemia and Myelodysplastic Syndromes After Radiation Therapy Are Similar to De Novo Disease and Differ From Other Therapy-Related Myeloid Neoplasms. J Clin Oncol 2012;30:2340
- Matutes et al. Mixed-phenotype acute leukemia: clinical and laboratory features and outcome in 100 patients defined according to the WHO 2008 classification. Blood 2011;117:3163(MPAL is a poor risk disease; authors recommend transplantation in 1st remission)
- Vasu et al. Ten-year outcome of patients with acute myeloid leukemia not treated with allogeneic transplantation in first complete remission. Blood Adv 2018;2:1645(16.6% of patients < 60 and 2.4% of those >60 disease free at 10 yrs; disease free survivors usually had CBF mutations)
- Mishra et al. Metabolism in acute myeloid leukemia: mechanistic insights and therapeutic targets. Blood 2023;141:1119
AML: clinical implications of molecular and cytogenetic changes
Prognosis
- Grimwade et al. Molecular landscape of acute myeloid leukemia in younger adults and its clinical relevance. Blood 2016;127:29
- Döhner and Gaidzik. Impact of genetic features on treatment decisions in AML. Hematology 2011;36
- Rockova et al. Risk stratification of intermediate-risk acute myeloid leukemia: integrative analysis of a multitude of gene mutation and gene expression markers. Blood 2011;118:1069
- Damm et al. Integrative prognostic risk score in acute myeloid leukemia with normal karyotype. Blood 2011;117:4561
- Klco et al. Association Between Mutation Clearance After Induction Therapy and Outcomes in Acute Myeloid Leukemia. JAMA 2015;314:811(Persistence of AML-assosiated mutations in remission marrow → worse prognosis)
- Mrózek and Bloomfield. Chromosome Aberrations, Gene Mutations and Expression Changes, and Prognosis in Adult Acute Myeloid Leukemia. Hematology 2006;169-77
- Mrózek et al. Clinical relevance of mutations and gene-expression changes in adult acute myeloid leukemia with normal cytogenetics: are we ready for a prognostically prioritized molecular classification? Blood 2007;109:431
- Welch and Link. Genomics of AML: clinical applications of next-generation sequencing. Hematology 2011;30
- Pikman and Stegmaier. Targeted therapy for fusion-driven high-risk acute leukemia. Blood 2018;132:1241
- Østgård et al. Epidemiology and Clinical Significance of Secondary and Therapy-Related Acute Myeloid Leukemia: A National Population-Based Cohort Study. J Clin Oncol 2015;33:3641(AML following CMML or MPD has dismal prognosis)
- Röllig et al. Does time from diagnosis to treatment affect the prognosis of patients with newly diagnosed acute myeloid leukemia? Blood 2020;136:823(No)
Cytogenetics
- Moorman et al. Karyotype is an independent prognostic factor in adult acute lymphoblastic leukemia (ALL): analysis of cytogenetic data from patients treated on the Medical Research Council (MRC) UKALLXII/Eastern Cooperative Oncology Group (ECOG) 2993 trial. Blood 2007;109:3189
- Byrd et al. Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461). Blood 2002;100:4325
- Marcucci et al. Abnormal Cytogenetics at Date of Morphologic Complete Remission Predicts Short Overall and Disease-Free Survival, and Higher Relapse Rate in Adult Acute Myeloid Leukemia: Results From Cancer and Leukemia Group B Study 8461. J Clin Oncol 2004;22:2410
- Farag et al. Pretreatment cytogenetics add to other prognostic factors predicting complete remission and long-term outcome in patients 60 years of age or older with acute myeloid leukemia: results from Cancer and Leukemia Group B 8461. Blood 2006;108:63
- Perrot et al. Dismal prognostic value of monosomal karyotype in elderly patients with acute myeloid leukemia: a GOELAMS study of 186 patients with unfavorable cytogenetic abnormalities. Blood 2011;118:679
- Haferlach et al. Prognostic value of monosomal karyotype in comparison to complex aberrant karyotype in acute myeloid leukemia: a study on 824 cases with aberrant karyotype. Blood 2012;119:2122(Monosomy or 4 or more cytogenetic abnormalities associated with very poor outcome)
- Kayser et al. Monosomal karyotype in adult acute myeloid leukemia: prognostic impact and outcome after different treatment strategies. Blood 2012;119:551(Poor response to induction therapy, 9% 4-year survival)
- Medeiros et al. Prognostic impact of monosomal karyotype in young adult and elderly acute myeloid leukemia: the Southwest Oncology Group (SWOG) experience. Blood 2010;116:2224(3% 4 yr survival!)
- Bacher et al. Multilineage dysplasia does not influence prognosis in CEBPA-mutated AML, supporting the WHO proposal to classify these patients as a unique entity. Blood 2012;119:4719
- Grimwade et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood 2010;116:354
- Middeke et al. Outcome of patients with abnl(17p) acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation. Blood 2014;123:2960(High relapse rate, low 3 year OS)
- Herold et al. Isolated trisomy 13 defines a homogeneous AML subgroup with high frequency of mutations in spliceosome genes and poor prognosis. Blood 2014;124:1304
Molecular analysis
- The Cancer Genome Atlas Research Network. Genomic and Epigenomic Landscapes of Adult De Novo Acute Myeloid Leukemia. NEJM 2013;368:2059(At least one “driver” mutation found in almost every case; average number of mutations per case around 13)
- Papaemmanuil et al. Genomic classification and prognosis in acute myeloid leukemia. NEJM 2016;374:2209(>5000 driver mutations identified; with editorial)
- Kewan et al. Molecular patterns identify distinct subclasses of myeloid neoplasia. Nat Comm 2023;14:3136
- Itzykson et al. Genetic identification of patients with AML older than 60 years achieving long-term survival with intensive chemotherapy. Blood 2021;138:507
- Metzeler et al. Spectrum and prognostic relevance of driver gene mutations in acute myeloid leukemia. Blood 2016;128:686(NPM1, FLT3, CEBPA, TP53, DNMT3A and RUNX1 most important risk factors)
- Klco et al. Association Between Mutation Clearance After Induction Therapy and Outcomes in Acute Myeloid Leukemia. JAMA 2015;314:811(Persistence of AML-assosiated mutations in remission marrow → worse prognosis)
- Grossman et al. A novel hierarchical prognostic model of AML solely based on molecular mutations. Blood 2012;120:2963
- Ommen et al. Strikingly different molecular relapse kinetics in NPM1c, PML-RARA, RUNX1-RUNX1T1, and CBFB-MYH11 acute myeloid leukemias. Blood 2010;115:198
- Lindsley et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood 2015;125:1367(Mutations in SRSF2, SF3B1, U2AF1, ZRSR2, ASXL1, EZH2, BCOR, or STAG2 associated with secondary AML, worse prognosis)
- Cairoli et al. Prognostic impact of c-KIT mutations in core binding factor leukemias: an Italian retrospective study. Blood 2006;107:3463
- Levis M. FLT3 mutations in acute myeloid leukemia: what is the best approach in 2013? Hematology 2013:220
- Gale et al. The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood 2008;111:2776(high FLT3 mutant level a strong negative prognostic factor)
- Ostronoff et al. Prognostic Significance of NPM1 Mutations in the Absence of FLT3–Internal Tandem Duplication in Older Patients With Acute Myeloid Leukemia: A SWOG and UK National Cancer Research Institute/Medical Research Council Report. J Clin Oncol 2015;33:1157 (NMP1 positive, FLT3-ITD negative genotype has favorable impact in patients 55-65, not in older patients)
- Bacher et al. Prognostic relevance of FLT3-TKD mutations in AML: the combination matters—an analysis of 3082 patients. Blood 2008;111:2527
- Bullinger et al. An FLT3 gene-expression signature predicts clinical outcome in normal karyotype AML. Blood 2008;111:4490(gene expression signature associated with FLT3 gene mutation a better predictor of outcome than FLT3 mutation status itself)
- Kayser et al. Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is associated with resistance to chemotherapy and inferior outcome. Blood 2009;114:2386
- Levis M. FLT3/ITD AML and the law of unintended consequences. Blood 2011;117:6987
- Falini et al. Cytoplasmic Nucleophosmin in Acute Myelogenous Leukemia with a Normal Karyotype. NEJM 2005;352:254
- Schnittger et al. Nucleophosmin gene mutations are predictors of favorable prognosis in acute myelogenous leukemia with a normal karyotype. Blood 2005;106:3733
- Schlenk et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. NEJM 2008;358:1909(mutations of nucleophosmin or CEBPA genes in the absence of FLT3-ITD mutations conferred better prognosis; with editorial)
- Falini et al. Acute myeloid leukemia with mutated nucleophosmin (NPM1): is it a distinct entity? Blood 2011;117:1109
- Falini et al NPM1-mutated acute myeloid leukemia: from bench to bedside. Blood 2020;136:1707
- Gaidzik et al. RUNX1 Mutations in Acute Myeloid Leukemia: Results From a Comprehensive Genetic and Clinical Analysis From the AML Study Group. J Clin Oncol 2011;29:1364
- Sood et al. Role of RUNX1 in hematological malignancies. Blood 2017;129:2070
- Shen et al. Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia. Blood 2011;118:5593
- Wouters and Delwel. Epigenetics and approaches to targeted epigenetic therapy in acute myeloid leukemia. Blood 2016;127:42
- Marcucci et al. MicroRNA expression in cytogenetically normal acute myeloid leukemia. NEJM 2008;358:1919(microRNA signature identified that gave improved prognosis in AML patients with high-risk molecular features; with editorial)
- Marcucci et al. The prognostic and functional role of microRNAs in acute myeloid leukemia. Blood 2011;117:1121
- Allan et al. Genetic variation in XPD predicts treatment outcome and risk of acute myeloid leukemia following chemotherapy. Blood 2004;104:3872
- Zuber et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 2011;478:524
- Kharas et al. Musashi-2 regulates normal hematopoiesis and promotes aggressive myeloid leukemia. Nat Med 2010;16:903
- Ley et al. DNMT3A mutations in acute myeloid leukemia. NEJM 2010;363:2424(DNA methyltransferase gene mutations found in one-third of those with intermediate-risk cytogenetic profile and were an independent predictor of poor outcome)
- Schnittger et al. IDH1 mutations are detected in 6.6% of 1414 AML patients and are associated with intermediate risk karyotype and unfavorable prognosis in adults younger than 60 years and unmutated NPM1 status. Blood 2010;116:5486
- Chou et al. TET2 mutation is an unfavorable prognostic factor in acute myeloid leukemia patients with intermediate-risk cytogenetics. Blood 2011;118:3803
- Metzeler et al. TET2 Mutations Improve the New European LeukemiaNet Risk Classification of Acute Myeloid Leukemia: A Cancer and Leukemia Group B Study. J Clin Oncol 2011;29:1373
- Quek et al. Clonal heterogeneity of acute myeloid leukemia treated with the IDH2 inhibitor enasidenib. Nat Med 2018;24:1167(Relapse via clonal evolution or selection of resistant clones, not via new IDH2 mutations)
- Tyner et al. Functional genomic landscape of acute myeloid leukaemia. Nature 2018;562:526(Response to specific drugs linked to mutational status)
Gene expression profiling
- Gentles et al. Association of a Leukemic Stem Cell Gene Expression Signature With Clinical Outcomes in Acute Myeloid Leukemia. JAMA 2010;304:2706
- Eisfeld et al. miR-3151 interplays with its host gene BAALC and independently affects outcome of patients with cytogenetically normal acute myeloid leukemia. Blood 2012;120:249.
- Gönen et al. CD25 expression status improves prognostic risk classification in AML independent of established biomarkers: ECOG phase 3 trial, E1900. Blood 2012;120:2297
- Bullinger et al. Use of Gene-Expression Profiling to Identify Prognostic Subclasses in Adult Acute Myeloid Leukemia. NEJM 2004;350:1605
- Haferlach et al. Global approach to the diagnosis of leukemia using gene expression profiling. Blood 2005;106:1189
- Valk et al. Prognostically Useful Gene-Expression Profiles in Acute Myeloid Leukemia. NEJM 2004;350:1617
- Wouters et al. A decade of genome-wide gene expression profiling in acute myeloid leukemia: flashback and prospects. Blood 2009;113:291
AML in the elderly
- Ossenkoppele and Löwenberg. How I treat the older patient with acute myeloid leukemia. Blood 2015;125:767
- Appelbaum et al. Age and acute myeloid leukemia. Blood 2006:107:3481 (Poor performance status + advanced age = 80% mortality within 30 days of starting Rx)
- Walter et al. Prediction of Early Death After Induction Therapy for Newly Diagnosed Acute Myeloid Leukemia With Pretreatment Risk Scores: A Novel Paradigm for Treatment Assignment. J Clin Oncol 2011;29:4417 (PS and age most important predictors of early death; in a multicomponent model age appears to be a surrogate for other risk factors)
- Bazinet et al. Evolving trends and outcomes in older patients with acute myeloid leukemia including allogeneic stem cell transplantation. Am J Hematol 2023;98:1383 (Adding venetoclax to low-intensity therapy increases CR rate from 48% to 72%. Frontline SCT improved OS in this retrospective single center study)
- Klepin et al. Geriatric assessment predicts survival for older adults receiving induction chemotherapy for acute myelogenous leukemia. Blood 2013;121:4287
- Fröhling et al. Cytogenetics and age are major determinants of outcome in intensively treated acute myeloid leukemia patients older than 60 years: results from AMLSG trial AML HD98-B. Blood 2006;108:3280(Patients >70 and those with unfavorable karyotype had median survival of 7 mo, 3 year overal survival 6%)
- Döhner et al. Genetic risk classification for adults with AML receiving less-intensive therapies: the 2024 ELN recommendations. Blood 2024;144:2169
- Döhner et al. Genetic risk stratification and outcomes among treatment-naive patients with AML treated with venetoclax and azacitidine. Blood 2024;144:2211
- Löwenberg et al. High-Dose Daunorubicin in Older Patients with Acute Myeloid Leukemia. NEJM 2009;361:1235(Higher CR rate, no more toxicity with 90 mg/m2 daunorubicin vs 45 mg/m2)
- Gardin et al. Superior Long-Term Outcome With Idarubicin Compared With High-Dose Daunorubicin in Patients With Acute Myeloid Leukemia Age 50 Years and Older. J Clin Oncol 2013;31:321
- Fehniger et al. A phase 2 study of high-dose lenalidomide as initial therapy for older patients with acute myeloid leukemia. Blood 2011;117:1828(30% response rate)
- Cashen et al. Multicenter, Phase II Study of Decitabine for the First-Line Treatment of Older Patients With Acute Myeloid Leukemia. J Clin Oncol 2010;28:556(24% CR rate; medial OS 7.7 mo)
- Fenaux et al. Azacitidine Prolongs Overall Survival Compared With Conventional Care Regimens in Elderly Patients With Low Bone Marrow Blast Count Acute Myeloid Leukemia. J Clin Oncol 2020;28:562(Azacitidine prolonged survival and reduced time spent in hospital)
- Dombret 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(Azacitidine increased median OS by about 4 mo compared to standard AML Rx)
- Nand et al. A phase 2 trial of azacitidine and gemtuzumab ozogamicin therapy in older patients with acute myeloid leukemia. Blood 2013;122:3432(44% of good-risk, 35% of poor-risk patients had CR)
- Huls et al. Azacitidine maintenance after intensive chemotherapy improves DFS in older AML patients. Blood 2019;133:1457
- Burnett et al. Clofarabine doubles the response rate in older patients with acute myeloid leukemia but does not improve survival. Blood 2013;122:1384
- Attar et al. Bortezomib Added to Daunorubicin and Cytarabine During Induction Therapy and to Intermediate-Dose Cytarabine for Consolidation in Patients With Previously Untreated Acute Myeloid Leukemia Age 60 to 75 Years: CALGB (Alliance) Study 10502. J Clin Oncol 2013;31:923(65% CR rate)
- Erba H. Prognostic factors in elderly patients with AML and the implications for treatment. Hematology 2007:429
- Krug et al. Complete remission and early death after intensive chemotherapy in patients aged 60 years or older with acute myeloid leukaemia: a web-based application for prediction of outcomes. Lancet 2010;376:2000(Web site – requires password)
- Prébet et al. Acute Myeloid Leukemia With Translocation (8;21) or Inversion (16) in Elderly Patients Treated With Conventional Chemotherapy: A Collaborative Study of the French CBF-AML Intergroup. J Clin Oncol 2009;27:4747(88% CR rate, 31% 5 year OS)
- Juliusson et al. Age and acute myeloid leukemia: real world data on decision to treat and outcomes from the Swedish Acute Leukemia Registry. Blood 2009; 113:4179(“Most AML patients up to 80 should be considered for intensive therapy”)
- Kantarjian et al. Intensive chemotherapy does not benefit most older patients (age 70 years or older) with acute myeloid leukemia. Blood 2010;116:4422(Median survival 4.6 mo, 8-week mortality 36%, 1-year survival 28%)
- Rowe et al. A phase 3 study of three induction regimens and of priming with GM-CSF in older adults with acute myeloid leukemia: a trial by the Eastern Cooperative Oncology Group. Blood 2004;103:479 (No difference among three different anthracyclines; no benefit from GM-CSF priming)
- Sekeres et al. Time from diagnosis to treatment initiation predicts survival in younger, but not older, acute myeloid leukemia patients. Blood 2009;113;28(Delaying treatment not harmful in elderly patients)
- Gardin et al. Postremission treatment of elderly patients with acute myeloid leukemia in first complete remission after intensive induction chemotherapy:results of the multicenter randomized Acute Leukemia French Association (ALFA) 9803 trial. Blood 2007;109:5129(6 cycles of outpatient treatment more successful than single course of high dose consolidation treatment)
- Clavio et al. Adding low-dose gemtuzumab ozogamicin to fludarabine, Ara-C and idarubicin (MY-FLAI) may improve disease-free and overall survival in elderly patients with non-M3 acute myeloid leukaemia: results of a prospective, pilot, multi-centre trial and comparison with a historical cohort of patients. Br J Haematol 2007; 138:186
- Burnett et al. Addition of Gemtuzumab Ozogamicin to Induction Chemotherapy Improves Survival in Older Patients With Acute Myeloid Leukemia. J Clin Oncol 2012;30:3924
- Amadori et al. Sequential Combination of Gemtuzumab Ozogamicin and Standard Chemotherapy in Older Patients With Newly Diagnosed Acute Myeloid Leukemia: Results of a Randomized Phase III Trial by the EORTC and GIMEMA Consortium (AML-17). J Clin Oncol 2013;31:4424(Adding GO produced no apparent benefit)
- Kadia et al. Cladribine and low-dose cytarabine alternating with decitabine as front-line therapy for elderly patients with acute myeloid leukaemia: a phase 2 single-arm trial. Lancet Haematol 2018;5:e411(58% CR rate, well-tolerated)
- Quintás-Cardama et al. Epigenetic therapy is associated with similar survival compared with intensive chemotherapy in older patients with newly diagnosed acute myeloid leukemia. Blood 2012;120:4840
- Tassara et al. Valproic acid in combination with all-trans retinoic acid and intensive therapy for acute myeloid leukemia in older patients. Blood 2014;123:4027(No difference in EFS or OS, but better PFS with addition of valproate)
- DiNardo et al. Safety and preliminary efficacy of venetoclax with decitabine or azacitidine in elderly patients with previously untreated acute myeloid leukaemia: a non-randomised, open-label, phase 1b study. Lancet Oncol 2018;19:216
- DiNardo et al. Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. NEJM 2020;383:617(With editorial)
- Dinardo et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood 2019;133:7(67% CR rate, median OS 17.5 mo)
- Pollyea et al. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat Med 2018;24:1859
- Wei et al. Venetoclax Combined With Low-Dose Cytarabine for Previously Untreated Patients With Acute Myeloid Leukemia: Results From a Phase Ib/II Study. J Clin Oncol 2019;37:1277
- Pigneux et al. Improved Survival by Adding Lomustine to Conventional Chemotherapy for Elderly Patients With AML Without Unfavorable Cytogenetics: Results of the LAM-SA 2007 FILO Trial. J Clin Oncol 2018;36:3203
- Swords et al. Pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, combined with azacitidine in patients with AML. Blood 2018;131:1415
- Fathi et al. A phase 1 trial of vadastuximab talirine combined with hypomethylating agents in patients with CD33-positive AML. Blood 2018;132:1125(70% CR rate in intermediate- and high risk AML, median age 75)
- Amadori et al. Gemtuzumab Ozogamicin Versus Best Supportive Care in Older Patients With Newly Diagnosed Acute Myeloid Leukemia Unsuitable for Intensive Chemotherapy: Results of the Randomized Phase III EORTC-GIMEMA AML-19 Trial. J Clin Oncol 2016;34:972(GO prolonged survival with “manageable” toxicity)
- de Botton et al. Enasidenib vs conventional care in older patients with late-stage mutant-IDH2 relapsed/refractory AML: a randomized phase 3 trial. Blood 2023;141:156 (Modest improvement in OS)
- DiNardo et al. Mutant Isocitrate Dehydrogenase 1 Inhibitor Ivosidenib in Combination With Azacitidine for Newly Diagnosed Acute Myeloid Leukemia. J Clin Oncol 2021;39:57
- Pigneux et al. Addition of Androgens Improves Survival in Elderly Patients With Acute Myeloid Leukemia: A GOELAMS Study. J Clin Oncol 2017;35:387(Adding androgen improved 5 year OS from 17% to 26% in patients 60+)
AML variants/subtypes
- Olopade et al. Clinical, morphologic and cytogenetic characteristics of 26 patients with acute erythroblastic leukemia. Blood 1992;80:2873
- Roumer et al. M0 AML, clinical and biologic features of the disease, including AML1 gene mutations: a report of 50 cases bye the Groupe Francais d’Hematologie Cellulaire (GFHC) and the Groupe Francais de Cytogenetiquie Hematologique (GFCH). Blood 2003;101:1277
- Hasserjian et al. Acute erythroid leukemia: a reassessment using criteria refined in the 2008 WHO classification. Blood 2010;115:1985
- Oki et al. Adult acute megakaryocytic leukemia: an analysis of 37 patients treated at M.D. Anderson Cancer Center. Blood 2006;107:880
- Weinberg et al. Clinical characterization of acute myeloid leukemia with myelodysplasia-related changes as defined by the 2008 WHO classification system. Blood 2009;113:1906
- Wolach and Stone. How I treat mixed-phenotype acute leukemia. Blood 2015;125:2477
- Surapally et al. Emerging therapies for inv(16) AML. Blood 2021;137:2579
AML treatment
- El Chaer et al. How I treat AML incorporating the updated classifications and guidelines. Blood 2023;141:2823
- Wei et al. How I treat patients with AML using azacitidine and venetoclax. Blood 2025;145:1237
- Issa et al. How I treat acute myeloid leukemia with differentiation therapy. Blood 2025;145:1251
- Roboz and Canaani. How I use maintenance therapy in acute myeloid leukemia. Blood 2025;145:1273
- DiNardo and Wei. How I treat acute myeloid leukemia in the era of new drugs. Blood 2020;135:85
- Green and Wang. How I treat secondary acute myeloid leukemia. Blood 2025;145:1260
- Burd et al. Precision medicine treatment in acute myeloid leukemia using prospective genomic profiling: feasibility and preliminary efficacy of the Beat AML Master Trial. Nat Med 2020;26:1852(No apparent harm from delaying therapy 1 week to obtain mutational profile prior to starting treatment in AML patients aged 60-92)
- Ofran et al. How I treat acute myeloid leukemia presenting with preexisting comorbidities. Blood 2016;128:488
- Krug et al. Increasing intensity of therapies assigned at diagnosis does not improve survival of adults with acute myeloid leukemia. Leukemia 2016;30:1230
- Rashidi et al. Maintenance therapy in acute myeloid leukemia: an evidence-based review of randomized trials. Blood 2016;128:763
- Huls et al. Menin inhibitors in the treatment of acute myeloid leukemia. Blood 2025;145:581
- Stein and Tallman. Emerging therapeutic drugs for AML. Blood 2016;127:71
- Pratz and Levis. How I treat FLT3-mutated AML. Blood 2017;129:565
- Erba et al. Quizartinib plus chemotherapy in newly diagnosed patients with FLT3-internal-tandem-duplication-positive acute myeloid leukaemia (QuANTUM-First): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2023;401:1571 (Median OS 32 mo with quizartinib, 15 mo without)
- Falini et al. How I diagnose and treat NPM1-mutated AML. Blood 2021;137:589
- Othman et al. Postinduction molecular MRD identifies patients with NPM1 AML who benefit from allogeneic transplant in first remission. Blood 2024;143:1931 (“Postinduction molecular MRD reliably identifies those patients who benefit from allogeneic transplant in first remission”)
- Forman and Rowe. The myth of the second remission of acute leukemia in the adult. Blood 2013;121:1077
- Venditti et al GIMEMA AML1310 trial of risk-adapted, MRD-directed therapy for young adults with newly diagnosed acute myeloid leukemia. Blood 2019;134:935
- Luskin et al. Benefit of high-dose daunorubicin in AML induction extends across cytogenetic and molecular groups. Blood 2016;127:1551
- Lee et al. Prospective Randomized Comparison of Idarubicin and High-Dose Daunorubicin in Induction Chemotherapy for Newly Diagnosed Acute Myeloid Leukemia. J Clin Oncol 2017;35:2754(Possible advantage of HD dauno in patients with FLT3 ITD)
- Bradstock et al. Idarubicin Dose Escalation During Consolidation Therapy for Adult Acute Myeloid Leukemia. J Clin Oncol 2017;35:1678(Increasing anthracycline dose during consolidation improved leukemia-free survival)
- Löwenberg et al. Cytarabine dose for acute myeloid leukemia. NEJM 2011;364:1027(2000 mg/m2 no more effective, and more toxic, than 1000 mg/m2)
- Löwenberg B. Sense and nonsense of high-dose cytarabine for acute myeloid leukemia. Blood 2013;121:26
- Willemze et al. High-Dose Cytarabine in Induction Treatment Improves the Outcome of Adult Patients Younger Than Age 46 Years With Acute Myeloid Leukemia: Results of the EORTC-GIMEMA AML-12 Trial. J Clin Oncol 2014;32:218(Modest improvement in survival for pts given HDAC vs standard dose cytarabine; benefit more pronounced in younger and high-risk pts)
- Ohtake et al. Randomized study of induction therapy comparing standard-dose idarubicin with high-dose daunorubicin in adult patients with previously untreated acute myeloid leukemia: the JALSG AML201 Study. Blood 2011;117:2358(50 mg/m2 daunorubicin x 5 days = 12 mg/m2 idarubicin x 3 days)
- Holowiecki et al. Cladribine, But Not Fludarabine, Added to Daunorubicin and Cytarabine During Induction Prolongs Survival of Patients With Acute Myeloid Leukemia: A Multicenter, Randomized Phase III Study. J Clin Oncol 2012;30:2441
- Löwenberg et al. Therapeutic value of clofarabine in younger and middle-aged (18-65 years) adults with newly diagnosed AML. Blood 2017;129:1636(Reduces relapse but does not improve survival)
- Miyawaki et al. A randomized comparison of 4 courses of standard-dose multiagent chemotherapy versus 3 courses of high-dose cytarabine alone in postremission therapy for acute myeloid leukemia in adults: the JALSG AML201 Study. Blood 2011;117:2366(2 regimens equally effective overall, HiDAC better with favorable cytogenetics)
- Hokland and Ommen. Towards individualized follow-up in adult acute myeloid leukemia in remission. Blood 2011;117:2577 (Role of MRD monitoring during remission)
- Bradstock et al. A randomized trial of high-versus conventional-dose cytarabine in consolidation chemotherapy for adult de novo acute myeloid leukemia in first remission after induction therapy containing high-dose cytarabine. Blood 2005;105:481
- Neubauer et al. Patients With Acute Myeloid Leukemia and RAS Mutations Benefit Most From Postremission High-Dose Cytarabine: A Cancer and Leukemia Group B Study. J Clin Oncol 2008;26:4603
- Lancet et al. CPX-351 versus 7+3 cytarabine and daunorubicin chemotherapy in older adults with newly diagnosed high-risk or secondary acute myeloid leukaemia: 5-year results of a randomised, open-label, multicentre, phase 3 trial. Lancet Haematol 2021;8:e481(Liposomal dauno+cytarabine superior to 7+3 in patients 60-75)
- Castaigne et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet 2012;379:1508(3 yr EFS 40.8% with low-dose fractionated Mylotarg, 17.1% without)
- Hills et al. Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials. Lancet Oncol 2014;15:986(Significant benefit in patients without advers cytogenetics)
- Burnett et al. Identification of Patients With Acute Myeloblastic Leukemia Who Benefit From the Addition of Gemtuzumab Ozogamicin: Results of the MRC AML15 Trial. J Clin Oncol 2011;29:369(Benefits patients with favorable cytogenetics)
- Petersdorf et al. A phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia. Blood 2013;121:4854(Adding gemtuzumab did not improve CR rate or OS)
- Fournier et al. Mutational profile and benefit of gemtuzumab ozogamicin in acute myeloid leukemia. Blood 2020;135:542(GO benefits patients with low- and intermediate-risk genotypes, and those with activating signaling mutations)
- Freeman et al. Fractionated vs single-dose gemtuzumab ozogamicin with determinants of benefit in older patients with AML: the UK NCRI AML18 trial. Blood 2023;142:1697
- Loo et al. Sorafenib plus intensive chemotherapy in newly diagnosed FLT3-ITD AML: a randomized, placebo-controlled study by the ALLG. Blood 2023;142:1960 (Pretransplant sorafenib did not improve DFS)
- Stone et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. NEJM 2017;377:454(Significant improval in OS and PFS with addition of midostaurin)
- Schlenk et al. Midostaurin added to chemotherapy and continued single-agent maintenance therapy in acute myeloid leukemia with FLT3-ITD. Blood 2019;133:840(Significant improvement in EFS versus historical controls)
- Döhner et al. Impact of NPM1/FLT3-ITD genotypes defined by the 2017 European LeukemiaNet in patients with acute myeloid leukemia. Blood 2020;135:371(Midostaurin benefits FLT3-ITD positive patients regardless of prognostic category)
- Welch et al. TP53 and Decitabine in Acute Myeloid Leukemia and Myelodysplastic Syndromes. NEJM 2016;375:2023(Presence of TP53 mutation predicted favorable response to treatment; with editorial)
- Sekeres et al. A phase 2 study of lenalidomide monotherapy in patients with deletion 5q acute myeloid leukemia: Southwest Oncology Group Study S0605. Blood 2011;118:523(14% overall response rate, medial overall survival 2 months in this group of older pts)
- Stein et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017;130:722(40% OR rate)
- Stein et al. Molecular remission and response patterns in patients with mutant-IDH2 acute myeloid leukemia treated with enasidenib. Blood 2019;133:676
- Amatangelo et al. Enasidenib induces acute myeloid leukemia cell differentiation to promote clinical response. Blood 2017;130:732
- Roboz et al. Ivosidenib induces deep durable remissions in patients with newly diagnosed IDH1-mutant acute myeloid leukemia. Blood 2020;135:463
- Stein et al. Ivosidenib or enasidenib combined with intensive chemotherapy in patients with newly diagnosed AML: a phase 1 study. Blood 2021;137:1792
- Montesinos et al. Ivosidenib and Azacitidine in IDH1-Mutated Acute Myeloid Leukemia. NEJM 2022;389:1519
- Illendula et al. A small-molecule inhibitor of the aberrant transcription factor CBFβ-SMMHC delays leukemia in mice. Science 2015;347:779(Promising experimental agent – with editorial)
- Stein et al. A phase 1 trial of vadastuximab talirine as monotherapy in patients with CD33-positive acute myeloid leukemia. Blood 2018;131:387
- Konopleva and Letai. BCL-2 inhibition in AML: an unexpected bonus? Blood 2018;132:1007(Venetoclax)
- Wei et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial. Blood 2020;135:2137 (Adding venetoclax to LDAC improved remission rate from 13% to 48%)
- Wang et al. Venetoclax plus 3 + 7 daunorubicin and cytarabine chemotherapy as first-line treatment for adults with acute myeloid leukaemia: a multicentre, single-arm, phase 2 trial. Lancet Haematol 2022;9:e415 (91% CR rate)
- Mantzaris et al. Venetoclax plus daunorubicin and cytarabine for newly diagnosed acute myeloid leukemia: results of a phase 1b study. Blood 2025;145:1870 (High rate of MRD-negative CR)
- Senapati et al. Venetoclax abrogates the prognostic impact of splicing factor gene mutations in newly diagnosed acute myeloid leukemia. Blood 2023;142:1647
- Xie et al. Venetoclax with decitabine as frontline treatment in younger adults with newly diagnosed ELN adverse-risk AML. Blood 2023;142:1323 (61% EFS @ 12 mo; promising as bridge to transplant)
- Thomas et al. Randomized Phase II Study of Clofarabine-Based Consolidation for Younger Adults With Acute Myeloid Leukemia in First Remission. J Clin Oncol 2017;35:1223(2 yr RFS 59% vs 47% with HDAC, but more toxic)
- Wei et al. Oral Azacitidine Maintenance Therapy for Acute Myeloid Leukemia in First Remission. NEJM 2020;383:2526(Median OS 25 mo vs 15 mo with placebo in pts 55+ who were not transplant candidates)
- Roboz et al. Oral azacitidine prolongs survival of patients with AML in remission independently of measurable residual disease status. Blood 2022;139:2145
- Döhner et al. Prognostic impact of NPM1 and FLT3 mutations in patients with AML in first remission treated with oral azacitidine. Blood 2022;140:1674 (Oral aza benefited patients in remission after intensive chemo regardless of mutation status)
- Wang et al. Phase 3 trial of gilteritinib plus azacitidine vs azacitidine for newly diagnosed FLT3mut+ AML ineligible for intensive chemotherapy. Blood 2022;140:1845 (Modest improvement in OS in gilteritinib group)
- Bakst et al. How I treat extramedullary acute myeloid leukemia. Blood 2011;118:3785
Treatment of relapsed or refractory AML
- Thol et al. How I treat refractory and relapsed acute myeloid leukemia. Blood 2024;143:11
- Gooptu et al. How I treat AML relapse after allogeneic HSCT. Blood 2025;145:2128
- Hospital et al. Core-binding factor acute myeloid leukemia in first relapse: a retrospective study from the French AML Intergroup. Blood 2014;124:1342(Gemtuzumab ozogamicin given before transplant improves outcome)
- Garzon et al. A phase 1 clinical trial of single-agent selinexor in acute myeloid leukemia. Blood 2017;129:3165(“Disease control rate” – improvement or disease stabilization – in 69%)
- DiNardo et al. Durable Remissions with Ivosidenib in IDH1-Mutated Relapsed or Refractory AML. NEJM 2018;378:2386(CR rate 22%, OR rate 42%; about a third of patients alive at 1 yr)
- Perl et al. Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated AML. Blood 2019;381:1728(Gilteritinib prolonged median OS from 5.6 mo to 9.3 mo with less toxicity)
- Daver et al. Venetoclax Plus Gilteritinib for FLT3-Mutated Relapsed/Refractory Acute Myeloid Leukemia. J Clin Oncol 2022;40:4048
- Perl et al. Follow-up of patients with R/R FLT3-mutation–positive AML treated with gilteritinib in the phase 3 ADMIRAL trial. Blood 2022;139:3366
- Uy et al. Flotetuzumab as salvage immunotherapy for refractory acute myeloid leukemia. Blood 2021;137:751(Bispecific Ab to CD3 and CD123, 30% OR rate, 12 mo OS 50%)
- Daver et al. Venetoclax and idasanutlin in relapsed/refractory AML: a nonrandomized, open-label phase 1b trial. Blood 2023;141:1265
APML
- Lallamand-Breietenbach and de Thé. Retinoic acid plus arsenic trioxide, the ultimate panacea for acute promyelocytic leukemia? Blood 2013;122:2008
- Sanz et al. Management of acute promyelocytic leukemia: updated recommendations from an expert panel of the European LeukemiaNet. Blood 2019;133:1630
- Sanz and Lo-Coco. Modern approaches to treating acute promyelocytic leukemia. J Clin Oncol 2011;29:495
- Lo-Coco and Ammatuna. The Biology of Acute Promyelocytic Leukemia and Its Impact on Diagnosis and Treatment. Hematology 2006;156-61
- Wei et al. Active Pin1 is a key target of all-trans retinoic acid in acute promyelocytic leukemia and breast cancer. Nat Med 2015;21:457
- Tallman et al. Does microgranular variant morphology of acute promyelocytic leukemia independently predict a less favorable outcome compared with classical M3 APL? A joint study of the North American Intergroup and the PETHEMA Group. Blood 2010;116:5650(No)
- Montesinos et al. Clinical significance of CD56 expression in patients with acute promyelocytic leukemia treated with all-trans retinoic acid and anthracycline-based regimens. Blood 2011;117:1799(Adverse prognostic feature)
- Licht J. Acute Promyelocytic Leukemia — Weapons of Mass Differentiation. NEJM 2009;360:928
- Ablain and de The. Revisiting the differentiation paradigm in acute promyelocytic leukemia. Blood 2011;117:5795
- Ablain et al. Activation of a promyelocytic leukemia–tumor protein 53 axis underlies acute promyelocytic leukemia cure. Nat Med 2014;20:167(With editorial; suggests inducing differentiation is not the sole basis of APL cure with arsenic or ATRA)
- Sanz et al. Management of acute promyelocytic leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet. Blood 2009;113:1875
- Adès et al. Is Cytarabine Useful in the Treatment of Acute Promyelocytic Leukemia? Results of a Randomized Trial From the European Acute Promyelocytic Leukemia Group. J Clin Oncol 2006;24:5703
- Adès et al. Treatment of newly diagnosed acute promyelocytic leukemia (APL): a comparison of French-Belgian-Swiss and PETHEMA results. Blood 2008;111:1078(Adding cytarabine to ATRA + ida may be beneficial in APL patients with WBC > 10K)
- Lo-Coco et al. Front-line treatment of acute promyelocytic leukemia with AIDA induction followed by risk-adapted consolidation for adults younger than 61 years: results of the AIDA-2000 trial of the GIMEMA Group. Blood 2010;116:3171
- Chen et al. From an old remedy to a magic bullet: molecular mechanisms underlying the therapeutic effects of arsenic in fighting leukemia. Blood 2011;117:6425
- Tallman et al. All-trans retinoic acid in acute promyelocytic leukemia: long-term outcome and prognostic factor analysis from the North American Intergroup protocol. Blood 2002;100:4298
- Tallman et al. Clinical description of 44 patients with acute promyelocytic leukemia who developed the retinoic acid syndrome. Blood 2000;95:90
- Montesinos et al. Differentiation syndrome in patients with acute promyelocytic leukemia treated with all-trans retinoic acid and anthracycline chemotherapy: characteristics, outcome, and prognostic factors. Blood 2009;113:775
- Sanz and Montesinos. How we prevent and treat differentiation syndrome in patients with acute promyelocytic leukemia. Blood 2014;123:2777
- Soignet et al. Complete remission after treatment of acute promyelocytic leukemia with arsenic trioxide. NEJM 1998;339:1341
- Sanz et al. Risk-adapted treatment of acute promyelocytic leukemia with all-trans-retinoic acid and anthracycline monochemotherapy: a multicenter study by the PETHEMA group. Blood 2004;103:1237
- Sanz et al. Risk-adapted treatment of acute promyelocytic leukemia with all-trans retinoic acid and anthracycline monochemotherapy: long-term outcome of the LPA 99 multicenter study by the PETHEMA Group. Blood 2008;112:3130(5 year DFS 84%)
- Sanz et al. Risk-adapted treatment of acute promyelocytic leukemia based on all-trans retinoic acid and anthracycline with addition of cytarabine in consolidation therapy for high-risk patients: further improvements in treatment outcome. Blood 2010;115:5137
- Avvisati et al. AIDA 0493 protocol for newly diagnosed acute promyelocytic leukemia: very long-term results and role of maintenance. Blood 2011;117:4716(12 year EFS 69%; no apparent benefit from maintenance therapy after molecular remission confirmed)
- Estey et al. Use of all-trans retinoic acid plus arsenic trioxide as an alternative to chemotherapy in untreated acute promyelocytic leukemia. Blood 2006;107:3469
- Burnett et al. Arsenic trioxide and all- trans retinoic acid treatment for acute promyelocytic leukaemia in all risk groups (AML17): results of a randomised, controlled, phase 3 trial. Lancet Oncol 2015;16:1295
- Russell et al. Attenuated arsenic trioxide plus ATRA therapy for newly diagnosed and relapsed APL: long-term follow-up of the AML17 trial. Blood 2018;132:1452
- Lo-Coco et al. Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. NEJM 2013;369:111(100% CR, 97% 2-yr EFS; less hematologic toxicity than ATRA+chemo, more liver problems)
- Platzbecker et al. Improved Outcomes With Retinoic Acid and Arsenic Trioxide Compared With Retinoic Acid and Chemotherapy in Non–High-Risk Acute Promyelocytic Leukemia: Final Results of the Randomized Italian-German APL0406 Trial. J Clin Oncol 2017;35:605
- Ravandi et al. Effective Treatment of Acute Promyelocytic Leukemia With All-Trans-Retinoic Acid, Arsenic Trioxide, and Gemtuzumab Ozogamicin. J Clin Oncol 2009;27:504(ATRA + ATO with or without GO can replace chemotherapy-containing regimens)
- Iland et al. All-trans-retinoic acid, idarubicin, and IV arsenic trioxide as initial therapy in acute promyelocytic leukemia (APML4). Blood 2012;120:1570(95% CR, 2 year OS 93%)
- Abaza et al. Long-term outcome of acute promyelocytic leukemia treated with all-trans-retinoic acid, arsenic trioxide, and gemtuzumab. Blood 2017;129:1283(5-year OS 88%; relapse is rare)
- Zhu et al. Oral Tetra-Arsenic Tetra-Sulfide Formula Versus Intravenous Arsenic Trioxide As First-Line Treatment of Acute Promyelocytic Leukemia: A Multicenter Randomized Controlled Trial. J Clin Oncol 2013;31:4216(Oral arsenic compound as effective and safe as IV ATO)
- Asou et al. A randomized study with or without intensified maintenance chemotherapy in patients with acute promyelocytic leukemia who have become negative for PML-RAR transcript after consolidation therapy: The Japan Adult Leukemia Study Group (JALSG) APL97 study(PCR negative patients had better outcome without maintenance)
- Adès et al. Very long-term outcome of acute promyelocytic leukemia after treatment with all-trans retinoic acid and chemotherapy: the European APL Group experience. Blood 2010; 115:1690(10 year survival 77%)
- Grimwade et al. Prospective Minimal Residual Disease Monitoring to Predict Relapse of Acute Promyelocytic Leukemia and to Direct Pre-Emptive Arsenic Trioxide Therapy. J Clin Oncol 2009;27:3650 (MRD monitoring was most powerful predictor of relapse. Early treatment with arsenic trioxide prevented frank relapse in most MRD positive pts)
- Chendamarai et al. Role of minimal residual disease monitoring in acute promyelocytic leukemia treated with arsenic trioxide in frontline therapy. Blood 2012;119:3413
- Au et al. Oral arsenic trioxide–based maintenance regimens for first complete remission of acute promyelocytic leukemia: a 10-year follow-up study. Blood 2011;118:6535
- Mathews et al. Single-Agent Arsenic Trioxide in the Treatment of Newly Diagnosed Acute Promyelocytic Leukemia: Long-Term Follow-Up Data. J Clin Oncol 2010;28:3866(80% 5 year DFS)
- Powell et al. Arsenic trioxide improves event-free and overall survival for adults with acute promyelocytic leukemia: North American Leukemia Intergroup Study C9710. Blood 2010;116:3751
- Zhu et al. The simpler, the better: oral arsenic for acute promyelocytic leukemia. Blood 2019;134:597
- Echaniz-Laguna et al. Mitochondrial myopathy caused by arsenic trioxide therapy. Blood 2012;119:4272
- Yanada et al. Phase 2 study of arsenic trioxide followed by autologous hematopoietic cell transplantation for relapsed acute promyelocytic leukemia. Blood 2013;121:3095
- Zhu et al. Resistance to arsenic in acute promyelocytic leukemia (letter). NEJM 2014;370:1864(PML gene mutations in arsenic-binding domain cause resistance)
- Mantha et al. Determinants of fatal bleeding during induction therapy for acute promyelocytic leukemia in the ATRA era. Blood 2017;129:1763(High white count predicts early hemorrhagic death)
- Cao et al. Promyelocytic extracellular chromatin exacerbates coagulation and fibrinolysis in acute promyelocytic leukemia. Blood 2017;129:1855
- de la Serna et al. Causes and prognostic factors of remission induction failure in patients with acute promyelocytic leukemia treated with all-trans retinoic acid and idarubicin. Blood 2008;111:3395.(Death during induction due to hemorrhage in 5%, infection in 2.3%, ATRA syndrome in 1.4%)
- Park et al. Early death rate in acute promyelocytic leukemia remains high despite all-trans retinoic acid. Blood 2011;118:1248(Overall early death rate 17.3%)
- Voso et al. Acute promyelocytic leukemia: long-term outcomes from the HARMONY project. Blood 2025;145:234 (ATRA/ATO improves outcomes but early death rate remains high)
ALL: biology/general
- Gökbuget et al. Diagnosis, prognostic factors, and assessment of ALL in adults: 2024 ELN recommendations from a European expert panel. Blood 2024;143:1891
- Gökbuget et al. Management of ALL in adults: 2024 ELN recommendations from a European expert panel. Blood 2024;143:1903
- Malard and Mohty. Acute lymphoblastic leukaemia. Lancet 2020;395:1146
- Hunger and Mullighan. Redefining ALL classification: toward detecting high-risk ALL and implementing precision medicine. Blood 2015;125:3977
- Simonin et al. NGS-based stratification refines the risk stratification in T-ALL and identifies a very-high-risk subgroup of patients. Blood 2024;144:1570
- Passet et al.Genetic subtypes of B-cell acute lymphoblastic leukemia in adults. Blood 2025;145:1451
- Pölönen et al. Classification and risk stratification in T-lineage acute lymphoblastic leukemia. Blood 2025;145:1464
- Forman and Rowe. The myth of the second remission of acute leukemia in the adult. Blood 2013;121:1077
- Gocho and Yang. Genetic defects in hematopoietic transcription factors and predisposition to acute lymphoblastic leukemia. Blood 2019;134:793
- Di Paola and Porter. ETV6-related thrombocytopenia and leukemia predisposition. Blood 2019;134:663(Predisposition to B-ALL)
- Pullarkat et al. Impact of cytogenetics on the outcome of adult acute lymphoblastic leukemia: results of Southwest Oncology Group 9400 study. Blood 2008;111:2563. (t(9;22), -7, +8, and 11q23 rearrangement had worse prognosis; cytogenetics most important prognostic factor in adult ALL)
- Moorman et al. A population-based cytogenetic study of adults with acute lymphoblastic leukemia. Blood 2010;115:206
- Roberts et al. Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia. NEJM 2014;371:1005
- Asnafi et al. NOTCH1/FBXW7 mutation identifies a large subgroup with favorable outcome in adult T-cell acute lymphoblastic leukemia (T-ALL): a Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL) study. Blood 2009;113:3918
- Thomas et al. Prognostic significance of CD20 expression in adults with de novo precursor B-lineage acute lymphoblastic leukemia. Blood 2009;113:6330(CD20 expression an independent predictor of poor outcome)
- Brüggemann et al. Has MRD monitoring superseded other prognostic factors in adult ALL? Blood 2012;120:4470
- Beldjord et al. Oncogenetics and minimal residual disease are independent outcome predictors in adult patients with acute lymphoblastic leukemia. Blood 2014;123: 3739
- Gökbuget et al. Adult patients with acute lymphoblastic leukemia and molecular failure display a poor prognosis and are candidates for stem cell transplantation and targeted therapies. Blood 2012;120:1868
- Girardi et al. The genetics and molecular biology of T-ALL. Blood 2017;129:1113
- Jain et al. Early T-cell precursor acute lymphoblastic leukemia/lymphoma (ETP-ALL/LBL) in adolescents and adults: a high-risk subtype. Blood 2016;127:1863
- Gökbuget et al. Outcome of relapsed adult lymphoblastic leukemia depends on response to salvage chemotherapy, prognostic factors, and performance of stem cell transplantation. Blood 2012;120:2032
- Jain et al. Ph-like acute lymphoblastic leukemia: a high-risk subtype in adults. Blood 2017;129:572
- Tasian et al. Philadelphia chromosome–like acute lymphoblastic leukemia. Blood 2017;130:2064
- Safavi and Paulsson. Near-haploid and low-hypodiploid acute lymphoblastic leukemia: two distinct subtypes with consistently poor prognosis. Blood 2017;129:420(“Near-haploid” = about 27 chromosomes, “low-hypodiploid” = about 36 chromosomes)
- Good et al. Single-cell developmental classification of B cell precursor acute lymphoblastic leukemia at diagnosis reveals predictors of relapse. Nat Med 2018;24:474(With editorial)
- Kourtis et al. Oncogenic hijacking of the stress response machinery in T cell acute lymphoblastic leukemia. Nat Med 2018;24:1157(Upregulation of heat shock proteins promotes leukemic cell survival)
- Benshang et al. Therapy-induced mutations drive the genomic landscape of relapsed acute lymphoblastic leukemia. Blood 2020;135:41(Early relapse driven by retained subclones, late relapse by chemo-induced mutations in pediatric ALL)
ALL: Treatment
- Basssan et al. New Approaches to the Management of Adult Acute Lymphoblastic Leukemia. J Clin Oncol 2018;33:3504
- Lim et al. How I treat newly diagnosed and refractory T-cell acute lymphoblastic lymphoma in children and young adults. Blood 2023;141:3019
- Marks and Rowntree. Management of adults with T-cell lymphoblastic leukemia. Blood 2017;129:1134
- Kopmar and Cassaday. How I prevent and treat central nervous system disease in adults with acute lymphoblastic leukemia. Blood 2023;141:1379
- Biossel and Baruchel. Acute lymphoblastic leukemia in adolescent and young adults: treat as adults or as children? Blood 2018;132:351 (Pediatric regimens improve outcomes)
- O’Dwyer et aL. Treatment strategies for adolescent and young adult patients with acute myeloid leukemia. Blood 2018;132:362
- Stock et al. A pediatric regimen for older adolescents and young adults with acute lymphoblastic leukemia: results of CALGB 10403. Blood 2019;133:1548
- Wolach and Stone. How I treat mixed-phenotype acute leukemia. Blood 2015;125:2477
- Gökbuget and Steffen. How I treat older patients with Ph/BCR-ABL–negative acute lymphoblastic leukemia. Blood 2025;145:53
- Curran and Stock. How I treat acute lymphoblastic leukemia in older adolescents and young adults. Blood 2015;125:3702
- Ribera et al. Chemotherapy or allogeneic transplantation in high-risk Philadelphia chromosome–negative adult lymphoblastic leukemia. Blood 2021;137:1879(MRD status after induction & early consolidation predicts need for allotransplant)
- Harrison CJ. Targeting signaling pathways in acute lymphoblastic leukemia: new insights. Hematology 2013:118
- Advani A.New immune strategies for the treatment of acute lymphoblastic leukemia: antibodies and chimeric antigen receptors. Hematology 2013:131
- Pikman and Stegmaier. Targeted therapy for fusion-driven high-risk acute leukemia. Blood 2018;132:1241
- Hunault et al. Better outcome of adult acute lymphoblastic leukemia after early genoidentical allogeneic bone marrow transplantation (BMT) than after late high-dose therapy and autologous BMT: a GOELAMS trial. Blood 2004;104:3028
- Rowe et al. Induction therapy for adults with acute lymphoblastic leukemia: results of more than 1500 patients from the international ALL trial: MRC UKALL XII/ECOG E2993. Blood 2005;106:3760(Overall survival 45% for patients who achieved CR with induction chemotherapy)
- Larson et al. A five-drug remission induction regimen with intensive consolidation for adults with acute lymphoblastic leukemia: cancer and leukemia group B study 8811. Blood 1995;85:2025
- Thomas et al. Chemoimmunotherapy With a Modified Hyper-CVAD and Rituximab Regimen Improves Outcome in De Novo Philadelphia Chromosome–Negative Precursor B-Lineage Acute Lymphoblastic Leukemia. J Clin Oncol 2010;28:3880(95% CR, 3 year OS 50%)
- Thomas et al. Outcome of Treatment in Adults With Acute Lymphoblastic Leukemia: Analysis of the LALA-94 Trial. J Clin Oncol 2004;22:4075(Allo-BMT superior in high-risk patients; auto-BMT no better than standard chemotherapy)
- O’Brien et al. High-Dose Vincristine Sulfate Liposome Injection for Advanced, Relapsed, and Refractory Adult Philadelphia Chromosome–Negative Acute Lymphoblastic Leukemia. J Clin Oncol 2013;31:676(20% CR, potential bridge to transplant, 5/65 pts long-term survivors)
- Fielding et al. Outcome of 609 adults after relapse of acute lymphoblastic leukemia (ALL); an MRC UKALL12/ECOG 2993 study. Blood 2007;109:944
- Möricke et al. Risk-adjusted therapy of acute lymphoblastic leukemia can decrease treatment burden and improve survival: treatment results of 2169 unselected pediatric and adolescent patients enrolled in the trial ALL-BFM 95. Blood 2008;111:4477
- Frey and Luger. How I treat adults with relapsed or refractory Philadelphia chromosome–negative acute lymphoblastic leukemia. Blood 2014;126:589
- Gökbuget et al. High single-drug activity of nelarabine in relapsed T-lymphoblastic leukemia/lymphoma offers curative option with subsequent stem cell transplantation. Blood 2011;118:3504
- Maury et al. Rutuximab in B-lineage adult acute lymphoblastic leukemia. NEJM 2016;375:1044(Rituximab significantly improved event-free survival)
- Chevallier et al. Trastuzumab for treatment of refractory/relapsed HER2-positive adult B-ALL: results of a phase 2 GRAALL study. Blood 2012;119:2474
- Kantarjian et al. Inotuzumab Ozogamicin versus Standard Therapy for Acute Lymphoblastic Leukemia. NEJM 2016;375:740(Higher CR rate, modest improvement in OS with inotuzumab in RR ALL; drug caused VOD in 11%)
- Jabbour et al. Phase 2 study of inotuzumab ozogamicin for measurable residual disease in acute lymphoblastic leukemia in remission. Blood 2024;143:417
- Topp et al. Long-term follow-up of hematologic relapse-free survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL. Blood 2012;120:5185
- Advani et al. SWOG 1318: A Phase II Trial of Blinatumomab Followed by POMP Maintenance in Older Patients With Newly Diagnosed Philadelphia Chromosome–Negative B-Cell Acute Lymphoblastic Leukemia. J Clin Oncol 2022;40:1574
- Topp et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol 2015;16:57(43% CR; 22% grade 3 or 4 neurotoxicity)
- Zugmaier et al. Long-term survival and T-cell kinetics in relapsed/refractory ALL patients who achieved MRD response after blinatumomab treatment. Blood 2015;126:2578
- Topp et al. Phase II Trial of the Anti-CD19 Bispecific T Cell–Engager Blinatumomab Shows Hematologic and Molecular Remissions in Patients With Relapsed or Refractory B-Precursor Acute Lymphoblastic Leukemia. J Clin Oncol 2014;32:4134
- Kantarjian et al. Blinatumomab versus Chemotherapy for Advanced Acute Lymphoblastic Leukemia. NEJM 2017;376:836(Median OS 7.7 mo with blinatumomab vs 4 mo with chemo)
- Martinelli et al. Complete Hematologic and Molecular Response in Adult Patients With Relapsed/Refractory Philadelphia Chromosome–Positive B-Precursor Acute Lymphoblastic Leukemia Following Treatment With Blinatumomab: Results From a Phase II, Single-Arm, Multicenter Study. J Clin Oncol 2017;35;1795
- Topp et al. Health-related quality of life in adults with relapsed/refractory acute lymphoblastic leukemia treated with blinatumomab. Blood 2018;131:2906(“Blinatumomab delays deterioration in adults with R/R ALL”)
- Gökbuget et al. Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia. Blood 2018;131:1522
- Litzow et al. Blinatumomab for MRD-Negative Acute Lymphoblastic Leukemia in Adults. NEJM 2024;391:320 (Risk of relapse or death reduced by 50% with adding blina to chemo)
- Maude et al. Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia. NEJM 2014;371:1507(27/30 patients had CR; 6-mo EFS 67%)
CAR-T therapy in ALL
- Badar et al. Incorporation of immunotherapy into frontline treatment for adults with B-cell precursor acute lymphoblastic leukemia. Blood 2025;145:1475
- Maude et al. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood 2015;125:4017
- Orlando et al. Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia. Nat Med 2018;24:1504
- Maude et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. NEJM 2018;378:439
- Gardner et al. Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood 2017;129:3322(Over 90% CR rate)
- Park et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. NEJM 2018;378:449
- Gardner et al. Acquisition of a CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy. Blood 2016;127:2406
- Fry et al. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat Med 2018;24:20
- Ghorashian et al. Enhanced CAR T cell expansion and prolonged persistence in pediatric patients with ALL treated with a low-affinity CD19 CAR. Nat Med 2019;25:1408
- Xu et al. CRISPR-Edited Stem Cells in a Patient with HIV and Acute Lymphocytic Leukemia. NEJM 2019;381:1240(Using CCR5-negative cells; with editorial)
- Chiesa et al. Base-Edited CAR7 T Cells for Relapsed T-Cell Acute Lymphoblastic Leukemia. NEJM 2023;389:899
- Jabbour et al. Monoclonal antibodies in acute lymphoblastic leukemia. Blood 2015;125:4010
- Shah et al. KTE-X19 anti-CD19 CAR T-cell therapy in adult relapsed/refractory acute lymphoblastic leukemia: ZUMA-3 phase 1 results. Blood 2021;138:11
- Qi et al. Efficacy and safety of CD19-specific CAR T cell–based therapy in B-cell acute lymphoblastic leukemia patients with CNSL. Blood 2022;139:3376
- Frey NV. Approval of brexucabtagene autoleucel for adults with relapsed and refractory acute lymphocytic leukemia. Blood 2022;140:11
- Roddie et al. Obecabtagene Autoleucel in Adults with B-Cell Acute Lymphoblastic Leukemia. NEJM 2024;391:2219
- Srinagesh et al. A phase 1 clinical trial of NKTR-255 with CD19-22 CAR T-cell therapy for refractory B-cell acute lymphoblastic leukemia. Blood 2024;144:1689
- Tran and Tasian. How I treat Philadelphia chromosome–like acute lymphoblastic leukemia in children, adolescents, and young adults. Blood 2025;145:20
BCR-ABL positive ALL
- Chiaretti and Foà. How I treat adult Ph+ ALL. Blood 2025;145:11
- Foà R. Ph-Positive Acute Lymphoblastic Leukemia — 25 Years of Progress. NEJM 2025; 391:1941
- Towatari et al. Combination of intensive chemotherapy and imatinib can rapidly induce high-quality complete remission for a majority of patients with newly diagnosed BCR-ABL-positive acute lymphoblastic leukemia. Blood 2004;104:3507
- Ottmann and Wassmann. Treatment of Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia. Hematology 2005:118-122
- Yanada et al. High Complete Remission Rate and Promising Outcome by Combination of Imatinib and Chemotherapy for Newly Diagnosed BCR-ABL-Positive Acute Lymphoblastic Leukemia: A Phase II Study by the Japan Adult Leukemia Study Group. J Clin Oncol 2006;24:460
- Short et al. Impact of complete molecular response on survival in patients with Philadelphia chromosome–positive acute lymphoblastic leukemia. Blood 2016;128:504(Patients who have CMR @ 3 mo have excellent long term outcomes without transplant)
- Wassmann et al. Alternating versus concurrent schedules of imatinib and chemotherapy as front-line therapy for Philadelphia-positive acute lymphoblastic leukemia (Ph+ALL). Blood 2006;108:1469
- de Labarthe et al. Imatinib combined with induction or consolidation chemotherapy in patients with de novo Philadelphia chromosome-positive acute lymphoblastic leukemia: results of the GRAAPH-2003 study. Blood 2007;109:1408
- Vignetti et al. Imatinib plus steroids induces complete remissions and prolonged survival in elderly Philadelphia chromosome–positive patients with acute lymphoblastic leukemia without additional chemotherapy: results of the Gruppo Italiano Malattie Ematologiche dell’Adulto (GIMEMA) LAL0201-B protocol. Blood 2007;109:3676
- Ottman et al. Dasatinib induces rapid hematologic and cytogenetic responses in adult patients with Philadelphia chromosome–positive acute lymphoblastic leukemia with resistance or intolerance to imatinib: interim results of a phase 2 study. Blood 2007;110:2309
- Ravandi et al. First report of phase 2 study of dasatinib with hyper-CVAD for the frontline treatment of patients with Philadelphia chromosome–positive (Ph+) acute lymphoblastic leukemia. Blood 2010;116:2070(2 yr OS 64%)
- Porkka et al. Dasatinib crosses the blood-brain barrier and is an efficient therapy for central nervous system Philadelphia chromosome–positive leukemia. Blood 2008;112:1005(imatinib does not cross BBB well, less effective)
- Foà et al. Dasatinib as first-line treatment for adult patients with Philadelphia chromosome–positive acute lymphoblastic leukemia. Blood 2011;118:6521(Dasatinib plus steroids causes complete hematologic remission in over 90% of patients)
- Rousselot et al. Dasatinib and low-intensity chemotherapy in elderly patients with Philadelphia chromosome–positive ALL. Blood 2016;128:774(36% 5 year OS)
- Fielding et al. UKALLXII/ECOG2993: addition of imatinib to a standard treatment regimen enhances long-term outcomes in Philadelphia positive acute lymphoblastic leukemia. Blood 2014;123:843(4 year OS improved from 22% to 38% with addition of imatinib)
- Chalandon et al. Randomized study of reduced-intensity chemotherapy combined with imatinib in adults with Ph-positive acute lymphoblastic leukemia. Blood 2015;125:3711(Less intense induction plus TKI as effective, less toxic than hyperCVAD plus TKI)
- Kim et al. Nilotinib combined with multiagent chemotherapy for newly diagnosed Philadelphia-positive acute lymphoblastic leukemia. Blood 2015;126:746(2 year OS 72%; MRD status early in remission predicted outcome)
- Chalandon et al. Nilotinib with or without cytarabine for Philadelphia-positive acute lymphoblastic leukemia. Blood 2024;143:2363
- Comoli et al. BCR-ABL–specific T-cell therapy in Ph+ ALL patients on tyrosine-kinase inhibitors. Blood 2017;129:582(Report of 3 patients, all obtained CR)
- Foà et al. Dasatinib–Blinatumomab for Ph-Positive Acute Lymphoblastic Leukemia in Adults. NEJM 2020;383:1613(88% DFS at 18 mo)
AML and ALL: complications
- Zuckerman et al. How I treat hematologic emergencies in adults with acute leukemia. Blood 2012;120:1993
- Wang et al. Management of hemostatic complications in acute leukemia: Guidance from the SSC of the ISTH. J Thromb Haemost 2020;18:3174
- Pui C. Central Nervous System Disease in Acute Lymphoblastic Leukemia: Prophylaxis and Treatment. Hematology 2006;142-6
- Rollig and Ehninger. How I treat hyperleukocytosis in acute myeloid leukemia. Blood 2015;125:3246
- Lichtman and Rowe. Hyperleukocytic leukemias: rheological, clinical, and therapeutic considerations. Blood 1982; 60:279
- Azoulay et al. How I manage acute respiratory failure in patients with hematological malignancies. Blood 2024:143:971
- Byrd et al. Extramedullary myeloid cell tumors in acute nonlymphocytic leukemia: a clinical review. J Clin Oncol 1995;13:1800
- Rank et al. Thromboembolism in acute lymphoblastic leukemia: results of NOPHO ALL2008 protocol treatment in patients aged 1 to 45 years. Blood 2018;131:2475(8% cumulative incidence; highest risk with asparaginase treatment)
- Zwicker et al.The prevention and management of asparaginase‐related venous thromboembolism in adults: Guidance from the SSC on Hemostasis and Malignancy of the ISTH. J Thromb Haemost 2020;18:278
- Aldoss and Douerl. How I treat the toxicities of pegasparaginase in adults with acute lymphoblastic leukemia. Blood 2020;135:987
- Ku et al. Venous thromboembolism in patients with acute leukemia: incidence, risk factors, and effect on survival. Blood 2009;113:3911
- Mitchell et al. Validation of a predictive model for identifying an increased risk for thromboembolism in children with acute lymphoblastic leukemia: results of a multicenter cohort study. Blood 2010;115: 4999(Asparaginase use, central catheters, steroid use, and thrombophilia risk factors for VTE)
- Orvain et al. Thromboembolism prophylaxis in adult patients with acute lymphoblastic leukemia treated in the GRAALL-2005 study. Blood 2020;136:328(Antithrombin/heparin prophylaxis partially effective; fibrinogen concentrates increase clotting risk)
- Libourel et al. Disseminated intravascular coagulation at diagnosis is a strong predictor for thrombosis in acute myeloid leukemia. Blood 2016;128:1854
- Kwaan H. Double hazard of thrombophilia and bleeding in leukemia. Hematology 2007:151
- Gernsheimer et al. Prophylactic tranexamic acid in patients with hematologic malignancy: a placebo-controlled, randomized clinical trial. Blood 2022;140:1254 (No apparent benefit)
- Hijiya et al. Cumulative Incidence of Secondary Neoplasms as a First Event After Childhood Acute Lymphoblastic Leukemia. JAMA 2007;297:1207(6% incidence of 2nd neoplasm after 30 yrs)
- Nielsen et al. Risk of thyroid cancer, brain cancer, and non-Hodgkin lymphoma after adult leukemia: a nationwide study. Blood 2011;118:4062