1
|
Wei Q, Hu S, Xu J, Loghavi S, Daver N, Toruner GA, Wang W, Medeiros LJ, Tang G. Detection of KMT2A Partial Tandem Duplication by Optical Genome Mapping in Myeloid Neoplasms: Associated Cytogenetics, Gene Mutations, Treatment Responses, and Patient Outcomes. Cancers (Basel) 2024; 16:4193. [PMID: 39766092 PMCID: PMC11674272 DOI: 10.3390/cancers16244193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
KMT2A partial tandem duplication (PTD) involves intragenic KMT2A duplications and has been associated with poorer prognosis. In this study, we evaluated KMT2A PTD in 1277 patients with hematological malignancies using optical genome mapping (OGM). KMT2A PTD was detected in 35 patients with acute myeloid leukemia (AML) (7%), 5 patients with myelodysplastic syndrome (MDS) (2.2%), and 5 patients with chronic myelomonocytic leukemia (CMML) (7.1%). The PTDs varied in size, region, and copy number. An Archer RNA fusion assay confirmed KMT2A PTD in all 25 patients tested: 15 spanning exons 2 to 8 and 10 spanning exons 2 to 10. Most patients exhibited a normal (n = 21) or non-complex (n = 20) karyotype. The most common chromosomal abnormalities included loss of 20q or 7q and trisomy 11/gain of 11q. All patients had gene mutations, with FLT3 ITD and DNMT3A prevalent in AML and DNMT3A and RUNX1 common in MDS and CMML. Among patients who received treatment and had at least one follow-up bone marrow evaluation, 82% of those with de novo AML achieved complete remission after initial induction chemotherapy, whereas 90% of patients with secondary or refractory/relapsed AML showed refractory or partial responses. All but one patient with MDS and CMML were refractory to therapy. We conclude that OGM is an effective tool for detecting KMT2A PTD. Neoplasms with KMT2A PTD frequently harbor gene mutations and display normal or non-complex karyotypes. Patients with KMT2A PTD are generally refractory to conventional therapy, except for de novo AML.
Collapse
Affiliation(s)
- Qing Wei
- Department of Hematopathology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA; (Q.W.); (S.H.); (J.X.); (S.L.); (G.A.T.); (W.W.); (L.J.M.)
| | - Shimin Hu
- Department of Hematopathology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA; (Q.W.); (S.H.); (J.X.); (S.L.); (G.A.T.); (W.W.); (L.J.M.)
| | - Jie Xu
- Department of Hematopathology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA; (Q.W.); (S.H.); (J.X.); (S.L.); (G.A.T.); (W.W.); (L.J.M.)
| | - Sanam Loghavi
- Department of Hematopathology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA; (Q.W.); (S.H.); (J.X.); (S.L.); (G.A.T.); (W.W.); (L.J.M.)
| | - Naval Daver
- Department of Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA;
| | - Gokce A. Toruner
- Department of Hematopathology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA; (Q.W.); (S.H.); (J.X.); (S.L.); (G.A.T.); (W.W.); (L.J.M.)
| | - Wei Wang
- Department of Hematopathology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA; (Q.W.); (S.H.); (J.X.); (S.L.); (G.A.T.); (W.W.); (L.J.M.)
| | - L. Jeffrey Medeiros
- Department of Hematopathology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA; (Q.W.); (S.H.); (J.X.); (S.L.); (G.A.T.); (W.W.); (L.J.M.)
| | - Guilin Tang
- Department of Hematopathology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA; (Q.W.); (S.H.); (J.X.); (S.L.); (G.A.T.); (W.W.); (L.J.M.)
| |
Collapse
|
2
|
Bhatnagar B, Kohlschmidt J, Orwick SJ, Buelow DR, Fobare S, Oakes CC, Kolitz JE, Uy G, Stock W, Powell BL, Nicolet D, Hertlein EK, Mrózek K, Blachly JS, Eisfeld AK, Baker SD, Byrd JC. Framework of clonal mutations concurrent with WT1 mutations in adults with acute myeloid leukemia: Alliance for Clinical Trials in Oncology study. Blood Adv 2023; 7:4671-4675. [PMID: 37603350 PMCID: PMC10448419 DOI: 10.1182/bloodadvances.2023010482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/19/2023] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Affiliation(s)
- Bhavana Bhatnagar
- Department of Hematology and Medical Oncology, West Virginia University Cancer Institute, Wheeling Hospital, Wheeling, WV
| | - Jessica Kohlschmidt
- Alliance Statistics and Data Management Center, The Ohio State University, Columbus, OH
| | - Shelley J. Orwick
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Daelynn R. Buelow
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Sydney Fobare
- Department of Internal Medicine, Medical Student Training Program, College of Medicine, The Ohio State University, Columbus, OH
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Christopher C. Oakes
- Clara D. Bloomfield Center, The Ohio State Comprehensive Cancer Center, Columbus, OH
| | | | - Geoff Uy
- Division of Oncology, Department of Medicine, Washington University, St. Louis, MO
| | - Wendy Stock
- Division of Hematology-Oncology, Department of Internal Medicine, University of Chicago, Chicago, IL
| | - Bayard L. Powell
- Division of Hematology-Oncology, Department of Internal Medicine, Wake Forest University Health System, Winston-Salem, NC
| | - Deedra Nicolet
- Alliance Statistics and Data Management Center, The Ohio State University, Columbus, OH
| | - Erin K. Hertlein
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Krzysztof Mrózek
- Clara D. Bloomfield Center, The Ohio State Comprehensive Cancer Center, Columbus, OH
| | - James S. Blachly
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH
| | - Ann-Kathrin Eisfeld
- Clara D. Bloomfield Center, The Ohio State Comprehensive Cancer Center, Columbus, OH
- Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH
| | - Sharyn D. Baker
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - John C. Byrd
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
3
|
Eisenmann ED, Stromatt JC, Fobare S, Huang KM, Buelow DR, Orwick S, Jeon JY, Weber RH, Larsen B, Mims AS, Hertlein E, Byrd JC, Baker SD. TP-0903 Is Active in Preclinical Models of Acute Myeloid Leukemia with TP53 Mutation/Deletion. Cancers (Basel) 2022; 15:29. [PMID: 36612026 PMCID: PMC9817780 DOI: 10.3390/cancers15010029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Acute myeloid leukemia (AML) with mutations in the tumor suppressor gene TP53 confers a dismal prognosis with 3-year overall survival of <5%. While inhibition of kinases involved in cell cycle regulation induces synthetic lethality in a variety of TP53 mutant cancers, this strategy has not been evaluated in mutant TP53 AML. Previously, we demonstrated that TP-0903 is a novel multikinase inhibitor with low nM activity against AURKA/B, Chk1/2, and other cell cycle regulators. Here, we evaluated the preclinical activity of TP-0903 in TP53 mutant AML cell lines, including a single-cell clone of MV4-11 containing a TP53 mutation (R248W), Kasumi-1 (R248Q), and HL-60 (TP 53 null). TP-0903 inhibited cell viability (IC50, 12−32 nM) and induced apoptosis at 50 nM. By immunoblot, 50 nM TP-0903 upregulated pChk1/2 and pH2AX, suggesting induction of DNA damage. The combination of TP-0903 and decitabine was additive in vitro, and in vivo significantly prolonged median survival compared to single-agent treatments in mice xenografted with HL-60 (vehicle, 46 days; decitabine, 55 days; TP-0903, 63 days; combination, 75 days) or MV4-11 (R248W) (51 days; 62 days; 81 days; 89 days) (p < 0.001). Together, these results provide scientific premise for the clinical evaluation of TP-0903 in combination with decitabine in TP53 mutant AML.
Collapse
Affiliation(s)
- Eric D. Eisenmann
- Division of Pharmaceutics and Pharmacology, The Ohio State University, Columbus, OH 43212, USA
| | - Jack C. Stromatt
- Division of Pharmaceutics and Pharmacology, The Ohio State University, Columbus, OH 43212, USA
| | - Sydney Fobare
- Division of Hematology, The Ohio State University, Columbus, OH 43212, USA
| | - Kevin M. Huang
- Division of Pharmaceutics and Pharmacology, The Ohio State University, Columbus, OH 43212, USA
| | - Daelynn R. Buelow
- Division of Pharmaceutics and Pharmacology, The Ohio State University, Columbus, OH 43212, USA
| | - Shelley Orwick
- Division of Pharmaceutics and Pharmacology, The Ohio State University, Columbus, OH 43212, USA
| | - Jae Yoon Jeon
- Division of Pharmaceutics and Pharmacology, The Ohio State University, Columbus, OH 43212, USA
| | - Robert H. Weber
- Division of Pharmaceutics and Pharmacology, The Ohio State University, Columbus, OH 43212, USA
| | - Bill Larsen
- Division of Pharmaceutics and Pharmacology, The Ohio State University, Columbus, OH 43212, USA
| | - Alice S. Mims
- Division of Hematology, The Ohio State University, Columbus, OH 43212, USA
| | - Erin Hertlein
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - John C. Byrd
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Sharyn D. Baker
- Division of Pharmaceutics and Pharmacology, The Ohio State University, Columbus, OH 43212, USA
| |
Collapse
|
4
|
Buelow DR, Bhatnagar B, Orwick SJ, Jeon JY, Eisenmann ED, Stromatt JC, Pabla NS, Blachly JS, Baker SD, Blaser BW. BMX kinase mediates gilteritinib resistance in FLT3-mutated AML through microenvironmental factors. Blood Adv 2022; 6:5049-5060. [PMID: 35797240 PMCID: PMC9631628 DOI: 10.1182/bloodadvances.2022007952] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/27/2022] [Indexed: 11/23/2022] Open
Abstract
Despite the clinical benefit associated with gilteritinib in relapsed/refractory acute myeloid leukemia (AML), most patients eventually develop resistance through unknown mechanisms. To delineate the mechanistic basis of resistance to gilteritinib, we performed targeted sequencing and scRNASeq on primary FLT3-ITD-mutated AML samples. Co-occurring mutations in RAS pathway genes were the most common genetic abnormalities, and unresponsiveness to gilteritinib was associated with increased expression of bone marrow-derived hematopoietic cytokines and chemokines. In particular, we found elevated expression of the TEK-family kinase, BMX, in gilteritinib-unresponsive patients pre- and post-treatment. BMX contributed to gilteritinib resistance in FLT3-mutant cell lines in a hypoxia-dependent manner by promoting pSTAT5 signaling, and these phenotypes could be reversed with pharmacological inhibition and genetic knockout. We also observed that inhibition of BMX in primary FLT3-mutated AML samples decreased chemokine secretion and enhanced the activity of gilteritinib. Collectively, these findings indicate a crucial role for microenvironment-mediated factors modulated by BMX in the escape from targeted therapy and have implications for the development of novel therapeutic interventions to restore sensitivity to gilteritinib.
Collapse
Affiliation(s)
- Daelynn R. Buelow
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Bhavana Bhatnagar
- West Virginia University Cancer Institute, Department of Hematology and Medical Oncology, Wheeling, WV; and
| | - Shelley J. Orwick
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Jae Yoon Jeon
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Eric D. Eisenmann
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Jack C. Stromatt
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Navjot Singh Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - James S. Blachly
- Division of Hematology, College of Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Sharyn D. Baker
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Bradley W. Blaser
- Division of Hematology, College of Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| |
Collapse
|
5
|
Next Generation Cytogenetics in Myeloid Hematological Neoplasms: Detection of CNVs and Translocations. Cancers (Basel) 2021; 13:cancers13123001. [PMID: 34203905 PMCID: PMC8232573 DOI: 10.3390/cancers13123001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Conventional cytogenetic approaches are the gold standard for the identification of chromosomal alterations in myeloid neoplasms. Next-generation sequencing panels are a new approach for the detection of copy number variations (CNV) or translocations. Here we report on a commercial panel utility including frequent mutations, CNVs and translocations in myeloid neoplasms. A total of 135 patients with myeloid neoplasms and three with acute lymphoblastic leukemia were analyzed by NGS. When comparing with gold standard techniques, 48 frequent alterations were detected by both methodologies, ten of them observed only by conventional methods and another eight only by NGS. Additionally, 38 secondary CNVs were detected in any of the genes included in the panel for mutational analysis. With those results we determine that NGS represents a reliable complementary source of information for the analysis of CNVs and translocations. Abstract Conventional cytogenetics are the gold standard for the identification of chromosomal alterations recurrent in myeloid neoplasms. Some next-generation sequencing (NGS) panels are designed for the detection of copy number variations (CNV) or translocations; however, their use is far from being widespread. Here we report on the results of a commercial panel including frequent mutations, CNVs and translocations in myeloid neoplasms. Frequent chromosomal alterations were analyzed by NGS in 135 patients with myeloid neoplasms and three with acute lymphoblastic leukemia. NGS analysis was performed using the enrichment-capture Myeloid Neoplasm-GeneSGKit (Sistemas Genómicos, Spain) gene panel including 35 genes for mutational analysis and frequent CNVs and translocations. NGS results were validated with cytogenetics and/or MLPA when possible. A total of 66 frequent alterations included in NGS panel were detected, 48 of them detected by NGS and cytogenetics. Ten of them were observed only by cytogenetics (mainly trisomy 8), and another eight only by NGS (mainly deletion of 12p). Aside from this, 38 secondary CNVs were detected in any of the genes included mainly for mutational analysis. NGS represents a reliable complementary source of information for the analysis of CNVs and translocations. Moreover, NGS could be a useful tool for the detection of alterations not observed by conventional cytogenetics.
Collapse
|
6
|
BCOR gene alterations in hematological diseases. Blood 2021; 138:2455-2468. [PMID: 33945606 DOI: 10.1182/blood.2021010958] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/14/2021] [Indexed: 11/20/2022] Open
Abstract
The BCL6 co-repressor (BCOR) is a transcription factor involved in the control of embryogenesis, mesenchymal stem cells function, hematopoiesis and lymphoid development. Recurrent somatic clonal mutations of the BCOR gene and its homologue BCORL1 have been detected in several hematological malignancies and aplastic anemia. They are scattered across the whole gene length and mostly represent frameshifts (deletions, insertions), nonsense and missence mutations. These disruptive events lead to the loss of full-length BCOR protein and to the lack or low expression of a truncated form of the protein, both consistent with the tumor suppressor role of BCOR. BCOR and BCORL1 mutations are similar to those causing two rare X-linked diseases: the oculo-facio-cardio-dental (OFCD) and the Shukla-Vernon syndromes, respectively. Here, we focus on the structure and function of normal BCOR and BCORL1 in normal hematopoietic and lymphoid tissues and review the frequency and clinical significance of the mutations of these genes in malignant and non-malignant hematological diseases. Moreover, we discuss the importance of mouse models to better understand the role of Bcor loss, alone and combined with alterations of other genes (e.g. Dnmt3a and Tet2), in promoting hematological malignancies and in providing a useful platform for the development of new targeted therapies.
Collapse
|
7
|
Feusier JE, Arunachalam S, Tashi T, Baker MJ, VanSant-Webb C, Ferdig A, Welm BE, Rodriguez-Flores JL, Ours C, Jorde LB, Prchal JT, Mason CC. Large-Scale Identification of Clonal Hematopoiesis and Mutations Recurrent in Blood Cancers. Blood Cancer Discov 2021; 2:226-237. [PMID: 34027416 DOI: 10.1158/2643-3230.bcd-20-0094] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is characterized by detectable hematopoietic-associated gene mutations in a person without evidence of hematologic malignancy. We sought to identify additional cancer-presenting mutations useable for CHIP detection by performing a data mining analysis of 48 somatic mutation studies reporting mutations at diagnoses of 7,430 adult and pediatric patients with hematologic malignancies. Following extraction of 20,141 protein-altering mutations, we identified 434 significantly recurrent mutation hotspots, 364 of which occurred at loci confidently assessable for CHIP. We then performed an additional large-scale analysis of whole exome sequencing data from 4,538 persons belonging to three non-cancer cohorts for clonal mutations. We found the combined cohort prevalence of CHIP with mutations identical to those reported at blood cancer mutation hotspots to be 1.8%, and that some of these CHIP mutations occurred in children. Our findings may help to improve CHIP detection and pre-cancer surveillance for both children and adults.
Collapse
Affiliation(s)
- Julie E Feusier
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Utah, Salt Lake City, UT, USA.,Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Sasi Arunachalam
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Utah, Salt Lake City, UT, USA.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Tsewang Tashi
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, UT, USA.,VA Medical Center, Salt Lake City, UT, USA
| | - Monika J Baker
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Utah, Salt Lake City, UT, USA
| | - Chad VanSant-Webb
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Utah, Salt Lake City, UT, USA
| | - Amber Ferdig
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Utah, Salt Lake City, UT, USA
| | - Bryan E Welm
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Surgery, University of Utah, Salt Lake City, UT, USA
| | | | - Christopher Ours
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Utah, Salt Lake City, UT, USA
| | - Lynn B Jorde
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Josef T Prchal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, UT, USA.,VA Medical Center, Salt Lake City, UT, USA
| | - Clinton C Mason
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
8
|
Testa U, Castelli G, Pelosi E. Isocitrate Dehydrogenase Mutations in Myelodysplastic Syndromes and in Acute Myeloid Leukemias. Cancers (Basel) 2020; 12:E2427. [PMID: 32859092 PMCID: PMC7564409 DOI: 10.3390/cancers12092427] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/03/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease generated by the acquisition of multiple genetic and epigenetic aberrations which impair the proliferation and differentiation of hematopoietic progenitors and precursors. In the last years, there has been a dramatic improvement in the understanding of the molecular alterations driving cellular signaling and biochemical changes determining the survival advantage, stimulation of proliferation, and impairment of cellular differentiation of leukemic cells. These molecular alterations influence clinical outcomes and provide potential targets for drug development. Among these alterations, an important role is played by two mutant enzymes of the citric acid cycle, isocitrate dehydrogenase (IDH), IDH1 and IDH2, occurring in about 20% of AMLs, which leads to the production of an oncogenic metabolite R-2-hydroxy-glutarate (R-2-HG); this causes a DNA hypermethylation and an inhibition of hematopoietic stem cell differentiation. IDH mutations differentially affect prognosis of AML patients following the location of the mutation and other co-occurring genomic abnormalities. Recently, the development of novel therapies based on the specific targeting of mutant IDH may contribute to new effective treatments of these patients. In this review, we will provide a detailed analysis of the biological, clinical, and therapeutic implications of IDH mutations.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (G.C.); (E.P.)
| | | | | |
Collapse
|
9
|
Bewersdorf JP, Shallis RM, Diadamo A, Gowda L, Podoltsev NA, Siddon A, Zeidan AM. Isolated trisomy 11 in patients with acute myeloid leukemia - is the prognosis not as grim as previously thought? Leuk Lymphoma 2020; 61:2254-2257. [PMID: 32338096 DOI: 10.1080/10428194.2020.1755858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Jan Philipp Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
| | - Rory M Shallis
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
| | - Autumn Diadamo
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Lohith Gowda
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
| | - Nikolai A Podoltsev
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA
| | - Alexa Siddon
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA.,Department of Pathology, Section of Hematopathology, Yale School of Medicine, New Haven, CT, USA
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine, New Haven, CT, USA.,Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT, USA
| |
Collapse
|
10
|
Vetro C, Haferlach T, Meggendorfer M, Stengel A, Jeromin S, Kern W, Haferlach C. Cytogenetic and molecular genetic characterization of KMT2A-PTD positive acute myeloid leukemia in comparison to KMT2A-Rearranged acute myeloid leukemia. Cancer Genet 2019; 240:15-22. [PMID: 31698332 DOI: 10.1016/j.cancergen.2019.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/23/2019] [Accepted: 10/28/2019] [Indexed: 12/19/2022]
Abstract
To define the biological differences in acute myeloid leukaemia (AML) with KMT2A gene involvements and their prognostic impact, we compared 190 de novo AML patients at diagnosis, 95 harbouring KMT2A-rearrangement (KMT2Ar) and 95 KMT2A-PTD by performing cytogenetic and molecular genetic analyses. Both AML subtypes had an unfavourable outcome, particularly in patients > 60 years. Patients with KMT2Ar were younger compared to patients with KMT2A-PTD (mean 52 vs 65 years, p < 0.001) and had a higher rate of additional cytogenetic abnormalities (ACA) (46% vs 25% of cases). In both groups, occurrence of ACA did not influence the overall survival (OS). Regarding molecular genetics, 66% of patients with KMT2Ar and 99% of patients with KMT2A-PTD had additional gene mutations. In multivariate analysis, KRAS mutations and 10p12 rearrangement resulted as adverse prognostic factors in KMT2Ar subgroup. In the KMT2A-PTD group, apart from age, only the occurrence of DNMT3A non-R882 mutations correlated with shorter OS.
Collapse
|
11
|
Clinical and molecular characterization of patients with acute myeloid leukemia and sole trisomies of chromosomes 4, 8, 11, 13 or 21. Leukemia 2019; 34:358-368. [PMID: 31462731 DOI: 10.1038/s41375-019-0560-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/01/2019] [Accepted: 07/15/2019] [Indexed: 12/13/2022]
Abstract
Sole trisomies of chromosomes 4, 8, 11, 13 and 21 account for 89-95% of all sole trisomies in adult AML patients. We analyzed clinical and molecular characteristics of 138 de novo AML patients with sole +4, +8, +11, +13 or +21, and compared them with AML patients with those trisomies occurring in addition to other chromosome abnormalities (non-sole trisomy) and with cytogenetically normal AML (CN-AML) patients. Mutations in methylation-related genes were most commonly observed within each sole trisomy group (+4, 55%; +8, 58%; +11, 71%; +13, 71%; +21, 75% of patients). Patients with sole trisomies, excluding +4, also had frequent mutations in spliceosome genes (+8, 43%; +11, 65%; +13, 65%; +21, 45% of patients). In contrast, +4 patients frequently had mutations in transcription factor genes (44%) and NPM1 (36%). While 48% of patients with sole trisomies harbored mutations in a spliceosome gene, spliceosome mutations were observed in only 24% of non-sole trisomy (n = 131, P < 0.001) and 19% of CN-AML patients (n = 716, P < 0.001). Our data suggest that mutations affecting methylation-related genes are a molecular hallmark of sole trisomies. Mutations in spliceosome genes were also commonly observed in many sole trisomy patients and represent a novel finding in this cytogenetic subgroup.
Collapse
|
12
|
Buelow DR, Pounds SB, Wang YD, Shi L, Li Y, Finkelstein D, Shurtleff S, Neale G, Inaba H, Ribeiro RC, Palumbo R, Garrison D, Orwick SJ, Blachly JS, Kroll K, Byrd JC, Gruber TA, Rubnitz JE, Baker SD. Uncovering the Genomic Landscape in Newly Diagnosed and Relapsed Pediatric Cytogenetically Normal FLT3-ITD AML. Clin Transl Sci 2019; 12:641-647. [PMID: 31350825 PMCID: PMC6853146 DOI: 10.1111/cts.12669] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/12/2019] [Indexed: 12/18/2022] Open
Abstract
Fms-like tyrosine kinase 3 (FLT3) internal tandem duplication (ITD) mutations, common in pediatric acute myeloid leukemia (AML), associate with early relapse and poor prognosis. Past studies have suggested additional cooperative mutations are required for leukemogenesis in FLT3-ITD+ AML. Using RNA sequencing and a next-generation targeted gene panel, we broadly characterize the co-occurring genomic alterations in pediatric cytogenetically normal (CN) FLT3-ITD+ AML to gain a deeper understanding of the clonal patterns and heterogeneity at diagnosis and relapse. We show that chimeric transcripts were present in 21 of 34 (62%) of de novo samples, 2 (6%) of these samples included a rare reoccurring fusion partner BCL11B. At diagnosis, the median number of mutations other than FLT3 per patient was 1 (range 0-3), which involved 8 gene pathways; WT1 and NPM1 mutations were frequently observed (35% and 24%, respectively). Fusion transcripts and high variant allele frequency (VAF) mutants, which included WT1, NPM1, SMARCA2, RAD21, and TYK2, were retained from diagnosis to relapse. We did observe reduction in VAF of simple or single mutation clones, but VAFs were preserved or expanded in more complex clones with multiple mutations. Our data provide the first insight into the genomic complexity of pediatric CN FLT3-ITD+ AML and could help stratify future targeted treatment strategies.
Collapse
Affiliation(s)
- Daelynn R Buelow
- Division of Pharmaceutics, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Stanley B Pounds
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Yong-Dong Wang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Lei Shi
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Yongjin Li
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Sheila Shurtleff
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Geoffrey Neale
- Hartwell Center, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Raul C Ribeiro
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Reid Palumbo
- Division of Pharmaceutics, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Dominique Garrison
- Division of Pharmaceutics, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Shelley J Orwick
- Division of Hematology, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - James S Blachly
- Division of Hematology, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Karl Kroll
- Division of Hematology, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - John C Byrd
- Division of Pharmaceutics, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA.,Division of Hematology, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Tanja A Gruber
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jeffrey E Rubnitz
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Sharyn D Baker
- Division of Pharmaceutics, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA.,Division of Hematology, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
13
|
Abstract
BCOR is a gene that encodes for an epigenetic regulator involved in the specification of cell differentiation and body structure development and takes part in the noncanonical polycomb repressive complex 1. This review provides a comprehensive summary of BCOR’s involvement in oncology, illustrating that various BCOR aberrations, such as the internal tandem duplications of the PCGF Ub-like fold discriminator domain and different gene fusions (mainly BCOR–CCNB3, BCOR–MAML3 and ZC3H7B–BCOR), represent driver elements of various sarcomas such as clear cell sarcoma of the kidney, primitive mesenchymal myxoid tumor of infancy, small round blue cell sarcoma, endometrial stromal sarcoma and histologically heterogeneous CNS neoplasms group with similar genomic methylation patterns known as CNS-HGNET-BCOR. Furthermore, other BCOR alterations (often loss of function mutations) recur in a large variety of mesenchymal, epithelial, neural and hematological tumors, suggesting a central role in cancer evolution.
Collapse
Affiliation(s)
- Annalisa Astolfi
- 'Giorgio Prodi' Cancer Research Center, University of Bologna, 40138 Bologna, Italy
| | - Michele Fiore
- Pediatric Oncology & Hematology Unit 'Lalla Seràgnoli', S Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Fraia Melchionda
- Pediatric Oncology & Hematology Unit 'Lalla Seràgnoli', S Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Valentina Indio
- 'Giorgio Prodi' Cancer Research Center, University of Bologna, 40138 Bologna, Italy
| | - Salvatore N Bertuccio
- Pediatric Oncology & Hematology Unit 'Lalla Seràgnoli', S Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Andrea Pession
- Pediatric Oncology & Hematology Unit 'Lalla Seràgnoli', S Orsola-Malpighi Hospital, 40138 Bologna, Italy.,Department of Medical & Surgical Sciences, University of Bologna, S Orsola-Malpighi Hospital, 40138 Bologna, Italy
| |
Collapse
|
14
|
Ferret Y, Boissel N, Helevaut N, Madic J, Nibourel O, Marceau-Renaut A, Bucci M, Geffroy S, Celli-Lebras K, Castaigne S, Thomas X, Terré C, Dombret H, Preudhomme C, Renneville A. Clinical relevance of IDH1/2 mutant allele burden during follow-up in acute myeloid leukemia. A study by the French ALFA group. Haematologica 2018; 103:822-829. [PMID: 29472349 PMCID: PMC5927984 DOI: 10.3324/haematol.2017.183525] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 02/16/2018] [Indexed: 12/27/2022] Open
Abstract
Assessment of minimal residual disease has emerged as a powerful prognostic factor in acute myeloid leukemia. In this study, we investigated the potential of IDH1/2 mutations as targets for minimal residual disease assessment in acute myeloid leukemia, since these mutations collectively occur in 15–20% of cases of acute myeloid leukemia and now represent druggable targets. We employed droplet digital polymerase chain reaction assays to quantify IDH1R132, IDH2R140, and IDH2R172 mutations on genomic DNA in 322 samples from 103 adult patients with primary IDH1/2 mutant acute myeloid leukemia and enrolled on Acute Leukemia French Association (ALFA) - 0701 or -0702 clinical trials. The median IDH1/2 mutant allele fraction in bone marrow samples was 42.3% (range, 8.2 – 49.9%) at diagnosis of acute myeloid leukemia, and below the detection limit of 0.2% (range, <0.2 – 39.3%) in complete remission after induction therapy. In univariate analysis, the presence of a normal karyotype, a NPM1 mutation, and an IDH1/2 mutant allele fraction <0.2% in bone marrow after induction therapy were statistically significant predictors of longer disease-free survival. In multivariate analysis, these three variables remained significantly predictive of disease-free survival. In 7/103 (7%) patients, IDH1/2 mutations persisted at high levels in complete remission, consistent with the presence of an IDH1/2 mutation in pre-leukemic hematopoietic stem cells. Five out of these seven patients subsequently relapsed or progressed toward myelodysplastic syndrome, suggesting that patients carrying the IDH1/2 mutation in a pre-leukemic clone may be at high risk of hematologic evolution.
Collapse
Affiliation(s)
- Yann Ferret
- Hematology Laboratory, Biology and Pathology Center, CHRU of Lille, France.,INSERM, UMR-S 1172, Cancer Research Institute of Lille, Paris, France.,University of Lille, F-59000, Paris, France
| | - Nicolas Boissel
- Hematology Department, Saint-Louis Hospital, AP-HP, Paris, France.,EA3518, Institut Universitaire d'Hématologie (IUH), University 7 Paris Diderot, Paris, France
| | - Nathalie Helevaut
- Hematology Laboratory, Biology and Pathology Center, CHRU of Lille, France
| | - Jordan Madic
- Circulating Biomarkers Laboratory, Curie Institute, Paris, France
| | - Olivier Nibourel
- Hematology Laboratory, Biology and Pathology Center, CHRU of Lille, France.,INSERM, UMR-S 1172, Cancer Research Institute of Lille, Paris, France.,University of Lille, F-59000, Paris, France
| | - Alice Marceau-Renaut
- Hematology Laboratory, Biology and Pathology Center, CHRU of Lille, France.,INSERM, UMR-S 1172, Cancer Research Institute of Lille, Paris, France.,University of Lille, F-59000, Paris, France
| | - Maxime Bucci
- Hematology Laboratory, Biology and Pathology Center, CHRU of Lille, France
| | - Sandrine Geffroy
- Hematology Laboratory, Biology and Pathology Center, CHRU of Lille, France
| | - Karine Celli-Lebras
- Acute Leukemia French Association (ALFA) coordination, Saint-Louis Hospital, AP-HP, Paris, France
| | | | - Xavier Thomas
- Hematology Department, Lyon Sud Hospital, Pierre Benite, France
| | - Christine Terré
- Cytogenetic Laboratory, Versailles Hospital, Le Chesnay, France
| | - Hervé Dombret
- Hematology Department, Saint-Louis Hospital, AP-HP, Paris, France.,EA3518, Institut Universitaire d'Hématologie (IUH), University 7 Paris Diderot, Paris, France
| | - Claude Preudhomme
- Hematology Laboratory, Biology and Pathology Center, CHRU of Lille, France.,INSERM, UMR-S 1172, Cancer Research Institute of Lille, Paris, France.,University of Lille, F-59000, Paris, France
| | - Aline Renneville
- Hematology Laboratory, Biology and Pathology Center, CHRU of Lille, France .,INSERM, UMR-S 1172, Cancer Research Institute of Lille, Paris, France.,University of Lille, F-59000, Paris, France
| |
Collapse
|
15
|
Yoo SH, Lim J, Byun JM, Park JH, Kim KH, Choi IS. Adult chronic myelomonocytic leukemia with trisomy 11: a case report. ACTA ACUST UNITED AC 2017; 24:e547-e550. [PMID: 29270065 DOI: 10.3747/co.24.3738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Chronic myelomonocytic leukemia (cmml) is an indolent disease in the category of myelodysplastic and myeloproliferative neoplasms, which can often evolve into acute leukemic neoplasms. Although cytogenetic abnormalities such as trisomy 8 or absence of chromosome Y are well known, few reports about cmml with trisomy 11 have been published. Here, we report a case of cmml with trisomy 11 as the sole chromosomal abnormality, resulting in a very poor outcome. Based on a bone marrow specimen, cmml-1 with trisomy 11 was diagnosed in a 79-year-old man presenting with anemia and atypical peripheral blood cells. Because of the patient's age, he was followed without receiving anticancer treatment. Two months after his diagnosis, the patient's leucocytosis and anemia rapidly worsened, with increasing numbers of immature peripheral cells, which was strongly suggestive of leukemic transformation. Because of acute kidney injury superimposed on chronic kidney disease that led to poor performance status, cytotoxic chemotherapy was not considered feasible, and the patient was transferred to a hospice care facility.
Collapse
Affiliation(s)
- S H Yoo
- Department of Internal Medicine, Seoul National University College of Medicine
| | - J Lim
- Department of Internal Medicine, Seoul Red Cross Hospital, and
| | - J M Byun
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - J H Park
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - K H Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - I S Choi
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| |
Collapse
|
16
|
The mutational oncoprint of recurrent cytogenetic abnormalities in adult patients with de novo acute myeloid leukemia. Leukemia 2017; 31:2211-2218. [PMID: 28321123 PMCID: PMC5628133 DOI: 10.1038/leu.2017.86] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/17/2017] [Accepted: 02/16/2017] [Indexed: 12/18/2022]
Abstract
Recurrent chromosomal abnormalities and gene mutations detected at the time of diagnosis of acute myeloid leukemia (AML) are associated with particular disease features, treatment response and survival of AML patients, and are used to denote specific disease entities in the World Health Organization classification of myeloid neoplasms and acute leukemia. However, large studies that integrate cytogenetic and comprehensive mutational information are scarce. We created a comprehensive oncoprint of mutations associated with recurrent cytogenetic findings by combining the information on mutational patterns of 80 cancer- and leukemia-associated genes with cytogenetic findings in 1603 adult patients with de novo AML. We show unique differences in the mutational profiles among major cytogenetic subsets, identify novel associations between recurrent cytogenetic abnormalities and both specific gene mutations and gene functional groups, and reveal differences in cytogenetic and mutational features between patients younger than 60 years and those aged 60 years or older. The identified associations between cytogenetic and molecular genetic data may help guide mutation testing in AML, and result in more focused application of targeted therapy in patients with de novo AML.
Collapse
|