1
|
Barone M, Patuelli A, Dicataldo M, Irno Consalvo M, Chirumbolo G, Bandini L, Atzeni G, Forte D, Cristiano G, Ottaviani E, Curti A, Buccisano F, Catani L, Arpinati M. Validation Study of Analytical Methods for Multiparameter Flow Cytometry-Based Measurable Residual Disease Assessment in Acute Myeloid Leukemia. Int J Mol Sci 2025; 26:4506. [PMID: 40429651 PMCID: PMC12110934 DOI: 10.3390/ijms26104506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/29/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
The standardization of multiparameter flow cytometry-based measurable residual disease (MFC-MRD) assessment in acute myeloid leukemia (AML) lacks clear criteria to define leukemia-associated immunophenotypes (LAIPs). In addition, the most specific/sensitive aberrations used to define LAIPs are often partially expressed by the leukemic clone at diagnosis, raising questions about their reliability for accurate MRD quantification. To address this, we investigated whether the quantification of LAIP+ cells reflects residual disease in cases of partial LAIP expression. The following two MFC-MRD approaches were evaluated by comparing their results to RT-qPCR for NPM1 mutations: (1) the LAIP-method, wherein all cells within the patient-specific template created at diagnosis are counted without further gating; (2) the LAIP-based different-from-normal (DfN)-method, wherein cells+ for LAIP-specific aberrant markers are further selected. A total of 125 bone marrow samples from 25 NPM1-mutated AML patients were studied. Our data demonstrate that the LAIP-based DfN-method improves the MFC-MRD accuracy and comparability with molecular MRD. ROC analysis identified cut-offs of 0.034% and 0.095% to discriminate positive/negative results in patients receiving intensive chemotherapy and hypomethylating agents, respectively. We also found distinct accuracy degrees based on the LAIP-specific aberrant markers used for MRD assessment. These results refine the MFC-MRD method and highlight the importance of therapy-specific MRD cut-offs and LAIP classification based on specificity and sensitivity.
Collapse
Affiliation(s)
- Martina Barone
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy; (A.P.); (G.A.); (E.O.); (A.C.); (L.C.); (M.A.)
| | - Agnese Patuelli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy; (A.P.); (G.A.); (E.O.); (A.C.); (L.C.); (M.A.)
| | - Michele Dicataldo
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (M.D.); (G.C.); (L.B.); (D.F.); (G.C.)
| | - Maria Irno Consalvo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (M.I.C.); (F.B.)
| | - Gabriella Chirumbolo
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (M.D.); (G.C.); (L.B.); (D.F.); (G.C.)
| | - Lorenza Bandini
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (M.D.); (G.C.); (L.B.); (D.F.); (G.C.)
| | - Giulia Atzeni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy; (A.P.); (G.A.); (E.O.); (A.C.); (L.C.); (M.A.)
| | - Dorian Forte
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (M.D.); (G.C.); (L.B.); (D.F.); (G.C.)
| | - Gianluca Cristiano
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (M.D.); (G.C.); (L.B.); (D.F.); (G.C.)
| | - Emanuela Ottaviani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy; (A.P.); (G.A.); (E.O.); (A.C.); (L.C.); (M.A.)
| | - Antonio Curti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy; (A.P.); (G.A.); (E.O.); (A.C.); (L.C.); (M.A.)
| | - Francesco Buccisano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; (M.I.C.); (F.B.)
| | - Lucia Catani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy; (A.P.); (G.A.); (E.O.); (A.C.); (L.C.); (M.A.)
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (M.D.); (G.C.); (L.B.); (D.F.); (G.C.)
| | - Mario Arpinati
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy; (A.P.); (G.A.); (E.O.); (A.C.); (L.C.); (M.A.)
| |
Collapse
|
2
|
Press RD, Dressel D, McBean M, Tiong IS, Anderson MW, Pride D, Raman A, Doom RR, Kaldate R. Evaluation of a New Closed-System Automated RT-qPCR Assay for the Rapid Detection and Monitoring of Common Nucleophosmin Mutations in Patients with Acute Myeloid Leukemia. Int J Mol Sci 2024; 25:7912. [PMID: 39063154 PMCID: PMC11277538 DOI: 10.3390/ijms25147912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Quantitative assessment of nucleophosmin 1 (NPM1) mutation status is integral to evaluating measurable residual disease (MRD) in NPM1-mutated acute myeloid leukemia (AML) patients. In a retrospective study, leftover peripheral blood (PB) specimens (n = 40) which were collected for routine clinical diagnostic evaluations of AML disease burden were tested by both a novel automated RT-qPCR quantitative NPM1 assay (Xpert NPM1 mutation assay) and the NPM1 mutA, mutB&D MutaQuant kit. Based on a Deming regression analysis, there was a high correlation (slope = 0.92; intercept = 0.12; Pearson's r = 0.982) between the quantitative results of the Xpert NPM1 mutation assay and the NPM1 mutA, mutB&D MutaQuant kit. The Xpert test quantitative results are thus highly correlated with the comparator method and the former has potential as a useful alternative for the monitoring of AML patients with a known NPM1 mutation.
Collapse
Affiliation(s)
- Richard D. Press
- Department of Pathology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Dana Dressel
- International Health Management Associates (IHMA), Schaumburg, IL 60173, USA;
| | - Michelle McBean
- Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (M.M.); (I.S.T.)
| | - Ing S. Tiong
- Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (M.M.); (I.S.T.)
| | | | - David Pride
- Department of Pathology, University of California, San Diego, CA 92103, USA;
| | - Aarthi Raman
- Cepheid, Sunnyvale, CA 94089, USA; (A.R.); (R.R.D.); (R.K.)
| | | | - Rajesh Kaldate
- Cepheid, Sunnyvale, CA 94089, USA; (A.R.); (R.R.D.); (R.K.)
| |
Collapse
|
3
|
Yang J, Zhu X, Zhang H, Fu Y, Li Z, Xing Z, Yu Y, Cao P, Le J, Jiang J, Li J, Wang H, Zhai X. Prognostic Factors of Pediatric Acute Myeloid Leukemia Patients with t(8;21) (q22;q22): A Single-Center Retrospective Study. CHILDREN (BASEL, SWITZERLAND) 2024; 11:605. [PMID: 38790600 PMCID: PMC11120327 DOI: 10.3390/children11050605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024]
Abstract
This retrospective study aimed to analyze the treatment effect and prognostic factors of pediatric acute myeloid leukemia (AML) patients with t(8;21). A total of 268 newly diagnosed pediatric AML (pAML) enrolled from 1 January 2005 to 31 December 2022 were retrospectively reviewed, and 50 (18.7%) patients harbored t(8;21) translocation. CR rate, OS, EFS, and RFS were assessed by multivariate Logistic and Cox regression models in these patients. Of the 50 patients, 2 patients abandoned treatment during the first induction course. Of the remaining 48 patients who received double-induction therapy and were included in the final analyses, CR1 and CR2 were 75.0% (36/48) and 95.8% (46/48), respectively. The overall three-year OS, EFS, and RFS were 68.4% (95% CI, 55.0-85.1), 64.2% (95% CI, 50.7-81.4), and 65.5% (95% CI, 51.9-82.8), respectively. The presence of loss of sex chromosome (LOS) at diagnosis (n = 21) was associated with a better 3-year OS [87.5% (95% CI, 72.7-100) vs. 52.7% (95% CI, 35.1-79.3), p = 0.0089], 3-year EFS [81.6% (95% CI, 64.7-100) vs. 49.7% (95% CI, 32.4-76.4), p = 0.023], and 3-year RFS [81.6% (95% CI, 64.7-100) vs. 51.7% (95% CI, 33.9-78.9), p = 0.036] than those without LOS (n = 27), and it was also an independent good prognostic factor of OS (HR, 0.08 [95% CI, 0.01-0.48], p = 0.005), EFS (HR, 0.22 [95% CI, 0.05-0.85], p = 0.029), and RFS (HR, 0.21 [95% CI, 0.05-0.90], p = 0.035). However, extramedullary leukemia (EML) featured the independent risk factors of inferior OS (HR, 10.99 [95% CI, 2.08-58.12], p = 0.005), EFS (HR, 4.75 [95% CI, 1.10-20.61], p = 0.037), and RFS (HR, 6.55 [95% CI, 1.40-30.63], p = 0.017) in pediatric individuals with t(8;21) AML. Further analysis of combining LOS with EML indicated that the EML+LOS- subgroup had significantly inferior OS (92.9%, [95% CI, 80.3-100]), EFS (86.2%, [95% CI, 70.0-100]), and RFS (86.2%, [95% CI, 80.3-100]) compared to the other three subgroups (all p < 0.001). LOS and EML are independent prognostic factors of OS, EFS, and RFS with t(8;21) pAML patients. LOS combined with EML may help improve risk stratification.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Xiaowen Zhai
- Department of Hematology and Oncology, National Children’s Medical Center, Children’s Hospital of Fudan University, Shanghai 201102, China
| |
Collapse
|
4
|
Leung B, Aung H, Nandini A, Abdulrasool G, Lau C, Seymour L. Analytical Validation of a 37-Gene Next-Generation Sequencing Panel for Myeloid Malignancies and Review of Initial Findings Incorporating Updated 2022 Diagnostic and Prognostic Guidelines. J Mol Diagn 2024; 26:399-412. [PMID: 38367765 DOI: 10.1016/j.jmoldx.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 10/23/2023] [Accepted: 01/16/2024] [Indexed: 02/19/2024] Open
Abstract
Myeloid neoplasms are clonal disorders that arise via acquisition of genetic mutations leading to excessive proliferation and defective differentiation. Mutational profiling is vital as it has implications for diagnosis, prognosis, and therapeutic decision-making. Next-generation sequencing (NGS) has become a mainstay in the evaluation of myeloid malignancies, as it enables efficient characterization of multiple genetic changes. Herein, the analytical validation of the 37-gene Archer VariantPlex Core Myeloid panel is reported, using 58 DNA specimens with 87 single-nucleotide variants and 23 insertions/deletions. The panel achieved good depth of coverage, 100% analytical sensitivity and specificity for single-nucleotide variants and insertions/deletions ≤21 bp, and 100% reproducibility, with a reportable limit of detection determined as 5%. The Archer NGS panel can accurately and reproducibly detect variants of clinical significance in myeloid neoplasms. A retrospective analysis of 535 clinical specimens tested with the Archer NGS panel showed a frequency and pattern of mutations across myeloid malignancies that were similar to other published studies. A review of the diagnostic classification of patients with acute myeloid leukemia and myelodysplastic syndrome using the World Health Organization 2017/2022 and International Consensus Classification 2022 guidelines, in addition to European LeukemiaNet 2017/2022 risk stratification of patients with acute myeloid leukemia, was also performed to assess the utility of the molecular information provided by the Archer NGS panel.
Collapse
Affiliation(s)
- Becky Leung
- Department of Haematology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; School of Medicine, Griffith University, Gold Coast, Queensland, Australia.
| | - Hnin Aung
- Department of Haematology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Adayapalam Nandini
- Department of Haematology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Ghusoon Abdulrasool
- Department of Haematology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Chiyan Lau
- Department of Haematology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Louise Seymour
- Department of Haematology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
5
|
Edsjö A, Gisselsson D, Staaf J, Holmquist L, Fioretos T, Cavelier L, Rosenquist R. Current and emerging sequencing-based tools for precision cancer medicine. Mol Aspects Med 2024; 96:101250. [PMID: 38330674 DOI: 10.1016/j.mam.2024.101250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/22/2024] [Indexed: 02/10/2024]
Abstract
Current precision cancer medicine is dependent on the analyses of a plethora of clinically relevant genomic aberrations. During the last decade, next-generation sequencing (NGS) has gradually replaced most other methods for precision cancer diagnostics, spanning from targeted tumor-informed assays and gene panel sequencing to global whole-genome and whole-transcriptome sequencing analyses. The shift has been impelled by a clinical need to assess an increasing number of genomic alterations with diagnostic, prognostic and predictive impact, including more complex biomarkers (e.g. microsatellite instability, MSI, and homologous recombination deficiency, HRD), driven by the parallel development of novel targeted therapies and enabled by the rapid reduction in sequencing costs. This review focuses on these sequencing-based methods, puts their emergence in a historic perspective, highlights their clinical utility in diagnostics and decision-making in pediatric and adult cancer, as well as raises challenges for their clinical implementation. Finally, the importance of applying sensitive tools for longitudinal monitoring of treatment response and detection of measurable residual disease, as well as future avenues in the rapidly evolving field of sequencing-based methods are discussed.
Collapse
Affiliation(s)
- Anders Edsjö
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden; Division of Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | - David Gisselsson
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden; Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Johan Staaf
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, Lund, Sweden
| | - Louise Holmquist
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden
| | - Thoas Fioretos
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden; Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden; Clinical Genomics Lund, Science for Life Laboratory, Lund University, Lund, Sweden
| | - Lucia Cavelier
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Clinical Genetics and Genomics, Karolinska University Hospital, Stockholm, Sweden
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Clinical Genetics and Genomics, Karolinska University Hospital, Stockholm, Sweden; Genomic Medicine Center Karolinska, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
6
|
Chea M, Rigolot L, Canali A, Vergez F. Minimal Residual Disease in Acute Myeloid Leukemia: Old and New Concepts. Int J Mol Sci 2024; 25:2150. [PMID: 38396825 PMCID: PMC10889505 DOI: 10.3390/ijms25042150] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Minimal residual disease (MRD) is of major importance in onco-hematology, particularly in acute myeloid leukemia (AML). MRD measures the amount of leukemia cells remaining in a patient after treatment, and is an essential tool for disease monitoring, relapse prognosis, and guiding treatment decisions. Patients with a negative MRD tend to have superior disease-free and overall survival rates. Considerable effort has been made to standardize MRD practices. A variety of techniques, including flow cytometry and molecular methods, are used to assess MRD, each with distinct strengths and weaknesses. MRD is recognized not only as a predictive biomarker, but also as a prognostic tool and marker of treatment efficacy. Expected advances in MRD assessment encompass molecular techniques such as NGS and digital PCR, as well as optimization strategies such as unsupervised flow cytometry analysis and leukemic stem cell monitoring. At present, there is no perfect method for measuring MRD, and significant advances are expected in the future to fully integrate MRD assessment into the management of AML patients.
Collapse
Affiliation(s)
- Mathias Chea
- Laboratoire d’Hématologie Biologique, Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (M.C.); (L.R.); (A.C.)
| | - Lucie Rigolot
- Laboratoire d’Hématologie Biologique, Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (M.C.); (L.R.); (A.C.)
- School of Medicine, Université Toulouse III Paul Sabatier, 31062 Toulouse, France
| | - Alban Canali
- Laboratoire d’Hématologie Biologique, Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (M.C.); (L.R.); (A.C.)
- School of Medicine, Université Toulouse III Paul Sabatier, 31062 Toulouse, France
| | - Francois Vergez
- Laboratoire d’Hématologie Biologique, Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (M.C.); (L.R.); (A.C.)
- School of Medicine, Université Toulouse III Paul Sabatier, 31062 Toulouse, France
| |
Collapse
|
7
|
Lin M, Zhao X, Chang Y, Zhao X. Current assessment and management of measurable residual disease in patients with acute lymphoblastic leukemia in the setting of CAR-T-cell therapy. Chin Med J (Engl) 2024; 137:140-151. [PMID: 38148315 PMCID: PMC10798764 DOI: 10.1097/cm9.0000000000002945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Indexed: 12/28/2023] Open
Abstract
ABSTRACT Chimeric antigen receptor (CAR)-modified T-cell therapy has achieved remarkable success in the treatment of acute lymphoblastic leukemia (ALL). Measurable/minimal residual disease (MRD) monitoring plays a significant role in the prognostication and management of patients undergoing CAR-T-cell therapy. Common MRD detection methods include flow cytometry (FCM), polymerase chain reaction (PCR), and next-generation sequencing (NGS), and each method has advantages and limitations. It has been well documented that MRD positivity predicts a poor prognosis and even disease relapse. Thus, how to perform prognostic evaluations, stratify risk based on MRD status, and apply MRD monitoring to guide individual therapeutic decisions have important implications in clinical practice. This review assesses the common and novel MRD assessment methods. In addition, we emphasize the critical role of MRD as a prognostic biomarker and summarize the latest studies regarding MRD-directed combination therapy with CAR-T-cell therapy and allogeneic hematopoietic stem cell transplantation (allo-HSCT), as well as other therapeutic strategies to improve treatment effect. Furthermore, this review discusses current challenges and strategies for MRD detection in the setting of disease relapse after targeted therapy.
Collapse
Affiliation(s)
- Minghao Lin
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Xiaosu Zhao
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Yingjun Chang
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Xiangyu Zhao
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| |
Collapse
|
8
|
Zheng Y, Pan L, Li J, Feng X, Li C, Zheng M, Mai H, Yang L, He Y, He X, Xu H, Wen H, Le S. Prognostic significance of multiparametric flow cytometry minimal residual disease at two time points after induction in pediatric acute myeloid leukemia. BMC Cancer 2024; 24:46. [PMID: 38195455 PMCID: PMC10775489 DOI: 10.1186/s12885-023-11784-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 12/21/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Prompt response to induction chemotherapy is a prognostic factor in pediatric acute myeloid leukemia. In this study, we aimed to evaluate the prognostic significance of multiparametric flow cytometry-minimal residual disease (MFC-MRD), assessed at the end of the first and second induction courses. METHODS MFC-MRD was performed at the end of the first induction (TP1) in 524 patients and second induction (TP2) in 467 patients who were treated according to the modified Medical Research Council (UK) acute myeloid leukemia 15 protocol. RESULTS Using a 0.1% cutoff level, patients with MFC-MRD at the two time points had lower event-free survival and overall survival. Only the TP2 MFC-MRD level could predict the outcome in a separate analysis of high and intermediate risks based on European LeukemiaNet risk stratification and KMT2A rearrangement. The TP2 MFC-MRD level could further differentiate the prognosis of patients into complete remission or non-complete remission based on morphological evaluation. Multivariate analysis indicated the TP2 MFC-MRD level as an independent adverse prognostic factor for event-free survival and overall survival. When comparing patients with MFC-MRD ≥ 0.1%, those who underwent hematopoietic stem cell transplant during the first complete remission had significantly higher 5-year event-free survival and overall survival and lower cumulative incidence of relapse than those who only received consolidation chemotherapy. CONCLUSIONS The TP2 MFC-MRD level can predict the outcomes in pediatric patients with acute myeloid leukemia and help stratify post-remission treatment.
Collapse
Affiliation(s)
- Yongzhi Zheng
- Department of Pediatric Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lili Pan
- Department of Pediatric Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jian Li
- Department of Pediatric Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoqin Feng
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunfu Li
- Nanfang-Chunfu Children's Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China
| | - Mincui Zheng
- Department of Pediatric Hematology/Oncology, Hematology and Oncology, Hunan Children's Hospital, Changsha, China
| | - Huirong Mai
- Department of Pediatric Hematology/Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Lihua Yang
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Yingyi He
- Department of Pediatric Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Xiangling He
- People's Hospital of Hunan Province, Changsha, China
| | - Honggui Xu
- Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Hong Wen
- The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Shaohua Le
- Department of Pediatric Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory On Hematology, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
9
|
Koo M, Song IC, Kim J, Kwon GC, Kim SY. Prognostic value of the mutation types and dynamics of FLT3-ITD in acute myeloid leukemia. Eur J Haematol 2023; 111:562-572. [PMID: 37435718 DOI: 10.1111/ejh.14044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/03/2023] [Accepted: 07/03/2023] [Indexed: 07/13/2023]
Abstract
OBJECTIVE The prognostic value of the mutation types and dynamics of FLT3-ITD in acute myeloid leukemia (AML) and other known factors were studied. METHODS Initial and follow-up samples from 45 AML patients with FLT3-ITD mutations were analyzed by fragment length analysis, Sanger sequencing, and next-generation sequencing. RESULTS Some patients (13%) had multiple FLT3-ITD mutations, and many of them had acute promyelocytic leukemia (APL). FLT3-ITD mutations were classified according to mutation types, including duplication-only FLT3-ITD (52%) and FLT3-ITD with duplications and insertions (dup + ins) (48%). The dup + ins FLT3-ITD variant was independently associated with poor prognosis among non-APL patients (odds ratio, 2.92) in addition to FLT3-ITD with ≥50% variant allele frequency (VAF). The VAFs of FLT3-ITD were low (median 2.2%) when detected during morphologic complete remission (CR) after conventional chemotherapy; however, in two patients treated with gilteritinib after relapse, the VAFs of FLT3-ITD were much higher (>95% and 8.1%) in the morphologic CR state. CONCLUSIONS The type of FLT3-ITD mutation is important in prognosis, and the dup + ins type of FLT3-ITD can be an indicator of poor prognosis. In addition, the FLT3-ITD mutation status may unexpectedly not match the morphologic examination results after gilteritinib treatment.
Collapse
Affiliation(s)
- Mosae Koo
- Department of Laboratory Medicine, Chungnam National University College of Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Ik-Chan Song
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University College of Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Jimyung Kim
- Department of Laboratory Medicine, Chungnam National University College of Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Gye Cheol Kwon
- Department of Laboratory Medicine, Chungnam National University College of Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Seon Young Kim
- Department of Laboratory Medicine, Chungnam National University College of Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
- Cancer Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| |
Collapse
|
10
|
De Cicco M, Lagreca I, Basso S, Barozzi P, Muscianisi S, Bianco A, Riva G, Di Vincenzo S, Pulvirenti C, Sapuppo D, Siciliano M, Rosti V, Candoni A, Zecca M, Forghieri F, Luppi M, Comoli P. Preclinical Validation of an Advanced Therapy Medicinal Product Based on Cytotoxic T Lymphocytes Specific for Mutated Nucleophosmin (NPM1 mut) for the Treatment of NPM1 mut-Acute Myeloid Leukemia. Cancers (Basel) 2023; 15:2731. [PMID: 37345068 DOI: 10.3390/cancers15102731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
Acute myeloid leukemia (AML) with nucleophosmin (NPM1) genetic mutations is the most common subtype in adult patients. Refractory or relapsed disease in unfit patients or after allogeneic hematopoietic stem cell transplantation (allo-HSCT) has a poor prognosis. NPM1-mutated protein, stably expressed on tumor cells but not on normal tissues, may serve as an ideal target for NPM1-mutated AML immunotherapy. The study aim was to investigate the feasibility of producing mutated-NPM1-specific cytotoxic T cells (CTLs) suitable for somatic cell therapy to prevent or treat hematologic relapse in patients with NPM1-mutated AML. T cells were expanded or primed from patient or donor peripheral blood mononuclear cells by NPM1-mutated protein-derived peptides, and tested for leukemia antigen-targeted cytotoxic activity, cytokine production and hematopoietic precursor inhibitory effect. We found that mutated-NPM1-specific CTLs, displaying specific cytokine production and high-level cytotoxicity against patients' leukemia blasts, and limited inhibitory activity in clonogenic assays, could be obtained from both patients and donors. The polyfunctional mutated-NPM1-specific CTLs included both CD8+ and CD4+ T cells endowed with strong lytic capacity. Our results suggest that mutated-NPM1-targeted CTLs may be a useful therapeutic option to control low-tumor burden relapse following conventional chemotherapy in older NPM1-mutated AML patients or eradicate persistent MRD after HSCT.
Collapse
Affiliation(s)
- Marica De Cicco
- SSD Cell Factory e Center for Advanced Therapies, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Ivana Lagreca
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, 41124 Modena, Italy
| | - Sabrina Basso
- SSD Cell Factory e Center for Advanced Therapies, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Patrizia Barozzi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, 41124 Modena, Italy
| | - Stella Muscianisi
- SSD Cell Factory e Center for Advanced Therapies, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- SC Pediatric Hematology/Oncology, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Alba Bianco
- SSD Cell Factory e Center for Advanced Therapies, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- SC Pediatric Hematology/Oncology, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Giovanni Riva
- Department of Laboratory Medicine and Pathology, Unità Sanitaria Locale, 41126 Modena, Italy
| | - Sara Di Vincenzo
- SSD Cell Factory e Center for Advanced Therapies, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Chiara Pulvirenti
- SSD Cell Factory e Center for Advanced Therapies, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- SC Pediatric Hematology/Oncology, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Davide Sapuppo
- SSD Cell Factory e Center for Advanced Therapies, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- SC Pediatric Hematology/Oncology, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Mariangela Siciliano
- SSD Cell Factory e Center for Advanced Therapies, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, General Medicine 2, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Anna Candoni
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, 41124 Modena, Italy
| | - Marco Zecca
- SC Pediatric Hematology/Oncology, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Fabio Forghieri
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, 41124 Modena, Italy
| | - Mario Luppi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, 41124 Modena, Italy
| | - Patrizia Comoli
- SSD Cell Factory e Center for Advanced Therapies, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, 41124 Modena, Italy
| |
Collapse
|
11
|
Pessoa FMCDP, Machado CB, Barreto IV, Sampaio GF, Oliveira DDS, Ribeiro RM, Lopes GS, de Moraes MEA, de Moraes Filho MO, de Souza LEB, Khayat AS, Moreira-Nunes CA. Association between Immunophenotypic Parameters and Molecular Alterations in Acute Myeloid Leukemia. Biomedicines 2023; 11:1098. [PMID: 37189716 PMCID: PMC10135936 DOI: 10.3390/biomedicines11041098] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 04/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a hematologic malignancy that occurs due to alterations such as genetic mutations, chromosomal translocations, or changes in molecular levels. These alterations can accumulate in stem cells and hematopoietic progenitors, leading to the development of AML, which has a prevalence of 80% of acute leukemias in the adult population. Recurrent cytogenetic abnormalities, in addition to mediating leukemogenesis onset, participate in its evolution and can be used as established diagnostic and prognostic markers. Most of these mutations confer resistance to the traditionally used treatments and, therefore, the aberrant protein products are also considered therapeutic targets. The surface antigens of a cell are characterized through immunophenotyping, which has the ability to identify and differentiate the degrees of maturation and the lineage of the target cell, whether benign or malignant. With this, we seek to establish a relationship according to the molecular aberrations and immunophenotypic alterations that cells with AML present.
Collapse
Affiliation(s)
- Flávia Melo Cunha de Pinho Pessoa
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Caio Bezerra Machado
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Igor Valentim Barreto
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Giulia Freire Sampaio
- Unichristus University Center, Faculty of Biomedicine, Fortaleza 60430-275, CE, Brazil
| | | | | | - Germison Silva Lopes
- Department of Hematology, César Cals General Hospital, Fortaleza 60015-152, CE, Brazil
| | - Maria Elisabete Amaral de Moraes
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Manoel Odorico de Moraes Filho
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Lucas Eduardo Botelho de Souza
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo 14040-900, SP, Brazil
| | - André Salim Khayat
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil
| | - Caroline Aquino Moreira-Nunes
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
- Unichristus University Center, Faculty of Biomedicine, Fortaleza 60430-275, CE, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil
| |
Collapse
|
12
|
Measurable residual disease in adult acute myeloid leukaemia: evaluation of a multidimensional 'radar' flow cytometric plot analysis method. Pathology 2023; 55:383-390. [PMID: 36725446 DOI: 10.1016/j.pathol.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/11/2022] [Accepted: 10/09/2022] [Indexed: 01/01/2023]
Abstract
Measurable residual disease (MRD) monitoring in acute myeloid leukaemia (AML) is becoming increasingly important and is predominantly performed by multiparameter flow cytometry (MFC) or quantitative polymerase chain reactions (RT-qPCR). We investigated the use of multidimensional plots (MD-MFC) for AML MRD monitoring in an adult cohort. AML MRD was determined using a novel MD-MFC method for 115 MRD samples. Results were correlated with traditional two-dimensional MFC (2D-MFC) and molecular methods. Using the standard cut-off of 0.1% CD45+ cells, concordance was 99/115 (p=0.332). Eighty-four of 115 were concordant using a very low reporting limit of 0.01% (p=0.216). MRD <0.1% by either method was present in 40 of 115 samples. Fifteen of 40 were MD-MFC positive and 2D-MFC negative. Of these two of 15 had a molecular MRD marker and both were positive. Molecular MRD markers were available in 36 of 115 cases. Twenty-one of 36 (58%) were concordant with MD-MFC. Eight of 36 had detectable molecular MRD only and eight of 36 had positive MD-MFC only. There was no correlation between either the MFC method and the molecular results. In summary, there is good correlation between MD- and 2D-MFC-MRD and no correlation between the MFC and molecular methods.
Collapse
|
13
|
Zhang YW, Su L, Tan YH, Lin H, Liu XL, Liu QJ, Sun JN, Zhang M, Du YZ, Song F, Han W, Gao SJ. Measurable residual disease detected by flow cytometry independently predicts prognoses of NPM1-mutated acute myeloid leukemia. Ann Hematol 2023; 102:337-347. [PMID: 36378304 DOI: 10.1007/s00277-022-05033-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia (AML) with NPM1 mutation is a distinct genetic entity with favorable outcomes. Nevertheless, emerging evidence suggests that NPM1-mutated AML is still a highly heterogeneous disorder. In this study, 266 patients with AML with NPM1 mutations were retrospectively analyzed to evaluate the associations between variant allele frequency (VAF) of NPM1 mutations, co-mutated genes, measurable residual disease (MRD), and patient outcomes. Multiparameter flow cytometry (MFC) and real-time quantitative polymerase chain reaction (RT-PCR) were used for monitoring MRD. Ultimately, 106 patients were included in the long-term follow-up period. Patients with high NPM1 VAF (≥ 42.43%) had poorer 2-year relapse-free survival (RFS) (55.7% vs. 70.2%, P = 0.017) and overall survival (OS) (63.7% vs. 82.0%, P = 0.027) than those with low VAF. DNMT3A mutations negatively influenced the outcomes of patients with NPM1 mutations. Patients with high DNMT3A VAF or NPM1/DNMT3A/FLT3-ITD triple mutations had shorter RFS and significantly lower OS than that in controls. After two cycles of chemotherapy, patients with positive MFC MRD results had lower RFS (MRD+ vs. MRD-:44.9% vs. 67.6%, P = 0.007) and OS (61.5% vs. 76.6%, P = 0.011) than those without positive MFC MRD results. In multivariate analysis, high NPM1 VAF (hazard ratio [HR] = 2.045; P = 0.034) and positive MRD after two cycles of chemotherapy (HR = 3.289; P = 0.003) were independent risk factors for RFS; MRD positivity after two cycles of chemotherapy (HR = 3.293; P = 0.008) independently predicted the OS of the patients. These results indicate that VAF of both NPM1 gene itself or certain co-occurring gene pre-treatment and MRD post-treatment are potential markers for restratifying the prognoses of patients AML having NPM1 mutations.
Collapse
Affiliation(s)
- Yun-Wei Zhang
- Hematology Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Long Su
- Hematology Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Ye-Hui Tan
- Hematology Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Hai Lin
- Hematology Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Xiao-Liang Liu
- Hematology Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Qiu-Ju Liu
- Hematology Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Jing-Nan Sun
- Hematology Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Ming Zhang
- Hematology Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Ya-Zhe Du
- Hematology Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Fei Song
- Hematology Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Wei Han
- Hematology Department, First Hospital of Jilin University, Changchun, 130021, China
| | - Su-Jun Gao
- Hematology Department, First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
14
|
Digital PCR as a New Method for Minimal Residual Disease Monitoring and Treatment Free Remission Management in Chronic Myeloid Leukemia Patients: Is It Reliable? HEMATO 2022. [DOI: 10.3390/hemato4010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The effective and sensitive monitoring of Minimal Residual Disease or Measurable Residual Disease (MRD) is a very important aspect in the management of patients affected by hematologic malignancies. The recent availability of new technologies has opened to the improvement of MRD monitoring. It is particularly relevant in patients affected by Chronic Myeloid Leukemia (CML). MRD monitoring is key in the management of CML patients thanks to the efficacy of TKIs therapy. Moreover, the policies of TKIs discontinuation aimed at treatment free remission are strongly based on the good selection of patients eligible for stopping TKIs therapy. The recently described application of digital PCR in CML patients monitoring seems to improve the accuracy and precision in the identification of optimal responders. The present review reports an overview on the application of digital PCR in the monitoring of MRD in CML and its impact on TKIs discontinuation trials and, consequently, on TFR success.
Collapse
|
15
|
Wang Y, Quesada AE, Zuo Z, Medeiros LJ, Yin CC, Li S, Xu J, Borthakur G, Li Y, Yang C, Abaza Y, Gao J, Lu X, You MJ, Zhang Y, Lin P. The Impact of Mutation of Myelodysplasia-Related Genes in De Novo Acute Myeloid Leukemia Carrying NPM1 Mutation. Cancers (Basel) 2022; 15:cancers15010198. [PMID: 36612194 PMCID: PMC9818485 DOI: 10.3390/cancers15010198] [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: 10/04/2022] [Revised: 12/21/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Background: The impact of gene mutations typically associated with myelodysplastic syndrome (MDS) in acute myeloid leukemia (AML) with NPM1 mutation is unclear. Methods: Using a cohort of 107 patients with NPM1-mutated AML treated with risk-adapted therapy, we compared survival outcomes of patients without MDS-related gene mutations (group A) with those carrying concurrent FLT3-ITD (group B) or with MDS-related gene mutations (group C). Minimal measurable disease (MMD) status assessed by multiparameter flow cytometry (MFC), polymerase chain reaction (PCR), and/or next-generation sequencing (NGS) were reviewed. Results: Among the 69 patients treated intensively, group C showed significantly inferior progression-free survival (PFS, p < 0.0001) but not overall survival (OS, p = 0.055) compared to group A. Though groups A and C had a similar MMD rate, group C patients had a higher relapse rate (p = 0.016). Relapse correlated with MMD status at the end of cycle 2 induction (p = 0.023). Survival of group C patients was similar to that of group B. Conclusion: MDS-related gene mutations are associated with an inferior survival in NPM1-mutated AML.
Collapse
Affiliation(s)
- Yi Wang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andres E. Quesada
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhuang Zuo
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - L. Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - C. Cameron Yin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shaoying Li
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Xu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yisheng Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chao Yang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yasmin Abaza
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Juehua Gao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xinyan Lu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - M. James You
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yizhuo Zhang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Correspondence: (Y.Z.); (P.L.); Tel.: +86-18622221239 (Y.Z.); +1-(713)-794-1746 (P.L.); Fax: +86-022-23340123 (Y.Z.); +1-(713)-563-2977 (P.L.)
| | - Pei Lin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (Y.Z.); (P.L.); Tel.: +86-18622221239 (Y.Z.); +1-(713)-794-1746 (P.L.); Fax: +86-022-23340123 (Y.Z.); +1-(713)-563-2977 (P.L.)
| |
Collapse
|
16
|
Ganzel C, Sun Z, Baslan T, Zhang Y, Gönen M, Abdel-Wahab OI, Racevskis J, Garrett-Bakelman F, Lowe SW, Fernandez HF, Ketterling R, Luger SM, Litzow M, Lazarus HM, Rowe JM, Tallman MS, Levine RL, Paietta E. Measurable residual disease by flow cytometry in acute myeloid leukemia is prognostic, independent of genomic profiling. Leuk Res 2022; 123:106971. [PMID: 36332294 PMCID: PMC9789386 DOI: 10.1016/j.leukres.2022.106971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 10/04/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022]
Abstract
Measurable residual disease (MRD) assessment provides a potent indicator of the efficacy of anti-leukemic therapy. It is unknown, however, whether integrating MRD with molecular profiling better identifies patients at risk of relapse. To investigate the clinical relevance of MRD in relation to a molecular-based prognostic schema, we measured MRD by flow cytometry in 189 AML patients enrolled in ECOG-ACRIN E1900 trial (NCT00049517) in morphologic complete remission (CR) (28.8 % of the original cohort) representing 44.4 % of CR patients. MRD positivity was defined as ≥ 0.1 % of leukemic bone marrow cells. Risk classification was based on standard cytogenetics, fluorescence-in-situ-hybridization, somatic gene analysis, and sparse whole genome sequencing for copy number ascertainment. At 84.6 months median follow-up of patients still alive at the time of analysis (range 47.0-120 months), multivariate analysis demonstrated that MRD status at CR (p = 0.001) and integrated molecular risk (p = 0.0004) independently predicted overall survival (OS). Among risk classes, MRD status significantly affected OS only in the favorable risk group (p = 0.002). Expression of CD25 (α-chain of the interleukin-2 receptor) by leukemic myeloblasts at diagnosis negatively affected OS independent of post-treatment MRD levels. These data suggest that integrating MRD with genetic profiling and pre-treatment CD25 expression may improve prognostication in AML.
Collapse
Affiliation(s)
- Chezi Ganzel
- Hematology Department, Shaare Zedek Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| | - Zhuoxin Sun
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Timour Baslan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yanming Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gönen
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar I Abdel-Wahab
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Janis Racevskis
- Department of Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Francine Garrett-Bakelman
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Departments of Medicine and Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, USA; University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Hugo F Fernandez
- Malignant Hematology and Cellular Therapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Rhett Ketterling
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Selina M Luger
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark Litzow
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Jacob M Rowe
- Hematology Department, Shaare Zedek Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Martin S Tallman
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
17
|
Deng X, Zhang M, Zhou J, Xiao M. Next-generation sequencing for MRD monitoring in B-lineage malignancies: from bench to bedside. Exp Hematol Oncol 2022; 11:50. [PMID: 36057673 PMCID: PMC9440501 DOI: 10.1186/s40164-022-00300-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/21/2022] [Indexed: 12/02/2022] Open
Abstract
Minimal residual disease (MRD) is considered the strongest relevant predictor of prognosis and an effective decision-making factor during the treatment of hematological malignancies. Remarkable breakthroughs brought about by new strategies, such as epigenetic therapy and chimeric antigen receptor-T (CAR-T) therapy, have led to considerably deeper responses in patients than ever, which presents difficulties with the widely applied gold-standard techniques of MRD monitoring. Urgent demands for novel approaches that are ultrasensitive and provide sufficient information have put a spotlight on high-throughput technologies. Recently, advances in methodology, represented by next-generation sequencing (NGS)-based clonality assays, have proven robust and suggestive in numerous high-quality studies and have been recommended by some international expert groups as disease-monitoring modalities. This review demonstrates the applicability of NGS-based clonality assessment for MRD monitoring of B-cell malignancies by summarizing the oncogenesis of neoplasms and the corresponding status of immunoglobulin (IG) rearrangements. Furthermore, we focused on the performance of NGS-based assays compared with conventional approaches and the interpretation of results, revealing directions for improvement and prospects in clinical practice.
Collapse
Affiliation(s)
- Xinyue Deng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China
| | - Meilan Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China.
| |
Collapse
|
18
|
Galimberti S, Balducci S, Guerrini F, Del Re M, Cacciola R. Digital Droplet PCR in Hematologic Malignancies: A New Useful Molecular Tool. Diagnostics (Basel) 2022; 12:1305. [PMID: 35741115 PMCID: PMC9221914 DOI: 10.3390/diagnostics12061305] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/21/2022] [Accepted: 05/22/2022] [Indexed: 01/27/2023] Open
Abstract
Digital droplet PCR (ddPCR) is a recent version of quantitative PCR (QT-PCR), useful for measuring gene expression, doing clonality assays and detecting hot spot mutations. In respect of QT-PCR, ddPCR is more sensitive, does not need any reference curve and can quantify one quarter of samples already defined as "positive but not quantifiable". In the IgH and TCR clonality assessment, ddPCR recapitulates the allele-specific oligonucleotide PCR (ASO-PCR), being not adapt for detecting clonal evolution, that, on the contrary, does not represent a pitfall for the next generation sequencing (NGS) technique. Differently from NGS, ddPCR is not able to sequence the whole gene, but it is useful, cheaper, and less time-consuming when hot spot mutations are the targets, such as occurs with IDH1, IDH2, NPM1 in acute leukemias or T315I mutation in Philadelphia-positive leukemias or JAK2 in chronic myeloproliferative neoplasms. Further versions of ddPCR, that combine different primers/probes fluorescences and concentrations, allow measuring up to four targets in the same PCR reaction, sparing material, time, and money. ddPCR is also useful for quantitating BCR-ABL1 fusion gene, WT1 expression, donor chimerism, and minimal residual disease, so helping physicians to realize that "patient-tailored therapy" that is the aim of the modern hematology.
Collapse
Affiliation(s)
- Sara Galimberti
- Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, 56126 Pisa, Italy; (S.G.); (S.B.); (F.G.); (M.D.R.)
| | - Serena Balducci
- Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, 56126 Pisa, Italy; (S.G.); (S.B.); (F.G.); (M.D.R.)
| | - Francesca Guerrini
- Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, 56126 Pisa, Italy; (S.G.); (S.B.); (F.G.); (M.D.R.)
| | - Marzia Del Re
- Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, 56126 Pisa, Italy; (S.G.); (S.B.); (F.G.); (M.D.R.)
| | - Rossella Cacciola
- Department of Clinical and Experimental Medicine, Section of Hemostasis, University of Catania, 95123 Catania, Italy
| |
Collapse
|
19
|
NPM1-mutation-based measurable residual disease assessment after completion of two courses of post-remission therapy is a valuable clinical predictor of the prognosis of acute myeloid leukemia. Int J Hematol 2022; 116:199-214. [DOI: 10.1007/s12185-022-03328-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 10/18/2022]
|
20
|
Increasing Role of Targeted Immunotherapies in the Treatment of AML. Int J Mol Sci 2022; 23:ijms23063304. [PMID: 35328721 PMCID: PMC8953556 DOI: 10.3390/ijms23063304] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 12/11/2022] Open
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults. The standard of care in medically and physically fit patients is intensive induction therapy. The majority of these intensively treated patients achieve a complete remission. However, a high number of these patients will experience relapse. In patients older than 60 years, the results are even worse. Therefore, new therapeutic approaches are desperately needed. One promising approach in high-risk leukemia to prevent relapse is the induction of the immune system simultaneously or after reduction of the initial tumor burden. Different immunotherapeutic approaches such as allogenic stem cell transplantation or donor lymphocyte infusions are already standard therapies, but other options for AML treatment are in the pipeline. Moreover, the therapeutic landscape in AML is rapidly changing, and in the last years, a number of immunogenic targets structures eligible for specific therapy, risk assessment or evaluation of disease course were determined. For example, leukemia-associated antigens (LAA) showed to be critical as biomarkers of disease state and survival, as well as markers of minimal residual disease (MRD). Yet many mechanisms and properties are still insufficiently understood, which also represents a great potential for this form of therapy. Therefore, targeted therapy as immunotherapy could turn into an efficient tool to clear residual disease, improve the outcome of AML patients and reduce the relapse risk. In this review, established but also emerging immunotherapeutic approaches for AML patients will be discussed.
Collapse
|
21
|
Voso MT, Ferrara F, Galimberti S, Rambaldi A, Venditti A. Diagnostic Workup of Acute Myeloid Leukemia: What Is Really Necessary? An Italian Survey. Front Oncol 2022; 12:828072. [PMID: 35251997 PMCID: PMC8893956 DOI: 10.3389/fonc.2022.828072] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/26/2022] [Indexed: 11/23/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with a wide variety of clinical presentations, morphological features, and immunophenotypes. The diagnostic approaches to AML that are adopted in Italy have been explored using an online Delphi-based process to expand the global discussion on mandatory tests for the correct diagnosis and, consequently, for optimal management of AML in clinical practice. The final results of the panel of Italian hematologists involved in this work highlight the importance of genetic evaluation for classification and risk stratification and firmly establish that karyotyping, fluorescence in situ hybridization in cases with non-evaluable karyotype, and molecular tests must be performed in every case of AML, regardless of age. Obtaining clinically relevant genetic data at diagnosis is the basis for the success of patient-tailored therapy. The Italian specialists also confirm the role of multidisciplinary diagnostics for AML, now mandatory and expected to become more important in the future context of “precision” medicine.
Collapse
Affiliation(s)
- Maria Teresa Voso
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
- *Correspondence: Maria Teresa Voso,
| | | | - Sara Galimberti
- Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Alessandro Rambaldi
- Department of Oncology-Hematology, University of Milan, Milan, Italy
- Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Adriano Venditti
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| |
Collapse
|
22
|
Revealing the Mysteries of Acute Myeloid Leukemia: From Quantitative PCR through Next-Generation Sequencing and Systemic Metabolomic Profiling. J Clin Med 2022; 11:jcm11030483. [PMID: 35159934 PMCID: PMC8836582 DOI: 10.3390/jcm11030483] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 12/13/2022] Open
Abstract
The efforts made in the last decade regarding the molecular landscape of acute myeloid leukemia (AML) have created the possibility of obtaining patients’ personalized treatment. Indeed, the improvement of accurate diagnosis and precise assessment of minimal residual disease (MRD) increased the number of new markers suitable for novel and targeted therapies. This progress was obtained thanks to the development of molecular techniques starting with real-time quantitative PCR (Rt-qPCR) passing through digital droplet PCR (ddPCR) and next-generation sequencing (NGS) up to the new attractive metabolomic approach. The objective of this surge in technological advances is a better delineation of AML clonal heterogeneity, monitoring patients without disease-specific mutation and designing customized post-remission strategies based on MRD assessment. In this context, metabolomics, which pertains to overall small molecules profiling, emerged as relevant access for risk stratification and targeted therapies improvement. In this review, we performed a detailed overview of the most popular modern methods used in hematological laboratories, pointing out their vital importance for MRD monitoring in order to improve overall survival, early detection of possible relapses and treatment efficacy.
Collapse
|
23
|
The Role of Nucleophosmin 1 ( NPM1) Mutation in the Diagnosis and Management of Myeloid Neoplasms. LIFE (BASEL, SWITZERLAND) 2022; 12:life12010109. [PMID: 35054502 PMCID: PMC8780493 DOI: 10.3390/life12010109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 11/17/2022]
Abstract
Nucleophosmin (NPM1) is a multifunctional protein with both proliferative and growth-suppressive roles in the cell. In humans, NPM1 is involved in tumorigenesis via chromosomal translocations, deletions, or mutation. Acute myeloid leukemia (AML) with mutated NPM1, a distinct diagnostic entity by the current WHO Classification of myeloid neoplasm, represents the most common diagnostic subtype in AML and is associated with a favorable prognosis. The persistence of NPM1 mutation in AML at relapse makes this mutation an ideal target for minimal measurable disease (MRD) detection. The clinical implication of this is far-reaching because NPM1-mutated AML is currently classified as being of standard risk, with the best treatment strategy (transplantation versus chemotherapy) yet undefined. Myeloid neoplasms with NPM1 mutations and <20% blasts are characterized by an aggressive clinical course and a rapid progression to AML. The pathological classification of these cases remains controversial. Future studies will determine whether NPM1 gene mutation may be sufficient for diagnosing NPM1-mutated AML independent of the blast count. This review aims to summarize the role of NPM1 in normal cells and in human cancer and discusses its current role in clinical management of AML and related myeloid neoplasms.
Collapse
|
24
|
Cenfra N, Lapietra G, Perrone S, Voso MT, Divona M, Mecarocci S, La Barbera EO, Cimino G. Azacitidine to Consolidate and Deepen the Therapeutic Response Achieved by Intensive Induction Treatment in a Young Patient Affected by NPM1mut-AML Who Has Become Ineligible for High-Dose Consolidation. Chemotherapy 2022; 67:24-28. [PMID: 35021172 DOI: 10.1159/000520205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/13/2021] [Indexed: 11/19/2022]
Abstract
Acute myeloid leukemia (AML) is the most common leukemia in adults. In spite of the most recent discoveries about the molecular landscape of this disease, the treatment of elderly and unfit young patients continues to be a great challenge. The hypomethylating agents (HMA) still represent an effective therapeutic option for these categories, especially for the low-risk subgroups. We report the case of a young patient with NPM1mut-AML who underwent a first cycle of intensive induction treatment, achieving a complete remission, but suffered from a serious life-threatening neurologic toxicity. Due to the ineligibility to further lines of intensive chemotherapy, we decided to consolidate the response with azacitidine, administered according to the regular schedule. The minimal residual disease (MRD), monitored through the NPM1 mutation at diagnosis, progressively decreased and became undetectable after 36 cycles of hypomethylating therapy. After 1 year from discontinuation of azacitidine, MRD remains undetectable. Therefore, HMA might still represent a feasible and effective option for patients with low-risk AML, especially when the standard chemotherapy is not indicated, or as maintenance therapy in nontransplantable patients.
Collapse
Affiliation(s)
- Natalia Cenfra
- Hematology Polo Universitario Pontino, S.M. Goretti Hospital, Latina, Italy
| | - Gianfranco Lapietra
- Department of Translational and Precision Medicine University "La Sapienza", Rome, Italy
| | - Salvatore Perrone
- Hematology Polo Universitario Pontino, S.M. Goretti Hospital, Latina, Italy
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | | | - Sergio Mecarocci
- Hematology Polo Universitario Pontino, S.M. Goretti Hospital, Latina, Italy
| | | | - Giuseppe Cimino
- Hematology Polo Universitario Pontino, S.M. Goretti Hospital, Latina, Italy.,Department of Translational and Precision Medicine University "La Sapienza", Rome, Italy
| |
Collapse
|
25
|
Zhou X, Cai Y, Yang J, Tong Y, Qiu H, Huang C, Zhou K, Xu X, Niu J, Xia X, Zhang Y, Shen C, Wei Y, Song X, Wan L. Lower Absolute Lymphocyte Count Before Conditioning Predicts High Relapse Risk in Patients After Haploidentical Peripheral Blood Stem Cell Transplantation With Low Dose Anti-Thymocyte Globulin/Post-Transplant Cyclophosphamide for GvHD Prophylaxis. Cell Transplant 2022; 31:9636897221079739. [PMID: 35225024 PMCID: PMC8894976 DOI: 10.1177/09636897221079739] [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] [Indexed: 11/15/2022] Open
Abstract
Standard anti-thymocyte globulin (ATG) weight-based dosing often resulted in highly variable ATG exposure, which had profound effects on relapse and survival, especially in recipients with relatively low absolute lymphocyte count (ALC) before conditioning. Data regarding rabbit ATG pharmacokinetics and pharmacodynamics in the setting of HLA-haploidentical peripheral blood stem cell transplantation (haplo-PBSCT) is lacking. We conducted a retrospective study on 90 consecutive patients who underwent haplo-PBSCT with low dose rabbit ATG (5 mg/kg) plus low dose post-transplant cyclophosphamide (50 mg/kg) based regimen for graft-versus-host disease (GvHD) prophylaxis. We compared serum concentration of ATG and post-transplant results between patients with ALC<500/μl and ALC≥500/μl before conditioning. Patients with ALC<500/μl had higher ATG concentrations, delayed immune reconstitution, lower incidence of grade II-IV acute GvHD (0 vs. 19.42%, P = 0.043), higher risk of Epstein-Barr virus infection within 100 days post-transplant (47.78% vs. 22.22%, P = 0.020) and 1-year relapse rate (33.33% vs.11.59%, P = 0.041), and lower 1-year overall survival (OS) (52.38% vs.79.71%, P = 0.004), 1-year relapse free survival (RFS) (47.62% vs. 75.36% for RFS, P = 0.014), and 1-year GvHD free relapse-free survival (GRFS) (42.89% vs. 65.22%, P = 0.043). ALC<500/μl before conditioning was a significant poor risk factor for relapse, OS, RFS, and GRFS.
Collapse
Affiliation(s)
- Xiao Zhou
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Yu Cai
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Jun Yang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Yin Tong
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Huiying Qiu
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Chongmei Huang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Kun Zhou
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Xiaowei Xu
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Jiahua Niu
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Xinxin Xia
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Ying Zhang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Chang Shen
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Yu Wei
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Xianmin Song
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| | - Liping Wan
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee (STCSM), Shanghai, China
| |
Collapse
|
26
|
Xu J, Wu W, Wu C, Mao Y, Qi X, Guo L, Lu R, Xie S, Lou J, Zhang Y, Ding Y, Guo Z, Zhang L, Liang N, Chen P, Zhang C, Tao M, Yu Z, Geng H, Xu M, Shi M, Wang L, Guo W, Zhao J, Li J, Shi L, Zhang Y, Qin Z, Chen J, Liu J, Ren J, Yang Z, Pan X, Lv Z, Dong H, Zhang J, Ou J, Li Z, Kaji K, Wang Y, Wang J, Wang Z. A large-scale, multicentered trial evaluating the sensitivity and specificity of digital PCR versus ARMS-PCR for detecting ctDNA-based EGFR p.T790M in non-small-cell lung cancer patients. Transl Lung Cancer Res 2021; 10:3888-3901. [PMID: 34858779 PMCID: PMC8577974 DOI: 10.21037/tlcr-21-564] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/16/2021] [Indexed: 12/27/2022]
Abstract
Background Developing liquid biopsy technology with higher sensitivity and specificity especially for low-frequency mutations remains crucial. This study demonstrated superior performance of the newly developed digital PCR (dPCR) kit for ctDNA-based EGFR p.T790M detection in metastatic non-small-cell lung cancer (NSCLC) against ARMS-PCR. Methods This large-scale, multi-centered diagnostic study recruited 1,045 patients including 1,029 patients diagnosed with advanced NSCLC and 16 patients with specific samples between April 1st 2018 and November 30th 2019. EGFR p.T790M in plasma samples from mNSCLC patients were tested using dPCR with ADx-ARMS PCR and Cobas®EGFR Mutation Test V2 as comparator assays to confirm cut-off value for dPCR and evaluate its performance against ARMS-PCR-based assays. Efficacy was evaluated for patients with EGFR p.T790M detected by dPCR or ARMS-PCR, who underwent Osimertinib treatment. Results The sensitivity, specificity, and concordance of dPCR against ADx-ARMS PCR was 98.15%, 88.66% and 90.16%, respectively for 1,026 plasma samples. Additional 9.26% patients were detected positive by dPCR. The majority of those samples had a mutation allele frequency between 0.1% and 1%. In 45 paired tissue and plasma samples, the sensitivity improved from 30.77% to 53.85% by dPCR with the specificity over 90%. The response of Osimertinib in 74 EGFR p.T790M-positive patients detected by dPCR, including 26 determined as negative by ARMS-PCR, were evaluated to have an ORR of 44.59% and a DCR of 90.54%. Conclusions dPCR is a sensitive and accurate tool for ctDNA-based EGFR p.T790M detection due to its significantly improved sensitivity without compromising specificity, and dPCR is equivalent to ARMS-PCR as a companion diagnostic tool while benefiting more patients under Osimertinib treatment. Trial Registration Chinese Clinical Trial Registry identifier: ChiCTR2100043147.
Collapse
Affiliation(s)
- Jiachen Xu
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yong Mao
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiaowei Qi
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Lin Guo
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Renquan Lu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Shuhong Xie
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Jiatao Lou
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Zhang
- Department of Respiratory Medicine, Nanjing Chest Hospital, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yiyan Ding
- Department of Respiratory Medicine, Nanjing Chest Hospital, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Zijian Guo
- Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Li Zhang
- Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Naixin Liang
- Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Peng Chen
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Clinical Research Center for Cancer, Tianjin, China
| | - Cuicui Zhang
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Clinical Research Center for Cancer, Tianjin, China
| | - Min Tao
- Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhengyuan Yu
- Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hua Geng
- Department of Pathology, Tianjin Chest Hospital, Tianjin, China
| | - Meilin Xu
- Department of Pathology, Tianjin Chest Hospital, Tianjin, China
| | - Meiqi Shi
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University/Jiangsu Cancer Hospital/Jiangsu Institute of Cancer Research, Nanjing, China
| | - Li Wang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University/Jiangsu Cancer Hospital/Jiangsu Institute of Cancer Research, Nanjing, China
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun Zhao
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Jianjie Li
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Lixia Shi
- Tianjin Haihe Hospital Tianjin Institute of Respiratory Diseases, Tianjin, China
| | - Yan Zhang
- Tianjin Haihe Hospital Tianjin Institute of Respiratory Diseases, Tianjin, China
| | - Zhonghua Qin
- Tianjin Haihe Hospital Tianjin Institute of Respiratory Diseases, Tianjin, China
| | - Jun Chen
- Tianjin Medical University General Hospital, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinghao Liu
- Tianjin Medical University General Hospital, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Ren
- Tianjin Medical University General Hospital, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhenlin Yang
- Department of Thoracic surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Pan
- Questgenomics, Nanjing, China
| | | | | | | | | | | | | | - Yan Wang
- Questgenomics, Nanjing, China.,Gnomegen, San Diego, CA, USA
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
27
|
Austin AE, Byrne M. Detecting and preventing post-hematopoietic cell transplant relapse in AML. Curr Opin Hematol 2021; 28:380-388. [PMID: 34534984 DOI: 10.1097/moh.0000000000000686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Relapsed disease is the primary cause of mortality for acute myeloid leukemia (AML) patients after allogeneic hematopoietic cell transplantation (HCT). This review outlines the most recent advances in the detection and prevention of AML relapse following allogeneic HCT. RECENT FINDINGS Conventional methods for predicting post-HCT relapse rely on the molecular and cytogenetics features present at diagnosis. These methods are slow to reflect a growing understanding of the molecular heterogeneity of AML and impact of new therapies on post-HCT outcomes. The use of measurable residual disease (MRD) techniques, including multiparameter flow cytometry and molecular testing, may improve the prognostic ability of these models and should be incorporated into post-HCT surveillance whenever possible.In the post-HCT setting, FLT3 inhibitor maintenance data indicate that effective therapies can improve post-HCT outcomes. Maintenance data with DNA methyltransferase inhibitor monotherapy is less compelling and outcomes may improve with combinations. Early interventions directed at preemptive management of MRD may further improve post-HCT outcomes. SUMMARY Post-HCT AML relapse prevention has evolved to include more sensitive measures of disease detection and novel therapies that may improve outcomes of poor-risk AML patients. Additional work is needed to maintain this progress.
Collapse
Affiliation(s)
| | - Michael Byrne
- Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
28
|
Aggarwal N, Alston ELJ, Pinkus GS, Weinberg OK. Role of immunohistochemistry and flow cytometry in minimal residual disease detection in NPM1-mutated AML. Int J Lab Hematol 2021; 44:e76-e78. [PMID: 34704364 DOI: 10.1111/ijlh.13722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/23/2021] [Accepted: 09/10/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Nidhi Aggarwal
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Erin L J Alston
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Geraldine S Pinkus
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Olga K Weinberg
- Department of Pathology, UT Southwestern Medical Center BioCenter, Dallas, Texas, USA
| |
Collapse
|
29
|
Bolandi SM, Pakjoo M, Beigi P, Kiani M, Allahgholipour A, Goudarzi N, Khorashad JS, Eiring AM. A Role for the Bone Marrow Microenvironment in Drug Resistance of Acute Myeloid Leukemia. Cells 2021; 10:2833. [PMID: 34831055 PMCID: PMC8616250 DOI: 10.3390/cells10112833] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with a poor prognosis and remarkable resistance to chemotherapeutic agents. Understanding resistance mechanisms against currently available drugs helps to recognize the therapeutic obstacles. Various mechanisms of resistance to chemotherapy or targeted inhibitors have been described for AML cells, including a role for the bone marrow niche in both the initiation and persistence of the disease, and in drug resistance of the leukemic stem cell (LSC) population. The BM niche supports LSC survival through direct and indirect interactions among the stromal cells, hematopoietic stem/progenitor cells, and leukemic cells. Additionally, the BM niche mediates changes in metabolic and signal pathway activation due to the acquisition of new mutations or selection and expansion of a minor clone. This review briefly discusses the role of the BM microenvironment and metabolic pathways in resistance to therapy, as discovered through AML clinical studies or cell line and animal models.
Collapse
Affiliation(s)
- Seyed Mohammadreza Bolandi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Mahdi Pakjoo
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Peyman Beigi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Mohammad Kiani
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Ali Allahgholipour
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Negar Goudarzi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
| | - Jamshid S. Khorashad
- Centre for Haematology, Hammersmith Hospital, Imperial College London, London W12 0HS, UK;
| | - Anna M. Eiring
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| |
Collapse
|
30
|
Lopez A, Patel S, Geyer JT, Racchumi J, Chadburn A, Simonson P, Ouseph MM, Inghirami G, Mencia-Trinchant N, Guzman ML, Gomez-Arteaga A, Lee S, Desai P, Ritchie EK, Roboz GJ, Tam W, Kluk MJ. Comparison of Multiple Clinical Testing Modalities for Assessment of NPM1-Mutant AML. Front Oncol 2021; 11:701318. [PMID: 34527579 PMCID: PMC8435844 DOI: 10.3389/fonc.2021.701318] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/12/2021] [Indexed: 11/13/2022] Open
Abstract
Background NPM1 mutation status can influence prognosis and management in AML. Accordingly, clinical testing (i.e., RT-PCR, NGS and IHC) for mutant NPM1 is increasing in order to detect residual disease in AML, alongside flow cytometry (FC). However, the relationship of the results from RT-PCR to traditional NGS, IHC and FC is not widely known among many practitioners. Herein, we aim to: i) describe the performance of RT-PCR compared to traditional NGS and IHC for the detection of mutant NPM1 in clinical practice, and also compare it to FC, and ii) provide our observations regarding the advantages and disadvantages of each approach in order to inform future clinical testing algorithms. Methods Peripheral blood and bone marrow samples collected for clinical testing at variable time points during patient management were tested by quantitative, real-time, RT-PCR and results were compared to findings from a Myeloid NGS panel, mutant NPM1 IHC and FC. Results RT-PCR showed superior sensitivity compared to NGS, IHC and FC with the main challenge of NGS, IHC and FC being the ability to identify a low disease burden (<0.5% NCN by RT-PCR). Nevertheless, the positive predictive value of NGS, IHC and FC were each ≥ 80% indicating that positive results by those assays are typically associated with RT-PCR positivity. IHC, unlike bulk methods (RT-PCR, NGS and FC), is able provide information regarding cellular/architectural context of disease in biopsies. FC did not identify any NPM1-mutated residual disease not already detected by RT-PCR, NGS or IHC. Conclusion Overall, our findings demonstrate that RT-PCR shows superior sensitivity compared to a traditional Myeloid NGS, suggesting the need for “deep-sequencing” NGS panels for NGS-based monitoring of residual disease in NPM1-mutant AML. IHC provides complementary cytomorphologic information to RT-PCR. Lastly, FC may not be necessary in the setting of post-therapy follow up for NPM1-mutated AML. Together, these findings can help inform future clinical testing algorithms.
Collapse
Affiliation(s)
- Amanda Lopez
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Sanjay Patel
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Julia T Geyer
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Joelle Racchumi
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Amy Chadburn
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Paul Simonson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Madhu M Ouseph
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Nuria Mencia-Trinchant
- Clinical and Translational Leukemia Program, Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Monica L Guzman
- Clinical and Translational Leukemia Program, Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Alexandra Gomez-Arteaga
- Clinical and Translational Leukemia Program, Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States.,Stem Cell Transplant Program, Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Sangmin Lee
- Clinical and Translational Leukemia Program, Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Pinkal Desai
- Clinical and Translational Leukemia Program, Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Ellen K Ritchie
- Clinical and Translational Leukemia Program, Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Gail J Roboz
- Clinical and Translational Leukemia Program, Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Wayne Tam
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Michael J Kluk
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
31
|
Neoantigen-Specific T-Cell Immune Responses: The Paradigm of NPM1-Mutated Acute Myeloid Leukemia. Int J Mol Sci 2021; 22:ijms22179159. [PMID: 34502069 PMCID: PMC8431540 DOI: 10.3390/ijms22179159] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023] Open
Abstract
The C-terminal aminoacidic sequence from NPM1-mutated protein, absent in normal human tissues, may serve as a leukemia-specific antigen and can be considered an ideal target for NPM1-mutated acute myeloid leukemia (AML) immunotherapy. Different in silico instruments and in vitro/ex vivo immunological platforms have identified the most immunogenic epitopes from NPM1-mutated protein. Spontaneous development of endogenous NPM1-mutated-specific cytotoxic T cells has been observed in patients, potentially contributing to remission maintenance and prolonged survival. Genetically engineered T cells, namely CAR-T or TCR-transduced T cells, directed against NPM1-mutated peptides bound to HLA could prospectively represent a promising therapeutic approach. Although either adoptive or vaccine-based immunotherapies are unlikely to be highly effective in patients with full-blown leukemia, these strategies, potentially in combination with immune-checkpoint inhibitors, could be promising in maintaining remission or preemptively eradicating persistent measurable residual disease, mainly in patients ineligible for allogeneic hematopoietic stem cell transplant (HSCT). Alternatively, neoantigen-specific donor lymphocyte infusion derived from healthy donors and targeting NPM1-mutated protein to selectively elicit graft-versus-leukemia effect may represent an attractive option in subjects experiencing post-HSCT relapse. Future studies are warranted to further investigate dynamics of NPM1-mutated-specific immunity and explore whether novel individualized immunotherapies may have potential clinical utility in NPM1-mutated AML patients.
Collapse
|
32
|
Hou C, Zhou L, Yang M, Jiang S, Shen H, Zhu M, Chen J, Miao M, Xu Y, Wu D. The Prognostic Value of Early Detection of Minimal Residual Disease as Defined by Flow Cytometry and Gene Mutation Clearance for Myelodysplastic Syndrome Patients After Myeloablative Allogeneic Hematopoietic Stem-Cell Transplantation. Front Oncol 2021; 11:700234. [PMID: 34422653 PMCID: PMC8374104 DOI: 10.3389/fonc.2021.700234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/14/2021] [Indexed: 01/17/2023] Open
Abstract
High relapse incidence remains a major problem for myelodysplastic syndrome (MDS) patients who have received an allogeneic hematopoietic stem-cell transplantation (allo-HSCT). We retrospectively analyzed the correlations between clinical outcomes and minimal residual disease (MRD) by using mutations (MUT) and flow cytometry (FCM) analysis of 115 MDS patients with allo-HSCT. We divided 115 MDS patients into four groups based on molecular genetics and FCM MRD results at day 30 post-HSCT. There were significant differences in the 2-year progression-free survival (PFS) between the FCMhigh MUTpos and FCMlow MUTneg groups (20% vs 79%, P < 0.001). In addition, by univariate analysis, we found that an IPSS-R score ≥4 pre-HSCT (HR, 5.061; P=0.007), DNMT3A mutations (HR, 2.291; P=0.052), TP53 mutations (HR, 3.946; P=0.011), and poor and very poor revised International Prognostic Scoring System (IPSS-R) cytogenetic risk (HR, 4.906; P < 0.001) were poor risk factors for PFS. In multivariate analysis, we found that an IPSS-R score ≥ 4 pre-HSCT (HR, 4.488; P=0.015), DNMT3A mutations (HR, 2.385; P=0.049), positive FCM MRD combined with persistence gene mutations at day 30 (HR, 5.198; P=0.013) were independent risk factors for disease progression. In conclusion, our data indicated that monitoring MRD by FCM combined with gene mutation clearance at day 30 could help in the prediction of disease progression for MDS patients after transplantation.
Collapse
Affiliation(s)
- Chang Hou
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Lili Zhou
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Menglu Yang
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shuhui Jiang
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongjie Shen
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mingqing Zhu
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jia Chen
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Miao Miao
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Xu
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Depei Wu
- Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
33
|
Holoubek A, Strachotová D, Otevřelová P, Röselová P, Heřman P, Brodská B. AML-Related NPM Mutations Drive p53 Delocalization into the Cytoplasm with Possible Impact on p53-Dependent Stress Response. Cancers (Basel) 2021; 13:cancers13133266. [PMID: 34209894 PMCID: PMC8269334 DOI: 10.3390/cancers13133266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Nucleophosmin (NPM) is one of the most abundant nucleolar proteins and its mutations frequently occur in acute myeloid leukemia (AML). The mutations cause aberrant cytoplasmic localization of mutated protein (NPMmut) and often mediate dislocation of NPM interaction partners. Tumor suppressor p53 is known to interact with NPM in response to genotoxic stress and its cytoplasmic localization is an unfavorable prognostic factor in cancers. This study aims to characterize the NPM-p53 interaction and to elucidate the effect of the NPM mutations on p53 localization and expression in live cells. In addition, the cellular dynamics of NPMmut and p53 after treatment with nuclear export inhibitor Selinexor is described and the mechanism of the Selinexor action proposed. Our results contribute to a better understanding of the oncogenic potential of NPM mutations. Abstract Nucleophosmin (NPM) interaction with tumor suppressor p53 is a part of a complex interaction network and considerably affects cellular stress response. The impact of NPM1 mutations on its interaction with p53 has not been investigated yet, although consequences of NPMmut-induced p53 export to the cytoplasm are important for understanding the oncogenic potential of these mutations. We investigated p53-NPM interaction in live HEK-293T cells by FLIM-FRET and in cell lysates by immunoprecipitation. eGFP lifetime-photoconversion was used to follow redistribution dynamics of NPMmut and p53 in Selinexor-treated cells. We confirmed the p53-NPMwt interaction in intact cells and newly documented that this interaction is not compromised by the NPM mutation causing displacement of p53 to the cytoplasm. Moreover, the interaction was not abolished for non-oligomerizing NPM variants with truncated oligomerization domain, suggesting that oligomerization is not essential for interaction of NPM forms with p53. Inhibition of the nuclear exporter XPO1 by Selinexor caused expected nuclear relocalization of both NPMmut and p53. However, significantly different return rates of these proteins indicate nontrivial mechanism of p53 and NPMmut cellular trafficking. The altered p53 regulation in cells expressing NPMmut offers improved understanding to help investigational strategies targeting these mutations.
Collapse
Affiliation(s)
- Aleš Holoubek
- Department of Proteomics, Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague, Czech Republic; (A.H.); (P.O.); (P.R.)
| | - Dita Strachotová
- Institute of Physics, Faculty of Mathematics and Physics, Charles University, Ke Karlovu 5, 121 16 Prague, Czech Republic;
| | - Petra Otevřelová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague, Czech Republic; (A.H.); (P.O.); (P.R.)
| | - Pavla Röselová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague, Czech Republic; (A.H.); (P.O.); (P.R.)
| | - Petr Heřman
- Institute of Physics, Faculty of Mathematics and Physics, Charles University, Ke Karlovu 5, 121 16 Prague, Czech Republic;
- Correspondence: (P.H.); (B.B.); Tel.: +420-951-551-461 (P.H.); +420-221-977-354 (B.B.)
| | - Barbora Brodská
- Department of Proteomics, Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague, Czech Republic; (A.H.); (P.O.); (P.R.)
- Correspondence: (P.H.); (B.B.); Tel.: +420-951-551-461 (P.H.); +420-221-977-354 (B.B.)
| |
Collapse
|
34
|
Pettersson L, Johansson Alm S, Almstedt A, Chen Y, Orrsjö G, Shah-Barkhordar G, Zhou L, Kotarsky H, Vidovic K, Asp J, Lazarevic V, Saal LH, Fogelstrand L, Ehinger M. Comparison of RNA- and DNA-based methods for measurable residual disease analysis in NPM1-mutated acute myeloid leukemia. Int J Lab Hematol 2021; 43:664-674. [PMID: 34053184 DOI: 10.1111/ijlh.13608] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 05/02/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Reverse transcriptase quantitative PCR (RT-qPCR) is considered the method of choice for measurable residual disease (MRD) assessment in NPM1-mutated acute myeloid leukemia (AML). MRD can also be determined with DNA-based methods offering certain advantages. We here compared the DNA-based methods quantitative PCR (qPCR), droplet digital PCR (ddPCR), and targeted deep sequencing (deep seq) with RT-qPCR. METHODS Of 110 follow-up samples from 30 patients with NPM1-mutated AML were analyzed by qPCR, ddPCR, deep seq, and RT-qPCR. To select DNA MRD cutoffs for bone marrow, we performed receiver operating characteristic analyses for each DNA method using prognostically relevant RT-qPCR cutoffs. RESULTS The DNA-based methods showed strong intermethod correlation, but were less sensitive than RT-qPCR. A bone marrow cutoff at 0.1% leukemic DNA for qPCR or 0.05% variant allele frequency for ddPCR and deep seq offered optimal sensitivity and specificity with respect to 3 log10 reduction of NPM1 transcripts and/or 2% mutant NPM1/ABL. With these cutoffs, MRD results agreed in 95% (191/201) of the analyses. Although more sensitive, RT-qPCR failed to detect leukemic signals in 10% of samples with detectable leukemic DNA. CONCLUSION DNA-based MRD techniques may complement RT-qPCR for assessment of residual leukemia. DNA-based methods offer high positive and negative predictive values with respect to residual leukemic NPM1 transcripts at levels of importance for response to treatment. However, moving to DNA-based MRD methods will miss a proportion of patients with residual leukemic RNA, but on the other hand some MRD samples with detectable leukemic DNA can be devoid of measurable leukemic RNA.
Collapse
Affiliation(s)
- Louise Pettersson
- Department of Clinical Sciences, Division of Pathology, Lund University, Skane University Hospital, Lund, Sweden.,Department of Pathology, Halland Hospital Halmstad, Region Halland, Halmstad, Sweden
| | - Sofie Johansson Alm
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Alvar Almstedt
- SciLife Clinical Genomics Gothenburg, Gothenburg, Sweden
| | - Yilun Chen
- Department of Clinical Sciences, Division of Oncology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Gustav Orrsjö
- Section for Hematology and Coagulation, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Giti Shah-Barkhordar
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Li Zhou
- Klinisk Patologi, Region Laboratories, Region Skåne, Lund, Sweden
| | - Heike Kotarsky
- Klinisk Patologi, Region Laboratories, Region Skåne, Lund, Sweden
| | - Karina Vidovic
- Department of Clinical Sciences, Division of Pathology, Lund University, Skane University Hospital, Lund, Sweden
| | - Julia Asp
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Vladimir Lazarevic
- Department of Hematology, Oncology and Radiation Physics, Lund University, Skane University Hospital, Lund, Sweden
| | - Lao H Saal
- Department of Clinical Sciences, Division of Oncology, Faculty of Medicine, Lund University, Lund, Sweden.,Lund University Cancer Center, Medicon Village, Lund, Sweden
| | - Linda Fogelstrand
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mats Ehinger
- Department of Clinical Sciences, Division of Pathology, Lund University, Skane University Hospital, Lund, Sweden.,Klinisk Patologi, Region Laboratories, Region Skåne, Lund, Sweden
| |
Collapse
|
35
|
Liu FJ, Cheng WY, Lin XJ, Wang SY, Jiang TY, Ma TT, Zhu YM, Shen Y. Measurable Residual Disease Detected by Multiparameter Flow Cytometry and Sequencing Improves Prediction of Relapse and Survival in Acute Myeloid Leukemia. Front Oncol 2021; 11:677833. [PMID: 34094982 PMCID: PMC8173083 DOI: 10.3389/fonc.2021.677833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
The clinically ideal time point and optimal approach for the assessment of measurable residual disease (MRD) in patients with acute myeloid leukemia (AML) are still inconclusive. We investigated the clinical value of multiparameter flow cytometry-based MRD (MFC MRD) after induction (n = 492) and two cycles of consolidation (n = 421). The latter time point was proved as a superior indicator with independent prognostic significance for both relapse-free survival (RFS, HR = 3.635, 95% CI: 2.433-5.431, P <0.001) and overall survival (OS: HR = 3.511, 95% CI: 2.191-5.626, P <0.001). Furthermore, several representative molecular MRD markers were compared with the MFC MRD. Both approaches can establish prognostic value in patients with NPM1 mutations, and FLT3, C-KIT, or N-RAS mutations involved in kinase-related signaling pathways, while the combination of both techniques further refined the risk stratification. The detection of RUNX1-RUNX1T1 fusion transcripts achieved a considerable net reclassification improvement in predicting the prognosis. Conversely, for patients with biallelic CEBPA or DNMT3A mutations, only the MFC method was recommended due to the poor prognostic discriminability in tracking mutant transcripts. In conclusion, this study demonstrated that the MFC MRD after two consolidation cycles independently predicted clinical outcomes, and the integration of MFC and molecular MRD should depend on different types of AML-related genetic lesions.
Collapse
Affiliation(s)
- Fu-Jia Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Yan Cheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Jing Lin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shi-Yang Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tian-Yi Jiang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting-Ting Ma
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong-Mei Zhu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Shen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Skou AS, Juul-Dam KL, Ommen HB, Hasle H. Peripheral blood molecular measurable residual disease is sufficient to identify patients with acute myeloid leukaemia with imminent clinical relapse. Br J Haematol 2021; 195:310-327. [PMID: 33851435 DOI: 10.1111/bjh.17449] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 01/03/2023]
Abstract
Longitudinal molecular measurable residual disease (MRD) sampling after completion of therapy serves as a refined tool for identification of imminent relapse of acute myeloid leukaemia (AML) among patients in long-term haematological complete remission. Tracking of increasing quantitative polymerase chain reaction MRD before cytomorphological reappearance of blasts may instigate individual management decisions and has paved the way for development of pre-emptive treatment strategies to substantially delay or perhaps even revert leukaemic regrowth. Traditionally, MRD monitoring is performed using repeated bone marrow aspirations, albeit the current European LeukemiaNet MRD recommendations acknowledge the use of peripheral blood as an alternative source for MRD assessment. Persistent MRD positivity in the bone marrow despite continuous morphological remission is frequent in both core binding factor leukaemias and nucleophosmin 1-mutated AML. In contrast, monthly assessment of MRD in peripheral blood superiorly separates patients with imminent haematological relapse from long-term remitters and may allow pre-emptive therapy of AML relapse.
Collapse
Affiliation(s)
- Anne-Sofie Skou
- Department of Paediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Hans B Ommen
- Department of Haematology, Aarhus University Hospital, Aarhus, Denmark
| | - Henrik Hasle
- Department of Paediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
37
|
Loghavi S, DiNardo CD, Furudate K, Takahashi K, Tanaka T, Short NJ, Kadia T, Konopleva M, Kanagal-Shamanna R, Farnoud NR, Pierce S, Khoury JD, Jorgensen JL, Patel KP, Daver N, Yilmaz M, Medeiros LJ, Kantarjian H, Ravandi F, Wang SA. Flow cytometric immunophenotypic alterations of persistent clonal haematopoiesis in remission bone marrows of patients with NPM1-mutated acute myeloid leukaemia. Br J Haematol 2021; 192:1054-1063. [PMID: 33618432 DOI: 10.1111/bjh.17347] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/22/2020] [Accepted: 12/14/2020] [Indexed: 01/07/2023]
Abstract
Clonal haematopoiesis (CH) in patients with acute myeloid leukaemia (AML) may persist beyond attaining complete remission. From a consecutive cohort of 67 patients with nucleophosmin 1-mutated (NPM1mut ) AML, we identified 50 who achieved NPM1mut clearance and had parallel multicolour flow cytometry (MFC) and next generation sequencing (NGS). In total, 13 (26%) cleared all mutations, 37 (74%) had persistent CH frequently involving DNA methyltransferase 3α (DNMT3A,70%), tet methylcytosine dioxygenase 2 (TET2, 27%), isocitrate dehydrogenase 2 (IDH2, 19%) and IDH1 (11%). A small number (<1%) of aberrant CD34+ myeloblasts, but immunophenotypically different from original AML blasts [herein referred to as a pre-leukaemic (PL) phenotype], was detected in 17 (49%) patients with CH, but not in any patients with complete clearance of all mutations (P = 0·0037). A PL phenotype was associated with higher mutation burden (P = 0·005). Persistent IDH2 and serine and arginine-rich splicing factor 2 (SRSF2) mutations were exclusively observed in PL+ CH+ cases (P = 0·016). Persistent dysplasia was seen exclusively in cases with a PL+ phenotype (29% vs. none; P = 0·04). The PL+ phenotype did not correlate with age, intensity of induction therapy or relapse-free survival. Post-remission CH in the setting of NPM1mut clearance is common and may result in immunophenotypic changes in myeloid progenitors. It is important to not misinterpret these cells as AML measurable residual disease (MRD).
Collapse
Affiliation(s)
- Sanam Loghavi
- The Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Ken Furudate
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA.,Department of Oral and Maxillofacial Surgery, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Koichi Takahashi
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Tomoyuki Tanaka
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas J Short
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Tapan Kadia
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Noushin R Farnoud
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sherry Pierce
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph D Khoury
- The Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey L Jorgensen
- The Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur P Patel
- The Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Musa Yilmaz
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - L Jeffrey Medeiros
- The Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop Kantarjian
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- The Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Sa A Wang
- The Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
38
|
Carbonell D, Suárez-González J, Chicano M, Andrés-Zayas C, Díez-Díez M, Rodríguez-Macías G, Muñiz P, Kwon M, Anguita J, Díez-Martín JL, Buño I, Martínez-Laperche C. Genetic biomarkers identify a subgroup of high-risk patients within low-risk NPM1-mutated acute myeloid leukemia. Leuk Lymphoma 2020; 62:1178-1186. [PMID: 33372822 DOI: 10.1080/10428194.2020.1863400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Although acute myeloid leukemia (AML) with NPM1mut/FLT3-ITDneg is a low-risk entity, its relapse rate remains high. Out of 333 AML patients, 27 were NPM1mut, and were analyzed in greater detail in order to find associations between clinical and molecular features and cumulative incidence of relapse. Next-generation sequencing (NGS) was performed on diagnosis and remission samples using two capture-based panels. The presence of the FLT3D835 variant at diagnosis and a qPCR value of NPM1mut ≥0.1% after induction chemotherapy were associated with an increased probability of relapse, especially if both conditions are present together. By contrast, patients in which the main clone found at diagnosis harbored NPM1 variant had a lower risk of relapse. Nineteen of the 85 variants found at diagnosis were detected by NGS in remission. AML Subgroup with NPM1mut/FLT3-ITDneg is a heterogeneous entity, which can be further risk-stratified based on molecular biomarkers.
Collapse
Affiliation(s)
- Diego Carbonell
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain.,Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| | - Julia Suárez-González
- Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain.,Genomics Unit, Gregorio Marañón General University Hospital, IiSGM, Madrid, Spain
| | - María Chicano
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain.,Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| | - Cristina Andrés-Zayas
- Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain.,Genomics Unit, Gregorio Marañón General University Hospital, IiSGM, Madrid, Spain
| | - Miriam Díez-Díez
- Genomics Unit, Gregorio Marañón General University Hospital, IiSGM, Madrid, Spain
| | | | - Paula Muñiz
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain.,Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| | - Mi Kwon
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain.,Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| | - Javier Anguita
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain.,Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| | - José Luis Díez-Martín
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain.,Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain.,Department of Medicine, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Ismael Buño
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain.,Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain.,Genomics Unit, Gregorio Marañón General University Hospital, IiSGM, Madrid, Spain.,Department of Cell Biology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Carolina Martínez-Laperche
- Department of Hematology, Gregorio Marañón General University Hospital, Madrid, Spain.,Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| |
Collapse
|
39
|
Allogeneic transplant can abrogate the risk of relapse in the patients of first remission acute myeloid leukemia with detectable measurable residual disease by next-generation sequencing. Bone Marrow Transplant 2020; 56:1159-1170. [PMID: 33279940 DOI: 10.1038/s41409-020-01165-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 10/16/2020] [Accepted: 11/19/2020] [Indexed: 11/09/2022]
Abstract
In patients with acute myeloid leukemia (AML) consolidation treatment options are between allogeneic hematopoietic stem cell transplantation (HCT) and chemotherapy, based on disease risk at the time of initial presentation and age. Measurable residual disease (MRD) following induction chemotherapy could be incorporated as a useful parameter for treatment decisions. The present study evaluated treatment outcomes according to the next-generation sequencing (NGS)-based MRD status and the type of consolidation therapy in patients with normal karyotype (NK)-AML. By sequencing 278 paired samples collected at diagnosis and first remission (CR1), we identified 361 mutations in 124 patients at diagnosis and tracked these at CR1. After excluding mutations associated with age-related clonal hematopoiesis, 82 mutations in 50 of the 124 patients (40.3%) were detected at CR1. Survival benefit was observed in favor of allogeneic HCT over chemotherapy consolidation in the MRDpos subgroup with respect to overall survival (HR 0.294, p = 0.003), relapse-free survival (HR 0.376, p = 0.015) and cumulative incidence of relapse (HR 0.279, p = 0.004) in multivariate analysis, but not in the MRDneg subgroup. In summary, these data support allogeneic HCT in NK-AML patients with detectable MRD by NGS in CR1. Randomized clinical trials will be required to confirm this observation.
Collapse
|
40
|
NPM1-Mutated Myeloid Neoplasms with <20% Blasts: A Really Distinct Clinico-Pathologic Entity? Int J Mol Sci 2020; 21:ijms21238975. [PMID: 33255988 PMCID: PMC7730332 DOI: 10.3390/ijms21238975] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022] Open
Abstract
Nucleophosmin (NPM1) gene mutations rarely occur in non-acute myeloid neoplasms (MNs) with <20% blasts. Among nearly 10,000 patients investigated so far, molecular analyses documented NPM1 mutations in around 2% of myelodysplastic syndrome (MDS) cases, mainly belonging to MDS with excess of blasts, and 3% of myelodysplastic/myeloproliferative neoplasm (MDS/MPN) cases, prevalently classified as chronic myelomonocytic leukemia. These uncommon malignancies are associated with an aggressive clinical course, relatively rapid progression to overt acute myeloid leukemia (AML) and poor survival outcomes, raising controversies on their classification as distinct clinico-pathologic entities. Furthermore, fit patients with NPM1-mutated MNs with <20% blasts could benefit most from upfront intensive chemotherapy for AML rather than from moderate intensity MDS-directed therapies, although no firm conclusion can currently be drawn on best therapeutic approaches, due to the limited available data, obtained from small and mainly retrospective series. Caution is also suggested in definitely diagnosing NPM1-mutated MNs with blast count <20%, since NPM1-mutated AML cases frequently present dysplastic features and multilineage bone marrow cells showing abnormal cytoplasmic NPM1 protein delocalization by immunohistochemical staining, therefore belonging to NPM1-mutated clone regardless of blast morphology. Further prospective studies are warranted to definitely assess whether NPM1 mutations may become sufficient to diagnose AML, irrespective of blast percentage.
Collapse
|
41
|
Panuzzo C, Signorino E, Calabrese C, Ali MS, Petiti J, Bracco E, Cilloni D. Landscape of Tumor Suppressor Mutations in Acute Myeloid Leukemia. J Clin Med 2020; 9:jcm9030802. [PMID: 32188030 PMCID: PMC7141302 DOI: 10.3390/jcm9030802] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 12/11/2022] Open
Abstract
Acute myeloid leukemia is mainly characterized by a complex and dynamic genomic instability. Next-generation sequencing has significantly improved the ability of diagnostic research to molecularly characterize and stratify patients. This detailed outcome allowed the discovery of new therapeutic targets and predictive biomarkers, which led to develop novel compounds (e.g., IDH 1 and 2 inhibitors), nowadays commonly used for the treatment of adult relapsed or refractory AML. In this review we summarize the most relevant mutations affecting tumor suppressor genes that contribute to the onset and progression of AML pathology. Epigenetic modifications (TET2, IDH1 and IDH2, DNMT3A, ASXL1, WT1, EZH2), DNA repair dysregulation (TP53, NPM1), cell cycle inhibition and deficiency in differentiation (NPM1, CEBPA, TP53 and GATA2) as a consequence of somatic mutations come out as key elements in acute myeloid leukemia and may contribute to relapse and resistance to therapies. Moreover, spliceosomal machinery mutations identified in the last years, even if in a small cohort of acute myeloid leukemia patients, suggested a new opportunity to exploit therapeutically. Targeting these cellular markers will be the main challenge in the near future in an attempt to eradicate leukemia stem cells.
Collapse
Affiliation(s)
- Cristina Panuzzo
- Department of Clinical and Biological Sciences, University of Turin, 10124 Turin, Italy; (C.P.); (E.S.); (C.C.); (M.S.A.); (J.P.)
| | - Elisabetta Signorino
- Department of Clinical and Biological Sciences, University of Turin, 10124 Turin, Italy; (C.P.); (E.S.); (C.C.); (M.S.A.); (J.P.)
| | - Chiara Calabrese
- Department of Clinical and Biological Sciences, University of Turin, 10124 Turin, Italy; (C.P.); (E.S.); (C.C.); (M.S.A.); (J.P.)
| | - Muhammad Shahzad Ali
- Department of Clinical and Biological Sciences, University of Turin, 10124 Turin, Italy; (C.P.); (E.S.); (C.C.); (M.S.A.); (J.P.)
| | - Jessica Petiti
- Department of Clinical and Biological Sciences, University of Turin, 10124 Turin, Italy; (C.P.); (E.S.); (C.C.); (M.S.A.); (J.P.)
| | - Enrico Bracco
- Department of Oncology, University of Turin, 10124 Turin, Italy;
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, 10124 Turin, Italy; (C.P.); (E.S.); (C.C.); (M.S.A.); (J.P.)
- Correspondence: ; Tel.: +39-011-9026610; Fax: +39-011-9038636
| |
Collapse
|
42
|
Gao MG, Ruan GR, Chang YJ, Liu YR, Qin YZ, Jiang Q, Jiang H, Huang XJ, Zhao XS. The predictive value of minimal residual disease when facing the inconsistent results detected by real-time quantitative PCR and flow cytometry in NPM1-mutated acute myeloid leukemia. Ann Hematol 2019; 99:73-82. [PMID: 31768677 DOI: 10.1007/s00277-019-03861-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 11/19/2019] [Indexed: 11/29/2022]
Abstract
For acute myeloid leukemia (AML) with nucleophosmin 1 mutation (NPM1m), multiparameter flow cytometry (FCM) and real-time quantitative polymerase chain reaction (RQ-PCR) are used to monitor minimal residual disease (MRD). However, the results of the two methods are sometimes inconsistent. This study was designed to analyze how to address the discordant results of FCM and RQ-PCR in AML patients undergoing chemotherapy, especially when positive FCM (FCM+) and negative NPM1m (NPM1m-) results are detected in the same sample. Our study included 93 AML patients with NPM1m positive (NPM1m+) who received chemotherapy but did not undergo hematopoietic stem cell transplantation. We monitored NPM1m and leukemia-associated immunophenotypes (LAIPs) by RQ-PCR and FCM, respectively, to assess MRD after each chemotherapy course. After each course of chemotherapy, all patients were classified into four groups based on the results of FCM and RQ-PCR: both negative (group 1, FCM-NPM1m-), single positive (group 2, FCM-NPM1m+; group 3, FCM+NPM1m-), or both positive (group 4, FCM+NPM1m+). The results showed that there was not a significant difference in the 2-year cumulative incidence of relapse (CIR) after each course of chemotherapy between group 2 and group 3. Furthermore, patients in groups 2 and 3 had a lower 2-year CIR than those in group 4 and a significantly higher 2-year CIR than those in group 1 after the first two courses. The patients in group 4 had a significantly higher 2-year CIR than those in group 1 after the first two courses. These results suggested that in the MRD monitoring process of AML patients, when the results of FCM and RQ-PCR are inconsistent (especially when FCM is positive and NPM1m is negative), these single-positive results still have predictive significance for relapse.
Collapse
Affiliation(s)
- Meng-Ge Gao
- National Clinical Research Center for Hematologic Disease, Peking University Institute of Hematology, Peking University People's Hospital, No. 11 South Street of Xizhimen, Xicheng District, Beijing, 100044, China
| | - Guo-Rui Ruan
- National Clinical Research Center for Hematologic Disease, Peking University Institute of Hematology, Peking University People's Hospital, No. 11 South Street of Xizhimen, Xicheng District, Beijing, 100044, China
| | - Ying-Jun Chang
- National Clinical Research Center for Hematologic Disease, Peking University Institute of Hematology, Peking University People's Hospital, No. 11 South Street of Xizhimen, Xicheng District, Beijing, 100044, China.,Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yan-Rong Liu
- National Clinical Research Center for Hematologic Disease, Peking University Institute of Hematology, Peking University People's Hospital, No. 11 South Street of Xizhimen, Xicheng District, Beijing, 100044, China
| | - Ya-Zhen Qin
- National Clinical Research Center for Hematologic Disease, Peking University Institute of Hematology, Peking University People's Hospital, No. 11 South Street of Xizhimen, Xicheng District, Beijing, 100044, China
| | - Qian Jiang
- National Clinical Research Center for Hematologic Disease, Peking University Institute of Hematology, Peking University People's Hospital, No. 11 South Street of Xizhimen, Xicheng District, Beijing, 100044, China
| | - Hao Jiang
- National Clinical Research Center for Hematologic Disease, Peking University Institute of Hematology, Peking University People's Hospital, No. 11 South Street of Xizhimen, Xicheng District, Beijing, 100044, China
| | - Xiao-Jun Huang
- National Clinical Research Center for Hematologic Disease, Peking University Institute of Hematology, Peking University People's Hospital, No. 11 South Street of Xizhimen, Xicheng District, Beijing, 100044, China.,Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiao-Su Zhao
- National Clinical Research Center for Hematologic Disease, Peking University Institute of Hematology, Peking University People's Hospital, No. 11 South Street of Xizhimen, Xicheng District, Beijing, 100044, China. .,Collaborative Innovation Center of Hematology, Peking University, Beijing, China.
| |
Collapse
|
43
|
Maurillo L, Bassan R, Cascavilla N, Ciceri F. Quality of Response in Acute Myeloid Leukemia: The Role of Minimal Residual Disease. Cancers (Basel) 2019; 11:cancers11101417. [PMID: 31548502 PMCID: PMC6826465 DOI: 10.3390/cancers11101417] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 12/22/2022] Open
Abstract
In the acute myeloid leukemia (AML) setting, research has extensively investigated the existence and relevance of molecular biomarkers, in order to better tailor therapy with newly developed agents and hence improve outcomes and/or save the patient from poorly effective therapies. In particular, in patients with AML, residual disease after therapy does reflect the sum of the contributions of all factors associated with diagnosis and post-diagnosis resistance. The evaluation of minimal/measurable residual disease (MRD) can be considered as a key tool to guide patient’s management and a promising endpoint for clinical trials. In this narrative review, we discuss MRD evaluation as biomarker for tailored therapy in AML patients; we briefly report current evidence on the use of MRD in clinical practice, and comment on the potential ability of MRD in the assessment of the efficacy of new molecules.
Collapse
Affiliation(s)
- Luca Maurillo
- Hematology Unit, Department of Biomedicine and Prevention, Fondazione Policlinico Tor Vergata, Hospital, 00133 Rome, Italy.
| | - Renato Bassan
- Hematology Unit, dell'Angelo Hospital and Santissimi Giovanni and Paolo Hospital, 30174 Mestre and Venice, Italy.
| | - Nicola Cascavilla
- Hematology Unit, Onco-hematology Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (FG), Italy.
| | - Fabio Ciceri
- Hematology and Bone Marrow Transplantation Unit, IRCCS S. Raffaele Scientific Institution, 20132 Milan, Italy.
| |
Collapse
|
44
|
Patel SS, Pinkus GS, Ritterhouse LL, Segal JP, Dal Cin P, Restrepo T, Harris MH, Stone RM, Hasserjian RP, Weinberg OK. High NPM1 mutant allele burden at diagnosis correlates with minimal residual disease at first remission in de novo acute myeloid leukemia. Am J Hematol 2019; 94:921-928. [PMID: 31148220 DOI: 10.1002/ajh.25544] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 05/21/2019] [Accepted: 05/28/2019] [Indexed: 12/28/2022]
Abstract
Acute myeloid leukemia (AML) with mutated NPM1 is a newly recognized separate entity in the revised 2016 WHO classification, and is associated with a favorable prognosis. While previous studies have evaluated NPM1 in a binary fashion, we recently demonstrated a significant independent negative prognostic effect of high NPM1 mutant allele burden (VAF) at diagnosis in a cohort of de novo AML patients. Although the importance of minimal residual disease (MRD) monitoring in NPM1-mutated AML has been well characterized, the potential relationship between diagnostic allele burden and MRD is unknown. We retrospectively evaluated for MRD at first remission (CR1). We used either next-generation sequencing (NGS) [n = 71], and/or immunohistochemistry (IHC) for mutant NPM1 (NPM1c) [n = 60], in a subset of patients from our recently examined cohort. We identified a statistically significant positive correlation between the VAF at diagnosis, and at CR1 (Spearman r = 0.4, P = .006), and enrichment for MRD in high diagnostic VAF patients (P = .05), as previously defined. IHC-positivity also correlated significantly with a higher median diagnostic NPM1 VAF (0.42 vs 0.39, P = .02), and with the VAF at CR1 (Spearman r = 0.7, P = .003). In multivariable analyses, both high diagnostic VAF (P = .003) and MRD (P = .02) were independent predictors of shorter event-free survival (EFS). Our findings suggest a relationship between the NPM1 mutant allele burden at diagnosis, and the presence of MRD at first remission. Our findings support IHC as a potentially useful adjunctive tool for disease monitoring.
Collapse
Affiliation(s)
- Sanjay S. Patel
- Department of PathologyBrigham and Women's Hospital Boston Massachusetts
| | | | | | - Jeremy P. Segal
- Division of Genomic and Molecular PathologyUniversity of Chicago Chicago Illinois
| | - Paola Dal Cin
- Department of PathologyBrigham and Women's Hospital Boston Massachusetts
| | - Tamara Restrepo
- Department of PathologyBoston Children's Hospital Boston Massachusetts
| | - Marian H. Harris
- Department of PathologyBoston Children's Hospital Boston Massachusetts
| | - Richard M. Stone
- Department of Medical OncologyDana‐Farber Cancer Institute Boston Massachusetts
| | | | - Olga K. Weinberg
- Department of PathologyBrigham and Women's Hospital Boston Massachusetts
- Department of PathologyBoston Children's Hospital Boston Massachusetts
| |
Collapse
|
45
|
Voso MT, Ottone T, Lavorgna S, Venditti A, Maurillo L, Lo-Coco F, Buccisano F. MRD in AML: The Role of New Techniques. Front Oncol 2019; 9:655. [PMID: 31396481 PMCID: PMC6664148 DOI: 10.3389/fonc.2019.00655] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 07/04/2019] [Indexed: 11/17/2022] Open
Abstract
In the context of precision medicine, assessment of minimal residual disease (MRD) has been used in acute myeloid leukemia (AML) to direct individual treatment programs, including allogeneic stem cell transplantation in patients at high-risk of relapse. One of the limits of this approach has been in the past the paucity of AML markers suitable for MRD assessment. Recently, the number of biomarkers has increased, due to the identification of highly specific leukemia-associated immunophenotypes by multicolor flow-cytometry, and of rare mutated gene sequences by digital droplet PCR, or next-generation sequencing (NGS). In addition, NGS allowed unraveling of clonal heterogeneity, present in AML at initial diagnosis or developing during treatment, which influences reliability of specific biomarkers, that may be unstable during the disease course. The technological advances have increased the application of MRD-based strategies to a significantly higher number of AML patients, and the information deriving from MRD assessment has been used to design individual post-remission protocols and pre-emptive treatments in patients with sub-clinical relapse. This led to the definition of MRD-negative complete remission as outcome definition in the recently published European Leukemianet MRD guidelines. In this review, we summarized the principles of modern technologies and their clinical applications for MRD detection in AML patients, according to the specific leukemic markers.
Collapse
Affiliation(s)
- Maria Teresa Voso
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
- Santa Lucia Foundation, IRCCS, Neuro-Oncohematology, Rome, Italy
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
- Santa Lucia Foundation, IRCCS, Neuro-Oncohematology, Rome, Italy
| | - Serena Lavorgna
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Adriano Venditti
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Luca Maurillo
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Francesco Lo-Coco
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Francesco Buccisano
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| |
Collapse
|
46
|
Du D, Zhu L, Wang Y, Ye X. [Expression of WT1 gene and its prognostic value in patients with acute myeloid leukemia]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2019; 48:50-57. [PMID: 31102358 PMCID: PMC10412419 DOI: 10.3785/j.issn.1008-9292.2019.02.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/10/2019] [Indexed: 06/09/2023]
Abstract
OBJECTIVE To investigate the expression of Wilms'tumor 1 (WT1) gene in patients with acute myeloid leukemia (AML), and to explore its application in predicting prognosis of AML in patients with wild or mutated nucleophosmin 1(NPM1) and Fms-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD). METHODS One hundred and sixty-seven newly diagnosed AML patients(exclued M3 type) were enrolled in this study. The survival of patients were analyzed with Kaplan-Meier method. The clinical data, laboratory findings and the survival of patients were analyzed and compared between patients with high WT1 expression (high-WT1 group) and those with low WT1 expression (low-WT1 group), as well as among the patients with NPM1 or FLT3-ITD wild type and mutants. RESULTS The overall response rates (ORR) in high-WT1 and low-WT1 groups were 65.9% (83/126) and 95.1% (39/41), respectively (P<0.01). Compared with the low-WT1 group, the high-WT1 group had lower 2-y overall survival (OS) rate (46.1% vs. 75.2%, P<0.05) and 2-y disease free survival (DFS) rate (43.5% vs. 68.5%, P<0.05). After induction chemotherapy, the patients with decreased WT1 gene expression ≥ 1log was associated with higher ORR and 2-y OS rate (all P<0.05), but the advantage of 2-y DFS rate was not shown (P>0.05). In patients with NPM1 wild type, the high-WT1 group had inferior ORR and 2-y OS rate (all P<0.05), while in the patients with FLT3-ITD wild type, the high-WT1 group had inferior ORR, 2-y OS rate and 2-y DFS rate (all P<0.05). In patients with NPM1 or FLT3 -ITD mutations, the WT1 expression had no significantly predicting values in treatment efficacy and survival (all P>0.05). CONCLUSIONS WT1 gene overexpression indicated poor prognosis of AML patients; the patients with decreased WT1 gene expression ≥ 1 log after the first induction therapy show better prognosis than those with<1 log. The WT1 gene expression level at diagnosis can be used as an unfavorable prognostic factor for AML patients with NPM1 or FLT3-ITD wild types.
Collapse
Affiliation(s)
- Dongfen Du
- Department of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Shaoxing Central Blood Station, Shaoxing 312000, Zhejiang Province, China
| | - Lixia Zhu
- Department of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yungui Wang
- Department of Hematology and Institute of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiujin Ye
- Department of Hematology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
47
|
Brodská B, Šašinková M, Kuželová K. Nucleophosmin in leukemia: Consequences of anchor loss. Int J Biochem Cell Biol 2019; 111:52-62. [PMID: 31009764 DOI: 10.1016/j.biocel.2019.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 12/17/2022]
Abstract
Nucleophosmin (NPM), one of the most abundant nucleolar proteins, has crucial functions in ribosome biogenesis, cell cycle control, and DNA-damage repair. In human cells, NPM occurs mainly in oligomers. It functions as a chaperone, undergoes numerous interactions and forms part of many protein complexes. Although NPM role in carcinogenesis is not fully elucidated, a variety of tumor suppressor as well as oncogenic activities were described. NPM is overexpressed, fused with other proteins, or mutated in various tumor types. In the acute myeloid leukemia (AML), characteristic mutations in NPM1 gene, leading to modification of NPM C-terminus, are the most frequent genetic aberration. Although multiple mutation types of NPM are found in AML, they are all characterized by aberrant cytoplasmic localization of the mutated protein. In this review, current knowledge of the structure and function of NPM is presented in relation to its interaction network, in particular to the interaction with other nucleolar proteins and with proteins active in apoptosis. Possible molecular mechanisms of NPM mutation-driven leukemogenesis and NPM therapeutic targeting are discussed. Finally, recent findings concerning the immunogenicity of the mutated NPM and specific immunological features of AML patients with NPM mutation are summarized.
Collapse
Affiliation(s)
- Barbora Brodská
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic
| | - Markéta Šašinková
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic
| | - Kateřina Kuželová
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic.
| |
Collapse
|
48
|
Salehzadeh S, Guerrini F, Pizzano U, Grassi S, Ciabatti E, Iovino L, Buda G, Caracciolo F, Benedetti E, Orciuolo E, Pelosini M, Consani G, Carulli G, Metelli MR, Martini F, Mazziotta F, Mazzantini E, Rossi P, Tavarozzi R, Ricci F, Petrini M, Galimberti S. The assessment of minimal residual disease versus that of somatic mutations for predicting the outcome of acute myeloid leukemia patients. Cancer Cell Int 2019; 19:83. [PMID: 30992690 PMCID: PMC6449954 DOI: 10.1186/s12935-019-0807-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/28/2019] [Indexed: 12/22/2022] Open
Abstract
Background In addition to morphological and cytogenetic features, acute myeloid leukemias are characterized by mutations that can be used for target-therapy; also the minimal/measurable residual disease (MRD) could be an important prognostic factor. The purpose of this retrospective study was to investigate if somatic mutations could represent an additional prognostic value in respect of MRD alone. Method At baseline, 98 patients were tested for NPM1, FLT3, and for WT1 expression; 31 for ASXL1, TET2, IDH1, IDH2, N-RAS, WT1, c-KIT, RUNX1, and DNMT3A. The same genes have been also tested after induction and consolidation. Results Overall, 60.2% of our patients resulted mutated: 24.5% carried mutations of FLT3-ITD, 38.7% of NPM1, 48.4% of c-KIT, 25.8% of N-RAS and 19.3% of IDH2. The probability of achieving a complete response (CR) was higher for younger patients, with low ELN risk score, NPM1-mutated, with low WT1 levels, and without FLT3. The presence of additional mutations represented a poor predictive factor: only 19% of these cases achieved CR in comparison to 43% of subjects without any of it. Concerning survival, it was conditioned by a lower ELN risk score, younger age, reduction > 1 log of the NPM1 mutational burden, disappearance of FLT3 mutations and lower WT1 expression. Regarding the role of the additional mutations, they impaired the outcome of 20% of the already MRD-negative patients. Concerning the possibility of predicting relapse, we observed an increase of the NPM1 mutational burden at the time-point immediately preceding the relapse (about 2 months earlier) in 50% of subjects. Similarly concerning WT1, an increase of its expression anticipated disease recurrence in 64% of cases. Conclusions We demonstrated that additional somatic mutations are able to impair outcome of the already MRD-negative subjects. About MRD, we suggest a prognostic role also for the WT1 expression. Finally, we considered as relevant the assessment of NPM1 quantity clearance instead of the presence/absence of mutations alone. Still remains in doubt the utility in terms of long-term prognosis of a baseline more complex mutational screening; we could hypothesize that it would be useful for those patients where other markers are not available or who reached the MRD negativity. Electronic supplementary material The online version of this article (10.1186/s12935-019-0807-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Serena Salehzadeh
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy.,4University of Rome Tor Vergata, Rome, Italy.,5Ospedale S. Chiara, UO Ematologia, Via Roma, 67, 56126 Pisa, Italy
| | - Francesca Guerrini
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Umberto Pizzano
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Susanna Grassi
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Elena Ciabatti
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Lorenzo Iovino
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Gabriele Buda
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Francesco Caracciolo
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Edoardo Benedetti
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Enrico Orciuolo
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Matteo Pelosini
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Giovanni Consani
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Giovanni Carulli
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | | | - Francesca Martini
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Francesco Mazziotta
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy.,2GeNOMEC School of Doctorate, University of Siena, Siena, Italy
| | - Elisa Mazzantini
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Pietro Rossi
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Rita Tavarozzi
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Federica Ricci
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Mario Petrini
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Sara Galimberti
- 1Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| |
Collapse
|
49
|
Ehinger M, Pettersson L. Measurable residual disease testing for personalized treatment of acute myeloid leukemia. APMIS 2019; 127:337-351. [PMID: 30919505 DOI: 10.1111/apm.12926] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022]
Abstract
This review summarizes - with the practicing hematologist in mind - the methods used to determine measurable residual disease (MRD) in everyday practice with some future perspectives, and the current knowledge about the prognostic impact of MRD on outcome in acute myeloid leukemia (AML), excluding acute promyelocytic leukemia. Possible implications for choice of MRD method, timing of MRD monitoring, and guidance of therapy are discussed in general and in some detail for certain types of leukemia with specific molecular markers to monitor, including core binding factor (CBF)-leukemias and NPM1-mutated leukemias.
Collapse
Affiliation(s)
- Mats Ehinger
- Department of Clinical Sciences, Pathology, Skane University Hospital, Lund University, Lund, Sweden
| | - Louise Pettersson
- Department of Pathology, Halland Hospital Halmstad, Region Halland, Halmstad, Sweden.,Faculty of Medicine, Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
50
|
Madanat YF, Gerds AT. Can allogeneic hematopoietic cell transplant cure therapy-related acute leukemia? Best Pract Res Clin Haematol 2019; 32:104-113. [DOI: 10.1016/j.beha.2019.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 02/18/2019] [Accepted: 02/22/2019] [Indexed: 01/16/2023]
|