1
|
Fokken H, Waclawski J, Kattre N, Kloos A, Müller S, Ettinger M, Kacprowski T, Heuser M, Maetzig T, Schwarzer A. A 19-color single-tube full spectrum flow cytometry assay for the detection of measurable residual disease in acute myeloid leukemia. Cytometry A 2024; 105:181-195. [PMID: 37984809 DOI: 10.1002/cyto.a.24811] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/22/2023]
Abstract
Multiparameter flow cytometry (MFC) has emerged as a standard method for quantifying measurable residual disease (MRD) in acute myeloid leukemia. However, the limited number of available channels on conventional flow cytometers requires the division of a diagnostic sample into several tubes, restricting the number of cells and the complexity of immunophenotypes that can be analyzed. Full spectrum flow cytometers overcome this limitation by enabling the simultaneous use of up to 40 fluorescent markers. Here, we used this approach to develop a good laboratory practice-conform single-tube 19-color MRD detection assay that complies with recommendations of the European LeukemiaNet Flow-MRD Working Party. We based our assay on clinically-validated antibody clones and evaluated its performance on an IVD-certified full spectrum flow cytometer. We measured MRD and normal bone marrow samples and compared the MRD data to a widely used reference MRD-MFC panel generating highly concordant results. Using our newly developed single-tube panel, we established reference values in healthy bone marrow for 28 consensus leukemia-associated immunophenotypes and introduced a semi-automated dimensionality-reduction, clustering and cell type identification approach that aids the unbiased detection of aberrant cells. In summary, we provide a comprehensive full spectrum MRD-MFC workflow with the potential for rapid implementation for routine diagnostics due to reduced cell requirements and ease of data analysis with increased reproducibility in comparison to conventional FlowMRD routines.
Collapse
Affiliation(s)
- Hendrik Fokken
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Julian Waclawski
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Nadine Kattre
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Arnold Kloos
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Sebastian Müller
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics of TU Braunschweig and Hannover Medical School, Braunschweig, Germany
- Braunschweig Integrated Centre for Systems Biology (BRICS), TU Braunschweig, Braunschweig, Germany
| | - Max Ettinger
- Department of Orthopedic Surgery, Hannover Medical School, Hannover, Germany
| | - Tim Kacprowski
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics of TU Braunschweig and Hannover Medical School, Braunschweig, Germany
- Braunschweig Integrated Centre for Systems Biology (BRICS), TU Braunschweig, Braunschweig, Germany
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Tobias Maetzig
- Department of Pediatric Hematology, Hannover Medical School, Hannover, Germany
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Adrian Schwarzer
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- CCC-MV and Department of Internal Medicine C, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
2
|
Singh N, Gupta A, Kumar S, Mawalankar G, Gupta B, Dhole N, Kori R, Singh A. Flow cytometric measurable residual disease in adult acute myeloid leukemia: a preliminary report from Eastern India. J Hematop 2023; 16:17-25. [PMID: 38175369 DOI: 10.1007/s12308-022-00527-2] [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: 10/15/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Presence of measurable residual disease (MRD) in acute myeloid leukemia (AML) is considered to be an independent predictor of relapse and poorer survival outcomes. MRD can be measured by flow cytometric, quantitative PCR, and NGS-based assays at varying sensitivities. There is scant Indian data on different aspects of MFC-MRD in AML including analysis strategies as well as molecular spectrum, clinical correlation, etc. This retrospective observational study included all newly diagnosed patients of acute myeloid leukemia in whom complete baseline diagnostic workup was available including flow cytometry and cytogenetic and molecular studies. Among patients with cytogenetic abnormalities (n = 25), no statistically significant correlation was observed between flow cytometric MRD positivity and presence of ≥ 3 mutations as well as relapsed disease. However, in AML patients with normal karyotype (n = 32), MRD positivity correlated strongly with relapsed status (p = 0.02), although no significant correlation was found with respect to FLT3 mutation, IDH mutation, NPM1 mutation, or complex genotype. Interestingly, 90.5% of MRD-positive patients belonged to ELN (2017) intermediate to high-risk category unlike only 9.5% in the good risk category (p = 0.0002). Median relapse-free survival was 8.5 months with a follow-up range of 3-24 months. On the basis of the observations of the present study, it can be clearly inferred that MRD status affects relapse status in the normal karyotype subgroup and can delineate patients who require stem cell transplantation in addition to molecular signatures.
Collapse
Affiliation(s)
- Neha Singh
- Hematopathology, Tata Memorial Center, Varanasi, India.
| | - Avinash Gupta
- Hematopathology, Tata Memorial Center, Varanasi, India
| | - Sujeet Kumar
- Adult Hematolymphoid Unit, Tata Memorial Center, Varanasi, India
| | | | - Bhumika Gupta
- Hematopathology, Tata Memorial Center, Varanasi, India
| | - Nilesh Dhole
- Hematopathology, Tata Memorial Center, Varanasi, India
| | | | - Anil Singh
- Adult Hematolymphoid Unit, Tata Memorial Center, Varanasi, India
| |
Collapse
|
3
|
Kamel AM, Elsharkawy NM, Kandeel EZ, Hanafi M, Samra M, Osman RA. Leukemia Stem Cell Frequency at Diagnosis Correlates With Measurable/Minimal Residual Disease and Impacts Survival in Adult Acute Myeloid Leukemia. Front Oncol 2022; 12:867684. [PMID: 35530356 PMCID: PMC9069678 DOI: 10.3389/fonc.2022.867684] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/03/2022] [Indexed: 11/17/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogenous disease in which the initiation and maintenance of the malignant clone is blamed on a rare population of leukemia stem cells (LSCs). The persistence of such a malignant population is referred to as measurable/minimal residual disease (MRD). Evaluation of MRD is the gold standard for follow-up of therapy and constitutes an independent prognostic parameter. As LSCs are the main contributor to the persistence of MRD, then MRD should correlate with the bulk of LSCs at the individual case level. MRD is measured at defined time points during therapy. However, LSCs can be evaluated at diagnosis, which ensures the advantage of early prediction of high-risk patients and allows for early therapeutic decisions. Using two simple four-color monoclonal antibody combinations (CD38/CD123/CD34/CD45 and CD90/CD133/CD45/CD33) and the prism function of the Coulter Navios flow cytometer, the frequency of LSC subsets was evaluated in 84 newly diagnosed adult AML patients. For each panel, 16 possible combinations were detected. Our results showed that there was extreme variability in the percentage of the LSC fraction between different cases, as well as at the individual case level. For each LSC subset, the median value was used to divide cases into low and high expressors. LSC subsets that showed an impact on overall survival (OS) and disease-free survival (DFS) included CD123+, CD 123+/CD34-, CD34-/CD38+/CD123+, CD34+/CD38-/CD123+, CD133+, and CD133+/CD33-. On multivariate analysis, only CD123 (p ≤ 0.001, SE = 0.266, HR = 2.8, 95% CI = 1.74.7) and CD133+/CD33- (p = 0.017, SE = 0.263, HR = 1.9, 95% CI = 1.1–3.1) retained their significance for OS. Likewise, only CD34+/CD38-/CD123+ (p ≤ 0.001, HR 2.3, SE: 0.499, 95% CI: 2.4–17.4) and CD133 (p = 0.015, HR 2.3, SE 0.34, 95% CI: 1.2–4.4) retained their statistical significance for DFS. The LSC frequency at diagnosis showed a moderate to strong correlation with MRD status at day 14 and day 28. In conclusion, the level of LSCs at diagnosis correlated with MRD status at day 14 and day 28 in AML patients and had a deleterious impact on OS and DFS. It may be used as an early marker for high-risk patients allowing for early therapeutic decisions.
Collapse
Affiliation(s)
- Azza M Kamel
- Department of Clinical Pathology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Nahla M Elsharkawy
- Department of Clinical Pathology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Eman Z Kandeel
- Department of Clinical Pathology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Marwa Hanafi
- Department of Clinical Pathology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohammed Samra
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Randa A Osman
- Department of Clinical Pathology, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
4
|
Buccisano F, Palmieri R, Piciocchi A, Arena V, Maurillo L, Del Principe MI, Paterno G, Irno-Consalvo MA, Ottone T, Divona M, Conti C, Fraboni D, Lavorgna S, Arcese W, Voso MT, Venditti A. Clinical relevance of an objective flow cytometry approach based on limit of detection and limit of quantification for measurable residual disease assessment in acute myeloid leukemia. A post-hoc analysis of the GIMEMA AML1310 trial. Haematologica 2022; 107:2823-2833. [PMID: 35295076 PMCID: PMC9713557 DOI: 10.3324/haematol.2021.279777] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
Using a multiparametric flow cytometry assay, we assessed the predictive power of a threshold calculated applying the criteria of limit of detection (LOD) and limit of quantitation (LOQ) in adult patients with acute myeloid leukemia. This was a post-hoc analysis of 261 patients enrolled in the GIMEMA AML1310 prospective trial. According to the protocol design, using the predefined measurable residual disease (MRD) threshold of 0.035% bone marrow residual leukemic cells (RLC) calculated on mononuclear cells, 154 (59%) of the 261 patients were negative (MRD <0.035%) and 107 (41%) were positive (MRD ≥0.035%). Using LOD and LOQ, we selected the following categories of patients: (i) LODneg if RLC were below the LOD (74; 28.4%); (ii) LODpos-LOQneg if RLC were between the LOD and LOQ (43; 16.5%); and (iii) LOQpos if RLC were above the LOQ (144; 54.4%). Two-year overall survival of these three categories of patients was 75.4%, 79.8% and 66.4%, respectively (P=0.1197). Given their superimposable outcomes, the LODneg and LODpos-LOQneg categories were combined. Two-year overall survival of LODneg/LODpos-LOQneg patients was 77.0% versus 66.4% of LOQpos individuals (P=0.043). This figure was challenged in univariate analysis (P=0.046, hazard ratio=1.6, 95% confidence interval: 1.01-2.54) which confirmed the independent role of the LOD-LOQ approach in determining overall survival. In the AML1310 protocol, using the threshold of 0.035%, 2-year overall survival of patients with MRD <0.035% and MRD ≥0.035% was 74.5% versus 66.4%, respectively (P=0.3521). In conclusion, the use of the LOD-LOQ method results in more sensitive detection of MRD that, in turn, translates into a more accurate recognition of patients with different outcomes.
Collapse
Affiliation(s)
- Francesco Buccisano
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma,FB and RP contributed equally as co-first authors
| | - Raffaele Palmieri
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma,FB and RP contributed equally as co-first authors
| | | | | | - Luca Maurillo
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | | | | | | | - Tiziana Ottone
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Mariadomenica Divona
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Consuelo Conti
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Daniela Fraboni
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Serena Lavorgna
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - William Arcese
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma,Rome Transplant Network, Rome, Italy
| | - Maria Teresa Voso
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| | - Adriano Venditti
- Ematologia, Dipartimento di Biomedicina e Prevenzione, “Tor Vergata” Università di Roma
| |
Collapse
|
5
|
ELN2017 risk stratification improves outcome prediction when applied to the prospective GIMEMA AML1310 protocol. Blood Adv 2021; 6:2510-2516. [PMID: 34731884 PMCID: PMC9043923 DOI: 10.1182/bloodadvances.2021005717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/05/2021] [Indexed: 11/20/2022] Open
Abstract
The 2017 version of the ELN recommendations, by integrating cytogenetics and mutational status of specific genes, sort out patients with Acute Myeloid Leukemia into 3 prognostically distinct risk categories: favorable (ELN2017-FR), intermediate (ELN2017-IR) and adverse (ELN2017-AR). We performed a post-hoc analysis of the GIMEMA AML1310 trial to investigate the applicability of the ELN2017 risk stratification to our study population. In this trial, after induction and consolidation, patients in complete remission were to receive autologous stem cell transplant (AuSCT) if categorized as favorable-risk or allogeneic stem cell transplant (ASCT) if adverse-risk. Intermediate-risk pts were to receive AuSCT or ASCT based on the post-consolidation levels of Measurable Residual Disease as measured by flow-cytometry. Risk categorization was originally conducted according to NCCN2009 recommendations. Among 500 patients, 445 (89%) were re-classified according to the ELN2017 criteria: ELN2017-FR (186/455; 41.8%), ELN2017-IR (179/445 40.2%) and ELN2017-AR (80/455; 18%); in 55 patients (11%) ELN2017 was not applicable (ELN2017-NC). Two-year overall survival (OS) was 68.8%, 51.3%, 45.8% and 42.8% for ELN2017-FR, ELN2017-IR, ELN2017-NC, and ELN2017-AR group, respectively (p<0.001). When comparing the two different transplant strategies in each ELN2017 risk category, a significant benefit of AuSCT over ASCT was observed among ELN2017-FR patients (2-years OS of 83.3% vs. 66.7%; p=0.0421). The two transplant procedures performed almost equally in the ELN2017-IR group (2-years OS of 73.9% vs. 70.8%; p=0.5552). This post-hoc analysis of the GIMEMA AML1310 trial, confirms that the ELN2017 classification is able to accurately discriminate patients with different outcomes and who may benefit from different transplant strategies.
Collapse
|
6
|
Talami A, Bettelli F, Pioli V, Giusti D, Gilioli A, Colasante C, Galassi L, Giubbolini R, Catellani H, Donatelli F, Maffei R, Martinelli S, Barozzi P, Potenza L, Marasca R, Trenti T, Tagliafico E, Comoli P, Luppi M, Forghieri F. How to Improve Prognostication in Acute Myeloid Leukemia with CBFB-MYH11 Fusion Transcript: Focus on the Role of Molecular Measurable Residual Disease (MRD) Monitoring. Biomedicines 2021; 9:biomedicines9080953. [PMID: 34440157 PMCID: PMC8391269 DOI: 10.3390/biomedicines9080953] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) carrying inv(16)/t(16;16), resulting in fusion transcript CBFB-MYH11, belongs to the favorable-risk category. However, even if most patients obtain morphological complete remission after induction, approximately 30% of cases eventually relapse. While well-established clinical features and concomitant cytogenetic/molecular lesions have been recognized to be relevant to predict prognosis at disease onset, the independent prognostic impact of measurable residual disease (MRD) monitoring by quantitative real-time reverse transcriptase polymerase chain reaction (qRT-PCR), mainly in predicting relapse, actually supersedes other prognostic factors. Although the ELN Working Party recently indicated that patients affected with CBFB-MYH11 AML should have MRD assessment at informative clinical timepoints, at least after two cycles of intensive chemotherapy and after the end of treatment, several controversies could be raised, especially on the frequency of subsequent serial monitoring, the most significant MRD thresholds (most commonly 0.1%) and on the best source to be analyzed, namely, bone marrow or peripheral blood samples. Moreover, persisting low-level MRD positivity at the end of treatment is relatively common and not predictive of relapse, provided that transcript levels remain stably below specific thresholds. Rising MRD levels suggestive of molecular relapse/progression should thus be confirmed in subsequent samples. Further prospective studies would be required to optimize post-remission monitoring and to define effective MRD-based therapeutic strategies.
Collapse
Affiliation(s)
- Annalisa Talami
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Francesca Bettelli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Valeria Pioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Davide Giusti
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Andrea Gilioli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Corrado Colasante
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Laura Galassi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Rachele Giubbolini
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Hillary Catellani
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Francesca Donatelli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Rossana Maffei
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Silvia Martinelli
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Patrizia Barozzi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Leonardo Potenza
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Roberto Marasca
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Unità Sanitaria Locale, 41126 Modena, Italy;
| | - Enrico Tagliafico
- Center for Genome Research, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy;
| | - Patrizia Comoli
- Pediatric Hematology/Oncology Unit and Cell Factory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, 27100 Pavia, Italy;
| | - Mario Luppi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
- Correspondence: (M.L.); (F.F.); Tel.: +39-059-4222447 (F.F.); Fax: +39-059-4222386 (F.F.)
| | - Fabio Forghieri
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, 41124 Modena, Italy; (A.T.); (F.B.); (V.P.); (D.G.); (A.G.); (C.C.); (L.G.); (R.G.); (H.C.); (F.D.); (R.M.); (S.M.); (P.B.); (L.P.); (R.M.)
- Correspondence: (M.L.); (F.F.); Tel.: +39-059-4222447 (F.F.); Fax: +39-059-4222386 (F.F.)
| |
Collapse
|
7
|
Buccisano F, Palmieri R, Piciocchi A, Maurillo L, Del Principe MI, Paterno G, Soddu S, Cerretti R, De Angelis G, Mariotti B, Irno Consalvo MA, Conti C, Fraboni D, Divona M, Ottone T, Lavorgna S, Panetta P, Voso MT, Arcese W, Venditti A. Use of Measurable Residual Disease to Evolve Transplant Policy in Acute Myeloid Leukemia: A 20-Year Monocentric Observation. Cancers (Basel) 2021; 13:1083. [PMID: 33802502 PMCID: PMC7959451 DOI: 10.3390/cancers13051083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/11/2021] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
Measurable residual disease (MRD) is increasingly employed as a biomarker of quality of complete remission (CR) in intensively treated acute myeloid leukemia (AML) patients. We evaluated if a MRD-driven transplant policy improved outcome as compared to a policy solely relying on a familiar donor availability. High-risk patients (adverse karyotype, FLT3-ITD) received allogeneic hematopoietic cell transplant (alloHCT) whereas for intermediate and low risk ones (CBF-AML and NPM1-mutated), alloHCT or autologous SCT was delivered depending on the post-consolidation measurable residual disease (MRD) status, as assessed by flow cytometry. For comparison, we analyzed a matched historical cohort of patients in whom alloHCT was delivered based on the sole availability of a matched sibling donor. Ten-years overall and disease-free survival were longer in the MRD-driven cohort as compared to the historical cohort (47.7% vs. 28.7%, p = 0.012 and 42.0% vs. 19.5%, p = 0.0003). The favorable impact of this MRD-driven strategy was evident for the intermediate-risk category, particularly for MRD positive patients. In the low-risk category, the significantly lower CIR of the MRD-driven cohort did not translate into a survival advantage. In conclusion, a MRD-driven transplant allocation may play a better role than the one based on the simple donor availability. This approach determines a superior outcome of intermediate-risk patients whereat in low-risk ones a careful evaluation is needed for transplant allocation.
Collapse
Affiliation(s)
- Francesco Buccisano
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Raffaele Palmieri
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | | | - Luca Maurillo
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Maria Ilaria Del Principe
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Giovangiacinto Paterno
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Stefano Soddu
- Centro Dati Fondazione GIMEMA, 00100 Rome, Italy; (A.P.); (S.S.)
| | - Raffaella Cerretti
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
- Rome Transplant Network, Tor Vergata University Hospital, 00133 Rome, Italy
| | - Gottardo De Angelis
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
- Rome Transplant Network, Tor Vergata University Hospital, 00133 Rome, Italy
| | - Benedetta Mariotti
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
- Rome Transplant Network, Tor Vergata University Hospital, 00133 Rome, Italy
| | - Maria Antonietta Irno Consalvo
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Consuelo Conti
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Daniela Fraboni
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Mariadomenica Divona
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Serena Lavorgna
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Paola Panetta
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| | - William Arcese
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
- Rome Transplant Network, Tor Vergata University Hospital, 00133 Rome, Italy
| | - Adriano Venditti
- Department of Biomedicine and Prevention, University Tor Vergata of Roma, 00133 Rome, Italy; (R.P.); (L.M.); (M.I.D.P.); (G.P.); (R.C.); (G.D.A.); (B.M.); (M.A.I.C.); (C.C.); (D.F.); (M.D.); (T.O.); (S.L.); (P.P.); (M.T.V.); (W.A.); (A.V.)
| |
Collapse
|
8
|
Abelson S, Zeng AGX, Nofech-Mozes I, Wang TT, Ng SWK, Minden MD, Pugh TJ, Awadalla P, Shlush LI, Murphy T, Chan SM, Dick JE, Bratman SV. Integration of intra-sample contextual error modeling for improved detection of somatic mutations from deep sequencing. SCIENCE ADVANCES 2020; 6:6/50/eabe3722. [PMID: 33298453 PMCID: PMC7725472 DOI: 10.1126/sciadv.abe3722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/23/2020] [Indexed: 06/12/2023]
Abstract
Sensitive mutation detection by next-generation sequencing is critical for early cancer detection, monitoring minimal/measurable residual disease (MRD), and guiding precision oncology. Nevertheless, because of artifacts introduced during library preparation and sequencing, the detection of low-frequency variants at high specificity is problematic. Here, we present Espresso, an error suppression method that considers local sequence features to accurately detect single-nucleotide variants (SNVs). Compared to other advanced error suppression techniques, Espresso consistently demonstrated lower numbers of false-positive mutation calls and greater sensitivity. We demonstrated Espresso's superior performance in detecting MRD in the peripheral blood of patients with acute myeloid leukemia (AML) throughout their treatment course. Furthermore, we showed that accurate mutation calling in a small number of informative genomic loci might provide a cost-efficient strategy for pragmatic risk prediction of AML development in healthy individuals. More broadly, we aim for Espresso to aid with accurate mutation detection in many other research and clinical settings.
Collapse
Affiliation(s)
- Sagi Abelson
- Ontario Institute for Cancer Research, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Andy G X Zeng
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ido Nofech-Mozes
- Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Ting Ting Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Stanley W K Ng
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Trevor J Pugh
- Ontario Institute for Cancer Research, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Philip Awadalla
- Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Liran I Shlush
- Division of Hematology, Rambam Healthcare Campus, Haifa, Israel
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Tracy Murphy
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Steven M Chan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - John E Dick
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Scott V Bratman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Mannelli F, Gianfaldoni G, Bencini S, Piccini M, Cutini I, Bonetti MI, Scappini B, Pancani F, Ponziani V, Chiarini M, Borlenghi E, Bassan R, Rossi G, Bosi A. Early peripheral blast cell clearance predicts minimal residual disease status and refines disease prognosis in acute myeloid leukemia. Am J Hematol 2020; 95:1304-1313. [PMID: 32697337 DOI: 10.1002/ajh.25942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/16/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022]
Abstract
Minimal residual disease (MRD) assessment in acute myeloid leukemia (AML) is increasingly used in risk stratification. However, several issues around this use are unresolved, including, among others, the most suitable time-point(s) for its application. Overall, late assessments appear more effective at distinguishing outcome but, in some studies, the early evaluations were already highly informative, anticipating the value of later ones. Our work integrated MRD with peripheral blast clearance (PBC), a treatment-related biomarker previously demonstrated to be a powerful predictor of response. From 2007 to 2014, we have studied 120 patients treated according to the NILG 02-06 trial and who achieved CR after induction. Patients in PBC-defined categories (separated by a 1.5-log threshold) showed significantly different probabilities of attaining MRD negativity, after either induction (MRD1) or consolidation (MRD2). Peripheral blast clearance combined with MRD1 largely anticipated MRD2-related information: when both biomarkers predicted chemosensitive disease (PBChigh /MRD1neg ), the rate of MRD2-negativity was 90%, and DFS and OS estimates were 68% and 76% at 3 years, respectively. When both markers were unfavorable (PBClow /MRD1pos ), rates of MRD2 negativity, DFS, and OS were 20%, 34%, and 24%, respectively, at 3 years. In fact, MRD2 added prognostic value only in cases with discordant PBC/MRD1 data. Our data support a reasoned timing for MRD-based therapeutic decisions, modulated on individual chemosensitivity, an approach we have implemented in a forthcoming prospective multi-center trial by Gruppo Italiano Malattie EMatologiche dell'Adulto (GIMEMA).
Collapse
Affiliation(s)
- Francesco Mannelli
- SOD Ematologia Università di Firenze, AOU Careggi Florence Italy
- Centro Ricerca e Innovazione Malattie Mieloproliferative (CRIMM) AOU Careggi Florence Italy
| | | | - Sara Bencini
- SOD Ematologia Università di Firenze, AOU Careggi Florence Italy
- Centro Diagnostico di Citofluorimetria e Immunoterapia AOU Careggi Florence Italy
| | - Matteo Piccini
- SOD Ematologia Università di Firenze, AOU Careggi Florence Italy
| | - Ilaria Cutini
- SOD Ematologia Università di Firenze, AOU Careggi Florence Italy
- SOD Terapie Cellulari e Medicina Trasfusionale AOU Careggi Florence Italy
| | - Maria Ida Bonetti
- SOD Ematologia Università di Firenze, AOU Careggi Florence Italy
- Centro Diagnostico di Citofluorimetria e Immunoterapia AOU Careggi Florence Italy
| | - Barbara Scappini
- SOD Ematologia Università di Firenze, AOU Careggi Florence Italy
| | - Fabiana Pancani
- SOD Ematologia Università di Firenze, AOU Careggi Florence Italy
| | - Vanessa Ponziani
- SOD Ematologia Università di Firenze, AOU Careggi Florence Italy
| | - Marco Chiarini
- Centro di Ricerca Emato‐Oncologica AIL (CREA) Spedali Civili Brescia Italy
- Laboratorio di Citofluorimetria; Dipartimento di Diagnostica di Laboratorio Spedali Civili Brescia Italy
| | | | - Renato Bassan
- Unità Operativa di Ematologia Ospedale dell'Angelo Mestre ‐ Venezia Italy
| | | | - Alberto Bosi
- SOD Ematologia Università di Firenze, AOU Careggi Florence Italy
| |
Collapse
|
10
|
Palmieri R, Buccisano F, Maurillo L, Del Principe MI, Paterno G, Venditti A, Martinelli G, Cerchione C. Current strategies for detection and approach to measurable residual disease in acute myeloid leukemia. Minerva Med 2020; 111:386-394. [PMID: 32955825 DOI: 10.23736/s0026-4806.20.07016-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Baseline cytogenetic/genetic features have been widely recognized to play a critical prognostic role in acute myeloid leukemia (AML) and have proven useful in designing risk-adapted treatment strategies. Nevertheless, to improve further the outcome of AML patients we are still in need of accurate methods to explore the quality of response and to adequately discriminate patients who are likely to relapse over time from those who are in deep and stable remission. In this view, is it well established that measurement of leukemic cells surviving chemotherapy (called measurable residual disease, MRD) during the course of treatment may be a reliable biomarker in predicting relapse. Detection of MRD relies on highly sensitive techniques, such as quantitative polymerase chain reaction and multiparametric flow cytometry, which, due to their levels of specificity and sensitivity, are increasingly included in the decision-making process of AML treatment. In the present manuscript, we will review the current techniques of MRD investigation and their clinical contribution to AML management.
Collapse
Affiliation(s)
- Raffaele Palmieri
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Francesco Buccisano
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy -
| | - Luca Maurillo
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | | | | | - Adriano Venditti
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Giovanni Martinelli
- Unit of Hematology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Claudio Cerchione
- Unit of Hematology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| |
Collapse
|
11
|
Tang W, Li Z, Li X, Huo Z. High CXCR2 expression predicts poor prognosis in adult patients with acute myeloid leukemia. Ther Adv Hematol 2020; 11:2040620720958586. [PMID: 32973988 PMCID: PMC7493249 DOI: 10.1177/2040620720958586] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
AIMS This study aimed to assess the associations between clinical parameters, long-term outcomes, and expression of chemokine receptor CXCR2 in patients with acute myeloid leukemia (AML). METHODS From May 2013 to May 2017, 83 adult patients newly diagnosed with AML in the Affiliated Hospital of BeiHua University and Jilin Chemical Hospital, were enrolled in this study. The expression of CXCR2 in bone marrow mononuclear cells was determined by quantitative real-time polymerase chain reaction (qRT-PCR). Clinical information and RNA-sequencing datasets of The Cancer Genome Atlas (TCGA) (n = 136) were obtained. The associations between clinical parameters, prognosis, and CXCR2 expression were analyzed. RESULTS From both cohorts, patients with AML with M4 and M5 subtypes showed higher CXCR2 expression levels than those with other French-American-British (FAB) subtypes. Patients with extramedullary leukemia infiltration had higher CXCR2 levels than those without. In our cohort, patients with high CXCR2 levels (⩾2.099) had lower relapse-free survival (RFS) (p < 0.000001) and overall survival (OS) (p = 0.000107) than those with low levels (<2.099). High CXCR2 levels (⩾2.082) also indicated a poor OS in the TCGA cohort but only in patients younger than 65 years (5-year OS: 7.7% versus 29.9% in those with CXCR2 levels < 2.082). High CXCR2 levels independently predicted poor prognosis in AML patients, as determined by Cox proportional hazards models. CONCLUSION Our results suggest that high CXCR2 expression associates with the monocytic lineage of AML and is an independent risk factor for poor patient prognosis.
Collapse
Affiliation(s)
- Wei Tang
- Department of Blood Transfusion, the Affiliated Hospital of BeiHua University, Jilin, Jilin Province, China
| | - Zunyan Li
- Department of Blood Transfusion, the Affiliated Hospital of BeiHua University, Jilin 132011, Jilin Province, China
| | - Xian Li
- Department of Hematology, Jilin Chemical Hospital, Jilin 132021, Jilin Province, China
| | - Zhonghua Huo
- Department of Blood Transfusion, the Affiliated Hospital of BeiHua University, Jilin, Jilin Province, China
| |
Collapse
|
12
|
Guo Y, Sun H, Zhang D, Zhao Y, Shi M, Yang M, Xing S, Fu X, Bin T, Lu B, Wu S, Xu X, Xu X, Chen Y, Zhao ZJ. Development of a highly sensitive method for detection of FLT3D835Y. Biomark Res 2020; 8:30. [PMID: 32817792 PMCID: PMC7424998 DOI: 10.1186/s40364-020-00210-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/04/2020] [Indexed: 12/05/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is a malignant hematological neoplasm of myeloid progenitor cells. Mutations of FLT3 in its tyrosine kinase domain (FLT3-TKD) are found in ~ 8% of patients with AML, with D835Y as the most common substitution. This mutation activates survival signals that drives the disease and is resistant to the first generation FLT3 inhibitors. Development of a highly sensitive method to detect FLT3D835Y is important to direct therapeutic options, predict prognosis, and monitor minimal residual disease in patients with AML. Methods and results In the present study, we developed a highly sensitive FLT3D835Y detection method by using the restriction fragment nested allele-specific PCR technique. The method consists of three steps: 1) initial amplification of DNA samples with PCR primers surrounding the FLT3D835Y mutation site, 2) digestion of the PCR products with restriction enzyme EcoRV that only cleaves the wild type allele, and 3) detection of FLT3D835Y by allele-specific PCR with nested primers. We were able to detect FLT3D835Y with a sensitivity of 0.001% by using purified plasmid DNAs and blood cell DNAs containing known proportions of FLT3D835Y. We analyzed blood cell DNA samples from 64 patients with AML and found six FLT3D835Y-positive cases, two of which could not be detected by conventional DNA sequencing methods. Importantly, the method was able to detect FLT3D835Y in a sample collected from a relapsed patient while the patient was in complete remission with negative MRD determined by flow cytometry. Therefore, our RFN-AS-PCR detected MRD after treatment that was missed by flow cytometry and Sanger DNA sequencing, by conventional methods. Conclusions We have developed a simple and highly sensitive method that will allow for detection of FLT3D835Y at a very low level. This method may have major clinical implications for treatment of AML.
Collapse
Affiliation(s)
- Yao Guo
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 Guangdong China.,Department of Pathology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd., BMSB 451, Oklahoma City, OK 73104 USA
| | - Honghua Sun
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 Guangdong China.,Clinical laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107 Guangdong China
| | - Dengyang Zhang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 Guangdong China
| | - Yuming Zhao
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 Guangdong China
| | - Mingxia Shi
- Department of Hematology, the First Affiliated Hospital of Kunming Medical University, Hematology Research Center of Yunnan Province, Kunming, 650032 China
| | - Ming Yang
- College of Life Sciences, Jilin University, Changchun, Jilin, 130012 China
| | - Shu Xing
- College of Life Sciences, Jilin University, Changchun, Jilin, 130012 China
| | - Xueqi Fu
- College of Life Sciences, Jilin University, Changchun, Jilin, 130012 China
| | - Ting Bin
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China, Shenzhen, 518107 Guangdong China
| | - Bo Lu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China, Shenzhen, 518107 Guangdong China
| | - Shunjie Wu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China, Shenzhen, 518107 Guangdong China
| | - Xiaojun Xu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China, Shenzhen, 518107 Guangdong China
| | - Xuesong Xu
- Clinical Laboratory of China-Japan Union Hospital, Jilin University, Changchun, Jilin, 130033 China
| | - Yun Chen
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 Guangdong China
| | - Zhizhuang Joe Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd., BMSB 451, Oklahoma City, OK 73104 USA
| |
Collapse
|
13
|
Is There Still a Role for Autologous Stem Cell Transplantation for the Treatment of Acute Myeloid Leukemia? Cancers (Basel) 2019; 12:cancers12010059. [PMID: 31878297 PMCID: PMC7016672 DOI: 10.3390/cancers12010059] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022] Open
Abstract
After intensive induction chemotherapy and complete remission achievement, patients with acute myeloid leukemia (AML) are candidates to receive either high-dose cytarabine-based regimens, or autologous (ASCT) or allogeneic (allo-SCT) hematopoietic stem cell transplantations as consolidation treatment. Pretreatment risk classification represents a determinant key of type and intensity of post-remission therapy. Current evidence indicates that allo-SCT represents the treatment of choice for high and intermediate risk patients if clinically eligible, and its use is favored by increasing availability of unrelated or haploidentical donors. On the contrary, the adoption of ASCT is progressively declining, although numerous studies indicate that in favorable risk AML the relapse rate is lower after ASCT than chemotherapy. In addition, the burden of supportive therapy and hospitalization favors ASCT. In this review, we summarize current indications (if any) to ASCT on the basis of molecular genetics at diagnosis and minimal residual disease evaluation after induction/consolidation phase. Finally, we critically discuss the role of ASCT in older patients with AML and acute promyelocytic leukemia.
Collapse
|
14
|
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: 91] [Impact Index Per Article: 15.2] [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
|
15
|
Prognostic impact of circulating tumor DNA status post–allogeneic hematopoietic stem cell transplantation in AML and MDS. Blood 2019; 133:2682-2695. [DOI: 10.1182/blood-2018-10-880690] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/07/2019] [Indexed: 12/11/2022] Open
Abstract
Abstract
This study was performed to assess the utility of tumor-derived fragmentary DNA, or circulating tumor DNA (ctDNA), for identifying high-risk patients for relapse of acute myeloid leukemia and myelodysplastic syndrome (AML/MDS) after undergoing myeloablative allogeneic hematopoietic stem cell transplantation (alloSCT). We retrospectively collected tumor and available matched serum samples at diagnosis and 1 and 3 months post-alloSCT from 53 patients with AML/MDS. After identifying driver mutations in 51 patients using next-generation sequencing, we designed at least 1 personalized digital polymerase chain reaction assay per case. Diagnostic ctDNA and matched tumor DNA exhibited excellent correlations with variant allele frequencies. Sixteen patients relapsed after a median of 7 months post-alloSCT. Both mutation persistence (MP) in bone marrow (BM) at 1 and 3 months post-alloSCT and corresponding ctDNA persistence (CP) in the matched serum (MP1 and MP3; CP1 and CP3, respectively) were comparably associated with higher 3-year cumulative incidence of relapse (CIR) rates (MP1 vs non-MP1, 72.9% vs 13.8% [P = .0012]; CP1 vs non-CP1, 65.6% vs 9.0% [P = .0002]; MP3 vs non-MP3, 80% vs 11.6% [P = .0002]; CP3 vs non-CP3, 71.4% vs 8.4% [P < .0001]). We subsequently evaluated whether subset analysis of patients with 3 genes associated with clonal hematopoiesis, DNMT3A, TET2, and ASXL1 (DTA), could also be helpful in relapse prediction. As a result, CP based on DTA gene mutations also had the prognostic effect on CIR. These results, for the first time, support the utility of ctDNA as a noninvasive prognostic biomarker in patients with AML/MDS undergoing alloSCT.
Collapse
|
16
|
Sui J, Chen Q, Zhang Y, Sheng Y, Wu J, Li J, Weng X, Chen B. Identifying leukemia-associated immunophenotype-based individualized minimal residual disease in acute myeloid leukemia and its prognostic significance. Am J Hematol 2019; 94:528-538. [PMID: 30734356 DOI: 10.1002/ajh.25431] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/01/2019] [Accepted: 02/05/2019] [Indexed: 12/25/2022]
Abstract
Based on the leukemia-associated immunophenotypes (LAIPs), minimal residual disease (MRD) related to the outcome can be detected by multiparameter flow cytometry in acute myeloid leukemia (AML) patients. Although 0.1% was commonly used as a cutoff value, measurable MRD or MRD level below 0.1% has also been associated with prognostic significance and more sensitive thresholds (<0.1%) are required for improving AML prognosis prediction. In this study, 292 adult patients diagnosed with AML (non-M3) were enrolled, 36 kinds of LAIPs were identified, and the baseline expression levels in normal or regenerating bone marrows of each kind of LAIP were established, which ranged from <2.00 × 10-5 to 5.71 × 10-4 . The baseline level of each LAIP was considered as the individual threshold for MRD assessment. MRD statuses stratified by 0.1% and individual threshold were termed as 0.1%-MRD and individual-MRD, respectively. The patients of individual-MRDneg showed significantly better survival compared with those of 0.1%-MRDneg /individual-MRDpos or 0.1%-MRDpos . Multivariate analysis showed that when time points of complete remission, post the first and second consolidation courses, were considered, only individual-MRD post second consolidation presented independent prognostic value. Notably, in patients of cytogenetic/molecular low-risk (LR) or intermediate-risk (IR), the individual-MRD status could identify the patients with significantly different outcomes, while 0.1%-MRD status could not. Furthermore, among the patients of the LR or IR group which received chemotherapy only, those with individual-MRDneg status presented favorable survival, which was comparable with that of the patients accepted allogeneic hematopoietic stem cell transplantation (ASCT). This approach is useful in the selection of an ASCT strategy for clinical practice.
Collapse
Affiliation(s)
- Jing‐Ni Sui
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Qiu‐Sheng Chen
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Yun‐Xiang Zhang
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Yan Sheng
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Jing Wu
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Jun‐Min Li
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Xiang‐Qin Weng
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| | - Bing Chen
- State Key Laboratory of Medical GenomicsShanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine 197 Rui Jin Road II, Shanghai China
| |
Collapse
|
17
|
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: 3] [Impact Index Per Article: 0.5] [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
|
18
|
Minimal/Measurable Residual Disease Monitoring in NPM1-Mutated Acute Myeloid Leukemia: A Clinical Viewpoint and Perspectives. Int J Mol Sci 2018; 19:ijms19113492. [PMID: 30404199 PMCID: PMC6274702 DOI: 10.3390/ijms19113492] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/02/2018] [Accepted: 11/03/2018] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) with NPM1 gene mutations is currently recognized as a distinct entity, due to its unique biological and clinical features. We summarize here the results of published studies investigating the clinical application of minimal/measurable residual disease (MRD) in patients with NPM1-mutated AML, receiving either intensive chemotherapy or hematopoietic stem cell transplantation. Several clinical trials have so far demonstrated a significant independent prognostic impact of molecular MRD monitoring in NPM1-mutated AML and, accordingly, the Consensus Document from the European Leukemia Net MRD Working Party has recently recommended that NPM1-mutated AML patients have MRD assessment at informative clinical timepoints during treatment and follow-up. However, several controversies remain, mainly with regard to the most clinically significant timepoints and the MRD thresholds to be considered, but also with respect to the optimal source to be analyzed, namely bone marrow or peripheral blood samples, and the correlation of MRD with other known prognostic indicators. Moreover, we discuss potential advantages, as well as drawbacks, of newer molecular technologies such as digital droplet PCR and next-generation sequencing in comparison to conventional RQ-PCR to quantify NPM1-mutated MRD. In conclusion, further prospective clinical trials are warranted to standardize MRD monitoring strategies and to optimize MRD-guided therapeutic interventions in NPM1-mutated AML patients.
Collapse
|
19
|
Circulating-free tumour DNA and the promise of disease phenotyping in hepatocellular carcinoma. Oncogene 2018; 37:4635-4638. [DOI: 10.1038/s41388-018-0262-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 03/23/2018] [Indexed: 02/08/2023]
|