1
|
Zhu C, Stiehl T. Modelling post-chemotherapy stem cell dynamics in the bone marrow niche of AML patients. Sci Rep 2024; 14:25060. [PMID: 39443599 PMCID: PMC11500015 DOI: 10.1038/s41598-024-75429-7] [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: 06/27/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
Acute myeloid leukemia (AML) is a stem cell-driven malignancy of the blood forming (hematopoietic) system. Despite of high dose chemotherapy with toxic side effects, many patients eventually relapse. The "7+3 regimen", which consists of 7 days of cytarabine in combination with daunorubicin during the first 3 days, is a widely used therapy protocol. Since peripheral blood cells are easily accessible to longitudinal sampling, significant research efforts have been undertaken to characterize and reduce adverse effects on circulating blood cells. However, much less is known about the impact of the 7+3 regimen on human hematopoietic stem cells and their physiological micro-environments, the so-called stem cell niches. One reason for this is the technical inability to observe human stem cells in vivo and the discomfort related to bone marrow biopsies. To better understand the treatment effects on human stem cells, we consider a mechanistic mathematical model of the stem cell niche before, during and after chemotherapy. The model accounts for different maturation stages of leukemic and hematopoietic cells and considers key processes such as cell proliferation, self-renewal, differentiation and therapy-induced cell death. In the model, hematopoietic (HSCs) and leukemic stem cells (LSCs) compete for a joint niche and respond to both systemic and niche-derived signals. We relate the model to clinical trial data from literature which longitudinally quantifies the counts of hematopoietic stem like (CD34+CD38-ALDH+) cells at diagnosis and after therapy. The proposed model can capture the clinically observed interindividual heterogeneity and reproduce the non-monotonous dynamics of the hematopoietic stem like cells observed in relapsing patients. Our model allows to simulate different scenarios proposed in literature such as therapy-related impairment of the stem cell niche or niche-mediated resistance. Model simulations suggest that during the post-therapy phase a more than 10-fold increase of hematopoietic stem-like cell proliferation rates is required to recapitulate the measured cell dynamics in patients achieving complete remission. We fit the model to data of 7 individual patients and simulate variations of the treatment protocol. These simulations are in line with the clinical finding that G-CSF priming can improve the treatment outcome. Furthermore, our model suggests that a decline of HSC counts during remission might serve as an indication for salvage therapy in patients lacking MRD (minimal residual disease) markers.
Collapse
Affiliation(s)
- Chenxu Zhu
- Institute for Computational Biomedicine-Disease Modeling, RWTH Aachen University, Aachen, Germany
| | - Thomas Stiehl
- Institute for Computational Biomedicine-Disease Modeling, RWTH Aachen University, Aachen, Germany.
- Department of Science and Environment, Roskilde University, Roskilde, Denmark.
- Centre for Mathematical Modeling-Human Health and Disease, Roskilde University, Roskilde, Denmark.
| |
Collapse
|
2
|
Sun Y, Zhu G, Zhong H. Minimal residual disease monitoring in acute myeloid leukemia: Focus on MFC-MRD and treatment guidance for elderly patients. Eur J Haematol 2024; 112:870-878. [PMID: 38342613 DOI: 10.1111/ejh.14187] [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: 11/24/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/13/2024]
Abstract
Acute myeloid leukemia (AML) is distinguished by clonal growth of myeloid precursor cells, which impairs normal hematopoiesis. Minimal residual disease (MRD) refers to the residual leukemia cells that persist after chemotherapy. Patients who test positive for MRD have a higher likelihood of experiencing a recurrence, regardless of the specific chemotherapy approach used. Multi-parameter flow cytometry (MFC), polymerase chain reaction (PCR), and next-generation sequencing (NGS) are commonly employed techniques for identifying MRD. In the context of AML, patients are frequently monitored for measurable residual disease via multi-parameter flow cytometry (MFC-MRD). In order to explore recent advancements in AML and MRD diagnosis, an extensive search of the PubMed database was conducted, focusing on relevant research in the past 20 years. This review aims to examine various MRD monitoring methods, the optimal time points for assessment, as well as different specimen types used. Additionally, it underscores the significance of MFC-MRD assessment in guiding the treatment of elderly AML.
Collapse
Affiliation(s)
- Yue Sun
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Gelan Zhu
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Hua Zhong
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
3
|
DeWolf S, Tallman MS, Rowe JM, Salman MY. What Influences the Decision to Proceed to Transplant for Patients With AML in First Remission? J Clin Oncol 2023; 41:4693-4703. [PMID: 37611216 PMCID: PMC10564290 DOI: 10.1200/jco.22.02868] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/12/2023] [Accepted: 06/14/2023] [Indexed: 08/25/2023] Open
Abstract
Although allogeneic hematopoietic cell transplantation (allo-HCT) remains the backbone of curative treatment for the majority of fit adults diagnosed with AML, there is indeed a subset of patients for whom long-term remission may be achieved without transplantation. Remarkable changes in our knowledge of AML biology in recent years has transformed the landscape of diagnosis, management, and treatment of AML. Specifically, markedly increased understanding of molecular characteristics of AML, the expanded application of minimal/measurable residual diseases testing, and an increased armamentarium of leukemia-directed therapeutic agents have created a new paradigm for the medical care of patients with AML. An attempt is herein made to decipher the decision to proceed to transplant for patients with AML in first complete remission on the basis of the current best available evidence. The focus is on factors affecting the biology and treatment of AML itself, rather than on variables related to allo-HCT, an area characterized by significant advancements that have reduced overall therapy-related complications. This review seeks to focus on areas of particular complexity, while simultaneously providing clarity on how our current knowledge and treatment strategies may, or may not, influence the decision to pursue allo-HCT in patients with AML.
Collapse
Affiliation(s)
- Susan DeWolf
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Martin S. Tallman
- Division of Hematology and Oncology Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Jacob M. Rowe
- Rambam Health Care Campus and Technion, Israel Institute of Technology, Haifa, Israel
- Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel
| | | |
Collapse
|
4
|
Li X, Tong X. Role of Measurable Residual Disease in Older Adult Acute Myeloid Leukemia. Clin Interv Aging 2023; 18:921-931. [PMID: 37313310 PMCID: PMC10258117 DOI: 10.2147/cia.s409308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
There is overwhelming evidence indicating that the use of measurable residual disease (MRD) as a biomarker provides critical prognostic information and that MRD may have a role in directing postremission decisions. There are a variety of assays for MRD assessment, such as multiparameter flow cytometry and molecular assessment of MRD, which present different characteristics in patients older than 60 years of age. Due to multiple reasons related to age, the progress of older adult AML patients is rarely investigated, especially with respect to MRD. In this review, we will clarify the characteristics of different assays for assessing MRD, focusing on its role as a risk-stratification biomarker to predict prognostic information and its role in optimal postremission therapy among older adult AML patients. These characteristics also provide guidance regarding the potential to apply personalized medicine in older adult AML patients.
Collapse
Affiliation(s)
- Xueyao Li
- Department of Hematology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Xiuzhen Tong
- Department of Hematology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
5
|
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
|
6
|
Small S, Oh TS, Platanias LC. Role of Biomarkers in the Management of Acute Myeloid Leukemia. Int J Mol Sci 2022; 23:14543. [PMID: 36498870 PMCID: PMC9741257 DOI: 10.3390/ijms232314543] [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: 09/30/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022] Open
Abstract
Despite many recent advances in treatment options, acute myeloid leukemia (AML) still has a high mortality rate. One important issue in optimizing outcomes for AML patients lies in the limited ability to predict response to specific therapies, duration of response, and likelihood of relapse. With evolving genetic characterization and improving molecular definitions, the ability to predict outcomes and long-term prognosis is slowly improving. The majority of the currently used prognostic assessments relate to molecular and chromosomal abnormalities, as well as response to initial therapy. These risk categories, however, do not account for a large amount of the variability in AML. Laboratory techniques now utilized in the clinic extend beyond bone marrow morphology and single gene sequencing, to next-generation sequencing of large gene panels and multiparameter flow cytometry, among others. Other technologic advances, such as gene expression analysis, have yet to demonstrate enough predictive and prognostic power to be employed in clinical medicine outside of clinical trials, but may be incorporated into the clinic in the future. In this review, we discuss the utility of current biomarkers, and present novel biomarker techniques and strategies that are in development for AML patients. Measurable residual disease (MRD) is a powerful prognostic tool that is increasingly being incorporated into clinical practice, and there are some exciting emerging biomarker technologies that have the potential to improve prognostic power in AML. As AML continues to be a difficult-to-treat disease with poor outcomes in many subtypes, advances in biomarkers that lead to better treatment decisions are greatly needed.
Collapse
Affiliation(s)
- Sara Small
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
- Division of Hematology-Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Timothy S. Oh
- Division of Hospital Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Leonidas C. Platanias
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
- Division of Hematology-Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
7
|
Real-World Experience of Measurable Residual Disease Response and Prognosis in Acute Myeloid Leukemia Treated with Venetoclax and Azacitidine. Cancers (Basel) 2022; 14:cancers14153576. [PMID: 35892834 PMCID: PMC9332730 DOI: 10.3390/cancers14153576] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/16/2022] [Accepted: 07/19/2022] [Indexed: 02/04/2023] Open
Abstract
The prognostic value of measurable residual disease (MRD) by flow cytometry in acute myeloid leukemia (AML) patients treated with non-intensive therapy is relatively unexplored. The clinical value of MRD threshold below 0.1% is also unknown after non-intensive therapy. In this study, MRD to a sensitivity of 0.01% was analyzed in sixty-three patients in remission after azacitidine/venetoclax treatment. Multivariable cox regression analysis identified prognostic factors associated with cumulative incidence of relapse (CIR), progression-free survival (PFS) and overall survival (OS). Patients who achieved MRD < 0.1% had a lower relapse rate than those who were MRD ≥ 0.1% at 18 months (13% versus 57%, p = 0.006). Patients who achieved an MRD-negative CR had longer median PFS and OS (not reached and 26.5 months) than those who were MRD-positive (12.6 and 10.3 months, respectively). MRD < 0.1% was an independent predictor for CIR, PFS, and OS, after adjusting for European Leukemia Net (ELN) risk, complex karyotype, and transplant (HR 5.92, 95% CI 1.34−26.09, p = 0.019 for PFS; HR 2.60, 95% CI 1.02−6.63, p = 0.046 for OS). Only an MRD threshold of 0.1%, and not 0.01%, was predictive for OS. Our results validate the recommended ELN MRD cut-off of 0.1% to discriminate between patients with improved CIR, PFS, and OS after azacitidine/venetoclax therapy.
Collapse
|
8
|
Falini B. AML risk models: where do we stand ? Am J Hematol 2022; 97:1124-1126. [PMID: 35856388 DOI: 10.1002/ajh.26666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/02/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Brunangelo Falini
- Institute of Hematology and CREO, University and Hospital of Perugia, Perugia, Italy
| |
Collapse
|
9
|
Molecular Measurable Residual Disease Assessment before Hematopoietic Stem Cell Transplantation in Pediatric Acute Myeloid Leukemia Patients: A Retrospective Study by the I-BFM Study Group. Biomedicines 2022; 10:biomedicines10071530. [PMID: 35884834 PMCID: PMC9313005 DOI: 10.3390/biomedicines10071530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/14/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a curative post-remission treatment in patients with acute myeloid leukemia (AML), but relapse after transplant is still a challenging event. In recent year, several studies have investigated the molecular minimal residual disease (qPCR-MRD) as a predictor of relapse, but the lack of standardized protocols, cut-offs, and timepoints, especially in the pediatric setting, has prevented its use in several settings, including before HSCT. Here, we propose the first collaborative retrospective I-BFM-AML study assessing qPCR-MRD values in pretransplant bone marrow samples of 112 patients with a diagnosis of AML harboring t(8;21)(q22; q22)RUNX1::RUNX1T1, or inv(16)(p13q22)CBFB::MYH11, or t(9;11)(p21;q23)KMT2A::MLLT3, or FLT3-ITD genetic markers. We calculated an ROC cut-off of 2.1 × 10−4 that revealed significantly increased OS (83.7% versus 57.1%) and EFS (80.2% versus 52.9%) for those patients with lower qPCR-MRD values. Then, we partitioned patients into three qPCR-MRD groups by combining two different thresholds, 2.1 × 10−4 and one lower cut-off of 1 × 10−2, and stratified patients into low-, intermediate-, and high-risk groups. We found that the 5-year OS (83.7%, 68.6%, and 39.2%, respectively) and relapse-free survival (89.2%, 73.9%, and 67.9%, respectively) were significantly different independent of the genetic lesion, conditioning regimen, donor, and stem cell source. These data support the PCR-based approach playing a clinical relevance in AML transplant management.
Collapse
|
10
|
CALCRL Gene is a Suitable Prognostic Factor in AML/ETO + AML Patients. JOURNAL OF ONCOLOGY 2022; 2022:3024360. [PMID: 35342399 PMCID: PMC8942673 DOI: 10.1155/2022/3024360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 01/16/2022] [Accepted: 03/01/2022] [Indexed: 11/17/2022]
Abstract
Introduction The t(8 ; 21) translocation is the most common chromosomal abnormality in human acute myeloid leukemia (AML) subtype 2 (M2), which forms the AML/ETO fusion gene. However, AML/ETO alone does not necessarily cause leukemia. Other factors are thought to contribute to the disease. Calcitonin receptor-like (CALCRL), a G-protein-coupled neuropeptide receptor, is involved in various biological processes, such as colony formation and drug resistance. Methods First, The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were used to determine any differences in CALCRL expression in AML patients with and without AML/ETO and the prognostic significance of CALCRL expression in AML patients was further evaluated. Next, we detected the CALCRL expression level in 67 AML/ETO+ AML patients and 16 patients with nonmalignant hematological diseases using qRT-PCR and identified its prognostic relevance. Results Individuals in the group expressing low levels of CALCRL had a longer median survival time. In AML/ETO+ AML patients, higher mRNA levels of CALCRL were observed before treatment, which decreased after the complete remission that followed multiple chemotherapy sessions. Clinical features indicated that more patients in the CALCRLhigh group also had c-kit mutations compared with patients in other groups. Overall survival (OS) was longer in patients with lower levels of CALCRL expression, especially in patients with c-kit mutations or with more blast cells in bone marrow (BM). In addition, a longer OS was observed in the CALCRLlow group after hematopoietic stem cell transplantation (HSCT). Conclusions This preliminary study indicates that CALCRL could serve as a suitable prognostic factor in AML/ETO+ AML patients.
Collapse
|
11
|
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: 2.0] [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
|
12
|
Pan X, Gao M, Sun Y, Zhou Y, Wang K, Wang Y, Xu L, Zhang X, Huang X, Zhao X. Significance of WT1 and multiparameter flow cytometry assessment in patients with chronic myelomonocytic leukemia receiving allogeneic hematopoietic stem cell transplantation. Int J Lab Hematol 2022; 44:510-517. [DOI: 10.1111/ijlh.13788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/10/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Xinan Pan
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Mengge Gao
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Yuqian Sun
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Yang Zhou
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Ke Wang
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Yu Wang
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Lanping Xu
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Xiaohui Zhang
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
| | - Xiaojun Huang
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
- Peking‐Tsinghua Center for Life Sciences Beijing China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies Chinese Academy of Medical Sciences Beijing China
| | - Xiao‐Su Zhao
- Peking University People’s HospitalPeking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell Transplantation Beijing China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies Chinese Academy of Medical Sciences Beijing China
| |
Collapse
|
13
|
Investigation of measurable residual disease in acute myeloid leukemia by DNA methylation patterns. Leukemia 2022; 36:80-89. [PMID: 34131280 PMCID: PMC8727289 DOI: 10.1038/s41375-021-01316-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 02/05/2023]
Abstract
Assessment of measurable residual disease (MRD) upon treatment of acute myeloid leukemia (AML) remains challenging. It is usually addressed by highly sensitive PCR- or sequencing-based screening of specific mutations, or by multiparametric flow cytometry. However, not all patients have suitable mutations and heterogeneity of surface markers hampers standardization in clinical routine. In this study, we propose an alternative approach to estimate MRD based on AML-associated DNA methylation (DNAm) patterns. We identified four CG dinucleotides (CpGs) that commonly reveal aberrant DNAm in AML and their combination could reliably discern healthy and AML samples. Interestingly, bisulfite amplicon sequencing demonstrated that aberrant DNAm patterns were symmetric on both alleles, indicating that there is epigenetic crosstalk between homologous chromosomes. We trained shallow-learning and deep-learning algorithms to identify anomalous DNAm patterns. The method was then tested on follow-up samples with and without MRD. Notably, even samples that were classified as MRD negative often revealed higher anomaly ratios than healthy controls, which may reflect clonal hematopoiesis. Our results demonstrate that targeted DNAm analysis facilitates reliable discrimination of malignant and healthy samples. However, since healthy samples also comprise few abnormal-classified DNAm reads the approach does not yet reliably discriminate MRD positive and negative samples.
Collapse
|
14
|
Huo Y, Guan XM, Dou Y, Wen XH, Guo YX, Shen YL, An XZ, Yu J. Prognostic significance of measurable residual disease based on multiparameter flow cytometry in childhood acute myeloid leukemia. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23:1111-1118. [PMID: 34753542 DOI: 10.7499/j.issn.1008-8830.2106102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
OBJECTIVES To study the prognostic value of measurable residual disease (MRD) for childhood acute myeloid leukemia (AML) by analyzing MRD-guided risk stratification therapy. METHODS A total of 93 children with AML were prospectively enrolled in this study. Chemotherapy with the 2015-AML-03 regimen was completed according to the risk stratification determined by genetic abnormality at initial diagnosis and MRD and bone marrow cytology after induction therapy I. Multiparameter flow cytometry was used to dynamically monitor MRD and analyze the prognostic effect of MRD on 3-year cumulative incidence of recurrence (CIR) rate, event-free survival (EFS) rate, and overall survival (OS) rate. RESULTS The 93 children with AML had a 3-year CIR rate of 48%±6%, a median time to recurrence of 11 months (range 2-32 months), a 3-year OS rate of 65%±6%, and a 3-year EFS rate of 50%±5%. After induction therapy I and intensive therapy I, the MRD-positive children had a significantly higher 3-year CIR rate and significantly lower 3-year EFS and OS rates than the MRD-negative children (P<0.05). There were no significant differences in 3-year CIR, EFS, and OS rates between the MRD-positive children with a low risk at initial diagnosis and the MRD-negative children after adjustment of chemotherapy intensity (P>0.05). The multivariate analysis showed that positive MRD after intensive treatment I was a risk factor for 3-year OS rate in children with AML (P<0.05). CONCLUSIONS MRD has predictive value for the prognosis of children with AML. Based on the MRD-guided risk stratification therapy, reasonable application of chemotherapy may improve the overall prognosis of children with AML.
Collapse
Affiliation(s)
- Ya Huo
- Department of Hematology and Oncology, Children's Hospital Affiliated to Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Children's Health and Disease Clinical Medicine Research Center/National International Science and Technology Cooperation Base for Critical Child Developmental Diseases/Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China (Yu J, 1808106657@qq. com)
| | - Xian-Min Guan
- Department of Hematology and Oncology, Children's Hospital Affiliated to Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Children's Health and Disease Clinical Medicine Research Center/National International Science and Technology Cooperation Base for Critical Child Developmental Diseases/Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China (Yu J, 1808106657@qq. com)
| | - Ying Dou
- Department of Hematology and Oncology, Children's Hospital Affiliated to Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Children's Health and Disease Clinical Medicine Research Center/National International Science and Technology Cooperation Base for Critical Child Developmental Diseases/Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China (Yu J, 1808106657@qq. com)
| | - Xian-Hao Wen
- Department of Hematology and Oncology, Children's Hospital Affiliated to Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Children's Health and Disease Clinical Medicine Research Center/National International Science and Technology Cooperation Base for Critical Child Developmental Diseases/Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China (Yu J, 1808106657@qq. com)
| | - Yu-Xia Guo
- Department of Hematology and Oncology, Children's Hospital Affiliated to Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Children's Health and Disease Clinical Medicine Research Center/National International Science and Technology Cooperation Base for Critical Child Developmental Diseases/Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China (Yu J, 1808106657@qq. com)
| | - Ya-Li Shen
- Department of Hematology and Oncology, Children's Hospital Affiliated to Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Children's Health and Disease Clinical Medicine Research Center/National International Science and Technology Cooperation Base for Critical Child Developmental Diseases/Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China (Yu J, 1808106657@qq. com)
| | - Xi-Zhou An
- Department of Hematology and Oncology, Children's Hospital Affiliated to Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Children's Health and Disease Clinical Medicine Research Center/National International Science and Technology Cooperation Base for Critical Child Developmental Diseases/Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China (Yu J, 1808106657@qq. com)
| | - Jie Yu
- Department of Hematology and Oncology, Children's Hospital Affiliated to Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Children's Health and Disease Clinical Medicine Research Center/National International Science and Technology Cooperation Base for Critical Child Developmental Diseases/Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China (Yu J, 1808106657@qq. com)
| |
Collapse
|
15
|
Clofarabine-fludarabine-busulfan in HCT for pediatric leukemia: an effective, low toxicity, TBI-free conditioning regimen. Blood Adv 2021; 6:1719-1730. [PMID: 34781362 PMCID: PMC8941455 DOI: 10.1182/bloodadvances.2021005224] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/10/2021] [Indexed: 11/20/2022] Open
Abstract
CloFluBu-conditioning results in encouraging EFS for ALL and AML, with low TRM, limited incidence of aGvHD and GF, and no cases of VOD. Minimal residual disease status prior to transplantation impacted outcome due to increased relapse risk in both AML and ALL patients.
We prospectively studied clofarabine-fludarabine-busulfan (CloFluBu)-conditioning in allogeneic hematopoietic cell therapy (HCT) for lymphoid and myeloid malignancies and hypothesized that CloFluBu provides a less toxic alternative to conventional conditioning regimens, with adequate antileukemic activity. All patients receiving their first HCT, from 2011-2019, were included and received CloFluBu. The primary endpoint was event-free survival (EFS). Secondary endpoints were overall survival (OS), graft-versus-host disease (GvHD)-relapse-free survival (GRFS), treatment-related mortality (TRM), cumulative incidence of relapse (CIR), acute and chronic GvHD (aGvHD and cGvHD), and veno-occlusive disease (VOD). Cox proportional hazard and Fine and Gray competing-risk models were used for data analysis. One hundred fifty-five children were included: 60 acute lymphoid leukemia (ALL), 69 acute myeloid leukemia (AML), and 26 other malignancies (mostly MDS-EB). The median age was 9.7 (0.5 to 18.6) years. Estimated 2-year EFS was 72.0% ± 6.0 in ALL patients, and 62.4% ± 6.0 in AML patients. TRM in the whole cohort was 11.0% ± 2.6, incidence of aGvHD 3 to 4 at 6 months was 12.3% ± 2.7, extensive cGvHD at 2 years was 6.4% ± 2.1. Minimal residual disease-positivity prior to HCT was associated with higher CIR, both in ALL and AML. CloFluBu showed limited toxicity and encouraging EFS. CloFluBu is a potentially less toxic alternative to conventional conditioning regimens. Randomized prospective studies are needed.
Collapse
|
16
|
[Chinese consensus on minimal residual disease detection and interpretation of patients with acute myeloid leukemia (2021)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 42:889-897. [PMID: 35045649 PMCID: PMC8763587 DOI: 10.3760/cma.j.issn.0253-2727.2021.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Indexed: 12/02/2022]
|
17
|
Naidoo M, Piercey O, Tie J. Circulating Tumour DNA and Colorectal Cancer: the Next Revolutionary Biomarker? Curr Oncol Rep 2021; 23:140. [PMID: 34735665 DOI: 10.1007/s11912-021-01137-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Improving outcomes for patients with colorectal cancer in both the adjuvant and metastatic setting has been challenging. Here, we review the current and future directions for using ctDNA in clinical practice. RECENT FINDINGS Circulating tumour DNA (ctDNA) with its ability to detect minimal residual disease is beginning to refine the way we assess recurrence risk in the adjuvant setting. We can potentially tailor treatments to reduce recurrence risk and minimize treatment toxicity. In the metastatic setting, ctDNA can provide a less invasive method of detecting clinically important genetic changes to guide molecularly targeted treatment and to identify mechanisms of molecular resistance. ctDNA can be a surrogate marker for treatment response and help guide the timing of anti-EGFR rechallenge. We await the results of the randomized clinical trials assessing clinical utility of ctDNA in both the adjuvant and metastatic setting before incorporating ctDNA into clinical practice.
Collapse
Affiliation(s)
- Mahendra Naidoo
- Department of Medical Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Parkville Melbourne, VIC 3000, Australia.
| | - Oliver Piercey
- Department of Medical Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Parkville Melbourne, VIC 3000, Australia
| | - Jeanne Tie
- Department of Medical Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Parkville Melbourne, VIC 3000, Australia.,Division of Personalised Oncology, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, 3052, Australia.,Department of Medical Oncology, Western Health, Melbourne, VIC, 3021, Australia.,Sir Peter MacCallum, Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| |
Collapse
|
18
|
Liu X, Liu X, Cai M, Luo A, He Y, Liu S, Zhang X, Yang X, Xu L, Jiang H. CircRNF220, not its linear cognate gene RNF220, regulates cell growth and is associated with relapse in pediatric acute myeloid leukemia. Mol Cancer 2021; 20:139. [PMID: 34702297 PMCID: PMC8549339 DOI: 10.1186/s12943-021-01395-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/17/2021] [Indexed: 02/08/2023] Open
Abstract
Background Circular RNAs (circRNAs) constitute a family of transcripts with unique structures and have been confirmed to be critical in tumorigenesis and to be potential biomarkers or therapeutic targets. However, only a few circRNAs have been functionally characterized in pediatric acute myeloid leukemia (AML). Methods Here, we investigated the expression pattern of circRNAs in pediatric AML using a circRNA microarray. The characteristics, potential diagnostic value, and prognostic significance of circRNF220 were evaluated. A series of functional experiments were performed to investigate the role of circRNF220 in primary pediatric AML cells. Then we investigated the aberrant transcriptional networks regulated by circRNF220 in primary AML cells by RNA-seq. Furthermore, biotin RNA pulldown assays were implemented to verify the relationship between circRNF220 and miR-30a. Results We identified a circRNA, circRNF220, which was specifically abundant in and accumulated in the peripheral blood and bone marrow of pediatric patients with AML. It could distinguish AML from ALL and other hematological malignancies with high sensitivity and specificity. Significantly, circRNF220 expression independently predicted prognosis, while high expression of circRNF220 was an unfavorable prognostic marker for relapse. Furthermore, we characterized the function of circRNF220 and found that circRNF220 knockdown specifically inhibited proliferation and promoted apoptosis in AML cell lines and primary cells. Mechanistically, circRNF220 may act as an endogenous sponge of miR-30a to sequester miR-30a and inhibit its activity, which increases the expression of its targets MYSM1 and IER2 and implicated in AML relapse. Conclusions Collectively, these findings demonstrated that circRNF220 could be highly efficient and specific for the accurate diagnosis of pediatric AML, with implications for relapse prediction. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-021-01395-7.
Collapse
Affiliation(s)
- Xiaodan Liu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China
| | - Xiaoping Liu
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Mansi Cai
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ailing Luo
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yingyi He
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China
| | - Sha Liu
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China
| | - Xiaohong Zhang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China
| | - Xu Yang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China.,Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ling Xu
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China.
| | - Hua Jiang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Zhujiang Newtown, Tianhe District, Guangzhou, 510623, Guangdong, China.
| |
Collapse
|
19
|
Hao Q, Liu X, Zhang Y, Zhang D, Li B, Wang J. MRD abnormal expression predict poor outcomes for refractory or relapsed acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation. J Clin Lab Anal 2021; 35:e23974. [PMID: 34432318 PMCID: PMC8529132 DOI: 10.1002/jcla.23974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/09/2021] [Accepted: 08/12/2021] [Indexed: 12/16/2022] Open
Abstract
We retrospectively analyzed data from 197 patients with refractory or relapsed acute myeloid leukemia (r/rAML) who underwent allo‐HCT between January 2013 and February 2020 in our center (patients with promyelocytic leukemia were excluded). Of all patients, 86 achieved a complete morphological remission (CR) before transplant, while 111 failed to do so (NR). In the CR group, 32 patients displayed minimal residual disease (MRD‐positive). According to their immunophenotype pre‐HCT, we divided the MRD‐positive group and NR group into three subgroups: MRD 0+ group (without any antigen abnormal expression of CD7+, CD56+, CD38−, or HLA‐DR−) 28 patients, MRD 1+ group (with one abnormal antigen expression of CD7+, CD56+, CD38−, or HLA‐DR−) 63 patients, MRD 2+ group (with two or more abnormal antigens expression of CD7+, CD56+, CD38‐, or HLA‐DR‐) 52 patients. 3‐year estimates of disease‐free survival (DFS) for MRD 0+, MRD 1+ and MRD 2+ patients were 59.5 ± 9.5%, 29.9 ± 6.1%, and 9.4 ± 5.1%, and 3‐year estimates of overall survival (OS) were 59.5 ± 9.5%, 34.5 ± 6.3%, and 14.5 ± 10.8%, respectively. Multivariate analysis adjusted for genetic risk, blast cell level, secondary disease, age, sex, and donor relationship pre‐HCT, the hazard ratios of abnormal expression of CD7+, CD56+, HLA‐DR−, and CD38‐ were 6.69 (range 2.08–21.52; p = 0.001) for DFS, 2.24 (range 1.21–4.14; p = 0.010) for OS, and 7.18 (range 2.23–23.10; p = 0.001) for relapse compared with CD7−, CD56−, HLA‐DR+, and CD38+ patients. Our finding suggested that abnormal expression of CD7+, CD56+, HLA‐DR−, and CD38− is associated with poor outcomes, and the more number of abnormal antigens expression predict worse outcomes.
Collapse
Affiliation(s)
- Qi Hao
- Department of Hematology, Aerospace Center Hospital, Beijing, China
| | - Xinyue Liu
- Department of Hematology, Aerospace Center Hospital, Beijing, China
| | - Yongping Zhang
- Department of Hematology, Aerospace Center Hospital, Beijing, China
| | - Dongmei Zhang
- Department of Hematology, Aerospace Center Hospital, Beijing, China
| | - Boran Li
- Department of Hematology, Aerospace Center Hospital, Beijing, China
| | - Jingbo Wang
- Department of Hematology, Aerospace Center Hospital, Beijing, China
| |
Collapse
|
20
|
Guolo F, Di Grazia C, Minetto P, Raiola AM, Clavio M, Miglino M, Tedone E, Contini P, Mangerini R, Kunkl A, Colombo N, Pugliese G, Carminati E, Marcolin R, Passannante M, Bagnasco S, Galaverna F, Lamparelli T, Ballerini F, Cagnetta A, Cea M, Gobbi M, Bacigalupo A, Lemoli RM, Angelucci E. Pre-transplant minimal residual disease assessment and transplant-related factors predict the outcome of acute myeloid leukemia patients undergoing allogeneic stem cell transplantation. Eur J Haematol 2021; 107:573-582. [PMID: 34297437 DOI: 10.1111/ejh.13694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/18/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022]
Abstract
We studied pretransplant minimal residual disease (MRD) in 224 patients (median age 44 years; range 17-65) with acute myeloid leukemia (AML) undergoing allogeneic stem cell transplant (HSCT) in complete remission. MRD was evaluated on marrow samples using multicolor flow cytometry and assessment of WT1 gene expression. Both methods showed a strong prognostic value and their combination allowed the identification of three groups of patients with different risk of relapse. In multivariate analysis, combined MRD was the only predictor of cumulative incidence of relapse, regardless of donor type, conditioning regimen, first or second CR at HSCT, HSCT year, and ELN risk group. Multivariate regression model showed that only negative combined MRD status (P < .001) and myeloablative conditioning (P = .004) were independently associated with better OS. Among MRD-positive patients, a reduced incidence of relapse was observed in patients receiving haplo transplant (P < .05) and in patients who showed grade II-IV aGVHD (P < .03). In patients with negative combined MRD, the intensity of conditioning regimen did not affect the overall favorable outcome. We suggest that pretransplant MRD evaluation combined with transplant-related factors can identify AML patients at higher risk for relapse and might help in defining the overall transplant strategy.
Collapse
Affiliation(s)
- Fabio Guolo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | | | - Paola Minetto
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | | | - Marino Clavio
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Maurizio Miglino
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | | | - Paola Contini
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | | | | | - Girolamo Pugliese
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Enrico Carminati
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Riccardo Marcolin
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Monica Passannante
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Samuele Bagnasco
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Federica Galaverna
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | | | - Filippo Ballerini
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Antonia Cagnetta
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Michele Cea
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Marco Gobbi
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | | | - Roberto Massimo Lemoli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | | |
Collapse
|
21
|
Shang L, Cai X, Sun W, Cheng Q, Mi Y. Time point-dependent concordance and prognostic significance of flow cytometry and real time quantitative PCR for measurable/minimal residual disease detection in acute myeloid leukemia with t(8;21)(q22;q22.1). CYTOMETRY PART B-CLINICAL CYTOMETRY 2021; 102:34-43. [PMID: 34232569 DOI: 10.1002/cyto.b.22028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Flow cytometry (FCM) and PCR are reliable methods for assessing minimal residual disease (MRD) in acute myeloid leukemia with t(8;21)(q22;q22.1). The aim of this study was to analyze the concordant rate of these two methods and their prognostic significance. METHODS PCR and FCM were simultaneously used for MRD analysis at four different time points on 450 BM samples from 124 patients with AML with t(8;21)(q22;q22.1). The four monitoring time points included post-induction (first), after the first consolidation (second) and the second consolidation (third), and at the end of chemotherapy or before Allo/Auto stem cell transplantation (fourth). RESULTS The concordant rates of the two methods were 33.06%, 25.81%, 49.59%, and 75.31%, respectively, and the main discordant cases were FCM-/PCR+ cases. At all monitoring time points, the MRD level ≥ 10-4 by FCM indicated a poor 3-year Relapse-Free Survival (RFS) (p < 0.001). More than 2-log MRD reduction by PCR after induction and more than 3-log reduction by PCR after the first consolidation remained the significant predictors of better RFS (p < 0.001). After the second consolidation, the negative MRD by PCR (<10-5) was also associated with improved RFS (p = 0.002). A > 1-log increase in PCR can effectively predict recurrence after molecular remission (p < 0.001). In the multivariate analysis, MRD≥0.01% by. FCM and less than 2-log MRD reduction by PCR after induction remained the significant predictors of poor RFS (p < 0.05). CONCLUSIONS FCM+ always indicates a poor prognosis. Sequential monitoring by PCR is of significance for evaluating prognosis. Our findings suggest a complementary role of two analyses in optimizing risk stratification in clinical practice.
Collapse
Affiliation(s)
- Lei Shang
- Department of Pathology and Lab Medicine, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Xiaojin Cai
- Department of Pathology and Lab Medicine, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Wanchen Sun
- Department of Pathology and Lab Medicine, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qingnian Cheng
- Department of Pathology and Lab Medicine, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yingchang Mi
- Department of Leukemia, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| |
Collapse
|
22
|
Abstract
Minimal or measurable residual disease (MRD) after therapy is the most important independent prognostic factor in acute myeloid leukemia. MRD measured by multiparametric flow cytometry and real-time quantitative polymerase chain reaction has been integrated into risk stratification and used to guide future treatment strategies. Recent technological advances have allowed the application of the novel molecular method, high-throughput sequencing, in MRD detection in clinical practice to improve sensitivity and specificity. Randomized studies are needed to address outstanding issues, including the optimal methods and timing of MRD testing and interlaboratory standardization to facilitate comparisons, to further improve MRD-directed interventions.
Collapse
Affiliation(s)
- Xueyan Chen
- Hematopathology, SCCA G7800, 825 Eastlake Ave E., Seattle, WA 98109, USA
| | - Sindhu Cherian
- Hematopathology, SCCA G7800, 825 Eastlake Ave E., Seattle, WA 98109, USA.
| |
Collapse
|
23
|
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: 0.7] [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
|
24
|
Liu Y, Wang G, Zhang J, Chen X, Xu H, Heng G, Chen J, Zhao Y, Li J, Ni Y, Zhang Y, Shan J, Qian C. CD9, a potential leukemia stem cell marker, regulates drug resistance and leukemia development in acute myeloid leukemia. Stem Cell Res Ther 2021; 12:86. [PMID: 33494824 PMCID: PMC7836575 DOI: 10.1186/s13287-021-02155-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 01/08/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Leukemia stem cells (LSCs) are responsible for the initiation, progression, and relapse of acute myeloid leukemia (AML). Therefore, a therapeutic strategy targeting LSCs is a potential approach to eradicate AML. In this study, we aimed to identify LSC-specific surface markers and uncover the underlying mechanism of AML LSCs. METHODS Microarray gene expression data were used to investigate candidate AML-LSC-specific markers. CD9 expression in AML cell lines, patients with AML, and normal donors was evaluated by flow cytometry (FC). The biological characteristics of CD9-positive (CD9+) cells were analyzed by in vitro proliferation, chemotherapeutic drug resistance, migration, and in vivo xenotransplantation assays. The molecular mechanism involved in CD9+ cell function was investigated by gene expression profiling. The effects of alpha-2-macroglobulin (A2M) on CD9+ cells were analyzed with regard to proliferation, drug resistance, and migration. RESULTS CD9, a cell surface protein, was specifically expressed on AML LSCs but barely detected on normal hematopoietic stem cells (HSCs). CD9+ cells exhibit more resistance to chemotherapy drugs and higher migration potential than do CD9-negative (CD9-) cells. More importantly, CD9+ cells possess the ability to reconstitute human AML in immunocompromised mice and promote leukemia growth, suggesting that CD9+ cells define the LSC population. Furthermore, we identified that A2M plays a crucial role in maintaining CD9+ LSC stemness. Knockdown of A2M impairs drug resistance and migration of CD9+ cells. CONCLUSION Our findings suggest that CD9 is a new biomarker of AML LSCs and is a promising therapeutic target.
Collapse
Affiliation(s)
- Yongliang Liu
- Center of Biological Therapy, Southwest Hospital, Army Medical University, Chongqing, China.,Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Guiqin Wang
- Center of Biological Therapy, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jiasi Zhang
- Department of Hematology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xue Chen
- Department of Hematology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Huailong Xu
- Center of Biological Therapy, Southwest Hospital, Army Medical University, Chongqing, China
| | - Gang Heng
- Center of Biological Therapy, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jun Chen
- Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Yongchun Zhao
- Chongqing Institute of Precision Medicine and Biotechnology Co., Ltd., Chongqing, China
| | - Jiatao Li
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Yuanli Ni
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Yingzi Zhang
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Juanjuan Shan
- Center of Biological Therapy, Southwest Hospital, Army Medical University, Chongqing, China. .,Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China.
| | - Cheng Qian
- Center of Biological Therapy, Southwest Hospital, Army Medical University, Chongqing, China. .,Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
25
|
Naidoo M, Gibbs P, Tie J. ctDNA and Adjuvant Therapy for Colorectal Cancer: Time to Re-Invent Our Treatment Paradigm. Cancers (Basel) 2021; 13:346. [PMID: 33477814 PMCID: PMC7832902 DOI: 10.3390/cancers13020346] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths worldwide. While there have been significant developments in the treatments for patients with metastatic CRC in recent years, improving outcomes in the adjuvant setting has been more challenging. Recent technological advances in circulating tumour DNA (ctDNA) assay with the ability to detect minimal residual disease (MRD) after curative intent surgery will fundamentally change how we assess recurrence risk and conduct adjuvant trials. Studies in non-metastatic CRC have now demonstrated the prognostic impact of ctDNA analysis after curative intent surgery over and above current standard of care clinicopathological criteria. This ability of ctDNA analysis to stratify patients into low- and very-high-risk groups provides a window of opportunity to personalise adjuvant treatment where escalation/de-escalation of adjuvant systemic therapy could potentially increase cure rates and also reduce treatment-related physical and financial toxicity. Emerging data suggest that conversion of ctDNA from detectable to undetectable after adjuvant chemotherapy may reflect treatment efficacy. This real-time assessment of treatment benefit could be used as a surrogate endpoint for adjuvant novel drug development. Several ctDNA-based randomized adjuvant trials are ongoing internationally to confirm the clinical utility of ctDNA in colorectal cancer.
Collapse
Affiliation(s)
- Mahendra Naidoo
- Peter MacCallum Cancer Centre, Department of Medical Oncology, Melbourne, VIC 3000, Australia;
| | - Peter Gibbs
- Division of Personalised Oncology, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia;
- Western Health, Department of Medical Oncology, Melbourne, VIC 3021, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Jeanne Tie
- Peter MacCallum Cancer Centre, Department of Medical Oncology, Melbourne, VIC 3000, Australia;
- Division of Personalised Oncology, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia;
- Western Health, Department of Medical Oncology, Melbourne, VIC 3021, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
26
|
Deak D, Gorcea-Andronic N, Sas V, Teodorescu P, Constantinescu C, Iluta S, Pasca S, Hotea I, Turcas C, Moisoiu V, Zimta AA, Galdean S, Steinheber J, Rus I, Rauch S, Richlitzki C, Munteanu R, Jurj A, Petrushev B, Selicean C, Marian M, Soritau O, Andries A, Roman A, Dima D, Tanase A, Sigurjonsson O, Tomuleasa C. A narrative review of central nervous system involvement in acute leukemias. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:68. [PMID: 33553361 PMCID: PMC7859772 DOI: 10.21037/atm-20-3140] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Acute leukemias (both myeloid and lymphoblastic) are a group of diseases for which each year more successful therapies are implemented. However, in a subset of cases the overall survival (OS) is still exceptionally low due to the infiltration of leukemic cells in the central nervous system (CNS) and the subsequent formation of brain tumors. The CNS involvement is more common in acute lymphocytic leukemia (ALL), than in adult acute myeloid leukemia (AML), although the rates for the second case might be underestimated. The main reasons for CNS invasion are related to the expression of specific adhesion molecules (VLA-4, ICAM-1, VCAM, L-selectin, PECAM-1, CD18, LFA-1, CD58, CD44, CXCL12) by a subpopulation of leukemic cells, called “sticky cells” which have the ability to interact and adhere to endothelial cells. Moreover, the microenvironment becomes hypoxic and together with secretion of VEGF-A by ALL or AML cells the permeability of vasculature in the bone marrow increases, coupled with the disruption of blood brain barrier. There is a single subpopulation of leukemia cells, called leukemia stem cells (LSCs) that is able to resist in the new microenvironment due to its high adaptability. The LCSs enter into the arachnoid, migrate, and intensively proliferate in cerebrospinal fluid (CSF) and consequently infiltrate perivascular spaces and brain parenchyma. Moreover, the CNS is an immune privileged site that also protects leukemic cells from chemotherapy. CD56/NCAM is the most important surface molecule often overexpressed by leukemic stem cells that offers them the ability to infiltrate in the CNS. Although asymptomatic or with unspecific symptoms, CNS leukemia should be assessed in both AML/ALL patients, through a combination of flow cytometry and cytological analysis of CSF. Intrathecal therapy (ITT) is a preventive measure for CNS involvement in AML and ALL, still much research is needed in finding the appropriate target that would dramatically lower CNS involvement in acute leukemia.
Collapse
Affiliation(s)
- Dalma Deak
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Nicolae Gorcea-Andronic
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Valentina Sas
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Pediatrics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Patric Teodorescu
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Catalin Constantinescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Intensive Care Unit, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Sabina Iluta
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Sergiu Pasca
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ionut Hotea
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Turcas
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Vlad Moisoiu
- Department of Neurosurgery, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Simona Galdean
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Jakob Steinheber
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Rus
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Sebastian Rauch
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cedric Richlitzki
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Bobe Petrushev
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Selicean
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Mirela Marian
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Olga Soritau
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Alexandra Andries
- Department of Radiology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Andrei Roman
- Department of Radiology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Radiology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Alina Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | | | - Ciprian Tomuleasa
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
27
|
Del Principe MI, Gatti A, Johansson U, Buccisano F, Brando B. ESCCA
/
ISCCA
protocol for the analysis of cerebrospinal fluid by multiparametric flow‐cytometry in hematological malignancies. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 100:269-281. [DOI: 10.1002/cyto.b.21981] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 10/14/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023]
Affiliation(s)
| | - Arianna Gatti
- Blood Transfusion Center Legnano General Hospital Legnano Italy
| | - Ulrika Johansson
- SI‐HMDS University Hospitals Bristol and Weston NHS Foundation Trust Bristol United Kingdom
| | - Francesco Buccisano
- Hematology, Department of BioMedicine and Prevention University of Rome “Tor Vergata” Rome Italy
| | - Bruno Brando
- Blood Transfusion Center Legnano General Hospital Legnano Italy
| |
Collapse
|
28
|
Shen H, Zhao Y, Shi Y, Sun J, Zhou D, Li L, Ye X, Xie W. The diagnostic and prognostic value of MRI in central nervous system involvement of acute myeloid leukemia: a retrospective cohort of 84 patients. ACTA ACUST UNITED AC 2020; 25:258-263. [PMID: 32567523 DOI: 10.1080/16078454.2020.1781500] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
ABSTRACT Objective: To assess the diagnostic and prognostic value of magnetic resonance imaging (MRI) in Acute Myeloid Leukemia (AML) complicated with central nervous system leukemia (CNSL). Methods: A total of 84 patients with AML and confirmed of CNSL from January 2010 to September 2019 were selected and underwent MRI scan. We retrospectively analyzed their MRI findings, summarized the imaging features of AML central infiltration, and assessed the guiding significance of MRI on diagnosis and prognosis of this disease. Results: A total of 52 patients (61.90%, 52/84) had abnormal MRI findings, of which 31 cases clearly indicated intracranial infiltration of leukemia. Among the 31 patients, the most common site of infiltration is parenchyma (19/31). Most MRI of these patients showed multiple lesions with low T1 signal and high T2 signal, which were more obvious on enhanced scan. Sensitivity of MRI in diagnosing AML central infiltration was 36.90%. Despite of its low sensitivity, it still had superior diagnostic value on some patients with false-negative CSF. The median disease-free survival (DFS) and overall survival (OS) time of patients with MRI clearly indicated central invasion were 4 and 9 months, respectively. But there was no significant difference in survival analysis compared with MRI negative patients (including abnormal but non-invasive). Conclusion: MRI manifestation of central infiltration in AML patients has certain characteristic findings, which is helpful to improve the diagnostic efficiency. Prognosis of MRI positive patients is relatively worse than that of MRI negative patients however there is no siginificant difference.
Collapse
Affiliation(s)
- Huafei Shen
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, People's Republic of China
| | - Yanchun Zhao
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, People's Republic of China
| | - Yuanfei Shi
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, People's Republic of China
| | - Jianai Sun
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, People's Republic of China
| | - De Zhou
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, People's Republic of China
| | - Li Li
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, People's Republic of China
| | - Xiujin Ye
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, People's Republic of China
| | - Wanzhuo Xie
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
29
|
Rotchanapanya W, Hokland P, Tunsing P, Owattanapanich W. Clinical Outcomes Based on Measurable Residual Disease Status in Patients with Core-Binding Factor Acute Myeloid Leukemia: A Systematic Review and Meta-Analysis. J Pers Med 2020; 10:jpm10040250. [PMID: 33256157 PMCID: PMC7711894 DOI: 10.3390/jpm10040250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 01/11/2023] Open
Abstract
Measurable residual disease (MRD) response during acute myeloid leukemia (AML) treatment is a gold standard for determining treatment strategy, especially in core-binding factor (CBL) AML. The aim of this study was to critically review the literature on MRD status in the CBF-AML to determine the overall impact of MRD status on clinical outcomes. Published studies in the MEDLINE and EMBASE databases from their inception up to 1 June 2019 were searched. The primary end-point was either overall survival (OS) or recurrence-free survival (RFS) between MRD negative and MRD positive CBF-AML patients. The secondary variable was cumulative incidence of relapse (CIR) between groups. Of the 736 articles, 13 relevant studies were included in this meta-analysis. The MRD negative group displayed more favorable recurrence-free survival (RFS) than those with MRD positivity, with a pooled odds ratio (OR) of 4.5. Moreover, OS was also superior in the MRD negative group, with a pooled OR of 7.88. Corroborating this, the CIR was statistically significantly lower in the MRD negative group, with a pooled OR of 0.06. The most common cutoff MRD level was 1 × 10−3. These results suggest that MRD assessment should be a routine investigation in clinical practice in this AML subset.
Collapse
Affiliation(s)
- Wannaphorn Rotchanapanya
- Division of Hematology, Department of Medicine, Chiangrai Prachanukroh Hospital, Chiang Rai 57000, Thailand;
| | - Peter Hokland
- Division of Hematology, Department of Clinical Medicine, Aarhus University Hospital, 8200 Aarhus N, Denmark;
| | - Pattaraporn Tunsing
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Weerapat Owattanapanich
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
- Correspondence: ; Tel.: +66-2419-4448
| |
Collapse
|
30
|
Aguirre-Ruiz P, Ariceta B, Viguria MC, Zudaire MT, Blasco-Iturri Z, Arnedo P, Aguilera-Diaz A, Jauregui A, Mañú A, Prosper F, Mateos MC, Fernández-Mercado M, Larráyoz MJ, Redondo M, Calasanz MJ, Vázquez I, Bandrés E. Assessment of Minimal Residual Disease by Next Generation Sequencing in Peripheral Blood as a Complementary Tool for Personalized Transplant Monitoring in Myeloid Neoplasms. J Clin Med 2020; 9:jcm9123818. [PMID: 33255857 PMCID: PMC7760908 DOI: 10.3390/jcm9123818] [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: 11/02/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/30/2022] Open
Abstract
Patients with myeloid neoplasms who relapsed after allogenic hematopoietic stem cell transplant (HSCT) have poor prognosis. Monitoring of chimerism and specific molecular markers as a surrogate measure of relapse is not always helpful; therefore, improved systems to detect early relapse are needed. We hypothesized that the use of next generation sequencing (NGS) could be a suitable approach for personalized follow-up post-HSCT. To validate our hypothesis, we analyzed by NGS, a retrospective set of peripheral blood (PB) DNA samples previously evaluated by high-sensitive quantitative PCR analysis using insertion/deletion polymorphisms (indel-qPCR) chimerism engraftment. Post-HCST allelic burdens assessed by NGS and chimerism status showed a similar time-course pattern. At time of clinical relapse in 8/12 patients, we detected positive NGS-based minimal residual disease (NGS-MRD). Importantly, in 6/8 patients, we were able to detect NGS-MRD at time points collected prior to clinical relapse. We also confirmed the disappearance of post-HCST allelic burden in non-relapsed patients, indicating true clinical specificity. This study highlights the clinical utility of NGS-based post-HCST monitoring in myeloid neoplasia as a complementary specific analysis to high-sensitive engraftment testing. Overall, NGS-MRD testing in PB is widely applicable for the evaluation of patients following HSCT and highly valuable to personalized early treatment intervention when mixed chimerism is detected.
Collapse
Affiliation(s)
- Paula Aguirre-Ruiz
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, 31008 Pamplona, Navarra, Spain; (P.A.-R.); (B.A.); (Z.B.-I.); (A.M.); (M.F.-M.); (M.J.L.); (M.J.C.)
| | - Beñat Ariceta
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, 31008 Pamplona, Navarra, Spain; (P.A.-R.); (B.A.); (Z.B.-I.); (A.M.); (M.F.-M.); (M.J.L.); (M.J.C.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain; (M.C.V.); (M.T.Z.); (A.A.-D.); (F.P.); (M.C.M.); (M.R.)
| | - María Cruz Viguria
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain; (M.C.V.); (M.T.Z.); (A.A.-D.); (F.P.); (M.C.M.); (M.R.)
- Hematology Department, Complejo Hospitalario de Navarra, 31008 Pamplona, Navarra, Spain; (P.A.); (A.J.)
| | - María Teresa Zudaire
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain; (M.C.V.); (M.T.Z.); (A.A.-D.); (F.P.); (M.C.M.); (M.R.)
- Hematology Department, Complejo Hospitalario de Navarra, 31008 Pamplona, Navarra, Spain; (P.A.); (A.J.)
| | - Zuriñe Blasco-Iturri
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, 31008 Pamplona, Navarra, Spain; (P.A.-R.); (B.A.); (Z.B.-I.); (A.M.); (M.F.-M.); (M.J.L.); (M.J.C.)
| | - Patricia Arnedo
- Hematology Department, Complejo Hospitalario de Navarra, 31008 Pamplona, Navarra, Spain; (P.A.); (A.J.)
| | - Almudena Aguilera-Diaz
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain; (M.C.V.); (M.T.Z.); (A.A.-D.); (F.P.); (M.C.M.); (M.R.)
- Advanced Genomics Laboratory, Hemato-Oncology, Center for Applied Medical Research (CIMA), 31008 Pamplona, Navarra, Spain
| | - Axier Jauregui
- Hematology Department, Complejo Hospitalario de Navarra, 31008 Pamplona, Navarra, Spain; (P.A.); (A.J.)
| | - Amagoia Mañú
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, 31008 Pamplona, Navarra, Spain; (P.A.-R.); (B.A.); (Z.B.-I.); (A.M.); (M.F.-M.); (M.J.L.); (M.J.C.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain; (M.C.V.); (M.T.Z.); (A.A.-D.); (F.P.); (M.C.M.); (M.R.)
| | - Felipe Prosper
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain; (M.C.V.); (M.T.Z.); (A.A.-D.); (F.P.); (M.C.M.); (M.R.)
- Advanced Genomics Laboratory, Hemato-Oncology, Center for Applied Medical Research (CIMA), 31008 Pamplona, Navarra, Spain
- Hematology Department, Clinica Universidad de Navarra (CUN), 31008 Pamplona, Navarra, Spain
| | - María Carmen Mateos
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain; (M.C.V.); (M.T.Z.); (A.A.-D.); (F.P.); (M.C.M.); (M.R.)
- Hematology Department, Complejo Hospitalario de Navarra, 31008 Pamplona, Navarra, Spain; (P.A.); (A.J.)
| | - Marta Fernández-Mercado
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, 31008 Pamplona, Navarra, Spain; (P.A.-R.); (B.A.); (Z.B.-I.); (A.M.); (M.F.-M.); (M.J.L.); (M.J.C.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain; (M.C.V.); (M.T.Z.); (A.A.-D.); (F.P.); (M.C.M.); (M.R.)
- Advanced Genomics Laboratory, Hemato-Oncology, Center for Applied Medical Research (CIMA), 31008 Pamplona, Navarra, Spain
| | - María José Larráyoz
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, 31008 Pamplona, Navarra, Spain; (P.A.-R.); (B.A.); (Z.B.-I.); (A.M.); (M.F.-M.); (M.J.L.); (M.J.C.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain; (M.C.V.); (M.T.Z.); (A.A.-D.); (F.P.); (M.C.M.); (M.R.)
| | - Margarita Redondo
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain; (M.C.V.); (M.T.Z.); (A.A.-D.); (F.P.); (M.C.M.); (M.R.)
- Hematology Department, Complejo Hospitalario de Navarra, 31008 Pamplona, Navarra, Spain; (P.A.); (A.J.)
| | - María José Calasanz
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, 31008 Pamplona, Navarra, Spain; (P.A.-R.); (B.A.); (Z.B.-I.); (A.M.); (M.F.-M.); (M.J.L.); (M.J.C.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain; (M.C.V.); (M.T.Z.); (A.A.-D.); (F.P.); (M.C.M.); (M.R.)
| | - Iria Vázquez
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, 31008 Pamplona, Navarra, Spain; (P.A.-R.); (B.A.); (Z.B.-I.); (A.M.); (M.F.-M.); (M.J.L.); (M.J.C.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain; (M.C.V.); (M.T.Z.); (A.A.-D.); (F.P.); (M.C.M.); (M.R.)
- Correspondence: (I.V.); (E.B.); Tel.: +34-948194700-1000 (I.V.)
| | - Eva Bandrés
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain; (M.C.V.); (M.T.Z.); (A.A.-D.); (F.P.); (M.C.M.); (M.R.)
- Hematology Department, Complejo Hospitalario de Navarra, 31008 Pamplona, Navarra, Spain; (P.A.); (A.J.)
- Correspondence: (I.V.); (E.B.); Tel.: +34-948194700-1000 (I.V.)
| |
Collapse
|
31
|
Gaut D, Mead M. Measurable residual disease in hematopoietic stem cell transplantation-eligible patients with acute myeloid leukemia: clinical significance and promising therapeutic strategies. Leuk Lymphoma 2020; 62:8-31. [DOI: 10.1080/10428194.2020.1827251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Daria Gaut
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Monica Mead
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
32
|
Guolo F, Fianchi L, Minetto P, Clavio M, Gottardi M, Galimberti S, Rizzuto G, Rondoni M, Bertani G, Dargenio M, Bilio A, Scappini B, Zappasodi P, Scattolin AM, Grimaldi F, Pietrantuono G, Musto P, Cerrano M, D'Ardia S, Audisio E, Cignetti A, Pasciolla C, Pavesi F, Candoni A, Gurreri C, Morselli M, Alati C, Fracchiolla N, Rossi G, Caizzi M, Carnevale-Schianca F, Tafuri A, Rossi G, Ferrara F, Pagano L, Lemoli RM. CPX-351 treatment in secondary acute myeloblastic leukemia is effective and improves the feasibility of allogeneic stem cell transplantation: results of the Italian compassionate use program. Blood Cancer J 2020; 10:96. [PMID: 33024084 PMCID: PMC7538937 DOI: 10.1038/s41408-020-00361-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/19/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023] Open
Abstract
Secondary acute myeloid leukemia (sAML) poorly responds to conventional treatments and allogeneic stem cell transplantation (HSCT). We evaluated toxicity and efficacy of CPX-351 in 71 elderly patients (median age 66 years) with sAML enrolled in the Italian Named (Compassionate) Use Program. Sixty days treatment-related mortality was 7% (5/71). The response rate at the end of treatment was: CR/CRi in 50/71 patients (70.4%), PR in 6/71 (8.5%), and NR in 10/71 (19.7%). After a median follow-up of 11 months relapse was observed in 10/50 patients (20%) and 12 months cumulative incidence of relapse (CIR) was 23.6%. Median duration of response was not reached. In competing risk analysis, CIR was reduced when HSCT was performed in first CR (12 months CIR of 5% and 37.4%, respectively, for patients receiving (=20) or not (=30) HSCT, p = 0.012). Twelve-months OS was 68.6% (median not reached). In landmark analysis, HSCT in CR1 was the only significant predictor of longer survival (12 months OS of 100 and 70.5%, for patients undergoing or not HSCT in CR1, respectively, p = 0.011). In conclusion, we extend to a real-life setting, the notion that CPX is an effective regimen for high risk AML patients and may improve the results of HSCT.
Collapse
Affiliation(s)
- Fabio Guolo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy. .,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy.
| | - Luana Fianchi
- Istituto di Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Paola Minetto
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Marino Clavio
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | | | - Sara Galimberti
- UO Ematologia, Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Giuliana Rizzuto
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Michela Rondoni
- U.O.C. di Ematologia, Azienda Unità Sanitaria Locale della Romagna, Ravenna, Italy
| | - Giambattista Bertani
- S.C. Ematologia, ASST Grande Ospedale Metropolitano, Niguarda Ca' Granda Milano, Milan, Italy
| | | | | | - Barbara Scappini
- Dipartimento di Oncologia-SODc Ematologia, Azienda Ospedaliero - Universitaria Careggi, Florence, Italy
| | - Patrizia Zappasodi
- Clinica Ematologica, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Francesco Grimaldi
- Dipartimento di Medicina Clinica e Chirurgia, AOU Federico II di Napoli, Naples, Italy
| | | | - Pellegrino Musto
- IRCCS Centro Oncologico della Basilicata, Rionero in Vulture, Potenza, Italy.,Unit of Hematology and Stem Cell Transplantation, AOU Policlinico Consorziale, "Aldo Moro" University, Bari, Italy
| | - Marco Cerrano
- Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, Turin, Italy
| | - Stefano D'Ardia
- Institute for Cancer Research and Treatment, University of Turin-School of Medicine, Turin, Italy
| | - Ernesta Audisio
- S.C. Ematologia2, Dipartimento di Ematologia e Oncologia, AO Città della Salute e della Scienza di Torino, Turin, Italy
| | - Alessandro Cignetti
- Divisione Universitaria di Ematologia e Terapie Cellulari, A.O. Ordine Mauriziano, Turin, Italy
| | | | - Francesca Pavesi
- Hematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Anna Candoni
- Clinica Ematologica, Centro Trapianti e Terapie Cellulari, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | - Carmela Gurreri
- U.O. Ematologia ed Immunologia Clinica, Azienda Ospedaliera di Padova, Padova, Italy
| | - Monica Morselli
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Modena, Italy
| | | | - Nicola Fracchiolla
- Oncoematologia, IRCCS Ca' Granda Ospedale Maggiore Policlinico and University of Milan, Milan, Italy
| | - Giovanni Rossi
- U.O. Ematologia, Casa Sollievo della Sofferenza IRCCS, San Giovanni Rotondo, Foggia, Italy
| | - Manuela Caizzi
- S.C. Ematologia Azienda Sanitaria Universitaria Integrata di Trieste, Ospedale Maggiore, Trieste, Italy
| | - Fabrizio Carnevale-Schianca
- Medical Oncology, Hematopoietic Stem Cells Unit, Turin Metropolitan Transplant Center, Candiolo Cancer Institute-FPO, IRCCS, Candiolo, Italy
| | - Agostino Tafuri
- Department of Clinical and Molecular Medicine & Hematology, Sant'Andrea - University Hospital - Sapienza - University of Rome, Rome, Italy
| | - Giuseppe Rossi
- SC Ematologia e Dipartimento di Oncologia Clinica, A.O. Spedali Civili, Brescia, Italy
| | | | - Livio Pagano
- Istituto di Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Roberto Massimo Lemoli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| |
Collapse
|
33
|
Daga S, Rosenberger A, Kashofer K, Heitzer E, Quehenberger F, Halbwedl I, Graf R, Krisper N, Prietl B, Höfler G, Reinisch A, Zebisch A, Sill H, Wölfler A. Sensitive and broadly applicable residual disease detection in acute myeloid leukemia using flow cytometry-based leukemic cell enrichment followed by mutational profiling. Am J Hematol 2020; 95:1148-1157. [PMID: 32602117 PMCID: PMC7540028 DOI: 10.1002/ajh.25918] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 12/19/2022]
Abstract
Persistent measurable residual disease (MRD) is an increasingly important prognostic marker in acute myeloid leukemia (AML). Currently, MRD is determined by multi-parameter flow cytometry (MFC) or PCR-based methods detecting leukemia-specific fusion transcripts and mutations. However, while MFC is highly operator-dependent and difficult to standardize, PCR-based methods are only available for a minority of AML patients. Here we describe a novel, highly sensitive and broadly applicable method for MRD detection by combining MFC-based leukemic cell enrichment using an optimized combinatorial antibody panel targeting CLL-1, TIM-3, CD123 and CD117, followed by mutational analysis of recurrently mutated genes in AML. In dilution experiments this method showed a sensitivity of 10-4 to 10-5 for residual disease detection. In prospectively collected remission samples this marker combination allowed for a median 67-fold cell enrichment with sufficient DNA quality for mutational analysis using next generation sequencing (NGS) or digital PCR in 39 out of 41 patients. Twenty-one samples (53.8%) tested MRD positive, whereas 18 (46.2%) were negative. With a median follow-up of 559 days, 71.4% of MRD positive (15/21) and 27.8% (5/18) of MRD negative patients relapsed (P = .007). The cumulative incidence of relapse (CIR) was higher for MRD positive patients (5-year CIR: 90.5% vs 28%, P < .001). In multivariate analysis, MRD positivity was a prominent factor for CIR. Thus, MFC-based leukemic cell enrichment using antibodies against CLL-1, TIM-3, CD123 and CD117 followed by mutational analysis allows high sensitive MRD detection and is informative on relapse risk in the majority of AML patients.
Collapse
Affiliation(s)
- Shruti Daga
- Division of HematologyMedical University of GrazGrazAustria
- CBmed Center of Biomarker Research in MedicineGrazAustria
| | | | - Karl Kashofer
- Division of PathologyMedical University of GrazGrazAustria
| | - Ellen Heitzer
- Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Franz Quehenberger
- Institute of Medical InformaticsStatistics and Documentation, Medical University of GrazGrazAustria
| | - Iris Halbwedl
- Division of PathologyMedical University of GrazGrazAustria
| | - Ricarda Graf
- Institute of Human GeneticsMedical University of GrazGrazAustria
| | - Nina Krisper
- CBmed Center of Biomarker Research in MedicineGrazAustria
| | - Barbara Prietl
- CBmed Center of Biomarker Research in MedicineGrazAustria
| | - Gerald Höfler
- Division of PathologyMedical University of GrazGrazAustria
| | | | - Armin Zebisch
- Division of HematologyMedical University of GrazGrazAustria
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of PharmacologyMedical University of GrazGrazAustria
| | - Heinz Sill
- Division of HematologyMedical University of GrazGrazAustria
| | - Albert Wölfler
- Division of HematologyMedical University of GrazGrazAustria
- CBmed Center of Biomarker Research in MedicineGrazAustria
| |
Collapse
|
34
|
DeWolf S, Tallman MS. How I treat relapsed or refractory AML. Blood 2020; 136:1023-1032. [PMID: 32518943 PMCID: PMC7453152 DOI: 10.1182/blood.2019001982] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 04/28/2020] [Indexed: 12/15/2022] Open
Abstract
Treatment of relapsed or refractory acute myeloid leukemia (AML) has presented challenges for hematologists for decades. Despite numerous clinical studies, outcomes are consistently disappointing with 5-year overall survival rates of ∼10%. Allogeneic hematopoietic cell transplantation at the time of second complete remission remains the only reliable option with curative potential. However, recent approval of several new agents has transformed treatment paradigms that had been in place for almost half a century in AML. This new therapeutic landscape provides the opportunity to revisit the approach to relapsed or refractory AML. Through illustrative cases, we describe our approach, which increasingly relies on specific disease biology. We focus on treatment outside of the context of clinical trials because such trials are not available in most parts of the world. Primarily, we consider age, fitness to tolerate intensive chemotherapy, remission duration, and presence of a targetable mutation to guide treatment. The coming years will inevitably bring new targets and agents that may prove most effective when combined with each other and/or chemotherapy. Future studies are needed to determine how best to implement this evolving armamentarium of treatment options, to elucidate mechanisms of resistance, and to continue the pursuit of novel drug discovery.
Collapse
Affiliation(s)
- Susan DeWolf
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; and
| | - Martin S Tallman
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; and
- Leukemia Service, Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
35
|
Multidimensional study of the heterogeneity of leukemia cells in t(8;21) acute myelogenous leukemia identifies the subtype with poor outcome. Proc Natl Acad Sci U S A 2020; 117:20117-20126. [PMID: 32747558 DOI: 10.1073/pnas.2003900117] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
t(8;21)(q22;q22) acute myelogenous leukemia (AML) is morphologically characterized by a continuum of heterogeneous leukemia cells from myeloblasts to differentiated myeloid elements. Thus, t(8;21) AML is an excellent model for studying heterogeneous cell populations and cellular evolution during disease progression. Using integrative analyses of immunophenotype, RNA-sequencing (RNA-seq), and single-cell RNA-sequencing (scRNA-seq), we identified three distinct intrapatient leukemic cell populations that were arrested at different stages of myeloid differentiation: CD34+CD117dim blasts, CD34+CD117bri blasts, and abnormal myeloid cells with partial maturation (AM). CD117 is also known as c-KIT protein. CD34+CD117dim cells were blocked in the G0/G1 phase at disease onset, presenting with the regular morphology of myeloblasts showing features of granulocyte-monocyte progenitors (GMP), and were drug-resistant to chemotherapy. Genes associated with cell migration and adhesion (LGALS1, EMP3, and ANXA 2) were highly expressed in the CD34+CD117dim population. CD34+CD117bri blasts were blocked a bit later than the CD34+CD117dim population in the hematopoietic differentiation stage and displayed high proliferation ability. AM cells, which bear abnormal myelocyte morphology, especially overexpressed granule genes AZU1, ELANE, and PRTN3 and were sensitive to chemotherapy. scRNA-seq at different time points identified CD34+CD117dim blasts as an important leukemic cluster that expanded at postrelapse refractory stage after several cycles of chemotherapy. Patients with t(8;21) AML with a higher proportion of CD34+CD117dim cells had significantly worse clinical outcomes than those with a lower CD34+CD117dim proportion. Univariate and multivariate analyses identified CD34+CD117dim proportion as an independent factor for poor disease outcome. Our study provides evidence for the multidimensional heterogeneity of t(8;21)AML and may offer new tools for future disease stratification.
Collapse
|
36
|
Venditti A, Gale RP, Buccisano F, Ossenkoppele G. Should persons with acute myeloid leukemia (AML) in 1st histological complete remission who are measurable residual disease (MRD) test positive receive an allotransplant? Leukemia 2020; 34:963-965. [PMID: 32132654 DOI: 10.1038/s41375-020-0780-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/01/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Adriano Venditti
- Department of Biomedicine and Prevention at University "Tor Vergata", Fondazione Policlinico Tor Vergata, Rome, Italy.
| | - Robert Peter Gale
- Haematology Research Centre, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Francesco Buccisano
- Department of Biomedicine and Prevention at University "Tor Vergata", Fondazione Policlinico Tor Vergata, Rome, Italy
| | - Gert Ossenkoppele
- Department of Hematology, VU University Medical Centre Amsterdam, Amsterdam, Noord-Holland, The Netherlands
| |
Collapse
|
37
|
Wang J, Lu R, Wu Y, Jia J, Gong L, Liu X, Lu S, Wang Y, Yan C, Liu K, Zhang X, Xu L, Jiang Q, Zhao X, Shi H, Lai Y, Huang X, Ruan G, Jiang H. Detection of measurable residual disease may better predict outcomes than mutations based on next-generation sequencing in acute myeloid leukaemia with biallelic mutations of CEBPA. Br J Haematol 2020; 190:533-544. [PMID: 32090321 DOI: 10.1111/bjh.16535] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 01/18/2020] [Indexed: 11/27/2022]
Abstract
Acute myeloid leukaemia (AML) patients with biallelic mutations of CEBPA (bi CEBPA) have a 30-50% relapse rate. This study established the value of mutations based on next-generation sequencing (NGS) and multiparameter flow cytometric measurable residual disease (MFC-MRD) detection and compared the outcomes. From 2014 to 2018, 124 newly diagnosed bi CEBPA AML patients were treated. The median age was 37·5 (16-69) years. The 3-year cumulative incidence of relapse (CIR), relapse-free survival (RFS) and overall survival (OS) were 33·0%, 64·7% and 84·3%, respectively. Patients without additional mutations and with GATA2 mutations were defined as 'NGS low risk', which was the only favourable independent factor for CIR and RFS of pretreatment parameters. Patients with sustained positive MRD after two consolidation cycles and MRD negative losses at any time were defined as 'MRD high risk', which was the only poor independent factor for CIR, RFS and OS, including pretreatment and post-treatment parameters. In CR2 and non-remission patients who underwent allo-HSCT, superior OS was achieved. We conclude that NGS low risk was a favourable factor in the analysis of pretreatment parameters. MRD risk stratification was an independent prognostic factor in pretreatment and post-treatment parameters. Relapsed patients still have a favourable outcome followed by allo-HSCT.
Collapse
Affiliation(s)
- Jing Wang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - RunQing Lu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - Ying Wu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - JinSong Jia
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - Lizhong Gong
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - XiaoHong Liu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - ShengYe Lu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - Yu Wang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - ChenHua Yan
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China.,Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
| | - KaiYan Liu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - XiaoHui Zhang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - LanPing Xu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China.,Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
| | - Qian Jiang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - XiaoSu Zhao
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China.,Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
| | - HongXia Shi
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - YueYun Lai
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - XiaoJun Huang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Beijing, China.,Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
| | - GuoRui Ruan
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| | - Hao Jiang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Beijing, China
| |
Collapse
|
38
|
Bussaglia E, Pratcorona M, Carricondo M, Sansegundo L, Rubio MA, Monter A, Brell A, Badell I, Esteve J, Arnan M, Talarn C, Tormo M, García A, Vall-Llovera F, Ortin X, Pedro C, Bargay J, Brunet S, Sierra J, Nomdedéu J. Application of a digital PCR method for WT1 to myeloid neoplasms in CR and deep ELN WT1 molecular response (< 10 copies). Ann Hematol 2020; 99:765-772. [PMID: 32062741 DOI: 10.1007/s00277-020-03910-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/13/2020] [Indexed: 11/30/2022]
Abstract
Bone marrow WT1 mRNA levels assessed by the ELN method are useful to establish prognostic correlations in myeloid malignancies treated with chemotherapy or hematopoietic stem cell transplantation (HCT). Those patients with WT1 levels below ten copies have a good outcome. However, some of these patients relapse. To further characterize this group of cases, we applied a new and sensitive digital (ddPCR) WT1 method. A consecutive series of 49 patients with treated myeloid malignancies and with an ELN WT1 quantitation of < 10 copies were included in the study. All cases (47 AML and 2 MDS) have received intensive chemotherapy or HCT. One to four micrograms of total RNA were retrotranscribed to obtain ≥ 10,000 ABL1 copies using the ELN protocol. Only those cases with a good quality cDNA were used in the ddPCR WT1 test. The ddPCR Gene Expression WT1 Assay of Bio-Rad© was used to perform the PCR amplification, and the microdroplets were quantified in the Bio-Rad's QX200 droplet reader. Eighteen patients showed a negative WT1 ddPCR assay (0 copies/μl), whereas 31 cases were positive (results ranged from 1 to 15.2 copies/μl). Survival analysis showed statistically significant differences in terms of OS between both groups, 83 ± 8% vs. 46 ± 9% (p = 0.024). A statistically significant correlation was also found between ddPCRWT1 results and CD123+ cell number detected by flow cytometry (p = 0.024). Larger series of patients tested with the current ddPCRWT1 method will solve whether it could be used to stratify patients with myeloid malignancies achieving deep WT1 molecular response (< 10 copies).
Collapse
Affiliation(s)
- E Bussaglia
- Hematology Lab, Hematology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Mas Casanovas, 9008041, Barcelona, Spain.,Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - M Pratcorona
- Hematology Lab, Hematology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Mas Casanovas, 9008041, Barcelona, Spain.,Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - M Carricondo
- Hematology Lab, Hematology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Mas Casanovas, 9008041, Barcelona, Spain.,Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - L Sansegundo
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Universitat de Barcelona, Barcelona, Spain
| | - M A Rubio
- Hematology Lab, Hematology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Mas Casanovas, 9008041, Barcelona, Spain.,Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - A Monter
- Hematology Lab, Hematology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Mas Casanovas, 9008041, Barcelona, Spain.,Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - A Brell
- Hematology Lab, Hematology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Mas Casanovas, 9008041, Barcelona, Spain.,Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - I Badell
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Pediatrics Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - J Esteve
- Hematology Department, Hospital Clínic, Barcelona, Spain
| | - M Arnan
- Hematology Department, ICO Hospitalet, L'Hospitalet de Llobregat, Spain
| | - C Talarn
- Hematology Department, Hospital Joan XXIII, Tarragona, Spain
| | - M Tormo
- Hematology Department, Hospital Clínic, Valencia, Spain
| | - A García
- Hematology Department, Hospital Arnau de Vilanova, Lleida, Spain
| | - F Vall-Llovera
- Hematology Department, Hospital de la Mutua de Terrassa, Terrassa, Spain
| | - X Ortin
- Hematology Department, Hospital Verge de La Cinta, Tortosa, Spain
| | - C Pedro
- Hematology Department, Parc de Salut Mar, Barcelona, Spain
| | - J Bargay
- Hematology Department, Hospital de Son Llatzer, Palma de Mallorca, Spain
| | - S Brunet
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Universitat de Barcelona, Barcelona, Spain
| | - J Sierra
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Universitat de Barcelona, Barcelona, Spain
| | - J Nomdedéu
- Hematology Lab, Hematology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Mas Casanovas, 9008041, Barcelona, Spain. .,Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
| |
Collapse
|
39
|
Kandeel EZ, El Sharkawy N, Hanafi M, Samra M, Kamel A. Tracing Leukemia Stem Cells and Their Influence on Clinical Course of Adult Acute Myeloid Leukemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:383-393. [PMID: 32201129 DOI: 10.1016/j.clml.2019.11.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/09/2019] [Accepted: 11/19/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Acute myeloid leukemia (AML) evolves from neoplastic transformation of stem cell disease termed "leukemia stem cells" (LSCs). An unsatisfactory response to AML therapy is determined by the presence of minimal residual disease (MRD). The predominance of LSCs might anticipate sustained MRD results. The present study aimed to demonstrate the effect of LSCs on MRD at induction days 14 and 28 on overall survival (OS) and disease-free survival (DFS) and to compare LSC expression with MRD status. PATIENTS AND METHODS A total of 84 patients with de novo adult AML underwent testing using LSC panels for CD38/CD123/CD34/CD45 and CD90/CD133/CD45/CD33 and different regular MRD panels. RESULTS At day 14 after induction, the high expression of CD123 and CD133 had adverse effects on both OS and DFS (P = .004 and P ≤ .001 and P ≤ .001 and P ≤ .001, respectively). Greater expression of CD34+/CD38-/CD123+ resulted in unfavorable OS and DFS (P ≤ .001 for both). Both CD34+/CD38-/CD123+ and CD34-/CD38+/CD123+ expression at day 14 after induction had an adverse effect on DFS only (P < .001 and P = .029, respectively). On multivariate analysis, CD133 expression and MRD status were independent prognostic parameters (hazard ratio [HR], 2.3; 95% confidence interval [CI], 1.2-4.4; P = .015; and HR, 2.9; 95% CI, 1.0-7.9; P = .041). At day 28 after induction, MRD and increased CD123+/CD34-, CD34+/CD38-/CD123+, CD133+/CD33- expression were associated with inferior OS (P = .016, P = .0035, P = .0.002, and P = .002, respectively). MRD and high expression of CD34+CD123+, CD133+/CD33-, CD34+/CD38-/CD123+ were associated with inferior DFS (P < .001, P = .002, P < .001, P < .001, respectively). On multivariate analysis, only CD133+/CD33- expression was the independent prognostic factor (HR, 3.1; 95% CI, 1.5-6.7; P = .003). CONCLUSIONS Estimation of LSC expression is a sensitive indicator of the response to therapy in adult patients with AML and might be a better prognosticator than the findings from regular MRD panels.
Collapse
Affiliation(s)
- Eman Z Kandeel
- Department of Clinical Pathology, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Nahla El Sharkawy
- Department of Clinical Pathology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Marwa Hanafi
- Department of Clinical Pathology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohamed Samra
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Azza Kamel
- Department of Clinical Pathology, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Despite advances in therapy over the past decades, overall survival for children with acute myeloid leukemia (AML) has not exceeded 70%. In this review, we highlight recent insights into risk stratification for patients with pediatric AML and discuss data driving current and developing therapeutic approaches. RECENT FINDINGS Advances in cytogenetics and molecular profiling, as well as improvements in detection of minimal residual disease after induction therapy, have informed risk stratification, which now relies heavily on these elements. The treatment of childhood AML continues to be based primarily on intensive, conventional chemotherapy. However, recent trials focus on limiting treatment-related toxicity through the identification of low-risk subsets who can safely receive fewer cycles of chemotherapy, allocation of hematopoietic stem-cell transplant to only high-risk patients and optimization of infectious and cardioprotective supportive care. SUMMARY Further incorporation of genomic and molecular data in pediatric AML will allow for additional refinements in risk stratification to enable the tailoring of treatment intensity. These data will also dictate the incorporation of molecularly targeted therapeutics into frontline treatment in the hope of improving survival while decreasing treatment-related toxicity.
Collapse
|
41
|
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: 1.8] [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
|
42
|
Del Principe MI, De Bellis E, Gurnari C, Buzzati E, Savi A, Consalvo MAI, Venditti A. Applications and efficiency of flow cytometry for leukemia diagnostics. Expert Rev Mol Diagn 2019; 19:1089-1097. [PMID: 31709836 DOI: 10.1080/14737159.2019.1691918] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: Multiparametric flow cytometry immunophenotype (MFCI) plays a crucial role in the diagnosis of acute leukemia (AL). Through the comprehensive assessment of surface and intracellular antigens expressed by blasts, MFCI permits to distinguish myeloid or B/T lymphoid AL, or AL of ambiguous lineages. By means of MFCI, the blasts can be characterized in bone marrow, peripheral blood, and body fluids, such as cerebrospinal fluid.Area covered: This review discusses how MFCI is currently applied in the diagnostic evaluation of AL; it also focuses on 'peculiar' issues such as the role of MFCI for the diagnosis of central nervous system leukemic involvement.Expert commentary: Despite the improved knowledge about the biology of AL, MFCI remains a fundamental tool to make a prompt and accurate diagnosis. MFCI also provides prognostic information for some antigens are associated with specific cytogenetic/genetic abnormalities and, recently, it became a powerful tool to evaluate the quality and depth of response (the so called 'measurable residual disease'). Its role as an efficient detector of residual disease paved the way to the investigation of tissues other than bone marrow and peripheral blood, demonstrating that even small amounts of AL appear to have a prognostic impact and may require personalized intervention.
Collapse
Affiliation(s)
- Maria Ilaria Del Principe
- Cattedra di Ematologia, Dipartimento di Biomedicina e Prevenzione, Università Tor Vergata, Roma, Italia.,Ematologia, Dipartimento di Onco-Ematologia, Fondazione Policlinico Tor Vergata, Roma, Italia
| | - Eleonora De Bellis
- Cattedra di Ematologia, Dipartimento di Biomedicina e Prevenzione, Università Tor Vergata, Roma, Italia.,Ematologia, Dipartimento di Onco-Ematologia, Fondazione Policlinico Tor Vergata, Roma, Italia
| | - Carmelo Gurnari
- Cattedra di Ematologia, Dipartimento di Biomedicina e Prevenzione, Università Tor Vergata, Roma, Italia.,Ematologia, Dipartimento di Onco-Ematologia, Fondazione Policlinico Tor Vergata, Roma, Italia
| | - Elisa Buzzati
- Cattedra di Ematologia, Dipartimento di Biomedicina e Prevenzione, Università Tor Vergata, Roma, Italia.,Ematologia, Dipartimento di Onco-Ematologia, Fondazione Policlinico Tor Vergata, Roma, Italia
| | - Arianna Savi
- Cattedra di Ematologia, Dipartimento di Biomedicina e Prevenzione, Università Tor Vergata, Roma, Italia.,Ematologia, Dipartimento di Onco-Ematologia, Fondazione Policlinico Tor Vergata, Roma, Italia
| | | | - Adriano Venditti
- Cattedra di Ematologia, Dipartimento di Biomedicina e Prevenzione, Università Tor Vergata, Roma, Italia.,Ematologia, Dipartimento di Onco-Ematologia, Fondazione Policlinico Tor Vergata, Roma, Italia
| |
Collapse
|
43
|
Komkov A, Miroshnichenkova A, Nugmanov G, Popov A, Pogorelyy M, Zapletalova E, Jelinkova H, Pospisilova S, Lebedev Y, Chudakov D, Olshanskaya Y, Plevova K, Maschan M, Mamedov I. High‐throughput sequencing of T‐cell receptor alpha chain clonal rearrangements at the DNA level in lymphoid malignancies. Br J Haematol 2019; 188:723-731. [DOI: 10.1111/bjh.16230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/04/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Alexander Komkov
- Department of Genomics of Adaptive Immunity Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry Moscow Russia
- Laboratory of Cytogenetics and Molecular Genetics Dmitry Rogachev National Medical and Research Centre of Paediatric Haematology, Oncology and Immunology Moscow Russia
| | - Anna Miroshnichenkova
- Laboratory of Cytogenetics and Molecular Genetics Dmitry Rogachev National Medical and Research Centre of Paediatric Haematology, Oncology and Immunology Moscow Russia
| | - Gaiaz Nugmanov
- Department of Genomics of Adaptive Immunity Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry Moscow Russia
| | - Alexander Popov
- Laboratory of Cytogenetics and Molecular Genetics Dmitry Rogachev National Medical and Research Centre of Paediatric Haematology, Oncology and Immunology Moscow Russia
| | - Mikhail Pogorelyy
- Department of Genomics of Adaptive Immunity Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry Moscow Russia
- Department of Molecular Technologies Pirogov Russian National Research Medical University Moscow Russia
| | - Eva Zapletalova
- Department of Internal Medicine, Haematology and Oncology University Hospital Brno and Faculty of Medicine Masaryk University Brno Czech Republic
| | - Hana Jelinkova
- Department of Internal Medicine, Haematology and Oncology University Hospital Brno and Faculty of Medicine Masaryk University Brno Czech Republic
| | - Sarka Pospisilova
- Department of Internal Medicine, Haematology and Oncology University Hospital Brno and Faculty of Medicine Masaryk University Brno Czech Republic
- Central European Institute of Technology Masaryk University Brno Czech Republic
| | - Yuri Lebedev
- Department of Genomics of Adaptive Immunity Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry Moscow Russia
- Department of Molecular Technologies Pirogov Russian National Research Medical University Moscow Russia
| | - Dmitriy Chudakov
- Department of Genomics of Adaptive Immunity Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry Moscow Russia
- Department of Molecular Technologies Pirogov Russian National Research Medical University Moscow Russia
- Central European Institute of Technology Masaryk University Brno Czech Republic
| | - Yulia Olshanskaya
- Laboratory of Cytogenetics and Molecular Genetics Dmitry Rogachev National Medical and Research Centre of Paediatric Haematology, Oncology and Immunology Moscow Russia
| | - Karla Plevova
- Department of Internal Medicine, Haematology and Oncology University Hospital Brno and Faculty of Medicine Masaryk University Brno Czech Republic
- Central European Institute of Technology Masaryk University Brno Czech Republic
| | - Michael Maschan
- Laboratory of Cytogenetics and Molecular Genetics Dmitry Rogachev National Medical and Research Centre of Paediatric Haematology, Oncology and Immunology Moscow Russia
| | - Ilgar Mamedov
- Department of Genomics of Adaptive Immunity Shemyakin‐Ovchinnikov Institute of Bioorganic Chemistry Moscow Russia
- Laboratory of Cytogenetics and Molecular Genetics Dmitry Rogachev National Medical and Research Centre of Paediatric Haematology, Oncology and Immunology Moscow Russia
- Department of Molecular Technologies Pirogov Russian National Research Medical University Moscow Russia
- Central European Institute of Technology Masaryk University Brno Czech Republic
| |
Collapse
|
44
|
Merle M, Fischbacher D, Liepert A, Grabrucker C, Kroell T, Kremser A, Dreyssig J, Freudenreich M, Schuster F, Borkhardt A, Kraemer D, Koehne CH, Kolb HJ, Schmid C, Schmetzer HM. Serum Chemokine-release Profiles in AML-patients Might Contribute to Predict the Clinical Course of the Disease. Immunol Invest 2019; 49:365-385. [PMID: 31535582 DOI: 10.1080/08820139.2019.1661429] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In cancer or hematologic disorders, chemokines act as growth- or survival factors, regulating hematopoiesis and angiogenesis, determining metastatic spread and controlling leukocyte infiltration into tumors to inhibit antitumor immune responses. The aim was to quantify the release of CXCL8, -9, -10, CCL2, -5, and IL-12 in AML/MDS-pts' serum by cytometric bead array and to correlate data with clinical subtypes and courses. Minimal differences in serum-levels subdivided into various groups (e.g. age groups, FAB-types, blast-proportions, cytogenetic-risk-groups) were seen, but higher release of CXCL8, -9, -10 and lower release of CCL2 and -5 tendentially correlated with more favorable subtypes (<50 years of age, <80% blasts in PB). Comparing different stages of the disease higher CCL5-release in persisting disease and a significantly higher CCL2-release at relapse were found compared to first diagnosis - pointing to a change of 'disease activity' on a chemokine level. Correlations with later on achieved response to immunotherapy and occurrence of GVHD were seen: Higher values of CXCL8, -9, -10 and CCL2 and lower CCL5-values correlated with achieved response to immunotherapy. Predictive cut-off-values were evaluated separating the groups in 'responders' and 'non-responders'. Higher levels of CCL2 and -5 but lower levels of CXCL8, -9, -10 correlated with occurrence of GVHD. We conclude, that in AML-pts' serum higher values of CXCL8, -9, -10 and lower values of CCL5 and in part of CCL2 correlate with more favorable subtypes and improved antitumor'-reactive function. This knowledge can contribute to develop immune-modifying strategies that promote antileukemic adaptive immune responses.
Collapse
Affiliation(s)
- M Merle
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - D Fischbacher
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - A Liepert
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - C Grabrucker
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - T Kroell
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - A Kremser
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - J Dreyssig
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - M Freudenreich
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany
| | - F Schuster
- Department for Pediatric Hematology and Oncology, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - A Borkhardt
- Department for Pediatric Hematology and Oncology, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - D Kraemer
- Department for Hematology, Municipal Hospital Oldenburg, Oldenburg, Germany
| | - C-H Koehne
- Department for Hematology, Municipal Hospital Oldenburg, Oldenburg, Germany
| | - H J Kolb
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany.,Helmholtz Center, Clinical Cooperative Group Human Cell Transplantation (CCG-HCT), Munich, Germany
| | - C Schmid
- Department for Hematology, University Hospital Augsburg, Augsburg, Germany
| | - H M Schmetzer
- Department for Hematopoietic Transplantations, Med III, University Hospital of Munich, Munich, Germany.,Helmholtz Center, Clinical Cooperative Group Human Cell Transplantation (CCG-HCT), Munich, Germany
| |
Collapse
|
45
|
|
46
|
Li Z, Liu Y, Wang Q, Chen L, Ma L, Hao S. Autologous Stem Cell Transplantation Is a Viable Postremission Therapy for Intermediate-Risk Acute Myeloid Leukemia in First Complete Remission in the Absence of a Matched Identical Sibling: A Meta-Analysis. Acta Haematol 2019; 141:164-175. [PMID: 30808826 PMCID: PMC6492512 DOI: 10.1159/000495206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/08/2018] [Indexed: 11/19/2022]
Abstract
BACKGROUND The preferred type of postremission therapy (PRT) for intermediate-risk acute myeloid leukemia (AML) in first complete remission (CR1) is a subject of continued debate. Although allogeneic stem cell transplantation (alloSCT) is regarded as a curative strategy for AML, the efficacy of autologous stem cell transplantation (autoSCT) for patients without a matched sibling donor (MSD) has remained controversial. METHODS To compare survival outcomes after alloSCT versus autoSCT for patients with intermediate-risk AML in CR1, we performed a meta-analysis of 11 clinical studies. The outcomes included relapse-free survival (RFS), overall survival (OS), relapse rate (RR), and treatment-related mortality (TRM). RESULTS Compared with autoSCT, alloSCT showed better RFS, OS, and RR benefits, but higher TRM. Subgroup analysis based on donor category (MSD and matched unrelated donor [MUD]) of alloSCT showed alloSCT from MSD rather than from MUD had better OS benefits compared to autoSCT. For fms-like tyrosine kinase 3 internal tandem duplications (FLT3-ITD) wild-type patients, alloSCT and autoSCT had comparable RFS and OS outcomes. CONCLUSION Our results suggest that, in the absence of an available MSD, autoSCT remains a viable PRT alternative for intermediate-risk AML in CR1, especially for FLT3-ITD wild-type patients.
Collapse
Affiliation(s)
- Zhichao Li
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yinmei Liu
- Department of Hospital Infection Management, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Qing Wang
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Linjun Chen
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Liyuan Ma
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Siguo Hao
- Department of Hematology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China,
| |
Collapse
|
47
|
Buldini B, Maurer-Granofszky M, Varotto E, Dworzak MN. Flow-Cytometric Monitoring of Minimal Residual Disease in Pediatric Patients With Acute Myeloid Leukemia: Recent Advances and Future Strategies. Front Pediatr 2019; 7:412. [PMID: 31681710 PMCID: PMC6798174 DOI: 10.3389/fped.2019.00412] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/25/2019] [Indexed: 01/10/2023] Open
Abstract
Minimal residual disease (MRD) by multiparametric flow cytometry (MFC) has been recently shown as a strong and independent prognostic marker of relapse in pediatric AML (pedAML) when measured at specific time points during Induction and/or Consolidation therapy. Hence, MFC-MRD has the potential to refine the current strategies of pedAML risk stratification, traditionally based on the cytogenetic and molecular genetic aberrations at diagnosis. Consequently, it may guide the modulation of therapy intensity and clinical decision making. However, the use of non-standardized protocols, including different staining panels, analysis, and gating strategies, may hamper a broad implementation of MFC-MRD monitoring in clinical routine. Besides, the thresholds of MRD positivity still need to be validated in large, prospective and multi-center clinical studies, as well as optimal time points of MRD assessment during therapy, to better discriminate patients with different prognosis. In the present review, we summarize the most relevant findings on MFC-MRD testing in pedAML. We examine the clinical significance of MFC-MRD and the recent advances in its standardization, including innovative approaches with an automated analysis of MFC-MRD data. We also touch upon other technologies for MRD assessment in AML, such as quantitative genomic breakpoint PCR, current challenges and future strategies to enable full incorporation of MFC-MRD into clinical practice.
Collapse
Affiliation(s)
- Barbara Buldini
- Laboratory of Hematology-Oncology, Department of Woman's and Child's Health, University of Padova, Padova, Italy
| | | | - Elena Varotto
- Laboratory of Hematology-Oncology, Department of Woman's and Child's Health, University of Padova, Padova, Italy
| | - Michael N Dworzak
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Vienna, Austria
| |
Collapse
|
48
|
Leukemia Stem Cells in the Pathogenesis, Progression, and Treatment of Acute Myeloid Leukemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1143:95-128. [DOI: 10.1007/978-981-13-7342-8_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
49
|
Platzbecker U, Middeke JM, Sockel K, Herbst R, Wolf D, Baldus CD, Oelschlägel U, Mütherig A, Fransecky L, Noppeney R, Bug G, Götze KS, Krämer A, Bochtler T, Stelljes M, Groth C, Schubert A, Mende M, Stölzel F, Borkmann C, Kubasch AS, von Bonin M, Serve H, Hänel M, Dührsen U, Schetelig J, Röllig C, Kramer M, Ehninger G, Bornhäuser M, Thiede C. Measurable residual disease-guided treatment with azacitidine to prevent haematological relapse in patients with myelodysplastic syndrome and acute myeloid leukaemia (RELAZA2): an open-label, multicentre, phase 2 trial. Lancet Oncol 2018; 19:1668-1679. [DOI: 10.1016/s1470-2045(18)30580-1] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/18/2018] [Accepted: 07/24/2018] [Indexed: 12/12/2022]
|
50
|
Fuda F, Chen W. Minimal/Measurable Residual Disease Detection in Acute Leukemias by Multiparameter Flow Cytometry. Curr Hematol Malig Rep 2018; 13:455-466. [DOI: 10.1007/s11899-018-0479-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|