1
|
Nakako S, Okamura H, Yokota I, Umemoto Y, Horiuchi M, Sakatoku K, Ido K, Makuuchi Y, Kuno M, Takakuwa T, Nishimoto M, Hirose A, Nakamae M, Nakashima Y, Koh H, Hino M, Nakamae H. Dynamic Relapse Prediction by Peripheral Blood WT1mRNA after Allogeneic Hematopoietic Cell Transplantation for Myeloid Neoplasms. Transplant Cell Ther 2024; 30:1088.e1-1088.e12. [PMID: 39147137 DOI: 10.1016/j.jtct.2024.08.008] [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: 04/21/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024]
Abstract
Although various relapse prediction models based on pretransplant information have been reported, they cannot update the predictive probability considering post-transplant patient status. Therefore, these models are not appropriate for deciding on treatment adjustment and preemptive intervention during post-transplant follow-up. A dynamic prediction model can update the predictive probability by considering the information obtained during follow-up. This study aimed to develop and assess a dynamic relapse prediction model after allogeneic hematopoietic cell transplantation (allo-HCT) for acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) using peripheral blood Wilms' tumor 1 messenger RNA (WT1mRNA). We retrospectively analyzed patients with AML or MDS who underwent allo-HCT at our institution. To develop dynamic models, we employed the landmarking supermodel approach, using age, refined disease risk index, conditioning intensity, and number of transplantations as pretransplant covariates and both pre- and post-transplant peripheral blood WT1mRNA levels as time-dependent covariates. Finally, we compared the predictive performances of the conventional and dynamic models by area under the time-dependent receiver operating characteristic curves. A total of 238 allo-HCT cases were included in this study. The dynamic model that considered all pretransplant WT1mRNA levels and their kinetics showed superior predictive performance compared to models that considered only pretransplant covariates or factored in both pretransplant covariates and post-transplant WT1mRNA levels without their kinetics; their time-dependent areas under the curve were 0.89, 0.73, and 0.87, respectively. The predictive probability of relapse increased gradually from approximately 90 days before relapse. Furthermore, we developed a web application to make our model user-friendly. This model facilitates real-time, highly accurate, and personalized relapse prediction at any time point after allo-HCT. This will aid decision-making during post-transplant follow-up by offering objective relapse forecasts for physicians.
Collapse
MESH Headings
- Humans
- Hematopoietic Stem Cell Transplantation/methods
- Male
- Female
- Middle Aged
- WT1 Proteins/blood
- WT1 Proteins/genetics
- Adult
- RNA, Messenger/blood
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/genetics
- Myelodysplastic Syndromes/therapy
- Myelodysplastic Syndromes/blood
- Transplantation, Homologous
- Retrospective Studies
- Recurrence
- Aged
- Young Adult
- Adolescent
- Neoplasm Recurrence, Local/blood
- Neoplasm Recurrence, Local/genetics
Collapse
Affiliation(s)
- Soichiro Nakako
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Okamura
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan; Department of Laboratory Medicine and Medical Informatics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan.
| | - Isao Yokota
- Department of Biostatistics, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Yukari Umemoto
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Mirei Horiuchi
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Kazuki Sakatoku
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Kentaro Ido
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan; Department of Laboratory Medicine and Medical Informatics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yosuke Makuuchi
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Masatomo Kuno
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Teruhito Takakuwa
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Mitsutaka Nishimoto
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Asao Hirose
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Mika Nakamae
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan; Department of Laboratory Medicine and Medical Informatics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yasuhiro Nakashima
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hideo Koh
- Department of Preventive Medicine and Environmental Health, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Masayuki Hino
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan; Department of Laboratory Medicine and Medical Informatics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hirohisa Nakamae
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
2
|
Duployez N, Preudhomme C. Monitoring molecular changes in the management of myelodysplastic syndromes. Br J Haematol 2024; 205:772-779. [PMID: 38934371 DOI: 10.1111/bjh.19614] [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: 04/18/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024]
Abstract
The ongoing or anticipated therapeutic advances as well as previous experience in other malignancies, including acute myeloid leukaemia, have made molecular monitoring a potential interesting tool for predicting outcomes and demonstrating treatment efficacy in patients with myelodysplastic syndromes (MDS). The important genetic heterogeneity in MDS has made challenging the establishment of recommendations. In this context, high-throughput/next-generation sequencing (NGS) has emerged as an attractive tool, especially in patients with high-risk diseases. However, its implementation in clinical practice still suffers from a lack of standardization in terms of sensitivity, bioinformatics and result interpretation. Data from literature, mostly gleaned from retrospective cohorts, show NGS monitoring when used appropriately could help clinicians to guide therapy, detect early relapse and predict disease evolution. Translating these observations into personalized patient management requires a prospective evaluation in clinical research and remains a major challenge for the next years.
Collapse
Affiliation(s)
- Nicolas Duployez
- Laboratory of Haematology, CHU Lille, Lille, France
- U1277 CANTHER (Cancer Heterogeneity Plasticity and Resistance to Therapies), University of Lille, INSERM, Lille, France
| | - Claude Preudhomme
- Laboratory of Haematology, CHU Lille, Lille, France
- U1277 CANTHER (Cancer Heterogeneity Plasticity and Resistance to Therapies), University of Lille, INSERM, Lille, France
| |
Collapse
|
3
|
Wu J, Yan H, Xiang C. Wilms' tumor gene 1 in hematological malignancies: friend or foe? Hematology 2023; 28:2254557. [PMID: 37668240 DOI: 10.1080/16078454.2023.2254557] [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: 03/27/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023] Open
Abstract
Wilms' tumor gene 1 (WT1) is a transcription and post-translational factor that has a crucial role in the biological and pathological processes of several human malignancies. For hematological malignancies, WT1 overexpression or mutation has been found in leukemia and myelodysplastic syndrome. About 70-90% of acute myeloid leukemia patients showed WT1 overexpression, and 6-15% of patients carried WT1 mutations. WT1 has been widely regarded as a marker for monitoring minimal residual disease in acute myeloid leukemia. Many researchers were interested in developing WT1 targeting therapy. In this review, we summarized biological and pathological functions, correlation with other genes and clinical features, prognosis value and targeting therapy of WT1 in hematological features.
Collapse
Affiliation(s)
- Jie Wu
- Department of Emergency Medicine, The Fifth People's Hospital of Huai'an and Huai'an Hospital Affiliated to Yangzhou University, Huai'an, People's Republic of China
| | - Hui Yan
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, People's Republic of China
| | - Chunli Xiang
- Department of General Medicine, The Affiliated Huai'an Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, People's Republic of China
| |
Collapse
|
4
|
Malagola M, Polverelli N, Beghin A, Bolda F, Comini M, Farina M, Morello E, Radici V, Accorsi Buttini E, Bernardi S, Re F, Leoni A, Bonometti D, Brugnoni D, Lanfranchi A, Russo D. Bone marrow CD34+ molecular chimerism as an early predictor of relapse after allogeneic stem cell transplantation in patients with acute myeloid leukemia. Front Oncol 2023; 13:1133418. [PMID: 36950550 PMCID: PMC10025489 DOI: 10.3389/fonc.2023.1133418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Background Minimal residual disease (MRD) monitoring is an important tool to optimally address post-transplant management of acute myeloid leukemia (AML) patients. Methods We retrospectively analyzed the impact of bone marrow CD34+ molecular chimerism and WT1 on the outcome of a consecutive series of 168 AML patients submitted to allogeneic stem cell transplantation. Results The cumulative incidence of relapse (CIR) was significantly lower in patients with donor chimerism on CD34+ cells ≥ 97.5% and WT1 < 213 copies/ABL x 10^4 both at 1st month (p=0.008 and p<0.001) and at 3rd month (p<0.001 for both). By combining chimerism and WT1 at 3rd month, 13 patients with chimerism < 97.5% or WT1 > 213 showed intermediate prognosis. 12 of these patients fell in this category because of molecular chimerism < 97.5% at a time-point in which WT1 was < 213. Conclusions Our results confirm that lineage-specific molecular chimerism and WT1 after allo-SCT (1st and 3rd month) are useful MRD markers. When considered together at 3rd month, CD34+ molecular chimerism could represent an earlier predictor of relapse compared to WT1. Further studies are necessary to confirm this preliminary observation.
Collapse
Affiliation(s)
- Michele Malagola
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- *Correspondence: Michele Malagola,
| | - Nicola Polverelli
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandra Beghin
- Stem Cell Laboratory, Section of Hematology and Blood Coagulation, Clinical Chemistry Laboratory, Diagnostics Department, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Federica Bolda
- Stem Cell Laboratory, Section of Hematology and Blood Coagulation, Clinical Chemistry Laboratory, Diagnostics Department, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Marta Comini
- Stem Cell Laboratory, Section of Hematology and Blood Coagulation, Clinical Chemistry Laboratory, Diagnostics Department, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Mirko Farina
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Enrico Morello
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Vera Radici
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Eugenia Accorsi Buttini
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Simona Bernardi
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Centro di Ricerca Emato-oncologico AIL (CREA) , “ASST-Spedali Civili” Hospital of Brescia, Brescia, Italy
| | - Federica Re
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Centro di Ricerca Emato-oncologico AIL (CREA) , “ASST-Spedali Civili” Hospital of Brescia, Brescia, Italy
| | - Alessandro Leoni
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Centro di Ricerca Emato-oncologico AIL (CREA) , “ASST-Spedali Civili” Hospital of Brescia, Brescia, Italy
| | - Davide Bonometti
- Department of Hematology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Duilio Brugnoni
- Department of Laboratory Diagnostics, ASST Spedali Civili, Brescia, Italy
| | - Arnalda Lanfranchi
- Stem Cell Laboratory, Section of Hematology and Blood Coagulation, Clinical Chemistry Laboratory, Diagnostics Department, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Domenico Russo
- Blood Diseases and Cell Therapies unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
5
|
High ME1 Expression Is a Molecular Predictor of Post-Transplant Survival of Patients with Acute Myeloid Leukemia. Cancers (Basel) 2022; 15:cancers15010296. [PMID: 36612292 PMCID: PMC9818450 DOI: 10.3390/cancers15010296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/09/2022] [Accepted: 12/26/2022] [Indexed: 01/04/2023] Open
Abstract
Several laboratory and clinical variables have been reported to be associated with the outcome of intensive chemotherapy for acute myeloid leukemia (AML), but only a few have been tested in the context of hematopoietic stem cell transplant (HSCT). This study aimed to identify genes whose expression of AML at diagnosis were associated with survival after HSCT. For this purpose, three publicly available adult AML cohorts (TCGA, BeatAML, and HOVON), whose patients were treated with intensive chemotherapy and then subjected to allogeneic or autologous HSCT, were included in this study. After whole transcriptome analysis, we identified ME1 as the only gene whose high expression was associated with shorter survival in patients subjected to HSCT. In addition, the inclusion of ME1 expression was able to improve the European LeukemiaNet risk stratification. Pathways related to lipid biosynthesis, mainly fatty acids, and cholesterol were positively correlated with ME1 expression. Furthermore, ME1 expression was associated with an M2 macrophage-enriched microenvironment, mature AML blasts hierarchy, and oxidative phosphorylation metabolism. Therefore, ME1 expression can be used as biomarker of poor response to HSCT in AML.
Collapse
|
6
|
Stanojevic M, Grant M, Vesely SK, Knoblach S, Kanakry CG, Nazarian J, Panditharatna E, Panchapakesan K, Gress RE, Holter-Chakrabarty J, Williams KM. Peripheral blood marker of residual acute leukemia after hematopoietic cell transplantation using multi-plex digital droplet PCR. Front Immunol 2022; 13:999298. [PMID: 36248870 PMCID: PMC9556966 DOI: 10.3389/fimmu.2022.999298] [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: 07/20/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Background Relapse remains the primary cause of death after hematopoietic cell transplantation (HCT) for acute leukemia. The ability to identify minimal/measurable residual disease (MRD) via the blood could identify patients earlier when immunologic interventions may be more successful. We evaluated a new test that could quantify blood tumor mRNA as leukemia MRD surveillance using droplet digital PCR (ddPCR). Methods The multiplex ddPCR assay was developed using tumor cell lines positive for the tumor associated antigens (TAA: WT1, PRAME, BIRC5), with homeostatic ABL1. On IRB-approved protocols, RNA was isolated from mononuclear cells from acute leukemia patients after HCT (n = 31 subjects; n = 91 specimens) and healthy donors (n = 20). ddPCR simultaneously quantitated mRNA expression of WT1, PRAME, BIRC5, and ABL1 and the TAA/ABL1 blood ratio was measured in patients with and without active leukemia after HCT. Results Tumor cell lines confirmed quantitation of TAAs. In patients with active acute leukemia after HCT (MRD+ or relapse; n=19), the blood levels of WT1/ABL1, PRAME/ABL1, and BIRC5/ABL1 exceeded healthy donors (p<0.0001, p=0.0286, and p=0.0064 respectively). Active disease status was associated with TAA positivity (1+ TAA vs 0 TAA) with an odds ratio=10.67, (p=0.0070, 95% confidence interval 1.91 - 59.62). The area under the curve is 0.7544. Changes in ddPCR correlated with disease response captured on standard of care tests, accurately denoting positive or negative disease burden in 15/16 (95%). Of patients with MRD+ or relapsed leukemia after HCT, 84% were positive for at least one TAA/ABL1 in the peripheral blood. In summary, we have developed a new method for blood MRD monitoring of leukemia after HCT and present preliminary data that the TAA/ABL1 ratio may may serve as a novel surrogate biomarker for relapse of acute leukemia after HCT.
Collapse
Affiliation(s)
- M. Stanojevic
- Department of Pediatrics, MedStar Georgetown University Hospital, Washington, DC, United States
| | - M. Grant
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA, United States
| | - S. K. Vesely
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - S. Knoblach
- Children’s Research Institute, Research Center for Genetic Medicine, Children’s National Health System, Washington, DC, United States
| | - C. G. Kanakry
- Experimental Transplantation and Immunotherapy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - J. Nazarian
- Children’s Research Institute, Research Center for Genetic Medicine, Children’s National Health System, Washington, DC, United States,Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - E. Panditharatna
- Department of Pediatric Oncology, Dana-Farber Boston Children’s Cancer and Blood Disorders Center, Boston, MA, United States
| | - K. Panchapakesan
- Children’s Research Institute, Research Center for Genetic Medicine, Children’s National Health System, Washington, DC, United States
| | - R. E. Gress
- Experimental Transplantation and Immunotherapy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - J. Holter-Chakrabarty
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Kirsten M. Williams
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA, United States,*Correspondence: Kirsten M. Williams,
| |
Collapse
|
7
|
Dynamic change in peripheral blood WT1 mRNA levels within three cycles of azacitidine predict treatment response in patients with high-risk myelodysplastic syndromes. Ann Hematol 2022; 101:1239-1250. [PMID: 35257209 DOI: 10.1007/s00277-022-04807-w] [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] [Received: 11/14/2021] [Accepted: 02/22/2022] [Indexed: 01/30/2023]
Abstract
Azacitidine (AZA) improves overall survival (OS) in patients with high-risk myelodysplastic syndromes (MDS). However, predictive factors for response to AZA remain largely unknown. To elucidate whether dynamic change in peripheral blood (PB) Wilms' Tumor 1 (WT1) mRNA levels could predict response to AZA, we retrospectively identified 75 treatment-naïve patients with high-risk MDS who received at least 3 cycles of AZA. We classified patients into 4 groups, low-increase (LI), low-stable (LS), high-decrease (HD), and high-stable (HS) based on the dynamic change in PB WT1 mRNA levels within 3 cycles of AZA. Cumulative incidence of overall response after 10 cycles of AZA was significantly higher in LS/HD than in HS/LI (75.5% vs 4.5%, P < 0.001). The median OS for LS/HD was 18.2 months (95% CI, 12.8-28.1 months), whereas it was 11.6 months for HS/LI (95% CI, 6.6-14.1 months; P < 0.001). Multivariate analysis demonstrated that poor-/very poor-IPSS-R cytogenetic risk and HS/LI were independently associated with poor OS (poor-/very poor-IPSS-R cytogenetic risk: HR, 2.26; 95% CI, 1.10-4.68, P = 0.03, HS/LI: HR, 2.32; 95% CI, 1.21-4.46, P = 0.01). Patients with HS/LI did not show any further response to continuous AZA, and they should be considered for alternative therapy from earlier cycles.
Collapse
|
8
|
Kitamura W, Fujii N, Nawa Y, Fujishita K, Sugiura H, Yoshioka T, Fujiwara Y, Usui Y, Fujii K, Fujiwara H, Asada N, Nishimori H, Matsuoka KI, Maeda Y. Possible prognostic impact of WT1 mRNA expression at day + 30 after haploidentical peripheral blood stem cell transplantation with posttransplant cyclophosphamide for patients with myeloid neoplasm: a multicenter study from the Okayama Hematological Study Group. Int J Hematol 2022; 115:515-524. [PMID: 35119651 DOI: 10.1007/s12185-022-03290-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Previous studies have revealed that relapse of myeloid neoplasms after allogeneic hematopoietic stem cell transplantation (allo-HSCT) could be predicted by monitoring Wilms' tumor 1 (WT1) mRNA expression. However, only a few studies have investigated patients who received human leukocyte antigen-haploidentical stem cell transplantation with posttransplant cyclophosphamide (PTCY-haplo). In this study, we investigated the relationship between WT1 mRNA levels and clinical outcomes in the PTCY-haplo group, and compared them with those in the conventional graft-versus-host disease prophylaxis group (conventional group). METHODS We retrospectively analyzed 130 patients who received their first allo-HSCT between April 2017 and December 2020, including 26 who received PTCY-haplo. RESULTS The WT1 mRNA expression level at day + 30 after allo-HSCT associated with increased risk of 1-year cumulative incidence of relapse (CIR) was ≥ 78 copies/μg RNA in the conventional group (p < 0.01) and ≥ 50 copies/μg RNA in the PTCY-haplo group (p = 0.03). CONCLUSIONS The appropriate cutoff level of WT1 mRNA at day + 30 after allo-HSCT for predicting prognosis in patients treated with PTCY-haplo may be < 50 copies/μg RNA.
Collapse
Affiliation(s)
- Wataru Kitamura
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Nobuharu Fujii
- Divison of Blood Transfusion, Okayama University Hospital, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Yuichiro Nawa
- Division of Hematology, Ehime Prefectural Central Hospital, 83, Kasuga-cho, Matsuyama, 790-0024, Japan
| | - Keigo Fujishita
- Department of Hematology and Blood Transfusion, Kochi Health Science Center, 2125-1, Ike, Kochi, 781-8555, Japan
| | - Hiroyuki Sugiura
- Department of Hematology, Chugoku Central Hospital, 148-13, Oazakamiiwanari, Miyuki-cho, Fukuyama, 720-0001, Japan
| | - Takanori Yoshioka
- Department of Hematology, National Hospital Organization Okayama Medical Center, 1711-1, Tamasu, Kita-ku, Okayama, 701-1192, Japan
| | - Yuki Fujiwara
- Department of Hematology and Oncology, Japanese Red Cross Society Himeji Hospital, 1-12-1, Shimoteno, Himeji, 670-8540, Japan
| | - Yoshiaki Usui
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Keiko Fujii
- Divison of Clinical Laboratory, Okayama University Hospital, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Hideaki Fujiwara
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Noboru Asada
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Hisakazu Nishimori
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Yoshinobu Maeda
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| |
Collapse
|
9
|
Shen MZ, Zhang XH, Xu LP, Wang Y, Yan CH, Chen H, Chen YH, Han W, Wang FR, Wang JZ, Zhao XS, Qin YZ, Chang YJ, Liu KY, Huang XJ, Mo XD. Preemptive Interferon-α Therapy Could Protect Against Relapse and Improve Survival of Acute Myeloid Leukemia Patients After Allogeneic Hematopoietic Stem Cell Transplantation: Long-Term Results of Two Registry Studies. Front Immunol 2022; 13:757002. [PMID: 35154096 PMCID: PMC8831731 DOI: 10.3389/fimmu.2022.757002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 01/10/2022] [Indexed: 12/26/2022] Open
Abstract
For allogeneic hematopoietic stem cell transplantation (allo-HSCT) recipients, preemptive interferon-α (IFN-α) therapy is considered as a useful method to eliminate the minimal residual disease (MRD). Our purpose is to assess the long-term efficacy of preemptive IFN-α therapy in acute myeloid leukemia (AML) patients following allo-HSCT based on two registry studies (#NCT02185261 and #NCT02027064). We would present the final data and unpublished results of long-term clinical outcomes with extended follow-up. We adopted polymerase chain reaction (PCR) and multiparameter flow cytometry (MFC) to monitor MRD, and a positive result of bone marrow specimen examined by either of them would be identified as the MRD-positive status. Subcutaneous injections of recombinant human IFN-α-2b were performed for 6 cycles, and prolonged IFN-α therapy could be permitted at the request of patients. The median cycles were 3.5 (range, 0.5-30.5) cycles. A total of 9 patients suffered from grade ≥3 toxicities (i.e., infectious: n = 6; hematologic: n = 3). The 6-year cumulative incidences of relapse and non-relapse mortality following IFN-α therapy were 13.0% (95% confidence interval [CI], 5.4-20.6%) and 3.9% (95%CI, 0.0-17.6%), respectively. The probability of disease-free survival at 6 years following IFN-α therapy was 83.1% (95%CI, 75.2-91.9%). The probability of overall survival at 6 years following IFN-α therapy was 88.3% (95%CI, 81.4-95.8%). The cumulative incidences of total chronic graft-versus-host disease (cGVHD) and severe cGVHD at 6 years following IFN-α therapy were 66.2% (95%CI, 55.5-77.0%) and 10.4% (95%CI, 3.6-17.2%), respectively. Multivariable analysis showed that an alternative donor was associated with a lower risk of relapse and the better disease-free survival. Thus, preemptive IFN-α therapy could clear MRD persistently, prevent relapse truly, and improve long-term survival in AML patients following allo-HSCT.
Collapse
Affiliation(s)
- Meng-Zhu Shen
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Lan-Ping Xu
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yu Wang
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Chen-Hua Yan
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Huan Chen
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yu-Hong Chen
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Wei Han
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Feng-Rong Wang
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Jing-Zhi Wang
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Su Zhao
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Ya-Zhen Qin
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Ying-Jun Chang
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Kai-Yan Liu
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao-Dong Mo
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Austin AE, Byrne M. Detecting and preventing post-hematopoietic cell transplant relapse in AML. Curr Opin Hematol 2021; 28:380-388. [PMID: 34534984 DOI: 10.1097/moh.0000000000000686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Relapsed disease is the primary cause of mortality for acute myeloid leukemia (AML) patients after allogeneic hematopoietic cell transplantation (HCT). This review outlines the most recent advances in the detection and prevention of AML relapse following allogeneic HCT. RECENT FINDINGS Conventional methods for predicting post-HCT relapse rely on the molecular and cytogenetics features present at diagnosis. These methods are slow to reflect a growing understanding of the molecular heterogeneity of AML and impact of new therapies on post-HCT outcomes. The use of measurable residual disease (MRD) techniques, including multiparameter flow cytometry and molecular testing, may improve the prognostic ability of these models and should be incorporated into post-HCT surveillance whenever possible.In the post-HCT setting, FLT3 inhibitor maintenance data indicate that effective therapies can improve post-HCT outcomes. Maintenance data with DNA methyltransferase inhibitor monotherapy is less compelling and outcomes may improve with combinations. Early interventions directed at preemptive management of MRD may further improve post-HCT outcomes. SUMMARY Post-HCT AML relapse prevention has evolved to include more sensitive measures of disease detection and novel therapies that may improve outcomes of poor-risk AML patients. Additional work is needed to maintain this progress.
Collapse
Affiliation(s)
| | - Michael Byrne
- Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
11
|
Vonk CM, Al Hinai ASA, Hanekamp D, Valk PJM. Molecular Minimal Residual Disease Detection in Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:5431. [PMID: 34771594 PMCID: PMC8582498 DOI: 10.3390/cancers13215431] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
Initial induction chemotherapy to eradicate the bulk of acute myeloid leukemia (AML) cells results in complete remission (CR) in the majority of patients. However, leukemic cells persisting in the bone marrow below the morphologic threshold remain unaffected and have the potential to proliferate and re-emerge as AML relapse. Detection of minimal/measurable residual disease (MRD) is a promising prognostic marker for AML relapse as it can assess an individual patients' risk profile and evaluate their response to treatment. With the emergence of molecular techniques, such as next generation sequencing (NGS), a more sensitive assessment of molecular MRD markers is available. In recent years, the detection of MRD by molecular assays and its association with AML relapse and survival has been explored and verified in multiple studies. Although most studies show that the presence of MRD leads to a worse clinical outcome, molecular-based methods face several challenges including limited sensitivity/specificity, and a difficult distinction between mutations that are representative of AML rather than clonal hematopoiesis. This review describes the studies that have been performed using molecular-based assays for MRD detection in the context of other MRD detection approaches in AML, and discusses limitations, challenges and opportunities.
Collapse
Affiliation(s)
- Christian M Vonk
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands
| | - Adil S A Al Hinai
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands
- National Genetic Center, Ministry of Health, Muscat 111, Oman
| | - Diana Hanekamp
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands
- Department of Hematology, Cancer Center VU University Medical Center, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
| | - Peter J M Valk
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands
| |
Collapse
|
12
|
Platzbecker U, Kubasch AS, Homer-Bouthiette C, Prebet T. Current challenges and unmet medical needs in myelodysplastic syndromes. Leukemia 2021; 35:2182-2198. [PMID: 34045662 PMCID: PMC8324480 DOI: 10.1038/s41375-021-01265-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/01/2021] [Accepted: 04/26/2021] [Indexed: 01/29/2023]
Abstract
Myelodysplastic syndromes (MDS) represent a heterogeneous group of myeloid neoplasms that are characterized by ineffective hematopoiesis, variable cytopenias, and a risk of progression to acute myeloid leukemia. Most patients with MDS are affected by anemia and anemia-related symptoms, which negatively impact their quality of life. While many patients with MDS have lower-risk disease and are managed by existing treatments, there currently is no clear standard of care for many patients. For patients with higher-risk disease, the treatment priority is changing the natural history of the disease by delaying disease progression to acute myeloid leukemia and improving overall survival. However, existing treatments for MDS are generally not curative and many patients experience relapse or resistance to first-line treatment. Thus, there remains an unmet need for new, more effective but tolerable strategies to manage MDS. Recent advances in molecular diagnostics have improved our understanding of the pathogenesis of MDS, and it is becoming clear that the diverse nature of genetic abnormalities that drive MDS demands a complex and personalized treatment approach. This review will discuss some of the challenges related to the current MDS treatment landscape, as well as new approaches currently in development.
Collapse
Affiliation(s)
- Uwe Platzbecker
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany.
- German MDS Study Group (D-MDS), Leipzig, Germany.
- The European Myelodysplastic Syndromes Cooperative Group (EMSCO), Leipzig, Germany.
| | - Anne Sophie Kubasch
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
- German MDS Study Group (D-MDS), Leipzig, Germany
- The European Myelodysplastic Syndromes Cooperative Group (EMSCO), Leipzig, Germany
| | | | | |
Collapse
|
13
|
Rautenberg C, Lauseker M, Kaivers J, Jäger P, Fischermanns C, Pechtel S, Haas R, Kobbe G, Germing U, Schroeder T. Prognostic impact of pretransplant measurable residual disease assessed by peripheral blood WT1-mRNA expression in patients with AML and MDS. Eur J Haematol 2021; 107:283-292. [PMID: 33987857 DOI: 10.1111/ejh.13664] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVE As peripheral blood (PB) Wilm's Tumor 1 (WT1)-mRNA expression is established as MRD-marker during conventional AML chemotherapy, impact of pretransplant WT1 expression remains unclear. Therefore, we aimed to assess prognostic impact of pretransplant WT1 expression on post-transplant outcome in patients with AML/MDS. METHODS In 64 AML/MDS patients, pretransplant WT1 expression was retrospectively analyzed using a standardized assay offering high sensitivity, specificity, and a validated cut-off. Patients were divided into three groups determined by pretransplant remission and WT1 expression. Post-transplant outcome of these groups was compared regarding cumulative incidence of relapse (CIR), relapse-free (RFS), and overall survival (OS). RESULTS Pretransplant forty-six patients (72%) showed hematologic remission, including 21 (46%) MRD-negative and 25 (54%) MRD-positive patients indicated by WT1 expression, while 18 refractory patients (28%) showed active disease. Two-year estimates of post-transplant CIR, RFS, and OS were similar in MRD-positive (61%, 37%, 54%) and refractory patients (70%, 26%, 56%), but significantly inferior compared with MRD-negative patients (10%, 89%, 90%). After multivariable adjustment, pretransplant MRD negativity measured by WT1 expression retained its prognostic impact on CIR (P = .008), RFS (P = .005), and OS (P = .049). CONCLUSIONS PB WT1 expression represents a useful method to estimate pretransplant MRD, which is highly predictable for post-transplant outcome and may help improving peri-transplant management in AML/MDS patients.
Collapse
Affiliation(s)
- Christina Rautenberg
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine - University, Duesseldorf, Germany
| | - Michael Lauseker
- Institute for Medical Information Processing, Biometry and Epidemiology, Ludwig-Maximilians-University, Munich, Germany
| | - Jennifer Kaivers
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine - University, Duesseldorf, Germany
| | - Paul Jäger
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine - University, Duesseldorf, Germany
| | - Carolin Fischermanns
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine - University, Duesseldorf, Germany
| | - Sabrina Pechtel
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine - University, Duesseldorf, Germany
| | - Rainer Haas
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine - University, Duesseldorf, Germany
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine - University, Duesseldorf, Germany
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine - University, Duesseldorf, Germany
| | - Thomas Schroeder
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine - University, Duesseldorf, Germany.,Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Medical Faculty, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
14
|
Descriptive and Functional Genomics in Acute Myeloid Leukemia (AML): Paving the Road for a Cure. Cancers (Basel) 2021; 13:cancers13040748. [PMID: 33670178 PMCID: PMC7916915 DOI: 10.3390/cancers13040748] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/24/2021] [Accepted: 02/01/2021] [Indexed: 12/18/2022] Open
Abstract
Over the past decades, genetic advances have allowed a more precise molecular characterization of AML with the identification of novel oncogenes and tumor suppressors as part of a comprehensive AML molecular landscape. Recent advances in genetic sequencing tools also enabled a better understanding of AML leukemogenesis from the preleukemic state to posttherapy relapse. These advances resulted in direct clinical implications with the definition of molecular prognosis classifications, the development of treatment recommendations based on minimal residual disease (MRD) measurement and the discovery of novel targeted therapies, ultimately improving AML patients' overall survival. The more recent development of functional genomic studies, pushed by novel molecular biology technologies (short hairpin RNA (shRNA) and CRISPR-Cas9) and bioinformatics tools design on one hand, along with the engineering of humanized physiologically relevant animal models on the other hand, have opened a new genomics era resulting in a greater knowledge of AML physiopathology. Combining descriptive and functional genomics will undoubtedly open the road for an AML cure within the next decades.
Collapse
|
15
|
How I treat measurable (minimal) residual disease in acute leukemia after allogeneic hematopoietic cell transplantation. Blood 2020; 135:1639-1649. [PMID: 31961921 DOI: 10.1182/blood.2019003566] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/18/2020] [Indexed: 12/12/2022] Open
Abstract
Although allogeneic hematopoietic cell transplantation (allo-HCT) is currently the standard curative treatment of acute leukemia, relapse remains unacceptably high. Measurable (minimal) residual disease (MRD) after allo-HCT may be used as a predictor of impending relapse and should be part of routine follow-up for transplanted patients. Patients with MRD may respond to therapies aiming to unleash or enhance the graft-versus-leukemia effect. However, evidence-based recommendations on how to best implement MRD testing and MRD-directed therapy after allo-HCT are lacking. Here, I describe our institutional approach to MRD monitoring for preemptive MRD-triggered intervention, using patient scenarios to illustrate the discussion.
Collapse
|
16
|
Antar AI, Otrock ZK, Abou Dalle I, El-Cheikh J, Bazarbachi A. Pharmacologic Therapies to Prevent Relapse of Acute Myeloid Leukemia After Allogeneic Hematopoietic Stem Cell Transplantation. Front Oncol 2020; 10:596134. [PMID: 33224890 PMCID: PMC7667262 DOI: 10.3389/fonc.2020.596134] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/09/2020] [Indexed: 12/29/2022] Open
Abstract
Relapse is the main cause of mortality in patients with acute myeloid leukemia (AML) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Adverse cytogenetic or molecular risk factors, as well as refractory disease or persistent measurable residual disease (MRD) at the time of transplantation are associated with an increased risk of recurrence. Salvage therapy for AML relapse after allo-HSCT is often limited to chemotherapy, donor lymphocyte infusions and/or second transplants and is rarely successful. Effective post-transplant preventive intervention in high risk AML may be crucial. The most frequent and promising approach is the use of post-transplant maintenance with hypomethylating agents or with FLT3 tyrosine kinase inhibitors when the target is present. Moreover, IDH1/IDH2 inhibitors and BCL-2 inhibitors in combination with other strategies are promising approaches in the maintenance setting. Here we summarize the current knowledge about the preemptive and prophylactic use of pharmacologic agents after allo-HSCT to prevent relapse of AML.
Collapse
Affiliation(s)
- Ahmad I. Antar
- Almoosa Specialist Hospital, Department of Internal Medicine, Division of Hematology-Oncology, Al-Ahsa, Saudi Arabia
- Department of Hematology and Oncology, Hammoud Hospital University Medical Center, Saida, Lebanon
| | - Zaher K. Otrock
- Department of Pathology and Laboratory Medicine, Henry Ford Hospital, Detroit, MI, United States
| | - Iman Abou Dalle
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Jean El-Cheikh
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
17
|
Cooperrider JH, Fulton N, Artz AS, Larson RA, Stock W, Kosuri S, Bishop M, Liu H. Phase I trial of maintenance selinexor after allogeneic hematopoietic stem cell transplantation for patients with acute myeloid leukemia and myelodysplastic syndrome. Bone Marrow Transplant 2020; 55:2204-2206. [PMID: 32376970 DOI: 10.1038/s41409-020-0925-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 11/08/2022]
Affiliation(s)
- Jennifer H Cooperrider
- Department of Internal Medicine, University of Chicago Medicine, 5841S Maryland Ave, Chicago, IL, 60637, USA
| | - Noreen Fulton
- Section of Hematology/Oncology, University of Chicago Medicine, 5841S Maryland Ave, Chicago, IL, 60637, USA
| | - Andrew S Artz
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Richard A Larson
- Section of Hematology/Oncology, University of Chicago Medicine, 5841S Maryland Ave, Chicago, IL, 60637, USA
| | - Wendy Stock
- Section of Hematology/Oncology, University of Chicago Medicine, 5841S Maryland Ave, Chicago, IL, 60637, USA
| | - Satyajit Kosuri
- Section of Hematology/Oncology, University of Chicago Medicine, 5841S Maryland Ave, Chicago, IL, 60637, USA
| | - Michael Bishop
- Section of Hematology/Oncology, University of Chicago Medicine, 5841S Maryland Ave, Chicago, IL, 60637, USA
| | - Hongtao Liu
- Section of Hematology/Oncology, University of Chicago Medicine, 5841S Maryland Ave, Chicago, IL, 60637, USA.
| |
Collapse
|
18
|
Valkova V, Vydra J, Markova M, Cerovska E, Vrana M, Marinov I, Cechova H, Cetkovsky P, Vitek A, Salek C. WT1 Gene Expression in Peripheral Blood Before and After Allogeneic Stem Cell Transplantation is a Clinically Relevant Prognostic Marker in AML - A Single-center 14-year Experience. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 21:e145-e151. [PMID: 33160932 DOI: 10.1016/j.clml.2020.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND This work summarizes our experience with WT1 monitoring before and after allogeneic hematopoietic stem cell transplantation (allo-HSCT). PATIENTS AND METHODS The expression of WT1 gene was measured by real-time polymerase chain reaction in peripheral blood according to the European Leukemia Net recommendations. Between May 2005 and August 2019, we analyzed 147 consecutive patients with acute myeloid leukemia with high WT1 expression at diagnosis, transplanted in first (CR1) or second (CR2) complete remission. RESULTS At the time of allo-HSCT, 107 patients had WT1-normal expression (WT1 ≤ 50 copies), and 40 patients had WT1-high expression. The median follow-up was 21 months. The estimated 5-year overall survival and event-free survival was significantly better in the WT1-normal cohort (65% and 57% vs. 37% and 25%; P = .0003 and P < .0001, respectively) and 5-year cumulative incidence of relapse was significantly lower in the WT1-normal group (19% vs. 53%; P < .0001). Five-year non-relapse mortality was not significantly different (20% and 23%). Multivariate analysis revealed WT1-high expression and acute graft-versus-host disease grade 3/4 as significantly negative prognostic factors for OS. Overall, 49 patients developed WT1 molecular relapse in the post-transplant period; in 14 cases, the therapeutic intervention was done. In all but 1 relapsed patient where WT1 minimal residual disease (MRD) was monitored (38 patients), we detected WT1-high levels (sensitivity of 97%). CONCLUSION The results of the analysis confirmed our previous experience that WT1 status before allo-HSCT is a strong prognostic factor for both OS and relapse risk. In addition, we confirmed the usefulness of this marker for MRD monitoring after allo-HSCT. The main advantage is the possibility of frequent MRD monitoring in peripheral blood and early bone marrow examination based on WT1-high expression.
Collapse
MESH Headings
- Adult
- Aged
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/metabolism
- Disease-Free Survival
- Feasibility Studies
- Female
- Follow-Up Studies
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Graft vs Host Disease/diagnosis
- Graft vs Host Disease/epidemiology
- Hematopoietic Stem Cell Transplantation/adverse effects
- Humans
- Incidence
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/therapy
- Male
- Middle Aged
- Neoplasm Recurrence, Local/epidemiology
- Neoplasm Recurrence, Local/genetics
- Neoplasm, Residual
- Prognosis
- Risk Assessment/methods
- Risk Factors
- Severity of Illness Index
- WT1 Proteins/blood
- WT1 Proteins/metabolism
- Young Adult
Collapse
Affiliation(s)
- Veronika Valkova
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Jan Vydra
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marketa Markova
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ela Cerovska
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Milena Vrana
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Iuri Marinov
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Hana Cechova
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Petr Cetkovsky
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Antonin Vitek
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Cyril Salek
- Department of Bone Marrow Transplant, Institute of Hematology and Blood Transfusion, Prague, Czech Republic; Institute of Clinical and Experimental Hematology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
19
|
Wilm's Tumor 1-guided preemptive treatment with hypomethylating agents for molecular relapse of AML and MDS after allogeneic transplantation. Bone Marrow Transplant 2020; 56:442-450. [PMID: 32873914 DOI: 10.1038/s41409-020-01039-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/01/2020] [Accepted: 08/18/2020] [Indexed: 12/25/2022]
Abstract
Hypomethylating agents (HMA) for relapsed acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) after allogeneic transplantation (allo-SCT) are most effective when used at the stage of molecular relapse. As Wilm's Tumor 1 (WT1)- expression has proven to serve as broadly applicable, sensitive and specific minimal residual disease (MRD) marker, we measured WT1-expression in 35 AML and MDS patients using a standardized assay for the guidance of therapy with HMA and donor lymphocyte infusions (DLI). Molecular relapse was detected in median 168 days post-transplant prompting therapy with a median of six HMA cycles and at least one DLI (n = 22, 63%). Hereby, 13 patients (37%) achieved major response (=MRD- complete remission [CR]), and 7 patients (20%) achieved minor response (=MRD+ CR), whereas 15 patients (43%) progressed into hematologic relapse. Two-year overall survival (OS) rate was 35% including 11 patients (31%) with ongoing MRD- remission for a median of 21 months. Patients with the major response after six cycles had significantly better OS suggesting that those not achieving MRD negativity after six cycles are candidates for alternative therapies. Combining MRD-monitoring of WT1-expression and preemptive therapy with HMA and DLI appears as a practicable and efficient approach for imminent relapse after allo-SCT.
Collapse
|
20
|
Chen S, Zeiser R. Novel Biomarkers for Outcome After Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2020; 11:1854. [PMID: 33013836 PMCID: PMC7461883 DOI: 10.3389/fimmu.2020.01854] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 07/09/2020] [Indexed: 12/29/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a well-established curative treatment for various malignant hematological diseases. However, its clinical success is substantially limited by major complications including graft-vs.-host disease (GVHD) and relapse of the underlying disease. Although these complications are known to lead to significant morbidity and mortality, standardized pathways for risk stratification of patients undergoing allo-HSCT are lacking. Recent advances in the development of diagnostic and prognostic tools have allowed the identification of biomarkers in order to predict outcome after allo-HSCT. This review will provide a summary of clinically relevant biomarkers that have been studied to predict the development of acute GVHD, the responsiveness of affected patients to immunosuppressive treatment and the risk of non-relapse mortality. Furthermore, biomarkers associated with increased risk of relapse and subsequent mortality will be discussed.
Collapse
Affiliation(s)
- Sophia Chen
- Department of Immunology, Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute, New York, NY, United States.,Department of Medicine I, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
21
|
Rautenberg C, Bergmann A, Germing U, Fischermanns C, Pechtel S, Kaivers J, Jäger P, Schuler E, Haas R, Kobbe G, Schroeder T. Prediction of Response and Survival Following Treatment with Azacitidine for Relapse of Acute Myeloid Leukemia and Myelodysplastic Syndromes after Allogeneic Hematopoietic Stem Cell Transplantation. Cancers (Basel) 2020; 12:cancers12082255. [PMID: 32806572 PMCID: PMC7464210 DOI: 10.3390/cancers12082255] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/04/2020] [Accepted: 08/08/2020] [Indexed: 12/23/2022] Open
Abstract
To provide long-term outcome data and predictors for response and survival, we retrospectively analyzed all 151 patients with relapse of myeloid neoplasms after allogeneic hematopoietic stem cell transplantation (allo-HSCT) who were uniformly treated with first-line azacitidine (Aza) salvage therapy at our center. Patients were treated for molecular (39%) or hematologic relapse (61%), with a median of 5 cycles of Aza and at least one donor lymphocyte infusion in 70% of patients. Overall response was 46%, with 41% achieving complete (CR) and 5% achieving partial remission. CR was achieved after a median of 4 cycles and lasted for a median of 11 months (range 0.9 to 120 months). With a median follow-up of 22 months (range: 1 to 122 months), the 2-year survival rate was 38% ± 9%, including 17 patients with ongoing remission for >5 years. Based on results from multivariate analyses, molecular relapse and time to relapse were integrated into a score, clearly dividing patients into 3 subgroups with CR rates of 71%, 39%, and 29%; and 2-year survival rates of 64%, 38%, and 27%, respectively. In the subgroup of MDS and secondary AML, receiving upfront transplantation was associated with superior response and survival, and therefore pretransplant strategy was integrated together with relapse type into a MDS–sAML-specific score. Overall, Aza enables meaningful responses and long-term survival, which is a predictable with a simple-to-use scoring system.
Collapse
|
22
|
The Proportional Relationship Between Pretransplant WT1 mRNA Levels and Risk of Mortality After Allogeneic Hematopoietic Cell Transplantation in Acute Myeloid Leukemia Not in Remission. Transplantation 2020; 103:2201-2210. [PMID: 30801534 DOI: 10.1097/tp.0000000000002662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The relationship between the expression levels of Wilms' tumor-1 gene (WT1) mRNA in peripheral blood before allogeneic hematopoietic cell transplantation (allo-HCT) and risk of mortality in acute myeloid leukemia (AML) patients in noncomplete remission (non-CR) remains quite elusive. METHODS We retrospectively assessed the impact of the pretransplant WT1 mRNA level on survival after allo-HCT in non-CR AML patients. RESULTS A total of 125 AML patients, including 46 non-CR patients (36.8%), were analyzed. On multivariate analysis of non-CR AML patients, WT1 mRNA ≥5000 copies/μg RNA was significantly related to increased risk of mortality (hazard ratio, 2.7; 95% confidence interval, 1.3-5.5; P = 0.008). Furthermore, in the entire cohort, log10-transformed WT1 mRNA before allo-HCT was found to be significantly associated with the increased risk of mortality irrespective of whether the disease status was CR or non-CR, using Akaike's information criterion. As the pretransplant WT1 mRNA level elevated, the hazard ratio of mortality monotonically increased in a nonlinear manner regardless of remission status, suggesting that WT1 mRNA level in peripheral blood might reflect tumor burden. CONCLUSIONS This study demonstrated that the pretransplant WT1 mRNA level was a powerful prognostic factor in allo-HCT even for non-CR AML patients, and there may be a WT1 mRNA threshold in non-CR patients for benefiting from allo-HCT.
Collapse
|
23
|
Luo P, Jing W, Yi K, Wu S, Zhou F. Wilms' tumor 1 gene in hematopoietic malignancies: clinical implications and future directions. Leuk Lymphoma 2020; 61:2059-2067. [PMID: 32401109 DOI: 10.1080/10428194.2020.1762884] [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] [Indexed: 02/08/2023]
Abstract
The Wilms' tumor 1 (WT1) gene is an important regulatory molecule that plays a vital role in cell growth and development. Initially, knowledge of WT1 was mostly limited to Wilms' tumor. Over the past years, numerous studies have shown that WT1 is aberrant expressed or mutated in hematopoietic malignancies, including acute leukemia (AL), myelodysplastic syndrome (MDS) and chronic myelogenous leukemia (CML). Currently, many studies focus on exploring the role of WT1 in hematopoietic malignancies. Such studies improve the understanding of hematopoietic malignancies, and the collection of data about WT1 expression or mutation in hematopoietic malignancies over the past years can facilitate the risk stratification of hematopoietic malignancies. In this review, we highlight the important role of WT1 in hematopoietic malignancies, discuss its potential clinical applications as a minimal residual disease (MRD) and prognostic biomarker, and evaluate the possible therapy target of WT1 in hematopoietic malignancies.
Collapse
Affiliation(s)
- Ping Luo
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei Jing
- Department of Clinical Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kezhen Yi
- Department of Clinical Laboratory Medicine & Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Sanyun Wu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
24
|
Malagola M, Bernardi S, Polverelli N, Russo D. Minimal residual disease monitoring in acute myeloid leukaemia: are we ready to move from bone marrow to peripheral blood? Br J Haematol 2020; 190:135-136. [PMID: 32191348 DOI: 10.1111/bjh.16579] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Michele Malagola
- Chair of Hematology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Unit of Blood Disease and Stem Cell Transplantation, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Simona Bernardi
- Chair of Hematology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Unit of Blood Disease and Stem Cell Transplantation, ASST Spedali Civili of Brescia, Brescia, Italy.,CREA Laboratory (Hematological-Research AIL Centre), ASST-Spedali Civili Brescia, Brescia, Italy
| | - Nicola Polverelli
- Chair of Hematology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Unit of Blood Disease and Stem Cell Transplantation, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Domenico Russo
- Chair of Hematology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Unit of Blood Disease and Stem Cell Transplantation, ASST Spedali Civili of Brescia, Brescia, Italy
| |
Collapse
|
25
|
Novel Disease Risk Model for Patients with Acute Myeloid Leukemia Receiving Allogeneic Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2020; 26:197-203. [DOI: 10.1016/j.bbmt.2019.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/01/2019] [Accepted: 09/04/2019] [Indexed: 02/04/2023]
|
26
|
Andreani G, Cilloni D. Strategies for minimal residual disease detection: current perspectives. BLOOD AND LYMPHATIC CANCER-TARGETS AND THERAPY 2019; 9:1-8. [PMID: 31807111 PMCID: PMC6855617 DOI: 10.2147/blctt.s172693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 12/13/2018] [Indexed: 11/25/2022]
Abstract
Currently, the post-remission treatment in acute leukemia is based on the genetic profile of leukemic cells at diagnosis (ie, FLT3 ITD positivity) and on the level of measurable residual disease (MRD) after induction and consolidation chemotherapy. Two methods are currently preferred for MRD evaluation in many centers: multiparameter flow cytometry and real-time quantitative PCR. Additional methods such as next-generation sequencing and digital PCR are under investigation, in an attempt to increase the sensitivity and thus allowing the detection of small clones. Many studies suggest that MRD positivity after chemotherapy is associated with negative prognosis, and the reappearance of MRD during follow-up allows impending relapse to be identified and consequently enables early intervention. Finally, MRD positivity before hematopoietic stem cell transplantation is predictive of the outcome. Although the significance of MRD in acute leukemia has been widely explored, the assessment of molecular MRD is not yet a routine practice. In this review, we describe the significance of MRD in different settings and the main markers and methods used for MRD detection.
Collapse
Affiliation(s)
- Giacomo Andreani
- Department of Clinical and Biological Sciences, University of Turin, Turin,Italy
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, Turin,Italy
| |
Collapse
|
27
|
Cloos J, Ossenkoppele GJ, Dillon R. Minimal residual disease and stem cell transplantation outcomes. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:617-625. [PMID: 31808862 PMCID: PMC6913494 DOI: 10.1182/hematology.2019000006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Risk classification and tailoring of treatment are essential for improving outcome for patients with acute myeloid leukemia or high-risk myelodysplastic syndrome. Both patient and leukemia-specific characteristics assessed using morphology, cytogenetics, molecular biology, and multicolor flow cytometry are relevant at diagnosis and during induction, consolidation, and maintenance phases of the treatment. In particular, minimal residual disease (MRD) during therapy has potential as a prognostic factor of outcome, determination of response to therapy, and direction of targeted therapy. MRD can be determined by cell surface markers using multicolor flow cytometry, whereas leukemia-specific translocations and mutations are measured using polymerase chain reaction-based techniques and recently using next-generation sequencing. All these methods of MRD detection have their (dis)advantages, and all need to be standardized, prospectively validated, and improved to be used for uniform clinical decision making and a potential surrogate end point for clinical trials testing novel treatment strategies. Important issues to be solved are time point of MRD measurement and threshold for MRD positivity. MRD is used for stem cell transplantation (SCT) selection in the large subgroup of patients with an intermediate risk profile. Patients who are MRD positive will benefit from allo-SCT. However, MRD-negative patients have a better chance of survival after SCT. Therefore, it is debated whether MRD-positive patients should be extensively treated to become MRD negative before SCT. Either way, accurate monitoring of potential residual or upcoming disease is mandatory. Tailoring therapy according to MRD monitoring may be the most successful way to provide appropriate specifically targeted, personalized treatment.
Collapse
Affiliation(s)
- Jacqueline Cloos
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, VUMC, Amsterdam, The Netherlands; and
| | - Gert J Ossenkoppele
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, VUMC, Amsterdam, The Netherlands; and
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College, London, United Kingdom
| |
Collapse
|
28
|
Bouvier A, Riou J, Thépot S, Sutra Del Galy A, François S, Schmidt A, Orvain C, Estienne MH, Villate A, Luque Paz D, Cottin L, Ribourtout B, Beucher A, Delneste Y, Ifrah N, Ugo V, Hunault-Berger M, Blanchet O. Quantitative chimerism in CD3-negative mononuclear cells predicts prognosis in acute myeloid leukemia patients after hematopoietic stem cell transplantation. Leukemia 2019; 34:1342-1353. [PMID: 31768015 DOI: 10.1038/s41375-019-0624-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 10/18/2019] [Accepted: 11/03/2019] [Indexed: 12/20/2022]
Abstract
Relapse is a major complication of acute myeloid leukemia (AML) after allogeneic hematopoietic stem cell transplantation (SCT). The objective of our study was to evaluate chimerism monitoring on the CD3-negative mononuclear cells by RQ-PCR to predict relapse of patients allografted for AML and to compare its performance with WT1 quantification. A cohort of 100 patients undergoing allogenic SCT for AML was retrospectively analyzed in a single institution. Patients without complete chimerism, defined as less than 0.01% of recipient's DNA in CD3-negative cells, had a significantly higher risk of relapse and a lower overall survival (p < 0.001). An increase in the percentage of recipient DNA in CD3-negative cells was associated with an increased risk of relapse (p < 0.001) but not with overall survival. Comparable performances between monitoring of CD3-negative cell chimerism and WT1 expression to predict relapse was observed up to more than 90 days before hematological relapse, with sensitivity of 82% and 78%, respectively, and specificity of 100% for both approaches. Quantitative specific chimerism of the CD3-negative mononuclear fraction, enriched in blastic cells, is a new and powerful tool for monitoring measurable residual disease and could be used for AML patients without available molecular markers.
Collapse
Affiliation(s)
- Anne Bouvier
- CHU Angers, Laboratoire d'Hématologie, Angers, France. .,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France. .,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.
| | - Jérémie Riou
- MINT, UMR INSERM 1066, CNRS 6021, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France
| | - Sylvain Thépot
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | | | - Sylvie François
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Aline Schmidt
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Corentin Orvain
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Marie-Hélène Estienne
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CHU Tours, Service d'Hématologie biologique, Tours, France
| | - Alban Villate
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CHU Tours, Service d'Hématologie et thérapie cellulaire, Tours, France
| | - Damien Luque Paz
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France
| | - Laurane Cottin
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,Université d'Angers, UFR Santé, Angers, France
| | - Bénédicte Ribourtout
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France
| | - Annaëlle Beucher
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France
| | - Yves Delneste
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,CHU Angers, Laboratoire d'Immunologie et Allergologie, Angers, France
| | - Norbert Ifrah
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Valérie Ugo
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France
| | - Mathilde Hunault-Berger
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Service des Maladies du sang, Angers, France
| | - Odile Blanchet
- CHU Angers, Laboratoire d'Hématologie, Angers, France.,Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL), Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Université d'Angers, UFR Santé, Angers, France.,CHU Angers, Centre de Ressources Biologiques, BB-0033-00038, Angers, France
| |
Collapse
|
29
|
Qin YZ, Wang Y, Xu LP, Zhang XH, Zhao XS, Liu KY, Huang XJ. Subgroup Analysis Can Optimize the Relapse-Prediction Cutoff Value for WT1 Expression After Allogeneic Hematologic Stem Cell Transplantation in Acute Myeloid Leukemia. J Mol Diagn 2019; 22:188-195. [PMID: 31751675 DOI: 10.1016/j.jmoldx.2019.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/03/2019] [Indexed: 01/06/2023] Open
Abstract
High WT1 expression after allogeneic hematologic stem cell transplantation (allo-HSCT) can strongly predict relapse in acute myeloid leukemia (AML). However, the cutoff values obtained have been inconsistent. Precise cutoff values may be optimized through subtype analysis; the RUNX1-RUNX1T1 fusion transcript provides an ideal reference. RUNX1-RUNX1T1 and WT1 transcript levels were simultaneously measured in 1299 bone marrow samples serially collected from 176 t(8;21) AML patients after allo-HSCT. The upper limit of the normal bone marrow WT1 level was 0.6%, which we previously reported to be the cutoff value for significant relapse prediction in AML as a whole. WT1 cutoff values of 0.6%, 1.2%, and 1.8% significantly differentiated patients in relapse after allo-HSCT. Nonetheless, patients with WT1 levels of 0.6% to 1.2% and those with levels of >1.2% and 1.8% after HSCT had rates of cumulative incidence of relapse similar to those with a continuous WT1 level of ≤0.6%, and both were significantly lower than that in patients with a WT1 level of >1.8%. WT1 expression was significantly related to RUNX1-RUNX1T1 transcript levels at WT1 levels of >1.8% but not at levels of 0.6% to 1.2% or >1.2% to 1.8%. Therefore, subgroup analysis can optimize the relapse-prediction cutoff value of WT1 expression. A cutoff level of 1.8% more accurately differentiates t(8;21) AML patients in relapse after allo-HSCT than does a cutoff level of 0.6%.
Collapse
Affiliation(s)
- Ya-Zhen Qin
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, People's Republic of China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, People's Republic of China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, People's Republic of China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, People's Republic of China
| | - Xiao-Su Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, People's Republic of China
| | - Kai-Yan Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, People's Republic of China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, People's Republic of China.
| |
Collapse
|
30
|
Qin YZ, Zhu LW, Lin S, Geng SX, Liu SW, Cheng H, Wu CY, Xiao M, Li XQ, Hu RP, Wang LL, Liu HY, Ma DX, Guan T, Ye YX, Niu T, Cen JN, Lu LS, Sun L, Yang TH, Wang YG, Li T, Wang Y, Li QH, Zhao XS, Li LD, Chen WM, Long LY, Huang XJ. [An interlaboratory comparison study on the detection of RUNX1-RUNX1T1 fusion transcript levels and WT1 transcript levels]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:889-894. [PMID: 31856435 PMCID: PMC7342382 DOI: 10.3760/cma.j.issn.0253-2727.2019.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Indexed: 02/05/2023]
Abstract
Objective: To investigate the current status and real performance of the detection of RUNX1-RUNX1T1 fusion transcript levels and WT1 transcript levels in China through interlaboratory comparison. Methods: Peking University People's Hospital (PKUPH) prepared the samples for comparison. That is, the fresh RUNX1-RUNX1T1 positive (+) bone morrow nucleated cells were serially diluted with RUNX1-RUNX1T1 negative (-) nucleated cells from different patients. Totally 23 sets with 14 different samples per set were prepared. TRIzol reagent was added in each tube and thoroughly mixed with cells for homogenization. Each laboratory simultaneously tested RUNX1-RUNX1T1 and WT1 transcript levels of one set of samples by real-time quantitative PCR method. All transcript levels were reported as the percentage of RUNX1-RUNX1T1 or WT1 transcript copies/ABL copies. Spearman correlation coefficient between the reported transcript levels of each participated laboratory and those of PKUPH was calculated. Results: ①RUNX1-RUNX1T1 comparison: 9 samples were (+) and 5 were (-) , the false negative and positive rates of the 20 participated laboratories were 0 (0/180) and 5% (5/100) , respectively. The reported transcript levels of all 9 positive samples were different among laboratories. The median reported transcript levels of 9 positive samples were from 0.060% to 176.7%, which covered 3.5-log. The ratios of each sample's highest to the lowest reported transcript levels were from 5.5 to 12.3 (one result which obviously deviated from other laboratories' results was not included) , 85% (17/20) of the laboratories had correlation coefficient ≥0.98. ②WT1 comparison: The median reported transcript levels of all 14 samples were from 0.17% to 67.6%, which covered 2.6-log. The ratios of each sample's highest to the lowest reported transcript levels were from 5.3-13.7, 62% (13/21) of the laboratories had correlation coefficient ≥0.98. ③ The relative relationship of the reported RUNX1-RUNX1T1 transcript levels between the participants and PKUPH was not always consistent with that of WT1 transcript levels. Both RUNX1-RUNX1T1 and WT1 transcript levels from 2 and 7 laboratories were individually lower than and higher than those of PKUPH, whereas for the rest 11 laboratories, one transcript level was higher than and the other was lower than that of PKUPH. Conclusion: The reported RUNX1-RUNX1T1 and WT1 transcript levels were different among laboratories for the same sample. Most of the participated laboratories reported highly consistent result with that of PKUPH. The relationship between laboratories of the different transcript levels may not be the same.
Collapse
Affiliation(s)
- Y Z Qin
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| | - L W Zhu
- Beijing Hightrust Diagnostics Co., Ltd, Beijing 100176, China
| | - S Lin
- Department of Hematology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - S X Geng
- Department of Hematology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - S W Liu
- Harbin Institute of Hematology and Oncology, Harbin 150010, China
| | - H Cheng
- Changhai Hospital, The Second Military Medical University, Shanghai 200433, China
| | - C Y Wu
- Institute of Hematology, Henan Provincial People's Hospital, Zhengzhou 450003, China
| | - M Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030
| | - X Q Li
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022
| | - R P Hu
- Department of Hematology, Bethune First Affiliated Hospital of Jilin University, Changchun 130021
| | - L L Wang
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - H Y Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - D X Ma
- Department of Hematology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - T Guan
- Department of Hematology, Shanxi Provincial Cancer Hospital, Taiyuan 030000, China
| | - Y X Ye
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - T Niu
- Department of Hematology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - J N Cen
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Disease, Suzhou 215006, China
| | - L S Lu
- Tianjin Sino-us Diagnostics Co., Ltd, Tianjin 301617, China
| | - L Sun
- Wuhan Kindstar Diagnostics Co., Ltd, Wuhan 430075, China
| | - T H Yang
- Department of Hematology, The First People's Hospital of Yunnan Province, Kunming 650034, China
| | - Y G Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China
| | - T Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Y Wang
- The First Hospital of China Medical University, Shenyang 110001, China
| | - Q H Li
- Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Hematologic Disease, Tianjin 300020, China
| | - X S Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| | - L D Li
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| | - W M Chen
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| | - L Y Long
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| | - X J Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| |
Collapse
|
31
|
Rautenberg C, Germing U, Pechtel S, Lamers M, Fischermanns C, Jäger P, Geyh S, Haas R, Kobbe G, Schroeder T. Prognostic impact of peripheral blood WT1-mRNA expression in patients with MDS. Blood Cancer J 2019; 9:86. [PMID: 31719523 PMCID: PMC6851368 DOI: 10.1038/s41408-019-0248-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/26/2019] [Accepted: 10/30/2019] [Indexed: 12/27/2022] Open
Abstract
Few reports suggested a prognostic impact of Wilms'Tumor-1 (WT1)-mRNA overexpression in MDS, but translation into clinical routine was hampered by limited patients numbers, differing sample sources, non-standardized methods/cut-offs. To evaluate whether WT1-mRNA expression yields additional prognostic information, we measured peripheral blood (PB) WT1-mRNA expression in 94 MDS using a standardized assay offering a validated cut-off to discriminate between normal and WT1-mRNA overexpression. Overall, 54 patients (57%) showed WT1-mRNA overexpression, while 40 patients (43%) had normal WT1-mRNA expression. This enabled discrimination between MDS and both healthy controls and non-MDS cytopenias. Furthermore, WT1-mRNA expression correlated with WHO 2016 subcategories and IPSS-R as indicated by mean WT1-mRNA expression and frequency of WT1-mRNA overexpressing patients within respective subgroups. Regarding the entire group, PB WT1-mRNA expression was associated with prognosis, as those patients showing WT1-mRNA overexpression had higher risk for disease progression and AML transformation and accordingly shorter progression-free, leukemia-free and overall survival in univariate analysis. In multivariate analysis, prognostic impact of PB WT1-mRNA expression status was independent of IPSS-R and enabled more precise prediction of PFS, but not OS, within IPSS-R very low/low and intermediate risk groups. Overall, measuring PB WT1-mRNA appears valuable to support diagnostics and refine prognostication provided by the IPSS-R.
Collapse
Affiliation(s)
- Christina Rautenberg
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Sabrina Pechtel
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Marius Lamers
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Carolin Fischermanns
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Paul Jäger
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Stefanie Geyh
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Rainer Haas
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Thomas Schroeder
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, Duesseldorf, Germany.
| |
Collapse
|
32
|
Mutated WT1, FLT3-ITD, and NUP98-NSD1 Fusion in Various Combinations Define a Poor Prognostic Group in Pediatric Acute Myeloid Leukemia. JOURNAL OF ONCOLOGY 2019; 2019:1609128. [PMID: 31467532 PMCID: PMC6699323 DOI: 10.1155/2019/1609128] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/24/2019] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia is a life-threatening malignancy in children and adolescents treated predominantly by risk-adapted intensive chemotherapy that is partly supported by allogeneic stem cell transplantation. Mutations in the WT1 gene and NUP98-NSD1 fusion are predictors of poor survival outcome/prognosis that frequently occur in combination with internal tandem duplications of the juxta-membrane domain of FLT3 (FLT3-ITD). To re-evaluate the effect of these factors in contemporary protocols, 353 patients (<18 years) treated in Germany with AML-BFM treatment protocols between 2004 and 2017 were included. Presence of mutated WT1 and FLT3-ITD in blasts (n=19) resulted in low 3-year event-free survival of 29% and overall survival of 33% compared to rates of 45-63% and 67-87% in patients with only one (only FLT3-ITD; n=33, only WT1 mutation; n=29) or none of these mutations (n=272). Including NUP98-NSD1 and high allelic ratio (AR) of FLT3-ITD (AR ≥0.4) in the analysis revealed very poor outcomes for patients with co-occurrence of all three factors or any of double combinations. All these patients (n=15) experienced events and the probability of overall survival was low (27%). We conclude that co-occurrence of WT1 mutation, NUP98-NSD1, and FLT3-ITD with an AR ≥0.4 as triple or double mutations still predicts dismal response to contemporary first- and second-line treatment for pediatric acute myeloid leukemia.
Collapse
|
33
|
Kobbe G, Schroeder T, Rautenberg C, Kaivers J, Gattermann N, Haas R, Germing U. Molecular genetics in allogeneic blood stem cell transplantation for myelodysplastic syndromes. Expert Rev Hematol 2019; 12:821-831. [DOI: 10.1080/17474086.2019.1645004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Guido Kobbe
- Departments of Hematology, Oncology and Clinical Immunology, Heinrich Heine University, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Thomas Schroeder
- Departments of Hematology, Oncology and Clinical Immunology, Heinrich Heine University, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Christina Rautenberg
- Departments of Hematology, Oncology and Clinical Immunology, Heinrich Heine University, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Jennifer Kaivers
- Departments of Hematology, Oncology and Clinical Immunology, Heinrich Heine University, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Norbert Gattermann
- Departments of Hematology, Oncology and Clinical Immunology, Heinrich Heine University, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Rainer Haas
- Departments of Hematology, Oncology and Clinical Immunology, Heinrich Heine University, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Ulrich Germing
- Departments of Hematology, Oncology and Clinical Immunology, Heinrich Heine University, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
34
|
Schuler E, Boughoufala S, Rautenberg C, Nachtkamp K, Dienst A, Fenk R, Haas R, Kondakci M, Germing U, Schroeder T, Kobbe G. Relapse patterns and treatment strategies in patients receiving allogeneic hematopoietic stem cell transplantation for myeloid malignancies. Ann Hematol 2019; 98:1225-1235. [DOI: 10.1007/s00277-019-03670-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 03/11/2019] [Indexed: 11/29/2022]
|
35
|
Rautenberg C, Germing U, Haas R, Kobbe G, Schroeder T. Relapse of Acute Myeloid Leukemia after Allogeneic Stem Cell Transplantation: Prevention, Detection, and Treatment. Int J Mol Sci 2019; 20:E228. [PMID: 30626126 PMCID: PMC6337734 DOI: 10.3390/ijms20010228] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) is a phenotypically and prognostically heterogeneous hematopoietic stem cell disease that may be cured in eligible patients with intensive chemotherapy and/or allogeneic stem cell transplantation (allo-SCT). Tremendous advances in sequencing technologies have revealed a large amount of molecular information which has markedly improved our understanding of the underlying pathophysiology and enables a better classification and risk estimation. Furthermore, with the approval of the FMS-like tyrosine kinase 3 (FLT3) inhibitor Midostaurin a first targeted therapy has been introduced into the first-line therapy of younger patients with FLT3-mutated AML and several other small molecules targeting molecular alterations such as isocitrate dehydrogenase (IDH) mutations or the anti-apoptotic b-cell lymphoma 2 (BCL-2) protein are currently under investigation. Despite these advances, many patients will have to undergo allo-SCT during the course of disease and depending on disease and risk status up to half of them will finally relapse after transplant. Here we review the current knowledge about the molecular landscape of AML and how this can be employed to prevent, detect and treat relapse of AML after allo-SCT.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Hematopoietic Stem Cell Transplantation
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/prevention & control
- Leukemia, Myeloid, Acute/therapy
- Mutation/genetics
- Neoplasm, Residual/diagnosis
- Neoplasm, Residual/drug therapy
- Recurrence
- Transplantation, Homologous
Collapse
Affiliation(s)
- Christina Rautenberg
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, 40225 Duesseldorf, Germany.
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, 40225 Duesseldorf, Germany.
| | - Rainer Haas
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, 40225 Duesseldorf, Germany.
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, 40225 Duesseldorf, Germany.
| | - Thomas Schroeder
- Department of Hematology, Oncology and Clinical Immunology, University of Duesseldorf, Medical Faculty, 40225 Duesseldorf, Germany.
| |
Collapse
|
36
|
Methods and role of minimal residual disease after stem cell transplantation. Bone Marrow Transplant 2018; 54:681-690. [PMID: 30116018 DOI: 10.1038/s41409-018-0307-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/28/2018] [Accepted: 06/13/2018] [Indexed: 11/08/2022]
Abstract
Relapse is the major cause of treatment failure after stem cell transplantation. Despite the fact that relapses occurred even if transplantation was performed in complete remission, it is obvious that minimal residual disease is present though not morphologically evident. Since adaptive immunotherapy by donor lymphocyte infusion or other novel cell therapies as well as less toxic drugs, which can be used after transplantation, the detection of minimal residual disease (MRD) has become a clinical important variable for outcome. Besides the increasing options to treat MRD, the most advanced technologies currently allow to detect residual malignant cells with a sensitivity of 10-5 to 10-6.Under the patronage of the European Society for Blood and Marrow Transplantation (EBMT) and the American Society for Blood and Marrow Transplantation (ASBMT) the 3rd workshop was held on 4/5 November 2016 in Hamburg/Germany, with the aim to present an up-to-date status of epidemiology and biology of relapse and to summarize the currently available options to prevent and treat post-transplant relapse. Here the current methods and role of minimal residual disease for myeloid and lymphoid malignancies are summarized.
Collapse
|