1
|
Feraille A, Etancelin P, Troche S, Jardin F, Buchbinder N, Schneider P, Dugay M, Rives N, Rondanino C, Dumont L. Detection of minimal residual disease in cryopreserved testicular tissue from (pre)pubertal boys with acute leukemia following first-line therapy. Hum Reprod 2025:deaf093. [PMID: 40389239 DOI: 10.1093/humrep/deaf093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/21/2025] [Indexed: 05/21/2025] Open
Abstract
STUDY QUESTION Is it feasible to detect minimal residual disease (MRD) in cryopreserved testicular tissue (TT) from (pre)pubertal boys diagnosed with acute leukemia using molecular biology techniques? SUMMARY ANSWER This pilot study demonstrates the feasibility of detecting MRD in cryopreserved TT, which could guide the choice of the safest techniques for fertility restoration. WHAT IS KNOWN ALREADY Fertility preservation through testicular tissue freezing (TTF) is offered to (pre)pubertal boys undergoing highly gonadotoxic treatment. However, the risk of reintroducing leukemic cells during fertility restoration has not been adequately addressed. To date, no study has evaluated the feasibility of detecting residual disease using molecular biology within cryopreserved, thawed and unfixed TT. STUDY DESIGN, SIZE, DURATION This pilot study analyzed cryopreserved TT from 14 (pre)pubertal boys diagnosed with acute lymphoblastic leukemia (ALL) or acute myeloblastic leukemia (AML) who had already received first-line chemotherapy and underwent TTF before hematopoietic stem cell transplantation. PARTICIPANTS/MATERIALS, SETTING, METHODS The study included cryopreserved TT from 14 (pre)pubertal boys. Molecular biology techniques, including RT-qPCR and qPCR, were used to detect oncogenic fusion genes or clonal rearrangements of immunoglobulin genes or T-cell receptor (Ig/TCR) in cryopreserved TT samples. MAIN RESULTS AND THE ROLE OF CHANCE MRD was identified in 36% (5 out of 14) of TT samples using molecular biology techniques. A 21% discordance was observed between conventional histopathology and molecular detection, with molecular methods showing higher sensitivity. No significant association was found between clinical or histological characteristics and MRD status in the TT. LIMITATIONS, REASONS FOR CAUTION This study is a pilot study with a small sample size of TT samples from patients with ALL or AML, which may limit the generalizability of the findings. Further studies with larger cohorts are needed to validate our data. WIDER IMPLICATIONS OF THE FINDINGS The detection of MRD in cryopreserved TT using molecular biology techniques could help guide the selection of the safest fertility restoration strategies for leukemic patients by minimizing the risk of reintroducing malignant cells. This approach underscores the importance of cryopreserving TT after complete remission of acute leukemia (AL). STUDY FUNDING/COMPETING INTEREST(S) The study was funded by Rouen University Hospital, GIRCI NO, French Biomedicine Agency, and Ligue National Contre le Cancer. The authors declare no competing interests. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Aurélie Feraille
- Biology of Reproduction-CECOS Laboratory, Rouen University Hospital, Team "Adrenal and Gonadal Pathophysiology", NorDIC, Inserm U1239, Univ Rouen Normandie, Rouen, France
| | | | - Shirley Troche
- Department of Genetic Oncology, Centre Henri Becquerel, Rouen, France
| | - Fabrice Jardin
- Department of Hematology, Centre Henri Becquerel, Rouen, France
- UMR918, Univ Rouen Normandie, Rouen, France
| | - Nimrod Buchbinder
- Department of Pediatric Hematology and Oncology, Rouen University Hospital, Univ Rouen Normandie, Rouen, France
| | - Pascale Schneider
- Department of Pediatric Hematology and Oncology, Rouen University Hospital, Univ Rouen Normandie, Rouen, France
| | - Magali Dugay
- Biology of Reproduction-CECOS Laboratory, Rouen University Hospital, Team "Adrenal and Gonadal Pathophysiology", NorDIC, Inserm U1239, Univ Rouen Normandie, Rouen, France
| | - Nathalie Rives
- Biology of Reproduction-CECOS Laboratory, Rouen University Hospital, Team "Adrenal and Gonadal Pathophysiology", NorDIC, Inserm U1239, Univ Rouen Normandie, Rouen, France
| | - Christine Rondanino
- Biology of Reproduction-CECOS Laboratory, Rouen University Hospital, Team "Adrenal and Gonadal Pathophysiology", NorDIC, Inserm U1239, Univ Rouen Normandie, Rouen, France
| | - Ludovic Dumont
- Biology of Reproduction-CECOS Laboratory, Rouen University Hospital, Team "Adrenal and Gonadal Pathophysiology", NorDIC, Inserm U1239, Univ Rouen Normandie, Rouen, France
| |
Collapse
|
2
|
Seferna K, Svaton M, Rennerova A, Skotnicova A, Reznickova L, Valova T, Sedlacek P, Riha P, Formankova R, Keslova P, Sramkova L, Stary J, Zuna J, Kolenova A, Salek C, Trka J, Fronkova E. NGS-MRD negativity in post-HSCT ALL spares unnecessary therapeutic interventions triggered by borderline qPCR results without an increase in relapse risk. Hemasphere 2025; 9:e70124. [PMID: 40201744 PMCID: PMC11978274 DOI: 10.1002/hem3.70124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/03/2025] [Accepted: 03/05/2025] [Indexed: 04/10/2025] Open
Abstract
Monitoring of minimal residual disease (MRD) after hematopoietic stem cell transplantation (HSCT) in patients with acute lymphoblastic leukemia (ALL) is vital for timely therapeutic intervention planning. However, interpreting low-positive results from the current standard method, quantitative PCR (qPCR) of immunoglobulin and T-cell receptor gene rearrangements (IG/TR), poses challenges due to the risk of false positivity caused by non-specific amplification. We aimed to improve MRD detection specificity using the next-generation amplicon sequencing (NGS) of IG/TR rearrangements for better relapse prediction. In pediatric and young adult ALL patients undergoing sequential post-HSCT MRD monitoring, we prospectively re-tested positive non-quantifiable qPCR results with NGS-MRD using the EuroClonality-NGS approach. We were able to confirm 13 out of 47 (27.7%) qPCR positive results using the more specific NGS-MRD method. Out of 10 patients with at least one MRD positivity confirmed by NGS, six relapsed (60%) 1-3.7 months after testing. Among 25 patients with all NGS-MRD results negative, two relapses occurred (8%) after 5.1 and 12.1 months. One-year RFS was 40% versus 96% and 3-year OS was 33.3% versus 94.4% for the NGS-positive and NGS-negative groups, respectively. The difference was not attributable to a varying rate of therapeutic interventions. Six patients out of 14 who had immunosuppressive treatment tapered or received donor lymphocyte infusion in response to MRD positivity developed significant graft versus host disease, leading to one fatality. This underscores the importance of enhancing the post-HSCT relapse risk prediction accuracy through NGS-MRD testing to avoid unnecessary interventions.
Collapse
Affiliation(s)
- Krystof Seferna
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Michael Svaton
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Andrea Rennerova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Aneta Skotnicova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Leona Reznickova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Tatana Valova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Petr Sedlacek
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Petr Riha
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Renata Formankova
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Petra Keslova
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Lucie Sramkova
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Jan Stary
- Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Jan Zuna
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Alexandra Kolenova
- Department of Pediatric Hematology and OncologyNational Institute of Children's Diseases and Comenius UniversityBratislavaSlovakia
| | - Cyril Salek
- Institute of Haematology and Blood TransfusionPragueCzechia
- Institute of Clinical and Experimental HaematologyFirst Faculty of Medicine, Charles UniversityPragueCzechia
| | - Jan Trka
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| | - Eva Fronkova
- CLIP—Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and OncologySecond Faculty of Medicine, Charles University and University Hospital MotolPragueCzechia
| |
Collapse
|
3
|
Kato M, Okamoto Y, Imamura T, Kada A, Saito AM, Iijima-Yamashita Y, Deguchi T, Ohki K, Fukushima T, Anami K, Sanada M, Taki T, Hashii Y, Inukai T, Kiyokawa N, Kosaka Y, Yoshida N, Yuza Y, Yanagimachi M, Watanabe K, Sato A, Imai C, Taga T, Adachi S, Horibe K, Manabe A, Koh K. JCCG ALL-B12: Evaluation of Intensified Therapies With Vincristine/Dexamethasone Pulses and Asparaginase and Augmented High-Dose Methotrexate for Pediatric B-ALL. J Clin Oncol 2025; 43:567-577. [PMID: 39531610 PMCID: PMC11809717 DOI: 10.1200/jco.24.00811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/06/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
PURPOSE The JCCG ALL-B12 clinical trial aimed to evaluate the effectiveness of unvalidated treatment phases for pediatric ALL and develop a safety-focused treatment framework. PATIENTS AND METHODS Patients age 1-19 years with newly diagnosed B-ALL were enrolled in this study. These patients were stratified into standard-risk (SR), intermediate-risk (IR), and high-risk (HR) groups. Randomized comparisons assessed the effectiveness of vincristine (VCR)/dexamethasone pulses in the SR group, evaluated the effects of L-asparaginase (ASP) intensification in the IR group, and compared standard consolidation including block-type treatment with experimental consolidation with high-dose methotrexate (HD-MTX) intensified with VCR and ASP in the HR group. RESULTS Of 1,936 patients enrolled, 1,804 were eligible for the experimental treatment. The overall 5-year event-free survival and overall survival rates were 85.2% (95% CI, 83.5 to 86.8) and 94.3% (95% CI, 93.1 to 95.3), respectively. The cumulative incidence of relapse and postremission nonrelapse mortality was 13.2% (95% CI, 11.6 to 14.8) and 0.6% (95% CI, 0.3 to 1.0), respectively. Random assignment in the SR group showed no significant benefit from pulse therapy. In the IR group, ASP intensification had limited effects. In the HR group, standard block therapy and HD-MTX yielded equivalent outcomes. CONCLUSION The ALL-B12 trial achieved favorable outcomes in a nationwide cohort by stratifying treatment on the basis of risk and balancing treatment intensity. This study not only demonstrated that existing standard of care can be further refined but also indicated that improvement in outcomes with intensified chemotherapy has reached a plateau.
Collapse
Affiliation(s)
- Motohiro Kato
- Department of Pediatrics, The University of Tokyo, Tokyo, Japan
| | - Yasuhiro Okamoto
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Toshihiko Imamura
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akiko Kada
- Clinical Research Center, NHO Nagoya Medical Center, Nagoya, Japan
| | - Akiko M. Saito
- Clinical Research Center, NHO Nagoya Medical Center, Nagoya, Japan
| | | | - Takao Deguchi
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Kentaro Ohki
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Takashi Fukushima
- Department of Child Health, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Pediatric Oncology and Hematology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Kenichi Anami
- Department of Medical Oncology, Hematology, and Infectious Diseases, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Masashi Sanada
- Clinical Research Center, NHO Nagoya Medical Center, Nagoya, Japan
| | - Tomohiko Taki
- Department of Medical Technology, Kyorin University Faculty of Health Sciences, Mitaka, Japan
| | - Yoshiko Hashii
- Department of Pediatrics, Osaka University, Suita, Japan
| | - Takeshi Inukai
- Department of Pediatrics, University of Yamanashi, Chuo, Japan
| | - Nobutaka Kiyokawa
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology and Oncology, Kobe Children's Hospital, Kobe, Japan
| | - Nao Yoshida
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Aichi Medical Center Nagoya First Hospital, Nagoya, Japan
| | - Yuki Yuza
- Department of Hematology-Oncology, Tokyo Metropolitan Children's Medical Center, Fuchu, Japan
| | - Masakatsu Yanagimachi
- Division of Hematology/Oncology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Kenichiro Watanabe
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Atsushi Sato
- Department of Hematology and Oncology, Miyagi Children's Hospital, Sendai, Japan
| | - Chihaya Imai
- Department of Pediatrics, University of Toyama, Toyama, Japan
| | - Takashi Taga
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Japan
| | - Souichi Adachi
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keizo Horibe
- Clinical Research Center, NHO Nagoya Medical Center, Nagoya, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| |
Collapse
|
4
|
Krstevska Bozhinovikj E, Matevska-Geshkovska N, Staninova Stojovska M, Gjorgievska E, Jovanovska A, Ridova N, Panovska Stavridis I, Kocheva S, Dimovski A. Presence of minimal residual disease determined by next-generation sequencing is not a reliable prognostic biomarker in children with acute lymphoblastic leukemia. Leuk Lymphoma 2025:1-8. [PMID: 39844437 DOI: 10.1080/10428194.2025.2456100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/23/2024] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
The role of next-generation sequencing (NGS) for minimal residual disease (MRD) assessment in pediatric acute lymphoblastic leukemia (ALL) is still under consideration. Fifty pediatric patients were prospectively evaluated for specific clonal rearrangements of immunoglobulin and T-cell receptor genes using NGS analysis at diagnosis and on days 33 and 78 from therapy onset. The prognostic value or the NGS-MRD status was analyzed after a median follow-up of 4 years. All but one patient with negative NGS-MRD status on day 33 are in clinical remission. A total of 29 (58%) patients were NGS-MRD positive on day 33, of which 9 (18%) patients remained positive on day 78. However, only a small percentage of the patients with positive NGS-MRD status on day 33 and day 78 relapsed: 21% (6/29) and 33% (3/9), respectively. Positive NGS-MRD status is not a reliable prognostic biomarker in children with ALL and warrants careful consideration in disease stratification.
Collapse
Affiliation(s)
- Elizabeta Krstevska Bozhinovikj
- Faculty of Pharmacy, Center for Biomolecular Pharmaceutical Analyses, University Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Nadica Matevska-Geshkovska
- Faculty of Pharmacy, Center for Biomolecular Pharmaceutical Analyses, University Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Marija Staninova Stojovska
- Faculty of Pharmacy, Center for Biomolecular Pharmaceutical Analyses, University Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Emilija Gjorgievska
- Faculty of Pharmacy, Center for Biomolecular Pharmaceutical Analyses, University Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Aleksandra Jovanovska
- Faculty of Medicine, University Clinic for Children's Diseases, University Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Nevenka Ridova
- Faculty of Medicine, University Clinic for Hematology, University Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Irina Panovska Stavridis
- Faculty of Medicine, University Clinic for Hematology, University Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Svetlana Kocheva
- Faculty of Medicine, University Clinic for Children's Diseases, University Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Aleksandar Dimovski
- Faculty of Pharmacy, Center for Biomolecular Pharmaceutical Analyses, University Ss. Cyril and Methodius in Skopje, Skopje, North Macedonia
- Research Center for Genetic Engineering and Biotechnology "Georgi D. Efremov", Macedonian Academy of Sciences and Arts, Skopje, North Macedonia
| |
Collapse
|
5
|
Niu FF, Gao C. [Current situation and prospect of minimal residual disease in pediatric T cell acute lymphoblastic leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2025; 46:97-102. [PMID: 40059690 PMCID: PMC11886430 DOI: 10.3760/cma.j.cn121090-20240701-00239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Indexed: 03/14/2025]
Abstract
Pediatric T-cell acute lymphoblastic leukemia (T-ALL) has been attracted much attention due to its high aggressiveness and complexity of treatment. Recently, with the development of technology and clinical research, the curative effect of T-ALL in children has been significantly improved. However, the presence of minimal residual disease (MRD) is still a key factor affecting the outcomes of children with T-ALL. With the continuous development of detection methods, the clinical applications of real-time quantitative PCR, multi-parameter flow cytometry, and high-throughput sequencing technology, MRD can be detected accurately to achieve personalized and precise treatment for each patient. The purpose of this article is to review the current detection methods of MRD in T-ALL, the clinical significance of MRD monitoring and evaluation in multi-agent combined chemotherapy and hematopoietic stem cell transplantation, and applying MRD to measure the responsiveness and effectiveness of new therapies in recent T-ALLs. Research directions and potential treatment strategies in the near future were also proposed.
Collapse
Affiliation(s)
- F F Niu
- Department of Clinical Laboratory Center, Beijing Key Laboratory of Pediatric Hematology Oncology, Key Laboratory of Major Diseases in Children Ministry of Education, Beijing Children's Hospital Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - C Gao
- Department of Clinical Laboratory Center, Beijing Key Laboratory of Pediatric Hematology Oncology, Key Laboratory of Major Diseases in Children Ministry of Education, Beijing Children's Hospital Capital Medical University, National Center for Children's Health, Beijing 100045, China
| |
Collapse
|
6
|
Engelmann R, Böttcher S. Flow Cytometric MRD Detection in Selected Mature B-Cell Malignancies. Methods Mol Biol 2025; 2865:145-188. [PMID: 39424724 DOI: 10.1007/978-1-0716-4188-0_7] [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] [Indexed: 10/21/2024]
Abstract
The quantification of submicroscopic minimal residual disease (MRD) after therapy proved to have independent prognostic significance in many mature B-cell malignancies. With the advent of routine benchtop cytometers capable of simultaneously analyzing ≥8 colors and with improved standardization, flow cytometry has become the method of choice for MRD assessments in some lymphoma entities. Herein we describe general aspects of flow cytometric standardization. Chronic lymphocytic leukemia (CLL) and multiple myeloma (MM) are used as examples to explain the technical standardization of flow cytometry for MRD detection according to EuroFlow strategies. MRD data acquisition and detailed analysis in MM and CLL is a particular focus of this chapter.
Collapse
Affiliation(s)
- Robby Engelmann
- Rostock University Medical Center, Division of Internal Medicine, Medical Clinic III - Hematology, Oncology and Palliative Medicine, Special Hematology Laboratory, Rostock, Germany
| | - Sebastian Böttcher
- Rostock University Medical Center, Division of Internal Medicine, Medical Clinic III - Hematology, Oncology and Palliative Medicine, Special Hematology Laboratory, Rostock, Germany.
| |
Collapse
|
7
|
Pott C, Brüggemann M, Ritgen M, van der Velden VHJ, van Dongen JJM. MRD Detection in B-Cell Non-Hodgkin Lymphomas Using Ig Gene Rearrangements and Chromosomal Translocations as Targets for Real-Time Quantitative PCR and ddPCR. Methods Mol Biol 2025; 2865:189-219. [PMID: 39424725 DOI: 10.1007/978-1-0716-4188-0_8] [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] [Indexed: 10/21/2024]
Abstract
Minimal residual disease (MRD) diagnostics is of high clinical relevance in patients with indolent B-cell non-Hodgkin lymphomas (B-NHL), B-cell chronic lymphocytic leukemia (CLL), and multiple myeloma and serves as a surrogate parameter to evaluate treatment effectiveness and long-term prognosis. Real-time quantitative PCR (RQ-PCR) targeting circulating lymphoma cells is still the gold standard for MRD detection in indolent B-NHL and currently the most sensitive and the most broadly applied method in follicular lymphoma (FL) and mantle cell lymphoma (MCL). Alternatively, droplet digital PCR (ddPCR) can be used for MRD monitoring in multiple myeloma, mantle cell lymphoma, CLL, and FL with comparable sensitivity, accuracy, and reproducibility.The most broadly applicable MRD target in B-NHL is the junctional regions of the rearranged immunoglobulin heavy (IGH) and light chain genes. Complete and incomplete IGH and additionally IG kappa light chain rearrangements can be used as targets for MRD. Next-generation sequencing (NGS) of IG-rearrangements (IG-NGS) as new sequencing-based technology can overcome the limitation of PCR-based approaches and has a potential for higher sensitivity. Chromosomal translocations like the t(14;18)(q32;q21) translocation associated with IGH::BCL2 fusion in FL and t(11;14)(q13;q32) translocation in MCL leading to the IGH::CCND1 fusion can be used as MRD target in selected lymphoma subtypes. In patients with CLL, both flow-cytometry and RQ-PCR are equally suited for MRD assessment as long as a sensitivity of 10-4 is achieved.MRD diagnostics targeting the IG loci is complex and requires extensive knowledge and experience because the junctional regions of each clonal rearranged gene have to be identified before the patient-specific PCR assays can be designed for MRD monitoring. In addition, the presence and load of somatic hypermutation within the rearranged IGH gene occurring during B-cell development of germinal center and post-germinal center B-cell lymphomas may hamper appropriate primer binding leading to false-negative results. The translocations mentioned above have the advantage that consensus forward primers and probes, both placed in the breakpoint regions of chromosome 18 in FL and chromosome 11 in MCL, can be used in combination with a reverse primer placed in the IGH joining region of chromosome 14. PCR-based methods using allele-specific primers can reach a high sensitivity of up to 10-5. This chapter provides all relevant background information and technical aspects for the complete laboratory process from detection of the clonal IG gene rearrangements and the chromosomal translocations at diagnosis to the actual MRD measurements in clinical follow-up samples of B-NHL. However, it should be noted that MRD diagnostics for clinical treatment protocols has to be accompanied by regular international quality control rounds to ensure the reproducibility and reliability of the MRD results. This is available by the EuroMRD network ( https://euromrd.org ), a subgroup of ESHLO ( https://eslho.org ).
Collapse
Affiliation(s)
- Christiane Pott
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany.
| | - Monika Brüggemann
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Matthias Ritgen
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Jacques J M van Dongen
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC, USAL-CSIC-FICUS) and Department of Medicine, University of Salamanca, Salamanca, Spain
- European Scientific foundation for Laboratory Hemato Oncology (ESLHO), Zutphen, The Netherlands
| |
Collapse
|
8
|
Yamanaka J, Ogawa C, Arakawa A, Deguchi T, Hori T, Kiyokawa N, Ueki H, Nishi M, Mochizuki S, Nishikawa T, Kumamoto T, Nishiuchi R, Kikuta A, Yamamoto S, Koh K, Hasegawa D, Ogawa A, Watanabe K, Sato A, Saito AM, Watanabe T, Manabe A, Horibe K, Goto H, Toyoda H. Outcomes in children with first-relapsed acute lymphoblastic leukemia in Japan: Results from JCCG Study JPLSG-ALL-R08. Pediatr Blood Cancer 2024; 71:e31319. [PMID: 39267231 DOI: 10.1002/pbc.31319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/04/2024] [Accepted: 08/28/2024] [Indexed: 09/17/2024]
Abstract
BACKGROUND/OBJECTIVES The Berlin-Frankfurt-Münster (BFM)-S classification is a crucial prognostic indicator in children experiencing first-relapsed acute lymphoblastic leukemia (ALL). Early molecular response to therapy, evaluated by measurable/minimal residual disease (MRD), has a significant impact on the survival of patients with childhood ALL. Applying risk stratification based on the BFM-S classification and MRD response after induction, the first nationwide prospective multicenter study, ALL-R08, was conducted in children with first-relapsed ALL in Japan. METHODS The ALL-R08 study comprised two parts: ALL-R08-I, an observational study aimed at obtaining an overall picture of outcomes in first-relapsed childhood ALL, and ALL-R08-II, a clinical trial for the non-T-ALL S2 risk group. In ALL-R08-II, patients with an MRD level of ≥10-3 at the end of induction therapy were assigned to undergo allogeneic hematopoietic stem cell transplantation (allo-HCT), whereas those with an MRD level less than 10-3 and isolated extramedullary relapse continued to receive chemotherapy. RESULTS In total, 163 patients were enrolled in the ALL-R08 study, and 82 and 81 patients were enrolled in the ALL-R08-I and the ALL-R08-II, respectively. In ALL-R08-I, the probability of 3-year event-free survival (EFS) for patients with S1, S2, S3, S4, and post-HCT groups was 83% ± 15%, 37% ± 11%, 28% ± 8%, 14% ± 7%, and 0%, respectively. In the ALL-R08-II trial, 3-year EFS in patients with post-induction MRD less than 10-3 and ≥10-3 was 70% ± 9% (n = 27) and 68% ± 8% (n = 31) (p = .591), respectively. CONCLUSIONS ALL-REZ BFM-type treatment is equally effective for children with first-relapsed ALL treated according to the Japanese frontline protocols and for children with first-relapsed ALL treated according to the BFM-type frontline protocols.
Collapse
Affiliation(s)
- Junko Yamanaka
- Department of Pediatrics, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Chitose Ogawa
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ayumu Arakawa
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Takao Deguchi
- Division of Cancer Immunodiagnostics, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Toshinori Hori
- Department of Pediatrics, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Nobutaka Kiyokawa
- Department of Pediatric Hematology and Oncology Research, Research Institute, National Center for Child Health and Development, Tokyo, Japan
| | - Hideaki Ueki
- Department of Pediatric Hematology/Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Masanori Nishi
- Department of Pediatrics, Faculty of Medicine, Saga University, Saga, Japan
| | - Shinji Mochizuki
- Department of Pediatrics, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Takuro Nishikawa
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Tadashi Kumamoto
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ritsuo Nishiuchi
- Department of Pediatrics, Kochi Health Sciences Center, Kochi, Japan
| | - Atsushi Kikuta
- Department of Pediatric Oncology, Fukushima Medical University Hospital, Fukushima, Japan
| | - Shohei Yamamoto
- Department of Pediatrics, Tokai University School of Medicine, Isehara, Japan
| | - Katsuyoshi Koh
- Department of Hematology/Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Daisuke Hasegawa
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan
| | - Atsushi Ogawa
- Department of Pediatrics, Niigata Cancer Center Hospital, Niigata, Japan
| | - Kenichiro Watanabe
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Atsushi Sato
- Department of Hematology/Oncology, Miyagi Children's Hospital, Sendai, Japan
| | - Akiko M Saito
- Clinical Research Center, NHO Nagoya Medical Center, Nagoya, Japan
| | - Tomoyuki Watanabe
- Department of Health and Nutritional Sciences, Faculty of Health Sciences, Aichi Gakuin University, Nisshin, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Keizo Horibe
- Clinical Research Center, NHO Nagoya Medical Center, Nagoya, Japan
| | - Hiroaki Goto
- Division of Hematology/Oncology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hidemi Toyoda
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
9
|
Doculara L, Evans K, Gooding JJ, Bayat N, Lock RB. Patient-Specific Circulating Tumor DNA for Monitoring Response to Menin Inhibitor Treatment in Preclinical Models of Infant Leukemia. Cancers (Basel) 2024; 16:3990. [PMID: 39682177 DOI: 10.3390/cancers16233990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND In infant KMT2A (MLL1)-rearranged (MLL-r) acute lymphoblastic leukemia (ALL), early relapse and treatment response are currently monitored through invasive repeated bone marrow (BM) biopsies. Circulating tumor DNA (ctDNA) in peripheral blood (PB) provides a minimally invasive alternative, allowing for more frequent disease monitoring. However, a poor understanding of ctDNA dynamics has hampered its clinical translation. We explored the predictive value of ctDNA for detecting minimal/measurable residual disease (MRD) and drug response in a patient-derived xenograft (PDX) model of infant MLL-r ALL. METHODS Immune-deficient mice engrafted with three MLL-r ALL PDXs were monitored for ctDNA levels before and after treatment with the menin inhibitor SNDX-50469. RESULTS The amount of ctDNA detected strongly correlated with leukemia burden during initial engraftment prior to drug treatment. However, following SNDX-50469 treatment, the leukemic burden assessed by either PB leukemia cells through flow cytometry or ctDNA levels through droplet digital polymerase chain reaction (ddPCR) was discrepant. This divergence could be attributed to the persistence of leukemia cells in the spleen and BM, highlighting the ability of ctDNA to reflect disease dynamics in key leukemia infiltration sites. CONCLUSIONS Notably, ctDNA analysis proved to be a superior predictor of MRD compared to PB assessment alone, especially in instances of low disease burden. These findings highlight the potential of ctDNA as a sensitive biomarker for monitoring treatment response and detecting MRD in infant MLL-r ALL.
Collapse
Affiliation(s)
- Louise Doculara
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Kathryn Evans
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| | - J Justin Gooding
- School of Chemistry, UNSW Sydney, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Narges Bayat
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
10
|
Bader MS, Kubetzko S, Schäfer BW, Arranto C, Drexler B, Halter J, Passweg JR, Medinger M. Minimal residual disease monitoring by Ig/TCR gene rearrangements predicts post-transplant relapse and survival in adult patients with acute lymphoblastic leukemia. Ann Hematol 2024; 103:4831-4833. [PMID: 39167183 PMCID: PMC11534960 DOI: 10.1007/s00277-024-05943-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Affiliation(s)
- Michael Stephan Bader
- Divisions of Hematology and Internal Medicine, Department of Medicine, University Hospital Basel, Petersgraben 4, Basel, CH-4031, Switzerland
- University of Basel, Basel, Switzerland
| | - Susanne Kubetzko
- Division of Oncology, University Children's Hospital, Zürich, Switzerland
| | | | - Christian Arranto
- Divisions of Hematology and Internal Medicine, Department of Medicine, University Hospital Basel, Petersgraben 4, Basel, CH-4031, Switzerland
- University of Basel, Basel, Switzerland
| | - Beatrice Drexler
- Divisions of Hematology and Internal Medicine, Department of Medicine, University Hospital Basel, Petersgraben 4, Basel, CH-4031, Switzerland
- University of Basel, Basel, Switzerland
| | - Jörg Halter
- Divisions of Hematology and Internal Medicine, Department of Medicine, University Hospital Basel, Petersgraben 4, Basel, CH-4031, Switzerland
- University of Basel, Basel, Switzerland
| | - Jakob R Passweg
- Divisions of Hematology and Internal Medicine, Department of Medicine, University Hospital Basel, Petersgraben 4, Basel, CH-4031, Switzerland
- University of Basel, Basel, Switzerland
| | - Michael Medinger
- Divisions of Hematology and Internal Medicine, Department of Medicine, University Hospital Basel, Petersgraben 4, Basel, CH-4031, Switzerland.
- University of Basel, Basel, Switzerland.
| |
Collapse
|
11
|
Bardini M, Fazio G, Abascal LC, Meyer C, Maglia O, Sala S, Palamini S, Rebellato S, Marschalek R, Rizzari C, Biondi A, Cazzaniga G. Prenatal origin of NUTM1 gene rearrangement in infant B-cell precursor acute lymphoblastic leukaemia. Br J Haematol 2024; 205:1883-1888. [PMID: 39099338 DOI: 10.1111/bjh.19685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/23/2024] [Indexed: 08/06/2024]
Abstract
Rearrangement of NUTM1 gene (NUTM1r) is one of the most frequent aberrations occurring in infants (younger than 1 year at diagnosis) with B-cell precursor Acute Lymphoblastic Leukaemia (BCP-ALL). In this study we had the unique opportunity to analyze the umbilical cord blood (UCB) sample from one infant patient with NUTM1r BCP-ALL. Herein we reported for the first time that NUTM1r infant ALL arise prenatally, as both the patient-specific CUX1::NUTM1 fusion gene, as well as two IG/TR leukaemic markers were already present and detectable in the patient's UCB at birth. Our results clearly demonstrate the prenatal origin of NUTM1r infant BCP-ALL.
Collapse
Affiliation(s)
- Michela Bardini
- Tettamanti Center, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Grazia Fazio
- Tettamanti Center, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy
| | | | - Claus Meyer
- DCAL/Institute of Pharmaceutical Biology, Goethe-University, Frankfurt/Main, Germany
| | - Oscar Maglia
- Tettamanti Center, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Simona Sala
- Tettamanti Center, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Sonia Palamini
- Tettamanti Center, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Stefano Rebellato
- Tettamanti Center, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Rolf Marschalek
- School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy
| | - Carmelo Rizzari
- Pediatrics, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Andrea Biondi
- School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy
- Pediatrics, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Giovanni Cazzaniga
- Tettamanti Center, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy
| |
Collapse
|
12
|
Domenech C, Kicinski M, De Moerloose B, Piette C, Chahla WA, Kornreich L, Pasquet M, Uyttebroeck A, Theron A, Poirée M, Arfeuille C, Bakkus M, Grardel N, Paillard C, Freycon C, Millot F, Simon P, Philippet P, Pluchart C, Suciu S, Rohrlich P, Ferster A, Bertrand Y, Cavé H, for the Children's Leukemia Group (CLG) of the European Organization for Research and Treatment of Cancer (EORTC). Results of the prospective EORTC Children Leukemia Group study 58081 in precursor B- and T-cell acute lymphoblastic leukemia. Hemasphere 2024; 8:e70025. [PMID: 39540141 PMCID: PMC11558101 DOI: 10.1002/hem3.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/28/2024] [Accepted: 08/07/2024] [Indexed: 11/16/2024] Open
Abstract
Here, we report the results of the prospective cohort study EORTC-CLG 58081 and compare them to the control arm of the randomized phase 3 trial EORTC-CLG 58951, on which treatment recommendations were built. In both studies, patients aged 1-18 years with BCR::ABL1 negative acute lymphoblastic leukemia of the B-lineage (B-ALL) or T-lineage (T-ALL) were treated using a BFM backbone without cranial irradiation. Similarly to the control arm of 58951, prednisolone (PRED) 60 mg/m2/day was used for induction therapy, but a few modifications were made. Dexamethasone (DXM) was used in average-risk 2 (AR2) T-ALL and B-ALL during induction, 10 and 6 mg/m2/day, respectively. Leucovorin rescue was delayed to 42 h instead of 36 h after initiation of high-dose methotrexate, and a postconsolidation MRD time point was added to stratify patients. Between 2011 and 2017, 835 patients were prospectively enrolled in the 58081 study. Overall, the 5-year event-free survival (EFS) was 84.8% versus 83.6% (hazard ratio [HR], 0.96 [95% confidence interval [CI]: 0.76-1.21]) for 58081 versus 58951 considered as a control group, respectively, 84.3% versus 84.9% (HR, 1.06 [99% CI: 0.75-1.49]) in B-ALL but 87.3% versus 76.6% (HR, 0.59 [99% CI: 0.28-1.24]) in T-ALL. The comparison between the two studies regarding EFS differed by risk group (p = 0.012). The HR was 2.15 (99% CI: 0.67-6.85) for very low-risk but 0.34 (99% CI: 0.13-0.89) for AR2. The particularly favorable results observed in the T-ALLs and AR2 subgroups suggest the benefit of using DXM in specific patient groups and highlight the importance of risk stratification.
Collapse
Affiliation(s)
- Carine Domenech
- Department of Pediatric Hematology‐Oncology, Institut d'Hématologie et d'Oncologie Pédiatrique, Hospices Civils de LyonUniversité Lyon1LyonFrance
| | | | - Barbara De Moerloose
- Department of Pediatric Hematology‐OncologyGhent University HospitalGhentBelgium
| | - Caroline Piette
- Department of Paediatrics, Division of Haematology‐OncologyUniversity Hospital Liège and University of LiègeLiègeBelgium
| | | | - Laure Kornreich
- Department of Hemato‐OncologyHUDERF‐HUB (ULB)BrusselsBelgium
| | | | - Anne Uyttebroeck
- Department of PediatricsUniversity Hospital GasthuisbergLeuvenBelgium
| | - Alexandre Theron
- Department of Pediatric Hematology OncologyCHU de MontpellierMontpellierFrance
| | | | - Chloé Arfeuille
- Département de GénétiqueAssistance Publique des Hôpitaux de Paris (AP‐HP), Hôpital Robert DebréParisFrance
- Department of Research‐INSERM UMR 1131Université Paris CitéFrance
| | - Marleen Bakkus
- Department of Molecular HematologyUZ BrusselBrusselsBelgium
| | | | - Catherine Paillard
- Department of Paediatric Haematology and OncologyCHU HautepierreStrasbourg
| | | | - Frédéric Millot
- Department of Pediatric Hematology‐OncologyCHUPoitiersFrance
| | - Pauline Simon
- Department of Pediatric Hematology‐OncologyCHRUBesançonFrance
| | - Pierre Philippet
- Department of Pediatric Hemato‐OncologyCHC MontLégiaLiègeBelgium
| | | | - Stefan Suciu
- Department of StatisticEORTC HeadquartersBrusselsBelgium
| | | | - Alina Ferster
- Department of Hemato‐OncologyHUDERF‐HUB (ULB)BrusselsBelgium
| | - Yves Bertrand
- Department of Pediatric Hematology‐Oncology, Institut d'Hématologie et d'Oncologie Pédiatrique, Hospices Civils de LyonUniversité Lyon1LyonFrance
| | - Hélène Cavé
- Département de GénétiqueAssistance Publique des Hôpitaux de Paris (AP‐HP), Hôpital Robert DebréParisFrance
- Department of Research‐INSERM UMR 1131Université Paris CitéFrance
| | | |
Collapse
|
13
|
Borlenghi E, Zollner T, Rossi G, Pagani C, Cattaneo C, Malagola M, Sottini A, Bertoli D, Tonelli M, Chiarini M, Leopaldo R, Passi A, Masina L, Federico F, Giupponi C, Tucci A. Older Adults with Ph Negative Acute Lymphoblastic Leukemia: A Monocentric Experience on 57 Patients Focusing on Treatment Intensity and Age-Related Prognosis. Mediterr J Hematol Infect Dis 2024; 16:e2024080. [PMID: 39534710 PMCID: PMC11556428 DOI: 10.4084/mjhid.2024.080] [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: 03/19/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Treating with curative intent elderly patients affected by acute lymphoblastic leukemia (ALL) is an unmet clinical need. Comorbidities, lower tolerance to organ toxicities, and poor disease-related prognostic features make it difficult to cure these patients.
Objectives: To analyze the outcome of ALL elderly patients aged over 55 years treated intensively or with the best supportive care (BSC) over the last 20 years.
Collapse
Affiliation(s)
| | | | | | - Chiara Pagani
- Unit of Hematology, ASST Spedali Civili, Brescia, Italy
| | | | - Michele Malagola
- Unit of Blood Diseases and Stem Cell Transplantation, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Alessandra Sottini
- Diagnostic Department, Clinical Chemistry Laboratory, ASST Spedali Civili, Brescia, Italy
| | - Diego Bertoli
- Diagnostic Department, Clinical Chemistry Laboratory, ASST Spedali Civili, Brescia, Italy
| | - Mariella Tonelli
- Diagnostic Department, Cytogenetic and Molecular Genetics Section, ASST Spedali Civili, Brescia, Italy
| | - Marco Chiarini
- Diagnostic Department, Clinical Chemistry Laboratory, ASST Spedali Civili, Brescia, Italy
| | | | - Angela Passi
- Unit of Hematology, ASST Spedali Civili, Brescia, Italy
| | | | | | - Carlotta Giupponi
- Diagnostic Department, Clinical Chemistry Laboratory, ASST Spedali Civili, Brescia, Italy
| | | |
Collapse
|
14
|
Boulland ML, Aliouat A, Jalaber E, Desmares A, Toujani S, Luque Paz D, Wiber M, Voirin E, Lachot S, Basinko A, Lambert WC, Carras S, Cousin E, Marchand T, de Tayrac M, Fest T, Houot R, Pastoret C. Tailored Digital PCR Follow-Up of Rare Fusion Transcripts after Initial Detection through RNA Sequencing in Hematological Malignancies. J Mol Diagn 2024; 26:1007-1017. [PMID: 39182671 DOI: 10.1016/j.jmoldx.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/05/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
Minimal residual disease (MRD) monitoring plays a pivotal role in the management of hematologic malignancies. Well-established molecular targets, such as PML::RARA, CBFB::MYH11, or RUNX1::RUNX1T1, are conventionally tracked by quantitative RT-PCR. Recently, a broader landscape of fusion transcripts has been unveiled through transcriptomic analysis. These newly discovered fusion transcripts may emerge as novel molecular markers for MRD quantification. In this study, we compared a targeted RNA-sequencing (RNA-seq) approach (FusionPlex) with a whole-transcriptomic strategy (Advanta RNA-Seq XT) for fusion detection in a training set of 21 samples. We evidenced a concordance of 100% for the detection of known fusions, and showed a good correlation for gene expression quantification between the two techniques (Spearman r = 0.77). Additionally, we prospectively evaluated the identification of fusions by targeted RNA-seq in a real-life series of 126 patients with hematological malignancy. At least one fusion transcript was detected for 60 patients (48%). We designed tailored digital PCR assays for 11 rare fusions, and validated this technique for MRD quantification with a limit of detection of <0.01%. The combination of RNA-seq and tailored digital PCR may become a new standard for MRD evaluation in patients lacking conventional molecular targets.
Collapse
Affiliation(s)
- Marie-Laure Boulland
- Hematology Laboratory, Rennes University Hospital, Rennes, France; Inserm U1236, Rennes University, Rennes, France
| | - Amyra Aliouat
- Genetics Laboratory, Rennes University Hospital, Rennes, France
| | - Elie Jalaber
- Clinical Hematology Department, Rennes University Hospital, Rennes, France
| | - Anne Desmares
- Hematology Laboratory, Rennes University Hospital, Rennes, France
| | - Saloua Toujani
- Cytogenetics and Cellular Biology Laboratory, Rennes University Hospital, Rennes, France
| | - Damien Luque Paz
- Angers, Nantes University, Angers University Hospital, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers (CRCI(2)NA), Angers, France
| | - Margaux Wiber
- Angers, Nantes University, Angers University Hospital, Inserm, CNRS, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers (CRCI(2)NA), Angers, France
| | - Emeline Voirin
- Hematology Laboratory, Tours University Hospital, Tours, France
| | | | - Audrey Basinko
- Cytogenetics Laboratory, Brest University Hospital, Brest, France
| | | | - Sylvain Carras
- Hematology Laboratory, Grenoble-Alpes University Hospital, La Tronche, France
| | - Elie Cousin
- Pediatric Onco-Hematology Department, Rennes University Hospital, Rennes, France
| | - Tony Marchand
- Inserm U1236, Rennes University, Rennes, France; Clinical Hematology Department, Rennes University Hospital, Rennes, France
| | - Marie de Tayrac
- Genetics Laboratory, Rennes University Hospital, Rennes, France
| | - Thierry Fest
- Hematology Laboratory, Rennes University Hospital, Rennes, France; Inserm U1236, Rennes University, Rennes, France
| | - Roch Houot
- Inserm U1236, Rennes University, Rennes, France; Clinical Hematology Department, Rennes University Hospital, Rennes, France
| | - Cédric Pastoret
- Hematology Laboratory, Rennes University Hospital, Rennes, France; Inserm U1236, Rennes University, Rennes, France.
| |
Collapse
|
15
|
Stutterheim J, van der Waarden R, de Groot-Kruseman HA, Sonneveld E, de Haas V, Dandis R, van der Schoot CE, van der Velden VHJ, Pieters R. Are measurable residual disease results after consolidation therapy useful in children with acute lymphoblastic leukemia? Leukemia 2024; 38:2376-2381. [PMID: 39256602 PMCID: PMC11518975 DOI: 10.1038/s41375-024-02386-5] [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: 10/04/2023] [Revised: 08/05/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024]
Abstract
Measurable residual disease (MRD) is regularly tested at later timepoints after the end of first consolidation (EOC) in children with acute lymphoblastic leukemia (ALL). The question remains whether this is useful for detecting (molecular) relapse. We investigated the clinical relevance of MRD after EOC in intermediate risk patients treated on DCOG-ALL-10 (n = 271) and DCOG-ALL-9 (n = 122), with MRD <0.05% at EOC. EOC MRD-negative patients (n = 178) had excellent outcomes, irrespective of MRD results at later timepoints; 6-year cumulative incidence of relapse (6-y CIR) of 7.4% (95% CI, 3.9%-12.3%) for those with MRD negativity at all later timepoints compared to 3.8% (95% CI, 0.3%-16.8%) for those with one or more later timepoints being positive (p = 0.51). Patients with positive EOC MRD (n = 91) of whom the subsequent timepoints were MRD negative (n = 43), had comparable good outcomes, 6-y CIR of 7.0% (95% CI, 1.8%-17.2%). In contrast, patients being MRD positive at EOC and MRD positive at one or more subsequent timepoints (n = 48) had a higher risk of relapse, 6-y CIR 29.4% (95% CI, 17.2%-42.8%), p < 0.001. These findings were confirmed in the validation cohort of ALL-9 as well as using the updated EuroMRD guidelines. In EOC MRD-negative patients, subsequent MRD measurements can be abandoned. For EOC MRD-positive patients the subsequent MRD measurement might be informative for further risk stratification.
Collapse
Affiliation(s)
- Janine Stutterheim
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| | | | - Hester A de Groot-Kruseman
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Dutch Childhood Oncology Group (DCOG), Utrecht, The Netherlands
| | - Edwin Sonneveld
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Dutch Childhood Oncology Group (DCOG), Utrecht, The Netherlands
| | - Valérie de Haas
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Dutch Childhood Oncology Group (DCOG), Utrecht, The Netherlands
| | - Rana Dandis
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory Amsterdam, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Dutch Childhood Oncology Group (DCOG), Utrecht, The Netherlands
| |
Collapse
|
16
|
Shen Q, Gong X, Feng Y, Hu Y, Wang T, Yan W, Zhang W, Qi S, Gale RP, Chen J. Measurable residual disease (MRD)-testing in haematological cancers: A giant leap forward or sideways? Blood Rev 2024; 68:101226. [PMID: 39164126 DOI: 10.1016/j.blre.2024.101226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024]
Abstract
Measurable residual disease (MRD)-testing is used in many haematological cancers to estimate relapse risk and to direct therapy. Sometimes MRD-test results are used for regulatory approval. However, some people including regulators wrongfully believe results of MRD-testing are highly accurate and of proven efficacy in directing therapy. We review MRD-testing technologies and evaluate the accuracy of MRD-testing for predicting relapse and the strength of evidence supporting efficacy of MRD-guided therapy. We show that at the individual level MRD-test results are often an inaccurate relapse predictor. Also, no convincing data indicate that increasing therapy-intensity based on a positive MRD-test reduces relapse risk or improves survival. We caution against adjusting therapy-intensity based solely on results of MRD-testing.
Collapse
Affiliation(s)
- Qiujin Shen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Xiaowen Gong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Yahui Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Yu Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Tiantian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Wen Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Wei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Saibing Qi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College of Science, Technology and Medicine, London, UK.
| | - Junren Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| |
Collapse
|
17
|
Revoltar M, van der Linde R, Cromer D, Gatt PN, Smith S, Fernandez MA, Vaughan L, Blyth E, Curnow J, Tegg E, Brown DA, Sasson SC. Indeterminate measurable residual disease by multiparameter flow cytometry is associated with an intermediate risk of clinical relapse in adult patients with acute leukaemia. Pathology 2024; 56:882-888. [PMID: 39025727 DOI: 10.1016/j.pathol.2024.04.009] [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: 08/26/2023] [Revised: 03/18/2024] [Accepted: 04/22/2024] [Indexed: 07/20/2024]
Abstract
Measurable residual disease (MRD) is useful for prognostication and for monitoring response to treatment in patients with acute leukaemia. MRD by multiparametric flow cytometry (MFC-MRD) utilises the leukaemia-associated immunophenotype (LAIP) and difference from normal (DfN) strategies to identify the leukaemic clone. Difficulties arise when the LAIP overlaps with normal regeneration, there is clonal evolution, or when the abnormal clone population is exceptionally small e.g., <0.01% of CD45+ cells. Such cases are reported as 'indeterminate'; however, there is little international consensus on this reporting. The relationship between clinical outcomes and indeterminate MFC-MRD is unknown. Here we determine the rate of indeterminate MFC-MRD reporting, its relationship to concurrent molecular MRD results when available, and to clinical outcomes to 12 months. We performed an internal audit of all adult testing for MFC-MRD between January and December 2021. A total of 153 consecutive patients with a diagnosis of acute leukaemia were included. Successive MFC-MRD results and clinical outcomes were recorded over a 12-month period from time of inclusion into the study. In total, 460 MFC-MRD tests from 153 patients were reviewed and 73 (16%) MFC-MRD tests from 54 (35%) patients were reported as indeterminate. The majority (70%) were at low levels between 0.01-0.1% of CD45+ cells. Compared to patients with a negative result, acute myeloid leukaemia (AML) was more frequent in patients who had an indeterminate MFC-MRD (70% vs 36%), and B-cell acute lymphoblastic leukaemia was less common (20% vs 55%). In patients with indeterminate MFC-MRD results, one-third had received either chemotherapy or allogeneic haemopoietic stem cell transplant (aHSCT) within the preceding 3 months. Agreement between MFC and molecular MRD testing was low. Patients with indeterminate MFC-MRD had leukaemia relapse rates below patients with a positive MFC-MRD, but greater than those with negative MFC-MRD (positive 33% vs indeterminate 21% vs negative 8%, p = 0.038). Overall, these findings indicate that indeterminate MFC-MRD results are more common in adults with AML and also in those who have received chemotherapy or aHSCT within the previous 3 months. We report for the first time that indeterminate MFC-MRD is a finding of potential clinical significance, which associates with a numerically higher median relapse rate within 12 months when compared to a negative MFC-MRD result.
Collapse
Affiliation(s)
- Maxine Revoltar
- Department of Laboratory Haematology, ICPMR, Westmead Hospital, NSW Health Pathology, Westmead, NSW, Australia; Department of Clinical Haematology, Westmead Hospital, Westmead, NSW, Australia.
| | - Riana van der Linde
- Department of Laboratory Haematology, ICPMR, Westmead Hospital, NSW Health Pathology, Westmead, NSW, Australia; Flow Cytometry Unit, ICPMR, NSW Health Pathology, Westmead Hospital, Westmead, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Deborah Cromer
- The Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Prudence N Gatt
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia; Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Sandy Smith
- Flow Cytometry Unit, ICPMR, NSW Health Pathology, Westmead Hospital, Westmead, NSW, Australia
| | - Marian A Fernandez
- Flow Cytometry Unit, ICPMR, NSW Health Pathology, Westmead Hospital, Westmead, NSW, Australia
| | - Lachlin Vaughan
- Department of Laboratory Haematology, ICPMR, Westmead Hospital, NSW Health Pathology, Westmead, NSW, Australia; Department of Clinical Haematology, Westmead Hospital, Westmead, NSW, Australia; Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Emily Blyth
- Department of Clinical Haematology, Westmead Hospital, Westmead, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia; Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Jennifer Curnow
- Department of Clinical Haematology, Westmead Hospital, Westmead, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Elizabeth Tegg
- Department of Laboratory Haematology, ICPMR, Westmead Hospital, NSW Health Pathology, Westmead, NSW, Australia; Flow Cytometry Unit, ICPMR, NSW Health Pathology, Westmead Hospital, Westmead, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - David A Brown
- Flow Cytometry Unit, ICPMR, NSW Health Pathology, Westmead Hospital, Westmead, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia; Westmead Institute for Medical Research, Westmead, NSW, Australia; Department of Clinical Immunology, Westmead Hospital, Westmead, NSW, Australia
| | - Sarah C Sasson
- Flow Cytometry Unit, ICPMR, NSW Health Pathology, Westmead Hospital, Westmead, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia; The Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
18
|
Kim R, Chalandon Y, Rousselot P, Cayuela JM, Huguet F, Balsat M, Passet M, Chevallier P, Hicheri Y, Raffoux E, Leguay T, Chantepie S, Maury S, Hayette S, Solly F, Braun T, De Prijck B, Cacheux V, Salanoubat C, Farnault L, Guibaud I, Lamarque M, Gastaud L, Lemasle E, Brissot E, Tavernier E, Bilger K, Villate A, Soulier J, Graux C, Lhéritier V, Dombret H, Boissel N, Clappier E. Significance of Measurable Residual Disease in Adult Philadelphia Chromosome-Positive ALL: A GRAAPH-2014 Study. J Clin Oncol 2024; 42:3140-3150. [PMID: 39028928 DOI: 10.1200/jco.24.00108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/19/2024] [Accepted: 05/03/2024] [Indexed: 07/21/2024] Open
Abstract
PURPOSE BCR::ABL1 quantification is widely regarded as the standard for monitoring measurable residual disease (MRD) in Philadelphia chromosome-positive (Ph+) ALL. However, recent evidence of BCR::ABL1 multilineage involvement questions the significance of BCR::ABL1 MRD. We aimed to define the prognostic role of MRD as assessed by BCR::ABL1 or lymphoid-specific immunoglobulin/T-cell receptor (IG/TR) gene markers. PATIENTS AND METHODS We conducted BCR::ABL1 and IG/TR quantification after each treatment cycle in 264 patients treated in the GRAAPH-2014 trial, which used four cycles of reduced-intensity chemotherapy with nilotinib, followed by hematopoietic stem-cell transplantation (HSCT). RESULTS Comparing BCR::ABL1 and IG/TR MRD revealed residual BCR::ABL1-positive non-ALL cells in 98 (43%) of 228 patients, defining multilineage Ph+ ALL. Despite poorer BCR::ABL1 responses, patients with multilineage Ph+ ALL had similar disease-free survival (DFS; hazard ratio [HR], 0.83 [95% CI, 0.49 to 1.41]; P = .50). Although BCR::ABL1 response failed to predict outcomes, IG/TR positivity (≥0.01%) was strongly associated with lower DFS (after cycle 2, HR, 2.49 [95% CI, 1.40 to 4.40]; P = .002; after cycle 4, HR, 4.13 [95% CI, 1.82 to 9.38]; P = .001). In multivariable analysis, both IG/TR positivity after cycle 2 and initial WBC count ≥30 × 109/L predicted poorer DFS, enabling to define a high-risk group having a 4-year DFS of 56.5% compared with 87.6% (HR, 3.72 [95% CI, 1.93 to 7.15]; P < .001). Moreover, allogeneic HSCT significantly improved DFS in the high-risk group (HR, 0.33 [95% CI, 0.18 to 0.60]; P < .001), whereas the standard-risk group had favorable outcomes regardless of allogeneic HSCT. CONCLUSION Our findings challenge the significance of BCR::ABL1 monitoring in adult Ph+ ALL and demonstrate the prognostic role of IG/TR MRD. This study provides a framework for using MRD to guide treatment strategies in adults with Ph+ ALL.
Collapse
Affiliation(s)
- Rathana Kim
- Hematology Laboratory, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis (IRSL), Paris, France
| | - Yves Chalandon
- Division of Hematology, Department of Oncology, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Swiss Group for Clinical Cancer Research (SAKK)
| | - Philippe Rousselot
- Hematology Department, Centre Hospitalier de Versailles, Unité mixte de recherche 1184 Commissariat à l'Energie Atomique, University Paris-Saclay, Le Chesnay, France
| | - Jean-Michel Cayuela
- Hematology Laboratory, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- EA 3518, Institut de Recherche Saint-Louis (IRSL), Université Paris Cité, Paris, France
| | - Françoise Huguet
- Hematology Department, Institut Universitaire de Cancer Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| | - Marie Balsat
- Hematology Department, Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre Benite, France
| | - Marie Passet
- Hematology Laboratory, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis (IRSL), Paris, France
| | | | - Yosr Hicheri
- Hematology Department, Institut Paoli-Calmettes, Aix Marseille Univ, CNRS, Inserm, CRCM, Marseille, France
| | - Emmanuel Raffoux
- Hematology Department, Hôpital Saint-Louis, AP-HP, Université Paris Cité, Paris, France
| | - Thibaut Leguay
- Hematology Department, CHU de Bordeaux, Hôpital du Haut-Levêque, Pessac, France
| | | | - Sébastien Maury
- Hematology Department, Hôpital Henri Mondor, AP-HP, Université Paris Est Créteil UPEC, Créteil, France
| | - Sandrine Hayette
- Hematology Laboratory, Hospices Civils de Lyon, Lyon-Sud Hospital, Pierre Benite, France
| | | | - Thorsten Braun
- Hematology Department, Hôpital Avicenne, AP-HP, Bobigny, France
| | | | | | - Celia Salanoubat
- Hematology Department, CH Sud Francilien, Corbeil-Essonnes, France
| | - Laure Farnault
- Hematology Department, Hôpital Universitaire de Marseille Conception, Marseille, France
| | - Isabelle Guibaud
- Hematology Department, CH de Metz, Hôpital de Mercy, Metz, France
| | - Mathilde Lamarque
- Hematology Department, CH Emile Muller de Mulhouse, Mulhouse, France
| | - Lauris Gastaud
- Hematology Department, Centre Antoine Lacassagne, Nice, France
| | - Emilie Lemasle
- Hematology Department, Centre Henri Becquerel, Rouen, France
| | - Eolia Brissot
- Hematology Department, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France
| | | | - Karine Bilger
- Hematology Department, CHU de Strasbourg, Hôpital Hautepierre, Strasbourg, France
| | - Alban Villate
- Hematology Department, CHU de Tours, Hôpital Bretonneau, Tours, France
| | - Jean Soulier
- Hematology Laboratory, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis (IRSL), Paris, France
| | - Carlos Graux
- Hematology Department, CHU UCL Namur Godinne, Yvoir, Belgium
| | - Véronique Lhéritier
- Coordination du Groupe GRAALL, Member of the French institute Carnot OPALE (the Organisation for Partnership in Leukemia Consortium), Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre Benite, France
| | - Hervé Dombret
- Hematology Department, Hôpital Saint-Louis, AP-HP, Université Paris Cité, Paris, France
| | - Nicolas Boissel
- Hematology Department, Hôpital Saint-Louis, AP-HP, Université Paris Cité, Paris, France
| | - Emmanuelle Clappier
- Hematology Laboratory, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis (IRSL), Paris, France
| |
Collapse
|
19
|
Huang XT, Wang CJ, Gao C, Xue TL, Zhao ZJ, Wang TY, Wu MY, Cui L, Zhang RD, Li ZG. Relationship between subtype-specific minimal residual disease level and long-term prognosis in children with acute lymphoblastic leukemia. Ann Hematol 2024; 103:3657-3665. [PMID: 38494553 DOI: 10.1007/s00277-024-05687-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
Minimal residual disease (MRD) based risk stratification criteria for specific genetic subtypes remained unclear in childhood acute lymphoblastic leukemia (ALL). Among 723 children with newly diagnosed ALL treated with the Chinese Children Leukemia Group CCLG-2008 protocol, MRD was assessed at time point 1 (TP1, at the end of induction) and TP2 (before consolidation treatment) and the MRD levels significantly differed in patients with different fusion genes or immunophenotypes (P all < 0.001). Moreover, the prognostic impact of MRD varied by distinct molecular subtypes. We stratified patients in each molecular subtype into two MRD groups based on the results. For patients carrying BCR::ABL1 or KMT2A rearrangements, we classified patients with MRD < 10-2 at both TP1 and TP2 as the low MRD group and the others as the high MRD group. ETV6::RUNX1+ patients with TP1 MRD < 10-3 and TP2 MRD-negative were classified as the low MRD group and the others as the high MRD group. For T-ALL, We defined children with TP1 MRD ≥ 10-3 as the high MRD group and the others as the low MRD group. The 10-year relapse-free survival of low MRD group was significantly better than that of high MRD group. We verified the prognostic impact of the subtype-specific MRD-based stratification in patients treated with the BCH-ALL2003 protocol. In conclusion, the subtype-specific MRD risk stratification may contribute to the precise treatment of childhood ALL.
Collapse
Affiliation(s)
- Xiao-Tong Huang
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Chan-Juan Wang
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Chao Gao
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Tian-Lin Xue
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Zi-Jing Zhao
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Tian-You Wang
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Min-Yuan Wu
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Lei Cui
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China.
| | - Rui-Dong Zhang
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China.
| | - Zhi-Gang Li
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China.
| |
Collapse
|
20
|
Tran V, Salafian K, Michaels K, Jones C, Reed D, Keng M, El Chaer F. MRD in Philadelphia Chromosome-Positive ALL: Methodologies and Clinical Implications. Curr Hematol Malig Rep 2024; 19:186-196. [PMID: 38888822 DOI: 10.1007/s11899-024-00736-9] [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] [Accepted: 06/08/2024] [Indexed: 06/20/2024]
Abstract
PURPOSE OF REVIEW Measurable residual disease (MRD) is integral in the management of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). This review discusses the current methods used to evaluate MRD as well as the interpretation, significance, and incorporation of MRD in current practice. RECENT FINDINGS New molecular technologies have allowed the detection of MRD to levels as low as 10- 6. The most used techniques to evaluate MRD are multiparametric flow cytometry (MFC), quantitative reverse transcription polymerase chain reaction (RT-qPCR), and high-throughput next-generation sequencing (NGS). Each method varies in terms of advantages, disadvantages, and MRD sensitivity. MRD negativity after induction treatment and after allogeneic hematopoietic cell transplantation (HCT) is an important prognostic marker that has consistently been shown to be associated with improved outcomes. Blinatumomab, a new targeted therapy for Ph + ALL, demonstrates high efficacy in eradicating MRD and improving patient outcomes. In the relapsed/refractory setting, the use of inotuzumab ozogamicin and tisagenlecleucel has shown promise in eradicating MRD. The presence of MRD has become an important predictive measure in Ph + ALL. Current studies evaluate the use of MRD in treatment decisions, especially in expanding therapeutic options for Ph + ALL, including tyrosine kinase inhibitors, targeted antibody therapies, chimeric antigen receptor cell therapy, and HCT.
Collapse
Affiliation(s)
- Valerie Tran
- Division of Hematology and Oncology, Department of Medicine, The University of Virginia, Charlottesville, VA, USA
| | - Kiarash Salafian
- Department of Medicine, The University of Virginia, Charlottesville, VA, USA
| | - Kenan Michaels
- Department of Medicine, The University of Virginia, Charlottesville, VA, USA
| | - Caroline Jones
- Division of Hematology and Oncology, Department of Medicine, The University of Virginia, Charlottesville, VA, USA
| | - Daniel Reed
- Division of Hematology and Oncology, Department of Medicine, The University of Virginia, Charlottesville, VA, USA
| | - Michael Keng
- Division of Hematology and Oncology, Department of Medicine, The University of Virginia, Charlottesville, VA, USA
| | - Firas El Chaer
- Division of Hematology and Oncology, Department of Medicine, The University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
21
|
Viscovo M, Clemmensen MDL, Fosso F, Maiolo E, Autore F, Laurenti L, Hohaus S, Chiusolo P. PlentiPlex™ MYD88 Waldenström lymphoma qPCR assay: A highly sensitive method for detection of MYD88 L265P mutation. Int J Lab Hematol 2024; 46:665-669. [PMID: 38390807 DOI: 10.1111/ijlh.14255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/21/2024] [Indexed: 02/24/2024]
Abstract
INTRODUCTION Agarose gel-based conventional and real-time allele-specific polymerase chain reaction (AS-PCR) assays are currently used for sensitive detection and quantification of MYD88 L265P mutation. Visual inspection of an agarose gel can often be ambiguous. We propose a new allele-specific quantification PCR (AS-qPCR) assay, PlentiPlex™ MYD88 Waldenström lymphoma qPCR assay, that uses Intercalating Nucleic Acid (INA®) technology for increased affinity and specificity. METHODS This study compares PlentiPlex™ MYD88 Waldenström lymphoma qPCR assay with conventional AS-PCR. We included a total of 102 peripheral and bone marrow blood samples from 94 patients with a lymphoproliferative disorder. Droplet digital PCR (ddPCR) was used as a third method in case of discrepancy. RESULTS A positive percent agreement of 100% (95% CI 0.92-1.0) and a negative percent agreement of 98% (95% CI 0.90-1.0) were found between the conventional AS-PCR and the AS-qPCR methods. Including the ddPCR results to validate the discrepant cases, the sensitivity and specificity of PlentiPlex™ MYD88 Waldenström lymphoma qPCR Assay were 1.0 (95% CI 0.97-1.0) and 1.0 (95% CI 0.96-1.0), respectively. CONCLUSION Our data demonstrate that PlentiPlex™ MYD88 Waldenström lymphoma qPCR assay is a fast, highly sensitive, and specific method for the detection of MYD88 L265P compared with conventional AS-PCR.
Collapse
Affiliation(s)
- Marcello Viscovo
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Rome, Italy
| | | | - Federica Fosso
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A Gemelli IRCC, Rome, Italy
| | - Elena Maiolo
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A Gemelli IRCC, Rome, Italy
| | - Francesco Autore
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A Gemelli IRCC, Rome, Italy
| | - Luca Laurenti
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Rome, Italy
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A Gemelli IRCC, Rome, Italy
| | - Stefan Hohaus
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Rome, Italy
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A Gemelli IRCC, Rome, Italy
| | - Patrizia Chiusolo
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Rome, Italy
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A Gemelli IRCC, Rome, Italy
| |
Collapse
|
22
|
Huang YJ, Chen SH, Liu HC, Jaing TH, Yeh TC, Kuo MC, Lin TL, Chen CC, Wang SC, Chang TK, Hsiao CC, Liang DC, Shih LY. Evaluation of next-generation sequencing for measurable residual disease monitoring in three major fusion transcript subtypes of B-precursor acute lymphoblastic leukaemia. Pathology 2024; 56:681-687. [PMID: 38719770 DOI: 10.1016/j.pathol.2024.02.008] [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: 03/28/2023] [Revised: 01/18/2024] [Accepted: 02/07/2024] [Indexed: 07/07/2024]
Abstract
The use of next-generation sequencing (NGS) for monitoring measurable residual disease (MRD) in acute lymphoblastic leukaemia (ALL) has been gaining traction. This study aimed to investigate the utility of NGS in MRD monitoring for the three major fusion transcript (FT) subtypes of B-precursor ALL (B-ALL). The MRD results for 104 bone marrow samples from 56 patients were analysed through NGS and real time quantitative reverse transcription PCR (RT-qPCR) for the three major FTs: BCR::ABL1, TCF3::PBX1, and ETV6::RUNX1. To validate the NGS approach, NGS-MRD was initially compared with allele-specific oligonucleotide-qPCR-MRD, and the coefficient of determination was good (R2=0.8158). A subsequent comparison of NGS-MRD with FT-MRD yielded a good coefficient of determination (R2=0.7690), but the coefficient varied by subtype. Specifically, the R2 was excellent for TCF3::PBX1 ALL (R2=0.9157), good for ETV6::RUNX1 ALL (R2=0.8606), and subpar for BCR::ABL1 ALL (R2=0.5763). The overall concordance between the two methods was 83.7%, and an excellent concordance rate of 95.8% was achieved for TCF3::PBX1 ALL. Major discordance, which was defined as a >1 log difference between discordant NGS-MRD and FT-MRD, occurred in 6.7% of the samples, with all but one sample being BCR::ABL1 ALL. Among the four non-transplanted patients with BCR::ABL1-MRD (+)/NGS-MRD (-), three did not relapse after long-term follow-up. Our finding indicates that NGS-MRD has a better prognostic impact than RT-qPCR-MRD in ETV6::RUNX1 and BCR::ABL1 ALL, whereas in TCF3::PBX1 ALL, both methods exhibit comparable efficacy.
Collapse
Affiliation(s)
- Ying-Jung Huang
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Shih-Hsiang Chen
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Hematology-Oncology, Chang Gung Children's Hospital at Linkou, Taoyuan, Taiwan
| | - Hsi-Che Liu
- Department of Hematology-Oncology, MacKay Children's Hospital and Mackay Medical College, Taipei, Taiwan
| | - Tang-Her Jaing
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Hematology-Oncology, Chang Gung Children's Hospital at Linkou, Taoyuan, Taiwan
| | - Ting-Chi Yeh
- Department of Hematology-Oncology, MacKay Children's Hospital and Mackay Medical College, Taipei, Taiwan
| | - Ming-Chung Kuo
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tung-Liang Lin
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chiu-Chen Chen
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Shih-Chung Wang
- Division of Pediatric Hematology-Oncology, Changhua Christian Children's Hospital, Changhua, Taiwan
| | - Te-Kau Chang
- Division of Pediatric Hematology and Oncology, China Medical University Children's Hospital, Taichung, Taiwan
| | - Chih-Cheng Hsiao
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Pediatrics, Chang Gung Memorial Hospital at Kaohsiung, Kaohsiung, Taiwan
| | - Der-Cherng Liang
- Department of Hematology-Oncology, MacKay Children's Hospital and Mackay Medical College, Taipei, Taiwan
| | - Lee-Yung Shih
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
23
|
Clerico M, Ferrero S, Alessandria B, Zaccaria GM, Genuardi E, Ragaini S, Tavarozzi R, Cavallo F, Hohaus S, Musuraca G, Carella AM, Stelitano C, Tani M, Gaidano G, Olivieri J, Usai SV, Galimberti S, Re F, Mian M, Castellino C, Pavone V, Evangelista A, Bruno B, Cortelazzo S, Passera R, Ladetto M. Stem cell collection and hematological recovery in the Fondazione Italiana Linfomi (FIL) MCL0208 clinical trial. Sci Rep 2024; 14:16946. [PMID: 39043871 PMCID: PMC11266400 DOI: 10.1038/s41598-024-67906-w] [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: 02/14/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
In the frontline high-dose phase 3 FIL-MCL0208 trial (NCT02354313), 8% of enrolled mantle cell lymphoma (MCL) patients could not be randomised to receive lenalidomide (LEN) maintenance vs observation after autologous stem cell transplantation (ASCT) due to inadequate hematological recovery and 52% of those who started LEN, needed a dose reduction due to toxicity. We therefore focused on the role played by CD34 + hematopoietic stem cells (PBSC) harvesting and reinfusion on toxicity and outcome. Overall, 90% (n = 245) of enrolled patients who underwent the first leukapheresis collected ≥ 4 × 106 PBSC/kg, 2.6% (n = 7) mobilized < 4 × 106 PBSC/kg and 7.7% (n = 21) failed the collection. Similar results were obtained for the planned second leukapheresis, with only one patient failing both attempts. Median count of reinfused PBSC was 5 × 106/kg and median time to recovery from neutropenia G4 was 10 days from ASCT. No impact of mobilizing subtype or number of reinfused PBSC on hematological recovery and LEN dose reduction was noted. At a median follow-up of 75 months from ASCT, PFS and OS of transplanted patients were 50% and 73%, respectively. A long lasting G4 neutropenia after ASCT (> 10 days) was associated with a worse outcome, both in terms of PFS and OS. In conclusion, although the harvesting procedures proved feasible for younger MCL patients, long-lasting cytopenia following ASCT remains a significant issue: this can hinder the administration of effective maintenance therapies, potentially increasing the relapse rate and negatively affecting survival outcomes.
Collapse
Affiliation(s)
- Michele Clerico
- Division of Hematology, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
| | - Simone Ferrero
- Division of Hematology, AOU "Città della Salute e della Scienza di Torino", Torino, Italy.
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy.
| | - Beatrice Alessandria
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Gian Maria Zaccaria
- Department of Electrical and Information Engineering (DEI), Polytechnic University of Bari, Bari, Italy
| | - Elisa Genuardi
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Simone Ragaini
- Division of Hematology, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Rita Tavarozzi
- SC Ematologia Azienda Ospedaliera Santi Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Federica Cavallo
- Division of Hematology, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Stefan Hohaus
- Institute of Hematology, Università Cattolica del Sacro Cuore and Fondazione Policlinico Univeristario A. Gemelli, Roma, Italy
| | - Gerardo Musuraca
- IRCCS, IRST Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori "Dino Amadori", Meldola, Italy
| | - Angelo Michele Carella
- Department of Hematology and Bone Marrow Transplant, IRCSS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Caterina Stelitano
- Department of Hematology, Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Monica Tani
- UOC di Ematologia, Ospedale S. Maria delle Croci, Ravenna, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Jacopo Olivieri
- Clinica Ematologica, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | | | - Sara Galimberti
- Division of Hematology, University Hospital of Pisa, Pisa, Italy
| | - Francesca Re
- Division of Immuno-Haematology, AOU Parma, Parma, Italy
| | - Michael Mian
- Innovation, Research and Teaching Service (SABES-ASDAA), Teaching Hospital of the Paracelsus Medical Private University (PMU), Bolzano, Italy
- College of Health Care-Professions Claudiana, Bozen, Italy
| | - Claudia Castellino
- Division of Hematology, Azienda Ospedaliera S. Croce e Carle, Cuneo, Italy
| | - Vincenzo Pavone
- Ospedale Pia Fondazione Cardinale Panico, Tricase, Lecce, Italy
| | - Andrea Evangelista
- Unit of Clinical Epidemiology, AOU "Città della Salute e della Scienza di Torino" and CPO Piemonte, Torino, Italy
| | - Benedetto Bruno
- Division of Hematology, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | | | - Roberto Passera
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Marco Ladetto
- SC Ematologia Azienda Ospedaliera Santi Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| |
Collapse
|
24
|
Stamatopoulos K, Pavlova S, Al‐Sawaf O, Chatzikonstantinou T, Karamanidou C, Gaidano G, Cymbalista F, Kater AP, Rawstron A, Scarfò L, Ghia P, Rosenquist R. Realizing precision medicine in chronic lymphocytic leukemia: Remaining challenges and potential opportunities. Hemasphere 2024; 8:e113. [PMID: 39035106 PMCID: PMC11260284 DOI: 10.1002/hem3.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/29/2024] [Accepted: 06/04/2024] [Indexed: 07/23/2024] Open
Abstract
Patients with chronic lymphocytic leukemia (CLL) exhibit diverse clinical outcomes. An expanding array of genetic tests is now employed to facilitate the identification of patients with high-risk disease and inform treatment decisions. These tests encompass molecular cytogenetic analysis, focusing on recurrent chromosomal alterations, particularly del(17p). Additionally, sequencing is utilized to identify TP53 mutations and to determine the somatic hypermutation status of the immunoglobulin heavy variable gene. Concurrently, a swift advancement of targeted treatment has led to the implementation of novel strategies for patients with CLL, including kinase and BCL2 inhibitors. This review explores both current and emerging diagnostic tests aimed at identifying high-risk patients who should benefit from targeted therapies. We outline existing treatment paradigms, emphasizing the importance of matching the right treatment to the right patient beyond genetic stratification, considering the crucial balance between safety and efficacy. We also take into consideration the practical and logistical issues when choosing a management strategy for each individual patient. Furthermore, we delve into the mechanisms underlying therapy resistance and stress the relevance of monitoring measurable residual disease to guide treatment decisions. Finally, we underscore the necessity of aggregating real-world data, adopting a global perspective, and ensuring patient engagement. Taken together, we argue that precision medicine is not the mere application of precision diagnostics and accessibility of precision therapies in CLL but encompasses various aspects of the patient journey (e.g., lifestyle exposures and comorbidities) and their preferences toward achieving true personalized medicine for patients with CLL.
Collapse
Affiliation(s)
- Kostas Stamatopoulos
- Centre for Research and Technology HellasInstitute of Applied BiosciencesThessalonikiGreece
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
| | - Sarka Pavlova
- Department of Internal Medicine, Hematology and Oncology, and Institute of Medical Genetics and GenomicsUniversity Hospital Brno and Medical Faculty, Masaryk UniversityBrnoCzech Republic
- Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Othman Al‐Sawaf
- Department I of Internal Medicine and German CLL Study Group, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)University of Cologne, Faculty of Medicine and University Hospital of CologneCologneGermany
- Francis Crick Institute LondonLondonUK
- Cancer Institute, University College LondonLondonUK
| | | | - Christina Karamanidou
- Centre for Research and Technology HellasInstitute of Applied BiosciencesThessalonikiGreece
| | - Gianluca Gaidano
- Division of Haematology, Department of Translational MedicineUniversity of Eastern PiedmontNovaraItaly
| | | | - Arnon P. Kater
- Department of Hematology, Cancer Center AmsterdamAmsterdam University Medical Centers, University of AmsterdamAmsterdamThe Netherlands
| | - Andy Rawstron
- Haematological Malignancy Diagnostic ServiceLeeds Teaching Hospitals TrustLeedsUK
| | - Lydia Scarfò
- Medical SchoolUniversità Vita Salute San RaffaeleMilanoItaly
- Strategic Research Program on CLLIRCCS Ospedale San RaffaeleMilanoItaly
| | - Paolo Ghia
- Medical SchoolUniversità Vita Salute San RaffaeleMilanoItaly
- Strategic Research Program on CLLIRCCS Ospedale San RaffaeleMilanoItaly
| | - Richard Rosenquist
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
- Clinical GeneticsKarolinska University HospitalStockholmSweden
| |
Collapse
|
25
|
Pulsoni A, Ferrero S, Tosti ME, Luminari S, Dondi A, Cavallo F, Merli F, Liberati AM, Cenfra N, Renzi D, Zanni M, Boccomini C, Ferreri AJM, Rattotti S, Zilioli VR, Bolis SA, Bernuzzi P, Musuraca G, Gaidano G, Perrone T, Stelitano C, Tucci A, Corradini P, Bigliardi S, Re F, Cencini E, Mannarella C, Mannina D, Celli M, Tani M, Annechini G, Assanto GM, Grapulin L, Guarini A, Cavalli M, De Novi LA, Bomben R, Ciabatti E, Genuardi E, Drandi D, Della Starza I, Arcaini L, Ricardi U, Gattei V, Galimberti S, Ladetto M, Foà R, Del Giudice I. Local radiotherapy and measurable residual disease-driven immunotherapy in patients with early-stage follicular lymphoma (FIL MIRO): final results of a prospective, multicentre, phase 2 trial. Lancet Haematol 2024; 11:e499-e509. [PMID: 38937025 DOI: 10.1016/s2352-3026(24)00143-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND The mainstay of treatment for early-stage follicular lymphoma is local radiotherapy, with a possible role for anti-CD20 monoclonal antibody (mAb). We aimed to evaluate the effect of these treatments using a measurable residual disease (MRD)-driven approach. METHODS This prospective, multicentre, phase 2 trial was conducted at 27 centres of the Fondazione Italiana Linfomi (FIL) in Italy. Eligible participants were adults (≥18 years) with newly diagnosed, histologically confirmed follicular lymphoma (stage I or II; grade I-IIIa). Patients were initially treated with 24 Gy involved-field radiotherapy over 12 days; those who were MRD-positive after radiotherapy or during follow-up received eight intravenous doses (1000 mg per dose; one dose per week) of the anti-CD20 mAb ofatumumab. The primary endpoint was the proportion of patients who were MRD-positive after involved-field radiotherapy and became MRD-negative after ofatumumab treatment. Patients were included in the primary endpoint analysis population if they were positive for BCL2::IGH rearrangement at enrolment in peripheral blood or bone marrow samples. MRD positivity was defined as the persistence of BCL2::IGH rearrangement in peripheral blood or bone marrow, assessed centrally by laboratories of the FIL MRD Network. The trial was registered with EudraCT, 2012-001676-11. FINDINGS Between May 2, 2015, and June 1, 2018, we enrolled 110 participants, of whom 106 (96%) were eligible and received involved-field radiotherapy. Of these, 105 (99%) were White, one (1%) was Black, 50 (47%) were male, and 56 (53%) were female. Of 105 participants in whom BCL2::IGH status was evaluable, 32 (30%) had a detectable BCL2::IGH rearrangement at baseline. After radiotherapy, 12 (40%) of 30 patients reached MRD-negative status, which was long-lasting (at least 36 or 42 months) in three (25%). In those who were MRD-positive after radiotherapy, ofatumumab induced MRD-negativity in 23 (92%; 95% CI 74-99) of 25 evaluable patients. After a median follow-up of 46·1 months (IQR 42·8-50·8), 14 (61%) of these 23 patients remain in complete response and are MRD-negative. The most common grade 3-4 adverse events were infusion-related reactions, observed in four patients. INTERPRETATION Local radiotherapy is frequently not associated with the eradication of follicular lymphoma. An MRD-driven, anti-CD20 monoclonal antibody consolidation enables molecular remission to be reached in almost all patients and is associated with a reduced incidence of relapse over time. A clinical advantage of an MRD-driven consolidation is therefore suggested. FUNDING AIRC Foundation for Cancer Research in Italy, Novartis International, and GlaxoSmithKline.
Collapse
Affiliation(s)
- Alessandro Pulsoni
- Hematology, Department of Translational and Precision Medicine, Sapienza University-Polo Pontino, S.M. Goretti Hospital, Latina, Italy.
| | - Simone Ferrero
- Hematology Division, Department of Molecular Biotechnologies and Health Sciences, University of Torino/AOU "Città della Salute e della Scienza di Torino", Turin, Italy
| | - Maria Elena Tosti
- Istituto Superiore di Sanità, National Center for Global Health, Rome, Italy
| | - Stefano Luminari
- Hematology Unit, Arcispedale S Maria Nuova, Azienda Unità Sanitaria Locale-IRCCS, Reggio Emilia, Italy; Department CHIMOMO, University of Modena and Reggio Emilia, Reggio Emilia, Italy
| | | | - Federica Cavallo
- Hematology Division, Department of Molecular Biotechnologies and Health Sciences, University of Torino/AOU "Città della Salute e della Scienza di Torino", Turin, Italy
| | - Francesco Merli
- Hematology Unit, Arcispedale S Maria Nuova, Azienda Unità Sanitaria Locale-IRCCS, Reggio Emilia, Italy
| | | | - Natalia Cenfra
- Hematology Unit, S Maria Goretti Hospital, AUSL Latina, Latina, Italy
| | - Daniela Renzi
- Hematology and Stem Cells Transplantation Unit, IRCCS Istituto Nazionale dei Tumori Regina Elena, Rome, Italy
| | - Manuela Zanni
- Division of Hematology, SS Antonio e Biagio Hospital, Alessandria, Italy
| | - Carola Boccomini
- SC Hematology, AOU Città della Salute e della Scienza, Turin, Italy
| | - Andrés J M Ferreri
- Lymphoma Unit, IRCCS San Raffaele Scientific Institute, University Vita-Salute San Raffaele, Milan, Italy
| | - Sara Rattotti
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Silvia Anna Bolis
- Struttura Complessa di Ematologia, Fondazione IRCCS San Gerardo dei Tintori-Monza, Monza, Italy
| | - Patrizia Bernuzzi
- Hematology Unit, Department of Onco-Hematology, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Gerardo Musuraca
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Tommasina Perrone
- Unit of Hematology with Transplantation, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Caterina Stelitano
- Department of Hematology, Azienda Ospedaliera Bianchi Melacrinò Morelli, Reggio Calabria, Italy
| | | | - Paolo Corradini
- Division of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Sara Bigliardi
- Onco-Hematology Department, Nuovo Ospedale Civile di Sassuolo, Sassuolo, Italy
| | | | - Emanuele Cencini
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese and University of Siena, Siena, Italy
| | | | - Donato Mannina
- Department of Hematology, Azienda Ospedaliera Papardo, Messina, Italy
| | | | - Monica Tani
- Hematology Unit, Santa Maria delle Croci Hospital, Ravenna, Italy
| | - Giorgia Annechini
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giovanni Manfredi Assanto
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Lavinia Grapulin
- Department of Radiotherapy, AOU Policlinico Umberto I, Rome, Italy
| | - Anna Guarini
- Hematology, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Marzia Cavalli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia Anna De Novi
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Riccardo Bomben
- Clinical and Experimental Onco-Hematology Unit, CRO Aviano National Cancer Institute, Aviano, Italy
| | - Elena Ciabatti
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elisa Genuardi
- Hematology Division, Department of Molecular Biotechnologies and Health Sciences, University of Torino/AOU "Città della Salute e della Scienza di Torino", Turin, Italy
| | - Daniela Drandi
- Hematology Division, Department of Molecular Biotechnologies and Health Sciences, University of Torino/AOU "Città della Salute e della Scienza di Torino", Turin, Italy
| | - Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy; AIL Roma, ODV, Rome, Italy
| | - Luca Arcaini
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Umberto Ricardi
- Radiation Oncology, Department of Oncology, University of Turin, Turin, Italy
| | - Valter Gattei
- Clinical and Experimental Onco-Hematology Unit, CRO Aviano National Cancer Institute, Aviano, Italy
| | - Sara Galimberti
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marco Ladetto
- Department of Translational Medicine, SCDU Ematologia AO SS Antonio e Biagio E Cesare Arrigo, Università del Piemonte Orientale, Alessandria, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ilaria Del Giudice
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
26
|
Kotrova M, Fronkova E, Svaton M, Drandi D, Schön F, Hoogeveen P, Hancock J, Skotnicova A, Schilhabel A, Eckert C, Clappier E, Cazzaniga G, Schäfer BW, van Dongen JJM, Ritgen M, Pott C, van der Velden VHJ, Trka J, Brüggemann M. The gray area of RQ-PCR-based measurable residual disease: subdividing the "positive, below quantitative range" category. Leukemia 2024; 38:1617-1620. [PMID: 38760480 PMCID: PMC11216996 DOI: 10.1038/s41375-024-02265-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 05/19/2024]
Affiliation(s)
- Michaela Kotrova
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Eva Fronkova
- CLIP-Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Michael Svaton
- CLIP-Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Austrian Academy of Sciences, CeMM Research Center for Molecular Medicine, Vienna, Austria
| | - Daniela Drandi
- Hematology Division, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Felix Schön
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Patricia Hoogeveen
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jeremy Hancock
- Bristol Genetics Laboratory, Southmead Hospital, Bristol, UK
| | - Aneta Skotnicova
- CLIP-Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Anke Schilhabel
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Cornelia Eckert
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Emmanuelle Clappier
- Hematology Laboratory, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Gianni Cazzaniga
- Tettamanti Cente, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Beat W Schäfer
- Department of Hematology, University Hospital, Zürich, Switzerland
| | - Jacques J M van Dongen
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Matthias Ritgen
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christiane Pott
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Vincent H J van der Velden
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jan Trka
- CLIP-Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Monika Brüggemann
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
27
|
Burmeister T, Ströh AS, Kehden B, Trautmann H, Meyer C, Marschalek R, Larghero P, Schwartz S, Steffen B, Spriewald B, Heinicke T, Jäkel N, Westermann J, Nachtkamp K, Viardot A, Topp MS, Neumann M, Baldus CD, Gökbuget N, Brüggemann M. Measurable residual disease quantification in adult patients with KMT2A-rearranged acute lymphoblastic leukemia. Leukemia 2024; 38:1600-1603. [PMID: 38519799 PMCID: PMC11216977 DOI: 10.1038/s41375-024-02209-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/25/2024]
Affiliation(s)
- Thomas Burmeister
- Department of Hematology, Oncology and Tumor Immunology, CVK, Charité-Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Aeint-Steffen Ströh
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Britta Kehden
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Heiko Trautmann
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Claus Meyer
- Diagnostic Center of Acute Leukemia (DCAL), Institute of Pharmaceutical Biology, Goethe University, Frankfurt, Germany
| | - Rolf Marschalek
- Diagnostic Center of Acute Leukemia (DCAL), Institute of Pharmaceutical Biology, Goethe University, Frankfurt, Germany
| | - Patrizia Larghero
- Diagnostic Center of Acute Leukemia (DCAL), Institute of Pharmaceutical Biology, Goethe University, Frankfurt, Germany
| | - Stefan Schwartz
- Department of Hematology, Oncology and Tumor Immunology, CBF, Charité Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Björn Steffen
- Department of Medicine II, Goethe University, University Hospital, Frankfurt, Germany
| | - Bernd Spriewald
- University Hospital Erlangen, Department of Internal Medicine 5, Hematology and Oncology, Erlangen, Germany
| | - Thomas Heinicke
- Department of Hematology and Oncology, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Nadja Jäkel
- Department of Hematology, Oncology, University Hospital, Halle/Saale, Germany
| | - Jörg Westermann
- Department of Hematology, Oncology and Tumor Immunology, CVK, Charité-Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kathrin Nachtkamp
- Department of Hematology, Oncology, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Andreas Viardot
- Department of Hematology, Oncology, University Hospital Ulm, Ulm, Germany
| | - Max S Topp
- Department of Hematology, Oncology, University Hospital Würzburg, Würzburg, Germany
| | - Martin Neumann
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Claudia D Baldus
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Nicola Gökbuget
- Department of Medicine II, Goethe University, University Hospital, Frankfurt, Germany
| | - Monika Brüggemann
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
28
|
Patel B, Kirkwood AA, Rowntree CJ, Alapi KZ, Barretta E, Clifton‐Hadley L, Creasey T, Lee S, Marks DI, Moorman AV, Morley N, Patrick P, Rana Z, Rijneveld A, Snowden JA, Fielding AK. Results from UKALL60+, a phase 2 study in older patients with untreated acute lymphoblastic leukemia. Hemasphere 2024; 8:e88. [PMID: 38846719 PMCID: PMC11154829 DOI: 10.1002/hem3.88] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 05/08/2024] [Indexed: 06/09/2024] Open
Affiliation(s)
- Bela Patel
- Centre for Haemato‐Oncology, Barts Cancer InstituteQueen Mary University of LondonLondonUK
| | - Amy A. Kirkwood
- CR UK and UCL Cancer Trial Centre, UCL Cancer Institute, UCLLondonUK
| | | | | | - Emilio Barretta
- Leukemia Research Cytogenetics Group, Translational and Clinical Research InstituteNewcastle UniversityNewcastle Upon TyneUK
| | | | - Tom Creasey
- Leukemia Research Cytogenetics Group, Translational and Clinical Research InstituteNewcastle UniversityNewcastle Upon TyneUK
| | - SooWah Lee
- Hematology, UCL Cancer InstituteLondonUK
| | | | - Anthony V. Moorman
- Leukemia Research Cytogenetics Group, Translational and Clinical Research InstituteNewcastle UniversityNewcastle Upon TyneUK
| | - Nicholas Morley
- Royal Hallamshire HospitalSheffield Teaching Hospital NHS Foundation TrustSheffieldUK
| | - Pip Patrick
- CR UK and UCL Cancer Trial Centre, UCL Cancer Institute, UCLLondonUK
| | - Zaynab Rana
- CR UK and UCL Cancer Trial Centre, UCL Cancer Institute, UCLLondonUK
| | | | - John A. Snowden
- Royal Hallamshire HospitalSheffield Teaching Hospital NHS Foundation TrustSheffieldUK
| | | |
Collapse
|
29
|
van der Velden VHJ, Dombrink I, Alten J, Cazzaniga G, Clappier E, Drandi D, Eckert C, Fronkova E, Hancock J, Kotrova M, Kraemer R, Montonen M, Pfeifer H, Pott C, Raff T, Trautmann H, Cavé H, Schäfer BW, van Dongen JJM, Trka J, Brüggemann M. Analysis of measurable residual disease by IG/TR gene rearrangements: quality assurance and updated EuroMRD guidelines. Leukemia 2024; 38:1315-1322. [PMID: 38744919 PMCID: PMC11147754 DOI: 10.1038/s41375-024-02272-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
Minimal/measurable residual disease (MRD) diagnostics using real-time quantitative PCR analysis of rearranged immunoglobulin and T-cell receptor gene rearrangements are nowadays implemented in most treatment protocols for patients with acute lymphoblastic leukemia (ALL). Within the EuroMRD Consortium, we aim to provide comparable, high-quality MRD diagnostics, allowing appropriate risk-group classification for patients and inter-protocol comparisons. To this end, we set up a quality assessment scheme, that was gradually optimized and updated over the last 20 years, and that now includes participants from around 70 laboratories worldwide. We here describe the design and analysis of our quality assessment scheme. In addition, we here report revised data interpretation guidelines, based on our newly generated data and extensive discussions between experts. The main novelty is the partial re-definition of the "positive below quantitative range" category by two new categories, "MRD low positive, below quantitative range" and "MRD of uncertain significance". The quality assessment program and revised guidelines will ensure reproducible and accurate MRD data for ALL patients. Within the Consortium, similar programs and guidelines have been introduced for other lymphoid diseases (e.g., B-cell lymphoma), for new technological platforms (e.g., digital droplet PCR or Next-Generation Sequencing), and for other patient-specific MRD PCR-based targets (e.g., fusion genes).
Collapse
Affiliation(s)
- Vincent H J van der Velden
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Isabel Dombrink
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Julia Alten
- Department of Pediatrics, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Giovanni Cazzaniga
- Centro Tettamanti, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine, University of Milano-Bicocca, Monza, Italy
| | - Emmanuelle Clappier
- Hematology Laboratory, Saint-Louis Hospital, Paris Cité University, Paris, France
- Université Paris-Cité, Paris, France
| | - Daniela Drandi
- Department of Molecular Biotechnology and health sciences, Hematology Division, University of Torino, Torino, Italy
| | - Cornelia Eckert
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eva Fronkova
- CLIP, Department of Pediatric Hematology and Oncology, Second Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Jeremy Hancock
- Bristol MRD Group, Bristol Genetics Laboratory, Southmead Hospital, Bristol, UK
| | - Michaela Kotrova
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Rebekka Kraemer
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Mirkka Montonen
- Tyks Laboratories, Genomics Department, Turku University Hospital, Turku, Finland
| | - Heike Pfeifer
- Department of Hematology, University Hospital Frankfurt, Frankfurt, Germany
| | - Christiane Pott
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thorsten Raff
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
- Military Medical City Hospital, Doha, Qatar
| | - Heiko Trautmann
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Hélène Cavé
- Hematology Laboratory, Saint-Louis Hospital, Paris Cité University, Paris, France
- Department of Genetics, University Hospital Robert Debré, Paris, France
| | | | - Jacques J M van Dongen
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CIC-IBMCC, USAL-CSIC-FICUS) and Department of Medicine, University of Salamanca, Salamanca, Spain
- European Scientific foundation for Laboratory Hemato Oncology (ESLHO), Zutphen, The Netherlands
- Department of Immunology, LUMC, Leiden, The Netherlands
| | - Jan Trka
- CLIP, Department of Pediatric Hematology and Oncology, Second Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Monika Brüggemann
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
30
|
Del Giudice I, Della Starza I, De Falco F, Gaidano G, Sportoletti P. Monitoring Response and Resistance to Treatment in Chronic Lymphocytic Leukemia. Cancers (Basel) 2024; 16:2049. [PMID: 38893168 PMCID: PMC11171231 DOI: 10.3390/cancers16112049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/09/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
The recent evolution in chronic lymphocytic leukemia (CLL) targeted therapies led to a progressive change in the way clinicians manage the goals of treatment and evaluate the response to treatment in respect to the paradigm of the chemoimmunotherapy era. Continuous therapies with BTK inhibitors achieve prolonged and sustained control of the disease. On the other hand, venetoclax and anti-CD20 monoclonal antibodies or, more recently, ibrutinib plus venetoclax combinations, given for a fixed duration, achieve undetectable measurable residual disease (uMRD) in the vast majority of patients. On these grounds, a time-limited MRD-driven strategy, a previously unexplored scenario in CLL, is being attempted. On the other side of the spectrum, novel genetic and non-genetic mechanisms of resistance to targeted treatments are emerging. Here we review the response assessment criteria, the evolution and clinical application of MRD analysis and the mechanisms of resistance according to the novel treatment strategies within clinical trials. The extent to which this novel evidence will translate in the real-life management of CLL patients remains an open issue to be addressed.
Collapse
Affiliation(s)
- Ilaria Del Giudice
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00161 Rome, Italy;
| | - Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00161 Rome, Italy;
- AIL Roma, ODV, 00161 Rome, Italy
| | - Filomena De Falco
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncological Research, University of Perugia, 06129 Perugia, Italy;
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy;
| | - Paolo Sportoletti
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncological Research, University of Perugia, 06129 Perugia, Italy;
| |
Collapse
|
31
|
Lebecque B, Besombes J, Dannus LT, De Antonio M, Cacheux V, Grèze V, Montagnon V, Veronese L, Tchirkov A, Tournilhac O, Berger MG, Veyrat-Masson R. Faster clinical decisions in B-cell acute lymphoblastic leukaemia: A single flow cytometric 12-colour tube improves diagnosis and minimal residual disease follow-up. Br J Haematol 2024; 204:1872-1881. [PMID: 38432068 DOI: 10.1111/bjh.19390] [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: 01/12/2024] [Revised: 02/22/2024] [Accepted: 02/24/2024] [Indexed: 03/05/2024]
Abstract
Assessing minimal residual disease (MRD) in B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) is essential for adjusting therapeutic strategies and predicting relapse. Quantitative polymerase chain reaction (qPCR) is the gold standard for MRD. Alternatively, flow cytometry is a quicker and cost-effective method that typically uses leukaemia-associated immunophenotype (LAIP) or different-from-normal (DFN) approaches for MRD assessment. This study describes an optimized 12-colour flow cytometry antibody panel designed for BCP-ALL diagnosis and MRD monitoring in a single tube. This method robustly differentiated hematogones and BCP-ALL cells using two specific markers: CD43 and CD81. These and other markers (e.g. CD73, CD66c and CD49f) enhanced the specificity of BCP-ALL cell detection. This innovative approach, based on a dual DFN/LAIP strategy with a principal component analysis method, can be used for all patients and enables MRD analysis even in the absence of a diagnostic sample. The robustness of our method for MRD monitoring was confirmed by the strong correlation (r = 0.87) with the qPCR results. Moreover, it simplifies and accelerates the preanalytical process through the use of a stain/lysis/wash method within a single tube (<2 h). Our flow cytometry-based methodology improves the BCP-ALL diagnosis efficiency and MRD management, offering a complementary method with considerable benefits for clinical laboratories.
Collapse
Affiliation(s)
- Benjamin Lebecque
- Hématologie Biologique, CHU Clermont-Ferrand, Estaing, Clermont-Ferrand, France
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Joevin Besombes
- Hématologie Biologique, CHU Clermont-Ferrand, Estaing, Clermont-Ferrand, France
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Louis-Thomas Dannus
- Hématologie Biologique, CHU Clermont-Ferrand, Estaing, Clermont-Ferrand, France
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Marie De Antonio
- Unité de Biostatistiques, Direction de la Recherche Clinique et de l'Innovation, Centre Hospitalier Universitaire de Clermont-Ferrand, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Victoria Cacheux
- Service de Thérapie Cellulaire et Hématologie Clinique Adulte, Clermont-Ferrand, France
| | - Victoria Grèze
- CHU Clermont-Ferrand, Service Hématologie Oncologie Pédiatrique, Hôpital Estaing, Clermont-Ferrand, France
| | - Valentin Montagnon
- Hématologie Biologique, CHU Clermont-Ferrand, Estaing, Clermont-Ferrand, France
| | - Lauren Veronese
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
- Cytogénétique Médicale, CHU Clermont-Ferrand, CHU Estaing, Clermont-Ferrand, France
| | - Andrei Tchirkov
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
- Cytogénétique Médicale, CHU Clermont-Ferrand, CHU Estaing, Clermont-Ferrand, France
| | - Olivier Tournilhac
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
- Service de Thérapie Cellulaire et Hématologie Clinique Adulte, Clermont-Ferrand, France
| | - Marc G Berger
- Hématologie Biologique, CHU Clermont-Ferrand, Estaing, Clermont-Ferrand, France
- Equipe d'Accueil EA7453 CHELTER, Université Clermont Auvergne, Clermont-Ferrand, France
| | | |
Collapse
|
32
|
Ueki H, Ogawa C, Goto H, Nishi M, Yamanaka J, Mochizuki S, Nishikawa T, Kumamoto T, Nishiuchi R, Kikuta A, Yamamoto S, Igarashi S, Sato A, Hori T, Saito AM, Watanabe T, Deguchi T, Manabe A, Horibe K, Toyoda H. TBI, etoposide, and cyclophosphamide conditioning for intermediate-risk relapsed childhood acute lymphoblastic leukemia. Int J Hematol 2024; 119:450-458. [PMID: 38267673 DOI: 10.1007/s12185-024-03710-6] [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: 04/26/2023] [Revised: 12/27/2023] [Accepted: 01/08/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND In children with intermediate-risk relapsed acute lymphoblastic leukemia (ALL), allogeneic hematopoietic stem cell transplantation (allo-HSCT) has markedly improved the outcome of patients with an unsatisfactory minimal residual disease (MRD) response. Total body irradiation (TBI), etoposide (ETP), and cyclophosphamide (CY) have been shown to be equivalent to or better than TBI + ETP for conditioning, so we hypothesized that even greater survival could be achieved due to recent advances in HSCT and supportive care. PROCEDURE We prospectively analyzed the efficacy and safety of allo-HSCT with a unified conditioning regimen of TBI + ETP + CY in children with intermediate-risk relapsed ALL, based on MRD in the bone marrow after induction, from the Japanese Pediatric Leukemia/Lymphoma Study Group (JPLSG) ALL-R08-II nationwide cohort (UMIN000002025). RESULTS Twenty patients with post-induction MRD ≥ 10-3 and two not evaluated for MRD underwent allo-HSCT. Engraftment was confirmed in all patients, and no transplantation-related mortality was observed. The 3-year event-free survival and overall survival rates after transplantation were 86.4% ± 7.3% and 95.5% ± 4.4%, respectively. CONCLUSION Allo-HSCT based on post-induction MRD with TBI + ETP + CY conditioning was feasible in Japanese children with intermediate-risk relapsed ALL.
Collapse
Affiliation(s)
- Hideaki Ueki
- Department of Pediatric Hematology/Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Chitose Ogawa
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroaki Goto
- Division of Hematology/Oncology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Masanori Nishi
- Department of Pediatrics, Faculty of Medicine, Saga University, Saga, Japan
| | - Junko Yamanaka
- Department of Pediatrics, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinji Mochizuki
- Department of Pediatrics, National Center for Global Health and Medicine, Tokyo, Japan
| | - Takuro Nishikawa
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Tadashi Kumamoto
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ritsuo Nishiuchi
- Department of Pediatrics, Kochi Health Sciences Center, Kochi, Japan
| | - Atsushi Kikuta
- Department of Pediatric Oncology, Fukushima Medical University Hospital, Fukushima, Japan
| | - Shohei Yamamoto
- Department of Pediatrics, Tokai University School of Medicine, Isehara, Japan
| | - Shunji Igarashi
- Department of Pediatric Hematology/Oncology, Japanese Red Cross Narita Hospital, Narita, Japan
| | - Atsushi Sato
- Department of Hematology/Oncology, Miyagi Children's Hospital, Sendai, Japan
| | - Toshinori Hori
- Department of Pediatrics, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Akiko M Saito
- Clinical Research Center, NHO Nagoya Medical Center, Nagoya, Japan
| | - Tomoyuki Watanabe
- Department of Nutritional Science, Faculty of Psychological and Physical Science, Aichi Gakuin University, Nisshin, Japan
| | - Takao Deguchi
- Division of Cancer Immunodiagnostics, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University, Sapporo, Japan
| | - Keizo Horibe
- Clinical Research Center, NHO Nagoya Medical Center, Nagoya, Japan
| | - Hidemi Toyoda
- Department of Pediatrics, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
| |
Collapse
|
33
|
Liu HC, Huang YJ, Jaing TH, Wu KH, Chen SH, Wang SC, Yeh TC, Hsiao CC, Chang TK, Yen HJ, Huang FL, Lin PC, Hou JY, Sheen JM, Liao YM, Chang TY, Chen YC, Chiou SS, Yang CP, Pui CH, Liang DC, Shih LY. Refining risk stratification in paediatric B-acute lymphoblastic leukaemia: Combining IKZF1 plus and Day 15 MRD positivity. Br J Haematol 2024; 204:1344-1353. [PMID: 38479427 DOI: 10.1111/bjh.19338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/29/2024] [Accepted: 02/01/2024] [Indexed: 04/11/2024]
Abstract
This study investigates the potential utility of IKZF1 deletion as an additional high-risk marker for paediatric acute lymphoblastic leukaemia (ALL). The prognostic impact of IKZF1 status, in conjunction with minimal/measurable residual disease (MRD), was evaluated within the MRD-guided TPOG-ALL-2013 protocol using 412 newly diagnosed B-ALL patients aged 1-18. IKZF1 status was determined using multiplex ligation-dependent probe amplification. IKZF1 deletions, when co-occurring with CDKN2A, CDKN2B, PAX5 or PAR1 region deletions in the absence of ERG deletions, were termed IKZF1plus. Both IKZF1 deletion (14.6%) and IKZF1plus (7.8%) independently predicted poorer outcomes in B-ALL. IKZF1plus was observed in 4.1% of Philadelphia-negative ALL, with a significantly lower 5-year event-free survival (53.9%) compared to IKZF1 deletion alone (83.8%) and wild-type IKZF1 (91.3%) (p < 0.0001). Among patients with Day 15 MRD ≥0.01%, provisional high-risk patients with IKZF1plus exhibited the worst outcomes in event-free survival (42.0%), relapse-free survival (48.0%) and overall survival (72.7%) compared to other groups (p < 0.0001). Integration of IKZF1plus and positive Day 15 MRD identified a subgroup of Philadelphia-negative B-ALL with a 50% risk of relapse. This study highlights the importance of assessing IKZF1plus alongside Day 15 MRD positivity to identify patients at increased risk of adverse outcomes, potentially minimizing overtreatment.
Collapse
Affiliation(s)
- Hsi-Che Liu
- Department of Hematology-Oncology, MacKay Children's Hospital and MacKay Medical College, Taipei, Taiwan
| | - Ying-Jung Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Tang-Her Jaing
- Department of Hematology-Oncology, Chang Gung Children's Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kang-Hsi Wu
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shih-Hsiang Chen
- Department of Hematology-Oncology, Chang Gung Children's Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Chung Wang
- Division of Pediatric Hematology-Oncology, Changhua Christian Children's Hospital, Changhua, Taiwan
| | - Ting-Chi Yeh
- Department of Hematology-Oncology, MacKay Children's Hospital and MacKay Medical College, Taipei, Taiwan
| | - Chih-Cheng Hsiao
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Pediatrics, Chang Gung Memorial Hospital at Kaohsiung, Kaohsiung, Taiwan
| | - Te-Kau Chang
- Division of Pediatric Hematology and Oncology, China Medical University Children's Hospital, Taichung, Taiwan
| | - Hsiu-Ju Yen
- Department of Pediatrics, Taipei Veterans General Hospital and School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Fang-Liang Huang
- Department of Pediatrics, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Pei-Chin Lin
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Jen-Yin Hou
- Department of Hematology-Oncology, MacKay Children's Hospital and MacKay Medical College, Taipei, Taiwan
| | - Jiunn-Ming Sheen
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Pediatrics, Chang Gung Memorial Hospital at Kaohsiung, Kaohsiung, Taiwan
- Department of Pediatrics, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
| | - Yu-Mei Liao
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Tsung-Yen Chang
- Department of Hematology-Oncology, Chang Gung Children's Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Chieh Chen
- Department of Pediatrics, Chang Gung Memorial Hospital at Kaohsiung, Kaohsiung, Taiwan
| | - Shyh-Shin Chiou
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chao-Ping Yang
- Department of Hematology-Oncology, Chang Gung Children's Hospital at Linkou, Taoyuan, Taiwan
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Der-Cherng Liang
- Department of Hematology-Oncology, MacKay Children's Hospital and MacKay Medical College, Taipei, Taiwan
| | - Lee-Yung Shih
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
34
|
Maurer-Granofszky M, Kohrer S, Fischer S, Schumich A, Nebral K, Larghero P, Meyer C, Mecklenbrauker A, Muhlegger N, Marschalek R, Haas OA, Panzer-Grumayer R, Dworzak MN. Genomic breakpoint-specific monitoring of measurable residual disease in pediatric non-standard-risk acute myeloid leukemia. Haematologica 2024; 109:740-750. [PMID: 37345487 PMCID: PMC10910191 DOI: 10.3324/haematol.2022.282424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 06/15/2023] [Indexed: 06/23/2023] Open
Abstract
Pediatric acute myeloid leukemia (AML) is a highly heterogeneous disease making standardized measurable residual disease (MRD) assessment challenging. Currently, patient-specific DNA-based assays are only rarely applied for MRD assessment in pediatric AML. We tested whether quantification of genomic breakpoint-specific sequences via quantitative polymerase chain reaction (gDNA-PCR) provides a reliable means of MRD quantification in children with non-standardrisk AML and compared its results to those obtained with state-of-the-art ten-color flow cytometry (FCM). Breakpointspecific gDNA-PCR assays were established according to Euro-MRD consortium guidelines. FCM-MRD assessment was performed according to the European Leukemia Network guidelines with adaptations for pediatric AML. Of 77 consecutively recruited non-standard-risk pediatric AML cases, 49 (64%) carried a chromosomal translocation potentially suitable for MRD quantification. Genomic breakpoint analysis returned a specific DNA sequence in 100% (41/41) of the cases submitted for investigation. MRD levels were evaluated using gDNA-PCR in 243 follow-up samples from 36 patients, achieving a quantitative range of at least 10-4 in 231/243 (95%) of samples. Comparing gDNA-PCR with FCM-MRD data for 183 bone marrow follow-up samples at various therapy timepoints showed a high concordance of 90.2%, considering a cut-off of ≥0.1%. Both methodologies outperformed morphological assessment. We conclude that MRD monitoring by gDNA-PCR is feasible in pediatric AML with traceable genetic rearrangements and correlates well with FCM-MRD in the currently applied clinically relevant range, while being more sensitive below that. The methodology should be evaluated in larger cohorts to pave the way for clinical application.
Collapse
Affiliation(s)
| | - Stefan Kohrer
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria; Labdia Labordiagnostik, Vienna
| | - Susanna Fischer
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria; Labdia Labordiagnostik, Vienna
| | - Angela Schumich
- St. Anna Children's Cancer Research Institute (CCRI), Vienna
| | - Karin Nebral
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria; Labdia Labordiagnostik, Vienna
| | - Patrizia Larghero
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia (DCAL), Goethe-University, Frankfurt/Main
| | - Claus Meyer
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia (DCAL), Goethe-University, Frankfurt/Main
| | - Astrid Mecklenbrauker
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria; Labdia Labordiagnostik, Vienna
| | - Nora Muhlegger
- St. Anna Children's Cancer Research Institute (CCRI), Vienna
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia (DCAL), Goethe-University, Frankfurt/Main
| | - Oskar A Haas
- St. Anna Children's Cancer Research Institute (CCRI), Vienna
| | | | - Michael N Dworzak
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria; Labdia Labordiagnostik, Vienna, Austria; St. Anna Children's Hospital, Department of Pediatrics, Medical University of Vienna, Vienna.
| |
Collapse
|
35
|
Hoster E, Delfau-Larue MH, Macintyre E, Jiang L, Stilgenbauer S, Vehling-Kaiser U, Salles G, Thieblemont C, Tilly H, Wirths S, Feugier P, Hübel K, Schmidt C, Ribrag V, Kluin-Nelemans JC, Dreyling M, Pott C. Predictive Value of Minimal Residual Disease for Efficacy of Rituximab Maintenance in Mantle Cell Lymphoma: Results From the European Mantle Cell Lymphoma Elderly Trial. J Clin Oncol 2024; 42:538-549. [PMID: 37992261 DOI: 10.1200/jco.23.00899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/17/2023] [Accepted: 09/06/2023] [Indexed: 11/24/2023] Open
Abstract
PURPOSE The outcome of older patients with mantle cell lymphoma (MCL) has improved by the introduction of immunochemotherapy, followed by rituximab (R)-maintenance. Assessment of minimal residual disease (MRD) represents a promising tool for individualized treatment decisions and was a prospectively planned part of the European MCL Elderly trial. We investigated how MRD status influenced the efficacy of R-maintenance and how MRD can enable tailored consolidation strategies. PATIENTS AND METHODS Previously untreated patients with MCL age 60 years or older have been randomly assigned to R versus interferon-alpha maintenance after response to rituximab, fludarabine, cyclophosphamide (R-FC) versus rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone (R-CHOP). MRD monitoring was performed by real-time quantitative polymerase chain reaction (qPCR) following EuroMRD guidelines. RESULTS A qPCR assay with a median sensitivity of 1 × 10-5 could be generated in 80% of 288 patients in an international, multicenter, multilaboratory setting. More extensive tumor dissemination facilitated the identification of a molecular marker. The efficacy of R-maintenance in clinical remission was confirmed for MRD-negative patients at the end of induction in terms of progression-free survival (PFS; hazard ratio [HR], 0.38 [95% CI, 0.21 to 0.63]) and overall survival (OS; HR, 0.37 [95% CI, 0.20 to 0.68]), particularly in R-CHOP-treated patients (PFS-HR, 0.23 [95% CI, 0.10 to 0.52]; OS-HR, 0.19 [95% CI, 0.07 to 0.52]). R-maintenance appeared less effective in MRD-positive patients (PFS-HR, 0.51 [95% CI, 0.26 to 1.02]) overall and after R-CHOP induction (PFS-HR, 0.59 [95% CI, 0.28 to 1.26]). R-FC achieved more frequent and faster MRD clearance compared with R-CHOP. MRD positivity in clinical remission after induction was associated with a short median time to clinical progression of approximately 1-1.7 years. CONCLUSION The results confirm the strong efficacy of R-maintenance in patients who are MRD-negative after induction. Treatment de-escalation for MRD-negative patients is discouraged by our results. More effective consolidation strategies should be explored in MRD-positive patients to improve their long-term prognosis.
Collapse
Affiliation(s)
- Eva Hoster
- Institute for Medical Informatics, Biometry, and Epidemiology (IBE), LMU Munich, Munich, Germany
- Department of Internal Medicine III, University Hospital LMU Munich, Munich, Germany
| | | | - Elizabeth Macintyre
- Laboratory of Onco-Hematology, Université Paris Cité, Institut Necker-Enfants Malades and Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Linmiao Jiang
- Institute for Medical Informatics, Biometry, and Epidemiology (IBE), LMU Munich, Munich, Germany
| | | | | | - Gilles Salles
- Hospices Civils de Lyon, Université Claude Bernard, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Hervé Tilly
- Department of Hematology and U1245, Centre Henri Becquerel, Rouen, France
| | - Stefan Wirths
- Department of Medicine II, University of Tübingen, Tübingen, Germany
| | - Pierre Feugier
- Department of Hematology and INSERM 1256, University of Lorraine, Vandoeuvre les Nancy, France
| | - Kai Hübel
- Klinik I für Innere Medizin, Universität zu Köln, Köln, Germany
| | - Christian Schmidt
- Department of Internal Medicine III, University Hospital LMU Munich, Munich, Germany
| | | | - Johanna C Kluin-Nelemans
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martin Dreyling
- Department of Internal Medicine III, University Hospital LMU Munich, Munich, Germany
| | - Christiane Pott
- Department of Medicine, University Hospital Schleswig-Holstein Campus Kiel/Christian-Albrechts University Kiel, Germany
| |
Collapse
|
36
|
Pott C, Jurinovic V, Trotman J, Kehden B, Unterhalt M, Herold M, Jagt RVD, Janssens A, Kneba M, Mayer J, Young M, Schmidt C, Knapp A, Nielsen T, Brown H, Spielewoy N, Harbron C, Bottos A, Mundt K, Marcus R, Hiddemann W, Hoster E. Minimal Residual Disease Status Predicts Outcome in Patients With Previously Untreated Follicular Lymphoma: A Prospective Analysis of the Phase III GALLIUM Study. J Clin Oncol 2024; 42:550-561. [PMID: 38096461 DOI: 10.1200/jco.23.00838] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/23/2023] [Accepted: 10/12/2023] [Indexed: 02/09/2024] Open
Abstract
PURPOSE We report an analysis of minimal residual/detectable disease (MRD) as a predictor of outcome in previously untreated patients with follicular lymphoma (FL) from the randomized, multicenter GALLIUM (ClinicalTrials.gov identifier: NCT01332968) trial. PATIENTS AND METHODS Patients received induction with obinutuzumab (G) or rituximab (R) plus bendamustine, or cyclophosphamide, doxorubicin, vincristine, prednisone (CHOP) or cyclophosphamide, vincristine, prednisone (CVP) chemotherapy, followed by maintenance with the same antibody in responders. MRD status was assessed at predefined time points (mid-induction [MI], end of induction [EOI], and at 4-6 monthly intervals during maintenance and follow-up). Patients with evaluable biomarker data at diagnosis were included in the survival analysis. RESULTS MRD positivity was associated with inferior progression-free survival (PFS) at MI (hazard ratio [HR], 3.03 [95% CI, 2.07 to 4.45]; P < .0001) and EOI (HR, 2.25 [95% CI, 1.53 to 3.32]; P < .0001). MRD response was higher after G- versus R-chemotherapy at MI (94.2% v 88.9%; P = .013) and at EOI (93.1% v 86.7%; P = .0077). Late responders (MI-positive/EOI-negative) had a significantly poorer PFS than early responders (MI-negative/EOI-negative; HR, 3.11 [95% CI, 1.75 to 5.52]; P = .00011). The smallest proportion of MRD positivity was observed in patients receiving bendamustine at MI (4.8% v 16.0% in those receiving CHOP; P < .0001). G appeared to compensate for less effective chemotherapy regimens, with similar MRD response rates observed across the G-chemo groups. During the maintenance period, more patients treated with R than with G were MRD-positive (R-CHOP, 20.7% v G-CHOP, 7.0%; R-CVP, 21.7% v G-CVP, 9.4%). Throughout maintenance, MRD positivity was associated with clinical relapse. CONCLUSION MRD status can determine outcome after induction and during maintenance, and MRD negativity is a prerequisite for long-term disease control in FL. The higher MRD responses after G- versus R-based treatment confirm more effective tumor cell clearance.
Collapse
Affiliation(s)
| | - Vindi Jurinovic
- Department of Internal Medicine III, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
- Institute for Medical Information Processing, Biometry, and Epidemiology, Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Judith Trotman
- Concord Repatriation General Hospital, University of Sydney, Sydney, NSW, Australia
| | - Britta Kehden
- University Hospital Schleswig-Holstein, Kiel, Germany
| | - Michael Unterhalt
- Department of Internal Medicine III, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | | | | | | | - Michael Kneba
- University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jiri Mayer
- University Hospital and Masaryk University, Brno, Czech Republic
| | - Moya Young
- East Kent Hospital, Canterbury, United Kingdom
| | - Christian Schmidt
- Department of Internal Medicine III, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | | | | | - Helen Brown
- Roche Products Ltd, Welwyn Garden City, United Kingdom
| | | | - Chris Harbron
- Roche Products Ltd, Welwyn Garden City, United Kingdom
| | | | | | | | - Wolfgang Hiddemann
- Department of Internal Medicine III, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Eva Hoster
- Department of Internal Medicine III, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
- Institute for Medical Information Processing, Biometry, and Epidemiology, Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
37
|
Jerkeman M, Kolstad A, Hutchings M, Pasanen A, Meriranta L, Niemann CU, Kragh Jørgensen RR, El-Galaly TC, Riise J, Leppä S, Christensen JH, Sonnevi K, Pedersen LB, Wader KF, Glimelius I. MRD-driven treatment with venetoclax-R2 in mantle cell lymphoma: the Nordic Lymphoma Group MCL7 VALERIA trial. Blood Adv 2024; 8:407-415. [PMID: 38113470 PMCID: PMC10827399 DOI: 10.1182/bloodadvances.2023011920] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023] Open
Abstract
ABSTRACT Despite improvements in treatment of mantle cell lymphoma (MCL), most patients eventually relapse. In this multicenter phase 1b/2 trial, we evaluated safety and efficacy of minimal residual disease (MRD)-driven venetoclax, lenalidomide, and rituximab (venetoclax-R2) in relapsed/refractory (R/R) MCL and explored the feasibility of stopping treatment in molecular remission. The primary end point was overall response rate (ORR) at 6 months. After dose escalation, the recommended phase 2 dose was lenalidomide 20 mg daily, days 1 to 21; venetoclax 600 mg daily after ramp-up; and rituximab 375 mg/m2 weekly for 4 weeks, then every 8 weeks. MRD monitoring by RQ-PCR was performed every 3 months. When MRD-negativity in the blood was reached, treatment was continued for another 3 months; if MRD-negativity was then confirmed, treatment was stopped. In total, 59 patients were enrolled, with a median age of 73 years. At 6 months, the ORR was 63% (29 complete remission [CR], 8 partial remission [PR]), and 40% (4 CR, 2 PR) for patients previously failing a Bruton tyrosine kinase (BTK) inhibitor. Median progression-free survival (PFS) was 21 months, with median overall survival of 31 months. TP53 mutation was associated with inferior PFS (P < .01). Overall, 28 patients (48%) discontinued treatment in molecular remission, and 25 remain MRD negative after a median of 17.4 months. Hematological toxicity was frequent, with 52 of 59 (88%) patients with G3-4 neutropenia and 21 of 59 (36%) patients with G3-4 thrombocytopenia. To conclude, MRD-driven venetoclax-R2 is feasible and tolerable and shows efficacy in R/R MCL, also after BTK inhibitor failure. This trial was registered at www.ClinicalTrials.gov as #NCT03505944.
Collapse
Affiliation(s)
- Mats Jerkeman
- Department of Oncology, Institute of Clinical Sciences, Lund University and Skane University Hospital SE22185, Lund, Sweden
| | - Arne Kolstad
- Department of Oncology, Innlandet Hospital, Brumunddal, Norway
| | | | - Annika Pasanen
- Department of Oncology, Helsinki University Hospital, Helsinki, Finland
| | - Leo Meriranta
- Department of Oncology, Helsinki University Hospital, Helsinki, Finland
| | | | - Rasmus Rask Kragh Jørgensen
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Tarec Christoffer El-Galaly
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Jon Riise
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Sirpa Leppä
- Department of Oncology, Helsinki University Hospital, Helsinki, Finland
| | | | - Kristina Sonnevi
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | | | - Karin Fahl Wader
- Department of Oncology, St Olav University Hospital, Trondheim, Norway
| | - Ingrid Glimelius
- Department of Immunology, Genetics and Pathology, Unit of Cancer Precision Medicine, Uppsala University, Uppsala, Sweden
| |
Collapse
|
38
|
Zaliova M, Zuna J, Winkowska L, Janotova I, Skorepova J, Lukes J, Meyer C, Marschalek R, Novak Z, Domansky J, Stary J, Sramkova L, Trka J. Genomic DNA-based measurable residual disease monitoring in pediatric acute myeloid leukemia: unselected consecutive cohort study. Leukemia 2024; 38:21-30. [PMID: 38001170 PMCID: PMC10776399 DOI: 10.1038/s41375-023-02083-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023]
Abstract
Measurable residual disease (MRD) monitoring in childhood acute myeloid leukemia (AML) is used to assess response to treatment and for early detection of imminent relapse. In childhood AML, MRD is typically evaluated using flow cytometry, or by quantitative detection of leukemia-specific aberrations at the mRNA level. Both methods, however, have significant limitations. Recently, we demonstrated the feasibility of MRD monitoring in selected subgroups of AML at the genomic DNA (gDNA) level. To evaluate the potential of gDNA-based MRD monitoring across all AML subtypes, we conducted a comprehensive analysis involving 133 consecutively diagnosed children. Integrating next-generation sequencing into the diagnostic process, we identified (presumed) primary genetic aberrations suitable as MRD targets in 97% of patients. We developed patient-specific quantification assays and monitored MRD in 122 children. The gDNA-based MRD monitoring via quantification of primary aberrations with a sensitivity of at least 10-4 was possible in 86% of patients; via quantification with sensitivity of 5 × 10-4, of secondary aberrations, or at the mRNA level in an additional 8%. Importantly, gDNA-based MRD exhibited independent prognostic value at early time-points in patients stratified to intermediate-/high-risk treatment arms. Our study demonstrates the broad applicability, feasibility, and clinical significance of gDNA-based MRD monitoring in childhood AML.
Collapse
Affiliation(s)
- Marketa Zaliova
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic.
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
- University Hospital Motol, Prague, Czech Republic.
| | - Jan Zuna
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Lucie Winkowska
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | | | - Justina Skorepova
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Julius Lukes
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Claus Meyer
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia (DCAL), Goethe-University, Frankfurt am Main, Germany
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia (DCAL), Goethe-University, Frankfurt am Main, Germany
| | - Zbynek Novak
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jiri Domansky
- Pediatric Oncology Department, University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Stary
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Lucie Sramkova
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Jan Trka
- CLIP (Childhood Leukaemia Investigation Prague), Prague, Czech Republic.
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
- University Hospital Motol, Prague, Czech Republic.
| |
Collapse
|
39
|
Menon H, Singh PK, Bagal B, Dolai T, Jain A, Chaudhri A. Minimal Residual Disease in the Management of B-Cell Acute Lymphoblastic Leukemia: A Systematic Review of Studies from Indian Settings. Indian J Hematol Blood Transfus 2024; 40:1-11. [PMID: 38312181 PMCID: PMC10831037 DOI: 10.1007/s12288-023-01641-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/06/2023] [Indexed: 04/07/2023] Open
Abstract
Minimal residual disease (MRD) has become an essential tool in the management of B-cell acute lymphoblastic leukemia (B-ALL) and aids in tailoring treatment strategies to suit specific patient needs. Although much progress has been made in this area, there is limited data on the use of MRD in the Indian context. Our objective was to identify relevant literature that discusses the utility of MRD in the management of B-cell ALL in adolescents and young adults (AYA) and adults in Indian settings. A systematic search and screening of articles were performed using the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. The primary data source was PubMed followed by Google Scholar for articles and conference proceedings. Of the 254 records screened, 24 records were retained for analysis. MRD monitoring had a significant role in the management of AYA/adult B-cell ALL patients. Variability of results was observed across these studies with respect to methods, techniques, and use. However, these studies evidenced and validated the importance of MRD assessment in risk-adapted management of B-cell ALL and highlighted the need for optimization. The advances in MRD diagnostics and applications are yet to be tested and adopted in Indian settings. Hence, there is a need for in-depth research to develop and optimize approaches for calibrating country-specific management strategies. The potential role of MRD assessments in anticipating relapse or treatment failures warrants more attention for the preemptive positioning of novel strategies involving immunotherapies.
Collapse
Affiliation(s)
- Hari Menon
- Hematology and Head Medical Oncology, St John’s National Academy of Health Sciences, Bangalore, Karnataka India
| | - Pawan Kumar Singh
- Hemato Oncology and Bone Marrow Transplant, BLK-Max Centre for Bone Marrow Transplant, Delhi, India
| | - Bhausaheb Bagal
- Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra India
| | - Tuphan Dolai
- Hematology Department, NRS Medical College and Hospital, Kolkata, West Bengal India
| | - Ankita Jain
- Oncology and Field Medical, Pfizer Oncology, Mumbai, Maharashtra India
| | | |
Collapse
|
40
|
Kim R, Bergugnat H, Pastoret C, Pasquier F, Raffoux E, Larcher L, Passet M, Grardel N, Delabesse E, Kubetzko S, Caye-Eude A, Meyer C, Marschalek R, Lafage-Pochitaloff M, Thiebaut-Bertrand A, Balsat M, Escoffre-Barbe M, Blum S, Baumann M, Banos A, Straetmans N, Gallego-Hernanz MP, Chalandon Y, Graux C, Soulier J, Leguay T, Hunault M, Huguet F, Lhéritier V, Dombret H, Boissel N, Clappier E. Genetic alterations and MRD refine risk assessment for KMT2A-rearranged B-cell precursor ALL in adults: a GRAALL study. Blood 2023; 142:1806-1817. [PMID: 37595275 DOI: 10.1182/blood.2023021501] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 08/20/2023] Open
Abstract
KMT2A-rearranged (KMT2A-r) B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is widely recognized as a high-risk leukemia in both children and adults. However, there is a paucity of data on adults treated in recent protocols, and the optimal treatment strategy for these patients is still a matter of debate. In this study, we set out to refine the prognosis of adult KMT2A-r BCP-ALL treated with modern chemotherapy regimen and investigate the prognostic impact of comutations and minimal residual disease (MRD). Of 1091 adult patients with Philadelphia-negative BCP-ALL enrolled in 3 consecutive trials from the Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL), 141 (12.9%) had KMT2A-r, with 5-year cumulative incidence of relapse (CIR) and overall survival (OS) rates of 40.7% and 53.3%, respectively. Molecular profiling highlighted a low mutational burden in this subtype, reminiscent of infant BCP-ALL. However, the presence of TP53 and/or IKZF1 alterations defined a subset of patients with significantly poorer CIR (69.3% vs 36.2%; P = .001) and OS (28.1% vs 60.7%; P = .006) rates. Next, we analyzed the prognostic implication of MRD measured after induction and first consolidation, using both immunoglobulin (IG) or T-cell receptor (TR) gene rearrangements and KMT2A genomic fusion as markers. In approximately one-third of patients, IG/TR rearrangements were absent or displayed clonal evolution during the disease course, compromising MRD monitoring. In contrast, KMT2A-based MRD was highly reliable and strongly associated with outcome, with early good responders having an excellent outcome (3-year CIR, 7.1%; OS, 92.9%). Altogether, our study reveals striking heterogeneity in outcomes within adults with KMT2A-r BCP-ALL and provides new biomarkers to guide risk-based therapeutic stratification.
Collapse
Affiliation(s)
- Rathana Kim
- Hematology Laboratory, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Hugo Bergugnat
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Cédric Pastoret
- Hematology Laboratory, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Florence Pasquier
- Department of Hematology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Emmanuel Raffoux
- Hematology Department, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Lise Larcher
- Hematology Laboratory, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Marie Passet
- Hematology Laboratory, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nathalie Grardel
- Hematology Laboratory, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Eric Delabesse
- Hematology Laboratory, Institut Universitaire de Cancer Toulouse-Oncopole, INSERM 1037, CNRS, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Susanne Kubetzko
- Department of Hematology, University Hospital of Zürich, Zürich, Switzerland
| | - Aurélie Caye-Eude
- Genetics Department, Molecular Genetics Unit, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| | - Claus Meyer
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia, Goethe University, Frankfurt/Main, Germany
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology/Diagnostic Center of Acute Leukemia, Goethe University, Frankfurt/Main, Germany
| | - Marine Lafage-Pochitaloff
- Laboratoire de Cytogénétique Hématologique, Hôpital Timone Enfant, Assistance Publique-Hôpitaux de Marseille, Aix-Marseille Université, Marseille, France
| | | | - Marie Balsat
- Department of Hematology, Hôpital Lyon Sud, Pierre Benite, France
| | | | - Sabine Blum
- Department of Hematology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Michael Baumann
- Klinik für Medizinische Onkologie und Hämatologie, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Anne Banos
- Department of Hematology, Centre Hospitalier de la Côte Basque, Bayonne, France
| | - Nicole Straetmans
- Department of Hematology, University Hospital Saint-Luc, Brussels, Belgium
| | | | - Yves Chalandon
- Division of Hematology, Department of Oncology, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland, for the Swiss Group for Clinical Cancer Research
- Swiss Group for Clinical Cancer Research
| | - Carlos Graux
- Department of Hematology, Université Catholique de Louvain, Centre Hospitalier Universitaire UCLouvain Namur-Godinne, Yvoir, Belgium
| | - Jean Soulier
- Hematology Laboratory, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Thibaut Leguay
- Department of Hematology, Centre Hospitalier Universitaire de Bordeaux, Hôpital du Haut-Levêque, Pessac, France
| | - Mathilde Hunault
- Département des Maladies du Sang, Centre Hospitalier Universitaire Angers, INSERM, CNRS, CRCI2NA, Fédération Hospitalo-Universitaire Grand Ouest Against Leukemia, Université d'Angers, Université de Nantes, Angers, France
| | - Françoise Huguet
- Department of Hematology, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire de Cancer Toulouse-Oncopole, Toulouse, France
| | - Véronique Lhéritier
- Coordination du Groupe Group for Research on Adult Acute Lymphoblastic Leukemia, Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre Benite, France
| | - Hervé Dombret
- Hematology Department, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Nicolas Boissel
- Hematology Department, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Emmanuelle Clappier
- Hematology Laboratory, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U944, CNRS UMR 7212 GenCellDis, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| |
Collapse
|
41
|
van der Linde R, Gatt PN, Smith S, Fernandez MA, Vaughan L, Blyth E, Curnow J, Brown DA, Tegg E, Sasson SC. Measurable Residual Disease (MRD) by Flow Cytometry in Adult B-Acute Lymphoblastic Leukaemia (B-ALL) and Acute Myeloid Leukaemia (AML): Correlation with Molecular MRD Testing and Clinical Outcome at One Year. Cancers (Basel) 2023; 15:5064. [PMID: 37894431 PMCID: PMC10605425 DOI: 10.3390/cancers15205064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Measurable residual disease (MRD) detected by flow cytometry (FC) is well established in paediatric B- lymphoblastic leukaemia (B-ALL) and adult chronic lymphocytic leukaemia (CLL), but its utility in adult B-ALL and adult acute myeloid leukaemia (AML) is less clear. In this prospective MRD study, one of the largest in Australia to date, we examined consecutive bone marrow aspirates from adult participants with B-ALL (n = 47) and AML (n = 87) sent for FC-MRD testing at a quaternary referral hospital in Sydney. FC-MRD results were correlated to corresponding Mol-MRD testing where available and clinical outcomes at three-month intervals over 1 year. B-ALL showed a moderate positive correlation (rs = 0.401, p < 0.001), while there was no correlation between FC-MRD and Mol-MRD for AML (rs = 0.13, p = 0.237). Five FC-MRD patterns were identified which had significant associations with relapse (X2(4) = 31.17(4), p > 0.001) and survival (X2(4) = 13.67, p = 0.008) in AML, but not in B-ALL. The three-month MRD results were also strongly associated with survival in AML, while the association in B-ALL was less evident. There was a moderate correlation between FC-MRD and Mol-MRD in B-ALL but not AML. The association of FC-MRD with relapse and survival was stronger in AML than in B-ALL. Overall, these findings suggest divergent utilities of FC-MRD in AML and B-ALL.
Collapse
Affiliation(s)
- Riana van der Linde
- Department of Laboratory Haematology, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (L.V.); (E.T.)
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
| | - Prudence N. Gatt
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
- Westmead Institute for Medical Research, University of Sydney, Sydney, NSW 2145, Australia
| | - Sandy Smith
- Flow Cytometry Unit, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (S.S.); (M.A.F.)
| | - Marian A. Fernandez
- Flow Cytometry Unit, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (S.S.); (M.A.F.)
| | - Lachlin Vaughan
- Department of Laboratory Haematology, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (L.V.); (E.T.)
- Department of Haematology, Western Sydney Local Health District, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Emily Blyth
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
- Westmead Institute for Medical Research, University of Sydney, Sydney, NSW 2145, Australia
- Department of Haematology, Western Sydney Local Health District, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Jennifer Curnow
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
- Department of Haematology, Western Sydney Local Health District, Westmead Hospital, Westmead, NSW 2145, Australia
| | - David A. Brown
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
- Westmead Institute for Medical Research, University of Sydney, Sydney, NSW 2145, Australia
- Department of Clinical Immunology and Immunopathology, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Elizabeth Tegg
- Department of Laboratory Haematology, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia; (L.V.); (E.T.)
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
| | - Sarah C. Sasson
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia; (P.N.G.); (E.B.); (J.C.); (D.A.B.); (S.C.S.)
- Department of Clinical Immunology and Immunopathology, Institute of Clinical Pathology and Medical Research, NSW Health Pathology, Westmead Hospital, Westmead, NSW 2145, Australia
| |
Collapse
|
42
|
Liao H, Jiang N, Yang Y, Zhang X, Chen J, Lai H, Zheng Q. Association of Minimal Residual Disease by a Single-Tube 8-Color Flow Cytometric Analysis With Clinical Outcome in Adult B-Cell Acute Lymphoblastic Leukemia: A Real-World Study Based on 486 Patients. Arch Pathol Lab Med 2023; 147:1186-1195. [PMID: 36508349 DOI: 10.5858/arpa.2022-0172-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2022] [Indexed: 09/29/2023]
Abstract
CONTEXT.— Minimal/measurable residual disease (MRD) measured by molecular and multiparametric flow cytometry (MFC) has been proven to be predictive of relapse and survival in patients with B-cell acute lymphoblastic leukemia (B-ALL). A universally applicable antibody panel at a low cost but without compromising sensitivity and power of prognosis prediction in adult B-ALL remains unestablished. OBJECTIVE.— To report our experience of using a single-tube 8-color MFC panel to measure the MRD status as a prognostic indicator in adult B-ALL patients. DESIGN.— We retrospectively analyzed the characteristics, MRD status, and prognosis of adult B-ALL based on a large real-world cohort of 486 patients during a 10-year period. RESULTS.— MRD assessed by MFC and polymerase chain reaction (PCR) assays for BCR-ABL+ patients showed concordant results in 74.2% of cases. MRD- status by our MFC panel could clearly predict a favorable relapse-free survival (RFS) and overall survival (OS) both at the end of induction and at the end of 1 consolidation course. Patients with continuous MRD- and with at least 1 MRD- result showed a favorable RFS and OS compared with those with at least 1 MRD+ result and continuous MRD+, respectively. CONCLUSIONS.— The single-tube 8-color MFC panel demonstrated a low cost, decent sensitivity, and comparability with polymerase chain reaction-MRD but an excellent performance in predicting RFS and OS, and thus could potentially be taken as a routine indicator in the evaluation of the treatment response for adult patients with B-ALL.
Collapse
Affiliation(s)
- Hongyan Liao
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Nenggang Jiang
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Ying Yang
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Xin Zhang
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Jiao Chen
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Hongli Lai
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Qin Zheng
- From the Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Jia MZ, Li WJ, Wang CJ, Zhang Q, Gao C, Huang XT, Zhu T, Zhang RD, Cui L, Li ZG. Tracing back of relapse clones by Ig/TCR gene rearrangements reveals complex patterns of recurrence in pediatric acute lymphoblastic leukemia. Int J Lab Hematol 2023; 45:717-725. [PMID: 37194559 DOI: 10.1111/ijlh.14100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/04/2023] [Indexed: 05/18/2023]
Abstract
INTRODUCTION Relapse remained the major obstacle to improving the prognosis of children with acute lymphoblastic leukemia (ALL). This study aimed to investigate the changing patterns of Ig/TCR gene rearrangements between diagnosis and relapse and the clinical relevance and to explore the mechanism of leukemic relapse. METHODS Clonal Ig/TCR gene rearrangements were screened by multiplex PCR amplification in 85 paired diagnostic and relapse bone marrow (BM) samples from children with ALL. The new rearrangements presented at relapse were quantitatively assessed by the RQ-PCR approach targeting the patient-specific junctional region sequence in 19 diagnostic samples. The relapse clones were further back-traced to diagnostic and follow-up BM samples from 12 patients. RESULTS Comparison of Ig/TCR gene rearrangements between diagnosis and relapse showed that 40 (57.1%) B-ALL and 5 (33.3%) T-ALL patients exhibited a change from diagnosis to relapse, and 25 (35.7%) B-ALL patients acquired new rearrangements at relapse. The new relapse rearrangements were present in 15 of the 19 (78.9%) diagnostic samples as shown by RQ-PCR, with a median level of 5.26 × 10-2 . The levels of minor rearrangements correlated with B immunophenotype, WBC counts, age at diagnosis, and recurrence time. Furthermore, back-tracing rearrangements in 12 patients identified three patterns of relapse clone dynamics, which suggested the recurrence mechanisms not only through clonal selection of pre-existing subclones but also through an ongoing clonal evolution during remission and relapse. CONCLUSION Backtracking Ig/TCR gene rearrangements in relapse clones of pediatric ALL revealed complex patterns of clonal selection and evolution for leukemic relapse.
Collapse
Affiliation(s)
- Ming-Zhu Jia
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Wei-Jing Li
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Chan-Juan Wang
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China
| | - Qing Zhang
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Chao Gao
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Xiao-Tong Huang
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Ting Zhu
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Rui-Dong Zhang
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China
| | - Lei Cui
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Zhi-Gang Li
- Hematologic Diseases Laboratory, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Pediatric Hematology-Oncology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
- Hematology Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| |
Collapse
|
44
|
Burmeister T, Gröger D, Gökbuget N, Spriewald B, Starck M, Elmaagacli A, Hoelzer D, Keller U, Schwartz S. Molecular characterization of TCF3::PBX1 chromosomal breakpoints in acute lymphoblastic leukemia and their use for measurable residual disease assessment. Sci Rep 2023; 13:15167. [PMID: 37704696 PMCID: PMC10499895 DOI: 10.1038/s41598-023-42294-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023] Open
Abstract
The translocation t(1;19)(q23;p13) with the resulting chimeric TCF3::PBX1 gene is the third most prevalent recurrent chromosomal translocation in acute lymphoblastic leukemia and accounts for 3-5% of cases. The molecular background of this translocation has been incompletely studied, especially in adult cases. We characterized the chromosomal breakpoints of 49 patients with TCF3::PBX1 and the corresponding reciprocal PBX1::TCF3 breakpoints in 15 cases at the molecular level, thus providing an extensive molecular overview of this translocation in a well-defined study patient population. Breakpoints were found to be remarkably clustered not only in TCF3 but also in PBX1. No association with DNA repeats or putative cryptic recombination signal sequence sites was observed. A simplified detection method for breakpoint identification was developed and the feasibility of patient-specific chromosomal break sites as molecular markers for detecting measurable residual disease (MRD) was explored. A highly sensitive generic real-time PCR for MRD assessment using these breakpoint sequences was established that could serve as a useful alternative to the classical method utilizing rearranged immune gene loci. This study provides the first extensive molecular data set on the chromosomal breakpoints of the t(1;19)/TCF3::PBX1 aberration in adult ALL. Based on the obtained data a generic MRD method was developed that has several theoretical advantages, including an on average higher sensitivity and a greater stability of the molecular marker in the course of disease.
Collapse
Affiliation(s)
- Thomas Burmeister
- Department of Hematology, Oncology and Tumor Immunology, CVK, Charité - Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Daniela Gröger
- Department of Hematology, Oncology and Tumor Immunology, CBF, Charité - Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nicola Gökbuget
- Medical Department 2, Goethe-Universität, Frankfurt, Germany
| | - Bernd Spriewald
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Michael Starck
- I. Medical Department, München Klinik Schwabing, Munich, Germany
| | - Ahmet Elmaagacli
- Department of Hematology, Oncology, Asklepios Klinik St. Georg, Hamburg, Germany
| | - Dieter Hoelzer
- Medical Department 2, Goethe-Universität, Frankfurt, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology and Tumor Immunology, CBF, Charité - Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan Schwartz
- Department of Hematology, Oncology and Tumor Immunology, CBF, Charité - Universitätsmedizin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
45
|
Kelm M, Darzentas F, Darzentas N, Kotrova M, Wessels W, Bendig S, Baldus CD, Lettau M, Gökbuget N, Kabelitz D, Brüggemann M, Chitadze G. Dominant T-cell Receptor Delta Rearrangements in B-cell Precursor Acute Lymphoblastic Leukemia: Leukemic Markers or Physiological γδ T Repertoire? Hemasphere 2023; 7:e948. [PMID: 37670805 PMCID: PMC10476800 DOI: 10.1097/hs9.0000000000000948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/20/2023] [Indexed: 09/07/2023] Open
Affiliation(s)
- Miriam Kelm
- Medical Department II, Hematology and Oncology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Germany
- University Cancer Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Kiel and Lübeck, Germany
| | - Franziska Darzentas
- Medical Department II, Hematology and Oncology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Germany
- University Cancer Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Kiel and Lübeck, Germany
| | - Nikos Darzentas
- Medical Department II, Hematology and Oncology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Germany
- University Cancer Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Kiel and Lübeck, Germany
| | - Michaela Kotrova
- Medical Department II, Hematology and Oncology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Germany
- University Cancer Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Kiel and Lübeck, Germany
| | - Wiebke Wessels
- Medical Department II, Hematology and Oncology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Germany
- University Cancer Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Kiel and Lübeck, Germany
| | - Sonja Bendig
- Medical Department II, Hematology and Oncology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Germany
- University Cancer Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Kiel and Lübeck, Germany
| | - Claudia D. Baldus
- Medical Department II, Hematology and Oncology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Germany
- University Cancer Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Kiel and Lübeck, Germany
| | - Marcus Lettau
- Medical Department II, Hematology and Oncology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Germany
- Institute of Immunology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Germany
| | - Nicola Gökbuget
- Department of Medicine II, Goethe University Hospital, Frankfurt, Germany
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Germany
| | - Monika Brüggemann
- Medical Department II, Hematology and Oncology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Germany
- University Cancer Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Kiel and Lübeck, Germany
| | - Guranda Chitadze
- Medical Department II, Hematology and Oncology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Germany
- University Cancer Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Kiel and Lübeck, Germany
| |
Collapse
|
46
|
Medina-Herrera A, Sarasquete ME, Jiménez C, Puig N, García-Sanz R. Minimal Residual Disease in Multiple Myeloma: Past, Present, and Future. Cancers (Basel) 2023; 15:3687. [PMID: 37509348 PMCID: PMC10377959 DOI: 10.3390/cancers15143687] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Responses to treatment have improved over the last decades for patients with multiple myeloma. This is a consequence of the introduction of new drugs that have been successfully combined in different clinical contexts: newly diagnosed, transplant-eligible or ineligible patients, as well as in the relapsed/refractory setting. However, a great proportion of patients continue to relapse, even those achieving complete response, which underlines the need for updated response criteria. In 2014, the international myeloma working group established new levels of response, prompting the evaluation of minimal residual disease (MRD) for those patients already in complete or stringent complete response as defined by conventional serological assessments: the absence of tumor plasma cells in 100,000 total cells or more define molecular and immunophenotypic responses by next-generation sequencing and flow cytometry, respectively. In this review, we describe all the potential methods that may be used for MRD detection based on the evidence found in the literature, paying special attention to their advantages and pitfalls from a critical perspective.
Collapse
Affiliation(s)
- Alejandro Medina-Herrera
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - María Eugenia Sarasquete
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Cristina Jiménez
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Noemí Puig
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Ramón García-Sanz
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| |
Collapse
|
47
|
Yu Z, Xie L, Zhang J, Lin H, Niu T. The evolution of minimal residual disease: key insights based on a bibliometric visualization analysis from 2002 to 2022. Front Oncol 2023; 13:1186198. [PMID: 37534257 PMCID: PMC10391156 DOI: 10.3389/fonc.2023.1186198] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/15/2023] [Indexed: 08/04/2023] Open
Abstract
Background The topic of minimal residual disease (MRD) has emerged as a crucial subject matter in the domain of oncology in recent years. The detection and monitoring of MRD have become essential for the diagnosis, treatment, and prognosis of various types of malignancy. Aims The purpose of this study is to explore the research trends, hotspots, and frontiers of MRD in the last two decades through bibliometric analysis. Methods We employed Web of Science databases to carry out a bibliometric visualization analysis of research on 8,913 academic papers about MRD research from 2002 to 2022. VOSviewer, CiteSpace, RStudio, and a bibliometric online analysis platform were mainly used to conduct co-occurrence analysis and cooperative relationship analysis of countries/regions, institutions, journals, and authors in the literature. Furthermore, co-occurrence, co-citation, and burst analyses of keyword and reference were also conducted to generate relevant knowledge maps. Results In the past 20 years, the number of MRD research papers has presented an overall rising trend, going through three stages: a plateau, development, and an explosion. The output of articles in the United States was notably superior and plays a dominant role in this field, and the Netherlands had the highest average citation per article. The most productive and influential institution was the University of Texas MD Anderson Cancer Center. Blood published the most papers and was the most cited journal. A collection of leading academics has come to the fore in the research field, the most prolific of which is Kantarjian HM. It was found that the application of MRD in "acute myeloid leukemia", "acute lymphoblastic leukemia", "multiple myeloma", as well as the detection technology of MRD, are the research hotspots and frontiers in this domain. Furthermore, we analyzed the co-citation network of references and found that the top 10 co-cited references were all associated with MRD in hematological malignancies. Conclusion This bibliometric visualization analysis conducted a thorough exploration into the research hotspots and trends in MRD from 2002 to 2022. Our findings can aid researchers in recognizing possible collaborations, guiding future research directions, and fostering the growth of MRD detection and monitoring technologies.
Collapse
Affiliation(s)
- Zhengyu Yu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Li Xie
- State Key Laboratory of Wildlife Quarantine and Surveillance (Sichuan), Technology Center of Chengdu Customs, Chengdu, China
| | - Jing Zhang
- State Key Laboratory of Wildlife Quarantine and Surveillance (Sichuan), Technology Center of Chengdu Customs, Chengdu, China
| | - Hua Lin
- State Key Laboratory of Wildlife Quarantine and Surveillance (Sichuan), Technology Center of Chengdu Customs, Chengdu, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
48
|
Fuchs S, Danßmann C, Klironomos F, Winkler A, Fallmann J, Kruetzfeldt LM, Szymansky A, Naderi J, Bernhart SH, Grunewald L, Helmsauer K, Rodriguez-Fos E, Kirchner M, Mertins P, Astrahantseff K, Suenkel C, Toedling J, Meggetto F, Remke M, Stadler PF, Hundsdoerfer P, Deubzer HE, Künkele A, Lang P, Fuchs J, Henssen AG, Eggert A, Rajewsky N, Hertwig F, Schulte JH. Defining the landscape of circular RNAs in neuroblastoma unveils a global suppressive function of MYCN. Nat Commun 2023; 14:3936. [PMID: 37402719 DOI: 10.1038/s41467-023-38747-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 05/12/2023] [Indexed: 07/06/2023] Open
Abstract
Circular RNAs (circRNAs) are a regulatory RNA class. While cancer-driving functions have been identified for single circRNAs, how they modulate gene expression in cancer is not well understood. We investigate circRNA expression in the pediatric malignancy, neuroblastoma, through deep whole-transcriptome sequencing in 104 primary neuroblastomas covering all risk groups. We demonstrate that MYCN amplification, which defines a subset of high-risk cases, causes globally suppressed circRNA biogenesis directly dependent on the DHX9 RNA helicase. We detect similar mechanisms in shaping circRNA expression in the pediatric cancer medulloblastoma implying a general MYCN effect. Comparisons to other cancers identify 25 circRNAs that are specifically upregulated in neuroblastoma, including circARID1A. Transcribed from the ARID1A tumor suppressor gene, circARID1A promotes cell growth and survival, mediated by direct interaction with the KHSRP RNA-binding protein. Our study highlights the importance of MYCN regulating circRNAs in cancer and identifies molecular mechanisms, which explain their contribution to neuroblastoma pathogenesis.
Collapse
Affiliation(s)
- Steffen Fuchs
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
- The German Cancer Consortium (DKTK), Partner Site Berlin, 10117, Berlin, Germany.
- The German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10178, Berlin, Germany.
- CRCT, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, 31037, Toulouse, France.
- Laboratoire d'Excellence Toulouse Cancer-TOUCAN, 31037, Toulouse, France.
| | - Clara Danßmann
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Filippos Klironomos
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Annika Winkler
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Jörg Fallmann
- Bioinformatics Group, Department of Computer Science, and Interdisciplinary Center for Bioinformatics, University of Leipzig, 04107, Leipzig, Germany
| | - Louisa-Marie Kruetzfeldt
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Annabell Szymansky
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Julian Naderi
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195, Berlin, Germany
| | - Stephan H Bernhart
- Bioinformatics Group, Department of Computer Science, and Interdisciplinary Center for Bioinformatics, University of Leipzig, 04107, Leipzig, Germany
| | - Laura Grunewald
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, 10117, Berlin, Germany
- The German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Konstantin Helmsauer
- Experimental and Clinical Research Center (ECRC) of the Charité and Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125, Berlin, Germany
| | - Elias Rodriguez-Fos
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Experimental and Clinical Research Center (ECRC) of the Charité and Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125, Berlin, Germany
| | - Marieluise Kirchner
- Core Unit Proteomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), 13125, Berlin, Germany
| | - Philipp Mertins
- Core Unit Proteomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), 13125, Berlin, Germany
| | - Kathy Astrahantseff
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Christin Suenkel
- Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Straße 28, 10115, Berlin, Germany
- Lonza Drug Product Services, 4057, Basel, Switzerland
| | - Joern Toedling
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, 10117, Berlin, Germany
- The German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Fabienne Meggetto
- CRCT, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, 31037, Toulouse, France
- Laboratoire d'Excellence Toulouse Cancer-TOUCAN, 31037, Toulouse, France
| | - Marc Remke
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Heinrich Heine University Düsseldorf, Medical Faculty, and University Hospital Düsseldorf, 40225, Düsseldorf, Germany
- The German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, 40225, Düsseldorf, Germany
- Institute of Neuropathology, Heinrich Heine University Düsseldorf, Medical Faculty, and University Hospital Düsseldorf, 40225, Düsseldorf, Germany
| | - Peter F Stadler
- Bioinformatics Group, Department of Computer Science, and Interdisciplinary Center for Bioinformatics, University of Leipzig, 04107, Leipzig, Germany
| | - Patrick Hundsdoerfer
- Department of Pediatric Oncology, Helios Klinikum Berlin-Buch, 13125, Berlin, Germany
| | - Hedwig E Deubzer
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, 10117, Berlin, Germany
- The German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10178, Berlin, Germany
- Experimental and Clinical Research Center (ECRC) of the Charité and Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125, Berlin, Germany
| | - Annette Künkele
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, 10117, Berlin, Germany
- The German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Peter Lang
- Department I - General Pediatrics, Hematology/Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Jörg Fuchs
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany
| | - Anton G Henssen
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, 10117, Berlin, Germany
- The German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- Experimental and Clinical Research Center (ECRC) of the Charité and Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, 10117, Berlin, Germany
- The German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10178, Berlin, Germany
| | - Nikolaus Rajewsky
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Systems Biology of Gene Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Straße 28, 10115, Berlin, Germany
| | - Falk Hertwig
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- The German Cancer Consortium (DKTK), Partner Site Berlin, 10117, Berlin, Germany
- The German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Johannes H Schulte
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
- The German Cancer Consortium (DKTK), Partner Site Berlin, 10117, Berlin, Germany.
- The German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10178, Berlin, Germany.
- Department I - General Pediatrics, Hematology/Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, 72076, Tuebingen, Germany.
| |
Collapse
|
49
|
Ariffin H, Chiew EKH, Oh BLZ, Lee SHR, Lim EH, Kham SKY, Abdullah WA, Chan LL, Foo KM, Lam JCM, Chan YH, Lin HP, Quah TC, Tan AM, Yeoh AEJ. Anthracycline-Free Protocol for Favorable-Risk Childhood ALL: A Noninferiority Comparison Between Malaysia-Singapore ALL 2003 and ALL 2010 Studies. J Clin Oncol 2023:JCO2202347. [PMID: 37276496 DOI: 10.1200/jco.22.02347] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 03/10/2023] [Accepted: 04/17/2023] [Indexed: 06/07/2023] Open
Abstract
PURPOSE To investigate whether, for children with favorable-risk B-cell precursor ALL (BCP-ALL), an anthracycline-free protocol is noninferior to a modified Berlin-Frankfurt-Muenster ALL-IC2002 protocol, which includes 120 mg/m2 of anthracyclines. PATIENTS AND METHODS Three hundred sixty-nine children with favorable-risk BCP-ALL (age 1-9 years, no extramedullary disease, and no high-risk genetics) who cleared minimal residual disease (≤0.01%) at the end of remission induction were enrolled into Ma-Spore (MS) ALL trials. One hundred sixty-seven standard-risk (SR) patients (34% of Malaysia-Singapore ALL 2003 study [MS2003]) were treated with the MS2003-SR protocol and received 120 mg/m2 of anthracyclines during delayed intensification while 202 patients (42% of MS2010) received an anthracycline-free successor protocol. The primary outcome was a noninferiority margin of 1.15 in 6-year event-free survival (EFS) between the MS2003-SR and MS2010-SR cohorts. RESULTS The 6-year EFS of MS2003-SR and MS2010-SR (anthracycline-free) cohorts was 95.2% ± 1.7% and 96.5% ± 1.5%, respectively (P = .46). The corresponding 6-year overall survival was 97.6% and 99.0% ± 0.7% (P = .81), respectively. The cumulative incidence of relapse was 3.6% and 2.6%, respectively (P = .42). After adjustment for race, sex, age, presenting WBC, day 8 prednisolone response, and favorable genetic subgroups, the hazard ratio for MS2010-SR EFS was 0.98 (95% CI, 0.84 to 1.14; P = .79), confirming noninferiority. Compared with MS2003-SR, MS2010-SR had significantly lower episodes of bacteremia (30% v 45.6%; P = .04) and intensive care unit admissions (1.5% v 9.5%; P = .004). CONCLUSION In comparison with MS2003-SR, the anthracycline-free MS2010-SR protocol is not inferior and was less toxic as treatment for favorable-risk childhood BCP-ALL.
Collapse
Affiliation(s)
| | | | - Bernice Ling Zhi Oh
- National University of Singapore, Singapore, Singapore
- National University Hospital, Singapore, Singapore
| | - Shawn Hsien Ren Lee
- National University of Singapore, Singapore, Singapore
- National University Hospital, Singapore, Singapore
| | | | | | | | - Lee Lee Chan
- Subang Jaya Medical Centre, Subang Jaya, Malaysia
| | - Koon Mian Foo
- KK Women's and Children's Hospital, Singapore, Singapore
| | | | | | - Hai Peng Lin
- Subang Jaya Medical Centre, Subang Jaya, Malaysia
| | - Thuan Chong Quah
- National University of Singapore, Singapore, Singapore
- National University Hospital, Singapore, Singapore
| | - Ah Moy Tan
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Allen Eng Juh Yeoh
- National University of Singapore, Singapore, Singapore
- National University Hospital, Singapore, Singapore
| |
Collapse
|
50
|
Metzner B, Müller TH, Casper J, Kimmich C, Köhne CH, Petershofen E, Renzelmann A, Thole R, Voss A, Dreyling M, Hoster E, Klapper W, Pott C. Long-term outcome in patients with mantle cell lymphoma following high-dose therapy and autologous stem cell transplantation. Eur J Haematol 2023. [PMID: 37094812 DOI: 10.1111/ejh.13985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/26/2023]
Abstract
BACKGROUND Long-term clinical and molecular remissions in patients with mantle cell lymphoma (MCL) after autologous stem cell transplantation (ASCT) have been evaluated in only a few studies. DESIGN AND METHODS Sixty-five patients with MCL received ASCT (54 first-line ASCT, 10 second-line ASCT, and 1 third-line ASCT). In the case of long-term remission (≥5 years; n = 27), peripheral blood was tested for minimal residual disease (MRD) by t(11;14)- and IGH-PCR at the last follow-up. RESULTS Ten-year overall survival (OS), progression-free survival (PFS), and freedom from progression (FFP) after first-line ASCT were 64%, 52%, and 59% versus after second-line ASCT 50%, 20%, and 20%, respectively. Five-year OS, PFS, and FFP for the first-line cohort were 79%, 63%, and 69%, respectively. Five-year OS, PFS, and FFP after second-line ASCT were 60%, 30%, and 30%, respectively. Treatment-related mortality (3 months after ASCT) was 1.5%. So far 26 patients developed sustained long-term clinical and molecular complete remissions of up to 19 years following ASCT in first treatment line. CONCLUSION Sustained long-term clinical and molecular remissions are achievable following ASCT.
Collapse
Affiliation(s)
- Bernd Metzner
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Thomas H Müller
- Red Cross Blood Transfusion Service NSTOB, Oldenburg, Germany
| | - Jochen Casper
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Christoph Kimmich
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Claus-Henning Köhne
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | | | - Andrea Renzelmann
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Ruth Thole
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Andreas Voss
- Department of Oncology and Hematology, Klinikum Oldenburg, University Clinic, Oldenburg, Germany
| | - Martin Dreyling
- Department of Internal Medicine III, University Hospital Munich, Grosshadern, Germany
| | - Eva Hoster
- Department of Internal Medicine III, University Hospital Munich, Grosshadern, Germany
| | - Wolfram Klapper
- Department of Pathology, Hematopathology Section and Lymph Node Registry, University of Schleswig-Holstein, Kiel, Germany
| | - Christiane Pott
- Department of Medicine II, University of Schleswig-Holstein, Kiel, Germany
| |
Collapse
|