1
|
Sutanto H, Pratiwi L, Romadhon PZ, Bintoro SUY. Advancing chronic myeloid leukemia research with next-generation sequencing: potential benefits, limitations, and future clinical integration. Hum Genet 2025; 144:481-503. [PMID: 40257486 DOI: 10.1007/s00439-025-02745-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025]
Abstract
Next-generation sequencing (NGS) has emerged as a powerful tool for advancing research in chronic myeloid leukemia (CML) by providing a deeper understanding of its genetic complexity. Beyond detecting the hallmark BCR::ABL1 fusion gene, NGS has enabled the identification of additional mutations associated with disease progression, therapy resistance, and clonal evolution. NGS also facilitates the detection of rare BCR::ABL1 fusion variants and cryptic rearrangements, offering a more refined genetic characterization of the disease. Additionally, it enhances the study of minimal residual disease (MRD) and evolving resistance patterns, which are crucial for developing targeted therapeutic strategies. However, challenges such as data interpretation, standardization, and cost constraints continue to limit the widespread application of NGS in routine research and clinical settings. This review explores the contributions of NGS to CML research, highlighting its role in uncovering novel genetic alterations, tracking clonal evolution, and identifying potential therapeutic targets. As sequencing technologies evolve, NGS is expected to further shape the future of CML research, providing critical insights that may ultimately refine disease management strategies.
Collapse
Affiliation(s)
- Henry Sutanto
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.
| | - Laras Pratiwi
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Pradana Zaky Romadhon
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Universitas Airlangga Hospital, Surabaya, Indonesia
| | - Siprianus Ugroseno Yudho Bintoro
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
| |
Collapse
|
2
|
Branford S, Apperley JF. Measurable residual disease in chronic myeloid leukemia. Haematologica 2022; 107:2794-2809. [PMID: 36453517 PMCID: PMC9713565 DOI: 10.3324/haematol.2022.281493] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Chronic myeloid leukemia is characterized by a single genetic abnormality resulting in a fusion gene whose mRNA product is easily detected and quantified by reverse-transcriptase polymerase chain reaction analysis. Measuring residual disease was originally introduced to identify patients relapsing after allogeneic stem cell transplantation but rapidly adopted to quantify responses to tyrosine kinase inhibitors. Real-time quantitative polymerase chain reaction is now an essential tool for the management of patients and is used to influence treatment decisions. In this review we track this development including the international collaboration to standardize results, discuss the integration of molecular monitoring with other factors that affect patients' management, and describe emerging technology. Four case histories describe varying scenarios in which the accurate measurement of residual disease identified patients at risk of disease progression and allowed appropriate investigations and timely clinical intervention.
Collapse
Affiliation(s)
- Susan Branford
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia,School of Medicine, University of Adelaide, Adelaide, Australia,Clinical and Health Sciences, University of South Australia, Adelaide, Australia,S. Branford
| | - Jane F. Apperley
- Department of Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK,Centre for Haematology, Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
3
|
Chen X, Song Q, Zhang B, Gao Y, Lou K, Liu Y, Wen W. A Rapid Digital PCR System with a Pressurized Thermal Cycler. MICROMACHINES 2021; 12:mi12121562. [PMID: 34945412 PMCID: PMC8708658 DOI: 10.3390/mi12121562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 12/15/2022]
Abstract
We designed a silicon-based fast-generated static droplets array (SDA) chip and developed a rapid digital polymerase chain reaction (dPCR) detection platform that is easy to load samples for fluorescence monitoring. By using the direct scraping method for sample loading, a droplet array of 2704 microwells with each volume of about 0.785 nL can be easily realized. It was determined that the sample loading time was less than 10 s with very simple and efficient characteristics. In this platform, a pressurized thermal cycling device was first used to solve the evaporation problem usually encountered for dPCR experiments, which is critical to ensuring the successful amplification of templates at the nanoliter scale. We used a gradient dilution of the hepatitis B virus (HBV) plasmid as the target DNA for a dPCR reaction to test the feasibility of the dPCR chip. Our experimental results demonstrated that the dPCR chip could be used to quantitatively detect DNA molecules. Furthermore, the platform can measure the fluorescence intensity in real-time. To test the accuracy of the digital PCR system, we chose three-channel silicon-based chips to operate real-time fluorescent PCR experiments on this platform.
Collapse
Affiliation(s)
- Xuee Chen
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong; (X.C.); (Q.S.)
| | - Qi Song
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong; (X.C.); (Q.S.)
- Guangzhou HKUST Fok Ying Tung Research Institute, Guangzhou 511458, China
| | - Beini Zhang
- Advanced Materials Thrust, Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong;
| | - Yibo Gao
- Zhuhai Shineway Biotech Co., Ltd., Zhuhai 519000, China;
| | - Kai Lou
- Guangzhou Kayja-Optics Technology Co., Ltd., Guangzhou 511458, China;
| | - Yiteng Liu
- Earth, Ocean and Atmospheric Sciences Thrust, Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong;
| | - Weijia Wen
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong; (X.C.); (Q.S.)
- Advanced Materials Thrust, Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong;
- Correspondence: ; Tel.: +852-2358-7979
| |
Collapse
|
4
|
Mughal TI, Psaila B, DeAngelo DJ, Saglio G, Van Etten RA, Radich JP. Interrogating the molecular genetics of chronic myeloproliferative malignancies for personalized management in 2021. Haematologica 2021; 106:1787-1793. [PMID: 33657787 PMCID: PMC8252942 DOI: 10.3324/haematol.2020.267252] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/13/2021] [Indexed: 12/15/2022] Open
Affiliation(s)
- Tariq I Mughal
- Tufts University Medical Center, Boston, MA, USA; University of Buckingham Medical School, Buckingham.
| | - Bethan Psaila
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford
| | | | | | | | | |
Collapse
|
5
|
Gutiérrez LG, Abelleyro MM, Ruiz MS, Anchordoqui MS, Freitas J, Bianchini M, De Brasi CD, Larripa IB. Development of an inverse-PCR approach for characterization of the major BCR-ABL1 breakpoint sequences on genomic DNA: proof of concept. Clin Chem Lab Med 2021; 59:e449-e453. [PMID: 34162038 DOI: 10.1515/cclm-2020-1482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 06/09/2021] [Indexed: 11/15/2022]
Affiliation(s)
- Leandro G Gutiérrez
- Instituto de Medicina Experimental (IMEX), CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Miguel M Abelleyro
- Instituto de Medicina Experimental (IMEX), CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - María Sol Ruiz
- Centro de Investigaciones Oncológicas-Fundación Cáncer (CIO-FUCA), Buenos Aires, Argentina
| | | | - Josefina Freitas
- Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Michele Bianchini
- Centro de Investigaciones Oncológicas-Fundación Cáncer (CIO-FUCA), Buenos Aires, Argentina
| | - Carlos D De Brasi
- Instituto de Medicina Experimental (IMEX), CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina.,Instituto de Investigaciones Hematológicas (IIHEMA), Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Irene B Larripa
- Instituto de Medicina Experimental (IMEX), CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| |
Collapse
|
6
|
Huggett JF. The Digital MIQE Guidelines Update: Minimum Information for Publication of Quantitative Digital PCR Experiments for 2020. Clin Chem 2021; 66:1012-1029. [PMID: 32746458 DOI: 10.1093/clinchem/hvaa125] [Citation(s) in RCA: 267] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/18/2020] [Indexed: 12/17/2022]
Abstract
Digital PCR (dPCR) has developed considerably since the publication of the Minimum Information for Publication of Digital PCR Experiments (dMIQE) guidelines in 2013, with advances in instrumentation, software, applications, and our understanding of its technological potential. Yet these developments also have associated challenges; data analysis steps, including threshold setting, can be difficult and preanalytical steps required to purify, concentrate, and modify nucleic acids can lead to measurement error. To assist independent corroboration of conclusions, comprehensive disclosure of all relevant experimental details is required. To support the community and reflect the growing use of dPCR, we present an update to dMIQE, dMIQE2020, including a simplified dMIQE table format to assist researchers in providing key experimental information and understanding of the associated experimental process. Adoption of dMIQE2020 by the scientific community will assist in standardizing experimental protocols, maximize efficient utilization of resources, and further enhance the impact of this powerful technology.
Collapse
|
7
|
Gliddon HD, Kaforou M, Alikian M, Habgood-Coote D, Zhou C, Oni T, Anderson ST, Brent AJ, Crampin AC, Eley B, Heyderman R, Kern F, Langford PR, Ottenhoff THM, Hibberd ML, French N, Wright VJ, Dockrell HM, Coin LJ, Wilkinson RJ, Levin M. Identification of Reduced Host Transcriptomic Signatures for Tuberculosis Disease and Digital PCR-Based Validation and Quantification. Front Immunol 2021; 12:637164. [PMID: 33763081 PMCID: PMC7982854 DOI: 10.3389/fimmu.2021.637164] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/03/2021] [Indexed: 12/18/2022] Open
Abstract
Recently, host whole blood gene expression signatures have been identified for diagnosis of tuberculosis (TB). Absolute quantification of the concentrations of signature transcripts in blood have not been reported, but would facilitate diagnostic test development. To identify minimal transcript signatures, we applied a transcript selection procedure to microarray data from African adults comprising 536 patients with TB, other diseases (OD) and latent TB (LTBI), divided into training and test sets. Signatures were further investigated using reverse transcriptase (RT)-digital PCR (dPCR). A four-transcript signature (GBP6, TMCC1, PRDM1, and ARG1) measured using RT-dPCR distinguished TB patients from those with OD (area under the curve (AUC) 93.8% (CI95% 82.2-100%). A three-transcript signature (FCGR1A, ZNF296, and C1QB) differentiated TB from LTBI (AUC 97.3%, CI95%: 93.3-100%), regardless of HIV. These signatures have been validated across platforms and across samples offering strong, quantitative support for their use as diagnostic biomarkers for TB.
Collapse
Affiliation(s)
- Harriet D Gliddon
- Section of Paediatrics, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom.,National Public Health Speciality Training Programme, South West, United Kingdom
| | - Myrsini Kaforou
- Section of Paediatrics, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Mary Alikian
- Imperial Molecular Pathology, Imperial Healthcare Trust, Hammersmith Hospital, London, United Kingdom.,Centre for Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Dominic Habgood-Coote
- Section of Paediatrics, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Chenxi Zhou
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Tolu Oni
- School of Public Health and Family Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Suzanne T Anderson
- Brighton and Sussex Medical School, Brighton, United Kingdom.,Brighton and Malawi Liverpool Wellcome Trust Unit, Blantyre, Malawi
| | - Andrew J Brent
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Amelia C Crampin
- Malawi Epidemiology and Intervention Research Unit, Chilumba, Malawi.,London School of Hygiene & Tropical Medicine, London, United Kingdom.,Karonga Prevention Study, Chilumba, Malawi
| | - Brian Eley
- Paediatric Infectious Diseases Unit, Red Cross War Memorial Children's Hospital, Cape Town, South Africa.,Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Robert Heyderman
- Division of Infection and Immunity, Faculty of Medical Sciences, University College London, London, United Kingdom
| | - Florian Kern
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom.,Brighton and Sussex University Hospitals National Health Service (NHS) Trust, Brighton, United Kingdom
| | - Paul R Langford
- Section of Paediatrics, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Martin L Hibberd
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Neil French
- Tropical and Infectious Disease Unit, Royal Liverpool and Broadgreen University Hospitals National Health Service (NHS) Trust, Liverpool, United Kingdom.,Centre for Global Vaccine Research, Institute of Infection & Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Victoria J Wright
- Section of Paediatrics, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Hazel M Dockrell
- Department of Immunology and Infection, and Tuberculosis (TB) Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Lachlan J Coin
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Robert J Wilkinson
- The Francis Crick Institute, London, United Kingdom.,Department of Medicine, Imperial College London, London, United Kingdom.,Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Michael Levin
- Section of Paediatrics, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
8
|
Branford S. Why is it critical to achieve a deep molecular response in chronic myeloid leukemia? Haematologica 2020; 105:2730-2737. [PMID: 33054104 PMCID: PMC7716360 DOI: 10.3324/haematol.2019.240739] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The primary goal of tyrosine kinase inhibitor (TKI) therapy for patients with chronic myeloid leukemia is survival, which is achieved by the vast majority of patients. The initial response to therapy provides a sensitive measure of future clinical outcome. Measurement of BCR-ABL1 transcript levels using real-time quantitative polymerase chain reaction standardized to the international reporting scale is now the principal recommended monitoring strategy. The method is used to assess early milestone responses and provides a guide for therapeutic intervention. When patients successfully traverse the critical first 12 months of TKI therapy, most will head towards another milestone response, deep molecular response (DMR, BCR-ABL1 ≤0.01%). DMR is essential for patients aiming to achieve treatment-free remission and a prerequisite for a trial of TKI discontinuation. The success of discontinuation trials has led to new treatment strategies in order for more patients to reach this milestone response. DMR has been incorporated into endpoints of clinical trials and is considered by some expert groups as the optimal treatment response. But is DMR a stable response and does it provide the ultimate protection against TKI resistance and death? Do we need to increase the sensitivity of detection of BCR-ABL1 to better identify the patients who would likely remain in treatment-free remission after TKI discontinuation? Is it necessary to switch current TKI therapy to a more potent inhibitor if the goal is to achieve DMR? These are issues that I will explore in this review.
Collapse
Affiliation(s)
- Susan Branford
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology; School of Pharmacy and Medical Science, Division of Health Sciences, University of South Australia; School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide and School of Biological Sciences, University of Adelaide, Adelaide.
| |
Collapse
|
9
|
Kockerols CC, Valk PJ, Levin MD, Pallisgaard N, Cornelissen JJ, Westerweel PE. Digital PCR for BCR-ABL1 Quantification in CML: Current Applications in Clinical Practice. Hemasphere 2020; 4:e496. [PMID: 33283168 PMCID: PMC7710259 DOI: 10.1097/hs9.0000000000000496] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023] Open
Abstract
Molecular monitoring of the BCR-ABL1 transcript for patients with chronic phase chronic myeloid leukemia (CML) has become increasingly demanding. Real-time quantitative PCR (qPCR) is the routinely used method, but has limitations in quantification accuracy due to its inherent technical variation. Treatment recommendations rely on specific BCR-ABL1 values set at timed response milestones, making precise measurement of BCR-ABL1 a requisite. Furthermore, the sensitivity of qPCR may be insufficient to reliably quantify low levels of residual BCR-ABL1 in patients in deep molecular response (DMR) who could qualify for an attempt to discontinue Tyrosine Kinase Inhibitor (TKI) therapy. We reviewed the current use of digital PCR (dPCR) as a promising alternative for response monitoring in CML. dPCR offers an absolute BCR-ABL1 quantification at various disease levels with remarkable precision and a clinical sensitivity reaching down to at least MR5.0. Moreover, dPCR has been validated in multiple studies as prognostic marker for successful TKI treatment discontinuation, while this could not be achieved using classical qPCR. dPCR may thus prospectively be the preferred method to reliably identify patients achieving treatment milestones after initiation of TKI therapy as well as for the selection and timing for TKI discontinuation.
Collapse
Affiliation(s)
| | - Peter J.M. Valk
- Department of Molecular Biology and Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mark-David Levin
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | | | - Jan J. Cornelissen
- Department of Molecular Biology and Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Peter E. Westerweel
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| |
Collapse
|
10
|
Kim IS. Minimal residual disease in acute lymphoblastic leukemia: technical aspects and implications for clinical interpretation. Blood Res 2020; 55:S19-S26. [PMID: 32719172 PMCID: PMC7386891 DOI: 10.5045/br.2020.s004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 12/16/2022] Open
Abstract
Minimal residual disease (MRD) monitoring has proven to be one of the fundamental independent prognostic factors for patients with acute lymphoblastic leukemia (ALL). Sequential monitoring of MRD using sensitive and specific methods, such as real-time quantitative polymerase chain reaction (qPCR) or flow cytometry (FCM), has improved the assessment of treatment response and is currently used for therapeutic stratification and early detection. Although both FCM and qPCR yield highly consistent results with sensitivities of 10‒4, each method has several limitations. For example, qPCR is time-consuming and laborious: designing primers that correspond to the immunoglobulin (IG) and T-cell receptor (TCR) gene rearrangements at diagnosis can take 3‒4 weeks. In addition, the evolution of additional clones beyond the first or index clone during therapy cannot be detected, which might lead to false-negative results. FCM requires experienced technicians and sometimes does not achieve a sensitivity of 10‒4. Accordingly, a next generation sequencing (NGS)-based method has been developed in an attempt to overcome these limitations. With the advent of high-throughput NGS technologies, a more in-depth analysis of IG and/or TCR gene rearrangements is now within reach, which impacts all applications of IG/TR analysis. However, standardization, quality control, and validation of this new technology are warranted prior to its incorporation into routine practice.
Collapse
Affiliation(s)
- In-Suk Kim
- Department of Laboratory Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| |
Collapse
|
11
|
Cumbo C, Anelli L, Specchia G, Albano F. Monitoring of Minimal Residual Disease (MRD) in Chronic Myeloid Leukemia: Recent Advances. Cancer Manag Res 2020; 12:3175-3189. [PMID: 32440215 PMCID: PMC7211966 DOI: 10.2147/cmar.s232752] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/23/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm caused by the BCR-ABL1 fusion gene generation as a consequence of the t(9;22)(q34;q11) rearrangement. The identification of the BCR-ABL1 transcript was of critical importance for both CML diagnosis and minimal residual disease (MRD) monitoring. In this review, we report the recent advances in the CML MRD monitoring based on RNA, DNA and protein analysis. The detection of the BCR-ABL1 transcript by the quantitative reverse-transcriptase polymerase chain reaction is the gold standard method, but other systems based on digital PCR or on GeneXpert technology have been developed. In the last years, DNA-based assays showed high sensitivity and specificity, and flow cytometric approaches for the detection of the BCR-ABL1 fusion protein have also been tested. Recently, new MRD monitoring systems based on the detection of molecular markers other than the BCR-ABL1 fusion were proposed. These approaches, such as the identification of CD26+ leukemic stem cells, microRNAs and mitochondrial DNA mutations, just remain preliminary and need to be implemented. In the precision medicine era, the constant improvement of the CML MRD monitoring practice could allow clinicians to choose the best therapeutic algorithm and a more accurate selection of CML patients eligible for the tyrosine kinase inhibitors discontinuation.
Collapse
Affiliation(s)
- Cosimo Cumbo
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, Bari 70124, Italy
| | - Luisa Anelli
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, Bari 70124, Italy
| | - Giorgina Specchia
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, Bari 70124, Italy
| | - Francesco Albano
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, Bari 70124, Italy
| |
Collapse
|
12
|
Saxena D, Sheikh S, Kao G, Binder ZA, Alonso-Basanta M, O'Rourke DM, Nasrallah MP, Dorsey JF. Rapid and ultrasensitive digital PCR (dPCR) profiling of EGFRvIII in tumor cells and tissues. Neurooncol Adv 2019; 1:vdz030. [PMID: 31807732 PMCID: PMC6881905 DOI: 10.1093/noajnl/vdz030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Amplification of the epidermal growth factor receptor (EGFR) gene is commonly found in glioblastoma (GBM). About 57% GBM overexpresses EGFR and are associated with tumor progression, poor prognosis, and shorter life expectancy. Molecular profiling of solid tumors usually takes several weeks and may be biased by intrinsic tumor heterogeneity. Methods The unique sequence created by the fusion of exon 1 and exon 8 in EGFRvIII was used to guide the design of primers and a Minor Groove Binder (MGB) probe. Extracted total RNA was reverse transcribed and pre-amplified by PCR, followed by detection of the EGFRvIII mutation by dPCR. Results The lowest limit of quantification of our EGFRvIII assay was 0.003%. The EGFRvIII variant was identified in patient-derived glioma neurosphere cell lines, xenograft mouse model, and patient-derived tumor specimens. The overall workflow can be accomplished within 24 hours. In certain samples, EGFRvIII was detected when next-generation sequencing was unable to identify the variant. This finding highlights the ability of the dPCR assay to identify EGFRvIII mutations in heterogeneous solid tumors such as GBM in a rapid fashion by profiling samples from spatially distinct areas of tumors from the same patient. Conclusions In this study, we developed a highly sensitive digital PCR (dPCR) platform and leveraged our assay to detect the variant III alteration of EGFR (EGFRvIII) and amplified EGFR in patient-derived glioma neurosphere cell lines, orthotopic xenograft GBM mouse models, and patient-derived tumor specimens in less than 24 hours from minute quantities of starting material.
Collapse
Affiliation(s)
- Deeksha Saxena
- Department of Radiation Oncology.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Abramson Cancer Center Glioblastoma Translational Center of Excellence, Penn Medicine, Philadelphia, PA
| | | | - Gary Kao
- Department of Radiation Oncology
| | - Zev A Binder
- Department of Neurosurgery.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Abramson Cancer Center Glioblastoma Translational Center of Excellence, Penn Medicine, Philadelphia, PA
| | | | - Donald M O'Rourke
- Department of Neurosurgery.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Abramson Cancer Center Glioblastoma Translational Center of Excellence, Penn Medicine, Philadelphia, PA
| | - MacLean P Nasrallah
- Department of Pathology and Laboratory Medicine, Division of Neuropathology.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Abramson Cancer Center Glioblastoma Translational Center of Excellence, Penn Medicine, Philadelphia, PA
| | - Jay F Dorsey
- Department of Radiation Oncology.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Abramson Cancer Center Glioblastoma Translational Center of Excellence, Penn Medicine, Philadelphia, PA
| |
Collapse
|
13
|
Pagani IS, Dang P, Saunders VA, Grose R, Shanmuganathan N, Kok CH, Carne L, Rwodzi Z, Watts S, McLean J, Braley J, Altamura H, Yeung DT, Branford S, Yong ASM, White DL, Hughes TP, Ross DM. Lineage of measurable residual disease in patients with chronic myeloid leukemia in treatment-free remission. Leukemia 2019; 34:1052-1061. [PMID: 31768016 DOI: 10.1038/s41375-019-0647-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/14/2019] [Accepted: 11/03/2019] [Indexed: 11/09/2022]
Abstract
Approximately half of patients with chronic myeloid leukemia (CML) in sustained deep molecular response who discontinue tyrosine kinase inhibitors (TKIs) remain in treatment-free remission (TFR). Some of these patients have measurable residual disease (MRD) by BCR-ABL1 mRNA testing, and most have detectable BCR-ABL1 DNA by highly sensitive methods. We used fluorescence-activated cell sorting and BCR-ABL1 DNA PCR to investigate the lineage of residual CML cells in TFR. Twenty patients in TFR for >1 year provided blood for sorting into granulocytes, monocytes, B cells, T cells, and NK cells. MRD was identified predominantly in the lymphoid compartment and never in granulocytes. B cells were more often BCR-ABL1 positive than T cells (18 vs 11/20 patients) and at higher levels (median 10-4.9 vs 10-5.7; P = 0.014). In 13 CML patients studied at diagnosis lymphocytes expressing BCR-ABL1 mRNA comprised a small proportion of total leukocytes. These data improve our understanding of TFR biology, since it is now clear that MRD in the blood of TFR patients need not imply the persistence of multipotent CML cells. Lineage-specific assessment of MRD could be explored as a means to improve the prediction of TFR.
Collapse
Affiliation(s)
- Ilaria S Pagani
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia.,School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Australasian Leukaemia and Lymphoma Group, Melbourne, VIC, Australia
| | - Phuong Dang
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - Verity A Saunders
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - Randall Grose
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - Naranie Shanmuganathan
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia.,School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Australasian Leukaemia and Lymphoma Group, Melbourne, VIC, Australia.,Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital, Adelaide, SA, Australia.,Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia.,Genetic and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Chung H Kok
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia.,School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Lisa Carne
- Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Zandy Rwodzi
- Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Sophie Watts
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - Jennifer McLean
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - Jodi Braley
- Genetic and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Haley Altamura
- Genetic and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - David T Yeung
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia.,School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Australasian Leukaemia and Lymphoma Group, Melbourne, VIC, Australia.,Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Susan Branford
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia.,Genetic and Molecular Pathology, SA Pathology, Adelaide, SA, Australia.,School of Biological Sciences, Faculty of Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Agnes S M Yong
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia.,School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Australasian Leukaemia and Lymphoma Group, Melbourne, VIC, Australia
| | - Deborah L White
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia.,School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Australasian Leukaemia and Lymphoma Group, Melbourne, VIC, Australia.,Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia.,School of Biological Sciences, Faculty of Sciences, University of Adelaide, Adelaide, SA, Australia.,School of Paediatrics, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Timothy P Hughes
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia.,School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Australasian Leukaemia and Lymphoma Group, Melbourne, VIC, Australia.,Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - David M Ross
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia. .,School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia. .,Australasian Leukaemia and Lymphoma Group, Melbourne, VIC, Australia. .,Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital, Adelaide, SA, Australia. .,Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia. .,Department of Haematology and Genetic Pathology, Flinders University and Medical Centre, Adelaide, SA, Australia.
| |
Collapse
|
14
|
Krumbholz M, Goerlitz K, Albert C, Lawlor J, Suttorp M, Metzler M. Large amplicon droplet digital PCR for DNA-based monitoring of pediatric chronic myeloid leukaemia. J Cell Mol Med 2019; 23:4955-4961. [PMID: 31199062 PMCID: PMC6653534 DOI: 10.1111/jcmm.14321] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 02/27/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022] Open
Abstract
Quantification of tumour‐specific molecular markers at the RNA and DNA level for treatment response monitoring is crucial for risk‐adapted stratification and guidance of individualized therapy in leukaemia and other malignancies. Most pediatric leukaemias and solid tumours of mesenchymal origin are characterized by a relatively low mutation burden at the single nucleotide level and the presence of recurrent chromosomal translocations. The genomic fusion sites resulting from translocations are stable molecular tumour markers; however, repeat‐rich DNA sequences flanking intronic breakpoints limit the design of high sensitivity PCR assays for minimal residual disease (MRD) monitoring. Here, we quantitatively evaluated the impact of repeat elements on assay selection and the feasibility of using extended amplicons (≤1330 bp) amplified by droplet digital PCR to monitor pediatric chronic myeloid leukaemia (CML). Molecular characterization of 178 genomic BCR‐ABL1 fusion sites showed that 64% were located within sequence repeat elements, impeding optimal primer/probe design. Comparative quantification of DNA and RNA BCR‐ABL1 copy numbers in 687 specimens from 55 pediatric patients revealed that their levels were highly correlated. The combination of droplet digital PCR, double quenched probes and extended amplicons represents a valuable tool for sensitive MRD assessment in CML and may be adapted to other translocation‐positive tumours.
Collapse
Affiliation(s)
- Manuela Krumbholz
- Department of Pediatrics, University Hospital Erlangen, Erlangen, Germany
| | - Katharina Goerlitz
- Department of Pediatrics, University Hospital Erlangen, Erlangen, Germany
| | - Christian Albert
- Department of Pediatrics, University Hospital Erlangen, Erlangen, Germany
| | - Jennifer Lawlor
- Department of Pediatrics, University Hospital Erlangen, Erlangen, Germany.,Department of Biology, Division of Genetics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Meinolf Suttorp
- Medical Faculty, Pediatric Hemato-Oncology, Technical University, Dresden, Germany
| | - Markus Metzler
- Department of Pediatrics, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
15
|
Pagani IS, Dang P, Kommers IO, Goyne JM, Nicola M, Saunders VA, Braley J, White DL, Yeung DT, Branford S, Hughes TP, Ross DM. BCR-ABL1 genomic DNA PCR response kinetics during first-line imatinib treatment of chronic myeloid leukemia. Haematologica 2018; 103:2026-2032. [PMID: 29976745 PMCID: PMC6269287 DOI: 10.3324/haematol.2018.189787] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/04/2018] [Indexed: 12/16/2022] Open
Abstract
Accurate quantification of minimal residual disease (MRD) during treatment of chronic myeloid leukemia (CML) guides clinical decisions. The conventional MRD method, RQ-PCR for BCR-ABL1 mRNA, reflects a composite of the number of circulating leukemic cells and the BCR-ABL1 transcripts per cell. BCR-ABL1 genomic DNA only reflects leukemic cell number. We used both methods in parallel to determine the relative contribution of the leukemic cell number to molecular response. BCR-ABL1 DNA PCR and RQ-PCR were monitored up to 24 months in 516 paired samples from 59 newly-diagnosed patients treated with first-line imatinib in the TIDEL-II study. In the first three months of treatment, BCR-ABL1 mRNA values declined more rapidly than DNA. By six months, the two measures aligned closely. The expression of BCR-ABL1 mRNA was normalized to cell number to generate an expression ratio. The expression of e13a2 BCR-ABL1 was lower than that of e14a2 transcripts at multiple time points during treatment. BCR-ABL1 DNA was quantifiable in 48% of samples with undetectable BCR-ABL1 mRNA, resulting in MRD being quantifiable for an additional 5-18 months (median 12 months). These parallel studies show for the first time that the rapid decline in BCR-ABL1 mRNA over the first three months of treatment is due to a reduction in both cell number and transcript level per cell, whereas beyond three months, falling levels of BCR-ABL1 mRNA are proportional to the depletion of leukemic cells.
Collapse
Affiliation(s)
- Ilaria S Pagani
- Cancer Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Australia
| | - Phuong Dang
- Cancer Theme, South Australian Health & Medical Research Institute, Adelaide, Australia
| | - Ivar O Kommers
- VU University Medical Center, Amsterdam, the Netherlands
| | - Jarrad M Goyne
- Cancer Theme, South Australian Health & Medical Research Institute, Adelaide, Australia
| | - Mario Nicola
- Genetic and Molecular Pathology, SA Pathology, Adelaide, Australia
| | - Verity A Saunders
- Cancer Theme, South Australian Health & Medical Research Institute, Adelaide, Australia
| | - Jodi Braley
- Genetic and Molecular Pathology, SA Pathology, Adelaide, Australia
| | - Deborah L White
- Cancer Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Australia.,School of Biological Sciences, Faculty of Sciences, University of Adelaide, Australia.,School of Paediatrics, Faculty of Health Sciences, University of Adelaide, Australia.,Health Sciences UniSA, Adelaide, Australia
| | - David T Yeung
- Cancer Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Australia.,Australasian Leukaemia and Lymphoma Group, Melbourne, Australia.,Department of Haematology, Royal Adelaide Hospital and SA Pathology, Australia
| | - Susan Branford
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Australia.,Genetic and Molecular Pathology, SA Pathology, Adelaide, Australia.,School of Paediatrics, Faculty of Health Sciences, University of Adelaide, Australia.,Centre for Cancer Biology, School of Pharmacy and Medical Science, University of South Australia, Adelaide, Australia
| | - Timothy P Hughes
- Cancer Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Australia.,Australasian Leukaemia and Lymphoma Group, Melbourne, Australia.,Department of Haematology, Royal Adelaide Hospital and SA Pathology, Australia.,Centre for Cancer Biology, School of Pharmacy and Medical Science, University of South Australia, Adelaide, Australia
| | - David M Ross
- Cancer Theme, South Australian Health & Medical Research Institute, Adelaide, Australia .,School of Medicine, Faculty of Health Sciences, University of Adelaide, Australia.,Australasian Leukaemia and Lymphoma Group, Melbourne, Australia.,Department of Haematology, Royal Adelaide Hospital and SA Pathology, Australia.,Centre for Cancer Biology, School of Pharmacy and Medical Science, University of South Australia, Adelaide, Australia.,Flinders University and Medical Centre, Adelaide, Australia
| |
Collapse
|
16
|
Cumbo C, Impera L, Minervini CF, Orsini P, Anelli L, Zagaria A, Coccaro N, Tota G, Minervini A, Casieri P, Brunetti C, Rossi AR, Parciante E, Specchia G, Albano F. Genomic BCR-ABL1 breakpoint characterization by a multi-strategy approach for "personalized monitoring" of residual disease in chronic myeloid leukemia patients. Oncotarget 2018. [PMID: 29541390 PMCID: PMC5834283 DOI: 10.18632/oncotarget.23971] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
For monitoring minimal residual disease (MRD) in chronic myeloid leukemia (CML) the most recommended method is quantitative RT-PCR (RT-qPCR) for measuring BCR-ABL1 transcripts. Several studies reported that a DNA-based assay enhances the sensitivity of detection of the BCR-ABL1 genomic rearrangement, even if its characterization results difficult. We developed a DNA-based method for detecting and quantifying residual BCR-ABL1 positive leukemic stem cells in CML patients. We propose two alternative approaches: the first one is a fluorescence in situ hybridization (FISH)-based step followed by Sanger sequencing; the second one employs MinION, a single molecule sequencer based on nanopore technology. Finally, after defining the BCR-ABL1 genomic junction, we performed the target CML patient–specific quantification, using droplet digital PCR (ddPCR). FISH and MinION steps, respectively, together with ddPCR analysis, greatly reduce the complexity that has impeded the use of “personalized monitoring” of CML in clinical practice. Our report suggests a feasible pipeline, in terms of costs and reproducibility, aimed at characterizing and quantifying the genomic BCR-ABL1 rearrangement during MRD monitoring in CML patients.
Collapse
Affiliation(s)
- Cosimo Cumbo
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| | - Luciana Impera
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| | | | - Paola Orsini
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| | - Luisa Anelli
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| | - Antonella Zagaria
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| | - Nicoletta Coccaro
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| | - Giuseppina Tota
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| | - Angela Minervini
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| | - Paola Casieri
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| | - Claudia Brunetti
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| | - Antonella Russo Rossi
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| | - Elisa Parciante
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| | - Giorgina Specchia
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| | - Francesco Albano
- Department of Emergency and Organ Transplantation, Hematology Section, University of Bari, 70124 Bari, Italy
| |
Collapse
|
17
|
Bai Y, Orfao A, Chim CS. Molecular detection of minimal residual disease in multiple myeloma. Br J Haematol 2017; 181:11-26. [PMID: 29265356 DOI: 10.1111/bjh.15075] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Despite the significantly higher complete remission rates and improved survival achieved in the last decade, multiple myeloma (MM) patients continue to relapse due to persistence of minimal residual disease (MRD). Generally, MRD refers to persistence of low levels of disease in the order of one tumour cell in ≥105 normal cells. Currently, molecular and immunophenotypic techniques are employed for MRD detection. This review focuses on MRD detection by molecular techniques, including allele-specific oligonucleotide polymerase chain reaction (ASO-PCR), next-generation sequencing (NGS) and digital PCR (dPCR), in addition to a brief description of, and comparison with, multiparameter flow cytometry. The basic principles, technical advantages and limitations, and the clinical impact of all three molecular techniques are reviewed and compared. They all have a sensitivity of at least 10-5 , among which ASO real-time quantitative PCR is the most well-standardized, and NGS carries the highest sensitivity and applicability, while dPCR is still under investigation. Furthermore, molecular MRD negativity is a favourable prognostic factor for survival of patients with MM. However, several challenges inherent to molecular detection of MRD still remain to be overcome, particularly false negativity and failure to detect extramedullary disease. Finally, detection of MRD from peripheral blood remains challenging.
Collapse
Affiliation(s)
- Yinlei Bai
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Spain
| | - Alberto Orfao
- Department of Medicine, University of Salamanca and University Hospital of Salamanca-IBSAL, Salamanca, Spain
| | - Chor Sang Chim
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Spain
| |
Collapse
|
18
|
High-throughput sequencing for noninvasive disease detection in hematologic malignancies. Blood 2017; 130:440-452. [PMID: 28600337 DOI: 10.1182/blood-2017-03-735639] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/25/2017] [Indexed: 12/20/2022] Open
Abstract
Noninvasive monitoring of minimal residual disease (MRD) has led to significant advances in personalized management of patients with hematologic malignancies. Improved therapeutic options and prolonged survival have further increased the need for sensitive tumor assessment that can inform treatment decisions and patient outcomes. At diagnosis or relapse of most hematologic neoplasms, malignant cells are often easily accessible in the blood as circulating tumor cells (CTCs), making them ideal targets to noninvasively profile the molecular features of each patient. In other cancer types, CTCs are generally rare and noninvasive molecular detection relies on circulating tumor DNA (ctDNA) shed from tumor deposits into circulation. The ability to precisely detect and quantify CTCs and ctDNA could minimize invasive procedures and improve prediction of clinical outcomes. Technical advances in MRD detection methods in recent years have led to reduced costs and increased sensitivity, specificity, and applicability. Among currently available tests, high-throughput sequencing (HTS)-based approaches are increasingly attractive for noninvasive molecular testing. HTS-based methods can simultaneously identify multiple genetic markers with high sensitivity and specificity without individual optimization. In this review, we present an overview of techniques used for noninvasive molecular disease detection in selected myeloid and lymphoid neoplasms, with a focus on the current and future role of HTS-based assays.
Collapse
|
19
|
Monitoring of childhood ALL using BCR-ABL1 genomic breakpoints identifies a subgroup with CML-like biology. Blood 2017; 129:2771-2781. [PMID: 28331056 DOI: 10.1182/blood-2016-11-749978] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 03/17/2017] [Indexed: 12/20/2022] Open
Abstract
We used the genomic breakpoint between BCR and ABL1 genes for the DNA-based monitoring of minimal residual disease (MRD) in 48 patients with childhood acute lymphoblastic leukemia (ALL). Comparing the results with standard MRD monitoring based on immunoglobulin/T-cell receptor (Ig/TCR) gene rearrangements and with quantification of IKZF1 deletion, we observed very good correlation for the methods in a majority of patients; however, >20% of children (25% [8/32] with minor and 12.5% [1/8] with major-BCR-ABL1 variants in the consecutive cohorts) had significantly (>1 log) higher levels of BCR-ABL1 fusion than Ig/TCR rearrangements and/or IKZF1 deletion. We performed cell sorting of the diagnostic material and assessed the frequency of BCR-ABL1-positive cells in various hematopoietic subpopulations; 12% to 83% of non-ALL B lymphocytes, T cells, and/or myeloid cells harbored the BCR-ABL1 fusion in patients with discrepant MRD results. The multilineage involvement of the BCR-ABL1-positive clone demonstrates that in some patients diagnosed with BCR-ABL1-positive ALL, a multipotent hematopoietic progenitor is affected by the BCR-ABL1 fusion. These patients have BCR-ABL1-positive clonal hematopoiesis resembling a chronic myeloid leukemia (CML)-like disease manifesting in "lymphoid blast crisis." The biological heterogeneity of BCR-ABL1-positive ALL may impact the patient outcomes and optimal treatment (early stem cell transplantation vs long-term administration of tyrosine-kinase inhibitors) as well as on MRD testing. Therefore, we recommend further investigations on CML-like BCR-ABL1-positive ALL.
Collapse
|
20
|
Alikian M, Gale RP, Apperley JF, Foroni L. Molecular techniques for the personalised management of patients with chronic myeloid leukaemia. BIOMOLECULAR DETECTION AND QUANTIFICATION 2017; 11:4-20. [PMID: 28331814 PMCID: PMC5348117 DOI: 10.1016/j.bdq.2017.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 12/28/2016] [Accepted: 01/18/2017] [Indexed: 12/25/2022]
Abstract
Chronic myeloid leukemia (CML) is the paradigm for targeted cancer therapy. RT-qPCR is the gold standard for monitoring response to tyrosine kinase-inhibitor (TKI) therapy based on the reduction of blood or bone marrow BCR-ABL1. Some patients with CML and very low or undetectable levels of BCR-ABL1 transcripts can stop TKI-therapy without CML recurrence. However, about 60 percent of patients discontinuing TKI-therapy have rapid leukaemia recurrence. This has increased the need for more sensitive and specific techniques to measure residual CML cells. The clinical challenge is to determine when it is safe to stop TKI-therapy. In this review we describe and critically evaluate the current state of CML clinical management, different technologies used to monitor measurable residual disease (MRD) focus on comparingRT-qPCR and new methods entering clinical practice. We discuss advantages and disadvantages of new methods.
Collapse
Key Words
- ABL1, Abelson murine leukaemia virus
- ALL, acute lymphoblastic leukaemia
- AP, accelerated phase
- ARQ, armored RNA Quant
- ATP, adenosine triphosphate
- BC, blast crisis
- BCR, breakpoint cluster region
- BM, bone marrow
- BMT, bone marrow transplantation
- Bp, base pair
- CAP, College of American Pathology
- CES, capillary electrophoresis sequencing
- CML
- CML, chronic myeloid leukaemia
- CMR, complete molecular response/remission
- CP, chronic phase
- DESTINY, De-Escalation and Stopping Treatment of Imatinib, Nilotinib or sprYcel in Chronic Myeloid Leukaemia
- DNA, deoxyribonucleic acid
- EAC, Europe Against Cancer
- ELN, European Leukaemia Net
- EURO-SKI, European Stop Tyrosine Kinase Inhibitor Study
- GUSB, glucuronidase beta gene
- IC, inhibotory concentration
- IRIS, interferon and cytarabine versus STI571
- IS, International Scale
- InDels, insertions and deletions
- KDa, Kilo Dalton
- Kbp, Kilo Base Pairs
- LPC, leukemic progenitor cells
- LSC, leukemic stem cell
- LoD, limit of detection
- LoQ, limit of quantification
- M-bcr, major-breakpoint cluster region
- MMR, major molecular response/remission
- MR, deep molecular response/remission
- MRD
- MRD, minimal residual disease
- Mbp, mega base pair
- Molecular monitoring
- NCCN, National Comprehensive Cancer Network
- NEQAS, National External Quality Assessement Service
- NGS
- NGS, next generation sequencing
- NTC, No Template Control
- PB, Peripheral Blood
- PCR, Polymerase Chain Reaction
- PFS, Progression Free Survival
- Ph, Philadelpia
- Q-PCR, quantitative polymerase chain reaction
- QC, Quality Control
- RT, reverse transcription
- RT-dPCR, reverse transcription-digital polymerase chain reaction
- RT-qPCR, reverse transcription-quantitative polymerase chain reaction
- SCT, stem cell transplant
- SMRT, single-molecule real-time sequencing
- STIM, stop imatinib
- TKD, tyrosine kinase domain
- TKI, tyrosine kinase inhibitor
- WHO, World Health Organisation
- ZMW, zero-mode wave-guided
- allo-SCT, Allogeneic Stem Cell Transplantation
- cDNA, coding or complimentary DNA
- dMIQE, Minimum Information for Publication of Quantitative Digital PCR Experiments
- dPCR
- dPCR, digital polymerase chain reaction
- emPCR, emulsion PCR
- gDNA, genomic deoxyribonucleic acid
- m-bcr, minor-breakpoint cluster region
- mRNA, messenger RNA
- nM, manomolar
- μ-bcr, micro-breakpoint cluster region
- μg, microgram
- μl, microliter
Collapse
Affiliation(s)
- Mary Alikian
- Centre for Haematology, Department of Medicine, Imperial College London Hammersmith Hospital, London UK; Imperial Molecular Pathology, Imperial College Healthcare Trust, Hammersmith Hospital, London, UK
| | - Robert Peter Gale
- Centre for Haematology, Department of Medicine, Imperial College London Hammersmith Hospital, London UK
| | - Jane F Apperley
- Centre for Haematology, Department of Medicine, Imperial College London Hammersmith Hospital, London UK
| | - Letizia Foroni
- Centre for Haematology, Department of Medicine, Imperial College London Hammersmith Hospital, London UK
| |
Collapse
|
21
|
Whale AS, Devonshire AS, Karlin-Neumann G, Regan J, Javier L, Cowen S, Fernandez-Gonzalez A, Jones GM, Redshaw N, Beck J, Berger AW, Combaret V, Dahl Kjersgaard N, Davis L, Fina F, Forshew T, Fredslund Andersen R, Galbiati S, González Hernández Á, Haynes CA, Janku F, Lacave R, Lee J, Mistry V, Pender A, Pradines A, Proudhon C, Saal LH, Stieglitz E, Ulrich B, Foy CA, Parkes H, Tzonev S, Huggett JF. International Interlaboratory Digital PCR Study Demonstrating High Reproducibility for the Measurement of a Rare Sequence Variant. Anal Chem 2017; 89:1724-1733. [DOI: 10.1021/acs.analchem.6b03980] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Alexandra S. Whale
- Molecular and Cell Biology Team, LGC, Queens
Road, Teddington, Middlesex TW11 0LY, United Kingdom
| | - Alison S. Devonshire
- Molecular and Cell Biology Team, LGC, Queens
Road, Teddington, Middlesex TW11 0LY, United Kingdom
| | - George Karlin-Neumann
- Digital Biology Center, Bio-Rad Laboratories, 5731 West Las Positas, Pleasanton, California 94588, United States
| | - Jack Regan
- Digital Biology Center, Bio-Rad Laboratories, 5731 West Las Positas, Pleasanton, California 94588, United States
| | - Leanne Javier
- Digital Biology Center, Bio-Rad Laboratories, 5731 West Las Positas, Pleasanton, California 94588, United States
| | - Simon Cowen
- Statistics
Team, LGC, Queens Road, Teddington, Middlesex TW11 0LY, United Kingdom
| | - Ana Fernandez-Gonzalez
- Molecular and Cell Biology Team, LGC, Queens
Road, Teddington, Middlesex TW11 0LY, United Kingdom
| | - Gerwyn M. Jones
- Molecular and Cell Biology Team, LGC, Queens
Road, Teddington, Middlesex TW11 0LY, United Kingdom
| | - Nicholas Redshaw
- Molecular and Cell Biology Team, LGC, Queens
Road, Teddington, Middlesex TW11 0LY, United Kingdom
| | - Julia Beck
- Chronix Biomedical, Goetheallee 8, 37073 Goettingen, Germany
| | - Andreas W. Berger
- Department
of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Valérie Combaret
- Laboratoire de Recherche
Translationnelle, Centre Léon-Bérard, Lyon, F-69008, France
| | - Nina Dahl Kjersgaard
- Sjællands Universitetshospital, Klinisk Biokemisk
Afdeling, Molekylærenhed, Sygehusvej 10, 4000 Roskilde, Denmark
| | - Lisa Davis
- Genoptix Inc., 1811 Aston Avenue, Carlsbad, California 92008, United States
| | - Frederic Fina
- Service
de Transfert d’Oncologie Biologique, Laboratoire de Biologie
Médicale, Faculte de médecine Nord, Boulevard Pierre
Dramard, Marseille 13916 cedex 20, France
| | - Tim Forshew
- UCL Cancer Institute, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6DD, United Kingdom
| | - Rikke Fredslund Andersen
- Department of Clinical Immunology and Biochemistry, Vejle Hospital, Kabbeltoft 25, 7100 Vejle, Denmark
| | - Silvia Galbiati
- Division of Genetics and
Cell Biology, IRCCS San Raffaele Scientific Institute, via Olgettina
60, 20132 Milano, Italy
| | - Álvaro González Hernández
- Department of Pediatrics and CIMA LAB Diagnostics, Clínica Universidad de Navarra, Avenida Pío XII 36, 31008 Pamplona, Spain
| | - Charles A. Haynes
- Michael Smith Laboratories, University of British Columbia, 301 Michael Smith Building, 2185 East Mall, Vancouver, BC V6T 1Z4, Canada
| | - Filip Janku
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics (Phase
I Clinical Trials Program), 1515 Holcombe Boulevard 0455, Houston, Texas 77030, United States
| | - Roger Lacave
- Solid Tumours Genomics Unit, Tenon Hospital, APHP and Université Pierre et Marie Curie, 4 rue
de la Chine, 75970 Paris, France
| | - Justin Lee
- Johns Hopkins, 1650 Orleans St., Baltimore, Maryland 21287, United States
| | - Vilas Mistry
- Department
of Cancer Studies, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester
Royal Infirmary, Leicester LE2 7LX, United Kingdom
| | - Alexandra Pender
- Lung Cancer Group, Division of Molecular
Pathology, The Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, United Kingdom
| | - Anne Pradines
- Institut Claudius Regaud − IUCTO, Laboratoire de Biologie Medicale Oncologique, 1 avenue Irène Joliot-Curie, Toulouse 31059 cedex 9, France
| | - Charlotte Proudhon
- Institut Curie, PSL Research University, SiRIC, Laboratory of Circulating Tumor Biomarkers, 26 rue d’Ulm, 75005 Paris, France
| | - Lao H. Saal
- Department of Oncology and Pathology, Lund University, Scheelevägen 2, MV 404-B2, SE-22381, Lund, Sweden
| | - Elliot Stieglitz
- Department of Pediatrics, Benioff Children’s Hospital, UCSF Helen Diller Family Comprehensive Cancer Center, 1450 Third Street, San Francisco, California 94158, United States
| | - Bryan Ulrich
- Dana Farber Cancer Institute, Belfer Center for Applied Cancer Science and Department
of Medical Oncology, Boston, Massachusetts 02115, United States
| | - Carole A. Foy
- Molecular and Cell Biology Team, LGC, Queens
Road, Teddington, Middlesex TW11 0LY, United Kingdom
| | - Helen Parkes
- Molecular and Cell Biology Team, LGC, Queens
Road, Teddington, Middlesex TW11 0LY, United Kingdom
| | - Svilen Tzonev
- Digital Biology Center, Bio-Rad Laboratories, 5731 West Las Positas, Pleasanton, California 94588, United States
| | - Jim F. Huggett
- Molecular and Cell Biology Team, LGC, Queens
Road, Teddington, Middlesex TW11 0LY, United Kingdom
- School of Biosciences & Medicine, Faculty of Health & Medical Science, University of Surrey, Guildford, Surrey GU2 7XH, United Kingdom
| |
Collapse
|
22
|
Siddique MR, Rutter AV, Wehbe K, Cinque G, Bellisola G, Sulé-Suso J. Effects of nilotinib on leukaemia cells using vibrational microspectroscopy and cell cloning. Analyst 2017; 142:1299-1307. [DOI: 10.1039/c6an01914e] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
S-FTIR and Raman microspectroscopies identify spectral markers of sensitivity/resistance to nilotinib in leukaemia cell clones.
Collapse
Affiliation(s)
- M. R. Siddique
- Institute for Science and Technology in Medicine
- Keele University
- Guy Hilton Research Centre
- Stoke on Trent ST4 7QB
- UK
| | - A. V. Rutter
- Institute for Science and Technology in Medicine
- Keele University
- Guy Hilton Research Centre
- Stoke on Trent ST4 7QB
- UK
| | - K. Wehbe
- Diamond Light Source
- Harwell Science and Innovation Campus
- Didcot
- UK
| | - G. Cinque
- Diamond Light Source
- Harwell Science and Innovation Campus
- Didcot
- UK
| | - G. Bellisola
- University Hospital of Verona
- Department of Pathology and Diagnostics
- Unit of Immunology
- I-37134 Verona
- Italy
| | - J. Sulé-Suso
- Institute for Science and Technology in Medicine
- Keele University
- Guy Hilton Research Centre
- Stoke on Trent ST4 7QB
- UK
| |
Collapse
|
23
|
Branford S. Molecular monitoring in chronic myeloid leukemia-how low can you go? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2016; 2016:156-163. [PMID: 27913475 PMCID: PMC6142476 DOI: 10.1182/asheducation-2016.1.156] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Molecular monitoring of BCR-ABL1 transcripts for patients with chronic myeloid leukemia (CML) is now used to assess response to tyrosine kinase inhibitors (TKIs), including treatment failure that mandates a change of therapy. Therefore, many centers have adopted the molecular technique for measuring BCR-ABL1 and rely on conversion of values to the international reporting scale for appropriate clinical interpretation. However, the technique has a degree of inherent variability despite standardized procedures, which means care should be taken by the clinician when assessing response based on BCR-ABL1 cutoff limits. The last few years have witnessed the emergence of a new molecular response target, which is the achievement and maintenance of a deep molecular response. The ability to achieve treatment-free remission for some patients has shifted the relevant boundary for molecular response. However, the definitive safe BCR-ABL1 transcript level and length of the maintenance phase after which treatment cessation can be attempted has not yet been determined. For patients with TKI resistance, BCR-ABL1 kinase domain mutation analysis remains an essential assessment to guide therapy. Furthermore, low-level mutation detection is clinically relevant for response prediction to subsequent TKI therapy for some patients. Multiple low-level mutations may be a biomarker of a clonally diverse disease with the propensity for resistance evolution. Overall, molecular monitoring, including low-level monitoring is a fundamental component of management for patients with CML.
Collapse
MESH Headings
- Drug Resistance, Neoplasm
- Fusion Proteins, bcr-abl/blood
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Protein Kinase Inhibitors/therapeutic use
- RNA, Messenger/blood
- RNA, Messenger/genetics
- RNA, Neoplasm/blood
- RNA, Neoplasm/genetics
Collapse
Affiliation(s)
- Susan Branford
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia; School of Pharmacy and Medical Science, University of South Australia, Adelaide, Australia; School of Medicine; and School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| |
Collapse
|
24
|
Marum JE, Branford S. Current developments in molecular monitoring in chronic myeloid leukemia. Ther Adv Hematol 2016; 7:237-251. [PMID: 27695615 PMCID: PMC5026293 DOI: 10.1177/2040620716657994] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Molecular monitoring plays an essential role in the clinical management of chronic myeloid leukemia (CML) patients, and now guides clinical decision making. Quantitative reverse-transcriptase-polymerase-chain-reaction (qRT-PCR) assessment of BCR-ABL1 transcript levels has become the standard of care protocol in CML. However, further developments are required to assess leukemic burden more efficiently, monitor minimal residual disease (MRD), detect mutations that drive resistance to tyrosine kinase inhibitor (TKI) therapy and identify predictors of response to TKI therapy. Cartridge-based BCR-ABL1 quantitation, digital PCR and next generation sequencing are examples of technologies which are currently being explored, evaluated and translated into the clinic. Here we review the emerging molecular methods/technologies currently being developed to advance molecular monitoring in CML.
Collapse
Affiliation(s)
- Justine Ellen Marum
- Centre for Cancer Biology, SA Pathology, Adelaide, Australia
- Division of Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Susan Branford
- Centre for Cancer Biology, SA Pathology, Adelaide, Australia
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA, Australia
- School of Medicine, University of Adelaide, SA, Adelaide, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
25
|
Moving treatment-free remission into mainstream clinical practice in CML. Blood 2016; 128:17-23. [PMID: 27013442 DOI: 10.1182/blood-2016-01-694265] [Citation(s) in RCA: 245] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/22/2016] [Indexed: 12/16/2022] Open
Abstract
The dramatic success of tyrosine kinase inhibitors (TKIs) has led to the widespread perception that chronic myeloid leukemia (CML) has become another chronic disease, where lifelong commitment to pharmacologic control is the paradigm. Recent trials demonstrate that some CML patients who have achieved stable deep molecular response can safely cease their therapy without relapsing (treatment free remission [TFR]). Furthermore, those who are unsuccessful in their cessation attempt can safely re-establish remission after restarting their TKI therapy. Based on the accumulated data on TFR, we propose that it is now time to change our approach for the many CML patients who have achieved a stable deep molecular response on long-term TKI therapy. Perhaps half of these patients could successfully achieve TFR if offered the opportunity. For many of these patients ongoing therapy is impairing quality of life and imposing a heavy financial burden while arguably achieving nothing. This recommendation is based on the evident safety of cessation attempts and TFR in the clinical trial setting. We acknowledge that there are potential risks associated with cessation attempts in wider clinical practice, but this should not deter us. Instead we need to establish criteria for safe and appropriate TKI cessation. Clinical trials will enable us to define the best strategies to achieve TFR, but clinicians need guidance today about how to approach this issue with their patients. We outline circumstances in which it would be in the patient's best interest to continue TKI, as well as criteria for a safe TFR attempt.
Collapse
|