51
|
Thakral D, Das N, Basnal A, Gupta R. Cell-free DNA for genomic profiling and minimal residual disease monitoring in Myeloma- are we there yet? AMERICAN JOURNAL OF BLOOD RESEARCH 2020; 10:26-45. [PMID: 32685257 PMCID: PMC7364270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/11/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE Multiple myeloma (MM), a plasma cell neoplasm, afflicts elder individuals accounting for 10% of hematologic malignancies. The MM plasma cells largely reside within the bone marrow niche and are accessible through an invasive bone marrow biopsy, which is challenging during serial monitoring of patients. In this setting, cell free DNA (cfDNA) may have a role to ascertain the molecular aberrations at diagnosis and in assessment of residual disease during therapy. The aim of this review was to explore the utility and current status of cfDNA in MM. METHOD PubMed was searched with terms including cell-free DNA, circulating-tumor DNA, Multiple Myeloma, diagnosis, genomic profiling, Minimal Residual Disease individually or in combination to shortlist the relevant studies. RESULT cfDNA serves as a non-invasive source of tumor-specific molecular biomarker, ctDNA that has immense potential in facilitating management of cancer patients. The mutation detection platforms for ctDNA include hybrid capture and ultra-deep sequencing. Hybrid capture allows full length gene sequencing for mutation and CNV detection. The disease progression can be monitored by profiling prognostic somatic copy number alterations by ultra-low pass whole genome sequencing of ctDNA cost-effectively. Evolution of both the laboratory protocols and bioinformatics tools may further improve the sensitivity of ctDNA detection for better disease management. Only a limited number of studies were available in MM exploring the potential utility of cfDNA. CONCLUSION In this review, we discuss the nuances and challenges associated with molecular evaluation of cfDNA and its potential role in diagnosis and monitoring of treatment response in MM.
Collapse
Affiliation(s)
- Deepshi Thakral
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences New Delhi, India
| | - Nupur Das
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences New Delhi, India
| | - Atul Basnal
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences New Delhi, India
| | - Ritu Gupta
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences New Delhi, India
| |
Collapse
|
52
|
Sanoja-Flores L, Flores-Montero J, Pérez-Andrés M, Puig N, Orfao A. Detection of Circulating Tumor Plasma Cells in Monoclonal Gammopathies: Methods, Pathogenic Role, and Clinical Implications. Cancers (Basel) 2020; 12:E1499. [PMID: 32521788 PMCID: PMC7352573 DOI: 10.3390/cancers12061499] [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: 05/01/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/28/2022] Open
Abstract
Cancer dissemination and distant metastasis most frequently require the release of tumor cells into the blood circulation, both in solid tumors and most hematological malignancies, including plasma cell neoplasms. However, detection of blood circulating tumor cells in solid tumors and some hematological malignancies, such as the majority of mature/peripheral B-cell lymphomas and monoclonal gammopathies, has long been a challenge due to their very low frequency. In recent years, the availability of highly-sensitive and standardized methods for the detection of circulating tumor plasma cells (CTPC) in monoclonal gammopathies, e.g., next-generation flow cytometry (NGF), demonstrated the systematic presence of CTPC in blood in virtually every smoldering (SMM) and symptomatic multiple myeloma (MM) patient studied at diagnosis, and in the majority of patients with newly-diagnosed monoclonal gammopathies of undetermined significance (MGUS). These methods set the basis for further detailed characterization of CTPC vs. their bone marrow counterpart in monoclonal gammopathies, to investigate their role in the biology of the disease, and to confirm their strong impact on patient outcome when measured both at diagnosis and after initiating therapy. Here, we review the currently available techniques for the detection of CTPC, and determine their biological features, physiopathological role and clinical significance in patients diagnosed with distinct diagnostic categories of plasma cell neoplasms.
Collapse
Affiliation(s)
- Luzalba Sanoja-Flores
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)- University of Salamanca, 37007 Salamanca, Spain; (L.S.-F.); (J.F.-M.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00400, Instituto Carlos III, 28029 Madrid, Spain
| | - Juan Flores-Montero
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)- University of Salamanca, 37007 Salamanca, Spain; (L.S.-F.); (J.F.-M.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00400, Instituto Carlos III, 28029 Madrid, Spain
| | - Martín Pérez-Andrés
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)- University of Salamanca, 37007 Salamanca, Spain; (L.S.-F.); (J.F.-M.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00400, Instituto Carlos III, 28029 Madrid, Spain
| | - Noemí Puig
- Department of Hematology, University Hospital of Salamanca, IBSAL, IBMCC (USAL-CSIC), 37007 Salamanca, Spain;
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00233, Instituto Carlos III, 28029 Madrid, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)- University of Salamanca, 37007 Salamanca, Spain; (L.S.-F.); (J.F.-M.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00400, Instituto Carlos III, 28029 Madrid, Spain
| |
Collapse
|
53
|
Kostopoulos IV, Ntanasis-Stathopoulos I, Gavriatopoulou M, Tsitsilonis OE, Terpos E. Minimal Residual Disease in Multiple Myeloma: Current Landscape and Future Applications With Immunotherapeutic Approaches. Front Oncol 2020; 10:860. [PMID: 32537439 PMCID: PMC7267070 DOI: 10.3389/fonc.2020.00860] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 04/30/2020] [Indexed: 01/10/2023] Open
Abstract
The basic principle that deeper therapeutic responses lead to better clinical outcomes in cancer has emerged technologies capable of detecting rare residual tumor cells. The need for ultra-sensitive approaches for minimal residual disease (MRD) detection is particularly evident in Multiple Myeloma (MM), where patients will ultimately relapse despite the achievement of complete remission, which is commonplace due to remarkable therapeutic advances. Consequently, current response criteria on MM have been amended based on MRD status and MRD negativity is now considered the most dominant prognostic factor and the most valuable indicator for a subsequent relapse. However, there are particular limitations and several aspects for MRD assessment that remain open. This review summarizes current data on MRD in the clinical management of MM, highlights open issues and discusses the challenges and the endless opportunities arising for both patients and clinicians. Furthermore, it focuses on the current status of MRD in clinical trials, its dynamics in addressing debatable aspects in the clinical handling and its potential role as the prevailing factor for future MRD-driven tailored therapies.
Collapse
Affiliation(s)
- Ioannis V Kostopoulos
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ourania E Tsitsilonis
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
54
|
Yegin ZA, Can F, Gökçen S, Sadioğlu RE, Özkurt ZN, İlhan Ç, Yağcı M. The Impact of Pre-transplant Cell-free DNA Levels on Leukemia Relapse and Transplant-related Complications in Allogeneic Hematopoietic Stem Cell Transplant Recipients. Balkan Med J 2020; 37:138-143. [PMID: 31970974 PMCID: PMC7161624 DOI: 10.4274/balkanmedj.galenos.2020.2019.8.25] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Background: Cell-free DNA, which may be considered as “liquid” biopsy, may serve as a diagnostic and prognostic marker not only in hematological malignancies but in solid tumors as well. Aims: To investigate the prognostic role of pre-transplant cell-free DNA levels in allogeneic hematopoietic stem cell transplant recipients. Study Design: Retrospective cohort study. Methods: A total of 177 allogeneic hematopoietic stem cell transplant recipients [median age: 36 (16-66) years; male/female: 111/66] with an initial diagnosis of acute leukemia were included in the study. Cell-free DNA was extracted from pre-transplant serum samples by using the MagNA Pure Compact Nucleic Acid Isolation Kit I with the MagNA Pure Compact instrument (Roche Diagnostics, Penzberg, Germany). Results: A positive correlation was demonstrated between cell-free DNA and age (p=0.018; r=0.177). Pre-transplant cell-free DNA levels were lower in bcr-abl (+) patients (p=0.001), while an adverse correlation was indicated between cell-free DNA and bcr-abl levels (p=0.001; r=−0.531). Acute lymphoblastic leukemia patients with bcr-abl positivity (p=0.001) or abnormal cytogenetics (p=0.038) represented significantly lower pre-transplant cell-free DNA levels. Cell-free DNA levels were lower in patients who developed sinusoidal obstruction syndrome (p=0.035). In terms of long-term complications, acute myeloid leukemia patients who experienced post-transplant relapse had significantly lower pre-transplant cell-free DNA levels (p=0.024). Overall survival was not statistically different between high- and low- cell-free DNA groups (45.2% vs 22.5; p=0.821). Conclusion: In general, low serum levels of pre-transplant cell-free DNA seem to be associated with transplant-related morbidities and may be considered an adverse prognostic factor for allogeneic hematopoietic stem cell transplant recipients.
Collapse
Affiliation(s)
- Zeynep Arzu Yegin
- Department of Hematology, Gazi University School of Medicine, Ankara, Turkey
| | - Ferda Can
- Department of Hematology, Gazi University School of Medicine, Ankara, Turkey
| | - Sanem Gökçen
- Department of Hematology, Gazi University School of Medicine, Ankara, Turkey
| | - Rezzan Eren Sadioğlu
- Department of Internal Medicine, Gazi University School of Medicine, Ankara, Turkey
| | - Zübeyde Nur Özkurt
- Department of Hematology, Gazi University School of Medicine, Ankara, Turkey
| | - Çiğdem İlhan
- Department of Hematology, Gazi University School of Medicine, Ankara, Turkey
| | - Münci Yağcı
- Department of Hematology, Gazi University School of Medicine, Ankara, Turkey
| |
Collapse
|
55
|
Ferreira B, Caetano J, Barahona F, Lopes R, Carneiro E, Costa-Silva B, João C. Liquid biopsies for multiple myeloma in a time of precision medicine. J Mol Med (Berl) 2020; 98:513-525. [PMID: 32246161 PMCID: PMC7198642 DOI: 10.1007/s00109-020-01897-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 12/17/2022]
Abstract
Multiple myeloma (MM) is a challenging, progressive, and highly heterogeneous hematological malignancy. MM is characterized by multifocal proliferation of neoplastic plasma cells in the bone marrow (BM) and sometimes in extramedullary organs. Despite the availability of novel drugs and the longer median overall survival, some patients survive more than 10 years while others die rapidly. This heterogeneity is mainly driven by biological characteristics of MM cells, including genetic abnormalities. Disease progressions are mainly due to the inability of drugs to overcome refractory disease and inevitable drug-resistant relapse. In clinical practice, a bone marrow biopsy, mostly performed in one site, is still used to access the genetics of MM. However, BM biopsy use is limited by its invasive nature and by often not accurately reflecting the mutational profile of MM. Recent insights into the genetic landscape of MM provide a valuable opportunity to implement precision medicine approaches aiming to enable better patient profiling and selection of targeted therapies. In this review, we explore the use of the emerging field of liquid biopsies in myeloma patients considering current unmet medical needs, such as assessing the dynamic mutational landscape of myeloma, early predictors of treatment response, and a less invasive response monitoring.
Collapse
Affiliation(s)
- Bruna Ferreira
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Joana Caetano
- Hemato-Oncology Unit, Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Filipa Barahona
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Raquel Lopes
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Emilie Carneiro
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Bruno Costa-Silva
- Systems Oncology Group, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Cristina João
- Hemato-Oncology Unit, Myeloma and Lymphoma Research Programme, Nova Medical School, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|
56
|
Blood monitoring of circulating tumor plasma cells by next generation flow in multiple myeloma after therapy. Blood 2020; 134:2218-2222. [PMID: 31697808 DOI: 10.1182/blood.2019002610] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
57
|
Long X, Xu Q, Lou Y, Li C, Gu J, Cai H, Wang D, Xu J, Li T, Zhou X, Xiao M, Wang Y, Mao X, Zhou J, Chen L. The utility of non-invasive liquid biopsy for mutational analysis and minimal residual disease assessment in extramedullary multiple myeloma. Br J Haematol 2020; 189:e45-e48. [PMID: 32191818 DOI: 10.1111/bjh.16440] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Xiaolu Long
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Qian Xu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Yaoyao Lou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Chunrui Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Jia Gu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Haodong Cai
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Di Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Jinhuan Xu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Tongjuan Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Xiaoxi Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Ying Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Xia Mao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Liting Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| |
Collapse
|
58
|
Bingham N, Spencer A. The role of cell free DNA and liquid biopsies in haematological conditions. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:521-531. [PMID: 35582436 PMCID: PMC8992501 DOI: 10.20517/cdr.2019.93] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cell free nucleic acids (CFNAs) are nucleic acids released from cells that circulate within bodily fluids. Recent advances in molecular techniques have led the ability to interrogate CFNAs in a clinically meaningful way, for example the identification and assessment of foetal CFNAs in maternal blood, allowing minimally invasive testing for foetal genetic abnormalities. The majority of CFNAs arise from haemopoietic cells, making it a particularly rich source of genetic information in haematological conditions. Furthermore, the innate genetic heterogeneity of haematological malignancies, as epitomised by multiple myeloma, lend itself well to “liquid biopsies”. This approach promises to provide a more wholistic assessment of whole disease genetics, especially when contrasted against the current gold-standard of single site tissue biopsies. This review briefly summarises the definitions and physiology of CFNAs, both cell free DNA (cfDNA) and extracellular RNA (exRNA), before exploring the literature surrounding the current and future roles of cfDNA in the haematological malignancies and patient care.
Collapse
Affiliation(s)
- Nicholas Bingham
- Australian Centre for Blood Diseases, Monash University, Victoria 3181, Australia
| | - Andrew Spencer
- Australian Centre for Blood Diseases, Monash University, Victoria 3181, Australia
| |
Collapse
|
59
|
Bolli N, Genuardi E, Ziccheddu B, Martello M, Oliva S, Terragna C. Next-Generation Sequencing for Clinical Management of Multiple Myeloma: Ready for Prime Time? Front Oncol 2020; 10:189. [PMID: 32181154 PMCID: PMC7057289 DOI: 10.3389/fonc.2020.00189] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/04/2020] [Indexed: 12/22/2022] Open
Abstract
Personalized treatment is an attractive strategy that promises increased efficacy with reduced side effects in cancer. The feasibility of such an approach has been greatly boosted by next-generation sequencing (NGS) techniques, which can return detailed information on the genome and on the transcriptome of each patient's tumor, thus highlighting biomarkers of response or druggable targets that may differ from case to case. However, while the number of cancers sequenced is growing exponentially, much fewer cases are amenable to a molecularly-guided treatment outside of clinical trials to date. In multiple myeloma, genomic analysis shows a variety of gene mutations, aneuploidies, segmental copy-number changes, translocations that are extremely heterogeneous, and more numerous than other hematological malignancies. Currently, in routine clinical practice we employ reduced FISH panels that only capture three high-risk features as part of the R-ISS. On the contrary, recent advances have suggested that extending genomic analysis to the full spectrum of recurrent mutations and structural abnormalities in multiple myeloma may have biological and clinical implications. Furthermore, increased efficacy of novel treatments can now produce deeper responses, and standard methods do not have enough sensitivity to stratify patients in complete biochemical remission. Consequently, NGS techniques have been developed to monitor the size of the clone to a sensitivity of up to a cell in a million after treatment. However, even these techniques are not within reach of standard laboratories. In this review we will recapitulate recent advances in multiple myeloma genomics, with special focus on the ones that may have immediate translational impact. We will analyze the benefits and pitfalls of NGS-based diagnostics, highlighting crucial aspects that will need to be taken into account before this can be implemented in most laboratories. We will make the point that a new era in myeloma diagnostics and minimal residual disease monitoring is close and conventional genetic testing will not be able to return the required information. This will mandate that even in routine practice NGS should soon be adopted owing to a higher informative potential with increasing clinical benefits.
Collapse
Affiliation(s)
- Niccolo Bolli
- Department of Clinical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
| | - Elisa Genuardi
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Bachisio Ziccheddu
- Department of Clinical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Marina Martello
- Seràgnoli Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Stefania Oliva
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Carolina Terragna
- Seràgnoli Institute of Hematology, Azienda Ospedaliero-Universitaria Sant'Orsola-Malpighi, Bologna, Italy
| |
Collapse
|
60
|
Hultcrantz M, Yellapantula V, Rustad EH. Genomic profiling of multiple myeloma: New insights and modern technologies. Best Pract Res Clin Haematol 2020; 33:101153. [PMID: 32139018 DOI: 10.1016/j.beha.2020.101153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023]
Abstract
Advances in technologies for genomic profiling, primarily with next generation sequencing, have lead to a better understanding of the complex genomic landscape in multiple myeloma. Integrated analysis of whole genome, exome and transcriptome sequencing has lead to new insights on disease drivers including translocations, copy number alterations, somatic mutations, and altered gene expression. Disease progression in multiple myeloma is largely driven by structural variations including the traditional immunoglobulin heavy chain (IGH) translocations and hyperdiploidy which are early events in myelomagenesis as well as more complex events spanning over multiple chromosomes and involving amplifications and deletions. In this review, we will discuss recent insights on the genomic landscape of multiple myeloma and their implications for disease progression and personalized treatment. We will review how sequencing assays compare to current clinical methods and give an overview of modern technologies for interrogating genomic aberrations.
Collapse
Affiliation(s)
- Malin Hultcrantz
- Myeloma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| | - Venkata Yellapantula
- Myeloma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Even H Rustad
- Myeloma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
61
|
Waldschmidt JM, Vijaykumar T, Knoechel B, Lohr JG. Tracking myeloma tumor DNA in peripheral blood. Best Pract Res Clin Haematol 2020; 33:101146. [PMID: 32139012 DOI: 10.1016/j.beha.2020.101146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/09/2020] [Indexed: 01/03/2023]
Abstract
Over the past years, the emergence of liquid biopsy technologies has dramatically expanded our ability to assess multiple myeloma without the need for invasive sampling. Interrogation of cell-free DNA from the peripheral blood recapitulates the mutational landscape at excellent concordance with matching bone marrow aspirates. It can quantify disease burden and identify previously undetected resistance mechanisms which may inform clinical management in real-time. The convenience of sample acquisition and storage provides strong procedural benefits over currently available testing. Further investigations will have to define the role of cell-free DNA as a diagnostic measure by determining clinically relevant tumor thresholds in comparison to existing routine parameters. This review presents an overview of currently available assays and discusses the clinical value, potential and limitations of cell-free DNA technologies for the assessment of this challenging disease.
Collapse
Affiliation(s)
- Johannes M Waldschmidt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Tushara Vijaykumar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Birgit Knoechel
- Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jens G Lohr
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
62
|
Vrabel D, Sedlarikova L, Besse L, Rihova L, Bezdekova R, Almasi M, Kubaczkova V, Brožová L, Jarkovsky J, Plonkova H, Jelinek T, Sandecka V, Stork M, Pour L, Sevcikova S, Hajek R. Dynamics of tumor-specific cfDNA in response to therapy in multiple myeloma patients. Eur J Haematol 2019; 104:190-197. [PMID: 31763708 PMCID: PMC7065130 DOI: 10.1111/ejh.13358] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 12/14/2022]
Abstract
Objectives Progress in multiple myeloma treatment allows patients to achieve deeper responses, for which the assessment of minimal residual disease (MRD) is critical. Typically, bone marrow samples are used for this purpose; however, this approach is site‐limited. Liquid biopsy represents a minimally invasive and more comprehensive technique that is not site‐limited, but equally challenging. Methods While majority of current data comes from short‐term studies, we present a long‐term study on blood‐based MRD monitoring using tumor‐specific cell‐free DNA detection by ASO‐qPCR. One hundred and twelve patients were enrolled into the study, but long‐term sampling and analysis were feasible only in 45 patients. Results We found a significant correlation of quantity of tumor‐specific cell‐free DNA levels with clinically meaningful events [induction therapy (P = .004); ASCT (P = .012)]. Moreover, length of cfDNA fragments is associated with better treatment response of patients. Conclusions These results support the concept of tumor‐specific cell‐free DNA as a prognostic marker.
Collapse
Affiliation(s)
- David Vrabel
- Babak Myeloma Group, Department of Pathophysiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Lenka Sedlarikova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, Czech Republic
| | - Lenka Besse
- Laboratory of Experimental Oncology, Department of Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Lucie Rihova
- Department of Clinical Hematology, University Hospital Brno, Brno, Czech Republic
| | - Renata Bezdekova
- Department of Clinical Hematology, University Hospital Brno, Brno, Czech Republic
| | - Martina Almasi
- Department of Clinical Hematology, University Hospital Brno, Brno, Czech Republic
| | - Veronika Kubaczkova
- Babak Myeloma Group, Department of Pathophysiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lucie Brožová
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jiri Jarkovsky
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Hana Plonkova
- Department of Hematooncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Tomas Jelinek
- Department of Hematooncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Viera Sandecka
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Martin Stork
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Ludek Pour
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Sabina Sevcikova
- Babak Myeloma Group, Department of Pathophysiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Roman Hajek
- Department of Hematooncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| |
Collapse
|
63
|
Manasanch EE. What to do with minimal residual disease testing in myeloma. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:137-141. [PMID: 31808833 PMCID: PMC6913484 DOI: 10.1182/hematology.2019000080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The role and use of minimal residual disease (MRD) testing has changed significantly over the past few years as it has become part of the routine care for response assessment in multiple myeloma. The most widely used standardized methods to assess MRD in myeloma in the bone marrow are multicolor flow cytometry and next-generation sequencing. Importantly, the depth of MRD negativity in the bone marrow correlates with improved progression-free survival and overall survival in myeloma. Whole-body position emission tomography-computed tomography and magnetic resonance imaging are also used to evaluate patchy and extramedullary disease, which may not be readily visible through bone marrow assessment. This article reviews a clinical case in which MRD testing, both in bone marrow and in functional imaging, is part of the standard of care. It also reviews the different modalities of MRD testing and current practice guidelines. Finally, patients with myeloma may be tested for MRD after treatment because this is part of the routine response assessment according to International Myeloma Working Group criteria and correlates with clinical outcomes. Important questions such as when to stop therapy for sustained MRD-negative patients or whether to change treatments for patients who go from MRD negative to positive without other evidence of disease relapse are being evaluated in clinical trials and remain controversial.
Collapse
Affiliation(s)
- Elisabet E Manasanch
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
64
|
Rustad EH, Misund K, Bernard E, Coward E, Yellapantula VD, Hultcrantz M, Ho C, Kazandjian D, Korde N, Mailankody S, Keats JJ, Akhlaghi T, Viny AD, Mayman DJ, Carroll K, Patel M, Famulare CA, Bruinink DH, Hutt K, Jacobsen A, Huang Y, Miller JE, Maura F, Papaemmanuil E, Waage A, Arcila ME, Landgren O. Stability and uniqueness of clonal immunoglobulin CDR3 sequences for MRD tracking in multiple myeloma. Am J Hematol 2019; 94:1364-1373. [PMID: 31571261 DOI: 10.1002/ajh.25641] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/12/2019] [Accepted: 09/16/2019] [Indexed: 12/26/2022]
Abstract
Minimal residual disease (MRD) tracking, by next generation sequencing of immunoglobulin sequences, is moving towards clinical implementation in multiple myeloma. However, there is only sparse information available to address whether clonal sequences remain stable for tracking over time, and to what extent light chain sequences are sufficiently unique for tracking. Here, we analyzed immunoglobulin repertoires from 905 plasma cell myeloma and healthy control samples, focusing on the third complementarity determining region (CDR3). Clonal heavy and/or light chain expression was identified in all patients at baseline, with one or more subclones related to the main clone in 3.2%. In 45 patients with 101 sequential samples, the dominant clonal CDR3 sequences remained identical over time, despite differential clonal evolution by whole exome sequencing in 49% of patients. The low frequency of subclonal CDR3 variants, and absence of evolution over time in active multiple myeloma, indicates that tumor cells at this stage are not under selective pressure to undergo antibody affinity maturation. Next, we establish somatic hypermutation and non-templated insertions as the most important determinants of light chain clonal uniqueness, identifying a potentially trackable sequence in the majority of patients. Taken together, we show that dominant clonal sequences identified at baseline are reliable biomarkers for long-term tracking of the malignant clone, including both IGH and the majority of light chain clones.
Collapse
Affiliation(s)
- Even H. Rustad
- Department of Medicine, Myeloma Service Memorial Sloan Kettering Cancer Center New York New York
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences Norwegian University of Science and Technology Trondheim Norway
| | - Kristine Misund
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences Norwegian University of Science and Technology Trondheim Norway
| | - Elsa Bernard
- Department of Epidemiology and Biostatistics Memorial Sloan Kettering Cancer Center New York New York
| | - Eivind Coward
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences Norwegian University of Science and Technology Trondheim Norway
| | - Venkata D. Yellapantula
- Department of Epidemiology and Biostatistics Memorial Sloan Kettering Cancer Center New York New York
| | - Malin Hultcrantz
- Department of Medicine, Myeloma Service Memorial Sloan Kettering Cancer Center New York New York
| | - Caleb Ho
- Department of Pathology Memorial Sloan Kettering Cancer Center New York New York
| | - Dickran Kazandjian
- Myeloma Program, Lymphoid Malignancies Branch, Center for Cancer Research National Cancer Institute, National Institutes of Health Bethesda Maryland
| | - Neha Korde
- Department of Medicine, Myeloma Service Memorial Sloan Kettering Cancer Center New York New York
| | - Sham Mailankody
- Department of Medicine, Myeloma Service Memorial Sloan Kettering Cancer Center New York New York
| | | | - Theresia Akhlaghi
- Department of Medicine, Myeloma Service Memorial Sloan Kettering Cancer Center New York New York
| | - Aaron D. Viny
- Department of Medicine, Leukemia Service Memorial Sloan Kettering Cancer Center New York New York
- Human Oncology & Pathogenesis Program Memorial Sloan Kettering Cancer Center New York New York
| | - David J. Mayman
- Adult Reconstruction and Joint Replacement Division Hospital for Special Surgery New York New York
| | - Kaitlin Carroll
- Adult Reconstruction and Joint Replacement Division Hospital for Special Surgery New York New York
| | - Minal Patel
- Center for Hematological Malignancies, Department of Medicine Memorial Sloan Kettering Cancer Center New York New York
| | - Christopher A. Famulare
- Center for Hematological Malignancies, Department of Medicine Memorial Sloan Kettering Cancer Center New York New York
| | | | | | | | | | | | - Francesco Maura
- Department of Medicine, Myeloma Service Memorial Sloan Kettering Cancer Center New York New York
| | - Elli Papaemmanuil
- Department of Epidemiology and Biostatistics Memorial Sloan Kettering Cancer Center New York New York
| | - Anders Waage
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences Norwegian University of Science and Technology Trondheim Norway
| | - Maria E. Arcila
- Department of Pathology Memorial Sloan Kettering Cancer Center New York New York
| | - Ola Landgren
- Department of Medicine, Myeloma Service Memorial Sloan Kettering Cancer Center New York New York
| |
Collapse
|
65
|
Wang N, Tesfaluul N, Li J, Gao X, Liu S, Yue B. Enrichment of circulating myeloma cells by immunomagnetic beads combined with flow cytometry for monitoring minimal residual disease and relapse in patients with multiple myeloma. Ann Hematol 2019; 98:2769-2780. [PMID: 31748925 DOI: 10.1007/s00277-019-03833-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 10/28/2019] [Indexed: 12/14/2022]
Abstract
Difficulty in regularly analyzing marrow myeloma cells (MMCs) and low frequency of circulating myeloma cells (CMCs) in blood presents challenges for monitoring minimal residual disease (MRD) in multiple myeloma (MM). We have developed a set of method for enrichment of CMCs by immunomagetic beads (IMB) combined with flow cytometry (IMB-FCM) based on CD38-APC/CD138-APC antibodies in U266-spiked samples and in 122 patient samples. U266 cell capture efficiency of CD38/CD138-IMB-FCM (6.960, 2.574) was 6- and 2-fold higher than that of FCM (1.032), and the sensitivity of FCM and IMB-FCM was 0.01% and 0.001%, respectively. In MM cohort, the positive rate of CMCs by IMB-FCM increased from 60.5~70.0 to 85~87.2% in newly diagnosed/relapsed and partial remission (PR) patients compared with by FCM (P < 0.05). Two complete remission (CR) patients contain certain amounts of CMCs by IMB-FCM while no CMCs and MMCs were detectable by FCM. Patients exhibiting PR and CR upon therapy had much lower CMC and MMC counts than newly diagnosed/relapsed patients (P < 0.005). Based on MRD measurement in BM and PB samples, all FCM-negative BM samples were also paired with FCM/IMB-FCM-negative PB samples among newly diagnosed, relapsed, and PR patients, and FCM-positive BM samples were accompanied by IMB-FCM-positive results in 88% of corresponding PB samples. CMCs strongly associated with other clinical biomarkers of disease burden, including elevated MMCs, β2-MG, sCrea, and DS and ISS stages, and more serious anemia, bone destruction, and renal impairment (P < 0.05). Logistic regression analysis revealed that elevated β2-MG and moderate-to-more anemia were significant risk factors for the presence of CMCs (P < 0.05). As a noninvasive "liquid biopsy" of monitoring MRD, the potential of IMB-FCM for CMC detection may complement or minimize bone marrow aspiration in future treatment of MM patients.
Collapse
Affiliation(s)
- Ningning Wang
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Laboratory Medicine, the First People's Hospital of Pingdingshan, Pingdingshan, Henan, China
| | - Nahom Tesfaluul
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jia Li
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaojuan Gao
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuai Liu
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Faculty of Laboratory Medicine, Zhengzhou University, Zhengzhou, Henan, China.,Key Laboratory Medicine of Henan Province, Faculty of Laboratory Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Baohong Yue
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China. .,Faculty of Laboratory Medicine, Zhengzhou University, Zhengzhou, Henan, China. .,Key Laboratory Medicine of Henan Province, Faculty of Laboratory Medicine, Zhengzhou University, Zhengzhou, Henan, China. .,Open Laboratory, Henan Province Key Subject of Clinical Medicine, Zhengzhou, Henan, China.
| |
Collapse
|
66
|
Myeloma MRD by deep sequencing from circulating tumor DNA does not correlate with results obtained in the bone marrow. Blood Adv 2019; 2:2811-2813. [PMID: 30355580 DOI: 10.1182/bloodadvances.2018025197] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 09/27/2018] [Indexed: 01/10/2023] Open
Abstract
Key Points
There is no correlation between ctDNA and bone marrow for MRD by NGS using only immunoglobulin gene rearrangements in myeloma patients.
Collapse
|
67
|
Simnica D, Schliffke S, Schultheiß C, Bonzanni N, Fanchi LF, Akyüz N, Gösch B, Casar C, Thiele B, Schlüter J, Lohse AW, Binder M. High-Throughput Immunogenetics Reveals a Lack of Physiological T Cell Clusters in Patients With Autoimmune Cytopenias. Front Immunol 2019; 10:1897. [PMID: 31497012 PMCID: PMC6713037 DOI: 10.3389/fimmu.2019.01897] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022] Open
Abstract
Autoimmune cytopenias (AIC) such as immune thrombocytopenia or autoimmune hemolytic anemia are claimed to be essentially driven by a dysregulated immune system. Using next-generation immunosequencing we profiled 59 T and B cell repertoires (TRB and IGH) of 25 newly diagnosed patients with primary or secondary (lymphoma-associated) AIC to test the hypothesis if these patients present a disease-specific immunological signature that could reveal pathophysiological clues and eventually be exploited as blood-based biomarker. Global TRB and IGH repertoire metrics as well as VJ gene usage distribution showed uniform characteristics for all lymphoma patients (high clonality and preferential usage of specific TRBV- and TRBJ genes), but no AIC-specific signature. Since T cell immune reactions toward antigens are unique and polyclonal, we clustered TCRβ clones in-silico based on target recognition using the GLIPH (grouping of lymphocyte interactions by paratope hotspots) algorithm. This analysis revealed a considerable lack of physiological T cell clusters in patients with primary AIC. Interestingly, this signature did not discriminate between the different subentities of AIC and was also found in an independent cohort of 23 patients with active autoimmune hepatitis. Taken together, our data suggests that the identified T cell cluster signature could represent a blood biomarker of autoimmune conditions in general and should be functionally validated in future studies.
Collapse
Affiliation(s)
- Donjete Simnica
- Department of Internal Medicine IV, Oncology and Hematology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Simon Schliffke
- Department of Oncology and Hematology, BMT With Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Christoph Schultheiß
- Department of Internal Medicine IV, Oncology and Hematology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | | | | | - Nuray Akyüz
- Department of Oncology and Hematology, BMT With Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Barbara Gösch
- Department of Oncology and Hematology, BMT With Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Christian Casar
- Department of Gastroenterology With Sections Infectious and Tropical Diseases, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Bioinformatics Core, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Benjamin Thiele
- Department of Oncology and Hematology, BMT With Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Janina Schlüter
- Department of Oncology and Hematology, BMT With Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ansgar W Lohse
- Department of Gastroenterology With Sections Infectious and Tropical Diseases, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Mascha Binder
- Department of Internal Medicine IV, Oncology and Hematology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
68
|
Genotyping circulating tumor DNA of pediatric Hodgkin lymphoma. Leukemia 2019; 34:151-166. [DOI: 10.1038/s41375-019-0541-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/20/2019] [Accepted: 06/17/2019] [Indexed: 12/21/2022]
|
69
|
Romano A, Palumbo GA, Parrinello NL, Conticello C, Martello M, Terragna C. Minimal Residual Disease Assessment Within the Bone Marrow of Multiple Myeloma: A Review of Caveats, Clinical Significance and Future Perspectives. Front Oncol 2019; 9:699. [PMID: 31482061 PMCID: PMC6710454 DOI: 10.3389/fonc.2019.00699] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022] Open
Abstract
There is an increasing clinical interest in the measure and achievement of minimal residual disease (MRD) negativity in the bone marrow of Multiple Myeloma (MM) patients, as defined equally either by Multicolor Flow Cytometry (MFC) or by Next Generation Sequencing (NGS) technologies. At present, modern technologies allow to detect up to one on 104 or on 105 or even on 106 cells, depending on their throughput. MFC approaches, which have been progressively improved up to the so-called Next Generation Flow (NGF), and NGS, which proved clear advantages over ASO-PCR, can detect very low levels of residual disease in the BM. These methods are actually almost superimposable, in terms of MRD detection power, supporting the lack of unanimous preference for either technique on basis of local availability. However, some technical issues are still open: the optimal assay to use to detect either phenotype (e.g., next generation multidimensional flow cytometry, imaging) or genotype aberrations (e.g., ASO-RQ PCR, digital droplet PCR, NGS) and their standardization, the sample source (BM or peripheral blood, PB) and its pre-processing (red-cell lysis vs. Ficoll, fresh vs. frozen samples, requirement of CD138+ cells enrichment). Overall, MRD negativity is considered as the most powerful predictor of favorable long-term outcomes in MM and is likely to represent the major driver of treatment strategies in the near future. In this manuscript, we reviewed the main pitfalls and caveats of MRD detection within bone marrow in MM patients after front-line therapy, highlighting the improving of the currently employed technology and describing alternative methods for MRD testing in MM, such as liquid biopsy.
Collapse
Affiliation(s)
- Alessandra Romano
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
| | - Giuseppe Alberto Palumbo
- Division of Hematology, Azienda Ospedaliero-Universitaria Policlinico Vittorio Emanuele di Catania, Catania, Italy
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie avanzate “G.F. Ingrassia,” University of Catania, Catania, Italy
| | - Nunziatina Laura Parrinello
- Division of Hematology, Azienda Ospedaliero-Universitaria Policlinico Vittorio Emanuele di Catania, Catania, Italy
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie avanzate “G.F. Ingrassia,” University of Catania, Catania, Italy
| | - Concetta Conticello
- Division of Hematology, Azienda Ospedaliero-Universitaria Policlinico Vittorio Emanuele di Catania, Catania, Italy
| | - Marina Martello
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale (DIMES), Università degli Studi di Bologna, Bologna, Italy
| | - Carolina Terragna
- Istituto di Ematologia “L.A.Seràgnoli,” Azienda Ospedaliera Sant'Orsola-Malpighi, Bologna, Italy
| |
Collapse
|
70
|
Monitoring tumour burden and therapeutic response through analysis of circulating tumour DNA and extracellular RNA in multiple myeloma patients. Leukemia 2019; 33:2022-2033. [PMID: 30992504 DOI: 10.1038/s41375-019-0469-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/17/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022]
Abstract
Monitoring tumour burden and therapeutic response through analyses of circulating cell-free tumour DNA (ctDNA) and extracellular RNA (exRNA) in multiple myeloma (MM) patients were performed in a Phase Ib trial of 24 relapsed/refractory patients receiving oral azacitidine in combination with lenalidomide and dexamethasone. Mutational characterisation of paired BM and PL samples at study entry identified that patients with a higher number of mutations or a higher mutational fractional abundance in PL had significantly shorter overall survival (OS) (p = 0.005 and p = 0.018, respectively). A decrease in ctDNA levels at day 5 of cycle 1 of treatment (C1D5) correlated with superior progression-free survival (PFS) (p = 0.017). Evaluation of exRNA transcripts of candidate biomarkers indicated that high CRBN levels coupled with low levels of SPARC at baseline were associated with shorter OS (p = 0.000003). IKZF1 fold-change <0.05 at C1D5 was associated with shorter PFS (p = 0.0051) and OS (p = 0.0001). Furthermore, patients with high baseline CRBN coupled with low fold-change at C1D5 were at the highest risk of progression (p = 0.0001). In conclusion, this exploratory analysis has provided the first demonstration in MM of ctDNA for predicting disease outcome and of the utility of exRNA as a biomarker of therapeutic response.
Collapse
|
71
|
Takamatsu H, Yoroidaka T, Fujisawa M, Kobori K, Hanawa M, Yamashita T, Murata R, Ueda M, Nakao S, Matsue K. Comparison of minimal residual disease detection in multiple myeloma by SRL 8-color single-tube and EuroFlow 8-color 2-tube multiparameter flow cytometry. Int J Hematol 2019; 109:377-381. [PMID: 30778767 DOI: 10.1007/s12185-019-02615-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/08/2019] [Accepted: 02/08/2019] [Indexed: 11/26/2022]
Abstract
We sought to determine the efficacy of a new, inexpensive, single-tube 8-color multiparameter flow cytometry (MFC) method (SRL-Flow), which is based on the EuroFlow next-generation flow (NGF) (tube 2 only), to assess minimal residual disease (MRD)-negative status. MRD-negative status is considered a treatment milestone in multiple myeloma (MM). We used 45 bone marrow samples from patients with MM, including 11 cases treated with anti-CD38 monoclonal antibody. The SRL-Flow sample preparation protocol was identical to that of EuroFlow-NGF. The antibody panel for SRL-Flow was as follows: CD138V450/CD27V500/CD38ME (multiepitope)FITC/CD56PE/CD45PerCP-Cy5.5/CD19PE-Cy7/cytoplasmic (Cy) immunoglobulin (Ig) κAPC/CyIgλAPC-H7. To identify abnormal plasma cells (aPCs) of patients with MM who received anti-CD38 monoclonal antibody, we used a panel of anti-CD45 and anti-CD138 antibodies (Abs) rather than a panel of anti-CD45 and anti-CD38 Abs. We comparatively analyzed the total nucleated cell numbers, total PC levels, and MRD levels between the SRL-Flow and EuroFlow-NGF. High correlations (r > 0.9) in total PC and MRD levels were noted among SRL-Flow, original EuroFlow-NGF (2 tubes), and EuroFlow-NGF (tube 2 only), suggesting that SRL-Flow is an inexpensive (< $200 USD/sample as of January of 2019) alternative to EuroFlow-NGF (< $350 USD/sample) for assessing MRD in MM.
Collapse
Affiliation(s)
- Hiroyuki Takamatsu
- Department of Hematology and Respirology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan.
| | - Takeshi Yoroidaka
- Department of Hematology and Respirology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Momoko Fujisawa
- Department of Hematology and Respirology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Kazuya Kobori
- Cellular Immunology Analysis Section, Genetic and Chromosome Analysis Department, SRL, Inc, Tokyo, Japan
| | - Masako Hanawa
- Cellular Immunology Analysis Section, Genetic and Chromosome Analysis Department, SRL, Inc, Tokyo, Japan
| | - Takeshi Yamashita
- Division of Internal Medicine, Keiju Kanazawa Hospital, Kanazawa, Japan
| | - Ryoichi Murata
- Division of Internal Medicine, Keiju Kanazawa Hospital, Kanazawa, Japan
| | - Mikio Ueda
- Division of Internal Medicine, Keiju Kanazawa Hospital, Kanazawa, Japan
| | - Shinji Nakao
- Department of Hematology and Respirology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Kosei Matsue
- Department of Hematology/Oncology, Kameda Medical Center, Kamogawa, Japan
| |
Collapse
|
72
|
New Molecular Technologies for Minimal Residual Disease Evaluation in B-Cell Lymphoid Malignancies. J Clin Med 2018; 7:jcm7090288. [PMID: 30231510 PMCID: PMC6162632 DOI: 10.3390/jcm7090288] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 09/10/2018] [Accepted: 09/14/2018] [Indexed: 12/19/2022] Open
Abstract
The clearance of malignant clonal cells significantly correlates with clinical outcomes in many hematologic malignancies. Accurate and high throughput tools for minimal residual disease (MRD) detection are needed to overcome some drawbacks of standard molecular techniques; such novel tools have allowed for higher sensitivity analyses and more precise stratification of patients, based on molecular response to therapy. In this review, we depict the recently introduced digital PCR and next-generation sequencing technologies, describing their current application for MRD monitoring in lymphoproliferative disorders. Moreover, we illustrate the feasibility of these new technologies to test less invasive and more patient-friendly tissues sources, such as "liquid biopsy".
Collapse
|
73
|
Caers J, Garderet L, Kortüm KM, O'Dwyer ME, van de Donk NWCJ, Binder M, Dold SM, Gay F, Corre J, Beguin Y, Ludwig H, Larocca A, Driessen C, Dimopoulos MA, Boccadoro M, Gramatzki M, Zweegman S, Einsele H, Cavo M, Goldschmidt H, Sonneveld P, Delforge M, Auner HW, Terpos E, Engelhardt M. European Myeloma Network recommendations on tools for the diagnosis and monitoring of multiple myeloma: what to use and when. Haematologica 2018; 103:1772-1784. [PMID: 30171031 PMCID: PMC6278986 DOI: 10.3324/haematol.2018.189159] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 08/27/2018] [Indexed: 01/04/2023] Open
Abstract
The diagnosis of multiple myeloma can be challenging, even for experienced physicians, and requires close collaboration between numerous disciplines (orthopedics, radiology, nuclear medicine, radiation therapy, hematology and oncology) before the final diagnosis of myeloma is made. The definition of multiple myeloma is based on the presence of clinical, biochemical, histopathological, and radiological markers of disease. Specific tests are needed both at presentation and during follow-up in order to reach the correct diagnosis and characterize the disease precisely. These tests can also serve prognostic purposes and are useful for follow-up of myeloma patients. Molecular analyses remain pivotal for defining high-risk myeloma and are used in updated patient stratifications, while minimal residual disease assessment via flow cytometry, molecular techniques and radiological approaches provides additional prognostic information on patients' long-term outcome. This pivotal information will guide our future treatment decisions in forthcoming clinical trials. The European Myeloma Network group updated their guidelines on different diagnostic recommendations, which should be of value to enable appropriate use of the recommendations both at diagnosis and during follow-up.
Collapse
Affiliation(s)
- Jo Caers
- Department of Hematology, University Hospital of Liege, Belgium .,Laboratory of Hematology, GIGA-I3, University of Liège, Belgium
| | | | - K Martin Kortüm
- Department of Internal Medicine II, University Hospital of Wuerzburg, Germany
| | - Michael E O'Dwyer
- Department of Hematology, National University of Ireland Galway, Ireland
| | | | - Mascha Binder
- Department of Internal Medicine II, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra Maria Dold
- Department of Medicine I, Hematology, Oncology & Stem Cell Transplantation, Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Francesca Gay
- Department of Hematology-Oncology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Jill Corre
- Unit for Genomics in Myeloma, Institut Universitaire du Cancer - Oncopole, Toulouse, France
| | - Yves Beguin
- Department of Hematology, University Hospital of Liege, Belgium.,Laboratory of Hematology, GIGA-I3, University of Liège, Belgium
| | - Heinz Ludwig
- Wilhelminen Cancer Research Institute, Vienna, Austria
| | - Alessandra Larocca
- Department of Hematology-Oncology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Christoph Driessen
- Department of Oncology and Hematology, Cantonal Hospital St. Gallen, Switzerland
| | | | - Mario Boccadoro
- Department of Hematology-Oncology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Martin Gramatzki
- Division of Stem Cell Transplantation and Immunotherapy, University of Kiel, Germany
| | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Wuerzburg, Germany
| | - Michele Cavo
- Seragnoli 'Institute of Hematology, Bologna University School of Medicine, Italy
| | - Hartmut Goldschmidt
- Department of Hematology, Rheumatology and Oncology, University Hospital Heidelberg, Germany.,National Center for Tumor Diseases, Heidelberg Medical University, Germany
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Michel Delforge
- Department of Hematology, University Hospital Leuven, Belgium
| | - Holger W Auner
- Centre for Haematology, Hammersmith Hospital, Imperial College London, UK
| | - Evangelos Terpos
- School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Monika Engelhardt
- Department of Medicine I, Hematology, Oncology & Stem Cell Transplantation, Medical Center, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
74
|
Biancon G, Gimondi S, Vendramin A, Carniti C, Corradini P. Noninvasive Molecular Monitoring in Multiple Myeloma Patients Using Cell-Free Tumor DNA: A Pilot Study. J Mol Diagn 2018; 20:859-870. [PMID: 30165206 DOI: 10.1016/j.jmoldx.2018.07.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/25/2018] [Accepted: 07/09/2018] [Indexed: 12/11/2022] Open
Abstract
Novel treatments for multiple myeloma (MM) have increased rates of complete response, raising interest in more accurate methods to evaluate residual disease. Cell-free tumor DNA (cfDNA) analysis may represent a minimally invasive approach complementary to multiparameter flow cytometry (MFC) and molecular methods on bone marrow aspirates. A sequencing approach using the Ion Torrent Personal Genome Machine was applied to identify clonal IGH gene rearrangements in tumor plasma cells (PCs) and in serial plasma samples of 25 patients with MM receiving second-line therapy. The same clonal IGH rearrangement identified in tumor PCs was detected in paired plasma samples, and levels of IGH cfDNA correlated with outcome and mirrored tumor dynamics evaluated using conventional laboratory parameters. In addition, IGH cfDNA levels reflected the number of PCs enumerated by MFC immunophenotyping even in the complete response context. Patients determined by MFC to be free of minimal residual disease were characterized by low frequencies of tumor clonotypes in cfDNA and longer survival. This pilot study supports the clinical applicability of the noninvasive monitoring of tumor levels in plasma samples of patients with MM by IGH sequencing.
Collapse
Affiliation(s)
- Giulia Biancon
- Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy; Department of Hematology, Università degli Studi di Milano, Milano, Italy
| | - Silvia Gimondi
- Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy; Department of Hematology, Università degli Studi di Milano, Milano, Italy
| | - Antonio Vendramin
- Department of Hematology, Università degli Studi di Milano, Milano, Italy
| | - Cristiana Carniti
- Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | - Paolo Corradini
- Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy; Department of Hematology, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
75
|
Brandt A, Matschke J, Fehrle W, von Wenserski L, Bokemeyer C, Illerhaus G, Binder M. A significant proportion of patients with primary central nervous system lymphoma harbor clonal bone marrow B-cells. Leuk Lymphoma 2018; 60:334-340. [DOI: 10.1080/10428194.2018.1482538] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Anna Brandt
- Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob Matschke
- Department of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wilfried Fehrle
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa von Wenserski
- Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerald Illerhaus
- Department of Hematology and Oncology, Klinikum Stuttgart, Stuttgart, Germany
| | - Mascha Binder
- Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
76
|
Bagratuni T, Ntanasis-Stathopoulos I, Gavriatopoulou M, Mavrianou-Koutsoukou N, Liacos C, Patseas D, Kanellias N, Migkou M, Ziogas DC, Eleutherakis-Papaiakovou E, Roussou M, Fotiou D, Terpos E, Kastritis E, Dimopoulos MA. Detection of MYD88 and CXCR4 mutations in cell-free DNA of patients with IgM monoclonal gammopathies. Leukemia 2018; 32:2617-2625. [PMID: 30026568 PMCID: PMC6286389 DOI: 10.1038/s41375-018-0197-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/29/2018] [Accepted: 06/07/2018] [Indexed: 12/15/2022]
Abstract
Liquid biopsyis being integrated into cancer diagnostics with profound therapeutic implications. However, its role in Waldenström's Macroglobulinemia (WM) and IgM monoclonal gammopathies is still unclear. In this study, we evaluated the role of peripheral blood (PB) cell-free DNA (cfDNA) in characterizing the mutational status of MYD88 and CXCR4 of patients with IgM monoclonal gammopathies. Paired bone marrow (BM) tumor DNA (tDNA) and PB cfDNA samples from 98 patients (9 MGUS, 45 with WM in remission, 44 with smoldering WM, newly diagnosed or relapsed WM) and 10 controls with non-IgM monoclonal gammopathies were analyzed. Regarding MYD88L265P mutation, 76 patients had paired tDNA and cfDNA informative samples. Among patients with WM in remission, 65% harbored the MYD88L265P mutation, whereas the corresponding percentage among smoldering/newly diagnosed or relapsed WM was 92%. The overall concordance rate was 94% (72/76). For CXCR4 mutations, 65 patients had paired informative tDNA and cfDNA samples. The overall concordance rate was 90% (59/65). All controls had wild-type MYD88 and CXCR4. In conclusion, PB cfDNA is a useful, minimally invasive, cost-effective, and time-effective tool for the identification of the presence of MYD88 and CXCR4 mutations in patients with IgM monoclonal gammopathies avoiding unnecessary BM assessment.
Collapse
Affiliation(s)
- Tina Bagratuni
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nefeli Mavrianou-Koutsoukou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Christine Liacos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Patseas
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Kanellias
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Magdalini Migkou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios C Ziogas
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Maria Roussou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina Fotiou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
77
|
Mithraprabhu S, Sirdesai S, Chen M, Khong T, Spencer A. Circulating Tumour DNA Analysis for Tumour Genome Characterisation and Monitoring Disease Burden in Extramedullary Multiple Myeloma. Int J Mol Sci 2018; 19:ijms19071858. [PMID: 29937522 PMCID: PMC6073672 DOI: 10.3390/ijms19071858] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 01/06/2023] Open
Abstract
Mutational characterisation in extramedullary multiple myeloma (EM-MM) patients is challenging due to inaccessible EM plasmacytomas, unsafe nature of multiple biopsies and the spatial and temporal genomic heterogeneity apparent in MM (Graphical abstract). Conventional monitoring of disease burden is through serum markers and PET-CT, however these modalities are sometimes inadequate (serum markers), not performed in a timely manner (PET-CT) and uninformative for identifying mutations driving disease progression. DNA released into the blood by tumour cells (ctDNA) contains the predominant clones derived from the multiple disease foci. Blood-derived ctDNA can, therefore, provide a holistic illustration of the major drivers of disease progression. In this report, the utility of ctDNA, as an adjunct to currently available modalities in EM-MM, is presented for a patient with EM and oligosecretory (OS) disease. Whole exome sequencing of contemporaneously acquired tumour tissue and matched ctDNA samples revealed the presence of spatial and temporal genetic heterogeneity and the identification of pathways associated with drug resistance. Longitudinal monitoring of plasma samples revealed that ctDNA can be utilised to define the dynamic clonal evolution co-existent with disease progression and as an adjunct non-invasive marker of tumour burden.
Collapse
Affiliation(s)
- Sridurga Mithraprabhu
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital⁻Monash University, Melbourne 3004, Australia.
- Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne 3004, Australia.
| | - Shreerang Sirdesai
- Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne 3004, Australia.
| | - Maoshan Chen
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital⁻Monash University, Melbourne 3004, Australia.
- Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne 3004, Australia.
| | - Tiffany Khong
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital⁻Monash University, Melbourne 3004, Australia.
- Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne 3004, Australia.
| | - Andrew Spencer
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital⁻Monash University, Melbourne 3004, Australia.
- Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne 3004, Australia.
- Department of Clinical Haematology, Monash University, Clayton 3800, Australia.
| |
Collapse
|
78
|
Soekojo CY, de Mel S, Ooi M, Yan B, Chng WJ. Potential Clinical Application of Genomics in Multiple Myeloma. Int J Mol Sci 2018; 19:ijms19061721. [PMID: 29890777 PMCID: PMC6032230 DOI: 10.3390/ijms19061721] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/02/2018] [Accepted: 06/07/2018] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma is a heterogeneous disease with different characteristics, and genetic aberrations play important roles in this heterogeneity. Studies have shown that these genetic aberrations are crucial in prognostication and response assessment; recent efforts have focused on their possible therapeutic implications. Despite many emerging studies being published, the best way to incorporate these results into clinical practice remains unclear. In this review paper we describe the different genomic techniques available, including the latest advancements, and discuss the potential clinical application of genomics in multiple myeloma.
Collapse
Affiliation(s)
- Cinnie Yentia Soekojo
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
| | - Sanjay de Mel
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
| | - Melissa Ooi
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
| | - Benedict Yan
- Department of Laboratory Medicine, National University Hospital, National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore.
| | - Wee Joo Chng
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore,14 Medical Drive, Singapore 117599, Singapore.
| |
Collapse
|
79
|
Genomic discovery and clonal tracking in multiple myeloma by cell-free DNA sequencing. Leukemia 2018; 32:1838-1841. [PMID: 29749395 DOI: 10.1038/s41375-018-0115-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/05/2018] [Accepted: 03/12/2018] [Indexed: 02/07/2023]
|
80
|
Stewart CM, Kothari PD, Mouliere F, Mair R, Somnay S, Benayed R, Zehir A, Weigelt B, Dawson SJ, Arcila ME, Berger MF, Tsui DW. The value of cell-free DNA for molecular pathology. J Pathol 2018; 244:616-627. [PMID: 29380875 DOI: 10.1002/path.5048] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/23/2018] [Accepted: 01/25/2018] [Indexed: 02/06/2023]
Abstract
Over the past decade, advances in molecular biology and genomics techniques have revolutionized the diagnosis and treatment of cancer. The technological advances in tissue profiling have also been applied to the study of cell-free nucleic acids, an area of increasing interest for molecular pathology. Cell-free nucleic acids are released from tumour cells into the surrounding body fluids and can be assayed non-invasively. The repertoire of genomic alterations in circulating tumour DNA (ctDNA) is reflective of both primary tumours and distant metastatic sites, and ctDNA can be sampled multiple times, thereby overcoming the limitations of the analysis of single biopsies. Furthermore, ctDNA can be sampled regularly to monitor response to treatment, to define the evolution of the tumour genome, and to assess the acquisition of resistance and minimal residual disease. Recently, clinical ctDNA assays have been approved for guidance of therapy, which is an exciting first step in translating cell-free nucleic acid research tests into clinical use for oncology. In this review, we discuss the advantages of cell-free nucleic acids as analytes in different body fluids, including blood plasma, urine, and cerebrospinal fluid, and their clinical applications in solid tumours and haematological malignancies. We will also discuss practical considerations for clinical deployment, such as preanalytical factors and regulatory requirements. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Caitlin M Stewart
- Marie-José and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Prachi D Kothari
- Marie-José and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Pediatric Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Florent Mouliere
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.,Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Richard Mair
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.,Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, UK.,Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, UK
| | - Saira Somnay
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sarah-Jane Dawson
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia.,Centre for Cancer Research, University of Melbourne, Victoria, Australia
| | - Maria E Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael F Berger
- Marie-José and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dana Wy Tsui
- Marie-José and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
81
|
Pugh TJ. Circulating Tumour DNA for Detecting Minimal Residual Disease in Multiple Myeloma. Semin Hematol 2018; 55:38-40. [PMID: 29759151 DOI: 10.1053/j.seminhematol.2018.03.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 03/05/2018] [Indexed: 12/23/2022]
Abstract
Circulating tumor DNA faithfully recapitulates somatic mutations detected in bone marrow aspirates from patients with newly diagnosed or relapsed or recurrent myeloma. Extending these methods to enable detection of minimal residual disease will require increased sensitivity and breadth of genomic assays to maximize information content from small quantities of cell-free DNA; as well as definition of a clinically meaningful ctDNA concentration in comparison with conventional bone marrow cell-count thresholds. This review describes the use of cell-free DNA sequencing in myeloma to date, identifies challenges associated with pushing limit of detection of these assays into the realm of detecting minimal residual disease, and describes potential strategies to overcome these challenges.
Collapse
Affiliation(s)
- Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Ontario Institute for Cancer Research, Toronto, ON, Canada.
| |
Collapse
|
82
|
Waldschmidt JM, Anand P, Knoechel B, Lohr JG. Comprehensive characterization of circulating and bone marrow-derived multiple myeloma cells at minimal residual disease. Semin Hematol 2018; 55:33-37. [PMID: 29759150 DOI: 10.1053/j.seminhematol.2018.02.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 02/21/2018] [Indexed: 01/04/2023]
Abstract
The presence or absence of minimal residual disease (MRD) in patients with multiple myeloma (MM) has emerged as a useful marker to determine the depth of remission. MRD negativity as an endpoint has been shown to be associated with improved progression-free survival in many studies. MRD detection is therefore part of numerous clinical trial protocols for MM. At the present time, two methodologies are most widely accepted for MRD detection: (1) multicolor flow cytometry and (2) next-generation sequencing-based clonotype detection. While both of those methodologies enable accurate quantification of MRD in the bone marrow (BM), with sensitivity as low as 10-5 to 10-6, there are several limitations to these methods. First, these approaches reveal the presence or absence of MRD but provide limited molecular information about MM. More comprehensive characterization of MM cells at the MRD stage may identify molecular mechanisms of drug resistance. Second, MRD detection in the BM is typically performed at one time point only, but more frequent detection may define the duration of the MRD status and thus refine its prognostic value. Third, less-invasive approaches that avoid the discomfort and risk associated with BM biopsy would be highly desirable, especially in elderly or frail patients. "Liquid biopsy" for the detection and characterization of circulating MM cells may address these issues. Although MRD detection in the peripheral blood at the same sensitivity as in the BM may be challenging, the identification of patients who do not achieve MRD negativity might reduce the need for BM biopsies. Here, we give an overview of approaches that have been described to detect and characterize MM cells when they occur at very low frequencies in the peripheral blood or in the BM, emphasizing recently described next-generation sequencing approaches for more comprehensive characterization of circulating MM cells.
Collapse
Affiliation(s)
- Johannes M Waldschmidt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Praveen Anand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Birgit Knoechel
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Jens G Lohr
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.
| |
Collapse
|
83
|
Gerber B, Manzoni M, Spina V, Bruscaggin A, Lionetti M, Fabris S, Barbieri M, Ciceri G, Pompa A, Forestieri G, Lerch E, Servida P, Bertoni F, Zucca E, Ghielmini M, Cortelezzi A, Cavalli F, Stussi G, Baldini L, Rossi D, Neri A. Circulating tumor DNA as a liquid biopsy in plasma cell dyscrasias. Haematologica 2018; 103:e245-e248. [PMID: 29472358 DOI: 10.3324/haematol.2017.184358] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Bernhard Gerber
- Division of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Martina Manzoni
- Department of Oncology and Hemato-oncology, University of Milan, Italy
| | - Valeria Spina
- Institute of Oncology Research, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Alessio Bruscaggin
- Institute of Oncology Research, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Marta Lionetti
- Department of Oncology and Hemato-oncology, University of Milan, Italy
| | - Sonia Fabris
- Hematology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marzia Barbieri
- Hematology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gabriella Ciceri
- Department of Oncology and Hemato-oncology, University of Milan, Italy
| | - Alessandra Pompa
- Hematology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gabriela Forestieri
- Institute of Oncology Research, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Erika Lerch
- Division of Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Paolo Servida
- Division of Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Emanuele Zucca
- Division of Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Michele Ghielmini
- Division of Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Agostino Cortelezzi
- Department of Oncology and Hemato-oncology, University of Milan, Italy.,Hematology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Franco Cavalli
- Institute of Oncology Research, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland.,Division of Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Georg Stussi
- Division of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Luca Baldini
- Department of Oncology and Hemato-oncology, University of Milan, Italy.,Hematology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Davide Rossi
- Division of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland .,Institute of Oncology Research, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Antonino Neri
- Department of Oncology and Hemato-oncology, University of Milan, Italy .,Hematology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
84
|
Zajec M, Jacobs JFM, Groenen PJTA, de Kat Angelino CM, Stingl C, Luider TM, De Rijke YB, VanDuijn MM. Development of a Targeted Mass-Spectrometry Serum Assay To Quantify M-Protein in the Presence of Therapeutic Monoclonal Antibodies. J Proteome Res 2018; 17:1326-1333. [DOI: 10.1021/acs.jproteome.7b00890] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Marina Zajec
- Department
of Neurology, Erasmus MC University Medical Center, Rotterdam 3015 GE, The Netherlands
- Department
of Clinical Chemistry, Erasmus MC University Medical Center, Rotterdam 3015 GE, The Netherlands
| | - Joannes F. M. Jacobs
- Department
of Laboratory Medicine, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands
| | | | - Corrie M. de Kat Angelino
- Department
of Laboratory Medicine, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands
| | - Christoph Stingl
- Department
of Neurology, Erasmus MC University Medical Center, Rotterdam 3015 GE, The Netherlands
| | - Theo M. Luider
- Department
of Neurology, Erasmus MC University Medical Center, Rotterdam 3015 GE, The Netherlands
| | - Yolanda B. De Rijke
- Department
of Clinical Chemistry, Erasmus MC University Medical Center, Rotterdam 3015 GE, The Netherlands
| | - Martijn M. VanDuijn
- Department
of Neurology, Erasmus MC University Medical Center, Rotterdam 3015 GE, The Netherlands
| |
Collapse
|
85
|
Abstract
Circulating tumor DNA (ctDNA) analysis is currently gaining momentum as an innovative methodology for characterizing the tumor genome and monitoring therapeutic efficacy in the multifocal, genetically and spatially heterogeneous plasma cell malignancy, multiple myeloma (MM). Circulating cell-free DNA (cfDNA), which consists of a combination of DNA derived from both tumor and normal cells, is present in extracellular bodily fluids. The presence of ctDNA within this admixture has been demonstrated recently in MM. In this chapter, we describe the routinely utilized methodology for the extraction and longitudinal analysis of specific mutations present in ctDNA derived from peripheral blood plasma of MM patients.
Collapse
Affiliation(s)
- Sridurga Mithraprabhu
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital-Monash University, Melbourne, VIC, Australia
- Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia
| | - Andrew Spencer
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital-Monash University, Melbourne, VIC, Australia.
- Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia.
- Department of Clinical Haematology, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
86
|
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
|
87
|
Mithraprabhu S, Spencer A. Circulating tumour DNA analysis in multiple myeloma. Oncotarget 2017; 8:90610-90611. [PMID: 29207579 PMCID: PMC5710860 DOI: 10.18632/oncotarget.21595] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Indexed: 12/12/2022] Open
Affiliation(s)
- Sridurga Mithraprabhu
- Andrew Spencer: Australian Centre for Blood Diseases, Monash University, Malignant Haematology & Stem Cell Transplantation, Alfred Hospital and Department of Clinical Haematology, Monash University, Melbourne, Australia
| | - Andrew Spencer
- Andrew Spencer: Australian Centre for Blood Diseases, Monash University, Malignant Haematology & Stem Cell Transplantation, Alfred Hospital and Department of Clinical Haematology, Monash University, Melbourne, Australia
| |
Collapse
|
88
|
Kubaczkova V, Vrabel D, Sedlarikova L, Besse L, Sevcikova S. Cell-free DNA - Minimally invasive marker of hematological malignancies. Eur J Haematol 2017; 99:291-299. [PMID: 28692178 DOI: 10.1111/ejh.12925] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2017] [Indexed: 12/13/2022]
Abstract
Although tumor cells are the most reliable source of tumor DNA, biopsy of the tumor is an invasive procedure that should be avoided in some cases. The main limitation of any biopsy is sampling of one tumor site, which may not represent all malignant clones due to the heterogeneity of the tumor. These clones respond to treatment differently and thus directly influence survival of the patient. Circulating cell-free DNA (cfDNA) is released from multiple tumor sites, reflects overall heterogeneity of the tumor, and correlates with its progression. Detection of tumor-specific genetic and epigenetic aberrations in cfDNA could have a direct impact on molecular diagnosis, prognosis, follow-up of disease, monitoring of minimal residual disease, and response to treatment. While most cfDNA data are still experimental, they are very promising. This review focuses on cfDNA in hematological malignancies.
Collapse
Affiliation(s)
- Veronika Kubaczkova
- Faculty of Medicine, Babak Myeloma Group, Department of Pathological Physiology, Masaryk University, Brno, Czech Republic
| | - David Vrabel
- Department of Chemistry and Toxicology, Veterinary Research Institute, Brno, Czech Republic
| | - Lenka Sedlarikova
- Faculty of Medicine, Babak Myeloma Group, Department of Pathological Physiology, Masaryk University, Brno, Czech Republic
| | - Lenka Besse
- Experimental Oncology and Hematology, Department of Oncology and Hematology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Sabina Sevcikova
- Faculty of Medicine, Babak Myeloma Group, Department of Pathological Physiology, Masaryk University, Brno, Czech Republic
| |
Collapse
|