1
|
Paiva B, Shi Q, Puig N, Cedena MT, Orfao A, Durie BGM, Munshi NC, San-Miguel J. Opportunities and challenges for MRD assessment in the clinical management of multiple myeloma. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01017-x. [PMID: 40195455 DOI: 10.1038/s41571-025-01017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2025] [Indexed: 04/09/2025]
Abstract
Measurable residual disease (MRD) assessment is, from the methodological point of view, ready for prime time in multiple myeloma (MM). Abundant evidence underscores the value of MRD status determined using highly sensitive next-generation flow cytometry and next-generation sequencing tests in evaluating response to treatment and, therefore, prognosis in patients with this disease. MRD response assessment and monitoring might present a range of opportunities for individualized patient management. Moreover, the considerable amounts of high-quality and standardized MRD data generated in clinical trials have led to the acceptance of MRD negativity as an early end point for accelerated regulatory approval of treatments for MM. The data leave no doubt that the efficacy of new regimens in inducing deeper and durable MRD-negative responses is connected with prolonged survival. Yet, several evidential, technical and practical challenges continue to limit the implementation of MRD-guided treatment strategies in routine practice, and the use of MRD as a surrogate end point remains controversial to some. In this Review, we draw on past and present research to propose opportunities for overcoming some of these challenges, and to accelerate the use of MRD assessment for improved clinical management of patients with MM.
Collapse
Affiliation(s)
- Bruno Paiva
- Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Cancer Center Clínica Universidad de Navarra, Pamplona, Spain.
| | - Qian Shi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Noemi Puig
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cancer (IBMCC, USAL-CSIC), CIBER-ONC number CB16/12/00233, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Maria-Teresa Cedena
- Instituto de Investigación imas12, CIBER-ONC number CB16/12/00369, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Alberto Orfao
- Department of Medicine and Cytometry Service, Centro de Investigación del Cancer (IBMCC, USAL-CSIC), CIBER-ONC number CB16/12/00400, University of Salamanca, Salamanca, Spain
| | - Brian G M Durie
- Division of Hematology/Oncology, Cedars-Sinai Outpatient Cancer Center, Los Angeles, CA, USA
| | - Nikhil C Munshi
- Basic and Correlative Science, Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, VA Boston Healthcare System, Boston, MA, USA
| | - Jesús San-Miguel
- Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Cancer Center Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
2
|
Zhang D, Chen X, Lin J, Jiang S, Fan M, Liu N, Huang Z, Wang J. Ultrasensitive Detection of Circulating Plasma Cells Using Surface-Enhanced Raman Spectroscopy and Machine Learning for Multiple Myeloma Monitoring. Anal Chem 2025; 97:4101-4110. [PMID: 39854732 DOI: 10.1021/acs.analchem.4c06244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
Multiple myeloma is a hematologic malignancy characterized by the proliferation of abnormal plasma cells in the bone marrow. Despite therapeutic advancements, there remains a critical need for reliable, noninvasive methods to monitor multiple myeloma. Circulating plasma cells (CPCs) in peripheral blood are robust and independent prognostic markers, but their detection is challenging due to their low abundance. Next-generation flow cytometry is commonly used for CPC detection but is not performed in routine clinical practice because it requires expensive instruments, is costly, and time-consuming. This study introduces a cost-effective, rapid surface-enhanced Raman spectroscopy (SERS) assay leveraging gold-deposited magnetic nanoparticles and plasmonic nanoparticles functionalized with anti-CD138 and anti-CD38 antibodies for detecting CPCs in peripheral blood samples. A portable optical device was used for signal recording, enhancing the potential for point-of-care applications. The developed assay is highly sensitive and specific, capable of detecting as few as one or two cells. The application of machine learning algorithms to SERS signal analysis yielded area under the curve values ranging from 0.90 to 0.95, demonstrating excellent performance in differentiating multiple myeloma patients from healthy donors. This SERS method provides a sensitive and accessible way for CPC detection, showing significant potential for multiple myeloma diagnosis, treatment monitoring, and prognosis prediction.
Collapse
Affiliation(s)
- Dechun Zhang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, Fujian 350117, China
| | - Xianling Chen
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Jia Lin
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Fujian Medical University Center of Translational Hematology, Fuzhou, Fujian 350001, China
| | - Shiyan Jiang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, Fujian 350117, China
| | - Min Fan
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, Fujian 350117, China
| | - Nenrong Liu
- Fujian Provincial Collaborative Innovation Center for Advanced High-Field Superconducting Materials and Engineering, Fujian Provincial Solar Energy Conversion and Energy Storage Engineering Technology Research Center, College of Physics and Energy, Fujian Normal University, Fuzhou, Fujian 350117, China
| | - Zufang Huang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, Fujian 350117, China
| | - Jing Wang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, Fujian 350117, China
| |
Collapse
|
3
|
Gutiérrez-González A, Del Hierro I, Cariaga-Martínez AE. Advancements in Multiple Myeloma Research: High-Throughput Sequencing Technologies, Omics, and the Role of Artificial Intelligence. BIOLOGY 2024; 13:923. [PMID: 39596878 PMCID: PMC11592186 DOI: 10.3390/biology13110923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/01/2024] [Accepted: 11/10/2024] [Indexed: 11/29/2024]
Abstract
Multiple myeloma is a complex and challenging type of blood cancer that affects plasma cells in the bone marrow. In recent years, the development of advanced research techniques, such as omics approaches-which involve studying large sets of biological data like genes and proteins-and high-throughput sequencing technologies, has allowed researchers to analyze vast amounts of genetic information rapidly and gain new insights into the disease. Additionally, the advent of artificial intelligence tools has accelerated data analysis, enabling more accurate predictions and improved treatment strategies. This review aims to highlight recent research advances in multiple myeloma made possible by these novel techniques and to provide guidance for researchers seeking effective approaches in this field.
Collapse
Affiliation(s)
| | | | - Ariel Ernesto Cariaga-Martínez
- DS-OMICS—Data Science and Omics, AI-Driven Biomedicine Group, Universidad Alfonso X el Sabio, 28619 Villanueva de la Cañada, Spain; (A.G.-G.); (I.D.H.)
| |
Collapse
|
4
|
Schroers-Martin JG, Alizadeh AA. Cell-Free DNA in Hematologic Malignancies. JCO Oncol Pract 2024; 20:1491-1499. [PMID: 39531844 DOI: 10.1200/op-24-00648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 08/27/2024] [Indexed: 11/16/2024] Open
Abstract
Liquid biopsy techniques using cell-free DNA (cfDNA) play an increasingly important role in the characterization and surveillance of solid tumors. For blood cancers, molecular response assessment techniques using circulating malignant cells or bone marrow aspirates are well established in clinical care. However, cfDNA has an emerging role in hematology as well, with the opportunity for disease assessment and quantification independent of circulating disease burden or invasive biopsies. In this review, we discuss key technologies and clinical data for the utilization of cfDNA in lymphomas, myeloma, and leukemias.
Collapse
Affiliation(s)
- Joseph G Schroers-Martin
- Department of Medicine, Divisions of Hematology & Oncology, Stanford University Medical Center, Stanford, CA
| | - Ash A Alizadeh
- Department of Medicine, Divisions of Hematology & Oncology, Stanford University Medical Center, Stanford, CA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA
- Stanford Cancer Institute, Stanford University, Stanford, CA
| |
Collapse
|
5
|
Schultheiß C, Paschold L, Mohebiany AN, Escher M, Kattimani YM, Müller M, Schmidt-Barbo P, Mensa-Vilaró A, Aróstegui JI, Boursier G, de Moreuil C, Hautala T, Willscher E, Jonas H, Chinchuluun N, Grosser B, Märkl B, Klapper W, Oommen PT, Gössling K, Hoffmann K, Tiegs G, Czernilofsky F, Dietrich S, Freeman A, Schwartz DM, Waisman A, Aksentijevich I, Binder M. A20 haploinsufficiency disturbs immune homeostasis and drives the transformation of lymphocytes with permissive antigen receptors. SCIENCE ADVANCES 2024; 10:eadl3975. [PMID: 39167656 PMCID: PMC11338232 DOI: 10.1126/sciadv.adl3975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 07/15/2024] [Indexed: 08/23/2024]
Abstract
Genetic TNFAIP3 (A20) inactivation is a classical somatic lymphoma lesion and the genomic trait in haploinsufficiency of A20 (HA20). In a cohort of 34 patients with HA20, we show that heterozygous TNFAIP3 loss skews immune repertoires toward lymphocytes with classical self-reactive antigen receptors typically found in B and T cell lymphomas. This skewing was mediated by a feed-forward tumor necrosis factor (TNF)/A20/nuclear factor κB (NF-κB) loop that shaped pre-lymphoma transcriptome signatures in clonally expanded B (CD81, BACH2, and NEAT1) or T (GATA3, TOX, and PDCD1) cells. The skewing was reversed by anti-TNF treatment but could also progress to overt lymphoma. Analysis of conditional TNFAIP3 knock-out mice reproduced the wiring of the TNF/A20/NF-κB signaling axis with permissive antigen receptors and suggested a distinct regulation in B and T cells. Together, patients with the genetic disorder HA20 provide an exceptional window into A20/TNF/NF-κB-mediated control of immune homeostasis and early steps of lymphomagenesis that remain clinically unrecognized.
Collapse
Affiliation(s)
- Christoph Schultheiß
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Lisa Paschold
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Alma Nazlie Mohebiany
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Moritz Escher
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Yogita Mallu Kattimani
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Melanie Müller
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Paul Schmidt-Barbo
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg, Germany
| | - Anna Mensa-Vilaró
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Juan Ignacio Aróstegui
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
| | - Guilaine Boursier
- Department of molecular and cytogenomics, Rare and Autoinflammatory Diseases Laboratory, CHU Montpellier, IRMB, University of Montpellier, INSERM, CEREMAIA, Montpellier, France
| | - Claire de Moreuil
- Department of Internal Medicine, CHU Brest, Université de Bretagne Occidentale, Brest, France
| | - Timo Hautala
- Research Unit of Biomedicine, University of Oulu and Department of Internal Medicine, Oulu University Hospital, Oulu, Finland
| | - Edith Willscher
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Hanna Jonas
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Namuun Chinchuluun
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Bianca Grosser
- Institute for Pathology, University Medical Center Augsburg, Augsburg, Germany
| | - Bruno Märkl
- Institute for Pathology, University Medical Center Augsburg, Augsburg, Germany
| | - Wolfram Klapper
- Institute of Pathology, Hematopathology Section, and Lymph Node Registry, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Prasad Thomas Oommen
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Center for Child and Adolescent Health, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Katharina Gössling
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Center for Child and Adolescent Health, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Katrin Hoffmann
- Institute for Human Genetics and Molecular Biology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Gisa Tiegs
- Institute for Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix Czernilofsky
- Department of Medicine V, Hematology, Oncology, and Rheumatology, University of Heidelberg, Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
| | - Sascha Dietrich
- Department of Medicine V, Hematology, Oncology, and Rheumatology, University of Heidelberg, Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematolgy, Oncology, and Immunolgy, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Alexandra Freeman
- Laboratory of Clinical Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Daniella M. Schwartz
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg, Germany
| |
Collapse
|
6
|
Quivoron C, Michot JM, Danu A, Lecourt H, Saada V, Saleh K, Vergé V, Cotteret S, Bernard OA, Ribrag V. Sensitivity, specificity, and accuracy of molecular profiling on circulating cell-free DNA in refractory or relapsed multiple myeloma patients, results of MM-EP1 study. Leuk Lymphoma 2024; 65:789-799. [PMID: 38433500 DOI: 10.1080/10428194.2024.2320258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/13/2024] [Indexed: 03/05/2024]
Abstract
As a promising alternative to bone marrow aspiration (BMA), mutational profiling on blood-derived circulating cell-free tumor DNA (cfDNA) is a harmless and simple technique to monitor molecular response and treatment resistance of patients with refractory/relapsed multiple myeloma (R/R MM). We evaluated the sensitivity and specificity of cfDNA compared to BMA CD138 positive myeloma plasma cells (PCs) in a series of 45 R/R MM patients using the 29-gene targeted panel (AmpliSeq) NGS. KRAS, NRAS, FAM46C, DIS3, and TP53 were the most frequently mutated genes. The average sensitivity and specificity of cfDNA detection were 65% and 97%, respectively. The concordance per gene between the two samples was good to excellent according to Cohen's κ coefficients interpretation. An increased number of mutations detected in cfDNA were associated with a decreased overall survival. In conclusion, we demonstrated cfDNA NGS analysis feasibility and accuracy in R/R MM patients who may benefit from early phase clinical trial.
Collapse
Affiliation(s)
- C Quivoron
- Translational Hematology Laboratory, AMMICa, INSERM US23/CNRS UAR3655, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM U1170, Université Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - J-M Michot
- INSERM U1170, Université Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
- Drug Development Department: Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
| | - A Danu
- Hematology Department, Gustave Roussy, Villejuif, France
| | - H Lecourt
- Translational Hematology Laboratory, AMMICa, INSERM US23/CNRS UAR3655, Gustave Roussy Cancer Campus, Villejuif, France
| | - V Saada
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - K Saleh
- Hematology Department, Gustave Roussy, Villejuif, France
| | - V Vergé
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - S Cotteret
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - O A Bernard
- INSERM U1170, Université Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - V Ribrag
- Translational Hematology Laboratory, AMMICa, INSERM US23/CNRS UAR3655, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM U1170, Université Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
- Drug Development Department: Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
- Hematology Department, Gustave Roussy, Villejuif, France
| |
Collapse
|
7
|
Wei JX, Shastri A, Sica RA, Mantzaris I, Kornblum N, Shah U, Janakiram M, Gritsman K, Verma A, Goldfinger M, Cooper D, Shah N. Impact of race and ethnicity on early mortality in multiple myeloma: a SEER analysis. Haematologica 2024; 109:1480-1486. [PMID: 37881838 PMCID: PMC11063841 DOI: 10.3324/haematol.2023.283304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
Over the past two decades, there have been significant advances in the treatment of multiple myeloma which has led to an improvement in overall survival.1,2 However, a notable proportion of patients continue to experience early mortality (EM), defined as 2 years from the time of diagnosis. This raises the possibility that improvements in myeloma survival have not extended equally to all groups. Using the latest data drawn from the Surveillance Epidemiology and End Results database of patients in the United States spanning 2000-2019, we study impact of important sociodemographic factors on EM. Through regression modeling, we demonstrate that patients diagnosed from 2000-2005, of older age, male sex, and of certain racial minority status (non-Hispanic Black and Hispanic) have higher odds of EM. Of these factors, minority status contributed to worse 2-year overall survival as well. We evaluate whether income, as a surrogate to access to care, could potentially explain this finding, but find that race has a distinct relationship with EM that is not modified by income. This is further reinforced by subgroup analysis. After characterizing groups vulnerable to EM, we examine reasons for these disparities and potential avenues to address them.
Collapse
Affiliation(s)
- John X Wei
- Department of Medicine, Montefiore/Albert Einstein College of Medicine, New York
| | - Aditi Shastri
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - R Alejandro Sica
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Ioannis Mantzaris
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Noah Kornblum
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Urvi Shah
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Murali Janakiram
- Division of Hematologic Malignancies, City of Hope National Medical Center, Duarte, CA
| | - Kira Gritsman
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Amit Verma
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Mendel Goldfinger
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Dennis Cooper
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Nishi Shah
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York.
| |
Collapse
|
8
|
Tembhare PR, Sriram H, Khanka T, Gawai S, Bagal B, Ghogale SG, Deshpande N, Girase K, Patil J, Hasan SK, Shetty D, Ghosh K, Chatterjee G, Rajpal S, Patkar NV, Jain H, Punatar S, Gokarn A, Nayak L, Mirgh S, Jindal N, Sengar M, Khattry N, Subramanian PG, Gujral S. Circulating tumor plasma cells and peripheral blood measurable residual disease assessment in multiple myeloma patients not planned for upfront transplant. Hemasphere 2024; 8:e63. [PMID: 38566804 PMCID: PMC10983024 DOI: 10.1002/hem3.63] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/16/2024] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Circulating tumor plasma cells (CTPCs) provide a noninvasive alternative for measuring tumor burden in newly diagnosed multiple myeloma (NDMM). Moreover, measurable residual disease (MRD) assessment in peripheral blood (PBMRD) can provide an ideal alternative to bone marrow MRD, which is limited by its painful nature and technical challenges. However, the clinical significance of PBMRD in NDMM still remains uncertain. Additionally, data on CTPC in NDMM patients not treated with transplant are scarce. We prospectively studied CTPC and PBMRD in 141 NDMM patients using highly sensitive multicolor flow cytometry (HS-MFC). PBMRD was monitored at the end of three cycles (PBMRD1) and six cycles (PBMRD2) of chemotherapy in patients with detectable baseline CTPC. Patients received bortezomib-based triplet therapy and were not planned for an upfront transplant. Among baseline risk factors, CTPC ≥ 0.01% was independently associated with poor progression-free survival (PFS) (hazard ratio [HR] = 2.77; p = 0.0047) and overall survival (OS) (HR = 2.9; p = 0.023) on multivariate analysis. In patients with detectable baseline CTPC, undetectable PBMRD at both subsequent time points was associated with longer PFS (HR = 0.46; p = 0.0037), whereas detectable PBMRD at any time point was associated with short OS (HR = 3.25; p = 0.004). Undetectable combined PBMRD (PBMRD1 and PBMRD2) outperformed the serum-immunofixation-based response. On multivariate analysis, detectable PBMRD at any time point was independently associated with poor PFS (HR = 2.0; p = 0.025) and OS (HR = 3.97; p = 0.013). Thus, our findings showed that CTPC and PBMRD assessment using HS-MFC provides a robust, noninvasive biomarker for NDMM patients not planned for an upfront transplant. Sequential PBMRD monitoring has great potential to improve the impact of the existing risk stratification and response assessment models.
Collapse
Affiliation(s)
- Prashant R. Tembhare
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Harshini Sriram
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Twinkle Khanka
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Sanghamitra Gawai
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Bhausaheb Bagal
- Department of Medical Oncology, Tata Memorial CentreHBNI UniversityMumbaiMaharashtraIndia
| | - Sitaram G. Ghogale
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Nilesh Deshpande
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Karishma Girase
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Jagruti Patil
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Syed Khaizer Hasan
- Hasan Laboratory, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Dhanalaxmi Shetty
- Department of Cancer Cytogenetics, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Kinjalka Ghosh
- Department of Biochemistry, Tata Memorial CentreHBNI UniversityMumbaiMaharashtraIndia
| | - Gaurav Chatterjee
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Sweta Rajpal
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Nikhil V. Patkar
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Hasmukh Jain
- Department of Medical Oncology, Tata Memorial CentreHBNI UniversityMumbaiMaharashtraIndia
| | - Sachin Punatar
- Bone Marrow Transplant Unit, Department of Medical Oncology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Anant Gokarn
- Bone Marrow Transplant Unit, Department of Medical Oncology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Lingaraj Nayak
- Bone Marrow Transplant Unit, Department of Medical Oncology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Sumeet Mirgh
- Bone Marrow Transplant Unit, Department of Medical Oncology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Nishant Jindal
- Bone Marrow Transplant Unit, Department of Medical Oncology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Manju Sengar
- Department of Medical Oncology, Tata Memorial CentreHBNI UniversityMumbaiMaharashtraIndia
| | - Navin Khattry
- Bone Marrow Transplant Unit, Department of Medical Oncology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Papagudi G. Subramanian
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Sumeet Gujral
- Department of Pathology, Tata Memorial Hospital, Tata Memorial CentreHBNI UniversityMumbaiMaharashtraIndia
| |
Collapse
|
9
|
Mithraprabhu S, Reynolds J, Quach H, Horvath N, Kerridge I, Khong T, Durie BG, Spencer A. Circulating tumor DNA and bone marrow minimal residual disease negativity confers superior outcome for multiple myeloma patients. Haematologica 2024; 109:974-978. [PMID: 37767561 PMCID: PMC10905075 DOI: 10.3324/haematol.2023.283831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Affiliation(s)
- Sridurga Mithraprabhu
- Australian Centre for Blood Diseases, Alfred Health - Monash University, Melbourne, Australia; Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne.
| | - John Reynolds
- Australian Centre for Blood Diseases, Alfred Health - Monash University, Melbourne, Australia; Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne
| | - Hang Quach
- St.Vincent's Hospital, University of Melbourne
| | | | | | - Tiffany Khong
- Australian Centre for Blood Diseases, Alfred Health - Monash University, Melbourne, Australia; Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne
| | - Brian Gm Durie
- Cedars-Sinai Comprehensive Cancer Center, Los Angeles, California
| | - Andrew Spencer
- Australian Centre for Blood Diseases, Alfred Health - Monash University, Melbourne, Australia; Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne.
| |
Collapse
|
10
|
Märkl F, Schultheiß C, Ali M, Chen SS, Zintchenko M, Egli L, Mietz J, Chijioke O, Paschold L, Spajic S, Holtermann A, Dörr J, Stock S, Zingg A, Läubli H, Piseddu I, Anz D, Minden MDV, Zhang T, Nerreter T, Hudecek M, Minguet S, Chiorazzi N, Kobold S, Binder M. Mutation-specific CAR T cells as precision therapy for IGLV3-21 R110 expressing high-risk chronic lymphocytic leukemia. Nat Commun 2024; 15:993. [PMID: 38307904 PMCID: PMC10837166 DOI: 10.1038/s41467-024-45378-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/22/2024] [Indexed: 02/04/2024] Open
Abstract
The concept of precision cell therapy targeting tumor-specific mutations is appealing but requires surface-exposed neoepitopes, which is a rarity in cancer. B cell receptors (BCR) of mature lymphoid malignancies are exceptional in that they harbor tumor-specific-stereotyped sequences in the form of point mutations that drive self-engagement of the BCR and autologous signaling. Here, we use a BCR light chain neoepitope defined by a characteristic point mutation (IGLV3-21R110) for selective targeting of a poor-risk subset of chronic lymphocytic leukemia (CLL) with chimeric antigen receptor (CAR) T cells. We develop murine and humanized CAR constructs expressed in T cells from healthy donors and CLL patients that eradicate IGLV3-21R110 expressing cell lines and primary CLL cells, but neither cells expressing the non-pathogenic IGLV3-21G110 light chain nor polyclonal healthy B cells. In vivo experiments confirm epitope-selective cytolysis in xenograft models in female mice using engrafted IGLV3-21R110 expressing cell lines or primary CLL cells. We further demonstrate in two humanized mouse models lack of cytotoxicity towards human B cells. These data provide the basis for advanced approaches of resistance-preventive and biomarker-guided cellular targeting of functionally relevant lymphoma driver mutations sparing normal B cells.
Collapse
Affiliation(s)
- Florian Märkl
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Christoph Schultheiß
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Murtaza Ali
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Shih-Shih Chen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | | | - Lukas Egli
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Juliane Mietz
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Obinna Chijioke
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Lisa Paschold
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Sebastijan Spajic
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Anne Holtermann
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Janina Dörr
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Sophia Stock
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Andreas Zingg
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Cancer Immunotherapy, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Cancer Immunotherapy, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Ignazio Piseddu
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - David Anz
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | | | - Tianjiao Zhang
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Thomas Nerreter
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Susana Minguet
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Munich, Research Center for Environmental Health (HMGU), Neuherberg, Germany.
| | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
11
|
Medina-Herrera A, Sarasquete ME, Jiménez C, Puig N, García-Sanz R. Minimal Residual Disease in Multiple Myeloma: Past, Present, and Future. Cancers (Basel) 2023; 15:3687. [PMID: 37509348 PMCID: PMC10377959 DOI: 10.3390/cancers15143687] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Responses to treatment have improved over the last decades for patients with multiple myeloma. This is a consequence of the introduction of new drugs that have been successfully combined in different clinical contexts: newly diagnosed, transplant-eligible or ineligible patients, as well as in the relapsed/refractory setting. However, a great proportion of patients continue to relapse, even those achieving complete response, which underlines the need for updated response criteria. In 2014, the international myeloma working group established new levels of response, prompting the evaluation of minimal residual disease (MRD) for those patients already in complete or stringent complete response as defined by conventional serological assessments: the absence of tumor plasma cells in 100,000 total cells or more define molecular and immunophenotypic responses by next-generation sequencing and flow cytometry, respectively. In this review, we describe all the potential methods that may be used for MRD detection based on the evidence found in the literature, paying special attention to their advantages and pitfalls from a critical perspective.
Collapse
Affiliation(s)
- Alejandro Medina-Herrera
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - María Eugenia Sarasquete
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Cristina Jiménez
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Noemí Puig
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Ramón García-Sanz
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| |
Collapse
|
12
|
Marx A, Osváth M, Szikora B, Pipek O, Csabai I, Nagy Á, Bödör C, Matula Z, Nagy G, Bors A, Uher F, Mikala G, Vályi-Nagy I, Kacskovics I. Liquid biopsy-based monitoring of residual disease in multiple myeloma by analysis of the rearranged immunoglobulin genes-A feasibility study. PLoS One 2023; 18:e0285696. [PMID: 37235573 DOI: 10.1371/journal.pone.0285696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
The need for sensitive monitoring of minimal/measurable residual disease (MRD) in multiple myeloma emerged as novel therapies led to deeper responses. Moreover, the potential benefits of blood-based analyses, the so-called liquid biopsy is prompting more and more studies to assess its feasibility. Considering these recent demands, we aimed to optimize a highly sensitive molecular system based on the rearranged immunoglobulin (Ig) genes to monitor MRD from peripheral blood. We analyzed a small group of myeloma patients with the high-risk t(4;14) translocation, using next-generation sequencing of Ig genes and droplet digital PCR of patient-specific Ig heavy chain (IgH) sequences. Moreover, well established monitoring methods such as multiparametric flow cytometry and RT-qPCR of the fusion transcript IgH::MMSET (IgH and multiple myeloma SET domain-containing protein) were utilized to evaluate the feasibility of these novel molecular tools. Serum measurements of M-protein and free light chains together with the clinical assessment by the treating physician served as routine clinical data. We found significant correlation between our molecular data and clinical parameters, using Spearman correlations. While the comparisons of the Ig-based methods and the other monitoring methods (flow cytometry, qPCR) were not statistically evaluable, we found common trends in their target detection. Regarding longitudinal disease monitoring, the applied methods yielded complementary information thus increasing the reliability of MRD evaluation. We also detected indications of early relapse before clinical signs, although this implication needs further verification in a larger patient cohort.
Collapse
Affiliation(s)
- Anita Marx
- Department of Immunology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Doctoral School of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Magdolna Osváth
- Department of Immunology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Doctoral School of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Bence Szikora
- Department of Immunology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Orsolya Pipek
- Department of Physics of Complex Systems, ELTE Eötvös Loránd University, Budapest, Hungary
| | - István Csabai
- Department of Physics of Complex Systems, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Ákos Nagy
- Department of Pathology and Experimental Cancer Research, HCEMM-SE Molecular Oncohematology Research Group, Semmelweis University, Budapest, Hungary
| | - Csaba Bödör
- Department of Pathology and Experimental Cancer Research, HCEMM-SE Molecular Oncohematology Research Group, Semmelweis University, Budapest, Hungary
| | - Zsolt Matula
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Ginette Nagy
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - András Bors
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Ferenc Uher
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Gábor Mikala
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - István Vályi-Nagy
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Imre Kacskovics
- Department of Immunology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
13
|
Buenache N, Sánchez-delaCruz A, Cuenca I, Giménez A, Moreno L, Martínez-López J, Rosa-Rosa JM. Identification of Immunoglobulin Gene Rearrangement Biomarkers in Multiple Myeloma through cfDNA-Based Liquid Biopsy Using tchDNA-Seq. Cancers (Basel) 2023; 15:cancers15112911. [PMID: 37296872 DOI: 10.3390/cancers15112911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by the clonal proliferation of pathogenic CD138+ plasma cells (PPCs) in bone marrow (BM). Recent years have seen a significant increase in the treatment options for MM; however, most patients who achieve complete the response ultimately relapse. The earlier detection of tumor-related clonal DNA would thus be very beneficial for patients with MM and would enable timely therapeutic interventions to improve outcomes. Liquid biopsy of "cell-free DNA" (cfDNA) as a minimally invasive approach might be more effective than BM aspiration not only for the diagnosis but also for the detection of early recurrence. Most studies thus far have addressed the comparative quantification of patient-specific biomarkers in cfDNA with PPCs and BM samples, which have shown good correlations. However, there are limitations to this approach, such as the difficulty in obtaining enough circulating free tumor DNA to achieve sufficient sensitivity for the assessment of minimal residual disease. Herein, we summarize current data on methodologies to characterize MM, and we present evidence that targeted capture hybridization DNA sequencing (tchDNA-Seq) can provide robust biomarkers in cfDNA, including immunoglobulin (IG) rearrangements. We also show that detection can be improved by prior purification of the cfDNA. Overall, liquid biopsies of cfDNA to monitor IG rearrangements have the potential to provide important diagnostic, prognostic, and predictive information in patients with MM.
Collapse
Affiliation(s)
- Natalia Buenache
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Andrea Sánchez-delaCruz
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Isabel Cuenca
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Alicia Giménez
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Laura Moreno
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Joaquín Martínez-López
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
- Department of Translational Haematology, Haematology Service, Hospital 12 de Octubre, 28041 Madrid, Spain
- Department of Medicine, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Spanish National Cancer Research Center (CNIO), 28034 Madrid, Spain
| | - Juan Manuel Rosa-Rosa
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
- Spanish National Cancer Research Center (CNIO), 28034 Madrid, Spain
| |
Collapse
|
14
|
Bou Zerdan M, Kassab J, Saba L, Haroun E, Bou Zerdan M, Allam S, Nasr L, Macaron W, Mammadli M, Abou Moussa S, Chaulagain CP. Liquid biopsies and minimal residual disease in lymphoid malignancies. Front Oncol 2023; 13:1173701. [PMID: 37228488 PMCID: PMC10203459 DOI: 10.3389/fonc.2023.1173701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 04/21/2023] [Indexed: 05/27/2023] Open
Abstract
Minimal residual disease (MRD) assessment using peripheral blood instead of bone marrow aspirate/biopsy specimen or the biopsy of the cancerous infiltrated by lymphoid malignancies is an emerging technique with enormous interest of research and technological innovation at the current time. In some lymphoid malignancies (particularly ALL), Studies have shown that MRD monitoring of the peripheral blood may be an adequate alternative to frequent BM aspirations. However, additional studies investigating the biology of liquid biopsies in ALL and its potential as an MRD marker in larger patient cohorts in treatment protocols are warranted. Despite the promising data, there are still limitations in liquid biopsies in lymphoid malignancies, such as standardization of the sample collection and processing, determination of timing and duration for liquid biopsy analysis, and definition of the biological characteristics and specificity of the techniques evaluated such as flow cytometry, molecular techniques, and next generation sequencies. The use of liquid biopsy for detection of minimal residual disease in T-cell lymphoma is still experimental but it has made significant progress in multiple myeloma for example. Recent attempt to use artificial intelligence may help simplify the algorithm for testing and may help avoid inter-observer variation and operator dependency in these highly technically demanding testing process.
Collapse
Affiliation(s)
- Maroun Bou Zerdan
- Department of Internal Medicine, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Joseph Kassab
- Cleveland Clinic, Research Institute, Cleveland, OH, United States
| | - Ludovic Saba
- Department of Hematology-Oncology, Myeloma and Amyloidosis Program, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL, United States
| | - Elio Haroun
- Department of Medicine, State University of New York (SUNY) Upstate Medical University, New York, NY, United States
| | | | - Sabine Allam
- Department of Medicine and Medical Sciences, University of Balamand, Balamand, Lebanon
| | - Lewis Nasr
- University of Texas MD Anderson Cancer Center, Texas, TX, United States
| | - Walid Macaron
- University of Texas MD Anderson Cancer Center, Texas, TX, United States
| | - Mahinbanu Mammadli
- Department of Internal Medicine, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | | | - Chakra P. Chaulagain
- Department of Hematology-Oncology, Myeloma and Amyloidosis Program, Maroone Cancer Center, Cleveland Clinic Florida, Weston, FL, United States
| |
Collapse
|
15
|
Talotta D, Almasri M, Cosentino C, Gaidano G, Moia R. Liquid biopsy in hematological malignancies: current and future applications. Front Oncol 2023; 13:1164517. [PMID: 37152045 PMCID: PMC10157039 DOI: 10.3389/fonc.2023.1164517] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
The assessment of the cancer mutational profile is crucial for patient management, stratification, and therapeutic decisions. At present, in hematological malignancies with a solid mass, such as lymphomas, tumor genomic profiling is generally performed on the tissue biopsy, but the tumor may harbor genetic lesions that are unique to other anatomical compartments. The analysis of circulating tumor DNA (ctDNA) on the liquid biopsy is an emerging approach that allows genotyping and monitoring of the disease during therapy and follow-up. This review presents the different methods for ctDNA analysis and describes the application of liquid biopsy in different hematological malignancies. In diffuse large B-cell lymphoma (DLBCL) and Hodgkin lymphoma (HL), ctDNA analysis on the liquid biopsy recapitulates the mutational profile of the tissue biopsy and can identify mutations otherwise absent on the tissue biopsy. In addition, changes in the ctDNA amount after one or two courses of chemotherapy significantly predict patient outcomes. ctDNA analysis has also been tested in myeloid neoplasms with promising results. In addition to mutational analysis, liquid biopsy also carries potential future applications of ctDNA, including the analysis of ctDNA fragmentation and epigenetic patterns. On these grounds, several clinical trials aiming at incorporating ctDNA analysis for treatment tailoring are currently ongoing in hematological malignancies.
Collapse
Affiliation(s)
| | | | | | | | - Riccardo Moia
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
16
|
Li S, Zhang E, Cai Z. Liquid biopsy by analysis of circulating myeloma cells and cell-free nucleic acids: a novel noninvasive approach of disease evaluation in multiple myeloma. Biomark Res 2023; 11:27. [PMID: 36890597 PMCID: PMC9997021 DOI: 10.1186/s40364-023-00469-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/26/2023] [Indexed: 03/10/2023] Open
Abstract
Multiple myeloma (MM) is an incurable hematological cancer with high spatial- and temporal-heterogeneity. Invasive single-point bone marrow sampling cannot capture the tumor heterogeneity and is difficult to repeat for serial assessments. Liquid biopsy is a technique for identifying and analyzing circulating MM cells and cell products produced by tumors and released into the circulation, allowing for the minimally invasive and comprehensive detection of disease burden and molecular alterations in MM and monitoring treatment response and disease progression. Furthermore, liquid biopsy can provide complementary information to conventional detection approaches and improve their prognostic values. This article reviewed the technologies and applications of liquid biopsy in MM.
Collapse
Affiliation(s)
- Shuchan Li
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79, Qingchun Road, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Enfan Zhang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79, Qingchun Road, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhen Cai
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79, Qingchun Road, Hangzhou, Zhejiang, China. .,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
17
|
Kriegsmann K, Manta C, Schwab R, Mai EK, Raab MS, Salwender HJ, Fenk R, Besemer B, Dürig J, Schroers R, von Metzler I, Hänel M, Mann C, Asemissen AM, Heilmeier B, Bertsch U, Huhn S, Müller-Tidow C, Goldschmidt H, Hundemer M. Comparison of bone marrow and peripheral blood aberrant plasma cell assessment by NGF in patients with MM. Blood Adv 2023; 7:379-383. [PMID: 35914229 PMCID: PMC9898596 DOI: 10.1182/bloodadvances.2022008005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 02/01/2023] Open
Affiliation(s)
- Katharina Kriegsmann
- Department of Hematology, Oncology, and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Calin Manta
- Department of Hematology, Oncology, and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Ricarda Schwab
- Department of Hematology, Oncology, and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Elias K. Mai
- Department of Hematology, Oncology, and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marc S. Raab
- Department of Hematology, Oncology, and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Hans J. Salwender
- Asklepios Tumorzentrum Hamburg, AK Altona and AK St. Georg, Hamburg, Germany
| | - Roland Fenk
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Britta Besemer
- Department of Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| | - Jan Dürig
- Department for Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Roland Schroers
- Department of Hematology and Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Ivana von Metzler
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Mathias Hänel
- Department of Internal Medicine III, Clinic Chemnitz, Chemnitz, Germany
| | - Christoph Mann
- Department for Hematology, Oncology and Immunology, University Hospital Gießen and Marburg, Marburg, Germany
| | - Anne M. Asemissen
- Department of Oncology, Hematology and Bone Marrow Transplant, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bernhard Heilmeier
- Clinic for Oncology and Hematology, Hospital Barmherzige Brueder Regensburg, Regensburg, Germany
| | - Uta Bertsch
- Department of Hematology, Oncology, and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefanie Huhn
- Department of Hematology, Oncology, and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Hematology, Oncology, and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Hartmut Goldschmidt
- Department of Hematology, Oncology, and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Hundemer
- Department of Hematology, Oncology, and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- Medizinisches Versorgungszentrum am Kreiskrankenhaus Bergstraße gGmbH, Heppenheim, Germany
| |
Collapse
|
18
|
Musso N, Romano A, Bonacci PG, Scandura G, Pandino C, Camarda M, Russo GI, Di Raimondo F, Cacciola E, Cacciola R. Label-Free Enrichment of Circulating Tumor Plasma Cells: Future Potential Applications of Dielectrophoresis in Multiple Myeloma. Int J Mol Sci 2022; 23:ijms231912052. [PMID: 36233350 PMCID: PMC9569623 DOI: 10.3390/ijms231912052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 12/05/2022] Open
Abstract
In multiple myeloma (MM), circulating tumor plasma cells (CTPCs) are an emerging prognostic factor, offering a promising and minimally invasive means for longitudinal patient monitoring. Recent advances highlight the complex biology of plasma cell trafficking, highlighting the phenotypic and genetic signatures of intra- and extra-medullary MM onset, making CTPC enumeration and characterization a new frontier of precision medicine for MM patients, requiring novel technological platforms for their standardized and harmonized detection. Dielectrophoresis (DEP) is an emerging label-free cell manipulation technique to separate cancer cells from healthy cells in peripheral blood samples, based on phenotype and membrane capacitance that could be successfully tested to enumerate and isolate CTPCs. Herein, we summarize preclinical data on DEP development for CTPC detection, as well as their clinical and research potential.
Collapse
Affiliation(s)
- Nicolò Musso
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
- StLab SRL, 95126 Catania, Italy
| | - Alessandra Romano
- Department of General Surgery and Surgical Medical Specialties, University of Catania, 95125 Catania, Italy
- Correspondence: ; Tel.: +39-095-378-2971
| | - Paolo Giuseppe Bonacci
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy
| | - Grazia Scandura
- Department of General Surgery and Surgical Medical Specialties, University of Catania, 95125 Catania, Italy
| | - Clarissa Pandino
- Department of General Surgery and Surgical Medical Specialties, University of Catania, 95125 Catania, Italy
| | | | - Giorgio Ivan Russo
- Urology Section, University of Catania, Via S. Sofia 78, 95125 Catania, Italy
| | - Francesco Di Raimondo
- Department of General Surgery and Surgical Medical Specialties, University of Catania, 95125 Catania, Italy
| | - Emma Cacciola
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy
- Hemostasis/Hematology Unit, A.O.U. Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy
| | - Rossella Cacciola
- Hemostasis/Hematology Unit, A.O.U. Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| |
Collapse
|
19
|
Steps towards a Multiple Myeloma Cure? J Pers Med 2022; 12:jpm12091451. [PMID: 36143236 PMCID: PMC9504254 DOI: 10.3390/jpm12091451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Multiple myeloma survival has increased in last 20 years because of new treatments, better clinical management due to novel diagnostic tools such as imaging, and better understanding of the disease, biologically and genetically. Novel drugs have been introduced that act with different therapeutic mechanisms, but so have novel therapeutic strategies such as consolidation and maintenance after autologous stem cell transplant. Imaging (such as PET-CT and MRI) has been applied at diagnosis and after therapy for minimal residual disease monitoring. Multiparametric flow and molecular NGS may detect, with high-sensitivity, residual monoclonal plasma cells in the bone marrow. With this novel therapeutic and biological approach, a considerable fraction of multiple myeloma patients can achieve durable remission or even MGUS-like regression, which can ultimately lead to disease disappearance. The big dogma, “Myeloma is an incurable disease”, is hopefully fading.
Collapse
|
20
|
Allegra A, Cancemi G, Mirabile G, Tonacci A, Musolino C, Gangemi S. Circulating Tumour Cells, Cell Free DNA and Tumour-Educated Platelets as Reliable Prognostic and Management Biomarkers for the Liquid Biopsy in Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14174136. [PMID: 36077672 PMCID: PMC9454477 DOI: 10.3390/cancers14174136] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Even though the presently employed biomarkers in the detection and management of multiple myeloma are demonstrating encouraging results, the mortality percentage of the malignancy is still elevated. Thus, searching for new diagnostic or prognostic markers is pivotal. Liquid biopsy allows the examination of circulating tumour DNA, cell-free DNA, extracellular RNA, and cell free proteins, which are released into the bloodstream due to the breakdown of tumour cells or exosome delivery. Liquid biopsy can now be applied in clinical practice to diagnose, and monitor multiple myeloma, probably allowing a personalized treatment of the disease. Abstract Liquid biopsy is one of the fastest emerging fields in cancer evaluation. Circulating tumour cells and tumour-originated DNA in plasma have become the new targets for their possible employ in tumour diagnosis, and liquid biopsy can define tumour burden without invasive procedures. Multiple Myeloma, one of the most frequent hematologic tumors, has been the target of therapeutic progresses in the last few years. Bone marrow aspirate is the traditional tool for diagnosis, prognosis, and genetic evaluation in multiple myeloma patients. However, this painful procedure presents a relevant drawback for regular disease examination as it requires an invasive practice. Moreover, new data demonstrated that a sole bone marrow aspirate is incapable of expressing the multifaceted multiple myeloma genetic heterogeneity. In this review, we report the emerging usefulness of the assessment of circulating tumour cells, cell-free DNA, extracellular RNA, cell-free proteins, extracellular vesicles, and tumour-educated platelets to evaluate the changing mutational profile of multiple myeloma, as early markers of disease, reliable predictors of prognosis, and as useful tools to perform less invasive monitoring in multiple myeloma.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
- Correspondence:
| | - Gabriella Cancemi
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
| | - Giuseppe Mirabile
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
| | - Alessandro Tonacci
- Clinical Physiology Institute, National Research Council of Italy (IFC-CNR), 56124 Pisa, Italy
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
21
|
Ferla V, Antonini E, Perini T, Farina F, Masottini S, Malato S, Marktel S, Lupo Stanghellini MT, Tresoldi C, Ciceri F, Marcatti M. Minimal residual disease detection by next-generation sequencing in multiple myeloma: Promise and challenges for response-adapted therapy. Front Oncol 2022; 12:932852. [PMID: 36052251 PMCID: PMC9426755 DOI: 10.3389/fonc.2022.932852] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/06/2022] [Indexed: 11/18/2022] Open
Abstract
Assessment of minimal residual disease (MRD) is becoming a standard diagnostic tool for curable hematological malignancies such as chronic and acute myeloid leukemia. Multiple myeloma (MM) remains an incurable disease, as a major portion of patients even in complete response eventually relapse, suggesting that residual disease remains. Over the past decade, the treatment landscape of MM has radically changed with the introduction of new effective drugs and the availability of immunotherapy, including targeted antibodies and adoptive cell therapy. Therefore, conventional serological and morphological techniques have become suboptimal for the evaluation of depth of response. Recently, the International Myeloma Working Group (IMWG) introduced the definition of MRD negativity as the absence of clonal Plasma cells (PC) with a minimum sensitivity of <10−5 either by next-generation sequencing (NGS) using the LymphoSIGHT platform (Sequenta/Adaptative) or by next-generation flow cytometry (NGF) using EuroFlow approaches as the reference methods. While the definition of the LymphoSIGHT platform (Sequenta/Adaptive) as the standard method derives from its large use and validation in clinical studies on the prognostic value of NGS-based MRD, other commercially available options exist. Recently, the LymphoTrack assay has been evaluated in MM, demonstrating a sensitivity level of 10−5, hence qualifying as an alternative effective tool for MRD monitoring in MM. Here, we will review state-of-the-art methods for MRD assessment by NGS. We will summarize how MRD testing supports clinical trials as a useful tool in dynamic risk-adapted therapy. Finally, we will also discuss future promise and challenges of NGS-based MRD determination for clinical decision-making. In addition, we will present our real-life single-center experience with the commercially available NGS strategy LymphoTrack-MiSeq. Even with the limitation of a limited number of patients, our results confirm the LymphoTrack-MiSeq platform as a cost-effective, readily available, and standardized workflow with a sensitivity of 10−5. Our real-life data also confirm that achieving MRD negativity is an important prognostic factor in MM.
Collapse
Affiliation(s)
- Valeria Ferla
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
- *Correspondence: Valeria Ferla,
| | - Elena Antonini
- Molecular Hematology Laboratory, San Raffaele Scientific Institute, Milan, Italy
| | - Tommaso Perini
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
- Age Related Diseases Laboratory, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Farina
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
| | - Serena Masottini
- Molecular Hematology Laboratory, San Raffaele Scientific Institute, Milan, Italy
| | - Simona Malato
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
| | - Sarah Marktel
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
| | | | - Cristina Tresoldi
- Molecular Hematology Laboratory, San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Magda Marcatti
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
22
|
Hultcrantz M, Rustad EH, Yellapantula V, Jacob A, Akhlaghi T, Korde N, Mailankody S, Lesokhin AM, Hassoun H, Smith EL, Lahoud OB, Landau HJ, Shah GL, Scordo M, Chung DJ, Giralt S, Papaemmanuil E, Landgren O. Capture Rate of V(D)J Sequencing for Minimal Residual Disease Detection in Multiple Myeloma. Clin Cancer Res 2022; 28:2160-2166. [PMID: 35553646 DOI: 10.1158/1078-0432.ccr-20-2995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/28/2020] [Accepted: 02/18/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Minimal residual disease (MRD) negativity is a strong predictor for outcome in multiple myeloma. To assess V(D)J clonotype capture using the updated Adaptive next-generation sequencing (NGS) MRD assay in a clinical setting, we analyzed baseline and follow-up samples from patients with multiple myeloma who achieved deep clinical responses. EXPERIMENTAL DESIGN A total of 159 baseline and 31 follow-up samples from patients with multiple myeloma were sequenced using the NGS MRD assay. Baseline samples were also sequenced using a targeted multiple myeloma panel (myTYPE). We estimated ORs with 95% confidence intervals (CI) for clonotypes detection using logistic regression. RESULTS The V(D)J clonotype capture rate was 93% in baseline samples with detectable genomic aberrations, indicating presence of tumor DNA, assessed through myTYPE. myTYPE-positive samples had significantly higher V(D)J clonotype detection rates in univariate (OR, 7.3; 95% CI, 2.8-22.6) and multivariate analysis (OR, 4.4; 95% CI, 1.4-16.9; P = 0.016). Higher disease burden was associated with higher probability of V(D)J clonotype capture, meanwhile no such association was found for age, gender, or type of heavy or light immunoglobulin chain. All V(D)J clonotypes detected at baseline were detected in MRD-positive samples indicating that the V(D)J clonotypes remained stable and did not undergo further rearrangements during follow-up. Of the 31 posttreatment samples, 12 were MRD-negative using the NGS MRD assay. CONCLUSIONS NGS for V(D)J rearrangements in multiple myeloma offers a reliable and sensitive method for MRD tracking with high detection rates in the clinical setting.
Collapse
Affiliation(s)
- Malin Hultcrantz
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York.,Karolinska Institute, Department of Medicine, Solna, Stockholm, Sweden
| | - Even H Rustad
- Department of Cancer Immunology, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Venkata Yellapantula
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | | | - Theresia Akhlaghi
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Neha Korde
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Sham Mailankody
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Alexander M Lesokhin
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Hani Hassoun
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Eric L Smith
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Oscar B Lahoud
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Heather J Landau
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Gunjan L Shah
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Michael Scordo
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - David J Chung
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Sergio Giralt
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Elli Papaemmanuil
- Department of Cancer Immunology, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Ola Landgren
- Myeloma Program, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| |
Collapse
|
23
|
Waldschmidt JM, Yee AJ, Vijaykumar T, Pinto Rengifo RA, Frede J, Anand P, Bianchi G, Guo G, Potdar S, Seifer C, Nair MS, Kokkalis A, Kloeber JA, Shapiro S, Budano L, Mann M, Friedman R, Lipe B, Campagnaro E, O’Donnell EK, Zhang CZ, Laubach JP, Munshi NC, Richardson PG, Anderson KC, Raje NS, Knoechel B, Lohr JG. Cell-free DNA for the detection of emerging treatment failure in relapsed/ refractory multiple myeloma. Leukemia 2022; 36:1078-1087. [PMID: 35027656 PMCID: PMC8983453 DOI: 10.1038/s41375-021-01492-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/24/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022]
Abstract
Interrogation of cell-free DNA (cfDNA) represents an emerging approach to non-invasively estimate disease burden in multiple myeloma (MM). Here, we examined low-pass whole genome sequencing (LPWGS) of cfDNA for its predictive value in relapsed/ refractory MM (RRMM). We observed that cfDNA positivity, defined as ≥10% tumor fraction by LPWGS, was associated with significantly shorter progression-free survival (PFS) in an exploratory test cohort of 16 patients who were actively treated on diverse regimens. We prospectively determined the predictive value of cfDNA in 86 samples from 45 RRMM patients treated with elotuzumab, pomalidomide, bortezomib, and dexamethasone in a phase II clinical trial (NCT02718833). PFS in patients with tumor-positive and -negative cfDNA after two cycles of treatment was 1.6 and 17.6 months, respectively (HR 7.6, P < 0.0001). Multivariate hazard modelling confirmed cfDNA as independent risk factor (HR 96.6, P = 6.92e-05). While correlating with serum-free light chains and bone marrow, cfDNA additionally discriminated patients with poor PFS among those with the same response by IMWG criteria. In summary, detectability of MM-derived cfDNA, as a measure of substantial tumor burden with therapy, independently predicts poor PFS and may provide refinement for standard-of-care response parameters to identify patients with poor response to treatment earlier than is currently feasible.
Collapse
Affiliation(s)
- Johannes M. Waldschmidt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Harvard Medical School, Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Andrew J. Yee
- Harvard Medical School, Boston, MA, USA,Massachusetts General Hospital, Boston, MA, USA
| | - Tushara Vijaykumar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ricardo A. Pinto Rengifo
- Broad Institute of MIT and Harvard, Cambridge, MA, USA,Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA,Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Julia Frede
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Harvard Medical School, Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Praveen Anand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Harvard Medical School, Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Giada Bianchi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Guangwu Guo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sayalee Potdar
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Charles Seifer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Monica S. Nair
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Antonis Kokkalis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Harvard Medical School, Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jake A. Kloeber
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | - Mason Mann
- Massachusetts General Hospital, Boston, MA, USA
| | | | - Brea Lipe
- University of Rochester, Rochester, NY, USA
| | | | - Elizabeth K. O’Donnell
- Harvard Medical School, Boston, MA, USA,Massachusetts General Hospital, Boston, MA, USA
| | - Cheng-Zhong Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA,Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA,Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Jacob P. Laubach
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Nikhil C. Munshi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Paul G. Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Kenneth C. Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Noopur S. Raje
- Harvard Medical School, Boston, MA, USA,Massachusetts General Hospital, 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, Boston, MA, USA
| | - Jens G. Lohr
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Harvard Medical School, Boston, MA, USA,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
24
|
Colmenares R, Álvarez N, Barrio S, Martínez-López J, Ayala R. The Minimal Residual Disease Using Liquid Biopsies in Hematological Malignancies. Cancers (Basel) 2022; 14:1310. [PMID: 35267616 PMCID: PMC8909350 DOI: 10.3390/cancers14051310] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/23/2022] [Accepted: 02/27/2022] [Indexed: 12/02/2022] Open
Abstract
The study of cell-free DNA (cfDNA) and other peripheral blood components (known as "liquid biopsies") is promising, and has been investigated especially in solid tumors. Nevertheless, it is increasingly showing a greater utility in the diagnosis, prognosis, and response to treatment of hematological malignancies; in the future, it could prevent invasive techniques, such as bone marrow (BM) biopsies. Most of the studies about this topic have focused on B-cell lymphoid malignancies; some of them have shown that cfDNA can be used as a novel way for the diagnosis and minimal residual monitoring of B-cell lymphomas, using techniques such as next-generation sequencing (NGS). In myelodysplastic syndromes, multiple myeloma, or chronic lymphocytic leukemia, liquid biopsies may allow for an interesting genomic representation of the tumor clones affecting different lesions (spatial heterogeneity). In acute leukemias, it can be helpful in the monitoring of the early treatment response and the prediction of treatment failure. In chronic lymphocytic leukemia, the evaluation of cfDNA permits the definition of clonal evolution and drug resistance in real time. However, there are limitations, such as the difficulty in obtaining sufficient circulating tumor DNA for achieving a high sensitivity to assess the minimal residual disease, or the lack of standardization of the method, and clinical studies, to confirm its prognostic impact. This review focuses on the clinical applications of cfDNA on the minimal residual disease in hematological malignancies.
Collapse
Affiliation(s)
- Rafael Colmenares
- Hematology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Imas12, 28041 Madrid, Spain; (R.C.); (N.Á.); (S.B.); (J.M.-L.)
| | - Noemí Álvarez
- Hematology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Imas12, 28041 Madrid, Spain; (R.C.); (N.Á.); (S.B.); (J.M.-L.)
- Hematological Malignancies Clinical Research Unit, CNIO, 28029 Madrid, Spain
| | - Santiago Barrio
- Hematology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Imas12, 28041 Madrid, Spain; (R.C.); (N.Á.); (S.B.); (J.M.-L.)
- Hematological Malignancies Clinical Research Unit, CNIO, 28029 Madrid, Spain
| | - Joaquín Martínez-López
- Hematology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Imas12, 28041 Madrid, Spain; (R.C.); (N.Á.); (S.B.); (J.M.-L.)
- Hematological Malignancies Clinical Research Unit, CNIO, 28029 Madrid, Spain
- Department of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, 28029 Madrid, Spain
| | - Rosa Ayala
- Hematology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Imas12, 28041 Madrid, Spain; (R.C.); (N.Á.); (S.B.); (J.M.-L.)
- Hematological Malignancies Clinical Research Unit, CNIO, 28029 Madrid, Spain
- Department of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, 28029 Madrid, Spain
| |
Collapse
|
25
|
Ye X, Li W, Zhang L, Yu J. Clinical Significance of Circulating Cell-Free DNA Detection in Multiple Myeloma: A Meta-Analysis. Front Oncol 2022; 12:852573. [PMID: 35252019 PMCID: PMC8894433 DOI: 10.3389/fonc.2022.852573] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Circulating cell-free DNA (cfDNA) detection, a non-invasive method, appears promising for genetic analyses as well as quantitative assessment of tumor burden in patients with cancer. Although the analysis of cfDNA for clinical prognosis and monitoring disease burden in multiple myeloma (MM) has been recently studied, the results are unclear. In this meta-analysis, we explored the clinical significance of circulating cfDNA detection in patients with MM. We searched PubMed, Embase, and the Cochrane Library for eligible studies published up until July 25, 2021. Diagnostic accuracy variables were calculated and analyzed using Meta-Disc, and prognostic data were analyzed using Review Manager. Overall, seven studies comprising 235 myeloma patients met our inclusion criteria. The overall sensitivity and specificity of cfDNA to detect minimal residual disease (MRD) were 0.58 and 0.91, respectively. Moreover, higher levels of cfDNA were associated with worse progression-free survival as well as with poor overall survival. Our meta-analysis revealed that ctDNA detection has an obvious advantage in terms of MRD detection specificity, but it showed no superiority over bone marrow assessment in terms of MRD detection sensitivity, and higher levels of cfDNA were indicative of worse prognosis in patients with MM. cfDNA detection is a non-invasive method and thus shows promise as a good alternative to BM biopsies for monitoring clonal evolution and tumor burden so as to guide the treatment of patients with MM.
Collapse
Affiliation(s)
- Xueshi Ye
- Department of Hematology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Xueshi Ye,
| | - Wanli Li
- Department of Orthopedics, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lifei Zhang
- Department of Hematology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junyao Yu
- Department of Hematology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
26
|
Alonso R, Lahuerta JJ. Tumor Reduction in Multiple Myeloma: New Concepts for New Therapeutics. Front Oncol 2022; 11:800309. [PMID: 35096603 PMCID: PMC8794792 DOI: 10.3389/fonc.2021.800309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/23/2021] [Indexed: 12/22/2022] Open
Abstract
The development of new resources for a more accurate diagnosis and response assessment in multiple myeloma has been a long process for decades, mainly since the middle of the 20th century. During this time, the succession of technical advances has run parallel to the better knowledge of disease biology and the availability of novel therapeutic strategies. The cornerstone of standardized criteria to uniformly evaluate the disease response in myeloma dates back to the 1990s when the key role of complete remission was established. Since then, different updates have been implemented according to available scientific evidences not always without certain controversies. The progressive improvements in survival results of myeloma patients and the growing quality of responses due to the novel therapies have led to the need of developing new tools for better monitoring of tumor burden. In this way, the concept of minimal residual disease and its key value based on the prognostic significance and the clinical relevance has been consolidated during the last years, overcoming the value of conventional response criteria or classical adverse prognosis markers. Nevertheless, its precise role in the clinical management of myeloma patients to detect early treatment failure and trigger early rescue strategies is still pending to be defined. In this review, we revisit the major milestones in the understanding of tumor reduction in multiple myeloma until the most recent imaging techniques or liquid biopsy approaches, including a critical view of conventional response criteria, whose backbone has remained unchanged during the last 20 years.
Collapse
Affiliation(s)
- Rafael Alonso
- Hematology Department, Hospital Universitario 12 de Octubre, CIBERONC CB16/12/00369, Madrid, Spain
| | - Juan José Lahuerta
- Instituto de Investigación Sanitaria, Hospital Universitario 12 de Octubre (imas12) CIBERONC CB16/12/00369, Madrid, Spain
| |
Collapse
|
27
|
Mack EKM, Hartmann S, Ross P, Wollmer E, Mann C, Neubauer A, Brendel C, Hoffmann J. Monitoring multiple myeloma in the peripheral blood based on cell-free DNA and circulating plasma cells. Ann Hematol 2022; 101:811-824. [PMID: 35106639 PMCID: PMC8913458 DOI: 10.1007/s00277-022-04771-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/16/2022] [Indexed: 11/25/2022]
Abstract
With the advent of novel, highly effective therapies for multiple myeloma (MM), classical serologic monitoring appears insufficient for response assessment and prediction of relapse. Moreover, serologic studies in MM are hampered by interference of therapeutic antibodies. The detection of malignant plasma cell clones by next generation sequencing (NGS) or multiparameter flow cytometry (MFC) circumvents these difficulties and can be performed in the peripheral blood (pB) by targeting circulating cell-free DNA (cfDNA) or circulating plasma cells (CPCs), thus also avoiding an invasive sampling procedure. Here, we applied NGS of VJ light chain (LC) rearrangements in cfDNA and MFC of magnetically-enriched CD138-positive CPCs (me-MFC) to investigate disease burden in unselected MM patients. Sequencing was successful for 114/130 (87.7%) cfDNA samples and me-MFC results were analyzable for 196/205 (95.6%) samples. MM clones were detectable in 38.9% of samples taken at initial diagnosis or relapse (ID/RD), but only in 11.8% of samples taken during complete remission (CR). Circulating MM plasma cells were present in 83.3% of ID/RD samples and 9.9% of CR samples. Residual disease assessment by NGS or me-MFC in samples taken during very good partial remission or CR was 80% concordant. Notably, 4/4 (NGS) and 5/8 (me-MFC) positive CR samples were from patients with oligo- or non-secretory myeloma. The time to progression was shorter if there was evidence of residual myeloma in the pB. Together, our findings indicate that our two novel analytical approaches accurately indicate the course of MM and may be particularly valuable for monitoring patients with serologically non-trackable disease.
Collapse
Affiliation(s)
- Elisabeth K M Mack
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany.
| | - Sören Hartmann
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Petra Ross
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Ellen Wollmer
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Christoph Mann
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Andreas Neubauer
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Cornelia Brendel
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Jörg Hoffmann
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany.
| |
Collapse
|
28
|
Charalampous C, Kourelis T. Minimal Residual Disease Assessment in Multiple Myeloma Patients: Minimal Disease With Maximal Implications. Front Oncol 2022; 11:801851. [PMID: 35155198 PMCID: PMC8825476 DOI: 10.3389/fonc.2021.801851] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/29/2021] [Indexed: 11/21/2022] Open
Abstract
Multiple Myeloma (MM), the second most common hematologic malignancy, has been the target of many therapeutic advances over the past two decades. The introduction of novel agents, such as proteasome inhibitors, immunomodulatory drugs, and monoclonal antibodies, along with autologous hematopoietic stem cell transplantation (ASCT) in the current standard of care, has increased the median survival of myeloma patients significantly. Nevertheless, a curative treatment option continues to elude us, and MM remains an incurable disease, with patients relapsing even after achieving deep conventionally defined responses, underscoring the need for the development of sensitive methods that will allow for proper identification and management of the patients with a higher probability of relapse. Accurate detection of Minimal Residual Disease (MRD) from a bone marrow biopsy represents a relatively new approach of evaluating response to treatment with data showing clear benefit from obtaining MRD(-) status at any point of the disease course. As life expectancy for patients with MM continues to increase and deep responses are starting to become the norm, establishing and refining the role of MRD in the disease course is more relevant than ever. This review examines the different methods used to detect MRD and discusses future considerations regarding the implementation in day-to-day clinical practice and as a prospective primary endpoint for clinical trials.
Collapse
|
29
|
Settino M, Cannataro M. Using MMRFBiolinks R-Package for Discovering Prognostic Markers in Multiple Myeloma. Methods Mol Biol 2022; 2401:289-314. [PMID: 34902136 DOI: 10.1007/978-1-0716-1839-4_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Multiple myeloma (MM) is the second most frequent hematological malignancy in the world although the related pathogenesis remains unclear. Gene profiling studies, commonly carried out through next-generation sequencing (NGS) and Microarrays technologies, represent powerful tools for discovering prognostic markers in MM. NGS technologies have made great leaps forward both economically and technically gaining in popularity. As NGS techniques becomes simpler and cheaper, researchers choose NGS over microarrays for more of their genomic applications. However, Microarrays still provide significant benefits with respect to NGS. For instance, RNA-Seq requires more complex bioinformatic analysis with respect to Microarray as well as it lacks of standardized protocols for analysis. Therefore, a synergy between the two technologies may be well expected in the future. In order to take up this challenge, a valid tool for integrative analysis of MM data retrieved through NGS techniques is MMRFBiolinks, a new R package for integrating and analyzing datasets from the Multiple Myeloma Research Foundation (MMRF) CoMMpass (Clinical Outcomes in MM to Personal Assessment of Genetic Profile) study, available at MMRF Researcher Gateway (MMRF-RG), and at the National Cancer Institute Genomic Data Commons (NCI-GDC) Data Portal. Instead of developing a completely new package from scratch, we decided to leverage TC-GABiolinks, an R/Bioconductor package, because it provides some useful methods to access and analyze MMRF-CoMMpass data. An integrative analysis workflow based on the usage of MMRFBiolinks is illustrated.In particular, it leads towards a comparative analysis of RNA-Seq data stored at GDC Data Portal that allows to carry out a Kaplan Meier (KM ) Survival Analysis and an enrichment analysis for a Differential Gene Expression (DGE) gene set.Furthermore, it deals with MMRF-RG data for analyzing the correlation between canonical variants and treatment outcome as well as treatment class. In order to show the potential of the workflow, we present two case studies. The former deals with data of MM Bone Marrow sample types available at GDC Data Portal. The latter deals with MMRF-RG data for analyzing the correlation between canonical variants in a gene set obtained from the case study 1 and the treatment outcome as well as the treatment class.
Collapse
Affiliation(s)
- Marzia Settino
- University Magna Graecia of Catanzaro, Catanzaro, Italy.
| | | |
Collapse
|
30
|
Russo GI, Musso N, Romano A, Caruso G, Petralia S, Lanzanò L, Broggi G, Camarda M. The Role of Dielectrophoresis for Cancer Diagnosis and Prognosis. Cancers (Basel) 2021; 14:198. [PMID: 35008359 PMCID: PMC8750463 DOI: 10.3390/cancers14010198] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 12/17/2022] Open
Abstract
Liquid biopsy is emerging as a potential diagnostic tool for prostate cancer (PC) prognosis and diagnosis. Unfortunately, most circulating tumor cells (CTC) technologies, such as AdnaTest or Cellsearch®, critically rely on the epithelial cell adhesion molecule (EpCAM) marker, limiting the possibility of detecting cancer stem-like cells (CSCs) and mesenchymal-like cells (EMT-CTCs) that are present during PC progression. In this context, dielectrophoresis (DEP) is an epCAM independent, label-free enrichment system that separates rare cells simply on the basis of their specific electrical properties. As compared to other technologies, DEP may represent a superior technique in terms of running costs, cell yield and specificity. However, because of its higher complexity, it still requires further technical as well as clinical development. DEP can be improved by the use of microfluid, nanostructured materials and fluoro-imaging to increase its potential applications. In the context of cancer, the usefulness of DEP lies in its capacity to detect CTCs in the bloodstream in their epithelial, mesenchymal, or epithelial-mesenchymal phenotype forms, which should be taken into account when choosing CTC enrichment and analysis methods for PC prognosis and diagnosis.
Collapse
Affiliation(s)
| | - Nicolò Musso
- Department of Biomedical and Biotechnological Science (BIOMETEC), University of Catania, 95123 Catania, Italy
- STLab s.r.l., Via Anapo 53, 95126 Catania, Italy;
| | - Alessandra Romano
- Haematological Section, University of Catania, 95125 Catania, Italy;
| | - Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (G.C.); (S.P.)
| | - Salvatore Petralia
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (G.C.); (S.P.)
| | - Luca Lanzanò
- Department of Physics and Astronomy “Ettore Majorana”, University of Catania, 95123 Catania, Italy;
| | - Giuseppe Broggi
- Pathology Section, Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| | | |
Collapse
|
31
|
Determination of MYD88L265P mutation fraction in IgM monoclonal gammopathies. Blood Adv 2021; 6:189-199. [PMID: 34788399 PMCID: PMC8753203 DOI: 10.1182/bloodadvances.2021005354] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/27/2021] [Indexed: 11/29/2022] Open
Abstract
Quantitative evaluation of the tumor load in patients with IgM monoclonal gammopathies in tDNA and cfDNA samples.
We describe a novel method for the detection of MYD88L265P mutation using a competitive allele-specific polymerase chain reaction (Cast-PCR) assay. This assay has a sensitivity of 1 × 10−3, is applicable in reactions containing very low amounts of DNA (as low as 20 pg), and allowed the detection of MYD88L265P somatic mutation in both tumor-derived DNA (tDNA) and cell-free DNA (cfDNA). In addition, using the Cast-PCR assay, we were able to determine the mutation allele fraction (MAF) in each tested sample. We then analyzed baseline tDNA and cfDNA samples from 163 patients (53 with immunoglobulin M monoclonal gammopathy of undetermined significance and 110 with Waldenström’s macroglobulinemia [WM], of whom 54 were asymptomatic and 56 were symptomatic) and also in sequential samples of 37 patients. MAF in both cfDNA and tDNA was higher among patients with symptomatic compared with asymptomatic WM and in those with asymptomatic WM compared with those with immunoglobulin M (IgM) monoclonal gammopathy of undetermined significance. In addition, the evaluation of sequential samples showed that MAF decreased after treatment, whereas it increased in patients who relapsed or progressed to symptomatic WM. Thus, Cast-PCR is a highly sensitive, cost-effective diagnostic tool for MYD88L265P detection, applicable in both tDNA and cfDNA samples, that also provides a quantitative evaluation of the tumor load in patients with IgM monoclonal gammopathies.
Collapse
|
32
|
Anderson KC, Auclair D, Adam SJ, Agarwal A, Anderson M, Avet-Loiseau H, Bustoros M, Chapman J, Connors DE, Dash A, Di Bacco A, Du L, Facon T, Flores-Montero J, Gay F, Ghobrial IM, Gormley NJ, Gupta I, Higley H, Hillengass J, Kanapuru B, Kazandjian D, Kelloff GJ, Kirsch IR, Kremer B, Landgren O, Lightbody E, Lomas OC, Lonial S, Mateos MV, Montes de Oca R, Mukundan L, Munshi NC, O'Donnell EK, Orfao A, Paiva B, Patel R, Pugh TJ, Ramasamy K, Ray J, Roshal M, Ross JA, Sigman CC, Thoren KL, Trudel S, Ulaner G, Valente N, Weiss BM, Zamagni E, Kumar SK. Minimal Residual Disease in Myeloma: Application for Clinical Care and New Drug Registration. Clin Cancer Res 2021; 27:5195-5212. [PMID: 34321279 PMCID: PMC9662886 DOI: 10.1158/1078-0432.ccr-21-1059] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/01/2021] [Accepted: 07/23/2021] [Indexed: 01/07/2023]
Abstract
The development of novel agents has transformed the treatment paradigm for multiple myeloma, with minimal residual disease (MRD) negativity now achievable across the entire disease spectrum. Bone marrow-based technologies to assess MRD, including approaches using next-generation flow and next-generation sequencing, have provided real-time clinical tools for the sensitive detection and monitoring of MRD in patients with multiple myeloma. Complementary liquid biopsy-based assays are now quickly progressing with some, such as mass spectrometry methods, being very close to clinical use, while others utilizing nucleic acid-based technologies are still developing and will prove important to further our understanding of the biology of MRD. On the regulatory front, multiple retrospective individual patient and clinical trial level meta-analyses have already shown and will continue to assess the potential of MRD as a surrogate for patient outcome. Given all this progress, it is not surprising that a number of clinicians are now considering using MRD to inform real-world clinical care of patients across the spectrum from smoldering myeloma to relapsed refractory multiple myeloma, with each disease setting presenting key challenges and questions that will need to be addressed through clinical trials. The pace of advances in targeted and immune therapies in multiple myeloma is unprecedented, and novel MRD-driven biomarker strategies are essential to accelerate innovative clinical trials leading to regulatory approval of novel treatments and continued improvement in patient outcomes.
Collapse
Affiliation(s)
- Kenneth C. Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Daniel Auclair
- Multiple Myeloma Research Foundation, Norwalk, Connecticut.,Corresponding Author: Daniel Auclair, Research, Multiple Myeloma Research Foundation, 383 Main Street, Norwalk, CT, 06851. E-mail:
| | - Stacey J. Adam
- Foundation for the National Institutes of Health, North Bethesda, Maryland
| | - Amit Agarwal
- US Medical Oncology, Bristol-Myers Squibb, Summit, New Jersey
| | | | - Hervé Avet-Loiseau
- Laboratoire d'Hématologie, Pôle Biologie, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Mark Bustoros
- Division of Hematology and Medical Oncology, Cornell University/New York Presbyterian Hospital, New York, New York
| | | | - Dana E. Connors
- Foundation for the National Institutes of Health, North Bethesda, Maryland
| | - Ajeeta Dash
- Takeda Pharmaceuticals, Cambridge, Massachusetts
| | | | - Ling Du
- GlaxoSmithKline, Collegeville, Pennsylvania
| | - Thierry Facon
- Department of Hematology, Lille University Hospital, Lille, France
| | - Juan Flores-Montero
- Cancer Research Center (IBMCC-CSIC/USAL-IBSAL); Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Francesca Gay
- Myeloma Unit, Division of Hematology, Azienda Ospedaliero Università Città della Salute e della Scienza, Torino, Italy
| | - Irene M. Ghobrial
- Preventative Cancer Therapies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Nicole J. Gormley
- Division of Hematologic Malignancies 2, Office of Oncologic Disease, Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland
| | - Ira Gupta
- GlaxoSmithKline, Collegeville, Pennsylvania
| | | | - Jens Hillengass
- Division of Hematology and Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - Bindu Kanapuru
- Division of Hematologic Malignancies 2, Office of Oncologic Disease, Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland
| | - Dickran Kazandjian
- Myeloma Program, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Gary J. Kelloff
- Division of Cancer Treatment and Diagnosis, NCI, NIH, Rockville, Maryland
| | - Ilan R. Kirsch
- Translational Medicine, Adaptive Biotechnologies, Seattle, Washington
| | | | - Ola Landgren
- Myeloma Program, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Elizabeth Lightbody
- Preventative Cancer Therapies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Oliver C. Lomas
- Preventative Cancer Therapies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Sagar Lonial
- Department of Hematology and Medical Oncology at Emory University School of Medicine, Atlanta, Georgia
| | | | | | | | - Nikhil C. Munshi
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - Alberto Orfao
- Cancer Research Center (IBMCC-CSIC/USAL-IBSAL); Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Bruno Paiva
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - Reshma Patel
- Janssen Research & Development, Spring House, Pennsylvania
| | - Trevor J. Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Karthik Ramasamy
- Cancer and Haematology Centre, Oxford University Hospitals, Oxford, United Kingdom
| | - Jill Ray
- BioOncology, Genentech Inc., South San Francisco, California
| | - Mikhail Roshal
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jeremy A. Ross
- Precision Medicine, Oncology, AbbVie, Inc., North Chicago, Illinois
| | | | | | - Suzanne Trudel
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | - Nancy Valente
- BioOncology, Genentech Inc., South San Francisco, California
| | | | - Elena Zamagni
- Seragnoli Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Shaji K. Kumar
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
33
|
Liquid biopsy: an evolving paradigm for the biological characterisation of plasma cell disorders. Leukemia 2021; 35:2771-2783. [PMID: 34262132 DOI: 10.1038/s41375-021-01339-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/14/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023]
Abstract
Liquid biopsies-a source of circulating cell-free nucleic acids, proteins and extracellular vesicles-are currently being explored for the quantitative and qualitative characterisation of the tumour genome and as a mode of non-invasive therapeutic monitoring in cancer. Emerging data suggest that liquid biopsies might offer a potentially simple, non-invasive, repeatable strategy for diagnosis, prognostication and therapeutic decision making in a genetically heterogeneous disease like multiple myeloma (MM), with particular applicability in subsets of patients where conventional markers of disease burden may be less informative. In this review, we describe the emerging utility of the evaluation of circulating tumour DNA, extracellular RNA, cell-free proteins and metabolites and extracellular vesicles in MM.
Collapse
|
34
|
Wallington-Beddoe CT, Mynott RL. Prognostic and predictive biomarker developments in multiple myeloma. J Hematol Oncol 2021; 14:151. [PMID: 34556161 PMCID: PMC8461914 DOI: 10.1186/s13045-021-01162-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 09/07/2021] [Indexed: 12/24/2022] Open
Abstract
New approaches to stratify multiple myeloma patients based on prognosis and therapeutic decision-making, or prediction, are needed since patients are currently managed in a similar manner regardless of individual risk factors or disease characteristics. However, despite new and improved biomarkers for determining the prognosis of patients, there is currently insufficient information to utilise biomarkers to intensify, reduce or altogether change treatment, nor to target patient-specific biology in a so-called predictive manner. The ever-increasing number and complexity of drug classes to treat multiple myeloma have improved response rates and so clinically useful biomarkers will need to be relevant in the era of such novel therapies. Therefore, the field of multiple myeloma biomarker development is rapidly progressing, spurred on by new technologies and therapeutic approaches, and underpinned by a deeper understanding of tumour biology with individualised patient management the goal. In this review, we describe the main biomarker categories in multiple myeloma and relate these to diagnostic, prognostic and predictive applications. ![]()
Collapse
Affiliation(s)
- Craig T Wallington-Beddoe
- College of Medicine and Public Health, Level 4, Flinders Centre for Innovation in Cancer, Flinders University, Bedford Park, SA, 5042, Australia. .,Flinders Medical Centre, Bedford Park, SA, 5042, Australia. .,Centre for Cancer Biology, SA Pathology and The University of South Australia, Adelaide, SA, 5000, Australia. .,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, 5000, Australia.
| | - Rachel L Mynott
- College of Medicine and Public Health, Level 4, Flinders Centre for Innovation in Cancer, Flinders University, Bedford Park, SA, 5042, Australia
| |
Collapse
|
35
|
O'Brien A, O'Halloran F, Mykytiv V. Minimal Residual Disease in Multiple Myeloma: Potential for Blood-Based Methods to Monitor Disease. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 22:e34-e40. [PMID: 34470720 DOI: 10.1016/j.clml.2021.07.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/25/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
In recent years, the life expectancy of Multiple Myeloma (MM) patients has substantially improved, but this cancer remains incurable with increasing incidence in the developed world. Most MM patients will eventually relapse due to residual drug-resistant cancerous cells that survive treatment, commonly referred to as minimal residual disease (MRD). Methods to improve MRD detection in MM patients are generating considerable interest as a means of monitoring patients' response to treatment. In clinical laboratories, these methods currently require bone marrow aspirates which are invasive and frequently miss detection of localised disease due to the spatial heterogeneity of disease infiltration. By simplifying serial sampling and allowing for the detection of extramedullary disease, a blood-based method could significantly impact treatment duration and intensity and minimise chemotherapy-induced toxicity. This review will describe the current blood-based techniques available to detect MRD in MM and compare their potential to evaluate patient prognosis and drive therapeutic decisions.
Collapse
Affiliation(s)
- Aisling O'Brien
- Dept. of Biological Sciences, Munster Technological University, Cork, Ireland; Dept. of Immunology, Cork University Hospital, Cork, Ireland.
| | - Fiona O'Halloran
- Dept. of Biological Sciences, Munster Technological University, Cork, Ireland
| | - Vitaliy Mykytiv
- Dept. of Haematology, Cork University Hospital, Cork, Ireland; Dept. of Medicine, University College Cork, Cork, Ireland
| |
Collapse
|
36
|
Yasui H, Kobayashi M, Sato K, Kondoh K, Ishida T, Kaito Y, Tamura H, Handa H, Tsukune Y, Sasaki M, Komatsu N, Tanaka N, Tanaka J, Kizaki M, Kawamata T, Makiyama J, Yokoyama K, Imoto S, Tojo A, Imai Y. Circulating cell-free DNA in the peripheral blood plasma of patients is an informative biomarker for multiple myeloma relapse. Int J Clin Oncol 2021; 26:2142-2150. [PMID: 34259983 DOI: 10.1007/s10147-021-01991-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/07/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Multiple myeloma (MM) is an incurable hematological malignancy. Despite the introduction of several novel drugs, most patients relapse. Biomarkers to identify the early signs of relapse will make it possible to adjust the therapeutic strategy before the disease worsens. Although understanding genetic changes is important for the treatment of MM, currently known biomarkers of relapse, including serum free-light chains and monoclonal paraproteins, are not associated with genetic changes. METHODS We therefore performed a multicenter study to examine the usefulness of circulating cell-free DNA (cfDNA) present in the peripheral blood (PB) plasma of patients as a biomarker for MM relapse. RESULTS We identified several driver mutations by combined analysis of next-generation sequencing and existing databases of candidate oncogenes. Furthermore, relapse was detected more sensitively by monitoring the circulating cfDNA with these driver mutations than by conventional serum free-light chain examination. CONCLUSION These results suggest the potential utility of cfDNA in the PB plasma of patients as a relevant early biomarker for MM relapse.
Collapse
Affiliation(s)
- Hiroshi Yasui
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masayuki Kobayashi
- Division of Molecular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Department of Hematology, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
| | - Kota Sato
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Kanya Kondoh
- Division of Molecular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tadao Ishida
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Yuta Kaito
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Hideto Tamura
- Department of Hematology, Nippon Medical School, Tokyo, Japan.,Division of Diabetes, Endocrinology and Hematology, Department of Internal Medicine, Dokkyo Medical University Saitama Medical Center, Mibu, Japan
| | - Hiroshi Handa
- Department of Medicine and Clinical Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yutaka Tsukune
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | - Makoto Sasaki
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | - Norio Komatsu
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | - Norina Tanaka
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| | - Junji Tanaka
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| | - Masahiro Kizaki
- Department of Hematology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Toyotaka Kawamata
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Junya Makiyama
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kazuaki Yokoyama
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seiya Imoto
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Arinobu Tojo
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Division of Molecular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichi Imai
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
37
|
Ogawa M, Yokoyama K, Imoto S, Tojo A. Role of Circulating Tumor DNA in Hematological Malignancy. Cancers (Basel) 2021; 13:2078. [PMID: 33923024 PMCID: PMC8123338 DOI: 10.3390/cancers13092078] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
With the recent advances in noninvasive approaches for cancer diagnosis and surveillance, the term "liquid biopsy" has become more familiar to clinicians, including hematologists. Liquid biopsy provides a variety of clinically useful genetic data. In this era of personalized medicine, genetic information is critical to early diagnosis, aiding risk stratification, directing therapeutic options, and monitoring disease relapse. The validity of circulating tumor DNA (ctDNA)-mediated liquid biopsies has received increasing attention. This review summarizes the current knowledge of liquid biopsy ctDNA in hematological malignancies, focusing on the feasibility, limitations, and key areas of clinical application. We also highlight recent advances in the minimal residual disease monitoring of leukemia using ctDNA. This article will be useful to those involved in the clinical practice of hematopoietic oncology.
Collapse
Affiliation(s)
- Miho Ogawa
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; (M.O.); (A.T.)
| | - Kazuaki Yokoyama
- Department of Hematology/Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan;
| | - Arinobu Tojo
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; (M.O.); (A.T.)
| |
Collapse
|
38
|
Paschold L, Willscher E, Bein J, Vornanen M, Eichenauer DA, Simnica D, Thiele B, Wickenhauser C, Rosenwald A, Bernd HW, Klapper W, Feller AC, Ott G, Fend F, Hartmann S, Binder M. Evolutionary clonal trajectories in nodular lymphocyte predominant Hodgkin lymphoma with high transformation risk. Haematologica 2021; 106:2654-2666. [PMID: 33882641 PMCID: PMC8485677 DOI: 10.3324/haematol.2021.278427] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Indexed: 11/09/2022] Open
Abstract
The B cell architecture of nodular lymphocyte predominant Hodgkin lymphoma (NLPHL) is complex since it is composed of malignant lymphocyte-predominant (LP) cells along with a B cell rich bystander environment. To gain insight into molecular determinants of transformation, we studied B cell evolutionary trajectories in lymphoma tissue from diagnosis to relapse or transformation to non-Hodgkin lymphoma by immunoglobulin heavy chain next-generation sequencing. NLPHL cases that later transformed showed higher age, IgD negativity, absence of the characteristic IGHV3/IGHD3/IGHJ6 LP rearrangement and high repertoire clonality. We constructed phylogenetic trees within the compartment of the malignant clone to investigate clonal evolution. In all relapsing cases, the LP rearrangement was identical at diagnosis and relapse. NLPHL cases with transformation showed more complex trajectories with strong intraclonal diversification. The dominant founder clone in transformations showed clonal evolution, if derived from the same cell of origin, or originated from a different cell of origin. Together, our data point to a significant role of antigenic drive in NLHPL transformations and identify high B cell repertoire clonality with dominant intraclonal LP cell diversification as a hallmark of transformation. Sequencing of the initial paraffin-embedded tissue may therefore be diagnostically applied to identify NLPHL cases with high transformation risk.
Collapse
Affiliation(s)
- Lisa Paschold
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale)
| | - Edith Willscher
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale)
| | - Julia Bein
- Dr. Senckenberg Institute of Pathology, Goethe University Hospital of Frankfurt Main, Theodor-Stern-Kai 7, D-60590 Frankfurt a. Main
| | - Martine Vornanen
- Department of Pathology, Tampere University Hospital and University of Tampere, Tampere 33520
| | - Dennis A Eichenauer
- University of Cologne, First Department of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Dusseldorf, Cologne, and German Hodgkin Study Group, University Hospital Cologne, Cologne
| | - Donjete Simnica
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale)
| | - Benjamin Thiele
- Department of Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Claudia Wickenhauser
- Institute of Pathology, Martin-Luther-University Halle-Wittenberg, Halle (Saale)
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg and Comprehensive Cancer Center (CCC) Mainfranken, Würzburg
| | | | - Wolfram Klapper
- Department of Pathology, Division of Hematopathology and Lymph Node Registry, Schleswig-Holstein Medical Center, Campus Kiel, Kiel
| | | | - German Ott
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart
| | - Falko Fend
- Institute of Pathology, Eberhard Karls University Tübingen, Tübingen
| | - Sylvia Hartmann
- Institute of Pathology, Eberhard Karls University Tübingen, Tübingen, Germany; Reference and Consultation Center for Lymph Node and Lymphoma Pathology, Goethe University, Frankfurt am Main
| | - Mascha Binder
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale).
| |
Collapse
|
39
|
|
40
|
Maclachlan KH, Came N, Diamond B, Roshal M, Ho C, Thoren K, Mayerhoefer ME, Landgren O, Harrison S. Minimal residual disease in multiple myeloma: defining the role of next generation sequencing and flow cytometry in routine diagnostic use. Pathology 2021; 53:385-399. [PMID: 33674146 DOI: 10.1016/j.pathol.2021.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022]
Abstract
For patients diagnosed with multiple myeloma (MM) there have been significant treatment advances over the past decade, reflected in an increasing proportion of patients achieving durable remissions. Clinical trials repeatedly demonstrate that achieving a deep response to therapy, with a bone marrow assessment proving negative for minimal residual disease (MRD), confers a significant survival advantage. To accurately assess for minute quantities of residual cancer requires highly sensitive methods; either multiparameter flow cytometry or next generation sequencing are currently recommended for MM response assessment. Under optimal conditions, these methods can detect one aberrant cell amongst 1,000,000 normal cells (a sensitivity of 10-6). Here, we will review the practical use of MRD assays in MM, including challenges in implementation for the routine diagnostic laboratory, standardisation across laboratories and clinical trials, the clinical integration of MRD status assessment into MM management and future directions for ongoing research.
Collapse
Affiliation(s)
- Kylee H Maclachlan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Haematology Service, Peter MacCallum Cancer Centre, East Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic, Australia.
| | - Neil Came
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic, Australia; Pathology Department, Peter MacCallum Cancer Centre, East Melbourne, Vic, Australia
| | - Benjamin Diamond
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mikhail Roshal
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Caleb Ho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katie Thoren
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marius E Mayerhoefer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ola Landgren
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Myeloma Program, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Simon Harrison
- Haematology Service, Peter MacCallum Cancer Centre, East Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic, Australia
| |
Collapse
|
41
|
Lim JK, Kuss B, Talaulikar D. Role of cell-free DNA in haematological malignancies. Pathology 2021; 53:416-426. [PMID: 33648721 DOI: 10.1016/j.pathol.2021.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 01/17/2021] [Indexed: 12/13/2022]
Abstract
Cell-free DNA (cfDNA) consists of fragments of double stranded DNA that are found in the circulation. They are released from the apoptosis of both normal haemopoietic cells and malignant cells. The use of cfDNA from easily accessible peripheral blood samples has created a new strategy in studying molecular genomics in haematological malignancies. Its use in diagnosis, prognosis and monitoring potentially precludes the need for repeated tissue samples, i.e., bone marrow biopsy or primary tissue biopsy. It also potentially provides a more comprehensive analysis of the disease as cfDNA are released from tumours from multiple sites of the body. While cfDNA research is still in its infancy, given its potential and the expansion in next generation sequencing (NGS) it has attracted a lot of attention in recent years. This review will focus on acute leukaemia, multiple myeloma and lymphoma and the potential diagnostic and prognostic implications of cfDNA, its role in response assessment and in detection of disease relapse.
Collapse
Affiliation(s)
- Jun K Lim
- Department of Haematology, The Canberra Hospital, Canberra, ACT, Australia
| | - Bryone Kuss
- Department of Molecular Medicine and Genetics, Flinders University/Flinders Medical Centre, SA Pathology Laboratories, Adelaide, SA, Australia
| | - Dipti Talaulikar
- Department of Haematology, The Canberra Hospital, Canberra, ACT, Australia; College of Health and Medicine, Australian National University, Canberra, ACT, Australia.
| |
Collapse
|
42
|
Correia RP, Puga RD, Muto NH, Lee MLDM, Torres DC, Hassan R, Bacal NS, Hamerschlak N, Campregher PV. High-throughput sequencing of immunoglobulin heavy chain for minimal residual disease detection in B-lymphoblastic leukemia. Int J Lab Hematol 2021; 43:724-731. [PMID: 33393719 DOI: 10.1111/ijlh.13453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/24/2020] [Accepted: 12/17/2020] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Minimal residual disease (MRD) is a cornerstone for stratification of upfront B-lymphoblastic leukemia (B-ALL) treatment protocols to decrease relapse risk. Although its detection by flow cytometry (FC) and real-time quantitative polymerase has clinical usefulness, evidence suggests that methods with increased sensitivity could lead to improved outcomes. The aim of this study was to develop an amplicon-based assay followed by high-throughput sequencing of the immunoglobulin heavy chain variable region for MRD detection in B-ALL. METHODS We analyzed 84 samples, 27 from diagnosis, 5 from relapse, 40 from post-treatment samples, and 12 from healthy controls. RESULTS Our assay was able to identify more neoplastic clones at diagnosis than Sanger sequencing including incomplete DJ rearrangements. From the 40 MRD samples evaluated 21 were positive by our new approach on high-throughput sequencing assay, but only 15 of these were positive by FC. The remaining 19 were negative by the two techniques. CONCLUSION We have developed a novel approach on high-sensitive assay for MRD detection in B-ALL, which could add clinical value in the management of patients, especially in cases negative for MRD by FC.
Collapse
Affiliation(s)
- Rodolfo P Correia
- Departments of Clinical Pathology Laboratory, Hematology and Hemotherapy, Research Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Renato D Puga
- Departments of Clinical Pathology Laboratory, Hematology and Hemotherapy, Research Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Nair H Muto
- Departments of Clinical Pathology Laboratory, Hematology and Hemotherapy, Research Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Davi C Torres
- Bone Marrow Transplantation Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Rocio Hassan
- Bone Marrow Transplantation Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Nydia S Bacal
- Departments of Clinical Pathology Laboratory, Hematology and Hemotherapy, Research Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil.,Centro de Hematologia de São Paulo, São Paulo, Brazil
| | - Nelson Hamerschlak
- Departments of Clinical Pathology Laboratory, Hematology and Hemotherapy, Research Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Paulo V Campregher
- Departments of Clinical Pathology Laboratory, Hematology and Hemotherapy, Research Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
43
|
Zuo X, Liu D. Progress in the application of minimal residual disease detection in multiple myeloma. J Hematop 2021. [DOI: 10.1007/s12308-020-00436-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
44
|
Da Vià MC, Ziccheddu B, Maeda A, Bagnoli F, Perrone G, Bolli N. A Journey Through Myeloma Evolution: From the Normal Plasma Cell to Disease Complexity. Hemasphere 2020; 4:e502. [PMID: 33283171 PMCID: PMC7710229 DOI: 10.1097/hs9.0000000000000502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
The knowledge of cancer origin and the subsequent tracking of disease evolution represent unmet needs that will soon be within clinical reach. This will provide the opportunity to improve patient's stratification and to personalize treatments based on cancer biology along its life history. In this review, we focus on the molecular pathogenesis of multiple myeloma (MM), a hematologic malignancy with a well-known multi-stage disease course, where such approach can sooner translate into a clinical benefit. We describe novel insights into modes and timing of disease initiation. We dissect the biology of the preclinical and pre-malignant phases, elucidating how knowledge of the genomics of the disease and the composition of the microenvironment allow stratification of patients based on risk of disease progression. Then, we explore cell-intrinsic and cell-extrinsic drivers of MM evolution to symptomatic disease. Finally, we discuss how this may relate to the development of refractory disease after treatment. By integrating an evolutionary view of myeloma biology with the recent acquisitions on its clonal heterogeneity, we envision a way to drive the clinical management of the disease based on its detailed biological features more than surrogates of disease burden.
Collapse
Affiliation(s)
- Matteo C. Da Vià
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Bachisio Ziccheddu
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Akihiro Maeda
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Filippo Bagnoli
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Clinical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giulia Perrone
- Department of Clinical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Niccolò Bolli
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Clinical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
45
|
Kim S, Yang NE, Jeong D, Yun J, Ryu S, Yoon S, Ahn Y, Lee DS. Minimal residual disease negativity by next-generation flow in non-CR myeloma patients. EJHAEM 2020; 1:563-566. [PMID: 35844993 PMCID: PMC9175670 DOI: 10.1002/jha2.103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 11/25/2022]
Abstract
Next-generation flow (NGF) has detected minimal residual disease (MRD) in numerous myeloma patients who achieve a complete response (CR). However, when MRD is not detected via NGF in non-CR patients, its clinical meaning is uncertain. Here, we investigated the correlation between MRD findings on NGF and the response criteria, paying special attention to patients with discrepant results. We performed NGS analysis of IgH rearrangements on bone marrow samples from the non-CR patients with negative MRD on NGF. NGS detected residual abnormal clones in those patients, suggesting that NGF and NGS should be used in a complementary manner for MRD investigation.
Collapse
Affiliation(s)
- Sung‐Min Kim
- Cancer Research InstituteSeoul National University College of MedicineSeoulSouth Korea
| | - Naery Euphrasia Yang
- Department of Laboratory MedicineSejong General HospitalBucheonRepublic of Korea
| | - Dajeong Jeong
- Department of Laboratory MedicineSeoul National University HospitalSeoulSouth Korea
| | - Jiwon Yun
- Department of Laboratory MedicineSeoul National University HospitalSeoulSouth Korea
| | - Sohee Ryu
- Department of Laboratory MedicineSeoul National University HospitalSeoulSouth Korea
| | - Sung‐Soo Yoon
- Department of Internal MedicineSeoul National University College of MedicineSeoulSouth Korea
| | - Yong‐Oon Ahn
- Cancer Research InstituteSeoul National University College of MedicineSeoulSouth Korea
| | - Dong Soon Lee
- Cancer Research InstituteSeoul National University College of MedicineSeoulSouth Korea
- Department of Laboratory MedicineSeoul National University HospitalSeoulSouth Korea
- Department of Laboratory MedicineSeoul National University College of MedicineSeoulSouth Korea
| |
Collapse
|
46
|
Simnica D, Ittrich H, Bockemeyer C, Stein A, Binder M. Targeting the Mutational Landscape of Bystander Cells: Drug-Promoted Blood Cancer From High-Prevalence Pre-neoplasias in Patients on BRAF Inhibitors. Front Oncol 2020; 10:540030. [PMID: 33042833 PMCID: PMC7517330 DOI: 10.3389/fonc.2020.540030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 08/13/2020] [Indexed: 01/07/2023] Open
Abstract
Drug-promoted cancers are increasingly recognized as a serious clinical problem in patients receiving BRAF inhibitory treatment. Here we report on a patient with BRAF mutant hairy cell leukemia and monoclonal B-cell lymphocytosis (MBL), who responded durably to BRAF/MEK inhibitors (BRAFi/MEKi) but experienced transformation of a RAS mutant MBL to chronic lymphocytic leukemia (CLL) with accelerated nodal progression. Hypothesizing that BRAFi triggered excessive MEK-ERK signaling in the MBL/CLL clone via the CRAF/RAS complex as previously described for BRAFi-induced cancers, BRAFi was discontinued inducing a rapid remission of the CLL on MEKi alone. Liquid biopsy monitoring showed a continuous increase of the MBL/CLL clone from the start of BRAFi/MEKi treatment followed by a rapid decline upon BRAFi withdrawal. Next-generation sequencing of a cohort of patients with MBL and monoclonal gammopathy of unclear significance (MGUS) revealed that almost one third of these cases harbored RAS mutations. In view of the population frequency of lymphatic pre-malignant conditions and the prevalence of RAS mutations in such cases, vigilant surveillance remains critical in patients treated with BRAF inhibitors.
Collapse
Affiliation(s)
- Donjete Simnica
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany.,Department of Oncology and Hematology, BMT with section Pneumology, Hubertus Wald Tumorzentrum/UCCH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Harald Ittrich
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bockemeyer
- Department of Oncology and Hematology, BMT with section Pneumology, Hubertus Wald Tumorzentrum/UCCH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander Stein
- Department of Oncology and Hematology, BMT with section Pneumology, Hubertus Wald Tumorzentrum/UCCH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Hematology-Oncology Practice Hamburg (HOPE), Hamburg, Germany
| | - Mascha Binder
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany.,Department of Oncology and Hematology, BMT with section Pneumology, Hubertus Wald Tumorzentrum/UCCH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
47
|
Gozzetti A, Raspadori D, Bacchiarri F, Sicuranza A, Pacelli P, Ferrigno I, Tocci D, Bocchia M. Minimal Residual Disease in Multiple Myeloma: State of the Art and Applications in Clinical Practice. J Pers Med 2020; 10:jpm10030120. [PMID: 32927719 PMCID: PMC7565263 DOI: 10.3390/jpm10030120] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Novel drugs have revolutionized multiple myeloma therapy in the last 20 years, with median survival that has doubled to up to 8–10 years. The introduction of therapeutic strategies, such as consolidation and maintenance after autologous stem cell transplants, has also ameliorated clinical results. The goal of modern therapies is becoming not only complete remission, but also the deepest possible remission. In this context, the evaluation of minimal residual disease by techniques such as next-generation sequencing (NGS) and next-generation flow (NGF) is becoming part of all new clinical trials that test drug efficacy. This review focuses on minimal residual disease approaches in clinical trials, with particular attention to real-world practices.
Collapse
|
48
|
Costa LJ, Derman BA, Bal S, Sidana S, Chhabra S, Silbermann R, Ye JC, Cook G, Cornell RF, Holstein SA, Shi Q, Omel J, Callander NS, Chng WJ, Hungria V, Maiolino A, Stadtmauer E, Giralt S, Pasquini M, Jakubowiak AJ, Morgan GJ, Krishnan A, Jackson GH, Mohty M, Mateos MV, Dimopoulos MA, Facon T, Spencer A, Miguel JS, Hari P, Usmani SZ, Manier S, McCarthy P, Kumar S, Gay F, Paiva B. International harmonization in performing and reporting minimal residual disease assessment in multiple myeloma trials. Leukemia 2020; 35:18-30. [DOI: 10.1038/s41375-020-01012-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 12/24/2022]
|
49
|
Ntanasis-Stathopoulos I, Gavriatopoulou M, Terpos E, Fotiou D, Kastritis E, Dimopoulos MA. Monitoring Plasma Cell Dyscrasias With Cell-free DNA Analysis. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e905-e909. [PMID: 32723621 DOI: 10.1016/j.clml.2020.06.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022]
Abstract
The analysis of cell-free tumor DNA (cfDNA) has emerged as a promising method to determine the evolving genomic landscape of the whole tumor compartment, mainly in solid malignancies. Plasma cell dyscrasias are characterized by complex and constantly changing genomic aberrations that are important in terms of prognosis, evaluation of the minimal residual disease, and response monitoring. In multiple myeloma, the detection of clonal immunoglobulin rearrangements and driver gene mutations in the cfDNA has shown high concordance rates with their identification in bone marrow-derived tumor DNA. In Waldenström macroglobulinemia, cfDNA can be a reliable alternative to bone marrow aspiration for determining the mutational status of the MYD88 and CXCR4 genes. Importantly, cfDNA can be representative of the whole bone marrow compartment and of extramedullary sites in contrast to the sampling of a single bone marrow site. However, standardization and validation of the techniques are necessary before integrating cfDNA in the clinical practice. Therefore, we encourage the conduction of clinical trials with novel cfDNA-based designs and the adoption of cfDNA-guided endpoints in order to precisely determine the role of cfDNA in the current management of plasma cell dyscrasias.
Collapse
Affiliation(s)
- Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Alexandra General Hospital, Athens, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Alexandra General Hospital, Athens, Greece.
| | - Evangelos Terpos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Alexandra General Hospital, Athens, Greece
| | - Despoina Fotiou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Alexandra General Hospital, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Alexandra General Hospital, Athens, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Alexandra General Hospital, Athens, Greece
| |
Collapse
|
50
|
Detection of the MYD88L265P and CXCR4S338X mutations by cell-free DNA in Waldenström macroglobulinemia. Ann Hematol 2020; 99:1763-1769. [DOI: 10.1007/s00277-020-04139-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 06/10/2020] [Indexed: 12/15/2022]
|