1
|
Mohty M, Avet-Loiseau H, Malard F, Harousseau JL. Potential future direction of measurable residual disease evaluation in multiple myeloma. Blood 2023; 142:1509-1517. [PMID: 37471603 DOI: 10.1182/blood.2023020284] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/22/2023] Open
Abstract
Multiple myeloma remains an incurable disease plagued by high relapse rates. Deeper and more sustainable responses, however, have been consistently shown to improve outcomes and could eventually pave the way to achieving a cure. Our understanding of disease response has surpassed complete response (CR), because the current definitions are suboptimal, and the treatment goal should aim even beyond stringent CR, toward molecular and flow CR, that is, measurable residual disease (MRD) negativity. It has been more than 20 years since the discrepancy in the outcome between patients in CR with and without MRD has been demonstrated, and the field has come a long way from multiparameter flow cytometry to next-generation flow and next-generation sequencing, able to detect up to a limit of detection of a single myeloma cell from 1 million healthy counterparts. This review aims to summarize the current available data regarding MRD but also its potential future use as a coprimary outcome both in clinical and trial settings as a survival surrogate as well as its use to evaluate treatment efficacy and for adaptive response-based and early-rescue therapy. Furthermore, we discuss whether these concepts are applicable in different settings (eg, newly diagnosed and relapsed/refractory myeloma, patients who are eligible and ineligible for tansplant, and standard- and high-risk disease).
Collapse
Affiliation(s)
- Mohamad Mohty
- UMRS 938, Saint-Antoine Research Center, INSERM, Sorbonne University, Paris, France
- Clinical Hematology and Cellular Therapy Department, Saint Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Hervé Avet-Loiseau
- Cancer Research Center of Toulouse, INSERM, Myeloma Genomics Lab University Cancer Institute Toulouse Oncopole, Toulouse, France
- Université Paul Sabatier, Toulouse, France
| | - Florent Malard
- UMRS 938, Saint-Antoine Research Center, INSERM, Sorbonne University, Paris, France
- Clinical Hematology and Cellular Therapy Department, Saint Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Luc Harousseau
- Centre René Gauducheau, Institut de Cancérologie de l'Ouest, Nantes-St Herblain, France
| |
Collapse
|
2
|
Jew S, Bujarski S, Regidor B, Emamy-Sadr M, Swift R, Eades B, Kim S, Eshaghian S, Berenson JR. Clinical Outcomes and Serum B-Cell Maturation Antigen Levels in a Real-World Unselected Population of Newly Diagnosed Multiple Myeloma Patients. Target Oncol 2023; 18:735-747. [PMID: 37682503 DOI: 10.1007/s11523-023-00990-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Progression-free survival (PFS) and overall survival (OS) of newly diagnosed multiple myeloma (MM) patients have been widely published in the clinical trials setting, but data published from real-world settings are limited. OBJECTIVE We determined the survival and factors that predict outcomes among 161 unselected, newly diagnosed MM patients whose frontline therapy was started at a single clinic specializing in the treatment of this B-cell malignancy. PATIENTS AND METHODS None of these patients underwent an autologous stem cell transplantation as part of their initial therapy and the population had a high proportion (35%) of cytogenetic high-risk patients. RESULTS With a median follow-up of 42.7 months, the cohort had a median PFS of 22.8 months and a median OS of 136.2 months. The 1-, 3-, and 5-year survival rates were 97.5%, 85.3%, and 76.2%, respectively. These results are considerably better than those reported from patients enrolled in clinical trials and those from countries with national registries. Age <65 years predicted for a longer OS (p = 0.0004). Baseline serum B-cell maturation antigen (sBCMA) levels were also assessed and showed median and mean levels of 320.3 ng/mL and 551.1 ng/mL, respectively. Furthermore, patients with baseline sBCMA levels in the lowest quartile (≤136.2 ng/mL) showed a longer PFS (p = 0.0262). CONCLUSION These results provide clinicians with a real-world understanding of the survival of unselected, newly diagnosed patients initiating therapy in a clinic specializing in the care of MM patients.
Collapse
Affiliation(s)
- Scott Jew
- Institute for Myeloma and Bone Cancer Research, Suite 300, 9201 W. Sunset Boulevard, West Hollywood, CA, 90069, USA
- Berenson Cancer Center, West Hollywood, CA, USA
| | - Sean Bujarski
- Institute for Myeloma and Bone Cancer Research, Suite 300, 9201 W. Sunset Boulevard, West Hollywood, CA, 90069, USA
- Berenson Cancer Center, West Hollywood, CA, USA
| | | | | | | | | | | | | | - James R Berenson
- Institute for Myeloma and Bone Cancer Research, Suite 300, 9201 W. Sunset Boulevard, West Hollywood, CA, 90069, USA.
- Berenson Cancer Center, West Hollywood, CA, USA.
- ONCOtherapeutics, West Hollywood, CA, USA.
- ONCOtracker, West Hollywood, CA, USA.
| |
Collapse
|
3
|
Solimando AG, Krebs M, Desantis V, Marziliano D, Caradonna IC, Morizio A, Argentiero A, Shahini E, Bittrich M. Breaking through Multiple Myeloma: A Paradigm for a Comprehensive Tumor Ecosystem Targeting. Biomedicines 2023; 11:2087. [PMID: 37509726 PMCID: PMC10377041 DOI: 10.3390/biomedicines11072087] [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: 04/10/2023] [Revised: 06/29/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Multiple myeloma (MM) is a cancerous condition characterized by the proliferation of plasma cells within the hematopoietic marrow, resulting in multiple osteolytic lesions. MM patients typically experience bone pain, kidney damage, fatigue due to anemia, and infections. Historically, MM was an incurable disease with a life expectancy of around three years after diagnosis. However, over the past two decades, the development of novel therapeutics has significantly improved patient outcomes, including response to treatment, remission duration, quality of life, and overall survival. These advancements include thalidomide and its derivatives, lenalidomide and pomalidomide, which exhibit diverse mechanisms of action against the plasma cell clone. Additionally, proteasome inhibitors such as bortezomib, ixazomib, and carfilzomib disrupt protein degradation, proving specifically toxic to cancerous plasma cells. Recent advancements also involve monoclonal antibodies targeting surface antigens, such as elotuzumab (anti-CS1) and daratumumab (anti-CD38), bispecific t-cell engagers such as teclistamab (anti-BCMA/CD3) and Chimeric antigen receptor T (CAR-T)-based strategies, with a growing focus on drugs that exhibit increasingly targeted action against neoplastic plasma cells and relevant effects on the tumor microenvironment.
Collapse
Affiliation(s)
- Antonio G Solimando
- Unit of Internal Medicine and Clinical Oncology "G. Baccelli", Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Markus Krebs
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, 97080 Würzburg, Germany
- Department of Urology and Pediatric Urology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Vanessa Desantis
- Department of Precision and Regenerative Medicine and Ionian Area, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Donatello Marziliano
- Unit of Internal Medicine and Clinical Oncology "G. Baccelli", Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Ingrid Catalina Caradonna
- Department of Precision and Regenerative Medicine and Ionian Area, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Arcangelo Morizio
- Orthopedics and Traumatology Unit ASL BA-Ospedale della Murgia "Fabio Perinei", 70022 Altamura, Italy
| | | | - Endrit Shahini
- Gastroenterology Unit, National Institute of Gastroenterology-IRCCS "Saverio de Bellis", 70013 Castellana Grotte, Italy
| | - Max Bittrich
- Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
4
|
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: 0] [Impact Index Per Article: 0] [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
|
5
|
Fitzpatrick MJ, Murali MR, Nardi V. Molecular Diagnostics of Plasma Cell Neoplasms. Surg Pathol Clin 2023; 16:401-410. [PMID: 37149365 DOI: 10.1016/j.path.2023.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Genetic characterization of myeloma at diagnosis by interphase fluorescence in situ hybridization and next-generation sequencing (NGS) can assist with risk stratification and treatment planning. Measurable residual disease (MRD) status after treatment, as evaluated by next-generation flow cytometry or NGS on bone marrow aspirate material, is one of the most important predictors of prognosis. Less-invasive tools for MRD assessment such as liquid biopsy approaches have also recently emerged as potential alternatives.
Collapse
|
6
|
Chow S, Kis O, Mulder DT, Danesh A, Bruce J, Wang TT, Reece D, Bhalis N, Neri P, Sabatini PJ, Keats J, Trudel S, Pugh TJ. Myeloma immunoglobulin rearrangement and translocation detection through targeted capture sequencing. Life Sci Alliance 2023; 6:e202201543. [PMID: 36328595 PMCID: PMC9644417 DOI: 10.26508/lsa.202201543] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022] Open
Abstract
Multiple myeloma is a plasma cell neoplasm characterized by clonal immunoglobulin V(D)J signatures and oncogenic immunoglobulin gene translocations. Additional subclonal genomic changes are acquired with myeloma progression and therapeutic selection. PCR-based methods to detect V(D)J rearrangements can have biases introduced by highly multiplexed reactions and primers undermined by somatic hypermutation, and are not readily extended to include mutation detection. Here, we report a hybrid-capture approach (CapIG-seq) targeting the 3' and 5' ends of the V and J segments of all immunoglobulin loci that enable the efficient detection of V(D)J rearrangements. We also included baits for oncogenic translocations and mutation detection. We demonstrate complete concordance with matched whole-genome sequencing and/or PCR clonotyping of 24 cell lines and report the clonal sequences for 41 uncharacterized cell lines. We also demonstrate the application to patient specimens, including 29 bone marrow and 39 cell-free DNA samples. CapIG-seq shows concordance between bone marrow and cfDNA blood samples (both contemporaneous and follow-up) with regard to the somatic variant, V(D)J, and translocation detection. CapIG-seq is a novel, efficient approach to examining genomic alterations in myeloma.
Collapse
Affiliation(s)
- Signy Chow
- University Health Network, Toronto, Canada
- Sunnybrook Health Sciences Centre, Toronto, Canada
- University of Toronto, Toronto, Canada
| | - Olena Kis
- University Health Network, Toronto, Canada
| | | | | | - Jeff Bruce
- University Health Network, Toronto, Canada
| | - Ting Ting Wang
- University Health Network, Toronto, Canada
- University of Toronto, Toronto, Canada
| | - Donna Reece
- University Health Network, Toronto, Canada
- University of Toronto, Toronto, Canada
| | | | | | - Peter Jb Sabatini
- University Health Network, Toronto, Canada
- University of Toronto, Toronto, Canada
| | - Jonathan Keats
- Translational Genomics Research Institute, City of Hope, AZ, USA
| | - Suzanne Trudel
- University Health Network, Toronto, Canada
- University of Toronto, Toronto, Canada
| | - Trevor J Pugh
- University Health Network, Toronto, Canada
- University of Toronto, Toronto, Canada
- Ontario Institute for Cancer Research, Toronto, Canada
| |
Collapse
|
7
|
Rath A, Panda T, Dass J, Seth T, Mahapatra M, Tyagi S. Minimal residual disease analysis in multiple myeloma: A single-center experience. JOURNAL OF APPLIED HEMATOLOGY 2023. [DOI: 10.4103/joah.joah_69_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
|
8
|
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: 0.5] [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
|
9
|
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: 5.5] [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
|
10
|
Fitzpatrick MJ, Nardi V, Sohani AR. Plasma cell myeloma: role of histopathology, immunophenotyping, and genetic testing. Skeletal Radiol 2022; 51:17-30. [PMID: 33687521 DOI: 10.1007/s00256-021-03754-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/27/2021] [Accepted: 03/02/2021] [Indexed: 02/02/2023]
Abstract
Myeloma is a malignant neoplasm of plasma cells with complex pathogenesis. Diagnosis and risk stratification require the integration of histology, radiology, serology, and genetic data. Bone marrow biopsies are essential for myeloma diagnosis by providing material for histologic and cytologic assessment as well as immunophenotypic and genetic studies. Flow cytometry and genetic studies are, in particular, becoming increasingly important for diagnosis, risk stratification, and assessment of treatment response. Myeloma has traditionally been characterized by recurrent cytogenetic abnormalities that can be divided into two subtypes: hyperdiploid, characterized by trisomies, and non-hyperdiploid, characterized by translocations involving chromosome 14. These abnormalities are thought to be primary events, initiating a premalignant state, which progresses to myeloma through the acquisition of secondary mutations. The emergence of next-generation sequencing has led to the discovery of numerous mutations and gene fusions that comprise the heterogenous genomic landscape of myeloma. As the underlying pathogenesis of myeloma continues to be delineated, possible therapeutic targets have also emerged. Herein, we describe the importance of histology, immunophenotype, and mutational analysis in the assessment of myeloma.
Collapse
Affiliation(s)
- Megan J Fitzpatrick
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, WRN 219, Boston, MA, 02114, USA
- Department of Pathology, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Valentina Nardi
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, WRN 219, Boston, MA, 02114, USA
- Department of Pathology, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Aliyah R Sohani
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, WRN 219, Boston, MA, 02114, USA.
- Department of Pathology, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA.
| |
Collapse
|
11
|
Ding H, Xu J, Lin Z, Huang J, Wang F, Yang Y, Cui Y, Luo H, Gao Y, Zhai X, Pang W, Zhang L, Zheng Y. Minimal residual disease in multiple myeloma: current status. Biomark Res 2021; 9:75. [PMID: 34649622 PMCID: PMC8515655 DOI: 10.1186/s40364-021-00328-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/17/2021] [Indexed: 02/08/2023] Open
Abstract
Multiple myeloma (MM) is a treatable plasma cell cancer with no cure. Clinical evidence shows that the status of minimal residual disease (MRD) after treatment is an independent prognostic factor of MM. MRD indicates the depth of post-therapeutic remission. In this review article, we outlined the major clinical trials that have determined the prognostic value of MRD in MM. We also reviewed different methods that were used for MM MRD assessment. Most important, we reviewed our current understanding of MM MRD biology. MRD studies strongly indicate that MRD is not a uniform declination of whole MM tumor population. Rather, MM MRD exhibits unique signatures of cytogenetic aberration and gene expression profiles, unlike those of MM cells before therapy. Diagnostic high-risk MM and low-risk MM exhibited a diversity of MRD features. Clonal evaluation may occur at the MRD stage in MM. The dynamics from the diagnostic MM to MRD correlate with the disease prognosis. Lastly, on the aspect of omics, we performed data-based analysis to address the biological features underlying the course of diagnostic-to-MRD MM. To summarize, the MRD stage of disease represents a critical step in MM pathogenesis and progression. Demonstration of MM MRD biology should help us to deal with the curative difficulties.
Collapse
Affiliation(s)
- Hong Ding
- Department of Hematology, West China Hospital, and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, #37 GuoXue Xiang Street, Chengdu, China
| | - Juan Xu
- Department of Hematology, West China Hospital, and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, #37 GuoXue Xiang Street, Chengdu, China
| | - Zhimei Lin
- Department of Hematology, West China Hospital, and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, #37 GuoXue Xiang Street, Chengdu, China.,Department of Hematology, The Affiliated Hospital of Chengdu University, Chengdu, China
| | - Jingcao Huang
- Department of Hematology, West China Hospital, and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, #37 GuoXue Xiang Street, Chengdu, China
| | - Fangfang Wang
- Department of Hematology, West China Hospital, and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, #37 GuoXue Xiang Street, Chengdu, China
| | - Yan Yang
- Department of Hematology, West China Hospital, and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, #37 GuoXue Xiang Street, Chengdu, China
| | - Yushan Cui
- Department of Hematology, West China Hospital, and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, #37 GuoXue Xiang Street, Chengdu, China
| | - Hongmei Luo
- Department of Hematology, West China Hospital, and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, #37 GuoXue Xiang Street, Chengdu, China
| | - Yuhan Gao
- Department of Hematology, West China Hospital, and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, #37 GuoXue Xiang Street, Chengdu, China
| | - Xinyu Zhai
- Department of Hematology, West China Hospital, and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, #37 GuoXue Xiang Street, Chengdu, China
| | - Weicui Pang
- Department of Hematology, West China Hospital, and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, #37 GuoXue Xiang Street, Chengdu, China
| | - Li Zhang
- Department of Hematology, West China Hospital, and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, #37 GuoXue Xiang Street, Chengdu, China.
| | - Yuhuan Zheng
- Department of Hematology, West China Hospital, and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, #37 GuoXue Xiang Street, Chengdu, China.
| |
Collapse
|
12
|
Bravo-Pérez C, Sola M, Teruel-Montoya R, García-Malo MD, Ortuño FJ, Vicente V, de Arriba F, Jerez A. Minimal Residual Disease in Multiple Myeloma: Something Old, Something New. Cancers (Basel) 2021; 13:4332. [PMID: 34503142 PMCID: PMC8430644 DOI: 10.3390/cancers13174332] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
The game-changing outcome effect, due to the generalized use of novel agents in MM, has cre-ated a paradigm shift. Achieving frequent deep responses has placed MM among those neoplasms where the rationale for assessing MRD is fulfilled. However, its implementation in MM has raised specific questions: how might we weight standard measures against deep MRD in the emerging CAR-T setting? Which high sensitivity method to choose? Are current response criteria still useful? In this work, we address lessons learned from the use of MRD in other neoplasms, the steps followed for the harmonization of current methods for comprehensively measuring MRD, and the challenges that new therapies and concepts pose in the MM clinical field.
Collapse
Affiliation(s)
- Carlos Bravo-Pérez
- Hematology and Medical Oncology Department, University Hospital Morales Meseguer, IMIB, 30003 Murcia, Spain; (C.B.-P.); (M.S.); (M.D.G.-M.); (F.J.O.); (V.V.); (F.d.A.); (A.J.)
| | - María Sola
- Hematology and Medical Oncology Department, University Hospital Morales Meseguer, IMIB, 30003 Murcia, Spain; (C.B.-P.); (M.S.); (M.D.G.-M.); (F.J.O.); (V.V.); (F.d.A.); (A.J.)
| | - Raúl Teruel-Montoya
- Hematology and Medical Oncology Department, University Hospital Morales Meseguer, IMIB, 30003 Murcia, Spain; (C.B.-P.); (M.S.); (M.D.G.-M.); (F.J.O.); (V.V.); (F.d.A.); (A.J.)
- CB15/00055-CIBERER, 30003 Murcia, Spain
| | - María Dolores García-Malo
- Hematology and Medical Oncology Department, University Hospital Morales Meseguer, IMIB, 30003 Murcia, Spain; (C.B.-P.); (M.S.); (M.D.G.-M.); (F.J.O.); (V.V.); (F.d.A.); (A.J.)
| | - Francisco José Ortuño
- Hematology and Medical Oncology Department, University Hospital Morales Meseguer, IMIB, 30003 Murcia, Spain; (C.B.-P.); (M.S.); (M.D.G.-M.); (F.J.O.); (V.V.); (F.d.A.); (A.J.)
| | - Vicente Vicente
- Hematology and Medical Oncology Department, University Hospital Morales Meseguer, IMIB, 30003 Murcia, Spain; (C.B.-P.); (M.S.); (M.D.G.-M.); (F.J.O.); (V.V.); (F.d.A.); (A.J.)
- CB15/00055-CIBERER, 30003 Murcia, Spain
| | - Felipe de Arriba
- Hematology and Medical Oncology Department, University Hospital Morales Meseguer, IMIB, 30003 Murcia, Spain; (C.B.-P.); (M.S.); (M.D.G.-M.); (F.J.O.); (V.V.); (F.d.A.); (A.J.)
| | - Andrés Jerez
- Hematology and Medical Oncology Department, University Hospital Morales Meseguer, IMIB, 30003 Murcia, Spain; (C.B.-P.); (M.S.); (M.D.G.-M.); (F.J.O.); (V.V.); (F.d.A.); (A.J.)
- CB15/00055-CIBERER, 30003 Murcia, Spain
| |
Collapse
|
13
|
Sato K, Okazuka K, Ishida T, Sakamoto J, Kaneko S, Nashimoto J, Uto Y, Ogura M, Yoshiki Y, Abe Y, Maeda A, Hamazaki H, Tsukada N, Hiragohri Y, Suzuki K. Minimal residual disease detection in multiple myeloma: comparison between BML single-tube 10-color multiparameter flow cytometry and EuroFlow multiparameter flow cytometry. Ann Hematol 2021; 100:2989-2995. [PMID: 34430990 DOI: 10.1007/s00277-021-04634-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/29/2021] [Indexed: 11/29/2022]
Abstract
Minimal residual disease (MRD)-negative status in multiple myeloma (MM) is associated with favorable outcomes. Although EuroFlow next-generation flow (NGF) is a global standard for MRD detection, its operating cost is high. Therefore, it is desirable to develop a less expensive method with equivalent sensitivity to that of EuroFlow-NGF. In this study, we compared the analytical ability of our BML 10-color multiparameter flow cytometry (MFC) to that of EuroFlow-NGF. Bone marrow samples collected from 51 patients with MM were subjected to MRD detection using BML 10-color-MFC and EuroFlow-NGF. Our antibody panel consisted of CD38 multiepitope, CD138, CD45, CD56, CD19, CD27, CD81, CD117, cytoplasmic immunoglobulin (cIg) κ, and cIgλ in a single tube. The median percentages of total plasma cells, as per 10-color-MFC and EuroFlow-NGF, were 0.2148% and 0.2200%, respectively, with a good correlation between the methods (r = 0.950). The median percentages of myeloma cells determined via 10-color-MFC and EuroFlow-NGF were 0.0012% and 0.0007%, respectively, with a strong correlation (r = 0.954). Our 10-color-MFC demonstrated high sensitivity to detect MRD; the results showed a good correlation with those obtained using EuroFlow-NGF. Therefore, our cost-effective single-tube MFC (approximately 100 USD/sample) is a promising alternative method for the detection of MRD in patients with MM.
Collapse
Affiliation(s)
- Kota Sato
- Department of Hematology, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo, Japan.
| | - Kiyoshi Okazuka
- Department of Hematology, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo, Japan
| | - Tadao Ishida
- Department of Hematology, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo, Japan
| | - Jun Sakamoto
- Cellular Immunology Section, BML Incorporation, Tokyo, Japan
| | - Shigeto Kaneko
- Cellular Immunology Section, BML Incorporation, Tokyo, Japan
| | - Junichiro Nashimoto
- Department of Hematology, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo, Japan
| | - Yui Uto
- Department of Hematology, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo, Japan
| | - Mizuki Ogura
- Department of Hematology, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo, Japan
| | - Yumiko Yoshiki
- Department of Hematology, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo, Japan
| | - Yu Abe
- Department of Hematology, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo, Japan
| | - Aki Maeda
- Cellular Immunology Section, BML Incorporation, Tokyo, Japan
| | | | - Nobuhiro Tsukada
- Department of Hematology, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo, Japan
| | - Yuji Hiragohri
- Cellular Immunology Section, BML Incorporation, Tokyo, Japan
| | - Kenshi Suzuki
- Department of Hematology, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo, Japan
| |
Collapse
|
14
|
Next-Generation Biomarkers in Multiple Myeloma: Understanding the Molecular Basis for Potential Use in Diagnosis and Prognosis. Int J Mol Sci 2021; 22:ijms22147470. [PMID: 34299097 PMCID: PMC8305153 DOI: 10.3390/ijms22147470] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/25/2021] [Accepted: 07/02/2021] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma (MM) is considered to be the second most common blood malignancy and it is characterized by abnormal proliferation and an accumulation of malignant plasma cells in the bone marrow. Although the currently utilized markers in the diagnosis and assessment of MM are showing promising results, the incidence and mortality rate of the disease are still high. Therefore, exploring and developing better diagnostic or prognostic biomarkers have drawn global interest. In the present review, we highlight some of the recently reported and investigated novel biomarkers that have great potentials as diagnostic and/or prognostic tools in MM. These biomarkers include angiogenic markers, miRNAs as well as proteomic and immunological biomarkers. Moreover, we present some of the advanced methodologies that could be utilized in the early and competent diagnosis of MM. The present review also focuses on understanding the molecular concepts and pathways involved in these biomarkers in order to validate and efficiently utilize them. The present review may also help in identifying areas of improvement for better diagnosis and superior outcomes of MM.
Collapse
|
15
|
Chopra S, Dunham T, Syrbu SI, Karandikar NJ, Darbro BW, Holman CJ. Utility of Flow Cytometry and Fluorescence In Situ Hybridization in Follow-up Monitoring of Plasma Cell Myeloma. Am J Clin Pathol 2021; 156:198-204. [PMID: 33437993 DOI: 10.1093/ajcp/aqaa224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES We sought to investigate the clinical utility of flow cytometry (FC) and fluorescence in situ hybridization (FISH) in the workup of myeloma. METHODS We retrospectively reviewed the reports of bone marrow biopsies received for myeloma evaluation between October 2015 and January 2019. RESULTS A total of 1,708 biopsy specimens from 469 myeloma patients (mean age, 64.5 years [SD, 9.3]; female, 41.4%) were reviewed. Both FC and FISH had comparable detection rates at the time of initial diagnosis (97.6% vs 98.8%) and for follow-up cases (28.6% vs 28.2%). FC and FISH results were concordant in 98.8% of the initial diagnosis cases and 89.6% of the follow-up cases. The FISH-positive (FISH+)/FC-negative (FC-) discordance and FISH-/FC+ discordance occurred among 81 (5.0%) and 87 (5.4%) follow-up cases. In comparison with all concordant cases, FISH+/FC- discordant cases were more likely to have received treatment with daratumumab (P < .05). CONCLUSIONS Plasma cell-enriched FISH and FC have comparable abnormal plasma cell detection rates, and approximately 10% of the follow-up cases have discordant FISH and FC results in which residual disease is detected by only one of these modalities. FISH testing should be considered for cases with negative FC, especially in patients who have received treatment with daratumumab or in cases in which there is concern about specimen adequacy.
Collapse
Affiliation(s)
| | - Timothy Dunham
- Stead Family Department of Pediatrics, University of Iowa Hospitals and Clinics, Iowa City
| | | | | | - Benjamin W Darbro
- Stead Family Department of Pediatrics, University of Iowa Hospitals and Clinics, Iowa City
| | | |
Collapse
|
16
|
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: 3.7] [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
|
17
|
Diamond BT, Rustad E, Maclachlan K, Thoren K, Ho C, Roshal M, Ulaner GA, Landgren CO. Defining the undetectable: The current landscape of minimal residual disease assessment in multiple myeloma and goals for future clarity. Blood Rev 2021; 46:100732. [PMID: 32771227 PMCID: PMC9928431 DOI: 10.1016/j.blre.2020.100732] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/05/2020] [Accepted: 07/06/2020] [Indexed: 01/19/2023]
Abstract
Multiple Myeloma, the second most prevalent hematologic malignancy, yet lacks an established curative therapy. However, overall response rate to modern four-drug regimens approaches 100%. Major efforts have thus focused on the measurement of minute quantities of residual disease (minimal residual disease or MRD) for prognostic metrics and therapeutic response evaluation. Currently, MRD is assessed by flow cytometry or by next generation sequencing to track tumor-specific immunoglobulin V(D)J rearrangements. These bone marrow-based methods can reach sensitivity thresholds of the identification of one neoplastic cell in 1,000,000 (10-6). New technologies are being developed to be used alone or in conjunction with established methods, including peripheral blood-based assays, mass spectrometry, and targeted imaging. Data is also building for MRD as a surrogate endpoint for overall survival. Here, we will address the currently utilized MRD assays, challenges in validation across labs and clinical trials, techniques in development, and future directions for successful clinical application of MRD in multiple myeloma.
Collapse
Affiliation(s)
| | | | | | | | - Caleb Ho
- Memorial Sloan Kettering Cancer Center, USA
| | | | | | | |
Collapse
|
18
|
Van der Straeten J, De Brouwer W, Kabongo E, Dresse MF, Fostier K, Schots R, Van Riet I, Bakkus M. Validation of a PCR-Based Next-Generation Sequencing Approach for the Detection and Quantification of Minimal Residual Disease in Acute Lymphoblastic Leukemia and Multiple Myeloma Using gBlocks as Calibrators. J Mol Diagn 2021; 23:599-611. [PMID: 33549860 DOI: 10.1016/j.jmoldx.2021.01.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 10/22/2022] Open
Abstract
Detection of minimal residual disease (MRD) to guide therapy has been a standard practice in treatment of childhood acute lymphoblastic leukemia (ALL) for decades. In multiple myeloma (MM), a clear correlation is found between absence of MRD and longer survival. Quantitative allele-specific oligonucleotide (qASO)-PCR is the standard molecular method for MRD detection in these hematologic malignant tumors. However, this technique has some drawbacks that can be overcome by next-generation sequencing (NGS). In this study, NGS is validated as an alternative method for qASO-PCR for MRD detection in both ALL and MM. MRD results obtained by NGS and qASO-PCR were compared in 59 and 39 bone marrow samples of 33 and 14 patients with ALL and MM, respectively. Our results indicate that the use of gBlocks as calibrators makes the NGS approach a powerful tool to quantify MRD. With an input of 400 ng of DNA (corresponding to approximately 7 × 104 cells), a limit of detection of 0.01% can be achieved. The specificity of the NGS-MRD technique was 100%, and a correlation with qASO-PCR for quantifiable MRD results of 0.93 and 0.91 was found in ALL and MM, respectively. Especially for MM, the higher applicability (100%) of the NGS-MRD protocol, compared with qASO-PCR (57%), was clearly demonstrated. These results demonstrate that NGS is an even better alternative to qASO-PCR.
Collapse
Affiliation(s)
- Jona Van der Straeten
- Molecular Hematology Laboratory, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Wouter De Brouwer
- Department of Hematology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Emmanuelle Kabongo
- Molecular Hematology Laboratory, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | | | - Karel Fostier
- Department of Hematology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Rik Schots
- Department of Hematology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ivan Van Riet
- Department of Hematology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Marleen Bakkus
- Molecular Hematology Laboratory, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium.
| |
Collapse
|
19
|
Devarakonda S, Jethava Y. Minimal residual disease in multiple myeloma: are we there yet? Int J Hematol Oncol 2020; 9:IJH29. [PMID: 33294170 PMCID: PMC7689536 DOI: 10.2217/ijh-2020-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
| | - Yogesh Jethava
- Division of Hematology, University of Iowa, Iowa city, IA 52242, USA
| |
Collapse
|
20
|
Medina A, Puig N, Flores-Montero J, Jimenez C, Sarasquete ME, Garcia-Alvarez M, Prieto-Conde I, Chillon C, Alcoceba M, Gutierrez NC, Oriol A, Rosinol L, Bladè J, Gironella M, Hernandez MT, Gonzalez-Calle V, Cedena MT, Paiva B, San-Miguel JF, Lahuerta JJ, Mateos MV, Martinez-Lopez J, Orfao A, Gonzalez M, Garcia-Sanz R. Comparison of next-generation sequencing (NGS) and next-generation flow (NGF) for minimal residual disease (MRD) assessment in multiple myeloma. Blood Cancer J 2020; 10:108. [PMID: 33127891 PMCID: PMC7603393 DOI: 10.1038/s41408-020-00377-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/14/2020] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
Detecting persistent minimal residual disease (MRD) allows the identification of patients with an increased risk of relapse and death. In this study, we have evaluated MRD 3 months after transplantation in 106 myeloma patients using a commercial next-generation sequencing (NGS) strategy (LymphoTrack®), and compared the results with next-generation flow (NGF, EuroFlow). The use of different marrow pulls and the need of concentrating samples for NGS biased the applicability for MRD evaluation and favored NGF. Despite that, correlation between NGS and NGF was high (R2 = 0.905). The 3-year progression-free survival (PFS) rates by NGS and NGF were longer for undetectable vs. positive patients (NGS: 88.7% vs. 56.6%; NGF: 91.4% vs. 50%; p < 0.001 for both comparisons), which resulted in a 3-year overall survival (OS) advantage (NGS: 96.2% vs. 77.3%; NGF: 96.6% vs. 74.9%, p < 0.01 for both comparisons). In the Cox regression model, NGS and NGF negativity had similar results but favoring the latter in PFS (HR: 0.20, 95% CI: 0.09–0.45, p < 0.001) and OS (HR: 0.21, 95% CI: 0.06–0.75, p = 0.02). All these results reinforce the role of MRD detection by different strategies in patient prognosis and highlight the use of MRD as an endpoint for multiple myeloma treatment.
Collapse
Affiliation(s)
- Alejandro Medina
- Departamento de Hematología, Hospital Universitario de Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Noemi Puig
- Departamento de Hematología, Hospital Universitario de Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Juan Flores-Montero
- Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Cristina Jimenez
- Departamento de Hematología, Hospital Universitario de Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), Salamanca, Spain
| | - M-Eugenia Sarasquete
- Departamento de Hematología, Hospital Universitario de Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), Salamanca, Spain.
| | - María Garcia-Alvarez
- Departamento de Hematología, Hospital Universitario de Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Isabel Prieto-Conde
- Departamento de Hematología, Hospital Universitario de Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Carmen Chillon
- Departamento de Hematología, Hospital Universitario de Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Miguel Alcoceba
- Departamento de Hematología, Hospital Universitario de Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Norma C Gutierrez
- Departamento de Hematología, Hospital Universitario de Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Albert Oriol
- Hospital Germans Trias i Pujol, Institut Català d'Oncología (ICO), Institut Josep Carreras, Badalona, Spain
| | - Laura Rosinol
- Hospital Clínic i Provincial, Institut de Investicacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Joan Bladè
- Hospital Clínic i Provincial, Institut de Investicacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | | | - Veronica Gonzalez-Calle
- Departamento de Hematología, Hospital Universitario de Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Maria-Teresa Cedena
- Hospital 12 de Octubre, i + 12, CNIO, Universidad Complutense, Madrid, Spain
| | - Bruno Paiva
- Clínica Universidad de Navarra (CUN), Centro de Investigación Médica Aplicada, IDISNA, CIBERONC, Pamplona, Spain
| | - Jesus F San-Miguel
- Clínica Universidad de Navarra (CUN), Centro de Investigación Médica Aplicada, IDISNA, CIBERONC, Pamplona, Spain
| | - Juan-Jose Lahuerta
- Hospital 12 de Octubre, i + 12, CNIO, Universidad Complutense, Madrid, Spain
| | - Maria-Victoria Mateos
- Departamento de Hematología, Hospital Universitario de Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), Salamanca, Spain
| | | | - Alberto Orfao
- Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Marcos Gonzalez
- Departamento de Hematología, Hospital Universitario de Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Ramon Garcia-Sanz
- Departamento de Hematología, Hospital Universitario de Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), Salamanca, Spain
| |
Collapse
|
21
|
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: 10.8] [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]
|
22
|
Flach J, Shumilov E, Joncourt R, Porret N, Novak U, Pabst T, Bacher U. Current concepts and future directions for hemato-oncologic diagnostics. Crit Rev Oncol Hematol 2020; 151:102977. [DOI: 10.1016/j.critrevonc.2020.102977] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 03/22/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023] Open
|
23
|
Sanoja-Flores L, Flores-Montero J, Pérez-Andrés M, Puig N, Orfao A. Detection of Circulating Tumor Plasma Cells in Monoclonal Gammopathies: Methods, Pathogenic Role, and Clinical Implications. Cancers (Basel) 2020; 12:E1499. [PMID: 32521788 PMCID: PMC7352573 DOI: 10.3390/cancers12061499] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/28/2022] Open
Abstract
Cancer dissemination and distant metastasis most frequently require the release of tumor cells into the blood circulation, both in solid tumors and most hematological malignancies, including plasma cell neoplasms. However, detection of blood circulating tumor cells in solid tumors and some hematological malignancies, such as the majority of mature/peripheral B-cell lymphomas and monoclonal gammopathies, has long been a challenge due to their very low frequency. In recent years, the availability of highly-sensitive and standardized methods for the detection of circulating tumor plasma cells (CTPC) in monoclonal gammopathies, e.g., next-generation flow cytometry (NGF), demonstrated the systematic presence of CTPC in blood in virtually every smoldering (SMM) and symptomatic multiple myeloma (MM) patient studied at diagnosis, and in the majority of patients with newly-diagnosed monoclonal gammopathies of undetermined significance (MGUS). These methods set the basis for further detailed characterization of CTPC vs. their bone marrow counterpart in monoclonal gammopathies, to investigate their role in the biology of the disease, and to confirm their strong impact on patient outcome when measured both at diagnosis and after initiating therapy. Here, we review the currently available techniques for the detection of CTPC, and determine their biological features, physiopathological role and clinical significance in patients diagnosed with distinct diagnostic categories of plasma cell neoplasms.
Collapse
Affiliation(s)
- Luzalba Sanoja-Flores
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)- University of Salamanca, 37007 Salamanca, Spain; (L.S.-F.); (J.F.-M.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00400, Instituto Carlos III, 28029 Madrid, Spain
| | - Juan Flores-Montero
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)- University of Salamanca, 37007 Salamanca, Spain; (L.S.-F.); (J.F.-M.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00400, Instituto Carlos III, 28029 Madrid, Spain
| | - Martín Pérez-Andrés
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)- University of Salamanca, 37007 Salamanca, Spain; (L.S.-F.); (J.F.-M.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00400, Instituto Carlos III, 28029 Madrid, Spain
| | - Noemí Puig
- Department of Hematology, University Hospital of Salamanca, IBSAL, IBMCC (USAL-CSIC), 37007 Salamanca, Spain;
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00233, Instituto Carlos III, 28029 Madrid, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)- University of Salamanca, 37007 Salamanca, Spain; (L.S.-F.); (J.F.-M.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Cáncer, CIBER-ONC number CB16/12/00400, Instituto Carlos III, 28029 Madrid, Spain
| |
Collapse
|
24
|
Kostopoulos IV, Ntanasis-Stathopoulos I, Gavriatopoulou M, Tsitsilonis OE, Terpos E. Minimal Residual Disease in Multiple Myeloma: Current Landscape and Future Applications With Immunotherapeutic Approaches. Front Oncol 2020; 10:860. [PMID: 32537439 PMCID: PMC7267070 DOI: 10.3389/fonc.2020.00860] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 04/30/2020] [Indexed: 01/10/2023] Open
Abstract
The basic principle that deeper therapeutic responses lead to better clinical outcomes in cancer has emerged technologies capable of detecting rare residual tumor cells. The need for ultra-sensitive approaches for minimal residual disease (MRD) detection is particularly evident in Multiple Myeloma (MM), where patients will ultimately relapse despite the achievement of complete remission, which is commonplace due to remarkable therapeutic advances. Consequently, current response criteria on MM have been amended based on MRD status and MRD negativity is now considered the most dominant prognostic factor and the most valuable indicator for a subsequent relapse. However, there are particular limitations and several aspects for MRD assessment that remain open. This review summarizes current data on MRD in the clinical management of MM, highlights open issues and discusses the challenges and the endless opportunities arising for both patients and clinicians. Furthermore, it focuses on the current status of MRD in clinical trials, its dynamics in addressing debatable aspects in the clinical handling and its potential role as the prevailing factor for future MRD-driven tailored therapies.
Collapse
Affiliation(s)
- Ioannis V Kostopoulos
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ourania E Tsitsilonis
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
25
|
Paiva B, Puig N, Cedena MT, Rosiñol L, Cordón L, Vidriales MB, Burgos L, Flores-Montero J, Sanoja-Flores L, Lopez-Anglada L, Maldonado R, de la Cruz J, Gutierrez NC, Calasanz MJ, Martin-Ramos ML, Garcia-Sanz R, Martinez-Lopez J, Oriol A, Blanchard MJ, Rios R, Martin J, Martinez-Martinez R, Sureda A, Hernandez MT, de la Rubia J, Krsnik I, Moraleda JM, Palomera L, Bargay J, Van Dongen JJ, Orfao A, Mateos MV, Blade J, San-Miguel JF, Lahuerta JJ. Measurable Residual Disease by Next-Generation Flow Cytometry in Multiple Myeloma. J Clin Oncol 2020; 38:784-792. [DOI: 10.1200/jco.19.01231] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Assessing measurable residual disease (MRD) has become standard with many tumors, but the clinical meaning of MRD in multiple myeloma (MM) remains uncertain, particularly when assessed by next-generation flow (NGF) cytometry. Thus, we aimed to determine the applicability and sensitivity of the flow MRD-negative criterion defined by the International Myeloma Working Group (IMWG). PATIENTS AND METHODS In the PETHEMA/GEM2012MENOS65 trial, 458 patients with newly diagnosed MM had longitudinal assessment of MRD after six induction cycles with bortezomib, lenalidomide, and dexamethasone (VRD), autologous transplantation, and two consolidation courses with VRD. MRD was assessed in 1,100 bone marrow samples from 397 patients; the 61 patients without MRD data discontinued treatment during induction and were considered MRD positive for intent-to-treat analysis. The median limit of detection achieved by NGF was 2.9 × 10−6. Patients received maintenance (lenalidomide ± ixazomib) according to the companion PETHEMA/GEM2014MAIN trial. RESULTS Overall, 205 (45%) of 458 patients had undetectable MRD after consolidation, and only 14 of them (7%) have experienced progression thus far; seven of these 14 displayed extraosseous plasmacytomas at diagnosis and/or relapse. Using time-dependent analysis, patients with undetectable MRD had an 82% reduction in the risk of progression or death (hazard ratio, 0.18; 95% CI, 0.11 to 0.30; P < .001) and an 88% reduction in the risk of death (hazard ratio, 0.12; 95% CI, 0.05 to 0.29; P < .001). Timing of undetectable MRD (after induction v intensification) had no impact on patient survival. Attaining undetectable MRD overcame poor prognostic features at diagnosis, including high-risk cytogenetics. By contrast, patients with Revised International Staging System III status and positive MRD had dismal progression-free and overall survivals (median, 14 and 17 months, respectively). Maintenance increased the rate of undetectable MRD by 17%. CONCLUSION The IMWG flow MRD-negative response criterion is highly applicable and sensitive to evaluate treatment efficacy in MM.
Collapse
Affiliation(s)
- Bruno Paiva
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada Instituto de Investigacion Sanitaria de Navarra, Pamplona, Spain
| | - Noemi Puig
- Hospital Universitario de Salamanca, Instituto de Investigacion Biomedica de Salamanca, Salamanca, Spain
| | | | - Laura Rosiñol
- Hospital Clínic Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lourdes Cordón
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - María-Belén Vidriales
- Hospital Universitario de Salamanca, Instituto de Investigacion Biomedica de Salamanca, Salamanca, Spain
| | - Leire Burgos
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada Instituto de Investigacion Sanitaria de Navarra, Pamplona, Spain
| | - Juan Flores-Montero
- Centro de Investigación Biomédica en Red de Cáncer, Instituto Carlos III, Madrid, Spain
- University of Salamanca, Salamanca, Spain
| | - Luzalba Sanoja-Flores
- Centro de Investigación Biomédica en Red de Cáncer, Instituto Carlos III, Madrid, Spain
- University of Salamanca, Salamanca, Spain
| | | | | | | | - Norma C. Gutierrez
- Hospital Universitario de Salamanca, Instituto de Investigacion Biomedica de Salamanca, Salamanca, Spain
| | - Maria-Jose Calasanz
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada Instituto de Investigacion Sanitaria de Navarra, Pamplona, Spain
| | | | - Ramón Garcia-Sanz
- Hospital Universitario de Salamanca, Instituto de Investigacion Biomedica de Salamanca, Salamanca, Spain
| | | | - Albert Oriol
- Institut Català d’Oncologia i Institut Josep Carreras, Badalona, Spain
| | | | - Rafael Rios
- Hospital Virgen de las Nieves, Granada, Spain
| | - Jesus Martin
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | | | - Anna Sureda
- Institut Català d'Oncologia L’Hospitalet, Barcelona, Spain
| | | | - Javier de la Rubia
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Catholic University of Valencia, Valencia, Spain
| | | | | | | | - Joan Bargay
- Hospital Son Llatzer, Palma de Mallorca, Spain
| | | | - Alberto Orfao
- Centro de Investigación Biomédica en Red de Cáncer, Instituto Carlos III, Madrid, Spain
- University of Salamanca, Salamanca, Spain
| | - Maria-Victoria Mateos
- Hospital Universitario de Salamanca, Instituto de Investigacion Biomedica de Salamanca, Salamanca, Spain
| | - Joan Blade
- Hospital Clínic Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jesús F. San-Miguel
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada Instituto de Investigacion Sanitaria de Navarra, Pamplona, Spain
| | | | | |
Collapse
|
26
|
Bolli N, Genuardi E, Ziccheddu B, Martello M, Oliva S, Terragna C. Next-Generation Sequencing for Clinical Management of Multiple Myeloma: Ready for Prime Time? Front Oncol 2020; 10:189. [PMID: 32181154 PMCID: PMC7057289 DOI: 10.3389/fonc.2020.00189] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/04/2020] [Indexed: 12/22/2022] Open
Abstract
Personalized treatment is an attractive strategy that promises increased efficacy with reduced side effects in cancer. The feasibility of such an approach has been greatly boosted by next-generation sequencing (NGS) techniques, which can return detailed information on the genome and on the transcriptome of each patient's tumor, thus highlighting biomarkers of response or druggable targets that may differ from case to case. However, while the number of cancers sequenced is growing exponentially, much fewer cases are amenable to a molecularly-guided treatment outside of clinical trials to date. In multiple myeloma, genomic analysis shows a variety of gene mutations, aneuploidies, segmental copy-number changes, translocations that are extremely heterogeneous, and more numerous than other hematological malignancies. Currently, in routine clinical practice we employ reduced FISH panels that only capture three high-risk features as part of the R-ISS. On the contrary, recent advances have suggested that extending genomic analysis to the full spectrum of recurrent mutations and structural abnormalities in multiple myeloma may have biological and clinical implications. Furthermore, increased efficacy of novel treatments can now produce deeper responses, and standard methods do not have enough sensitivity to stratify patients in complete biochemical remission. Consequently, NGS techniques have been developed to monitor the size of the clone to a sensitivity of up to a cell in a million after treatment. However, even these techniques are not within reach of standard laboratories. In this review we will recapitulate recent advances in multiple myeloma genomics, with special focus on the ones that may have immediate translational impact. We will analyze the benefits and pitfalls of NGS-based diagnostics, highlighting crucial aspects that will need to be taken into account before this can be implemented in most laboratories. We will make the point that a new era in myeloma diagnostics and minimal residual disease monitoring is close and conventional genetic testing will not be able to return the required information. This will mandate that even in routine practice NGS should soon be adopted owing to a higher informative potential with increasing clinical benefits.
Collapse
Affiliation(s)
- Niccolo Bolli
- Department of Clinical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
| | - Elisa Genuardi
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Bachisio Ziccheddu
- Department of Clinical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Marina Martello
- Seràgnoli Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Stefania Oliva
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Carolina Terragna
- Seràgnoli Institute of Hematology, Azienda Ospedaliero-Universitaria Sant'Orsola-Malpighi, Bologna, Italy
| |
Collapse
|
27
|
Takamatsu H. Clinical value of measurable residual disease testing for multiple myeloma and implementation in Japan. Int J Hematol 2020; 111:519-529. [DOI: 10.1007/s12185-020-02828-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/15/2020] [Indexed: 11/24/2022]
|
28
|
Oliva S, D'Agostino M, Boccadoro M, Larocca A. Clinical Applications and Future Directions of Minimal Residual Disease Testing in Multiple Myeloma. Front Oncol 2020; 10:1. [PMID: 32076595 PMCID: PMC7006453 DOI: 10.3389/fonc.2020.00001] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/02/2020] [Indexed: 12/20/2022] Open
Abstract
In the last years, the life expectancy of multiple myeloma (MM) patients has substantially improved thanks to the availability of many new drugs. Our ability to induce deep responses has improved as well, and the treatment goal in patients tolerating treatment moved from the delay of progression to the induction of the deepest possible response. As a result of these advances, a great scientific effort has been made to redefine response monitoring, resulting in the development and validation of high-sensitivity techniques to detect minimal residual disease (MRD). In 2016, the International Myeloma Working Group (IMWG) updated MM response categories defining MRD-negative responses both in the bone marrow (assessed by next-generation flow cytometry or next-generation sequencing) and outside the bone marrow. MRD is an important factor independently predicting prognosis during MM treatment. Moreover, using novel combination therapies, MRD-negative status can be achieved in a fairly high percentage of patients. However, many questions regarding the clinical use of MRD status remain unanswered. MRD monitoring can guide treatment intensity, although well-designed clinical trials are needed to demonstrate this potential. This mini-review will focus on currently available techniques and data on MRD testing and their potential future applications.
Collapse
Affiliation(s)
- Stefania Oliva
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Cittá Della Salute e Della Scienza di Torino, Turin, Italy
| | - Mattia D'Agostino
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Cittá Della Salute e Della Scienza di Torino, Turin, Italy
| | - Mario Boccadoro
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Cittá Della Salute e Della Scienza di Torino, Turin, Italy
| | - Alessandra Larocca
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Cittá Della Salute e Della Scienza di Torino, Turin, Italy
| |
Collapse
|
29
|
Kunacheewa C, Lee HC, Patel K, Thomas S, Amini B, Srour S, Bashir Q, Nieto Y, Qazilbash MH, Weber DM, Feng L, Orlowski RZ, Lin P, Manasanch EE. Minimal Residual Disease Negativity Does Not Overcome Poor Prognosis in High-Risk Multiple Myeloma: A Single-Center Retrospective Study. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e221-e238. [PMID: 32037287 DOI: 10.1016/j.clml.2020.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 12/28/2019] [Accepted: 01/02/2020] [Indexed: 01/14/2023]
Abstract
BACKGROUND Minimal residual disease (MRD) is a standard measurement for response assessment in multiple myeloma (MM). Despite new treatments, high-risk MM patients continue to have poor prognosis. We evaluated the effect of MRD negativity in high-risk versus standard-risk patients. PATIENTS AND METHODS We retrospectively evaluated all consecutive MM patients who underwent routine MRD testing by 1-tube 8-color advanced flow cytometry with 2,000,000 events and sensitivity level 10-5 at our center from 2015 to 2018 after initial therapy. Kaplan-Meier and log-rank test were used to assess survival estimates and differences between study groups. RESULTS One hundred thirty-six patients with MRD testing after initial therapy or autologous stem-cell transplantation were identified. At a median follow-up of 14 months (range, 1-36 months), progression-free survival and overall survival were significantly worse in high-risk versus standard-risk patients. During the study period, 50% of high-risk group had experienced disease progression (relapse and/or death) versus 20% in the standard-risk group (P = .0006). No patients with standard-risk died, but 4 (14%) in the high-risk group did (P = .0007). Regardless of MRD status, high-risk patients had statistically significant worse progression-free survival than standard-risk patients. At median follow-up, those with disease 10% standard-risk/MRD negative; 20% standard-risk/MRD positive; 40% high-risk/MRD negative; and 45% high-risk/MRD positive had either experienced relapse or died (P = .0041). MRD status did not significantly affect overall survival in either group (P = .0914); however, longer follow-up is needed to assess survival. CONCLUSION Genetic abnormalities remain a powerful prognostic indicator for MM, regardless of MRD status. For newly diagnosed MM patients treated with novel triple-drug initial therapy and frontline autologous stem-cell transplantation, MRD-negative status did not mitigate the poor-prognosis outcomes of high-risk MM patients.
Collapse
Affiliation(s)
- Chutima Kunacheewa
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hans C Lee
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Krina Patel
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sheeba Thomas
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Behrang Amini
- Division of Diagnostic Imaging, Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Samer Srour
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Qaiser Bashir
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yago Nieto
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Muzzaffar H Qazilbash
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Donna M Weber
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lei Feng
- Department of Statistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Pei Lin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elisabet E Manasanch
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
30
|
Vrabel D, Sedlarikova L, Besse L, Rihova L, Bezdekova R, Almasi M, Kubaczkova V, Brožová L, Jarkovsky J, Plonkova H, Jelinek T, Sandecka V, Stork M, Pour L, Sevcikova S, Hajek R. Dynamics of tumor-specific cfDNA in response to therapy in multiple myeloma patients. Eur J Haematol 2019; 104:190-197. [PMID: 31763708 PMCID: PMC7065130 DOI: 10.1111/ejh.13358] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 12/14/2022]
Abstract
Objectives Progress in multiple myeloma treatment allows patients to achieve deeper responses, for which the assessment of minimal residual disease (MRD) is critical. Typically, bone marrow samples are used for this purpose; however, this approach is site‐limited. Liquid biopsy represents a minimally invasive and more comprehensive technique that is not site‐limited, but equally challenging. Methods While majority of current data comes from short‐term studies, we present a long‐term study on blood‐based MRD monitoring using tumor‐specific cell‐free DNA detection by ASO‐qPCR. One hundred and twelve patients were enrolled into the study, but long‐term sampling and analysis were feasible only in 45 patients. Results We found a significant correlation of quantity of tumor‐specific cell‐free DNA levels with clinically meaningful events [induction therapy (P = .004); ASCT (P = .012)]. Moreover, length of cfDNA fragments is associated with better treatment response of patients. Conclusions These results support the concept of tumor‐specific cell‐free DNA as a prognostic marker.
Collapse
Affiliation(s)
- David Vrabel
- Babak Myeloma Group, Department of Pathophysiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Lenka Sedlarikova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, Czech Republic
| | - Lenka Besse
- Laboratory of Experimental Oncology, Department of Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Lucie Rihova
- Department of Clinical Hematology, University Hospital Brno, Brno, Czech Republic
| | - Renata Bezdekova
- Department of Clinical Hematology, University Hospital Brno, Brno, Czech Republic
| | - Martina Almasi
- Department of Clinical Hematology, University Hospital Brno, Brno, Czech Republic
| | - Veronika Kubaczkova
- Babak Myeloma Group, Department of Pathophysiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lucie Brožová
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jiri Jarkovsky
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Hana Plonkova
- Department of Hematooncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Tomas Jelinek
- Department of Hematooncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Viera Sandecka
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Martin Stork
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Ludek Pour
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | - Sabina Sevcikova
- Babak Myeloma Group, Department of Pathophysiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Roman Hajek
- Department of Hematooncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| |
Collapse
|
31
|
Bai Y, Chim CS. Ficolled bone marrow is superior to bone marrow buffy coat for detection of minimal residual disease in multiple myeloma. ACTA ACUST UNITED AC 2019; 24:533-537. [PMID: 31280705 DOI: 10.1080/16078454.2019.1637574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Buffy coat and ficoll of bone marrow (BM) are viable options for the study of minimal residual disease (MRD) in multiple myeloma (MM). As yet, there is no data about the superiority of either sample types. Herein, we aimed to address this issue. METHODS Forty pairs of ficolled BMs and BM buffy coats of 19 MM patients were studied for MRD by allele-specific oligonucleotide real-time quantitative PCR, with patient-specific primers/probes whenever appropriate. RESULTS There were 41 pairs of MRD data for comparison analysis due to one patient with biclonal disease. MRD levels in ficolls and buffy coats were highly concordant (rs = 0.936, P < 0.0001), with 31 (76%) and seven (17%) pairs being concomitantly MRD-positive or -negative. On the other hand, apart from the 16 pairs being both MRD-negative, or -positive but not quantifiable in ficolls and buffy coats, majority (n = 22, 88%) had higher MRD levels in ficolled BMs than BM buffy coats. Furthermore, in 17 pairs, in which MRD was quantifiable in both, MRD levels in ficolled BMs were 3.1 times those of BM buffy coats (median, 567/105 vs. 184/105, P = 0.001). CONCLUSION Taken together, ficolled BM is more sensitive than BM buffy coat for MRD detection in MM, hence should be recommended.
Collapse
Affiliation(s)
- Yinlei Bai
- a Institute for Immunology and School of Medicine , Tsinghua University , Beijing , People's Republic of China
| | - Chor Sang Chim
- b Department of Medicine , The University of Hong Kong , Hong Kong , People's Republic of China
| |
Collapse
|
32
|
Yao WQ, Zhu MQ, Yan LZ, Jin S, Shang JJ, Yao Y, Yan S, Liu Y, Wu DP, Fu CC. [Clinical implication of minimal residual disease monitoring by 10-color flow cytometry in multiple myeloma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:720-725. [PMID: 31648471 PMCID: PMC7342455 DOI: 10.3760/cma.j.issn.0253-2727.2019.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
目的 采用十色流式细胞术监测多发性骨髓瘤(MM)患者治疗后骨髓微小残留病(MRD),探讨其对MM患者预后的预测价值。 方法 回顾性分析2015年7月至2017年7月苏州大学附属第一医院收治的150例MM患者的临床资料,十色流式细胞术检测MRD水平。 结果 ①诱导治疗结束后采用十色流式细胞术监测MRD的87例MM患者中,MRD阴性者34例(39.1%)。自体造血干细胞移植后1年内行十色流式细胞术监测MRD的69例患者中,MRD阴性者34例(49.3%),诱导治疗结束和移植后MRD阴性患者的无进展生存(PFS)期优于MRD阳性患者(未达到对21个月,P=0.022;未达到对18个月,P<0.001)。②根据移植前后MRD的动态改变将MM患者分为MRD持续阴性、阳性转阴性、持续阳性、阴性转阳性四组,四组的2年PFS率分别为83%、82%、44%、0(P=0.002)。③多因素分析显示诱导治疗结束后的MRD水平是影响PFS的独立预后因素[P=0.002,HR=4.808(95%CI 1.818~12.718)]。 结论 治疗后MRD转阴提示更好的临床预后,MRD水平对于MM患者的预后价值优于血清学疗效评估,可以联合R-ISS分期及细胞遗传学异常评估患者预后。
Collapse
Affiliation(s)
- W Q Yao
- The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou 215006, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Impact of Minimal Residual Disease Detection by Next-Generation Flow Cytometry in Multiple Myeloma Patients with Sustained Complete Remission after Frontline Therapy. Hemasphere 2019; 3:e300. [PMID: 31976475 PMCID: PMC6924563 DOI: 10.1097/hs9.0000000000000300] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/27/2019] [Accepted: 09/15/2019] [Indexed: 02/05/2023] Open
Abstract
Minimal residual disease (MRD) was monitored in 52 patients with sustained CR (≥2 years) after frontline therapy using next-generation flow (NGF) cytometry. 25% of patients initially MRD- reversed to MRD+. 56% of patients in sustained CR were MRD+; 45% at the level of 10−5; 17% at 10−6. All patients who relapsed during follow-up were MRD+ at the latest MRD assessment, including those with ultra-low tumor burden. MRD persistence was associated with specific phenotypic profiles: higher erythroblasts’ and tumor-associated monocytes/macrophages’ predominance in the bone marrow niche. NGF emerges as a suitable method for periodic, reproducible, highly-sensitive MRD-detection at the level of 10−6.
Collapse
|
34
|
Romano A, Palumbo GA, Parrinello NL, Conticello C, Martello M, Terragna C. Minimal Residual Disease Assessment Within the Bone Marrow of Multiple Myeloma: A Review of Caveats, Clinical Significance and Future Perspectives. Front Oncol 2019; 9:699. [PMID: 31482061 PMCID: PMC6710454 DOI: 10.3389/fonc.2019.00699] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022] Open
Abstract
There is an increasing clinical interest in the measure and achievement of minimal residual disease (MRD) negativity in the bone marrow of Multiple Myeloma (MM) patients, as defined equally either by Multicolor Flow Cytometry (MFC) or by Next Generation Sequencing (NGS) technologies. At present, modern technologies allow to detect up to one on 104 or on 105 or even on 106 cells, depending on their throughput. MFC approaches, which have been progressively improved up to the so-called Next Generation Flow (NGF), and NGS, which proved clear advantages over ASO-PCR, can detect very low levels of residual disease in the BM. These methods are actually almost superimposable, in terms of MRD detection power, supporting the lack of unanimous preference for either technique on basis of local availability. However, some technical issues are still open: the optimal assay to use to detect either phenotype (e.g., next generation multidimensional flow cytometry, imaging) or genotype aberrations (e.g., ASO-RQ PCR, digital droplet PCR, NGS) and their standardization, the sample source (BM or peripheral blood, PB) and its pre-processing (red-cell lysis vs. Ficoll, fresh vs. frozen samples, requirement of CD138+ cells enrichment). Overall, MRD negativity is considered as the most powerful predictor of favorable long-term outcomes in MM and is likely to represent the major driver of treatment strategies in the near future. In this manuscript, we reviewed the main pitfalls and caveats of MRD detection within bone marrow in MM patients after front-line therapy, highlighting the improving of the currently employed technology and describing alternative methods for MRD testing in MM, such as liquid biopsy.
Collapse
Affiliation(s)
- Alessandra Romano
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
| | - Giuseppe Alberto Palumbo
- Division of Hematology, Azienda Ospedaliero-Universitaria Policlinico Vittorio Emanuele di Catania, Catania, Italy
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie avanzate “G.F. Ingrassia,” University of Catania, Catania, Italy
| | - Nunziatina Laura Parrinello
- Division of Hematology, Azienda Ospedaliero-Universitaria Policlinico Vittorio Emanuele di Catania, Catania, Italy
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie avanzate “G.F. Ingrassia,” University of Catania, Catania, Italy
| | - Concetta Conticello
- Division of Hematology, Azienda Ospedaliero-Universitaria Policlinico Vittorio Emanuele di Catania, Catania, Italy
| | - Marina Martello
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale (DIMES), Università degli Studi di Bologna, Bologna, Italy
| | - Carolina Terragna
- Istituto di Ematologia “L.A.Seràgnoli,” Azienda Ospedaliera Sant'Orsola-Malpighi, Bologna, Italy
| |
Collapse
|
35
|
Alonso R, Cedena MT, Gómez‐Grande A, Ríos R, Moraleda JM, Cabañas V, Moreno MJ, López‐Jiménez J, Martín F, Sanz A, Valeri A, Jiménez A, Sánchez R, Lahuerta JJ, Martínez‐López J. Imaging and bone marrow assessments improve minimal residual disease prediction in multiple myeloma. Am J Hematol 2019; 94:853-861. [PMID: 31074033 DOI: 10.1002/ajh.25507] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 05/04/2019] [Accepted: 05/07/2019] [Indexed: 01/01/2023]
Abstract
The value of minimal residual disease (MRD) status by bone marrow and imaging analysis as independent prognostic factors has been well established in multiple myeloma (MM). Nevertheless data about their potential complementarity for a more accurate assessment are limited. With this aim, we retrospectively analyzed the prediction of outcome with the combination of PET-CT and MRD, assessed by multiparameter flow cytometry (MFC) in 103 patients with newly diagnosed MM. We confirmed the benefit in terms of progression-free survival (PFS), linked to the achievement of negativity by MFC (hazard ratio [HR] 0.53; 95% confidence interval [CI]: 0.28-0.98), and PET-CT (HR 0.18; 95% CI: 0.09-0.36) individually. By combining both techniques, patients who became MRD-/PET-, with a median of PFS 92 months, had significant prolonged median PFS (P < .001). This is compared with MRD+/PET- and PET+ patients (median PFS of 45 and 28 months, respectively). We observed a significant difference (P = .003) in overall survival (OS) outcomes between MRD-/PET- and MRD+/PET- patients (4-year OS 94.2% and 100%, respectively), vs PET+ patients (4-year OS 73.8%). All survival results were confirmed in a conditional landmark analysis. These findings support the potential complementarity between PET-CT and MFC, and highlight their better predictive capability when improving sensitivity.
Collapse
Affiliation(s)
- Rafael Alonso
- Department of Hematology , Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, Departamento de MedicinaH12O CNIO Clinical Research Hematology Unit; CIBERONC Madrid Spain
| | - María Teresa Cedena
- Department of Hematology , Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, Departamento de MedicinaH12O CNIO Clinical Research Hematology Unit; CIBERONC Madrid Spain
| | - Adolfo Gómez‐Grande
- Department of Nuclear MedicineHospital Universitario 12 de Octubre Madrid Spain
| | - Rafael Ríos
- Department of HematologyHospital Universitario Virgen de las Nieves Granada Spain
| | - José María Moraleda
- Department of HematologyHospital Clínico Universitario Virgen de la Arrixaca Murcia Spain
| | - Valentín Cabañas
- Department of HematologyHospital Clínico Universitario Virgen de la Arrixaca Murcia Spain
| | - María José Moreno
- Department of HematologyHospital Clínico Universitario Virgen de la Arrixaca Murcia Spain
| | | | - Fernando Martín
- Department of HematologyHospital Universitario Ramón y Cajal Madrid Spain
| | - Alejandro Sanz
- Department of HematologyHospital Universitario Ramón y Cajal Madrid Spain
| | - Antonio Valeri
- Department of Hematology , Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, Departamento de MedicinaH12O CNIO Clinical Research Hematology Unit; CIBERONC Madrid Spain
| | - Ana Jiménez
- Department of Hematology , Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, Departamento de MedicinaH12O CNIO Clinical Research Hematology Unit; CIBERONC Madrid Spain
| | - Ricardo Sánchez
- Department of Hematology , Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, Departamento de MedicinaH12O CNIO Clinical Research Hematology Unit; CIBERONC Madrid Spain
| | - Juan José Lahuerta
- Department of Hematology , Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, Departamento de MedicinaH12O CNIO Clinical Research Hematology Unit; CIBERONC Madrid Spain
| | - Joaquín Martínez‐López
- Department of Hematology , Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, Departamento de MedicinaH12O CNIO Clinical Research Hematology Unit; CIBERONC Madrid Spain
| |
Collapse
|
36
|
Berger N, Kim-Schulze S, Parekh S. Minimal Residual Disease in Multiple Myeloma: Impact on Response Assessment, Prognosis and Tumor Heterogeneity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1100:141-159. [PMID: 30411265 DOI: 10.1007/978-3-319-97746-1_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Multiple Myeloma (MM) therapy has evolved rapidly over the past decade. With current multidrug combinations and autologous transplant, rates of overall response exceed 90% and complete response (CR) more than 50% in some studies. Unfortunately, despite higher rates of CR, relapse rates remain high suggesting that persistent disease may not be measured by current techniques. Traditionally, response rates were defined by urine and serum protein electrophoresis, immunofixation and histopathological absence of clonal plasma cells in the bone marrow. Currently, there are several validated sensitive assays to evaluate for MRD (minimal residual disease); multiparameter flow cytometry (MFC) including nextgeneration flow cytometry (NGF), next-generation sequencing (NGS), and allele specific oligonucleotide quantitative polymerase chain reaction (ASO-qPCR). These methods have provided a means to quantitatively assess residual disease and accurately prognosticate PFS and OS in myeloma. In this chapter, we will discuss the current techniques for MRD detection as well as describe techniques that are emerging for improved characterization of drug resistant residual populations that could be adapted for MRD monitoring in the future. While improved therapies are able to eradicate the dominant clone, resistant sub-clones persist and remain undetectable even by MRD techniques. Characterization of these clones will help design therapies against drug-resistant clones and move us closer to a cure in MM.
Collapse
Affiliation(s)
- Natalie Berger
- Hematology/Oncology Fellowship Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- Department of Oncological Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samir Parekh
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
37
|
Soh KT, Wallace PK. Monitoring of Measurable Residual Disease in Multiple Myeloma by Multiparametric Flow Cytometry. ACTA ACUST UNITED AC 2019; 90. [PMID: 31608132 DOI: 10.1002/cpcy.63] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent interest in high sensitivity multiple myeloma (MM) measurable residual disease (MRD) testing is a direct consequence of the high-quality responses achieved using novel therapeutic agents and better treatment strategies. Traditional diagnostic measures such as immunohistochemistry and morphology have detection sensitivities of only 10-2 - 10-3, which do not reliably predict progression free survival (PFS) or overall survival (OS) after these treatments. Contemporary monitoring of MM MRD has switched to more sensitive platforms such as quantitative allele-specific oligonucleotide polymerase chain reaction (ASO-qPCR), next-generation sequencing (NGS), and multiparametric flow cytometry (MFC). Though both ASO-qPCR and NGS have excellent detection sensitivities (10-5 - 10-6), both technologies have lower applicability when compared to MFC. Conventional MFC can easily reach a detection sensitivity of 10-4 and when optimized can achieve a sensitivity of 10-5 - 10-6. Current consensus guidelines require a minimum of 2 million and recommend 5 million events be acquired to reach a minimum sensitivity of 10-5. As conventional immunophenotyping protocols are unable to attain these numbers, alternative MFC staining procedures are required. This manuscript describes two high-sensitivity MFC approaches that can be used for MM MRD testing.
Collapse
Affiliation(s)
- Kah Teong Soh
- Roswell Park Comprehensive Cancer Center, Department of Flow and Image Cytometry, Elm & Carlton Streets, Buffalo, New York 14263
| | - Paul K Wallace
- Roswell Park Comprehensive Cancer Center, Department of Flow and Image Cytometry, Elm & Carlton Streets, Buffalo, New York 14263
| |
Collapse
|
38
|
Yang PY, Liu MM, Fan HQ, Yang YP, Han W, Yu XY, Yue TT, Su KJ, Guo Q, Gao SJ, Jin FY. [The prognostic significance of dynamic monitoring of minimal residual disease (MRD) status in patients with newly-diagnosed multiple myeloma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:584-588. [PMID: 32397022 PMCID: PMC7364907 DOI: 10.3760/cma.j.issn.0253-2727.2019.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Indexed: 11/05/2022]
Abstract
Objective: To evaluate the prognostic value of kinetic changes in minimal residual disease (MRD) status, as well as its relationship with risk stratification, therapeutic response and treatment in patients with newly-diagnosed multiple myeloma (MM) . Methods: A total of 135 patients with newly-diagnosed MM were screened, and 105 patients who achieved VGPR or more as the best responses were included into this study. The MRD status was determined by multiparameter flow cytometry (MFC) at multiple intervals after two cycles of treatment until clinical relapse, death, or last follow-up. The statistical methods included Kaplan-Meier analysis, Cox regression, etc. Results: ①In all 135 patients, 57.8% (78/135) patients achieved MRD negativity (MRD(-)) after treatment. In 105 patients who achieved VGPR and thus included in this study, the MRD(-) rate was 72.4% (76/105) , with a median interval of 3 months from starting treatment to achievement of MRD(-) status. ②The 2-year PFS rate of patients with MRD(-) status was significantly higher than that of MRD(+) status (62.2% vs 41.3%, P=0.001) , while MRD persistence (MRD(+)) was an independent factor for poor prognosis (multivariate analysis for PFS: P=0.044, HR=3.039, 95%CI 1.029-8.974) . ③Loss of MRD(-) status (i.e., MRD reappearance) showed inferior outcomes compared with MRD sustained negative ones, the PFS was 18 months versus not reach (P<0.001) and the OS was not reach for both (P=0.002) . ④The 2-year PFS and OS rates of patients with duration of MRD(-)status≥12 months were significantly higher than those of the control group (PFS: 77.7% vs 36.7%, P<0.001; OS: 96.4% vs 57.9%, P<0.001 respectively) . Duration of MRD(-) status was associated with a marked reduction in risk of relapse or death (univariate analysis for PFS: P<0.001, HR=0.865, 95%CI 0.815-0.918; for OS: P=0.001, HR=0.850, 95%CI 0.741-0.915 respectively) . ⑤Moreover, even in patients carrying high-risk cytogenetic abnormalities (CA) or ineligible for ASCT, MRD negativity remained its prognostic value to predict PFS (high-risk CA medianPFS: not reach vs 19 months, P=0.006; ineligible for ASCT medianPFS: not reach vs 25 months, P=0.052 respectively) . ⑥Last, treatment with the bortezomib-based regimens contributed to prolonged MRD(-) duration (median MRD(-) duratio: 25 months vs 10 months, P=0.034) . Conclusion: Our findings supported MRD(+) status as an independent poor prognostic factor in MM patients, which implicated that duration of MRD(-) status also played a significant role in evaluation of prognosis, while loss of MRD(-)status might serve as an early biomarker for relapse. Therefore, monitoring of MRD kinetics might more precisely predict prognosis, as well as guide treatment decision, especially for when to start retreatment in relapsed patients.
Collapse
Affiliation(s)
- P Y Yang
- Department of Hematology, the First Hospital of Jilin University, Changchun 130021, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
|
40
|
Owen RG. Minimal residual disease (mrd) in multiple myeloma: prognostic and therapeutic implications (including imaging). Hemasphere 2019; 3:HemaSphere-2019-0059. [DOI: 10.1097/hs9.0000000000000243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 04/01/2019] [Indexed: 11/25/2022] Open
|
41
|
Blum A, Haussmann K, Streitz M, Schlickeiser S, Tietze-Buerger C, Blau IW, Uharek L. Standardized assay for assessment of minimal residual disease in blood, bone marrow and apheresis from patients with plasma cell myeloma. Sci Rep 2019; 9:2922. [PMID: 30814612 PMCID: PMC6393516 DOI: 10.1038/s41598-019-39631-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 01/24/2019] [Indexed: 12/21/2022] Open
Abstract
The recent advances in myeloma treatment result in significantly better outcomes, defined as increased progression free survival (PFS) and overall survival (OS). Since there is a proven correlation between the extend of response and prolonged survival, there is an urgent need for highly sensitive assays for the detection of minimal residual disease (MRD). Next generation flow cytometry has become a valuable approach for sensitive evaluation of the depth of complete response (CR). Here, we report the diagnostic performance and validation results of a single-tube 9-color panel assay. The validation design included intra-assay analysis measuring accuracy, inter-assay analysis estimating method’s linearity and precision and inter-assay analysis evaluating repeatability. Furthermore, in inter-operator analysis assessed the comparability of the result analysis of different operators. Staining stability was evaluated in age-of-stain experiments. Our validation results show that a reliable detection of residual myeloma cells is feasible to a detection level of 10−5 with a single-tube assay for a variety of materials (peripheral blood, bone marrow and stem cell apheresis). This study establishes highly sensitive, fully standardized approach for MRD detection in myeloma that is ready for implementation in routine diagnostic laboratories.
Collapse
Affiliation(s)
- Agnieszka Blum
- Charité Stem Cell Facility, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, Berlin, 13353, Germany.
| | - Katy Haussmann
- Charité Stem Cell Facility, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Mathias Streitz
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Stephan Schlickeiser
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Carola Tietze-Buerger
- Charité Stem Cell Facility, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Igor Wolfgang Blau
- Department of Haematology, Oncology, Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Lutz Uharek
- Charité Stem Cell Facility, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, Berlin, 13353, Germany
| |
Collapse
|
42
|
Abstract
The quantification of submicroscopic minimal residual disease (MRD) after therapy proved to have independent prognostic significance in many mature B-cell malignancies. With the advent of routine benchtop cytometers capable of simultaneously analyzing ≥4 colors and with improved standardization, flow cytometry has become the method of choice for MRD assessments in some lymphoma entities. Herein we describe general aspects of flow cytometric standardization. Chronic lymphocytic leukemia and multiple myeloma (MM) are used as examples to explain the technical standardization of flow cytometry for MRD detection according to EuroFlow strategies. MRD data acquisition and detailed analysis using a newly developed approach (so-called next generation flow, NGF) in MM is a particular focus of this chapter.
Collapse
|
43
|
Scott SD, Fletcher M, Whitehouse H, Whitby L, Yuan C, Mazzucchelli S, Lin P, de Tute R, Dorwal P, Wallace PK, Tembhare P, Arroz M, Snowden JA, Chantry AD, Barnett D. Assessment of plasma cell myeloma minimal residual disease testing by flow cytometry in an international inter‐laboratory study: Is it ready for primetime use? CYTOMETRY PART B-CLINICAL CYTOMETRY 2018; 96:201-208. [DOI: 10.1002/cyto.b.21754] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 10/26/2018] [Accepted: 11/15/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Stuart D. Scott
- UK NEQAS for Leucocyte ImmunophenotypingSheffield Teaching Hospitals Sheffield UK
- Department of Oncology and Metabolism, Faculty of Medicine, Dentistry and HealthUniversity of Sheffield Sheffield UK
| | - Matthew Fletcher
- UK NEQAS for Leucocyte ImmunophenotypingSheffield Teaching Hospitals Sheffield UK
| | - Helen Whitehouse
- UK NEQAS for Leucocyte ImmunophenotypingSheffield Teaching Hospitals Sheffield UK
| | - Liam Whitby
- UK NEQAS for Leucocyte ImmunophenotypingSheffield Teaching Hospitals Sheffield UK
| | - Constance Yuan
- Clinical Flow Cytometry Laboratory, Laboratory of PathologyCCR, NCI, NIH Bethesda Maryland
| | - Silvia Mazzucchelli
- Department of Haematology and Flow CytometrySynlab Suisse SA Bioggio Switzerland
| | - Pei Lin
- Department of HematopathologyMD Anderson Cancer Center Houston Texas
| | - Ruth de Tute
- HMDS, Department of HaematologySt. James's Institute of Oncology Leeds UK
| | - Pranav Dorwal
- Flow Cytometry LaboratoryWaikato Hospital Hamilton New Zealand
| | - Paul K. Wallace
- Department of Flow and Image CytometryRoswell Park Cancer Institute Buffalo New York
| | - Prashant Tembhare
- Hematopathology LaboratoryTata Memorial Center Mumbai Maharashtra India
| | - Maria Arroz
- Flow Cytometry Laboratory, Department of Clinical PathologyCHLO S. Francisco Xavier Hospital Lisbon Portugal
| | - John A. Snowden
- Department of Oncology and Metabolism, Faculty of Medicine, Dentistry and HealthUniversity of Sheffield Sheffield UK
- Department of HaematologySheffield Teaching Hospitals NHS Foundation Trust Sheffield UK
| | - Andrew D. Chantry
- Department of Oncology and Metabolism, Faculty of Medicine, Dentistry and HealthUniversity of Sheffield Sheffield UK
- Department of HaematologySheffield Teaching Hospitals NHS Foundation Trust Sheffield UK
| | - David Barnett
- UK NEQAS for Leucocyte ImmunophenotypingSheffield Teaching Hospitals Sheffield UK
- Department of Oncology and Metabolism, Faculty of Medicine, Dentistry and HealthUniversity of Sheffield Sheffield UK
- Department of HaematologySheffield Teaching Hospitals NHS Foundation Trust Sheffield UK
| |
Collapse
|
44
|
Minimal residual disease by flow cytometry and allelic‐specific oligonucleotide real‐time quantitative polymerase chain reaction in patients with myeloma receiving lenalidomide maintenance: A pooled analysis. Cancer 2018; 125:750-760. [DOI: 10.1002/cncr.31854] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/14/2018] [Accepted: 10/12/2018] [Indexed: 11/07/2022]
|
45
|
Levin A, Hari P, Dhakal B. Novel biomarkers in multiple myeloma. Transl Res 2018; 201:49-59. [PMID: 30301522 DOI: 10.1016/j.trsl.2018.05.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/10/2018] [Accepted: 05/22/2018] [Indexed: 11/24/2022]
Abstract
Significant advancements have been made in the molecular mechanisms of myelomagenesis, diagnostic methods, prognostication, and the treatment options in multiple myeloma (MM) over the last decade. Despite these, MM remains a heterogeneous disease with differing outcomes. As myeloma treatment landscape continues to expand, personalized treatment that provides maximum benefit to a specific patient becomes more important. In the last few years, serum monoclonal proteins including the serum-free light chain assays, imaging, and cytogenetics have been used to predict the outcomes of MM patients receiving different types of therapies. With the development of novel technologies, more sensitive detection of residual disease using flow cytometry and next-generation sequencing has been possible. In addition, liquid biopsies using circulating tumor cells, tumor DNA, and novel immune biomarkers are potentially being investigated. These novel potential biomarkers not only accurately detect the mutational landscape of different cancers compared to standard methods but also serve as prognostic and predictive biomarkers for disease relapse and response to therapy. It is likely that we will be able to offer more targeted and risk-adapted therapeutic approach to patients with MM at different stages of their disease guided by these potential biomarkers.
Collapse
Affiliation(s)
- Adam Levin
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Parameswaran Hari
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Binod Dhakal
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
46
|
Shires K, Van Wyk T. The role of Cancer/Testis Antigens in Multiple Myeloma pathogenesis and their application in disease monitoring and therapy. Crit Rev Oncol Hematol 2018; 132:17-26. [PMID: 30447924 DOI: 10.1016/j.critrevonc.2018.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/22/2018] [Accepted: 09/12/2018] [Indexed: 12/17/2022] Open
Abstract
A unique group of genes, encoding tumour associated antigens, known as the Cancer/Testis Antigens (CTAs), have been explored as novel markers of disease progression and as targets of immunotherapy in several cancers, including the haematological malignancy Multiple Myeloma (MM). This review aims to update the knowledge of CTA involvement in MM pathogenesis and how their potential as biomarkers for disease monitoring and targets of immunotherapy has been explored in the MM disease arena. Despite the initial promise of these antigens, their use as immunotherapy targets has not been successful, yet with a greater understanding of their role in disease pathogenesis they may still have a significant role to play as biomarkers of disease and therapeutic targets.
Collapse
Affiliation(s)
- Karen Shires
- Division of Haematology, Department of Pathology, University of Cape Town and National Health Laboratory Service/Groote Schuur Hospital, Cape Town, South Africa.
| | - Teagan Van Wyk
- Department of Medicine, University of Cape Town, South Africa
| |
Collapse
|
47
|
Caers J, Garderet L, Kortüm KM, O'Dwyer ME, van de Donk NWCJ, Binder M, Dold SM, Gay F, Corre J, Beguin Y, Ludwig H, Larocca A, Driessen C, Dimopoulos MA, Boccadoro M, Gramatzki M, Zweegman S, Einsele H, Cavo M, Goldschmidt H, Sonneveld P, Delforge M, Auner HW, Terpos E, Engelhardt M. European Myeloma Network recommendations on tools for the diagnosis and monitoring of multiple myeloma: what to use and when. Haematologica 2018; 103:1772-1784. [PMID: 30171031 PMCID: PMC6278986 DOI: 10.3324/haematol.2018.189159] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 08/27/2018] [Indexed: 01/04/2023] Open
Abstract
The diagnosis of multiple myeloma can be challenging, even for experienced physicians, and requires close collaboration between numerous disciplines (orthopedics, radiology, nuclear medicine, radiation therapy, hematology and oncology) before the final diagnosis of myeloma is made. The definition of multiple myeloma is based on the presence of clinical, biochemical, histopathological, and radiological markers of disease. Specific tests are needed both at presentation and during follow-up in order to reach the correct diagnosis and characterize the disease precisely. These tests can also serve prognostic purposes and are useful for follow-up of myeloma patients. Molecular analyses remain pivotal for defining high-risk myeloma and are used in updated patient stratifications, while minimal residual disease assessment via flow cytometry, molecular techniques and radiological approaches provides additional prognostic information on patients' long-term outcome. This pivotal information will guide our future treatment decisions in forthcoming clinical trials. The European Myeloma Network group updated their guidelines on different diagnostic recommendations, which should be of value to enable appropriate use of the recommendations both at diagnosis and during follow-up.
Collapse
Affiliation(s)
- Jo Caers
- Department of Hematology, University Hospital of Liege, Belgium .,Laboratory of Hematology, GIGA-I3, University of Liège, Belgium
| | | | - K Martin Kortüm
- Department of Internal Medicine II, University Hospital of Wuerzburg, Germany
| | - Michael E O'Dwyer
- Department of Hematology, National University of Ireland Galway, Ireland
| | | | - Mascha Binder
- Department of Internal Medicine II, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra Maria Dold
- Department of Medicine I, Hematology, Oncology & Stem Cell Transplantation, Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Francesca Gay
- Department of Hematology-Oncology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Jill Corre
- Unit for Genomics in Myeloma, Institut Universitaire du Cancer - Oncopole, Toulouse, France
| | - Yves Beguin
- Department of Hematology, University Hospital of Liege, Belgium.,Laboratory of Hematology, GIGA-I3, University of Liège, Belgium
| | - Heinz Ludwig
- Wilhelminen Cancer Research Institute, Vienna, Austria
| | - Alessandra Larocca
- Department of Hematology-Oncology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Christoph Driessen
- Department of Oncology and Hematology, Cantonal Hospital St. Gallen, Switzerland
| | | | - Mario Boccadoro
- Department of Hematology-Oncology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Martin Gramatzki
- Division of Stem Cell Transplantation and Immunotherapy, University of Kiel, Germany
| | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Wuerzburg, Germany
| | - Michele Cavo
- Seragnoli 'Institute of Hematology, Bologna University School of Medicine, Italy
| | - Hartmut Goldschmidt
- Department of Hematology, Rheumatology and Oncology, University Hospital Heidelberg, Germany.,National Center for Tumor Diseases, Heidelberg Medical University, Germany
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Michel Delforge
- Department of Hematology, University Hospital Leuven, Belgium
| | - Holger W Auner
- Centre for Haematology, Hammersmith Hospital, Imperial College London, UK
| | - Evangelos Terpos
- School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Monika Engelhardt
- Department of Medicine I, Hematology, Oncology & Stem Cell Transplantation, Medical Center, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
48
|
Abstract
The primary hurdle in the path to curing multiple myeloma (MM) is defining a validated minimal residual disease (MRD) and its utility in the therapeutic decision making. A better definition of MRD will aid in tailoring MM therapy further to address the clonal heterogeneity and genomic instability and overcome patient's ineffective immune surveillance. MRD analysis can define the logical endpoint for maintenance therapy, in addition also aids in providing a better clinical end point for studies comparing novel agents in myeloma. MRD is a surrogate for the survival in MM. Guidelines for global incorporation of MRD in myeloma are fraught with lack of standardization, universal availability and abridged physicians' understanding of MRD modalities. We aimed at addressing some of the frequently asked questions in the MRD assessment and will also place in perspective some arguments in favor of MRD assessment in routine practice and clinical trial scenario.
Collapse
Affiliation(s)
- Uday Yanamandra
- Department of Hematology and Stem Cell Transplant, Army Hospital - Research and Referral, New Delhi, Delhi, IN 110010
| | - Shaji K. Kumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA 55905
| |
Collapse
|
49
|
Soekojo CY, de Mel S, Ooi M, Yan B, Chng WJ. Potential Clinical Application of Genomics in Multiple Myeloma. Int J Mol Sci 2018; 19:ijms19061721. [PMID: 29890777 PMCID: PMC6032230 DOI: 10.3390/ijms19061721] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/02/2018] [Accepted: 06/07/2018] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma is a heterogeneous disease with different characteristics, and genetic aberrations play important roles in this heterogeneity. Studies have shown that these genetic aberrations are crucial in prognostication and response assessment; recent efforts have focused on their possible therapeutic implications. Despite many emerging studies being published, the best way to incorporate these results into clinical practice remains unclear. In this review paper we describe the different genomic techniques available, including the latest advancements, and discuss the potential clinical application of genomics in multiple myeloma.
Collapse
Affiliation(s)
- Cinnie Yentia Soekojo
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
| | - Sanjay de Mel
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
| | - Melissa Ooi
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
| | - Benedict Yan
- Department of Laboratory Medicine, National University Hospital, National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore.
| | - Wee Joo Chng
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore,14 Medical Drive, Singapore 117599, Singapore.
| |
Collapse
|
50
|
Soh KT, Tario JD, Wallace PK. Diagnosis of Plasma Cell Dyscrasias and Monitoring of Minimal Residual Disease by Multiparametric Flow Cytometry. Clin Lab Med 2018; 37:821-853. [PMID: 29128071 DOI: 10.1016/j.cll.2017.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Plasma cell dyscrasia (PCD) is a heterogeneous disease that has seen a tremendous change in outcomes due to improved therapies. Over the past few decades, multiparametric flow cytometry has played an important role in the detection and monitoring of PCDs. Flow cytometry is a high-sensitivity assay for early detection of minimal residual disease (MRD) that correlates well with progression-free survival and overall survival. Before flow cytometry can be effectively implemented in the clinical setting, sample preparation, panel configuration, analysis, and gating strategies must be optimized to ensure accurate results. Current consensus methods and reporting guidelines for MRD testing are discussed.
Collapse
Affiliation(s)
- Kah Teong Soh
- Department of Flow and Image Cytometry, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| | - Joseph D Tario
- Department of Flow and Image Cytometry, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Paul K Wallace
- Department of Flow and Image Cytometry, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| |
Collapse
|