1
|
Wijnands C, Armony G, Noori S, Gloerich J, Bonifay V, Caillon H, Luider TM, Brehmer S, Pfennig L, Srikumar T, Trede D, Kruppa G, Dejoie T, van Duijn MM, van Gool AJ, Jacobs JFM, Wessels HJCT. An automated workflow based on data independent acquisition for practical and high-throughput personalized assay development and minimal residual disease monitoring in multiple myeloma patients. Clin Chem Lab Med 2024; 62:2507-2518. [PMID: 38872409 DOI: 10.1515/cclm-2024-0306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVES Minimal residual disease (MRD) status in multiple myeloma (MM) is an important prognostic biomarker. Personalized blood-based targeted mass spectrometry detecting M-proteins (MS-MRD) was shown to provide a sensitive and minimally invasive alternative to MRD-assessment in bone marrow. However, MS-MRD still comprises of manual steps that hamper upscaling of MS-MRD testing. Here, we introduce a proof-of-concept for a novel workflow using data independent acquisition-parallel accumulation and serial fragmentation (dia-PASEF) and automated data processing. METHODS Using automated data processing of dia-PASEF measurements, we developed a workflow that identified unique targets from MM patient sera and personalized protein sequence databases. We generated patient-specific libraries linked to dia-PASEF methods and subsequently quantitated and reported M-protein concentrations in MM patient follow-up samples. Assay performance of parallel reaction monitoring (prm)-PASEF and dia-PASEF workflows were compared and we tested mixing patient intake sera for multiplexed target selection. RESULTS No significant differences were observed in lowest detectable concentration, linearity, and slope coefficient when comparing prm-PASEF and dia-PASEF measurements of serial dilutions of patient sera. To improve assay development times, we tested multiplexing patient intake sera for target selection which resulted in the selection of identical clonotypic peptides for both simplex and multiplex dia-PASEF. Furthermore, assay development times improved up to 25× when measuring multiplexed samples for peptide selection compared to simplex. CONCLUSIONS Dia-PASEF technology combined with automated data processing and multiplexed target selection facilitated the development of a faster MS-MRD workflow which benefits upscaling and is an important step towards the clinical implementation of MS-MRD.
Collapse
Affiliation(s)
- Charissa Wijnands
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gad Armony
- Translational Metabolic Laboratory, Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Somayya Noori
- Department of Neurology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Jolein Gloerich
- Translational Metabolic Laboratory, Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Hélène Caillon
- Biochemistry Laboratory, Hospital of Nantes, Nantes, France
| | - Theo M Luider
- Department of Neurology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | - Thomas Dejoie
- Biochemistry Laboratory, Hospital of Nantes, Nantes, France
| | - Martijn M van Duijn
- Department of Neurology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Alain J van Gool
- Translational Metabolic Laboratory, Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joannes F M Jacobs
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans J C T Wessels
- Translational Metabolic Laboratory, Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
2
|
Langerhorst P, Baerenfaenger M, Kulkarni P, Nadal S, Wijnands C, Post MA, Noori S, vanDuijn MM, Joosten I, Dejoie T, van Gool AJ, Gloerich J, Lefeber DJ, Wessels HJCT, Jacobs JFM. N-linked glycosylation of the M-protein variable region: glycoproteogenomics reveals a new layer of personalized complexity in multiple myeloma. Clin Chem Lab Med 2024; 62:1626-1635. [PMID: 38332688 DOI: 10.1515/cclm-2023-1189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/28/2024] [Indexed: 02/10/2024]
Abstract
OBJECTIVES Multiple myeloma (MM) is a plasma cell malignancy characterized by a monoclonal expansion of plasma cells that secrete a characteristic M-protein. This M-protein is crucial for diagnosis and monitoring of MM in the blood of patients. Recent evidence has emerged suggesting that N-glycosylation of the M-protein variable (Fab) region contributes to M-protein pathogenicity, and that it is a risk factor for disease progression of plasma cell disorders. Current methodologies lack the specificity to provide a site-specific glycoprofile of the Fab regions of M-proteins. Here, we introduce a novel glycoproteogenomics method that allows detailed M-protein glycoprofiling by integrating patient specific Fab region sequences (genomics) with glycoprofiling by glycoproteomics. METHODS Glycoproteogenomics was used for the detailed analysis of de novo N-glycosylation sites of M-proteins. First, Genomic analysis of the M-protein variable region was used to identify de novo N-glycosylation sites. Subsequently glycopeptide analysis with LC-MS/MS was used for detailed analysis of the M-protein glycan sites. RESULTS Genomic analysis uncovered a more than two-fold increase in the Fab Light Chain N-glycosylation of M-proteins of patients with Multiple Myeloma compared to Fab Light Chain N-glycosylation of polyclonal antibodies from healthy individuals. Subsequent glycoproteogenomics analysis of 41 patients enrolled in the IFM 2009 clinical trial revealed that the majority of the Fab N-glycosylation sites were fully occupied with complex type glycans, distinguishable from Fc region glycans due to high levels of sialylation, fucosylation and bisecting structures. CONCLUSIONS Together, glycoproteogenomics is a powerful tool to study de novo Fab N-glycosylation in plasma cell dyscrasias.
Collapse
Affiliation(s)
- Pieter Langerhorst
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Melissa Baerenfaenger
- Department of Neurology, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
- Division of BioAnalytical Chemistry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Purva Kulkarni
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Simon Nadal
- CY Cergy Paris Université, CNRS, BioCIS, Cergy-Pontoise, France
| | - Charissa Wijnands
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Merel A Post
- Department of Neurology, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Somayya Noori
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Martijn M vanDuijn
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Irma Joosten
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas Dejoie
- Biochemistry Laboratory, Centre Hospitalier Universitaire (CHU), Nantes, France
| | - Alain J van Gool
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jolein Gloerich
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dirk J Lefeber
- Department of Neurology, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans J C T Wessels
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joannes F M Jacobs
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
3
|
Aguilan JT, Lim J, Racine-Brzostek S, Fischer J, Silvescu C, Cornett S, Nieves E, Mendu DR, Aliste CM, Semple S, Angeletti R, Weiss LM, Cole A, Prystowsky M, Pullman J, Sidoli S. Effect of dynamic exclusion and the use of FAIMS, DIA and MALDI-mass spectrometry imaging with ion mobility on amyloid protein identification. Clin Proteomics 2024; 21:47. [PMID: 38961380 PMCID: PMC11223398 DOI: 10.1186/s12014-024-09500-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024] Open
Abstract
Amyloidosis is a disease characterized by local and systemic extracellular deposition of amyloid protein fibrils where its excessive accumulation in tissues and resistance to degradation can lead to organ failure. Diagnosis is challenging because of approximately 36 different amyloid protein subtypes. Imaging methods like immunohistochemistry and the use of Congo red staining of amyloid proteins for laser capture microdissection combined with liquid chromatography tandem mass spectrometry (LMD/LC-MS/MS) are two diagnostic methods currently used depending on the expertise of the pathology laboratory. Here, we demonstrate a streamlined in situ amyloid peptide spatial mapping by Matrix Assisted Laser Desorption Ionization-Mass Spectrometry Imaging (MALDI-MSI) combined with Trapped Ion Mobility Spectrometry for potential transthyretin (ATTR) amyloidosis subtyping. While we utilized the standard LMD/LC-MS/MS workflow for amyloid subtyping of 31 specimens from different organs, we also evaluated the potential introduction in the MS workflow variations in data acquisition parameters like dynamic exclusion, or testing Data Dependent Acquisition combined with High-Field Asymmetric Waveform Ion Mobility Spectrometry (DDA FAIMS) versus Data Independent Acquisition (DIA) for enhanced amyloid protein identification at shorter acquisition times. We also demonstrate the use of Mascot's Error Tolerant Search and PEAKS de novo sequencing for the sequence variant analysis of amyloidosis specimens.
Collapse
Affiliation(s)
- Jennifer T Aguilan
- Laboratory for Macromolecular Analysis and Proteomics Facility, Albert Einstein College of Medicine, New York, 10461, USA
- Department of Pathology, Albert Einstein College of Medicine, New York, 10461, USA
- Montefiore Medical Center, Moses and Weiler Campus, New York, 10461, USA
| | - Jihyeon Lim
- Janssen Research and Development, Malvern, PA, USA
| | | | | | | | | | - Edward Nieves
- Laboratory for Macromolecular Analysis and Proteomics Facility, Albert Einstein College of Medicine, New York, 10461, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Damodara Rao Mendu
- Clinical Chemistry Laboratory, Mount Sinai School of Medicine, New York, USA
| | - Carlos-Madrid Aliste
- Laboratory for Macromolecular Analysis and Proteomics Facility, Albert Einstein College of Medicine, New York, 10461, USA
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, New York, 10461, USA
| | | | - Ruth Angeletti
- Laboratory for Macromolecular Analysis and Proteomics Facility, Albert Einstein College of Medicine, New York, 10461, USA
| | - Louis M Weiss
- Department of Pathology, Albert Einstein College of Medicine, New York, 10461, USA
- Montefiore Medical Center, Moses and Weiler Campus, New York, 10461, USA
| | - Adam Cole
- Montefiore Medical Center, Moses and Weiler Campus, New York, 10461, USA
| | - Michael Prystowsky
- Department of Pathology, Albert Einstein College of Medicine, New York, 10461, USA
- Montefiore Medical Center, Moses and Weiler Campus, New York, 10461, USA
| | - James Pullman
- Montefiore Medical Center, Moses and Weiler Campus, New York, 10461, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
4
|
Szalat R, Anderson K, Munshi N. Role of minimal residual disease assessment in multiple myeloma. Haematologica 2024; 109:2049-2059. [PMID: 38328864 PMCID: PMC11215375 DOI: 10.3324/haematol.2023.284662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/31/2024] [Indexed: 02/09/2024] Open
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by clonal proliferation of plasma cells. MM is a heterogeneous disease, featured by various molecular subtypes with different outcomes. With the advent of very efficient therapies including monoclonal antibodies, bispecific T-cell engagers and chimeric antigen receptor T cells (CAR T cells), most MM patients now have a prolonged survival. However, the disease remains incurable, and a subgroup of high-risk patients continue to have early relapse and short survival. Novel and highly sensitive methods have been developed allowing the detection of minimal residual disease (MRD) during or after treatment. Achievement of MRD negativity is a strong and independent prognostic factor in both prospective randomized clinical trials and in the real-world setting. While MRD assessment is now a validated endpoint in clinical trials, its incorporation in clinical practice is not yet established and its potential impact on guiding therapy remains under in-depth evaluation. Here we discuss the different methods available for MRD assessment and the role of MRD evaluation in MM management.
Collapse
Affiliation(s)
- Raphael Szalat
- Section of Hematology and Medical Oncology, Boston University School of Medicine and Boston Medical Center, Boston, MA.
| | - Kenneth Anderson
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Nikhil Munshi
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
5
|
Giles HV, Karunanithi K. Performance Characteristics and Limitations of the Available Assays for the Detection and Quantitation of Monoclonal Free Light Chains and New Emerging Methodologies. Antibodies (Basel) 2024; 13:19. [PMID: 38534209 DOI: 10.3390/antib13010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Light chain measurements form an essential component of the testing strategy for the detection and monitoring of patients with suspected and/or proven plasma cell disorders. Urine-based electrophoretic assays remain at the centre of the international guidelines for response assessment but the supplementary role of serum-free light chain (FLC) assays in response assessment and the detection of disease progression due to their increased sensitivity has been increasingly recognised since their introduction in 2001. Serum FLC assays have also been shown to be prognostic across the spectrum of plasma cell disorders and are now incorporated into risk stratification scores for patients with monoclonal gammopathy of undetermined significance (MGUS), smouldering multiple myeloma, and light chain amyloidosis (AL amyloidosis), as well as being incorporated into the criteria for defining symptomatic multiple myeloma. There are now multiple different commercially available serum FLC assays available with differing performance characteristics, which are discussed in this review, along with the implications of these for patient monitoring. Finally, newer methodologies for the identification and characterisation of monoclonal FLC, including modifications to electrophoretic techniques, mass spectrometry-based assays and Amylite, are also described along with the relevant published data available regarding the performance of each assay.
Collapse
Affiliation(s)
- Hannah V Giles
- Department of Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2SY, UK
- Instute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Kamaraj Karunanithi
- Department of Clinical Haematology, University Hospitals North Midlands NHS Trust, Royal Stoke Hospital, Newcastle Road, Stoke-on-Trent ST4 6QG, UK
- School of Medicine, Keele University, Keele, Newcastle-under-Lyme ST5 5BG, UK
| |
Collapse
|
6
|
Wijnands C, Langerhorst P, Noori S, Keizer-Garritsen J, Wessels HJ, Gloerich J, Bonifay V, Caillon H, Luider TM, van Gool AJ, Dejoie T, VanDuijn MM, Jacobs JF. M-protein diagnostics in multiple myeloma patients using ultra-sensitive targeted mass spectrometry and an off-the-shelf calibrator. Clin Chem Lab Med 2024; 62:540-550. [PMID: 37823394 PMCID: PMC10808047 DOI: 10.1515/cclm-2023-0781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023]
Abstract
OBJECTIVES Minimal residual disease status in multiple myeloma is an important prognostic biomarker. Recently, personalized blood-based targeted mass spectrometry (MS-MRD) was shown to provide a sensitive and minimally invasive alternative to measure minimal residual disease. However, quantification of MS-MRD requires a unique calibrator for each patient. The use of patient-specific stable isotope labelled (SIL) peptides is relatively costly and time-consuming, thus hindering clinical implementation. Here, we introduce a simplification of MS-MRD by using an off-the-shelf calibrator. METHODS SILuMAB-based MS-MRD was performed by spiking a monoclonal stable isotope labeled IgG, SILuMAB-K1, in the patient serum. The abundance of both M-protein-specific peptides and SILuMAB-specific peptides were monitored by mass spectrometry. The relative ratio between M-protein peptides and SILuMAB peptides allowed for M-protein quantification. We assessed linearity, sensitivity and reproducibility of SILuMAB-based MS-MRD in longitudinally collected sera from the IFM-2009 clinical trial. RESULTS A linear dynamic range was achieved of over 5 log scales, allowing for M-protein quantification down to 0.001 g/L. The inter-assay CV of SILuMAB-based MS-MRD was on average 11 %. Excellent concordance between SIL- and SILuMAB-based MS-MRD was shown (R2>0.985). Additionally, signal intensity of spiked SILuMAB can be used for quality control purpose to assess system performance and incomplete SILuMAB digestion can be used as quality control for sample preparation. CONCLUSIONS Compared to SIL peptides, SILuMAB-based MS-MRD improves the reproducibility, turn-around-times and cost-efficacy of MS-MRD without diminishing its sensitivity and specificity. Furthermore, SILuMAB can be used as a MS-MRD quality control tool to monitor sample preparation efficacy and assay performance.
Collapse
Affiliation(s)
- Charissa Wijnands
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pieter Langerhorst
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Somayya Noori
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Hans J.C.T. Wessels
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jolein Gloerich
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Hélène Caillon
- Biochemistry Laboratory, Hospital of Nantes, Nantes, France
| | - Theo M. Luider
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Alain J. van Gool
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas Dejoie
- Biochemistry Laboratory, Hospital of Nantes, Nantes, France
| | - Martijn M. VanDuijn
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Joannes F.M. Jacobs
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
7
|
Muccio S, Hirtz C, Descloux S, Fedeli O, Macé S, Lehmann S, Vialaret J. A sensitive high-resolution mass spectrometry method for quantifying intact M-protein light chains in patients with multiple myeloma. Clin Chim Acta 2024; 552:117634. [PMID: 37980975 DOI: 10.1016/j.cca.2023.117634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023]
Abstract
To determine the disease status and the response to treatment for patients with multiple myeloma, measuring serum M-protein levels is a widely used alternative to invasive punctures to count malignant plasma cells in the bone marrow. However, the quantification of this monoclonal antibody, which varies from patient to patient, poses significant analytical challenges. This paper describes a sensitive and specific mass spectrometry assay that addresses two objectives: to overcome the potential interference of biotherapeutics in the measurement of M-proteins, and to determine the depth of response to treatment by assessing minimal residual disease. After immunocapture of immunoglobulins and free light chains in serum, heavy and light chains were dissociated by chemical reduction and separated by liquid chromatography. M-proteins were analyzed by high-resolution mass spectrometry using a method combining a full MS scan for isotyping and identification and a targeted single ion monitoring scan for quantification. This method was able to discriminate M-protein from the therapeutic antibody in all patient samples analyzed and allowed quantification of M-protein with a LLOQ of 2.0 to 3.5 µg/ml in 5 out of 6 patients. This methodology appears to be promising for assessing minimal residual disease with sufficient sensitivity, specificity, and throughput.
Collapse
Affiliation(s)
- Stéphane Muccio
- Sanofi, TMED-BCB, 371 rue du Professeur Blayac, 34184 Montpellier, France.
| | - Christophe Hirtz
- Montpellier Univ, IRMB CHU, INM INSERM, 80 avenue Augustin Fliche, 34295 Montpellier, France
| | - Sandrine Descloux
- Sanofi, TMED-BCB, 371 rue du Professeur Blayac, 34184 Montpellier, France
| | - Olivier Fedeli
- Sanofi, TMED-BCB, 371 rue du Professeur Blayac, 34184 Montpellier, France
| | - Sandrine Macé
- Sanofi, TMED-PMO, 1 avenue Pierre Brossolette, 91385 Chilly-Mazarin, France
| | - Sylvain Lehmann
- Montpellier Univ, IRMB CHU, INM INSERM, 80 avenue Augustin Fliche, 34295 Montpellier, France
| | - Jérôme Vialaret
- Montpellier Univ, IRMB CHU, INM INSERM, 80 avenue Augustin Fliche, 34295 Montpellier, France
| |
Collapse
|
8
|
Wijnands C, Noori S, Donk NWCJVD, VanDuijn MM, Jacobs JFM. Advances in minimal residual disease monitoring in multiple myeloma. Crit Rev Clin Lab Sci 2023; 60:518-534. [PMID: 37232394 DOI: 10.1080/10408363.2023.2209652] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/07/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023]
Abstract
Multiple myeloma (MM) is characterized by the clonal expansion of plasma cells and the excretion of a monoclonal immunoglobulin (M-protein), or fragments thereof. This biomarker plays a key role in the diagnosis and monitoring of MM. Although there is currently no cure for MM, novel treatment modalities such as bispecific antibodies and CAR T-cell therapies have led to substantial improvement in survival. With the introduction of several classes of effective drugs, an increasing percentage of patients achieve a complete response. This poses new challenges to traditional electrophoretic and immunochemical M-protein diagnostics because these methods lack sensitivity to monitor minimal residual disease (MRD). In 2016, the International Myeloma Working Group (IMWG) expanded their disease response criteria with bone marrow-based MRD assessment using flow cytometry or next-generation sequencing in combination with imaging-based disease monitoring of extramedullary disease. MRD status is an important independent prognostic marker and its potential as a surrogate endpoint for progression-free survival is currently being studied. In addition, numerous clinical trials are investigating the added clinical value of MRD-guided therapy decisions in individual patients. Because of these novel clinical applications, repeated MRD evaluation is becoming common practice in clinical trials as well as in the management of patients outside clinical trials. In response to this, novel mass spectrometric methods that have been developed for blood-based MRD monitoring represent attractive minimally invasive alternatives to bone marrow-based MRD evaluation. This paves the way for dynamic MRD monitoring to allow the detection of early disease relapse, which may prove to be a crucial factor in facilitating future clinical implementation of MRD-guided therapy. This review provides an overview of state-of-the-art of MRD monitoring, describes new developments and applications of blood-based MRD monitoring, and suggests future directions for its successful integration into the clinical management of MM patients.
Collapse
Affiliation(s)
- Charissa Wijnands
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Somayya Noori
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | | | - Martijn M VanDuijn
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Joannes F M Jacobs
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
9
|
Jia X, Liu X, Yang W. Predictive Value Analysis of Serum Ig A, Ig G, and TNF- α in Recurrence of Multiple Myeloma. DISEASE MARKERS 2022; 2022:2095696. [PMID: 36277989 PMCID: PMC9581636 DOI: 10.1155/2022/2095696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/23/2022] [Accepted: 09/30/2022] [Indexed: 09/07/2024]
Abstract
Objective The study is aimed at analyzing the predictive value of serum Ig A, Ig G, and TNF-α in the recurrence of multiple myeloma (MM). Methods 136 patients with MM treated in our hospital from January 2010 to January 2017 were followed up for 5 years. Finally, 100 patients who met the inclusion and exclusion criteria and had the complete follow-up visit were selected as the study subjects, with the recurrence of MM as endpoint event, and the observation was taken until the occurrence of endpoint event in patients or the termination of this study. They were divided into the recurrence group (RG) and the nonrecurrence group (NRG) according to whether the endpoint event occurred. The venous blood of patients was collected at the first diagnosis and subsequent visit (at the time of recurrence or termination of the study) to measure the Ig A and Ig G using a full automatic special protein analyzer and the TNF-α level by enzyme-linked immunosorbent assay. The data obtained in this study were analyzed by univariate analysis to choose the factors with difference in statistical significance to draw the ROC curve, and the areas under the curve (AUC) were recorded to analyze the potential mechanism of Ig A, Ig G, and TNF-α in predicting the recurrence of MM. Results After follow-up visit, there were 62 patients with recurrence (62.0%) and 38 patients without recurrence (38.0%), with no obvious difference in gender, age, body weight, and immune classification between the two groups (P > 0.05). Compared with the NRG, the levels of soluble interleukin-2 receptor (sIL-2R) and β 2-microglobulin (β 2-MG) in the RG at the first diagnosis were distinctly higher (P < 0.001); the levels of Ig A, Ig G, and TNF-α in the RG at the first diagnosis were visibly higher (P < 0.05); and the levels of Ig A, Ig G, and TNF-α in the RG at the subsequent visit were clearly higher (P < 0.05). There was a correlation between Ig G, Ig A, and TNF-α and β 2-MG at the first diagnosis and the subsequent visit (P < 0.05); there was a correlation between Ig G and TNF-α, and sIL-2R at the first diagnosis and the subsequent visit (P < 0.05); and there was a correlation between Ig A and sIL-2R at the subsequent visit (P < 0.05). The AUC of Ig G, Ig A, and TNF-α in predicting the MM at the first diagnosis were 0.772, 0.776, and 0.778, respectively. Conclusion The serum Ig A, Ig G, and TNF-α had a predictive value in the recurrence of MM, and TNF-α was correlated with sIL-2R and β 2-MG, with the highest AUC and the best predictive value.
Collapse
Affiliation(s)
- Xinyan Jia
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiangxin Liu
- Department of Hematology, Ji'an Hospital of Shanghai East Hospital, Ji'an, 343000 Jiangxi, China
| | - Wenzhong Yang
- Department of Hematology, Shanghai Punan Hospital of Pudong New District, Shanghai 200125, China
| |
Collapse
|
10
|
Retrospective Longitudinal Monitoring of Multiple Myeloma Patients by Mass Spectrometry Using Archived Serum Protein Electrophoresis Gels and De Novo Sequence Analysis. Hemasphere 2022; 6:e758. [PMID: 35935609 PMCID: PMC9348860 DOI: 10.1097/hs9.0000000000000758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/03/2022] [Indexed: 11/26/2022] Open
|
11
|
Giles HV, Wechalekar A, Pratt G. The potential role of mass spectrometry for the identification and monitoring of patients with plasma cell disorders: Where are we now and which questions remain unanswered? Br J Haematol 2022; 198:641-653. [PMID: 35514140 DOI: 10.1111/bjh.18226] [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: 01/24/2022] [Revised: 04/08/2022] [Accepted: 04/20/2022] [Indexed: 11/28/2022]
Abstract
Mass spectrometry (MS) techniques provide a highly sensitive methodology for the assessment and monitoring of paraproteins compared to standard electrophoretic techniques. The International Myeloma Working Group (IMWG) recently approved the use of intact light chain matrix-assisted laser desorption/ionisation time-of-flight mass spectrometry (MALDI-TOF MS) in lieu of immunofixation in the clinical assessment of patients and the assessment of patients enrolled on clinical trials. The increased sensitivity of these assays may help to detect and monitor monoclonal proteins (MP) in many patients with previously non-measurable disease, will reduce complete response (CR) rates and increase detection of low-level MP. The ability to track the unique mass or amino acid sequence of the MP also eliminates interference from therapeutic monoclonal antibodies (tmAbs) in most patients with IgG kappa myeloma. The intact light chain assays also provide structural information about the monoclonal light chain, including the presence of N-linked glycosylation, which has been shown to be commoner on amyloidogenic light chains and may have prognostic significance in monoclonal gammopathy of undetermined significance (MGUS). In this review, we discuss these issues alongside differences in the analytical and practical aspects related to the different MS assays under development and the challenges for MS.
Collapse
Affiliation(s)
- Hannah V Giles
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,University of Birmingham, Birmingham, UK
| | - Ashutosh Wechalekar
- Royal Free London NHS Foundation Trust, London, UK.,University College London, London, UK
| | - Guy Pratt
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,University of Birmingham, Birmingham, UK
| |
Collapse
|
12
|
Dasari S, Kohlhagen MC, Dispenzieri A, Willrich MAV, Snyder MR, Kourelis TV, Lust JA, Mills JR, Kyle RA, Murray DL. Detection of Plasma Cell Disorders by Mass Spectrometry: A Comprehensive Review of 19,523 Cases. Mayo Clin Proc 2022; 97:294-307. [PMID: 34887112 DOI: 10.1016/j.mayocp.2021.07.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 01/14/2023]
Abstract
OBJECTIVES To verify the analytical performance of a new mass spectrometry-based method, termed MASS-FIX, when screening for plasma cell disorders in a routine clinical laboratory. PATIENTS AND METHODS Results from 19,523 unique patients tested for an M-protein between July 24, 2018, and March 6, 2020, by a combination serum protein electrophoresis (SPEP) and MASS-FIX were examined for consistency with pretest implementation performance. MASS-FIX's ability to verify abnormal results from SPEP and free light chain measurements was then compared with that of immunofixation electrophoresis (IFE) using a separate cohort of 52,586 patients tested by SPEP/IFE during the same period. RESULTS Overall, 62.4% of our cohort was negative for an M-protein. Importantly, 7.3% of all specimens had an M spike on SPEP (0.1 to 8.5 g/dL) and MASS-FIX detected an M-protein in all these samples. Of all samples, 30.3% had M-proteins that were detected by MASS-FIX but the SPEP finding was too small for quantification. Of the positive samples, 5.7% contained a therapeutic monoclonal antibody. Of the positive samples, 4.1% had an N-glycosylated light chain (biomarker of high-risk plasma cell disorders). MASS-FIX confirmed a higher percentage of SPEP abnormalities than IFE. MASS-FIX was slightly more sensitive than IFE when confirming an M-protein in samples with an abnormal free light chain ratio. MASS-FIX had a very low sample repeat rate (1.5%). MASS-FIX was highly automatable resulting in a higher number of samples/technologist/day than IFE (∼30% more). CONCLUSION Overall, MASS-FIX was successful in maintaining validation characteristics. MASS-FIX was more sensitive in confirming SPEP abnormalities when compared with IFE. Ability to detect therapeutic monoclonal antibodies and glycosylated light chains was distinctly advantageous.
Collapse
Affiliation(s)
- Surendra Dasari
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Mindy C Kohlhagen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Angela Dispenzieri
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Department of Medicine, Mayo Clinic, Rochester, MN
| | - Maria A V Willrich
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Melissa R Snyder
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - John A Lust
- Department of Medicine, Mayo Clinic, Rochester, MN
| | - John R Mills
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - David L Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN.
| |
Collapse
|
13
|
Redefining Non-measurable Multiple Myeloma Using Mass Spectrometry. Blood 2021; 139:946-950. [PMID: 34871382 DOI: 10.1182/blood.2021013794] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/15/2021] [Indexed: 11/20/2022] Open
|
14
|
Du J, Zhuang J. Major advances in the treatment of multiple myeloma in American Society of Hematology annual meeting 2020. Chronic Dis Transl Med 2021; 7:220-226. [PMID: 34786541 PMCID: PMC8579022 DOI: 10.1016/j.cdtm.2021.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Indexed: 01/01/2023] Open
Abstract
Treatment of multiple myeloma (MM) has advanced dramatically in the past two decades. However, under the conditions of the COVID-19 pandemic, treatment strategies have been modified accordingly. Numerous novel agents, updated trials, and major advances in myeloma have been reported in the American Society of Hematology 2020 annual meeting, either for transplant-eligible or ineligible patients. Hot topics such as the significance of autologous stem cell transplantation (ASCT), development of novel agents, and chimeric antigen receptor-T (CAR-T) cells have been widely discussed. The triplet regimen bortezomib, lenalidomide, and dexamethasone (VRd) is recommended as the standard first-line treatment, and the addition of a fourth drug improves efficacy and survival. The value of ASCT remains undoubtful, even in the era of quadruplet induction. Dual-drug maintenance, including proteasome inhibitors and immunomodulatory drugs, overcomes unfavorable outcomes in high-risk patients. For relapsed/refractory myeloma (RRMM) patients, novel agents such as selinexor and venetoclax are superior. CAR-T cells and other cell-surface-targeted therapies also appear promising.
Collapse
Affiliation(s)
- Jianhua Du
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Junling Zhuang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
15
|
Langerhorst P, Noori S, Zajec M, De Rijke YB, Gloerich J, van Gool AJ, Caillon H, Joosten I, Luider TM, Corre J, VanDuijn MM, Dejoie T, Jacobs JFM. Multiple Myeloma Minimal Residual Disease Detection: Targeted Mass Spectrometry in Blood vs Next-Generation Sequencing in Bone Marrow. Clin Chem 2021; 67:1689-1698. [PMID: 34643690 DOI: 10.1093/clinchem/hvab187] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/18/2021] [Indexed: 01/11/2023]
Abstract
BACKGROUND Minimal residual disease (MRD) status assessed on bone marrow aspirates is a major prognostic biomarker in multiple myeloma (MM). In this study we evaluated blood-based targeted mass spectrometry (MS-MRD) as a sensitive, minimally invasive alternative to measure MM disease activity. METHODS Therapy response of 41 MM patients in the IFM-2009 clinical trial (NCT01191060) was assessed with MS-MRD on frozen sera and compared to routine state-of-the-art monoclonal protein (M-protein) diagnostics and next-generation sequencing (NGS-MRD) at 2 time points. RESULTS In all 41 patients we were able to identify clonotypic M-protein-specific peptides and perform serum-based MS-MRD measurements. MS-MRD is significantly more sensitive to detect M-protein compared to either electrophoretic M-protein diagnostics or serum free light chain analysis. The concordance between NGS-MRD and MS-MRD status in 81 paired bone marrow/sera samples was 79%. The 50% progression-free survival (PFS) was identical (49 months) for patients who were either NGS-positive or MS-positive directly after maintenance treatment. The 50% PFS was 69 and 89 months for NGS-negative and MS-negative patients, respectively. The longest 50% PFS (96 months) was observed in patients who were MRD-negative for both methods. MS-MRD relapse during maintenance treatment was significantly correlated to poor PFS (P < 0.0001). CONCLUSIONS Our data indicate proof-of-principle that MS-MRD evaluation in blood is a feasible, patient friendly alternative to NGS-MRD assessed on bone marrow. Clinical validation of the prognostic value of MS-MRD and its complementary value in MRD-evaluation of patients with MM is warranted in an independent larger cohort.
Collapse
Affiliation(s)
- Pieter Langerhorst
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Somayya Noori
- Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marina Zajec
- Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Yolanda B De Rijke
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jolein Gloerich
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Alain J van Gool
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hélène Caillon
- Laboratoire de Biochimie, Centre Hospitalier Universitaire (CHU), Nantes, France
| | - Irma Joosten
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Theo M Luider
- Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jill Corre
- Unité de Génomique du Myélome, Institute Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Martijn M VanDuijn
- Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Thomas Dejoie
- Laboratoire de Biochimie, Centre Hospitalier Universitaire (CHU), Nantes, France
| | - Joannes F M Jacobs
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
16
|
Anderson KC, Auclair D, Adam SJ, Agarwal A, Anderson M, Avet-Loiseau H, Bustoros M, Chapman J, Connors DE, Dash A, Di Bacco A, Du L, Facon T, Flores-Montero J, Gay F, Ghobrial IM, Gormley NJ, Gupta I, Higley H, Hillengass J, Kanapuru B, Kazandjian D, Kelloff GJ, Kirsch IR, Kremer B, Landgren O, Lightbody E, Lomas OC, Lonial S, Mateos MV, Montes de Oca R, Mukundan L, Munshi NC, O'Donnell EK, Orfao A, Paiva B, Patel R, Pugh TJ, Ramasamy K, Ray J, Roshal M, Ross JA, Sigman CC, Thoren KL, Trudel S, Ulaner G, Valente N, Weiss BM, Zamagni E, Kumar SK. Minimal Residual Disease in Myeloma: Application for Clinical Care and New Drug Registration. Clin Cancer Res 2021; 27:5195-5212. [PMID: 34321279 PMCID: PMC9662886 DOI: 10.1158/1078-0432.ccr-21-1059] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/01/2021] [Accepted: 07/23/2021] [Indexed: 01/07/2023]
Abstract
The development of novel agents has transformed the treatment paradigm for multiple myeloma, with minimal residual disease (MRD) negativity now achievable across the entire disease spectrum. Bone marrow-based technologies to assess MRD, including approaches using next-generation flow and next-generation sequencing, have provided real-time clinical tools for the sensitive detection and monitoring of MRD in patients with multiple myeloma. Complementary liquid biopsy-based assays are now quickly progressing with some, such as mass spectrometry methods, being very close to clinical use, while others utilizing nucleic acid-based technologies are still developing and will prove important to further our understanding of the biology of MRD. On the regulatory front, multiple retrospective individual patient and clinical trial level meta-analyses have already shown and will continue to assess the potential of MRD as a surrogate for patient outcome. Given all this progress, it is not surprising that a number of clinicians are now considering using MRD to inform real-world clinical care of patients across the spectrum from smoldering myeloma to relapsed refractory multiple myeloma, with each disease setting presenting key challenges and questions that will need to be addressed through clinical trials. The pace of advances in targeted and immune therapies in multiple myeloma is unprecedented, and novel MRD-driven biomarker strategies are essential to accelerate innovative clinical trials leading to regulatory approval of novel treatments and continued improvement in patient outcomes.
Collapse
Affiliation(s)
- Kenneth C. Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Daniel Auclair
- Multiple Myeloma Research Foundation, Norwalk, Connecticut.,Corresponding Author: Daniel Auclair, Research, Multiple Myeloma Research Foundation, 383 Main Street, Norwalk, CT, 06851. E-mail:
| | - Stacey J. Adam
- Foundation for the National Institutes of Health, North Bethesda, Maryland
| | - Amit Agarwal
- US Medical Oncology, Bristol-Myers Squibb, Summit, New Jersey
| | | | - Hervé Avet-Loiseau
- Laboratoire d'Hématologie, Pôle Biologie, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Mark Bustoros
- Division of Hematology and Medical Oncology, Cornell University/New York Presbyterian Hospital, New York, New York
| | | | - Dana E. Connors
- Foundation for the National Institutes of Health, North Bethesda, Maryland
| | - Ajeeta Dash
- Takeda Pharmaceuticals, Cambridge, Massachusetts
| | | | - Ling Du
- GlaxoSmithKline, Collegeville, Pennsylvania
| | - Thierry Facon
- Department of Hematology, Lille University Hospital, Lille, France
| | - Juan Flores-Montero
- Cancer Research Center (IBMCC-CSIC/USAL-IBSAL); Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Francesca Gay
- Myeloma Unit, Division of Hematology, Azienda Ospedaliero Università Città della Salute e della Scienza, Torino, Italy
| | - Irene M. Ghobrial
- Preventative Cancer Therapies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Nicole J. Gormley
- Division of Hematologic Malignancies 2, Office of Oncologic Disease, Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland
| | - Ira Gupta
- GlaxoSmithKline, Collegeville, Pennsylvania
| | | | - Jens Hillengass
- Division of Hematology and Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - Bindu Kanapuru
- Division of Hematologic Malignancies 2, Office of Oncologic Disease, Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland
| | - Dickran Kazandjian
- Myeloma Program, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Gary J. Kelloff
- Division of Cancer Treatment and Diagnosis, NCI, NIH, Rockville, Maryland
| | - Ilan R. Kirsch
- Translational Medicine, Adaptive Biotechnologies, Seattle, Washington
| | | | - Ola Landgren
- Myeloma Program, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Elizabeth Lightbody
- Preventative Cancer Therapies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Oliver C. Lomas
- Preventative Cancer Therapies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Sagar Lonial
- Department of Hematology and Medical Oncology at Emory University School of Medicine, Atlanta, Georgia
| | | | | | | | - Nikhil C. Munshi
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - Alberto Orfao
- Cancer Research Center (IBMCC-CSIC/USAL-IBSAL); Cytometry Service (NUCLEUS) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Bruno Paiva
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada (CIMA), Instituto de Investigacion Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - Reshma Patel
- Janssen Research & Development, Spring House, Pennsylvania
| | - Trevor J. Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Karthik Ramasamy
- Cancer and Haematology Centre, Oxford University Hospitals, Oxford, United Kingdom
| | - Jill Ray
- BioOncology, Genentech Inc., South San Francisco, California
| | - Mikhail Roshal
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jeremy A. Ross
- Precision Medicine, Oncology, AbbVie, Inc., North Chicago, Illinois
| | | | | | - Suzanne Trudel
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | - Nancy Valente
- BioOncology, Genentech Inc., South San Francisco, California
| | | | - Elena Zamagni
- Seragnoli Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Shaji K. Kumar
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
17
|
Liyasova M, McDonald Z, Taylor P, Gorospe K, Xu X, Yao C, Liu Q, Yang L, Atenafu EG, Piza G, Ma B, Reece D, Trudel S. A Personalized Mass Spectrometry-Based Assay to Monitor M-Protein in Patients with Multiple Myeloma (EasyM). Clin Cancer Res 2021; 27:5028-5037. [PMID: 34210683 PMCID: PMC9401514 DOI: 10.1158/1078-0432.ccr-21-0649] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/27/2021] [Accepted: 06/24/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE M-protein is a well-established biomarker used for multiple myeloma monitoring. Current improvements in multiple myeloma treatment created the need to monitor minimal residual disease (MRD) with high sensitivity. Measuring residual levels of M-protein in serum by MS was established as a sensitive assay for disease monitoring. In this study we evaluated the performance of EasyM-a noninvasive, sensitive, MS-based assay for M-protein monitoring. EXPERIMENTAL DESIGN Twenty-six patients enrolled in MCRN-001 clinical trial of two high-dose alkylating agents as conditioning followed by lenalidomide maintenance were selected for the study. All selected patients achieved complete responses (CR) during treatment, whereas five experienced progressive disease on study. The M-protein of each patient was first sequenced from the diagnostic serum using our de novo protein sequencing platform. The patient-specific M-protein peptides were then measured by targeted MS assay to monitor the response to treatment. RESULTS The M-protein doubling over 6 months measured by EasyM could predict the relapse in 4 of 5 relapsed patients 2 to 11 months earlier than conventional testing. In 21 disease-free patients, the M-protein was still detectable by EasyM despite normal FLC and MRD negativity. Importantly, of 72 MRD negative samples with CR status, 62 were positive by EasyM. The best sensitivity achieved by EasyM, detecting 0.58 mg/L of M-protein, was 1,000- and 200-fold higher compared with serum protein electrophoresis and immunofixation electrophoresis, respectively. CONCLUSIONS EasyM was demonstrated to be a noninvasive, sensitive assay with superior performance compared with other assays, making it ideal for multiple myeloma monitoring and relapse prediction.
Collapse
Affiliation(s)
| | | | - Paul Taylor
- Rapid Novor, Inc., Kitchener, Ontario, Canada
| | | | - Xin Xu
- Rapid Novor, Inc., Kitchener, Ontario, Canada
| | - Chenyu Yao
- Rapid Novor, Inc., Kitchener, Ontario, Canada
| | - Qixin Liu
- Rapid Novor, Inc., Kitchener, Ontario, Canada
| | | | | | - Giovanni Piza
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Bin Ma
- University of Waterloo, Waterloo, Ontario, Canada
| | - Donna Reece
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Suzanne Trudel
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Corresponding Author: Suzanne Trudel, Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON, Canada M5G 2M9. E-mail:
| |
Collapse
|
18
|
Dunphy K, O’Mahoney K, Dowling P, O’Gorman P, Bazou D. Clinical Proteomics of Biofluids in Haematological Malignancies. Int J Mol Sci 2021; 22:ijms22158021. [PMID: 34360786 PMCID: PMC8348619 DOI: 10.3390/ijms22158021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022] Open
Abstract
Since the emergence of high-throughput proteomic techniques and advances in clinical technologies, there has been a steady rise in the number of cancer-associated diagnostic, prognostic, and predictive biomarkers being identified and translated into clinical use. The characterisation of biofluids has become a core objective for many proteomic researchers in order to detect disease-associated protein biomarkers in a minimally invasive manner. The proteomes of biofluids, including serum, saliva, cerebrospinal fluid, and urine, are highly dynamic with protein abundance fluctuating depending on the physiological and/or pathophysiological context. Improvements in mass-spectrometric technologies have facilitated the in-depth characterisation of biofluid proteomes which are now considered hosts of a wide array of clinically relevant biomarkers. Promising efforts are being made in the field of biomarker diagnostics for haematologic malignancies. Several serum and urine-based biomarkers such as free light chains, β-microglobulin, and lactate dehydrogenase are quantified as part of the clinical assessment of haematological malignancies. However, novel, minimally invasive proteomic markers are required to aid diagnosis and prognosis and to monitor therapeutic response and minimal residual disease. This review focuses on biofluids as a promising source of proteomic biomarkers in haematologic malignancies and a key component of future diagnostic, prognostic, and disease-monitoring applications.
Collapse
Affiliation(s)
- Katie Dunphy
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Kelly O’Mahoney
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland; (K.O.); (P.O.)
| | - Paul Dowling
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Peter O’Gorman
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland; (K.O.); (P.O.)
| | - Despina Bazou
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland; (K.O.); (P.O.)
- Correspondence:
| |
Collapse
|
19
|
Abstract
The diagnosis of myeloma and other plasma cell disorders has traditionally been done with the aid of electrophoretic methods, whereas amyloidosis has been characterized by immunohistochemistry. Mass spectrometry has recently been established as an alternative to these traditional methods and has been proved to bring added benefit for patient care. These newer mass spectrometry-based methods highlight some of the key advantages of modern proteomic methods and how they can be applied to the routine care of patients.
Collapse
Affiliation(s)
- David L Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| | - Surendra Dasari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
20
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021. [DOI: 10.37349/etat.2020.00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3University of Montpellier, UFR Medicine, 34093 Montpellier, France 4 Institut Universitaire de France (IUF), 75000 Paris France
| |
Collapse
|
21
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:65-106. [PMID: 36046090 PMCID: PMC9400753 DOI: 10.37349/etat.2021.00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 01/06/2021] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3UFR Medicine, University of Montpellier, 34093 Montpellier, France 4Institut Universitaire de France (IUF), 75000 Paris, France
| |
Collapse
|
22
|
Murray DL, Puig N, Kristinsson S, Usmani SZ, Dispenzieri A, Bianchi G, Kumar S, Chng WJ, Hajek R, Paiva B, Waage A, Rajkumar SV, Durie B. Mass spectrometry for the evaluation of monoclonal proteins in multiple myeloma and related disorders: an International Myeloma Working Group Mass Spectrometry Committee Report. Blood Cancer J 2021; 11:24. [PMID: 33563895 PMCID: PMC7873248 DOI: 10.1038/s41408-021-00408-4] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/10/2020] [Accepted: 12/01/2020] [Indexed: 01/30/2023] Open
Abstract
Plasma cell disorders (PCDs) are identified in the clinical lab by detecting the monoclonal immunoglobulin (M-protein) which they produce. Traditionally, serum protein electrophoresis methods have been utilized to detect and isotype M-proteins. Increasing demands to detect low-level disease and new therapeutic monoclonal immunoglobulin treatments have stretched the electrophoretic methods to their analytical limits. Newer techniques based on mass spectrometry (MS) are emerging which have improved clinical and analytical performance. MS is gaining traction into clinical laboratories, and has replaced immunofixation electrophoresis (IFE) in routine practice at one institution. The International Myeloma Working Group (IMWG) Mass Spectrometry Committee reviewed the literature in order to summarize current data and to make recommendations regarding the role of mass spectrometric methods in diagnosing and monitoring patients with myeloma and related disorders. Current literature demonstrates that immune-enrichment of immunoglobulins coupled to intact light chain MALDI-TOF MS has clinical characteristics equivalent in performance to IFE with added benefits of detecting additional risk factors for PCDs, differentiating M-protein from therapeutic antibodies, and is a suitable replacement for IFE for diagnosing and monitoring multiple myeloma and related PCDs. In this paper we discuss the IMWG recommendations for the use of MS in PCDs.
Collapse
Affiliation(s)
- David L Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| | - Noemi Puig
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | | | - Saad Z Usmani
- Department of Hematologic Oncology & Blood Disorders, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA
| | - Angela Dispenzieri
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Department of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Giada Bianchi
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shaji Kumar
- Department of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Wee Joo Chng
- Cancer Science Institute of Singapore, NUS, Singapore, Singapore
- Yong Loo Lin School of Medicine, NUS, Singapore, Singapore
- National University Cancer Institute, Singapore, Singapore
| | - Roman Hajek
- Department of Hematooncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Bruno Paiva
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada (CIMA), IDISNA, Pamplona, Spain
| | - Anders Waage
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Hematology, St. Olav's University Hospital, Trondheim, Norway
| | | | - Brian Durie
- Department of Hematology, Cedars-Sinai Outpatient Cancer Center, Los Angeles, CA, USA
| |
Collapse
|
23
|
Zuo X, Liu D. Progress in the application of minimal residual disease detection in multiple myeloma. J Hematop 2021. [DOI: 10.1007/s12308-020-00436-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|