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
|
Lee HW, Kim SM, Park HD. Evaluating the HYDRASHIFT 2/4 Daratumumab assay: a powerful approach to assess treatment response in multiple myeloma. Clin Chem Lab Med 2024; 62:2223-2232. [PMID: 38630027 DOI: 10.1515/cclm-2024-0416] [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: 03/03/2024] [Accepted: 04/08/2024] [Indexed: 09/28/2024]
Abstract
OBJECTIVES This study evaluates the HYDRASHIFT assay's effectiveness in mitigating daratumumab interference on serum protein tests during multiple myeloma (MM) treatment, aiming to ensure an accurate assessment of treatment response. METHODS We analyzed 113 serum samples from 68 MM patients undergoing daratumumab treatment, employing both standard IF and the HYDRASHIFT assay. The assay's precision was determined through intra-day and inter-day variability assessments, while its specificity was verified using serum samples devoid of daratumumab. Comparative analysis of IF results, before and after the application of the HYDRASHIFT assay, facilitated the categorization of treatment responses in alignment with the International Myeloma Working Group's response criteria. RESULTS The precision underscored the assay's consistent repeatability and reproducibility, successfully eliminating interference of daratumumab-induced Gκ bands. Specificity assessments demonstrated the assay's capability to distinguish daratumumab from both isatuximab and naturally occurring M-proteins. Of the analyzed cases, 91 exhibited successful migration of daratumumab-induced Gκ bands, thereby enhancing the accuracy of treatment response classification. The remaining 22 cases did not show a visible migration complex, likely due to the low concentration of daratumumab in the serum. These findings underscore the assay's critical role in distinguishing daratumumab from endogenous M-protein, particularly in samples with a single Gκ band on standard IF, where daratumumab and endogenous M-protein had co-migrated. CONCLUSIONS The HYDRASHIFT assay demonstrates high precision, specificity, and utility in the accurate monitoring of treatment responses in MM patients receiving daratumumab. This assay represents a significant advancement in overcoming the diagnostic challenges posed by daratumumab interference.
Collapse
Affiliation(s)
- Hyun-Woo Lee
- Department of Laboratory Medicine and Genetics, 36626 Samsung Medical Center , Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sang-Mi Kim
- Department of Laboratory Medicine and Genetics, 36626 Samsung Medical Center , Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyung-Doo Park
- Department of Laboratory Medicine and Genetics, 36626 Samsung Medical Center , Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Yee AJ. An ATLAS to map MRD with peripheral blood. Blood 2024; 144:919-920. [PMID: 39207809 DOI: 10.1182/blood.2024025189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Affiliation(s)
- Andrew J Yee
- Massachusetts General Hospital Cancer Center and Harvard Medical School
| |
Collapse
|
4
|
Canil G, Miolo G, Simula M, Rupolo M, Steffan A, Corona G. Quantitative assessment of daratumumab in serum via intact light chain measurement using liquid chromatography-high resolution mass spectrometry: a method suitable for therapeutic drug monitoring. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:4240-4246. [PMID: 38780038 DOI: 10.1039/d4ay00404c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Daratumumab, a pivotal treatment for multiple myeloma, exhibits considerable inter-patient variability in pharmacological clinical outcomes, likely attributed to serum concentration that may underscore the need for its therapeutic drug monitoring. This study aims to develop and validate a straightforward analytical method for quantifying daratumumab in serum, focusing on intact light chain determination, using liquid chromatography high-resolution mass spectrometry. The sample preparation involved immunoglobulin enrichment using Melon gel followed by a reduction step to dissociate the light from the heavy chains of immunoglobulins. The latter were then separated using a MabPac RP 2.1 × 50 mm chromatographic column and the intact light chains were detected and quantified using a Q Exactive Orbitrap mass spectrometer operating in ESI-positive ion mode at 17 500 resolution. The method demonstrated excellent linearity (R2 > 0.992) across a serum concentration range of 100 to 2000 μg mL-1 and good precision and accuracy: intra- and interday relative errors ranged from -5.1% to 6.5%, with a relative standard deviation of less than 5.8%. Clinical suitability was confirmed by analyzing 80 clinical samples from multiple myeloma patients treated with 1800 mg of daratumumab. 99% of the samples fell within the analytical range with a mean daratumumab concentration evaluated before the next administration (Ctrough) of 398 μg mL-1. These findings highlighted that intact light chain monoclonal antibody quantification could be a valid and robust alternative to either immunoassays or to LC-MS/MS targeting peptides for measuring daratumumab in clinical samples, positioning it as a suitable method for therapeutic drug monitoring applications.
Collapse
Affiliation(s)
- Giovanni Canil
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico (CRO), IRCCS Aviano, 33081 Aviano, Italy.
| | - Gianmaria Miolo
- Medical Oncology and Cancer Prevention Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Mariapaola Simula
- Clinical Pathology Unit, ASFO General Hospital, 33170 Pordenone, Italy
| | - Maurizio Rupolo
- Oncohematology and Cell Therapy Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico (CRO), IRCCS Aviano, 33081 Aviano, Italy.
| | - Giuseppe Corona
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico (CRO), IRCCS Aviano, 33081 Aviano, Italy.
| |
Collapse
|
5
|
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
|
6
|
Shuai W, Wu X, Chen C, Zuo E, Chen X, Li Z, Lv X, Wu L, Chen C. Rapid diagnosis of rheumatoid arthritis and ankylosing spondylitis based on Fourier transform infrared spectroscopy and deep learning. Photodiagnosis Photodyn Ther 2024; 45:103885. [PMID: 37931694 DOI: 10.1016/j.pdpdt.2023.103885] [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: 09/13/2023] [Revised: 09/26/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
OBJECTIVE Rheumatoid arthritis and Ankylosing spondylitis are two common autoimmune inflammatory rheumatic diseases that negatively affect activities of daily living and can lead to structural and functional disability, reduced quality of life. Here, this study utilized Fourier transform infrared (FTIR) spectroscopy on dried serum samples and achieved early diagnosis of rheumatoid arthritis and ankylosing spondylitis based on deep learning models. METHOD A total of 243 dried serum samples were collected in this study, including 81 samples each from ankylosing spondylitis, rheumatoid arthritis, and healthy controls. Three multi-scale convolutional modules with different specifications were designed based on the multi-scale convolutional neural network (MSCNN) to effectively fuse the local features to enhance the generalization ability of the model. The FTIR was then combined with the MSCNN model to achieve a non-invasive, fast, and accurate diagnosis of ankylosing spondylitis, rheumatoid arthritis, and healthy controls. RESULTS Spectral analysis shows that the curves and waveforms of the three spectral graphs are similar. The main differences are distributed in the spectral regions of 3300-3250 cm-1, 3000-2800 cm-1, 1750-1500 cm-1, and 1500-1300 cm-1, which represent: Amides, fatty acids, cholesterol, proteins with a carboxyl group, amide II, free amino acids, and polysaccharides. Four classification models, namely artificial neural network (ANN), convolutional neural network (CNN), improved AlexNet model, and multi-scale convolutional neural network (MSCNN) were established. Through comparison, it was found that the diagnostic AUC value of the MSCNN model was 0.99, and the accuracy rate was as high as 0.93, which was much higher than the other three models. CONCLUSION The study demonstrated the superiority of MSCNN in distinguishing ankylosing spondylitis from rheumatoid arthritis and healthy controls. FTIR may become a rapid, sensitive, and non-invasive means of diagnosing rheumatism.
Collapse
Affiliation(s)
- Wei Shuai
- College of Software, Xinjiang University, Urumqi, China
| | - Xue Wu
- Department of Rheumatology and Immunology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China; Xinjiang Clinical Research Center for Rheumatoid arthritis, Urumqi, China
| | - Chen Chen
- College of Information Science and Engineering, Xinjiang University, Urumqi, China
| | - Enguang Zuo
- College of Information Science and Engineering, Xinjiang University, Urumqi, China
| | - Xiaomei Chen
- Department of Rheumatology and Immunology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China; Xinjiang Clinical Research Center for Rheumatoid arthritis, Urumqi, China
| | - Zhengfang Li
- Department of Rheumatology and Immunology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China; Xinjiang Clinical Research Center for Rheumatoid arthritis, Urumqi, China
| | - Xiaoyi Lv
- College of Software, Xinjiang University, Urumqi, China; Key Laboratory of signal detection and processing, Xinjiang University, Urumqi, China
| | - Lijun Wu
- Department of Rheumatology and Immunology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China; Xinjiang Clinical Research Center for Rheumatoid arthritis, Urumqi, China.
| | - Cheng Chen
- College of Software, Xinjiang University, Urumqi, China.
| |
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
|
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
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
Muccio S, Tavernier A, Rouchon MC, Roccon A, Dai S, Finn G, Macé S, Boutet V, Fedeli O. Validated Method Based on Immunocapture and Liquid Chromatography Coupled to High-Resolution Mass Spectrometry to Eliminate Isatuximab Interference with M-Protein Measurement in Serum. Anal Chem 2021; 93:15236-15242. [PMID: 34762405 DOI: 10.1021/acs.analchem.1c03410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In multiple myeloma (MM) disease, malignant plasma cells produce excessive quantities of a monoclonal immunoglobulin (Ig), known as M-protein. M-protein levels are measured in the serum of patients with MM using electrophoresis techniques to determine the response to treatment. However, therapeutic monoclonal antibodies, such as isatuximab, may confound signals using electrophoresis assays. We developed a robust assay based on immunocapture and liquid chromatography coupled to high-resolution mass spectrometry (IC-HPLC-HRMS) in order to eliminate this interference. Following immunocapture of Ig and free light chains (LC) in serum, heavy chains (HC) and LC were dissociated using dithiothreitol, sorted by liquid chromatography and analyzed using HRMS (Q-Orbitrap). This method allowed the M-proteins to be characterized and the signals from isatuximab and M-proteins to be discriminated. As M-protein is specific to each patient, no standards were available for absolute quantification. We therefore used alemtuzumab (an IgG kappa mAb) as a surrogate analyte for the semiquantification of M-protein in serum. This assay was successfully validated in terms of selectivity/specificity, accuracy/precision, robustness, dilution linearity, and matrix variability from 10.0 to 200 μg/mL in human serum. This method was used for clinical assessment of samples and eliminated potential interference due to isatuximab when monitoring patients with MM.
Collapse
Affiliation(s)
- Stéphane Muccio
- Sanofi R&D, 371 Rue du Professeur Blayac, 34184 Montpellier, France
| | | | | | - Alain Roccon
- Sanofi R&D, 371 Rue du Professeur Blayac, 34184 Montpellier, France
| | - Shujia Dai
- Sanofi Translational Sciences, 640 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Greg Finn
- Sanofi Oncology, 640 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Sandrine Macé
- Sanofi R&D, 1 Avenue Pierre Brossolette, 91385 Chilly-Mazarin, France
| | - Valérie Boutet
- Sanofi R&D, 1 Avenue Pierre Brossolette, 91385 Chilly-Mazarin, France
| | - Olivier Fedeli
- Sanofi R&D, 371 Rue du Professeur Blayac, 34184 Montpellier, France
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Noori S, Verkleij CPM, Zajec M, Langerhorst P, Bosman PWC, de Rijke YB, Zweegman S, VanDuijn M, Luider T, van de Donk NWCJ, Jacobs JFM. Monitoring the M-protein of multiple myeloma patients treated with a combination of monoclonal antibodies: the laboratory solution to eliminate interference. Clin Chem Lab Med 2021; 59:1963-1971. [PMID: 34392637 DOI: 10.1515/cclm-2021-0399] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/08/2021] [Indexed: 01/01/2023]
Abstract
OBJECTIVES The therapeutic monoclonal antibody (t-mAb) daratumumab, used to treat multiple myeloma (MM) patients, interferes with routine, electrophoretic based M-protein diagnostics. Electrophoretic response assessment becomes increasingly difficult when multiple t-mAbs are combined for use in a single patient. This is the first study to address the analytical challenges of M-protein monitoring when multiple t-mAbs are combined. METHODS In this proof-of-principle study we evaluate two different methods to monitor M-protein responses in three MM patients, who receive both daratumumab and nivolumab. The double hydrashift assay aims to resolve t-mAb interference on immunofixation. The MS-MRD (mass spectrometry minimal residual disease) assay measures clonotypic peptides to quantitate both M-protein and t-mAb concentrations. RESULTS After exposure to daratumumab and nivolumab, both t-mAbs become visible on immunofixation electrophoresis (IFE) as two IgG-kappa bands that migrate close to each other at the cathodal end of the γ-region. In case the M-protein co-migrates with these t-mAbs, the observed interference was completely abolished with the double IFE hydrashift assay. In all three patients the MS-MRD assay was also able to distinguish the M-protein from the t-mAbs. Additional advantage of the MS-MRD assay is that this multiplex assay is more sensitive and allows quantitative M-protein-, daratumumab- and nivolumab-monitoring. CONCLUSIONS Daratumumab and nivolumab interfere with electrophoretic M-protein diagnostics. However, the M-protein can be distinguished from both t-mAbs by use of a double hydrashift assay. The MS-MRD assay provides an alternative method that allows sensitive and simultaneous quantitative monitoring of both the M-protein and t-mAbs.
Collapse
Affiliation(s)
- Somayya Noori
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Christie P M Verkleij
- Department of Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, 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
| | - Pieter Langerhorst
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Patricia W C Bosman
- Department of Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Yolanda B de Rijke
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Martijn VanDuijn
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Theo Luider
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Niels W C J van de Donk
- Department of Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Joannes F M Jacobs
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
17
|
O'Brien A, O'Halloran F, Mykytiv V. Minimal Residual Disease in Multiple Myeloma: Potential for Blood-Based Methods to Monitor Disease. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 22:e34-e40. [PMID: 34470720 DOI: 10.1016/j.clml.2021.07.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/25/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
In recent years, the life expectancy of Multiple Myeloma (MM) patients has substantially improved, but this cancer remains incurable with increasing incidence in the developed world. Most MM patients will eventually relapse due to residual drug-resistant cancerous cells that survive treatment, commonly referred to as minimal residual disease (MRD). Methods to improve MRD detection in MM patients are generating considerable interest as a means of monitoring patients' response to treatment. In clinical laboratories, these methods currently require bone marrow aspirates which are invasive and frequently miss detection of localised disease due to the spatial heterogeneity of disease infiltration. By simplifying serial sampling and allowing for the detection of extramedullary disease, a blood-based method could significantly impact treatment duration and intensity and minimise chemotherapy-induced toxicity. This review will describe the current blood-based techniques available to detect MRD in MM and compare their potential to evaluate patient prognosis and drive therapeutic decisions.
Collapse
Affiliation(s)
- Aisling O'Brien
- Dept. of Biological Sciences, Munster Technological University, Cork, Ireland; Dept. of Immunology, Cork University Hospital, Cork, Ireland.
| | - Fiona O'Halloran
- Dept. of Biological Sciences, Munster Technological University, Cork, Ireland
| | - Vitaliy Mykytiv
- Dept. of Haematology, Cork University Hospital, Cork, Ireland; Dept. of Medicine, University College Cork, Cork, Ireland
| |
Collapse
|
18
|
McDonald Z, Taylor P, Liyasova M, Liu Q, Ma B. Mass Spectrometry Provides a Highly Sensitive Noninvasive Means of Sequencing and Tracking M-Protein in the Blood of Multiple Myeloma Patients. J Proteome Res 2021; 20:4176-4185. [PMID: 34242034 DOI: 10.1021/acs.jproteome.0c01022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The amino acid sequence of the M-protein for multiple myeloma is unique compared to the polyclonal antibodies in patients' blood. This uniqueness is exploited to develop an ultrasensitive M-protein detection method utilizing mass spectrometry (MS). The method involves the de novo amino acid sequencing of the full-length M-protein, and a targeted MS/MS assay to detect and quantify the unique M-protein sequence in serum samples. Healthy control serum spiked with NISTmAb and serial samples from an MM patient were used to demonstrate the ability of the platform to sequence and monitor a target M-protein. The de novo NISTmAb protein sequence obtained matched the published sequence, confirming the ability of the platform to accurately sequence a target M-protein in serum. NISTmAb was quantified down to 0.0002 g/dL in serum, a level hundreds of times more sensitive than conventional blood-based tests such as SPEP and IFE. The M-protein in the patient sample could be quantified throughout complete remission, demonstrating the utility of the assay to track M-protein considerably beyond the sensitivities of current blood-based tests. Notably, the assay detected a 2-fold rise in M-protein levels 10 months before any changes were detected by conventional IFE. The MS-based assay is highly sensitive, noninvasive, and requires only a small amount of serum, less than 100 μL. Sequencing data is deposited into PRIDE with identifier PXD022784, and quantification data can be found in Panorama Public with identifier PXD022980.
Collapse
Affiliation(s)
- Zac McDonald
- Rapid Novor, Inc., Kitchener, Ontario N2G 4X8, Canada
| | - Paul Taylor
- Rapid Novor, Inc., Kitchener, Ontario N2G 4X8, Canada
| | | | - Qixin Liu
- Rapid Novor, Inc., Kitchener, Ontario N2G 4X8, Canada
| | - Bin Ma
- University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
19
|
Santockyte R, Jin C, Pratt J, Ammar R, Desai K, Bolisetty M, Das P, Popa-McKiver M, Puig O. Sensitive multiple myeloma disease monitoring by mass spectrometry. Blood Cancer J 2021; 11:78. [PMID: 33927186 PMCID: PMC8084958 DOI: 10.1038/s41408-021-00473-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 03/22/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022] Open
Affiliation(s)
- Rasa Santockyte
- Translational Medicine, Bristol Myers Squibb, Lawrence Township, NJ, USA.
| | - Chelsea Jin
- Biometrics and Data Sciences, Bristol Myers Squibb, Lawrence Township, NJ, USA
| | - James Pratt
- Biometrics and Data Sciences, Bristol Myers Squibb, Lawrence Township, NJ, USA
| | - Ron Ammar
- Translational Medicine, Bristol Myers Squibb, Lawrence Township, NJ, USA
| | - Keyur Desai
- Biometrics and Data Sciences, Bristol Myers Squibb, Lawrence Township, NJ, USA
| | - Mohan Bolisetty
- Translational Medicine, Bristol Myers Squibb, Lawrence Township, NJ, USA
| | - Prianka Das
- Hematology Clinical Development, Bristol Myers Squibb, Lawrence Township, NJ, USA
| | - Mihaela Popa-McKiver
- Hematology Clinical Development, Bristol Myers Squibb, Lawrence Township, NJ, USA
| | - Oscar Puig
- Translational Medicine, Bristol Myers Squibb, Lawrence Township, NJ, USA. .,Eli Lilly and Company, 450 East 29th St, New York, NY, 10016, USA.
| |
Collapse
|
20
|
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
|
21
|
Langerhorst P, Brinkman AB, VanDuijn MM, Wessels HJCT, Groenen PJTA, Joosten I, van Gool AJ, Gloerich J, Scheijen B, Jacobs JFM. Clonotypic Features of Rearranged Immunoglobulin Genes Yield Personalized Biomarkers for Minimal Residual Disease Monitoring in Multiple Myeloma. Clin Chem 2021; 67:867-875. [PMID: 33709101 DOI: 10.1093/clinchem/hvab017] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/12/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Due to improved treatment, more patients with multiple myeloma (MM) reach a state of minimal residual disease (MRD). Different strategies for MM MRD monitoring include flow cytometry, allele-specific oligonucleotide-quantitative PCR, next-generation sequencing, and mass spectrometry (MS). The last 3 methods rely on the presence and the stability of a unique immunoglobulin fingerprint derived from the clonal plasma cell population. For MS-MRD monitoring it is imperative that MS-compatible clonotypic M-protein peptides are identified. To support implementation of molecular MRD techniques, we studied the presence and stability of these clonotypic features in the CoMMpass database. METHODS An analysis pipeline based on MiXCR and HIGH-VQUEST was constructed to identify clonal molecular fingerprints and their clonotypic peptides based on transcriptomic datasets. To determine the stability of the clonal fingerprints, we compared the clonal fingerprints during disease progression for each patient. RESULTS The analysis pipeline to establish the clonal fingerprint and MS-suitable clonotypic peptides was successfully validated in MM cell lines. In a cohort of 609 patients with MM, we demonstrated that the most abundant clone harbored a unique clonal molecular fingerprint and that multiple unique clonotypic peptides compatible with MS measurements could be identified for all patients. Furthermore, the clonal immunoglobulin gene fingerprints of both the light and heavy chain remained stable during MM disease progression. CONCLUSIONS Our data support the use of the clonal immunoglobulin gene fingerprints in patients with MM as a suitable MRD target for MS-MRD analyses.
Collapse
Affiliation(s)
- Pieter Langerhorst
- Laboratory Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands.,Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Arie B Brinkman
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Martijn M VanDuijn
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Hans J C T Wessels
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Patricia J T A Groenen
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Irma Joosten
- Laboratory Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Alain J van Gool
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jolein Gloerich
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Blanca Scheijen
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Joannes F M Jacobs
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
22
|
Clearing drug interferences in myeloma treatment using mass spectrometry. Clin Biochem 2021; 92:61-66. [PMID: 33691184 DOI: 10.1016/j.clinbiochem.2021.02.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/29/2021] [Accepted: 02/22/2021] [Indexed: 01/26/2023]
Abstract
OBJECTIVE To explore the possibility of using a combination of a rapid MALDI-TOF-MS method (Mass-Fix) in conjunction with higher resolution LC-ESI-QTOF-MS (miRAMM) measurements to discriminate the IgG kappa M-protein from daratumumab, elotuzumab and isatuximab in myeloma patients. DESIGN & METHODS 86 patients with an IgG kappa M-protein were spiked with therapeutic levels of the drugs and examined by Mass-Fix and miRAMM to establish the percent of cases that could be resolved by each method. The method was then applied to 21 samples from patients receiving one of the drugs. RESULTS Mass-Fix was capable of resolving the t-mAb from M-protein for 87 percent of the spiked samples. For the cases unresolved by Mass-Fix, miRAMM was capable of resolving the remaining drug interferences. The 21 IgG kappa myeloma patients that were receiving the drugs were all resolved by Mass-Fix. CONCLUSION This proposed algorithm allows use of a clinical available assay (Mass-Fix) while maximizing the number of cases that can accurately resolve the t-mAb from the M-protein.
Collapse
|
23
|
Using MALDI-TOF mass spectrometry in peripheral blood for the follow up of newly diagnosed multiple myeloma patients treated with daratumumab-based combination therapy. Clin Chim Acta 2021; 516:136-141. [PMID: 33545108 DOI: 10.1016/j.cca.2021.01.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/31/2020] [Accepted: 01/25/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Daratumumab-based combination therapies have shown high rates of complete response (CR) and minimal residual disease negativity in patients with multiple myeloma. However, daratumumab, an IgGκ monoclonal antibody, interferes with electrophoretic techniques making it difficult to reliably define residual disease versus CR, especially in patients with IgGκ multiple myeloma. METHODS Enrichment with polyclonal sheep antibody-coated magnetic microparticles combined with MALDI-TOF mass spectrometry (MALDI-TOF MS) analysis was used to detect M-proteins in serial samples from newly diagnosed multiple myeloma patients treated with daratumumab-based therapy. The performance of the MALDI-TOF MS assay was compared to that of a routine test panel (serum protein electrophoresis (SPEP), immunofixation (IFE) and serum free light chain (FLC)). RESULTS Comparison of MALDI-TOF MS to SPEP/IFE/FLC showed a concordance of 84.9% (p < 0.001). When MALDI-TOF MS and FLC results were combined, the M-protein detection rate was the same or better than the routine test panel. For the 9 patients who obtained CR during follow-up, MALDI-TOF MS detected an M-protein in 46% of subsequent samples. Daratumumab could be distinguished from the M-protein in 215/222 samples. CONCLUSION MALDI-TOF MS is useful in assessing CR in patients treated with monoclonal antibody-based therapies.
Collapse
|
24
|
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
|
25
|
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
|
26
|
Santockyte R, Puig O, Zheng N, Ouyang Z, Titsch C, Zhang YJ, Pillutla R, Zeng J. High-Throughput Therapeutic Antibody Interference-Free High-Resolution Mass Spectrometry Assay for Monitoring M-Proteins in Multiple Myeloma. Anal Chem 2020; 93:834-842. [DOI: 10.1021/acs.analchem.0c03357] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Rasa Santockyte
- Department of Translational Medicine, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Oscar Puig
- Department of Translational Medicine, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Naiyu Zheng
- Department of Translational Medicine, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Zheng Ouyang
- Department of Translational Medicine, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Craig Titsch
- Department of Translational Medicine, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Yang J. Zhang
- Department of Translational Medicine, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Renuka Pillutla
- Department of Translational Medicine, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Jianing Zeng
- Department of Translational Medicine, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| |
Collapse
|
27
|
Zajec M, Langerhorst P, VanDuijn MM, Gloerich J, Russcher H, van Gool AJ, Luider TM, Joosten I, de Rijke YB, Jacobs JFM. Mass Spectrometry for Identification, Monitoring, and Minimal Residual Disease Detection of M-Proteins. Clin Chem 2020; 66:421-433. [PMID: 32031591 DOI: 10.1093/clinchem/hvz041] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/13/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Monoclonal gammopathies (MGs) are plasma cell disorders defined by the clonal expansion of plasma cells, resulting in the characteristic excretion of a monoclonal immunoglobulin (M-protein). M-protein detection and quantification are integral parts of the diagnosis and monitoring of MGs. Novel treatment modalities impose new challenges on the traditional electrophoretic and immunochemical methods that are routinely used for M-protein diagnostics, such as interferences from therapeutic monoclonal antibodies and the need for increased analytical sensitivity to measure minimal residual disease. CONTENT Mass spectrometry (MS) is ideally suited to accurate mass measurements or targeted measurement of unique clonotypic peptide fragments. Based on these features, MS-based methods allow for the analytically sensitive measurement of the patient-specific M-protein. SUMMARY This review provides a comprehensive overview of the MS methods that have been developed recently to detect, characterize, and quantify M-proteins. The advantages and disadvantages of using these techniques in clinical practice and the impact they will have on the management of patients with MGs are discussed.
Collapse
Affiliation(s)
- M Zajec
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.,Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - P Langerhorst
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M M VanDuijn
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - J Gloerich
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - H Russcher
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - A J van Gool
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - T M Luider
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - I Joosten
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Y B de Rijke
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - J F M Jacobs
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
28
|
Cerebrospinal Fluid Penetrance of Daratumumab in Leptomeningeal Multiple Myeloma. Hemasphere 2020; 4:e413. [PMID: 32904033 PMCID: PMC7448363 DOI: 10.1097/hs9.0000000000000413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/06/2020] [Indexed: 11/26/2022] Open
|
29
|
Martins CO, Huet S, Yi SS, Ritorto MS, Landgren O, Dogan A, Chapman JR. Mass Spectrometry-Based Method Targeting Ig Variable Regions for Assessment of Minimal Residual Disease in Multiple Myeloma. J Mol Diagn 2020; 22:901-911. [PMID: 32302778 DOI: 10.1016/j.jmoldx.2020.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 03/25/2020] [Accepted: 04/01/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma is a systemic malignancy of monoclonal plasma cells that accounts for 10% of hematologic cancers. With development of highly effective therapies for multiple myeloma, minimal residual disease (MRD) assessment has emerged as an important end point for management decisions. Currently, serologic assays lack the sensitivity for MRD assessment, and invasive bone marrow sampling with flow cytometry or molecular methods has emerged as the gold standard. We report a sensitive and robust targeted mass spectrometry proteomics method to detect MRD in serum, without the need of invasive, sequential bone marrow aspirates. The method detects Ig-derived clonotypic tryptic peptides predicted by sequencing the clonal plasma cell Ig genes. A heavy isotope-labeled Ig internal standard is added to patient serum at a known concentration, the Ig is enriched in a light chain type specific manner, and proteins are digested and analyzed by targeted mass spectrometry. Peptides from the constant regions of the λ or κ light chains, Ig heavy chains, and clonotypic peptides unique to the patient monoclonal Igs are targeted. This technique is highly sensitive and specific for the patient-specific monoclonal Igs, even in samples negative by multiparametric flow cytometry. Our method can accurately and precisely detect monoclonal protein in serum of patients treated for myeloma and has broad implications for management of hematologic patients.
Collapse
Affiliation(s)
- Carlo O Martins
- Hematopathology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sarah Huet
- Hematopathology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - San S Yi
- Hematopathology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria S Ritorto
- Hematopathology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ola Landgren
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmet Dogan
- Hematopathology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jessica R Chapman
- Hematopathology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
30
|
Complete Depletion of Daratumumab Interference in Serum Samples from Plasma Cell Myeloma Patients Improves the Detection of Endogenous M-Proteins in a Preliminary Study. Diagnostics (Basel) 2020; 10:diagnostics10040219. [PMID: 32295157 PMCID: PMC7235820 DOI: 10.3390/diagnostics10040219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/11/2020] [Accepted: 04/08/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Therapeutic humanized IgG1 kappa monoclonal antibody (t-mAb), daratumumab (DARA) is a Food and Drug Administration approved drug for the treatment of relapsed/refractory plasma cell myeloma (PCM). DARA appears on serum protein electrophoresis (SPEP) and on serum immunofixation (sIFE) as an IgG kappa monoclonal immunoglobulin protein (M-protein), complicating the assessment of the patients' response to therapy. A more ominous threat to patient safety can occur with the misinterpretation of the presence of a small t-mAb spike as being the residual product of the patient's neoplastic clone, presented either as oligoclonality or new clonality, which could result in incorrect interpretation of failure to achieve remission. METHODS In this report, we describe a novel and cost-effective technique based on biotinylated recombinant CD38 and streptavidin-coated magnetic beads to capture and remove residual DARA present in PCM patient serum samples. The treated samples are then run like regular samples on SPEP and sIFE. We validated this simple technique in DARA-spiked PCM samples and patient samples on DARA treatment. RESULTS Our simple capture technique completely extracted DARA in all of the tested serum specimens and allowed the assessment of residual M-protein without DARA interference. The results were reproducible and highly specific for DARA, and did not have any impact on endogenous M-protein migration and quantification by SPEP and sIFE. The cost of this technique is much lower and it can be performed in-house with a very short turnaround time compared to the currently available alternative methods. There is a great need for such reflex technologies to avoid interpretation errors. CONCLUSIONS This method is an effective way to eliminate DARA interference in SPEP and sIFE, and can be easily implemented in any clinical laboratory without any patent restriction. This simple technique can be adopted for other t-mAbs using their respective ligands and will help to reduce additional doses of toxic treatment and further testing in patients on t-mAbs with a false positive M-protein spike.
Collapse
|
31
|
Tracking of low disease burden in multiple myeloma: Using mass spectrometry assays in peripheral blood. Best Pract Res Clin Haematol 2020; 33:101142. [PMID: 32139008 DOI: 10.1016/j.beha.2020.101142] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 01/09/2020] [Indexed: 11/21/2022]
Abstract
Efforts over the last 5 years have demonstrated that it is technically feasible to detect low levels of monoclonal proteins in peripheral blood using mass spectrometry. These methods are based on the fact that an M-protein has a specific amino acid sequence, and therefore, a specific mass. This mass can be tracked over time and can serve as a surrogate marker of the presence of clonal plasma cells. This review describes the use of mass spectrometry to detect M-proteins in multiple myeloma to date, identifies the challenges of using this biomarker, and describes potential strategies to overcome these challenges. We discuss the work that must be done for these techniques to be incorporated into clinical practice for tracking of low disease burden in multiple myeloma.
Collapse
|
32
|
Zajec M, Jacobs JFM, de Kat Angelino CM, Dekker LJM, Stingl C, Luider TM, De Rijke YB, VanDuijn MM. Integrating Serum Protein Electrophoresis with Mass Spectrometry, A New Workflow for M-Protein Detection and Quantification. J Proteome Res 2020; 19:2845-2853. [DOI: 10.1021/acs.jproteome.9b00705] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Marina Zajec
- Department of Neurology, Erasmus MC University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Clinical Chemistry, Erasmus MC University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Joannes F. M. Jacobs
- Department of Laboratory Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Corrie M. de Kat Angelino
- Department of Laboratory Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Lennard J. M. Dekker
- Department of Neurology, Erasmus MC University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Christoph Stingl
- Department of Neurology, Erasmus MC University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Theo M. Luider
- Department of Neurology, Erasmus MC University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Yolanda B. De Rijke
- Department of Clinical Chemistry, Erasmus MC University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Martijn M. VanDuijn
- Department of Neurology, Erasmus MC University Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
33
|
Fleming CK, Swarttouw T, de Kat Angelino CM, Jacobs JF, Russcher H. Method comparison of four clinically available assays for serum free light chain analysis. ACTA ACUST UNITED AC 2019; 58:85-94. [DOI: 10.1515/cclm-2019-0533] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/29/2019] [Indexed: 12/22/2022]
Abstract
Abstract
Background
Serum free light chain (sFLC) measurements are increasingly important in the context of screening for monoclonal gammopathies, prognostic stratification and monitoring of therapy responses. In this study we have performed a method comparison of four sFLC assays that are currently available for routine clinical use.
Methods
In a retrospective study, sFLC analyses were performed on a cohort that included 139 patients with various monoclonal gammopathies and 54 control sera without an M-protein. Method comparisons of the following four FLC assays were performed: Freelite (Binding Site), N-Latex FLC (Siemens), Seralite (Abingdon Health) and Sebia FLC (Sebia).
Results
Bland-Altman agreement analysis showed biases varying between −0.1 and 16.2 mg/L for κFLC, −6.0 and 6.8 mg/L for λFLC and −0.04 and 0.38 for the ratio of the involved to uninvolved FLC. Strong agreements were observed for FLC-concentrations below 100 mg/L. The clinical concordance of the κ/λFLC-ratio of the four methods varied between 86% and 92%. Significant quantitative differences were observed between the different methods, mainly in sera with high FLC concentrations. Most assays consistently overestimated FLC concentrations compared to SPE.
Conclusions
Good overall clinical concordances were observed between the four sFLC assays that were compared in this study. Although good agreements were observed between the FLC assays, significant absolute differences in FLC concentrations in individual patients can be seen, particularly at higher FLC concentrations. Because of inequivalent absolute sFLC values between the methods in individual patients, none of the four sFLC assays can be used interchangeably.
Collapse
Affiliation(s)
- Chérina K.A. Fleming
- Department of Clinical Chemistry, Erasmus MC , University Medical Center Rotterdam , Rotterdam , The Netherlands
| | - Tim Swarttouw
- Department of Clinical Chemistry, Erasmus MC , University Medical Center Rotterdam , Rotterdam , The Netherlands
| | | | - Joannes F.M. Jacobs
- Department of Laboratory Medicine , Radboud University Medical Center , Nijmegen , The Netherlands
| | - Henk Russcher
- Department of Clinical Chemistry, Erasmus MC , University Medical Center Rotterdam , Rotterdam , The Netherlands
| |
Collapse
|
34
|
MacMullan MA, Dunn ZS, Graham NA, Yang L, Wang P. Quantitative Proteomics and Metabolomics Reveal Biomarkers of Disease as Potential Immunotherapy Targets and Indicators of Therapeutic Efficacy. Theranostics 2019; 9:7872-7888. [PMID: 31695805 PMCID: PMC6831481 DOI: 10.7150/thno.37373] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/19/2019] [Indexed: 02/07/2023] Open
Abstract
Quantitative mass spectrometry (MS) continues to deepen our understanding of the immune system, quickly becoming the gold standard for obtaining high-throughput, quantitative data on biomolecules. The development of targeted and multiplexed assays for biomarker quantification makes MS an attractive tool both for diagnosing diseases and for quantifying the effects of immunotherapeutics. Because of its accuracy, the use of MS for identifying biomarkers of disease reduces the potential for misdiagnosis and overtreatment. Advances in workflows for sample processing have drastically reduced processing time and complexities due to sample preparation, making MS a more accessible technology. In this review, we present how recent developments in proteomics and metabolomics make MS an essential component of enhancing and monitoring the efficacy of immunotherapeutic treatments.
Collapse
Affiliation(s)
- Melanie A. MacMullan
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California
| | - Zachary S. Dunn
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California
| | - Nicholas A. Graham
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, California
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California
| |
Collapse
|
35
|
A universal solution for eliminating false positives in myeloma due to therapeutic monoclonal antibody interference. Blood 2018; 132:670-672. [DOI: 10.1182/blood-2018-05-848986] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|