1
|
Liang H, Li Y, Liu C, Wang H, Ren Y, Sun F, Xue M, Zhu G, Zhou Y. Raman spectroscopy of dried serum for the detection of rapid noninvasive multiple myeloma. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 328:125448. [PMID: 39579726 DOI: 10.1016/j.saa.2024.125448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/10/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024]
Abstract
Raman spectroscopy has recently emerged as an attractive focus of research interest in studies of hematological diseases. However, comprehensive Raman spectral analyses of serum samples from patients with multiple myeloma (MM) or other forms of lymphatic neoplastic disease are lacking. In this study, laser Raman spectroscopy and orthogonal partial least-squares discrimination analysis (OPLS-DA) were employed to develop a simple, noninvasive approach to MM diagnosis based on dried serum samples. To that end, systematic OPLS-DA analyses of dried serum from 35 patients with MM and 13 healthy controls were performed, revealing clear differences between these two groups in terms of the resultant serum spectral data. Specifically, significant reductions in the intensities of Raman peaks corresponding to nucleic acids (726, 781, 1579 cm-1), proteins (621, 1603, 1616 cm-1), lipids (1437, 1443, 1446 cm-1) and carotenoids (957, 1160, 1520 cm-1) were observed in MM, together with increases in the intensities of peaks corresponding to carbohydrates (920, 1123 cm-1) and collagen (1345 cm-1). Through combined analyses of serological indices associated with metabolic activity, MM patients were confirmed to exhibit elevated serum glucose levels and decreased levels of high-density lipoprotein cholesterol. These results offer a spectroscopic foundation for the relationships between MM classification and serological testing data, offering new evidence that can guide the early and efficient identification and characterization of this deadly cancer type. This exploratory study thus offers insight into the potential utility of Raman spectroscopy as a tool for the noninvasive detection of specific subtypes of MM.
Collapse
Affiliation(s)
- Haoyue Liang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Yihan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Chang Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Haoyu Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Yansong Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Fanfan Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Mei Xue
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Guoqing Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600, China.
| | - Yuan Zhou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600, China.
| |
Collapse
|
2
|
Lee J, Choi JH, Kim EH, Im J, Hwang H, Yang S, Lee JH, Lee K, Song J, Park S, Song SH. Detecting M-Protein via Mass Spectrometry and Affinity Beads: Enrichment With Mixed Kappa-Lambda Beads Enables Prompt Application in Clinical Laboratories. Ann Lab Med 2024; 44:518-528. [PMID: 39161319 PMCID: PMC11375182 DOI: 10.3343/alm.2024.0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/23/2024] [Accepted: 06/27/2024] [Indexed: 08/21/2024] Open
Abstract
Background Detecting monoclonal protein (M-protein), a hallmark of plasma cell disorders, traditionally relies on methods such as protein electrophoresis, immune-electrophoresis, and immunofixation electrophoresis (IFE). Mass spectrometry (MS)-based methods, such as matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) and electrospray ionization-quadrupole time-of-flight (ESI-qTOF) MS, have emerged as sensitive methods. We explored the M-protein-detection efficacies of different MS techniques. Methods To isolate immunoglobulin and light chain proteins, six types of beads (IgG, IgA, IgM, kappa, lambda, and mixed kappa and lambda) were used to prepare samples along with CaptureSelect nanobody affinity beads (NBs). After purification, both MALDI-TOF MS and liquid chromatography coupled with Synapt G2 ESI-qTOF high-resolution MS analysis were performed. We purified 25 normal and 25 abnormal IFE samples using NBs and MALDI-TOF MS (NB-MALDI-TOF). Results Abnormal samples showed monoclonal peaks, whereas normal samples showed polyclonal peaks. The IgG and mixed kappa and lambda beads showed monoclonal peaks following the use of daratumumab (an IgG/kappa type of monoclonal antibody) with both MALDI-TOF and ESI-qTOF MS analysis. The limits of detection for MALDI-TOF MS and ESI-qTOF MS were established as 0.1 g/dL and 0.025 g/dL, respectively. NB-MALDI-TOF and IFE exhibited comparable sensitivity and specificity (92% and 92%, respectively). Conclusions NBs for M-protein detection, particularly with mixed kappa-lambda beads, identified monoclonal peaks with both MALDI-TOF and ESI-qTOF analyses. Qualitative analysis using MALDI-TOF yielded results comparable with that of IFE. NB-MALDI-TOF might be used as an alternative method to replace conventional tests (such as IFE) to detect M-protein with high sensitivity.
Collapse
Affiliation(s)
- Jikyo Lee
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jung Hoon Choi
- Digital OMICs Research Center, Korea Basic Science Institute, Cheongju, Korea
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Eun-Hee Kim
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jihyun Im
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Heeyoun Hwang
- Digital OMICs Research Center, Korea Basic Science Institute, Cheongju, Korea
| | - Seojin Yang
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Joon Hee Lee
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kyunghoon Lee
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Junghan Song
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Seungman Park
- Department of Laboratory Medicine, National Cancer Center, Goyang, Korea
| | - Sang Hoon Song
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
3
|
Ranjit Anderson N, Korczyk D, Mollee P. Improving care for systemic light-chain amyloidosis patients: is a multidisciplinary approach best? Expert Rev Hematol 2024; 17:567-579. [PMID: 39155770 DOI: 10.1080/17474086.2024.2388184] [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: 04/08/2024] [Revised: 07/21/2024] [Accepted: 07/31/2024] [Indexed: 08/20/2024]
Abstract
INTRODUCTION Light chain (AL) amyloidosis is a rare and complex disease which can affect various systems of the body. In common with many rare and multisystemic diseases, the breadth of diagnostic, clinical, and supportive expertise required to care for such patients is best met by a multidisciplinary team. AREAS COVERED We outline different phases of the patients' journey, including diagnosis, staging, treatment, and response assessment, to highlight common clinical issues best resolved by a multidisciplinary approach. EXPERT OPINION To extend the benefit of multidisciplinary care to the majority of patients with AL amyloidosis, innovative healthcare models such as telehealth and multisite multidisciplinary team meetings need to be implemented. The need for a multidisciplinary approach where such a wide array of healthcare skills is required also highlights the shortcomings of our current diagnostic and monitoring assays. Better access to diagnostic and subtyping assays is necessary. The ability to characterize and measure the causative amyloidogenic light chain as well as imaging techniques to accurately diagnose and monitor response to therapy is also needed and is currently an area of research focus.
Collapse
Affiliation(s)
- Nirija Ranjit Anderson
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Dariusz Korczyk
- Faculty of Medicine, University of Queensland, Brisbane, Australia
- Department of Cardiology, Princess Alexandra Hospital, Brisbane, Australia
- Queensland Amyloidosis Centre, Princess Alexandra Hospital, Brisbane, Australia
| | - Peter Mollee
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Australia
- Queensland Amyloidosis Centre, Princess Alexandra Hospital, Brisbane, Australia
| |
Collapse
|
4
|
Bomsztyk J, Ravichandran S, Giles HV, Wright N, Berlanga O, Khwaja J, Mahmood S, Wisniowski B, Cohen O, Foard D, Gilbertson J, Rauf MU, Starr N, Martinez-Naharro A, Venneri L, Whelan C, Fontana M, Hawkins PN, Gillmore JD, Lachmann H, Harding S, Pratt G, Wechalekar AD. Complete responses in AL amyloidosis are unequal: the impact of free light chain mass spectrometry in AL amyloidosis. Blood 2024; 143:1259-1268. [PMID: 38194690 DOI: 10.1182/blood.2023022399] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 01/11/2024] Open
Abstract
ABSTRACT Amyloidogenic serum free light chains (sFLCs) drive disease progression in AL amyloidosis. Matrix-assisted laser desorption/ionization time of flight mass spectrometry-based FLC assay (FLC-MS) has greater sensitivity than conventional sFLC assays allowing for the detection of serological residual disease. We report the utility of FLC-MS in a large series of patients with AL amyloidosis assessing the impact of FLC-MS negativity after treatment on overall survival (OS) and organ response rates. Serum samples were analyzed using FLC-MS at diagnosis and at 6 and 12 months after treatment. The impact of FLC-MS negativity over standard hematologic responses on survival and organ response was assessed. A total of 487 patients were included; 290 (59%) and 349 (71.5%) had cardiac and renal involvement, respectively. There was 100% concordance between the light chain (LC) fibril type and LC isotype identified by FLC-MS. At 6 and 12 months, 81 (16.6%) and 101 (20.7%) were FLC-MS negative. Of those achieving a conventional hematologic complete response (CR) at 6 and 12 months, 45 (27.7%) and 64 (39%) were FLC-MS negative. At 12 months, median OS for CR + FLC-MS negative was not reached vs 108 months in CR + FLC-MS positive (P = .024). At 12 months, 70% of patients with FLC-MS negativity (vs 50% FLC-MS positive) achieved a cardiac response (P = .015). In a multivariate analysis, FLC-MS negativity at 12 months was an independent predictor of better outcomes. FLC-MS can detect persistent monoclonal light chains in a significant proportion of patients in a conventional hematologic CR. FLC-MS assessment promises to be a new standard for response assessment in AL amyloidosis.
Collapse
Affiliation(s)
- Joshua Bomsztyk
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
| | | | - Hannah V Giles
- University Hospital Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- University of Birmingham, Birmingham, United Kingdom
| | | | | | | | - Shameem Mahmood
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
- University College London Hospital, London, United Kingdom
| | - Brendan Wisniowski
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
- University College London Hospital, London, United Kingdom
| | - Oliver Cohen
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
| | - Darren Foard
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
| | - Janet Gilbertson
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
| | - Muhammad U Rauf
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
| | - Neasa Starr
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
| | | | - Lucia Venneri
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
| | - Carol Whelan
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
| | - Marianna Fontana
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
| | - Philip N Hawkins
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
| | - Julian D Gillmore
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
| | - Helen Lachmann
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
| | | | - Guy Pratt
- University Hospital Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- University of Birmingham, Birmingham, United Kingdom
| | - Ashutosh D Wechalekar
- National Amyloid Centre, Royal Free Hospital, London, United Kingdom
- University College London Hospital, London, United Kingdom
| |
Collapse
|
5
|
Fan H, Wang B, Shi L, Pan N, Yan W, Xu J, Gong L, Li L, Liu Y, Du C, Cui J, Zhu G, Deng S, Sui W, Xu Y, Yi S, Hao M, Zou D, Chen X, Qiu L, An G. Monitoring Minimal Residual Disease in Patients with Multiple Myeloma by Targeted Tracking Serum M-Protein Using Mass Spectrometry (EasyM). Clin Cancer Res 2024; 30:1131-1142. [PMID: 38170583 PMCID: PMC10940853 DOI: 10.1158/1078-0432.ccr-23-2767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/10/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024]
Abstract
PURPOSE We investigated both the clinical utilities and the prognostic impacts of the clonotypic peptide mass spectrometry (MS)-EasyM, a blood-based minimal residual disease (MRD) monitoring protocol in multiple myeloma. EXPERIMENTAL DESIGN A total of 447 sequential serum samples from 56 patients with multiple myeloma were analyzed using EasyM. Patient-specific M-protein peptides were sequenced from diagnostic samples; sequential samples were quantified by EasyM to monitor the M-protein. The performance of EasyM was compared with serum immunofixation electrophoresis (IFE), bone marrow multiparameter flow cytometry (MFC), and next-generation flow cytometry (NGF) detection. The optimal balance of EasyM sensitivity/specificity versus NGF (10-5 sensitivity) was determined and the prognostic impact of MS-MRD status was investigated. RESULTS Of the 447 serum samples detected and measured by EasyM, 397, 126, and 92 had time-matching results for comparison with serum IFE, MFC-MRD, and NGF-MRD, respectively. Using a dotp >0.9 as the MS-MRD positive, sensitivity was 99.6% versus IFE and 100.0% versus MFC and NGF. Using an MS negative cutoff informed by ROC analysis (<1.86% of that at diagnosis), EasyM sensitivity remained high versus IFE (88.3%), MFC (85.1%), and NGF (93.2%), whereas specificity increased to 90.4%, 55.8%, and 93.2%, respectively. In the multivariate analysis, older diagnostic age was an independent predictor for progression-free survival [PFS; high risk (HR), 3.15; 1.26-7.86], the best MS-MRD status (MS-MRD negative) was independent predictor for both PFS (HR, 0.25; 0.12-0.52) and overall survival (HR, 0.16; 0.06-0.40). CONCLUSIONS EasyM is a highly sensitive and minimal invasive method of MRD monitoring in multiple myeloma; MS-MRD had significant predictive ability for survival outcomes.
Collapse
Affiliation(s)
- Huishou Fan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Bing Wang
- Shanghai Kuaixu Biotechnology Co., Ltd., Shanghai, China
| | - Lihui Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ni Pan
- Shanghai Kuaixu Biotechnology Co., Ltd., Shanghai, China
| | - Wenqiang Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jingyu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lixin Gong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lingna Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yuntong Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Chenxing Du
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jian Cui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Guoqing Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhui Deng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhua Yi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Mu Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiequn Chen
- Department of Hematology, Affiliated Hospital of Northwest University, Institute of Hematology, Northwest University, Xian, Shaanxi, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Gang An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
6
|
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 PMCID: PMC10967543 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
|
7
|
Guan L, Su W, Zhong J, Qiu L. M-protein detection by mass spectrometry for minimal residual disease in multiple myeloma. Clin Chim Acta 2024; 552:117623. [PMID: 37924928 DOI: 10.1016/j.cca.2023.117623] [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: 08/07/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 11/06/2023]
Abstract
Multiple myeloma (MM) is characterized by excessive production of monoclonal immunoglobulins (M proteins). Routine screening methods for M proteins to assess prognosis are unable to detect low levels of M proteins produced by residual tumor cells, ie, minimal residual disease (MRD). Assessment of MRD can be conducted by examining residual tumor cells in bone marrow or circulating M proteins. Advances in mass spectrometry have enabled reliable and highly sensitive detection of low abundance serum biomarkers making it a viable and significantly less invasive approach. Mass spectrometry can achieve dynamic monitoring of MRD and identify therapeutic monoclonal antibodies as well as oligoclonal proteins. In this review we summarize mass spectrometry methods in M protein detection and their applications of MRD detection in MM.
Collapse
Affiliation(s)
- Lihua Guan
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, PR China
| | - Wei Su
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, PR China
| | - Jian Zhong
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, PR China
| | - Ling Qiu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, PR China; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing 100730, PR China.
| |
Collapse
|
8
|
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
|
9
|
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: 3.0] [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
|
10
|
Hermouet S, Bigot-Corbel E, Harb J. Determination of the target of monoclonal immunoglobulins: a novel diagnostic tool for individualized MGUS therapy, and prevention and therapy of smoldering and multiple myeloma. Front Immunol 2023; 14:1253363. [PMID: 38022528 PMCID: PMC10644846 DOI: 10.3389/fimmu.2023.1253363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/11/2023] [Indexed: 12/01/2023] Open
Abstract
Subsets of patients diagnosed with a monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM) or multiple myeloma (MM), present with a monoclonal immunoglobulin (Ig) specific for an infectious pathogen, including hepatitis C and B viruses (HCV, HBV), Helicobacter pylori and several Herpesviruses. Such cases are likely initiated by infection, since in the context of HCV- or HBV-infected patients, antiviral therapy can lead to the disappearance of antigenic stimulation, control of clonal plasma cells, and reduced or suppressed monoclonal Ig production. Complete remission has been obtained with anti-HCV therapy in refractory MM with a HCV-specific monoclonal Ig, and antiviral treatments significantly improved the probability of survival of MM patients infected with HCV or HBV prior to the diagnosis of MM. Monoclonal Igs may also target glucolipids, particularly glucosylsphingosine (GlcSph), and GlcSph-reducing therapy can lead to complete remission in SMM and MM patients presenting with a GlcSph-specific monoclonal Ig. The present review describes the importance of determining the target of the monoclonal Ig of MGUS, SMM and MM patients, and discusses the efficacy of target-reducing treatments in the management of MGUS, SMM and MM cases who present with a monoclonal Ig reactive against a treatable infectious pathogen or GlcSph.
Collapse
Affiliation(s)
- Sylvie Hermouet
- Nantes Université, INSERM, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, Nantes, France
- Laboratoire d’Hématologie, CHU Nantes, Nantes, France
| | - Edith Bigot-Corbel
- Nantes Université, INSERM, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, Nantes, France
- Laboratoire de Biochimie, CHU Nantes, Nantes, France
| | - Jean Harb
- Laboratoire de Biochimie, CHU Nantes, Nantes, France
| |
Collapse
|
11
|
Watson E, Ramasamy K. Flicking the switch in myeloma MRD. Blood 2023; 141:557-558. [PMID: 36757729 DOI: 10.1182/blood.2022018394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
12
|
Casadonte R, Kriegsmann J, Kriegsmann M, Kriegsmann K, Torcasio R, Gallo Cantafio ME, Viglietto G, Amodio N. A Comparison of Different Sample Processing Protocols for MALDI Imaging Mass Spectrometry Analysis of Formalin-Fixed Multiple Myeloma Cells. Cancers (Basel) 2023; 15:cancers15030974. [PMID: 36765932 PMCID: PMC9913598 DOI: 10.3390/cancers15030974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Sample processing of formalin-fixed specimens constitutes a major challenge in molecular profiling efforts. Pre-analytical factors such as fixative temperature, dehydration, and embedding media affect downstream analysis, generating data dependent on technical processing rather than disease state. In this study, we investigated two different sample processing methods, including the use of the cytospin sample preparation and automated sample processing apparatuses for proteomic analysis of multiple myeloma (MM) cell lines using imaging mass spectrometry (IMS). In addition, two sample-embedding instruments using different reagents and processing times were considered. Three MM cell lines fixed in 4% paraformaldehyde were either directly centrifuged onto glass slides using cytospin preparation techniques or processed to create paraffin-embedded specimens with an automatic tissue processor, and further cut onto glass slides for IMS analysis. The number of peaks obtained from paraffin-embedded samples was comparable between the two different sample processing instruments. Interestingly, spectra profiles showed enhanced ion yield in cytospin compared to paraffin-embedded samples along with high reproducibility compared to the sample replicate.
Collapse
Affiliation(s)
- Rita Casadonte
- Proteopath GmbH, 54296 Trier, Germany
- Correspondence: (R.C.); (N.A.)
| | - Jörg Kriegsmann
- Proteopath GmbH, 54296 Trier, Germany
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, 3500 Krems, Austria
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, Heidelberg University, 69120 Heidelberg, Germany
| | - Roberta Torcasio
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | | | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
- Correspondence: (R.C.); (N.A.)
| |
Collapse
|
13
|
Claveau JS, Murray DL, Dispenzieri A, Kapoor P, Binder M, Buadi F, Dingli D, Fonder A, Gertz M, Gonsalves W, Hayman S, Hobbs M, Hwa YL, Kourelis T, Lacy M, Leung N, Lin Y, Warsame R, Kyle RA, Rajkumar V, Kumar SK. Value of bone marrow examination in determining response to therapy in patients with multiple myeloma in the context of mass spectrometry-based M-protein assessment. Leukemia 2023; 37:1-4. [PMID: 36482129 DOI: 10.1038/s41375-022-01779-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022]
Affiliation(s)
| | - David L Murray
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Laboratory, Mayo Clinic, Rochester, MN, USA
| | | | | | - Moritz Binder
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Francis Buadi
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - David Dingli
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Amie Fonder
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Morie Gertz
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Miriam Hobbs
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Yi Lisa Hwa
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Martha Lacy
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Nelson Leung
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Yi Lin
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Rahma Warsame
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Robert A Kyle
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Shaji K Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|