1
|
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
|
2
|
Galior KD, Ladwig PM, Snyder MR, Algeciras-Schimnich A, Bornhorst JA, Block DR, Baumann NA, Willrich MAV. Lack of observed interference by therapeutic monoclonal antibodies in select commonly utilized immunoassays. Clin Biochem 2023; 121-122:110685. [PMID: 37972806 DOI: 10.1016/j.clinbiochem.2023.110685] [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: 09/06/2023] [Revised: 10/17/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Therapeutic monoclonal antibodies (tmabs) have been hypothesized to interfere with immunoassay measurements, although studies investigating this potential new class of interference are lacking. This study evaluated the effects of tmabs used in cancers ipilimumab (Bristol Myers Squibb), nivolumab (Bristol Myers Squibb), pembrolizumab (Merck) and autoimmune disorders adalimumab (AbbVie), infliximab (Janssen) and vedolizumab (Takeda) in common immunoassays used in the clinical laboratory. METHODS Residual sera from 10 randomly chosen patients were split into two tubes and spiked with same volume (approximately 5 % final volume) of either saline (control) or 6 tmabs (final concentration of 100 μg/mL each). Concentrations from sixteen analytes in 19 different assays were assessed: TSH (Roche and Beckman), free thyroxine (Roche and Siemens), cortisol (Beckman), Cancer Antigens (CA): CA19-9 (Beckman), CA15-3 (Roche), CA125 (Roche), and CA27.29 (Siemens), carcinoembryonic antigen (Beckman), alpha-fetoprotein (Beckman), thyroglobulin (Beckman) and thyroglobulin antibodies (Beckman), thyroid peroxidase antibody (Beckman), beta-human chorionic gonadotropin (Roche and Beckman), total prostate-specific antigen (Roche), parathyroid hormone (Roche) and antinuclear antibodies IgG (Werfen). The tmab spiked residual sera were compared with matched saline spiked sera and percent error was assessed against allowable total error defined from biological variation or CLIA limits. RESULTS None of the tested immunoassays were affected by the presence of the tmabs, in samples within or outside assay reference intervals. The median % error among all immunoassays ranged between -2.0% (for TSH) to 2.7% (for TPO Ab assay). CONCLUSION These findings demonstrate no detectable tmab interference for the assessed immunoassays using spiked preparations of the tmabs in residual human sera. The findings are limited to the tmabs and immunoassays studied here.
Collapse
Affiliation(s)
- Kornelia D Galior
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA.
| | - Paula M Ladwig
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Melissa R Snyder
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Joshua A Bornhorst
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Darci R Block
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Nikola A Baumann
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
3
|
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
|
4
|
Teuwen JTJ, Ritzen LFL, Knapen-Portz YM, Ludwiczek PK, Damoiseaux JGMC, van Beers JJBC, de Boer D. Identifying therapeutic monoclonal antibodies using target protein collision electrophoresis reflex assay to separate the wheat from the chaff. J Immunol Methods 2023; 522:113552. [PMID: 37652294 DOI: 10.1016/j.jim.2023.113552] [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: 06/13/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Monoclonal gammopathies are characterized by the presence of monoclonal immunoglobulins, also known as M-proteins. Therapeutic monoclonal antibodies (t-mAbs) can interfere in laboratory assays used to monitor the state of disease, such as serum protein electrophoresis (SPE) and immunofixation electrophoresis (IFE). To establish a correct interpretation of IFE, Target protein-Collision Immunofixation Electrophoresis Reflex Assay (T-CIERA) was developed to identify t-mAbs in IFE. Here we demonstrate that T-CIERA is applicable to a wide variety of t-mAbs for which the target protein is commercially available. Moreover, the shift observed was characteristic for each t-mAb, and T-CIERA enabled the identification of multiple t-mAbs sharing a common target protein. Additionally, the lower limit of detection (LLOD) was determined objectively, and T-CIERA demonstrated an adequate LLOD for all tested t-mAbs. Furthermore, T-CIERA was also successfully applied to serum samples obtained from patients receiving daratumumab, isatuximab, elotuzumab, and durvalumab treatment. In conclusion, T-CIERA is a suitable reflex assay for identifying a wide variety of t-mAbs, including those for which no commercial assay is available to deal with their interference. Moreover, CD38-CIERA could serve as an alternative or complementary test to the commercially available Hydrashift assay kits. T-CIERA would enable laboratories without mass spectrometry equipment and expertise in this area to distinguish between drug and disease to improve clinical response monitoring and diagnosis of monoclonal gammopathies.
Collapse
Affiliation(s)
- Jules T J Teuwen
- Central Diagnostic Laboratory, section Protein Chemistry, Maastricht University Medical Center+, PO Box 5800, 6202 AZ Maastricht, the Netherlands.
| | - Lucas F L Ritzen
- Central Diagnostic Laboratory, section Protein Chemistry, Maastricht University Medical Center+, PO Box 5800, 6202 AZ Maastricht, the Netherlands
| | - Yvon M Knapen-Portz
- Central Diagnostic Laboratory, section Protein Chemistry, Maastricht University Medical Center+, PO Box 5800, 6202 AZ Maastricht, the Netherlands.
| | - Patricia K Ludwiczek
- Central Diagnostic Laboratory, section Protein Chemistry, Maastricht University Medical Center+, PO Box 5800, 6202 AZ Maastricht, the Netherlands.
| | - Jan G M C Damoiseaux
- Central Diagnostic Laboratory, section Protein Chemistry, Maastricht University Medical Center+, PO Box 5800, 6202 AZ Maastricht, the Netherlands.
| | - Joyce J B C van Beers
- Central Diagnostic Laboratory, section Protein Chemistry, Maastricht University Medical Center+, PO Box 5800, 6202 AZ Maastricht, the Netherlands.
| | - Douwe de Boer
- Central Diagnostic Laboratory, section Protein Chemistry, Maastricht University Medical Center+, PO Box 5800, 6202 AZ Maastricht, the Netherlands.
| |
Collapse
|
5
|
Manthei DM. Abatacept as a Mimic of Gamma Heavy Chain Disease. Clin Chem 2023:hvad074. [PMID: 37317952 DOI: 10.1093/clinchem/hvad074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/05/2023] [Indexed: 06/16/2023]
Affiliation(s)
- David M Manthei
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, United States
| |
Collapse
|
6
|
Li J, Xu A, Xie W, Li B, Yan C, Xia Y, Liang C, Ji L. MALDI-TOF-MS for rapid screening analysis of M-protein in serum. Front Oncol 2022; 12:1073479. [PMID: 36591514 PMCID: PMC9797962 DOI: 10.3389/fonc.2022.1073479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Monoclonal immunoglobin (M-protein) is a serum biomarker for the diagnosis of plasma cell dyscrasias. Despite limitation of analytical sensitivity and resolution, serum protein electrophoresis and immunofixation electrophoresis are still the front-line tests for the detection of M-proteins. Herein, we developed a MALDI-TOF Mass spectrometry-based method for the screening test of M-proteins in human serum. Based on the unique mass signature of different immunoglobin isotypes, M-Proteins could be rapidly identified and typed. The method demonstrated with high analytical performance and throughput, rapid and simple, which could be a new choice for the diagnosis of plasma cell dyscrasias.
Collapse
Affiliation(s)
- Jie Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China,*Correspondence: Jie Li, ; Chao Liang, ; Ling Ji,
| | - Anping Xu
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Weijie Xie
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Bowen Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Cunliang Yan
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yong Xia
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Chao Liang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China,Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China,State Key Laboratory of Proteomics, National Center for Protein Science (Beijing), Beijing Institute of Lifeomics, Beijing, China,*Correspondence: Jie Li, ; Chao Liang, ; Ling Ji,
| | - Ling Ji
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China,*Correspondence: Jie Li, ; Chao Liang, ; Ling Ji,
| |
Collapse
|
7
|
Baloda V, Shurin MR, Wheeler SE. Pilot Verification of a Novel Approach to Remove Electrophoretic Interference of the Therapeutic Monoclonal Antibody Daratumumab. J Appl Lab Med 2022; 7:910-915. [DOI: 10.1093/jalm/jfab174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/15/2021] [Indexed: 11/14/2022]
Abstract
Abstract
Introduction
The advent of therapeutic monoclonal antibodies (tmAbs) in treatment of multiple myeloma poses unique challenges for the clinical laboratory. These tmAbs may appear as a detectable monoclonal protein by electrophoretic methods resulting in misinterpretation or inability to measure therapeutic responses in some patients, and there are currently limited techniques for identifying interference. In this study we performed a preliminary assessment of the SPIFE anti-daratumumab (SPIFE anti-Dara) reagent to determine whether it would be a feasible aid in resolving the interference of tmAbs with serum protein electrophoresis (SPE) and immunofixation electrophoresis (IFE).
Methods
We performed a pilot study with 20 serum samples and clinical correlates. All samples had a characteristic daratumumab electrophoretic pattern (cathodal IgG/κ). A pre-electrophoretic sample treatment was performed with SPIFE anti-Dara. The reagent is a derivatized anti-Dara that forms multiple antibody/daratumumab complexes. SPE and IFE technical procedures were performed on Helena SPIFE 3000 according to the manufacturer instructions.
Results
Of the 20 patients, 14 patients were identified to be on daratumumab therapy. In 14/14 of cases, the daratumumab interference was successfully removed both from SPE and IFE assays. Disease associated M-protein was still visible after pretreatment, and quantification of M-protein may be possible with the use of SPIFE anti-Dara procedure.
Discussion
SPIFE anti-Dara is a promising method to remove the interference of therapeutic monoclonal antibody daratumumab with SPE and IFE results in clinical laboratories and warrants further assessment.
Collapse
Affiliation(s)
- Vandana Baloda
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Michael R Shurin
- Departments of Immunology and Pathology, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sarah E Wheeler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
8
|
Fatica EM, Martinez M, Ladwig PM, Murray JD, Kohlhagen MC, Kyle RA, Kourelis T, Lust JA, Snyder MR, Dispenzieri A, Murray DL, Willrich MAV. MALDI-TOF mass spectrometry can distinguish immunofixation bands of the same isotype as monoclonal or biclonal proteins. Clin Biochem 2021; 97:67-73. [PMID: 34384797 DOI: 10.1016/j.clinbiochem.2021.08.001] [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: 05/14/2021] [Revised: 07/15/2021] [Accepted: 08/05/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Plasma cell disorders (PCDs) are typically characterized by excessive production of a single immunoglobulin, defined as a monoclonal protein (M-protein). Some patients have more than one identifiable M-protein, termed biclonal. Traditional immunofixation electrophoresis (IFE) cannot distinguish if two bands of the same isotype represent biclonal proteins or M-proteins with some other feature. A novel assay using immunoenrichment coupled to matrix-assisted laser desorption ionization time-of-flight mass-spectrometry (Mass-Fix) was applied to determine whether two bands of the same isotype represented (1) monomers and dimers of a single M-protein, (2) an M-protein plus a therapeutic monoclonal antibody (t-mAb), (3) an M-protein with light chain glycosylation, or (4) two distinct biclonal M-proteins. METHODS Patient samples with two bands of the same isotype identified by IFE were enriched using nanobodies against IgG, IgA, IgM, or κ and λ light chains then analyzed by Mass-Fix. Light chain masses were used to differentiate IgGκ M-proteins from t-mAbs. Mass differences between peaks were calculated to identify N-glycosylation or matrix adducts. High-resolution mass spectrometry was used as a comparator method in a subset of samples. RESULTS Eighty-one residual samples were collected. For IgA, 93% (n = 25) were identified as monoclonal. For IgG, 67% (n = 24) were monoclonal, and 33% (n = 12) were truly biclonal. Among the monoclonal IgGs, the second band represented a glycosylated form for 21% (n = 5), while 33% (n = 8) had masses consistent with a t-mAb. 44% (n = 8) of IgM samples were biclonal, and 56% (n = 10) were monoclonal, of which one was glycosylated. CONCLUSIONS We demonstrate the utility of mass spectrometry in the characterization of multiple IFE bands of the same isotype. Improved reporting accuracy of M-proteins is useful for monitoring of patients with PCDs.
Collapse
Affiliation(s)
- Erica M Fatica
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Mark Martinez
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Paula M Ladwig
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Josiah D Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Mindy C Kohlhagen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Robert A Kyle
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - Taxiarchis Kourelis
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States; Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - John A Lust
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States; Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - Melissa R Snyder
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Angela Dispenzieri
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States; Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - David L Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Maria A V Willrich
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
9
|
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
|
10
|
Liu L, Shurin MR, Wheeler SE. A novel approach to remove interference of therapeutic monoclonal antibody with serum protein electrophoresis. Clin Biochem 2020; 75:40-47. [PMID: 31669513 PMCID: PMC6928417 DOI: 10.1016/j.clinbiochem.2019.10.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/03/2019] [Accepted: 10/14/2019] [Indexed: 01/29/2023]
Abstract
OBJECTIVES Multiple myeloma (MM) is characterized by malignant growth of plasma cells, usually producing a monoclonal antibody (mAb). New treatments for MM include therapeutic monoclonal antibodies (tmAbs), but patients treated with tmAb demonstrate interference on serum electrophoresis (SPE) and immunoprecipitation electrophoresis (IEP). Evaluation of treatment efficacy and determination of MM remission include SPE and IEP which identifies mAb, but cannot differentiate between disease associated mAb and tmAb. We hypothesized that tmAb could be removed from patient sera before testing by SPE and IEP to provide accurate diagnoses for clinicians. DESIGN AND METHODS We developed the Antigen Specific therapeutic monoclonal Antibody Depletion Assay (ASADA), that utilizes magnetic beads coated with the cognate antigen of the tmAbs, to deplete two different tmAb (daratumumab, elotuzumab) from saline and patient sera and assessed for complete removal of tmAb by SPE and IEP. RESULTS We found that tmAb could be efficiently removed from saline and patient sera. ASADA demonstrated acceptable analytical specificity and sensitivity in IEP. Recovery of appropriate quantitative values by SPE was demonstrated with clinically acceptable precision. A single bead cocktail could be used to treat both daratumumab and elotuzumab. CONCLUSIONS This demonstrates proof of principle that ASADA can be used to remove current and future tmAb from patient sera, regardless of platform. This research provides for accurate diagnosis, disease monitoring, and remission status in MM patients being treated with tmAb.
Collapse
Affiliation(s)
- Li Liu
- University of Pittsburgh Medical Center, Department of Pathology, Clinical Laboratory Building, 3477 Euler Way, Pittsburgh, PA 15213, United States.
| | - Michael R Shurin
- University of Pittsburgh Medical Center, Department of Pathology, Clinical Laboratory Building, 3477 Euler Way, Pittsburgh, PA 15213, United States; University of Pittsburgh, Departments of Pathology and Immunology, Clinical Laboratory Building, 3477 Euler Way, Pittsburgh, PA 15213, United States.
| | - Sarah E Wheeler
- University of Pittsburgh Medical Center, Department of Pathology, Clinical Laboratory Building, 3477 Euler Way, Pittsburgh, PA 15213, United States; University of Pittsburgh, Department of Pathology, Clinical Laboratory Building, 3477 Euler Way, Pittsburgh, PA 15213, United States.
| |
Collapse
|