1
|
Ianosi-Irimie MR, Willrich MAV, Edmonston TB. Patient with a New Small IgG Kappa Monoclonal Protein. Clin Chem 2025; 71:430-431. [PMID: 40037406 DOI: 10.1093/clinchem/hvae152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/23/2024] [Indexed: 03/06/2025]
Affiliation(s)
- Monica R Ianosi-Irimie
- Department of Pathology and Laboratory Services, Cooper University Hospital, Camden, NJ, United States
- Cooper Medical School at Rowan University, Camden, NJ, United States
| | - Maria Alice V Willrich
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Tina B Edmonston
- Department of Pathology and Laboratory Services, Cooper University Hospital, Camden, NJ, United States
- Cooper Medical School at Rowan University, Camden, NJ, United States
| |
Collapse
|
2
|
Barnidge D, Sakrikar D, Kubicki T, Derman BA, Jakubowiak AJ, Lakos G. Distinguishing Daratumumab from Endogenous Monoclonal Proteins in Serum from Multiple Myeloma Patients Using an Automated Mass Spectrometry System. J Appl Lab Med 2025; 10:235-249. [PMID: 39699179 DOI: 10.1093/jalm/jfae142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/21/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Therapeutic monoclonal antibodies (t-mAbs) may interfere with electrophoresis-based methods used to monitor multiple myeloma (MM), which can create inaccurate results. Matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry is an alternative to gels distinguishing between endogenous M-proteins and t-mAbs based on molecular mass. METHODS Serum samples (n = 109) from 34 MM patients receiving Dara-KRd were collected 14 or 28 days postdaratumumab administration. Samples were analyzed using the EXENT® Analyzer that combines automated immunopurification and MALDI-TOF MS for the isotyping and quantification of monoclonal immunoglobulins. RESULTS Daratumumab was identified in 103 out of 109 samples (94.5%). In all IgGλ (n = 8), IgAκ (n = 8), and IgAλ (n = 2) patients, the M-protein and daratumumab were detected. Of the IgGκ patients (n = 18), 5 patients had a total of 6 samples where the M-protein was detected but daratumumab was not. There was no difference in the detection rate of daratumumab between samples taken 14 and 28 days postadministration with the median daratumumab concentration being 0.95 and 0.54 g/L, respectively. A precision study was also performed on 25 replicates containing 1 g/L daratumumab in serum where a coefficient of variation of 4.2% was observed as determined by the EXENT Analyzer. CONCLUSIONS The Immunoglobulin Isotypes (GAM: IgG, IgA, and IgM) for the EXENT Analyzer detected and distinguished a daratumumab kappa light chain peak from an M-protein light chain peak in MM patient serum when resolved by the mass spectrometer.
Collapse
Affiliation(s)
- David Barnidge
- The Binding Site, part of Thermo Fisher Scientific, Rochester, MN, United States
| | - Dhananjay Sakrikar
- The Binding Site, part of Thermo Fisher Scientific, Rochester, MN, United States
| | - Tadeusz Kubicki
- Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - Benjamin A Derman
- Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | | | - Gabriella Lakos
- The Binding Site, part of Thermo Fisher Scientific, Rochester, MN, United States
| |
Collapse
|
3
|
Murray DL, Willrich MAV. Applications of Mass Spectrometry Proteomic Methods to Immunoglobulins in the Clinical Laboratory. Clin Chem 2024; 70:1422-1435. [PMID: 39667030 DOI: 10.1093/clinchem/hvae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/16/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Immunoglobulin (Ig) measurements in the clinical laboratory have been traditionally performed by nephelometry, turbidimetry, electrophoresis, and ELISA assays. Mass spectrometry (MS) measurements have the potential to provide deeper insights on the nature of these markers. CONTENT Different approaches-top-down, middle-down, or bottom-up-have been described for measuring specific Igs for endogenous monoclonal immunoglobulins (M-proteins) and exogenous therapeutic monoclonal antibody therapies (t-mAbs). Challenges arise in distinguishing the Ig of interest from the polyclonal Ig background. MS is emerging as a practical method to provide quantitative analysis and information about structural and clonal features that are not easily determined by current clinical laboratory methods. This review discusses clinically implemented examples, including isotyping and quantification of M-proteins and quantitation of t-mAbs within the polyclonal Ig background, as examples of how MS can enhance our detection and characterization of Igs. SUMMARY This review of current clinically available MS proteomic tests for Igs highlights both analytical and nonanalytical challenges for implementation. Given the new insight into Igs from these methods, it is hoped that vendors, laboratorians, healthcare providers, and payment systems can work to overcome these challenges and advance the care of patients.
Collapse
Affiliation(s)
- David L Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Maria A V Willrich
- Department of Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| |
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
|
Kubicki T, Derman BA, Dytfeld D, Jakubowiak AJ. Measurable residual disease in peripheral blood in myeloma: dream or reality. Curr Opin Oncol 2023; 35:574-580. [PMID: 37621165 DOI: 10.1097/cco.0000000000000987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
PURPOSE OF REVIEW Therapeutic advancements in multiple myeloma have led to increasingly deeper and more durable responses, creating a need for highly sensitive and applicable techniques for measurable residual disease (MRD) assessment. Bone marrow assays can deeply assess for MRD, but it is not conducive to performing frequent and dynamic evaluations, which may be needed for MRD-adapted treatment approaches. Recently, numerous techniques for MRD assessment in peripheral blood have come under investigation, and their integration into routine clinical practice is eagerly anticipated. RECENT FINDINGS The identification of circulating tumor cells (CTCs), evaluation of cell-free DNA, and measuring monoclonal protein concentration with mass spectrometry are promising research areas for assessing myeloma in peripheral blood. CTCs assessment and cell-free DNA may carry prognostic significance, but they lack the sensitivity of bone marrow-based techniques. Mass spectrometry has already been implemented in clinical practice in certain centers, but its full potential has yet to be fully realized. This review focuses on recent developments in these fields, emphasizing the potential future roles of these assessments. SUMMARY MRD assessment in peripheral blood is still in the development stage but holds promise for not only complementing bone marrow based evaluations but also potential for improving sensitivity.
Collapse
|
6
|
Malik S, Toong C. Monoclonal antibody interference in electrophoresis. Pathology 2023; 55:163-165. [PMID: 36210228 DOI: 10.1016/j.pathol.2022.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/22/2022] [Accepted: 05/22/2022] [Indexed: 01/24/2023]
Affiliation(s)
- Sameer Malik
- Department of Immunology, NSW Health Pathology, Liverpool, NSW, Australia; Department of Immunology, Liverpool Hospital, Liverpool, NSW, Australia.
| | - Catherine Toong
- Department of Immunology, NSW Health Pathology, Liverpool, NSW, Australia; Department of Immunology, Liverpool Hospital, Liverpool, NSW, Australia; University of NSW, Sydney, NSW, Australia
| |
Collapse
|
7
|
Baloda V, McCreary EK, Goscicki BK, Shurin MR, Wheeler SE. Tixagevimab Plus Cilgavimab Does Not Affect the Interpretation of Electrophoretic and Free Light Chain Assays. Am J Clin Pathol 2023; 159:10-13. [PMID: 36469049 DOI: 10.1093/ajcp/aqac137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/06/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES There is concern that the anti-severe acute respiratory syndrome coronavirus 2 therapeutic monoclonal antibodies, used as preexposure prophylaxis in patients with multiple myeloma, may appear as a detectable monoclonal protein by electrophoretic methods, resulting in misinterpretation or inability to measure therapeutic responses in some patients. In this pilot study, we characterize the effect of tixagevimab plus cilgavimab (Evusheld; T + C) on interpretation of serum protein electrophoresis (SPE), immunofixation electrophoresis (IFE), and serum free light chain (sFLC) assays. METHODS We performed spiking experiments with T + C at serum maximum concentration following a 300-mg dose (1× Cmax) and at 10 times the concentration of Cmax (10× Cmax) with pooled serum samples. SPE and IFE technical procedures were performed on the SPIFE 3000, and sFLC and immunoglobulin G1 (IgG1) subtype quantitation was performed on the Optilite. RESULTS T + C-associated interference was not visible as an M-spike in normogammaglobulinemic pooled samples. Hypogammaglobulemic pooled samples at 10× Cmax demonstrated an M-spike in SPE and immunoglobulin Gκ pattern in IFE. No increases were noted in the results of sFLC or IgG1 levels. CONCLUSIONS This study indicates that T + C at pharmacologic Cmax is unlikely to interfere with SPE, IFE, sFLC, or IgG1 analyses when spiked into patient serum samples, but further evaluation of recently injected patients may be warranted.
Collapse
Affiliation(s)
- Vandana Baloda
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Erin K McCreary
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Breana K Goscicki
- Department of Pharmacy, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Michael R Shurin
- Departments of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.,Departments of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah E Wheeler
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.,Departments of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
8
|
Aydin O, Aykas F. Therapeutic Monoclonal Antibody Interference in Monoclonal Gammopathy Monitoring: a Denosumab Experience. Lab Med 2022; 54:e95-e97. [PMID: 36221992 DOI: 10.1093/labmed/lmac129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Abstract
A 73-year-old woman was diagnosed with a lambda light chain myeloma. A follow-up immunofixation electrophoresis showed a monoclonal immunoglobulin (Ig)G kappa in addition to the regular lambda band. A monoclonal antibody therapy interference was suspected but her VRD (bortezomib, lenalidomide, dexamethasone) regimen did not include such a medication. Later it was learned that she was prescribed denosumab, a monoclonal human antibody agent to treat bone lesions. The IgG kappa band disappeared 7 months after the first and 4 months after the last dose of denosumab, confirming a case of interference. This case once again emphasizes the importance of delta check and close communication between clinicians to avoid a false result in electrophoresis. It also describes the migration pattern of denosumab. As therapeutic antibodies gain approval and enter into common clinical practice, drug interference will complicate electrophoresis testing in diagnosis and patient follow-up.
Collapse
Affiliation(s)
- Ozgur Aydin
- Antalya Research and Education Hospital, Central Laboratory , Antalya , Turkey
| | - Fatma Aykas
- Antalya Research and Education Hospital, Hematology Clinic , Antalya , Turkey
| |
Collapse
|
9
|
Murray DL. Bringing mass spectrometry into the care of patients with multiple myeloma. Int J Hematol 2022; 115:790-798. [PMID: 35471500 DOI: 10.1007/s12185-022-03364-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/19/2022] [Accepted: 04/19/2022] [Indexed: 10/18/2022]
Abstract
Serum protein electrophoresis methods are widely employed to detect, quantify and isotype M-proteins for multiple myeloma patients. Increasing clinical demands to detect residual disease and interferences from new therapeutic monoclonal antibody treatments have stretched electrophoretic methods to their analytical limits. Newer techniques to detect M-proteins using mass spectrometry (MS) are emerging with improved clinical and analytical performance. These techniques are beginning to gain traction within the routine clinical lab testing. This review describes these MS methods with attention to the current and future roles such testing could play in the care of multiple myeloma patients.
Collapse
Affiliation(s)
- David L Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55906, USA.
| |
Collapse
|
10
|
Dasari S, Kohlhagen MC, Dispenzieri A, Willrich MAV, Snyder MR, Kourelis TV, Lust JA, Mills JR, Kyle RA, Murray DL. Detection of Plasma Cell Disorders by Mass Spectrometry: A Comprehensive Review of 19,523 Cases. Mayo Clin Proc 2022; 97:294-307. [PMID: 34887112 DOI: 10.1016/j.mayocp.2021.07.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 01/14/2023]
Abstract
OBJECTIVES To verify the analytical performance of a new mass spectrometry-based method, termed MASS-FIX, when screening for plasma cell disorders in a routine clinical laboratory. PATIENTS AND METHODS Results from 19,523 unique patients tested for an M-protein between July 24, 2018, and March 6, 2020, by a combination serum protein electrophoresis (SPEP) and MASS-FIX were examined for consistency with pretest implementation performance. MASS-FIX's ability to verify abnormal results from SPEP and free light chain measurements was then compared with that of immunofixation electrophoresis (IFE) using a separate cohort of 52,586 patients tested by SPEP/IFE during the same period. RESULTS Overall, 62.4% of our cohort was negative for an M-protein. Importantly, 7.3% of all specimens had an M spike on SPEP (0.1 to 8.5 g/dL) and MASS-FIX detected an M-protein in all these samples. Of all samples, 30.3% had M-proteins that were detected by MASS-FIX but the SPEP finding was too small for quantification. Of the positive samples, 5.7% contained a therapeutic monoclonal antibody. Of the positive samples, 4.1% had an N-glycosylated light chain (biomarker of high-risk plasma cell disorders). MASS-FIX confirmed a higher percentage of SPEP abnormalities than IFE. MASS-FIX was slightly more sensitive than IFE when confirming an M-protein in samples with an abnormal free light chain ratio. MASS-FIX had a very low sample repeat rate (1.5%). MASS-FIX was highly automatable resulting in a higher number of samples/technologist/day than IFE (∼30% more). CONCLUSION Overall, MASS-FIX was successful in maintaining validation characteristics. MASS-FIX was more sensitive in confirming SPEP abnormalities when compared with IFE. Ability to detect therapeutic monoclonal antibodies and glycosylated light chains was distinctly advantageous.
Collapse
Affiliation(s)
- Surendra Dasari
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Mindy C Kohlhagen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Angela Dispenzieri
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Department of Medicine, Mayo Clinic, Rochester, MN
| | - Maria A V Willrich
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Melissa R Snyder
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - John A Lust
- Department of Medicine, Mayo Clinic, Rochester, MN
| | - John R Mills
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - David L Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN.
| |
Collapse
|
11
|
Monoclonal gammopathy detection and current technologies. Cancer Biomark 2022. [DOI: 10.1016/b978-0-12-824302-2.00005-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
12
|
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: 7] [Impact Index Per Article: 1.8] [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
|
13
|
Lengfeld J, Zhang H, Stoesz S, Murali R, Pass F, Greene MI, Goel PN, Grover P. Challenges in Detection of Serum Oncoprotein: Relevance to Breast Cancer Diagnostics. BREAST CANCER-TARGETS AND THERAPY 2021; 13:575-593. [PMID: 34703307 PMCID: PMC8524259 DOI: 10.2147/bctt.s331844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/02/2021] [Indexed: 11/23/2022]
Abstract
Breast cancer is a highly prevalent malignancy that shows improved outcomes with earlier diagnosis. Current screening and monitoring methods have improved survival rates, but the limitations of these approaches have led to the investigation of biomarker evaluation to improve early diagnosis and treatment monitoring. The enzyme-linked immunosorbent assay (ELISA) is a specific and robust technique ideally suited for the quantification of protein biomarkers from blood or its constituents. The continued clinical relevancy of this assay format will require overcoming specific technical challenges, including the ultra-sensitive detection of trace biomarkers and the circumventing of potential assay interference due to the expanding use of monoclonal antibody (mAb) therapeutics. Approaches to increasing the sensitivity of ELISA have been numerous and include employing more sensitive substrates, combining ELISA with the polymerase chain reaction (PCR), and incorporating nanoparticles as shuttles for detection antibodies and enzymes. These modifications have resulted in substantial boosts in the ability to detect extremely low levels of protein biomarkers, with some systems reliably detecting antigen at sub-femtomolar concentrations. Extensive utilization of mAb therapies in oncology has presented an additional contemporary challenge for ELISA, particularly when both therapeutic and assay antibodies target the same protein antigen. Resolution of issues such as epitope overlap and steric hindrance requires a rational approach to the design of diagnostic antibodies that takes advantage of modern antibody generation pipelines, epitope binning techniques and computational methods to strategically target biomarker epitopes. This review discusses technical strategies in ELISA implemented to date and their feasibility to address current constraints on sensitivity and problems with interference in the clinical setting. The impact of these recent advancements will depend upon their transformation from research laboratory protocols into facile, reliable detection systems that can ideally be replicated in point-of-care devices to maximize utilization and transform both the diagnostic and therapeutic monitoring landscape.
Collapse
Affiliation(s)
- Justin Lengfeld
- Martell Diagnostic Laboratories, Inc., Roseville, MN, 55113, USA
| | - Hongtao Zhang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Steven Stoesz
- Martell Diagnostic Laboratories, Inc., Roseville, MN, 55113, USA
| | - Ramachandran Murali
- Department of Biomedical Sciences, Research Division of Immunology; Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Franklin Pass
- Martell Diagnostic Laboratories, Inc., Roseville, MN, 55113, USA
| | - Mark I Greene
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Peeyush N Goel
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Payal Grover
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
14
|
Xu S, Liu Y, Wen L, Zhao L, Deng X, Rong R, Lu J. [Application of Hydrashift 2/4 daratumumab assay in eliminating interference of daratumumab on serum immunofixation electrophoresis]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 42:840-845. [PMID: 34788924 PMCID: PMC8607023 DOI: 10.3760/cma.j.issn.0253-2727.2021.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Indexed: 11/05/2022]
Abstract
Objective: To investigate the interference of daratumumab on immunofixation electrophoresis after treating plasma cell diseases and methods to eliminate the interference. Methods: Serum samples of eight patients with plasma cell diseases treated with daratumumab in Peking University People's Hospital from April 2020 to March 2021 were collected for standard immunofixation electrophoresis and Hydrashift 2/4 daratumumab assay. Results: After treatment, 81.3% (13/16) of the samples showed drug-induced monoclonal antibodies (IgG-κ) . The samples without drug-induced monoclonal bands were related to individual differences, administration intervals, and immunoglobulin levels. Among the samples with IgG-κ monoclonal bands, 76.9% (10/13) could be directly identified as endogenous or exogenous monoclonal bands by immunofixation electrophoresis, and the others (3/13) could be identified by Hydrashift 2/4 daratumumab assay. Conclusion: Hydrashift 2/4 daratumumab assay can remove the band of daratumumab on the immunofixation electrophoresis and help with efficacy evaluation.
Collapse
Affiliation(s)
- S Xu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing 100044, China
| | - Y Liu
- Department of Hematology, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| | - L Wen
- Department of Hematology, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| | - L Zhao
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing 100044, China
| | - X Deng
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing 100044, China
| | - R Rong
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing 100044, China
| | - J Lu
- Department of Hematology, Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China Center for Collaborative Innovation in Hematology, Suzhou 215006,China
| |
Collapse
|
15
|
Huls F, Schroeder L, Keren DF. Expression of Daratumumab and Elotuzumab Migration by Capillary Electrophoresis Relative to Transferrin Improves Precision of Their Identification. J Appl Lab Med 2021; 5:419-422. [PMID: 32445376 DOI: 10.1093/jalm/jfz008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 11/14/2022]
Affiliation(s)
- Forest Huls
- Division of General Internal Medicine, Department of Medicine, the University of Alabama, Vestavia Hills, AL
| | - Lee Schroeder
- Department of Pathology, the University of Michigan, Ann Arbor, MI
| | - David F Keren
- Department of Pathology, the University of Michigan, Ann Arbor, MI
| |
Collapse
|
16
|
Liu L, Wertz WJ, Kondisko A, Shurin MR, Wheeler SE. Incidence and Management of Therapeutic Monoclonal Antibody Interference in Monoclonal Gammopathy Monitoring. J Appl Lab Med 2021; 5:29-40. [PMID: 32445341 DOI: 10.1373/jalm.2019.029009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 05/28/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND The treatment of multiple myeloma (MM) has been revolutionized by the introduction of therapeutic monoclonal antibodies (tmAbs). Daratumumab, a human IgG1/κ tmAb against CD38 on plasma cells, has improved overall survival in refractory MM and was recently approved as a frontline therapy for MM. Work on tmAb interference with serum protein electrophoresis (SPE) during MM monitoring has failed to provide information for laboratories on incidence of interference and effective methods of managing the interference at a practicable level. We aimed to evaluate daratumumab and elotuzumab interference in a large academic hospital setting and implement immediate solutions. METHODS We identified and chart reviewed all cases of possible daratumumab interference by electrophoretic pattern (120 of 1317 total cases over 3 months). We retrospectively reviewed SPE cases in our laboratory to assess clinical implications of tmAb interference before the laboratory was aware of tmAb treatment. We supplemented samples with daratumumab and elotuzumab to determine the limits of detection and run free light chain analysis. RESULTS Approximately 9% (120 of 1317) of tested cases have an SPE and/or immunofixation electrophoresis (IFE) pattern consistent with daratumumab, but only approximately 47% (56) of these cases were associated with daratumumab therapy. Presence of daratumumab led to physician misinterpretation of SPE/IFE results. Limits of daratumumab detection varied with total serum gammaglobulin concentrations, but serum free light chain analysis was unaffected. CONCLUSIONS Clinical laboratories currently rely on interference identification by electrophoretic pattern, which may be insufficient and is inefficient. Critical tools in preventing misinterpretation efficiently include physician education, pharmacy notifications, separate order codes, and interpretive comments.
Collapse
Affiliation(s)
- Li Liu
- Department of Pathology, Division of Clinical Immunopathology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - William J Wertz
- Department of Enterprise Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Anthony Kondisko
- Department of Enterprise Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Michael R Shurin
- Department of Pathology, Division of Clinical Immunopathology, University of Pittsburgh Medical Center, Pittsburgh, PA.,Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA.,Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Sarah E Wheeler
- Department of Pathology, Division of Clinical Immunopathology, University of Pittsburgh Medical Center, Pittsburgh, PA.,Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA
| |
Collapse
|
17
|
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
|
18
|
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: 12] [Impact Index Per Article: 3.0] [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
|
19
|
Gastélum-Cano JM, Fragoso-Flores J, Noffal-Nuño VM, Deffis-Court M. An unusual pattern in serum protein electrophoresis to take in mind: A case report. Pract Lab Med 2021; 24:e00200. [PMID: 33490350 PMCID: PMC7810760 DOI: 10.1016/j.plabm.2020.e00200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 12/28/2020] [Indexed: 01/21/2023] Open
Abstract
Here we described a case of an asymptomatic 73 years-old female patient in geriatric routine consultation, whose laboratory testing showed hyperproteinemia with accompanying hyperglobulinemia. A diagnosis of BGUS was made only after a correlation among SPEP, densitometry tracing and IFE results was established, evidencing a second peak, that was less evident and not reported at first. These biclonal conditions are of very low incidence in the clinical laboratory, requiring the laboratory professional to have particular skills for their identification. As far as is known, clinical findings in BGUS are similar to those found in MGUS. However, they remain not well understood. Therefore, for an accurate diagnosis of BGUS, the clinical laboratory technician must be trained and sensitized to detect a second M - protein as a band or peak; taking in mind the possible different scenarios in heavy and light chain typing.
Collapse
Affiliation(s)
- José María Gastélum-Cano
- Laboratorios Ruiz, Mártires Del 2 de Octubre 808, Anzures, 72530, Puebla, Mexico
- Universidad Popular Autónoma Del Estado de Puebla, 21 Sur 1103, Barrio Santiago, 72410, Puebla, Mexico
| | - Jaime Fragoso-Flores
- Laboratorios Ruiz, Mártires Del 2 de Octubre 808, Anzures, 72530, Puebla, Mexico
- Universidad Popular Autónoma Del Estado de Puebla, 21 Sur 1103, Barrio Santiago, 72410, Puebla, Mexico
| | | | - Marcela Deffis-Court
- Hospital Médica Sur, Puente de Piedra 150, Toriello Guerra, 14050, Ciudad de México, Mexico
| |
Collapse
|
20
|
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: 0.8] [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
|
21
|
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: 103] [Impact Index Per Article: 25.8] [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
|
22
|
Evolving Role of Daratumumab: From Backbencher to Frontline Agent. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:572-587. [DOI: 10.1016/j.clml.2020.03.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/08/2020] [Accepted: 03/19/2020] [Indexed: 12/11/2022]
|
23
|
Kohlhagen M, Dasari S, Willrich M, Hetrick M, Netzel B, Dispenzieri A, Murray DL. Automation and validation of a MALDI-TOF MS (Mass-Fix) replacement of immunofixation electrophoresis in the clinical lab. Clin Chem Lab Med 2020; 59:155-163. [PMID: 32745067 DOI: 10.1515/cclm-2020-0581] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/03/2020] [Indexed: 12/14/2022]
Abstract
Objectives A matrix assisted laser desorption ionization time of flight mass spectrometry (MALDI-TOF MS) method (Mass-Fix) as a replacement for gel-based immunofixation (IFE) has been recently described. To utilize Mass-Fix clinically, a validated automated method was required. Our aim was to automate the pre-analytical processing, improve positive specimen identification and ergonomics, reduce paper data storage and increase resource utilization without increasing turnaround time. Methods Serum samples were batched and loaded onto a liquid handler along with reagents and a barcoded sample plate. The pre-analytical steps included: (1) Plating immunopurification beads. (2) Adding 10 μl of serum. (3) Bead washing. (4) Eluting the immunoglobulins (Igs), and reducing to separate the heavy and light Ig chains. The resulting plate was transferred to a second low-volume liquid handler for MALDI plate spotting. MALDI-TOF mass spectra were collected. Integrated in-house developed software was utilized for sample tracking, driving data acquisition, data analysis, history tracking, and result reporting. A total of 1,029 residual serum samples were run using the automated system and results were compared to prior electrophoretic results. Results The automated Mass-Fix method was capable of meeting the validation requirements of concordance with IFE, limit of detection (LOD), sample stability and reproducibility with a low repeat rate. Automation and integrated software allowed a single user to process 320 samples in an 8 h shift. Software display facilitated identification of monoclonal proteins. Additionally, the process maintains positive specimen identification, reduces manual pipetting, allows for paper free tracking, and does not significantly impact turnaround time (TAT). Conclusions Mass-Fix is ready for implementation in a high-throughput clinical laboratory.
Collapse
Affiliation(s)
- Mindy Kohlhagen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Surendra Dasari
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Maria Willrich
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - MeLea Hetrick
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Brian Netzel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Angela Dispenzieri
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - David L Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
24
|
Shah N, Aiello J, Avigan DE, Berdeja JG, Borrello IM, Chari A, Cohen AD, Ganapathi K, Gray L, Green D, Krishnan A, Lin Y, Manasanch E, Munshi NC, Nooka AK, Rapoport AP, Smith EL, Vij R, Dhodapkar M. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of multiple myeloma. J Immunother Cancer 2020; 8:e000734. [PMID: 32661116 PMCID: PMC7359060 DOI: 10.1136/jitc-2020-000734] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2020] [Indexed: 12/24/2022] Open
Abstract
Outcomes in multiple myeloma (MM) have improved dramatically in the last two decades with the advent of novel therapies including immunomodulatory agents (IMiDs), proteasome inhibitors and monoclonal antibodies. In recent years, immunotherapy for the treatment of MM has advanced rapidly, with the approval of new targeted agents and monoclonal antibodies directed against myeloma cell-surface antigens, as well as maturing data from late stage trials of chimeric antigen receptor CAR T cells. Therapies that engage the immune system to treat myeloma offer significant clinical benefits with durable responses and manageable toxicity profiles, however, the appropriate use of these immunotherapy agents can present unique challenges for practicing physicians. Therefore, the Society for Immunotherapy of Cancer convened an expert panel, which met to consider the current role of approved and emerging immunotherapy agents in MM and provide guidance to the oncology community by developing consensus recommendations. As immunotherapy evolves as a therapeutic option for the treatment of MM, these guidelines will be updated.
Collapse
Affiliation(s)
- Nina Shah
- Division of Hematology-Oncology, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jack Aiello
- Patient Empowerment Network, San Jose, California, USA
| | - David E Avigan
- Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Jesus G Berdeja
- Department of Medicine, Sarah Cannon Research Institute, Nashville, Tennessee, USA
| | - Ivan M Borrello
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center of Johns Hopkins, Baltimore, Maryland, USA
| | - Ajai Chari
- Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Adam D Cohen
- Department of Medicine, Abramson Cancer Center at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Karthik Ganapathi
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Lissa Gray
- University of California San Francisco, San Francisco, CA, USA
| | - Damian Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Amrita Krishnan
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Multiple Myeloma Center for Clinical Research, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Yi Lin
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Elisabet Manasanch
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nikhil C Munshi
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Ajay K Nooka
- Department of Hematology/Oncology, Emory University, Atlanta, Georgia, USA
| | - Aaron P Rapoport
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Eric L Smith
- Myeloma Service and Cellular Therapeutics Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ravi Vij
- Division of Medical Oncology, Siteman Cancer Center, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Madhav Dhodapkar
- School of Medicine, Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
25
|
Moore LM, Cho S, Thoren KL. MALDI-TOF mass spectrometry distinguishes daratumumab from M-proteins. Clin Chim Acta 2019; 492:91-94. [PMID: 30790545 PMCID: PMC6524149 DOI: 10.1016/j.cca.2019.02.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/08/2019] [Accepted: 02/17/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Daratumumab, a therapeutic IgG kappa monoclonal antibody, can cause a false positive interference on electrophoretic assays that are routinely used to monitor patients with monoclonal gammopathies. In this study, we evaluate the ability of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) to distinguish daratumumab from disease-related IgG kappa monoclonal proteins (M-protein). METHODS Waste clinical samples from 31 patients who were receiving daratumumab and had a history of IgG kappa monoclonal gammopathy were collected. Immunoglobulins were purified from serum and analyzed by MALDI-TOF MS. Mass spectra were assessed for the presence of distinct monoclonal proteins. For samples in which only one monoclonal peak was identified near the expected m/z of daratumumab, the Hydrashift 2/4 Daratumumab Assay was used to confirm the presence of an M-protein. RESULTS Using MALDI-TOF MS, daratumumab could be distinguished from M-proteins in 26 out of 31 samples (84%). Results from 2 samples were inconclusive since the M-protein was not detected by the Hydrashift assay and may also be undetectable by MALDI-TOF MS. Comparatively, daratumumab was distinguishable from M-proteins in 14 out of 31 samples (45%) by immunofixation. CONCLUSIONS MALDI-TOF MS offers greater specificity compared to immunofixation for distinguishing daratumumab from M-proteins.
Collapse
Affiliation(s)
- Lauren M Moore
- Clinical Chemistry Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, 327 E. 64th St., New York, NY 10065, United States of America
| | - Sun Cho
- Clinical Chemistry Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, 327 E. 64th St., New York, NY 10065, United States of America
| | - Katie L Thoren
- Clinical Chemistry Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, 327 E. 64th St., New York, NY 10065, United States of America.
| |
Collapse
|
26
|
Lázár-Molnár E, Delgado JC. Implications of Monoclonal Antibody Therapeutics Use for Clinical Laboratory Testing. Clin Chem 2019; 65:393-405. [DOI: 10.1373/clinchem.2016.266973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/10/2018] [Indexed: 12/15/2022]
Abstract
Abstract
BACKGROUND
Monoclonal antibody therapeutics (MATs) represent a rapidly expanding class of biological drugs used to treat a variety of diseases. The widespread use of MATs increasingly affects clinical laboratory medicine.
CONTENT
This review provides an overview of MATs currently approved for clinical use in the US, starting from basic biology of antibodies to the engineering, pharmacokinetic and pharmacodynamic properties, nomenclature, and production of MATs. Immunogenicity and the production of antidrug antibodies (ADAs) play a major role in loss of therapeutic response and the development of treatment failure to certain MATs. Laboratory-based monitoring for MATs and detection of ADAs represent emerging needs for optimizing the use of MATs to achieve the best outcomes at affordable cost. In addition, the increased use of MATs affects clinical laboratory testing by interference of MATs with clinical laboratory tests across different areas of laboratory medicine, including histocompatibility, blood bank, and monoclonal protein testing.
SUMMARY
The number of MATs is rapidly growing each year to address previously unmet clinical needs. Laboratory monitoring of MATs and detecting ADAs represent expanding areas of laboratory testing. Test-based strategies allow for treatment optimization at the level of the individual patient, thus providing a personalized medicine approach. In addition, clinical laboratories must be aware that the increasing use of MATs affects laboratory testing and be ready to implement methods to eliminate or mitigate interference with clinical tests.
Collapse
Affiliation(s)
- Eszter Lázár-Molnár
- ARUP Laboratories, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - Julio C Delgado
- ARUP Laboratories, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| |
Collapse
|
27
|
Tang F, Malek E, Math S, Schmotzer CL, Beck RC. Interference of Therapeutic Monoclonal Antibodies With Routine Serum Protein Electrophoresis and Immunofixation in Patients With Myeloma: Frequency and Duration of Detection of Daratumumab and Elotuzumab. Am J Clin Pathol 2018; 150:121-129. [PMID: 29901687 DOI: 10.1093/ajcp/aqy037] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVES We investigated the frequency and pattern of detection of therapeutic monoclonal antibodies (t-mAbs) daratumumab and elotuzumab by routine serum protein electrophoresis (SPE) and immunofixation (IF) in treated patients with myeloma. METHODS Detection of t-mAb was assessed in 22 patients by retrospective review of SPE/IF ordered prior to, during, and after 26 individual courses of therapy. RESULTS t-mAb was distinguishable from M-protein in 16 of 26 courses, with daratumumab detected in nine of nine and elotuzumab in six of seven patients. t-mAb was detected on first follow-up SPE/IF in 12 patients, with earliest detection 7 days after therapy initiation and latest detection 70 days after therapy. t-mAb persisted throughout induction therapy in most patients, with loss of detection during maintenance daratumumab. CONCLUSIONS When distinguishable from M-protein, t-mAbs are detectable in 93% of treated patients as soon as 7 days after the initial dose and are consistently observed throughout induction therapy, warranting increased monitoring and careful interpretation of SPE/IF.
Collapse
Affiliation(s)
- Felicia Tang
- School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Ehsan Malek
- School of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Hematology/Oncology, University Hospitals of Cleveland Medical Center, Cleveland, OH
| | - Susan Math
- Department of Pathology, University Hospitals of Cleveland Medical Center, Cleveland, OH
| | - Christine L Schmotzer
- School of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Pathology, University Hospitals of Cleveland Medical Center, Cleveland, OH
| | - Rose C Beck
- School of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Pathology, University Hospitals of Cleveland Medical Center, Cleveland, OH
| |
Collapse
|
28
|
Willrich MAV. Therapeutic Monoclonal Antibodies in the Clinical Laboratory. J Appl Lab Med 2017; 2:454-457. [PMID: 33636855 DOI: 10.1373/jalm.2016.022160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/06/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Maria Alice Vieira Willrich
- Department of Laboratory Medicine and Pathology, Division of Clinical Biochemistry and Immunology, Mayo Clinic, Rochester, MN
| |
Collapse
|
29
|
Keren DF, McCudden CR, Booth RA. Editorial on laboratory diagnosis and management of plasma cell dyscrasias special issue. Clin Biochem 2017; 51:1-3. [PMID: 29111449 DOI: 10.1016/j.clinbiochem.2017.10.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 10/25/2017] [Indexed: 11/16/2022]
Affiliation(s)
- David F Keren
- Division of Clinical Pathology, Department of Pathology, The University of Michigan Hospital and Health Systems, USA
| | - Christopher R McCudden
- Dept. of Pathology & Lab. Medicine, Division of Biochemistry, University of Ottawa, Canada.
| | - Ronald A Booth
- Dept. of Pathology & Lab. Medicine, Division of Biochemistry, University of Ottawa, Canada
| |
Collapse
|