1
|
Lee HW, Kim SM, Park HD. Evaluating the HYDRASHIFT 2/4 Daratumumab assay: a powerful approach to assess treatment response in multiple myeloma. Clin Chem Lab Med 2024; 62:2223-2232. [PMID: 38630027 DOI: 10.1515/cclm-2024-0416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 04/08/2024] [Indexed: 09/28/2024]
Abstract
OBJECTIVES This study evaluates the HYDRASHIFT assay's effectiveness in mitigating daratumumab interference on serum protein tests during multiple myeloma (MM) treatment, aiming to ensure an accurate assessment of treatment response. METHODS We analyzed 113 serum samples from 68 MM patients undergoing daratumumab treatment, employing both standard IF and the HYDRASHIFT assay. The assay's precision was determined through intra-day and inter-day variability assessments, while its specificity was verified using serum samples devoid of daratumumab. Comparative analysis of IF results, before and after the application of the HYDRASHIFT assay, facilitated the categorization of treatment responses in alignment with the International Myeloma Working Group's response criteria. RESULTS The precision underscored the assay's consistent repeatability and reproducibility, successfully eliminating interference of daratumumab-induced Gκ bands. Specificity assessments demonstrated the assay's capability to distinguish daratumumab from both isatuximab and naturally occurring M-proteins. Of the analyzed cases, 91 exhibited successful migration of daratumumab-induced Gκ bands, thereby enhancing the accuracy of treatment response classification. The remaining 22 cases did not show a visible migration complex, likely due to the low concentration of daratumumab in the serum. These findings underscore the assay's critical role in distinguishing daratumumab from endogenous M-protein, particularly in samples with a single Gκ band on standard IF, where daratumumab and endogenous M-protein had co-migrated. CONCLUSIONS The HYDRASHIFT assay demonstrates high precision, specificity, and utility in the accurate monitoring of treatment responses in MM patients receiving daratumumab. This assay represents a significant advancement in overcoming the diagnostic challenges posed by daratumumab interference.
Collapse
Affiliation(s)
- Hyun-Woo Lee
- Department of Laboratory Medicine and Genetics, 36626 Samsung Medical Center , Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sang-Mi Kim
- Department of Laboratory Medicine and Genetics, 36626 Samsung Medical Center , Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyung-Doo Park
- Department of Laboratory Medicine and Genetics, 36626 Samsung Medical Center , Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| |
Collapse
|
2
|
van de Donk NWCJ, Zweegman S. Monoclonal Antibodies in the Treatment of Multiple Myeloma. Hematol Oncol Clin North Am 2024; 38:337-360. [PMID: 38151402 DOI: 10.1016/j.hoc.2023.12.002] [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] [Indexed: 12/29/2023]
Abstract
The incorporation of monoclonal antibodies into backbone regimens has substantially improved the clinical outcomes of patients with newly diagnosed and relapsed/refractory multiple myeloma (MM). Although the SLAMF7-targeting antibody elotuzumab has no single- agent activity, there is clinical synergy between elotuzumab and immunomodulatory drugs in patients with relapsed/refractory disease. Daratumumab and isatuximab are CD38-targeting antibodies which have single-agent activity and a favorable safety profile, which make these agents an attractive component of combination regimens. Monoclonal antibodies may cause infusion-related reactions, but with subcutaneous administration these are less frequently observed. All therapeutic antibodies may interfere with assessment of complete response. Next-generation Fc-engineered monoclonal antibodies are in development with the potential to further improve the outcome of patients with MM.
Collapse
Affiliation(s)
- Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands.
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
| |
Collapse
|
3
|
Cárdenas MC, García-Sanz R, Puig N, Pérez-Surribas D, Flores-Montero J, Ortiz-Espejo M, de la Rubia J, Cruz-Iglesias E. Recommendations for the study of monoclonal gammopathies in the clinical laboratory. A consensus of the Spanish Society of Laboratory Medicine and the Spanish Society of Hematology and Hemotherapy. Part I: Update on laboratory tests for the study of monoclonal gammopathies. Clin Chem Lab Med 2023; 61:2115-2130. [PMID: 37477188 DOI: 10.1515/cclm-2023-0326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/29/2023] [Indexed: 07/22/2023]
Abstract
Monoclonal gammopathies (MG) are characterized by the proliferation of plasma cells that produce identical abnormal immunoglobulins (intact or some of their subunits). This abnormal immunoglobulin component is called monoclonal protein (M-protein), and is considered a biomarker of proliferative activity. The identification, characterization and measurement of M-protein is essential for the management of MG. We conducted a systematic review of the different tests and measurement methods used in the clinical laboratory for the study of M-protein in serum and urine, the biochemistry and hematology tests necessary for clinical evaluation, and studies in bone marrow, peripheral blood and other tissues. This review included literature published between 2009 and 2022. The paper discusses the main methodological characteristics and limitations, as well as the purpose and clinical value of the different tests used in the diagnosis, prognosis, monitoring and assessment of treatment response in MG. Included are methods for the study of M-protein, namely electrophoresis, measurement of immunoglobulin levels, serum free light chains, immunoglobulin heavy chain/light chain pairs, and mass spectrometry, and for the bone marrow examination, morphological analysis, cytogenetics, molecular techniques, and multiparameter flow cytometry.
Collapse
Affiliation(s)
- María C Cárdenas
- Department of Clinical Analysis, Hospital Clinico San Carlos, Madrid, Spain
- Protein Commission, Spanish Society of Laboratory Medicine (SEQCML), Barcelona, Spain
| | - Ramón García-Sanz
- Hematology Department, University Hospital of Salamanca, Research Biomedical Institute of Salamanca (IBSAL), CIBERONC and Center for Cancer Research-IBMCC (University of Salamanca-CSIC), Salamanca, Spain
- Spanish Society of Hematology and Hemotherapy (SEHH), Madrid, Spain
| | - Noemí Puig
- Hematology Department, University Hospital of Salamanca, Research Biomedical Institute of Salamanca (IBSAL), CIBERONC and Center for Cancer Research-IBMCC (University of Salamanca-CSIC), Salamanca, Spain
- Spanish Society of Hematology and Hemotherapy (SEHH), Madrid, Spain
| | - David Pérez-Surribas
- Laboratori Pasteur, Andorra La Vella, Andorra
- Protein Commission, Spanish Society of Laboratory Medicine (SEQCML), Barcelona, Spain
| | - Juan Flores-Montero
- Hematology Department, University Hospital of Salamanca, Research Biomedical Institute of Salamanca (IBSAL), CIBERONC and Center for Cancer Research-IBMCC (University of Salamanca-CSIC), Salamanca, Spain
- Spanish Society of Hematology and Hemotherapy (SEHH), Madrid, Spain
| | - María Ortiz-Espejo
- Department of Clinical Analysis, Hospital Universitario Marqués de Valdecilla, Santander, Spain
- Protein Commission, Spanish Society of Laboratory Medicine (SEQCML), Barcelona, Spain
| | - Javier de la Rubia
- Hematology Department, Hospital Universitario y Politécnico La Fe & Universidad Católica de Valencia, Instituto de Investigación Sanitaria La Fe Centro de Investigación Biomédica en Red de Cáncer, CIBERONC CB16/12/00284, Instituto de Salud Carlos III, Valencia, Spain
- Spanish Society of Hematology and Hemotherapy (SEHH), Madrid, Spain
| | - Elena Cruz-Iglesias
- Department of Laboratory Medicine, Osakidetza Basque Health Service, Basurto University Hospital, Bilbao, Spain
- Protein Commission, Spanish Society of Laboratory Medicine (SEQCML), Barcelona, Spain
| |
Collapse
|
4
|
Mele G, Di Renzo N, Cascavilla N, Carella AM, Guarini A, Mazza P, Melillo L, Pavone V, Tarantini G, Curci P, Falcone AP, Germano C, Mele A, Merchionne F, Palazzo G, Palumbo G, Quinto AM, Reddiconto G, Rossini B, Spina A, Sgherza N, Specchia G, Musto P, Pastore D. Real-world evidence on the use of daratumumab, bortezomib and dexamethasone (DVd) in lenalidomide-refractory myeloma patients: subgroup analysis of the multicenter retrospective experience by "rete ematologica pugliese". Leuk Lymphoma 2023; 64:1715-1718. [PMID: 37417467 DOI: 10.1080/10428194.2023.2232487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 06/24/2023] [Indexed: 07/08/2023]
Affiliation(s)
| | | | - Nicola Cascavilla
- Haematology, Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo (Foggia), Italy
| | - Angelo Michele Carella
- Haematology, Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo (Foggia), Italy
| | | | | | | | - Vincenzo Pavone
- Haematology, Ospedale Cardinale Panico, Tricase (Lecce), Italy
| | | | - Paola Curci
- Policlinico, University of Bari Medical School, Bari, Italy
| | | | - Candida Germano
- Haematology, Ospedale Monsignor R. Dimiccoli, Barletta (Bari), Italy
| | - Anna Mele
- Haematology, Ospedale Cardinale Panico, Tricase (Lecce), Italy
| | | | | | | | | | - Giovanni Reddiconto
- Haematology, Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo (Foggia), Italy
| | | | | | - Nicola Sgherza
- Policlinico, University of Bari Medical School, Bari, Italy
| | | | | | | |
Collapse
|
5
|
Pape LJ, Hambach J, Gebhardt AJ, Rissiek B, Stähler T, Tode N, Khan C, Weisel K, Adam G, Koch-Nolte F, Bannas P. CD38-specific nanobodies allow in vivo imaging of multiple myeloma under daratumumab therapy. Front Immunol 2022; 13:1010270. [PMID: 36389758 PMCID: PMC9647632 DOI: 10.3389/fimmu.2022.1010270] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/03/2022] [Indexed: 08/30/2023] Open
Abstract
RATIONALE Recent studies have demonstrated the feasibility of CD38-specific antibody constructs for in vivo imaging of multiple myeloma. However, detecting multiple myeloma in daratumumab-pretreated patients remains difficult due to overlapping binding epitopes of the CD38-specific imaging antibody constructs and daratumumab. Therefore, the development of an alternative antibody construct targeting an epitope of CD38 distinct from that of daratumumab is needed. We report the generation of a fluorochrome-conjugated nanobody recognizing such an epitope of CD38 to detect myeloma cells under daratumumab therapy in vitro, ex vivo, and in vivo. METHODS We conjugated the CD38-specific nanobody JK36 to the near-infrared fluorescent dye Alexa Fluor 680. The capacity of JK36AF680 to bind and detect CD38-expressing cells pretreated with daratumumab was evaluated on CD38-expressing tumor cell lines in vitro, on primary myeloma cells from human bone marrow biopsies ex vivo, and in a mouse tumor model in vivo. RESULTS Fluorochrome-labeled nanobody JK36AF680 showed specific binding to CD38-expressing myeloma cells pretreated with daratumumab in vitro and ex vivo and allowed for specific imaging of CD38-expressing xenografts in daratumumab-pretreated mice in vivo. CONCLUSIONS Our study demonstrates that a nanobody recognizing a distinct, non-overlapping epitope of CD38 allows the specific detection of myeloma cells under daratumumab therapy in vitro, ex vivo, and in vivo.
Collapse
Affiliation(s)
- Luca Julius Pape
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Hambach
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Josephine Gebhardt
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Rissiek
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Stähler
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Natalie Tode
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cerusch Khan
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katja Weisel
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerhard Adam
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Bannas
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
6
|
Leleu X, Martin T, Weisel K, Schjesvold F, Iida S, Malavasi F, Manier S, Chang-Ki Min, Ocio EM, Pawlyn C, Perrot A, Quach H, Richter J, Spicka I, Yong K, Richardson PG. Anti-CD38 antibody therapy for patients with relapsed/refractory multiple myeloma: differential mechanisms of action and recent clinical trial outcomes. Ann Hematol 2022; 101:2123-2137. [PMID: 35943588 PMCID: PMC9463192 DOI: 10.1007/s00277-022-04917-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 07/02/2022] [Indexed: 11/30/2022]
Abstract
CD38 is a transmembrane glycoprotein that functions both as a receptor and an ectoenzyme, playing key roles in the regulation of calcium signaling and migration of immune cells to tumor microenvironments. High expression on multiple myeloma (MM) cells and limited expression on normal cells makes CD38 an ideal target for the treatment of MM patients. Two monoclonal antibodies directed at CD38, isatuximab and daratumumab, are available for use in patients with relapsed and/or refractory MM (RRMM); daratumumab is also approved in newly diagnosed MM and light-chain amyloidosis. Clinical experience has shown that anti-CD38 antibody therapy is transforming treatment of MM owing to its anti-myeloma efficacy and manageable safety profile. Isatuximab and daratumumab possess similarities and differences in their mechanisms of action, likely imparted by their binding to distinct, non-overlapping epitopes on the CD38 molecule. In this review, we present the mechanistic properties of these two antibodies and outline available evidence on their abilities to induce adaptive immune responses and modulate the bone marrow niche in MM. Further, we discuss differences in regulatory labeling between these two agents and analyze recent key clinical trial results, including evidence in patients with underlying renal impairment and other poor prognostic factors. Finally, we describe the limited existing evidence for the use of isatuximab or daratumumab after disease progression on prior anti-CD38 mono- or combination therapy, highlighting the need for additional clinical evaluations to define optimal anti-CD38 antibody therapy selection and sequencing in RRMM.
Collapse
Affiliation(s)
- Xavier Leleu
- Service d'Hématologie Et Thérapie Cellulaire, CHU and CIC Inserm 1402, Poitiers Cedex, France.
| | - Thomas Martin
- Department of Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Katja Weisel
- University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Fredrik Schjesvold
- Oslo Myeloma Center, Department of Hematology, KG Jebsen Center for B Cell Malignancies, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University, Nagoya, Japan
| | - Fabio Malavasi
- Department of Medical Sciences, University of Torino Medical School, Fondazione Ricerca Molinette, Turin, Italy
| | - Salomon Manier
- Department of Hematology, CHU, Universite de Lille, Lille, France
| | - Chang-Ki Min
- Department of Hematology, College of Medicine, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Enrique M Ocio
- Hospital Universitario Marqués de Valdecilla (IDIVAL), Universidad de Cantabria, Santander, Spain
| | - Charlotte Pawlyn
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Aurore Perrot
- Department of Hematology, Institut Universitaire du Cancer de Toulouse, Toulouse, France
| | - Hang Quach
- Clinical Haematology Service, St Vincent's Hospital, University of Melbourne, Melbourne, Australia
| | - Joshua Richter
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Mount Sinai, New York, NY, USA
| | - Ivan Spicka
- Department of Medicine, Department of Hematology, First Faculty of Medicine, Charles University and General Hospital, Prague, Czech Republic
| | - Kwee Yong
- Department of Haematology, University College, Hospitals NHS Foundation Trust, London, UK
| | - Paul G Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
7
|
Liang S, Feng W, Ma H, Zhang L, Jia C. False positive results: a challenge for laboratory physicians and hematologists in treating multiple myeloma with daratumumab. Hematology 2022; 27:332-336. [PMID: 35255237 DOI: 10.1080/16078454.2022.2045723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Daratumumab injection was approved by China in 2019 for the treatment of recurrent or refractory multiple myeloma. However, the molecular weight of daratumumab, an immunoglobin G1 kappa human monoclonal antibody, was similar to that of M protein and could not be distinguished from IgG κ M protein in SPEP and SIFE. It might lead to false-positive detection resulting in misdiagnose and confusing evaluation of therapeutic response, especially for patients with IgG κ M proteins. Herein, we reported two cases encountered in our daily clinical work. These two case reports could serve as a reminder to global hematologists who have not yet started or just begun to use the drug of daratumumab.
Collapse
Affiliation(s)
- Shanshan Liang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Weihua Feng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hongbing Ma
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Li Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chengyao Jia
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
8
|
Perincheri S. Tumor Microenvironment of Lymphomas and Plasma Cell Neoplasms: Broad Overview and Impact on Evaluation for Immune Based Therapies. Front Oncol 2021; 11:719140. [PMID: 34956859 PMCID: PMC8692247 DOI: 10.3389/fonc.2021.719140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/16/2021] [Indexed: 12/20/2022] Open
Abstract
Lymphomas and plasma cell neoplasms are a heterogenous group of malignancies derived from lymphocytes. They are a significant cause of patient morbidity and mortality. Advances in morphologic, immunophenotypic and molecular techniques have led to better understanding of the pathogenesis and diagnosis of these neoplasms. Advances in treatment, particularly immune-based therapies, increasingly allow for targeted therapies of these diseases. Mechanistic studies using animal models and clinical trials have revealed the importance of the tumor microenvironment on disease pathogenesis, progression, and response to therapy in these malignancies. Simultaneous progress in diagnostic techniques has made it feasible to generate high-resolution, high-throughput data from the tumor microenvironment with spatial context. As the armamentarium of targeted therapies and diagnostic techniques grows, there is potential to harness these advances to better stratify patients for targeted therapies, including immune-based therapies, in hematologic malignancies.
Collapse
Affiliation(s)
- Sudhir Perincheri
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
9
|
Theodorakakou F, Dimopoulos MA, Kastritis E. Daratumumab plus CyBorD for patients with newly diagnosed light chain (AL) amyloidosis. Ther Adv Hematol 2021; 12:20406207211058334. [PMID: 34840708 PMCID: PMC8613887 DOI: 10.1177/20406207211058334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022] Open
Abstract
Primary systemic immunoglobulin light chain (AL) amyloidosis is caused by a plasma cell clone of, usually low, malignant potential that expresses CD38 molecules on their surface. Treatment of AL amyloidosis is based on the elimination of the plasma cell clone. The combination of cyclophosphamide–bortezomib–dexamethasone (CyBorD) is the most widely used and is considered a standard of care; however, complete hematologic response rates and organ response rates remain unsatisfactory. Daratumumab, an anti-CD38 monoclonal antibody, has demonstrated encouraging results, with rapid and deep responses, in patients with relapsed or refractory AL amyloidosis as monotherapy with a favorable toxicity profile. The large phase-III, randomized, ANDROMEDA study evaluated the addition of daratumumab to CyBorD in previously untreated patients with AL amyloidosis and demonstrated that addition of daratumumab can substantially improve hematologic complete response rates, survival free from major organ deterioration or hematologic progression, and organ responses. In this review, we discuss the role of daratumumab in the treatment of AL amyloidosis, its mechanism of action, and the results of ANDROMEDA study that led to the first approved therapy for AL amyloidosis.
Collapse
Affiliation(s)
- Foteini Theodorakakou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
10
|
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
|
11
|
El Hussein S, Wang W, Wang SA, Fang H, Garces S, Tashakori M, Lyapichev KA, Qiu L, Wang X, Loghavi S, Medeiros LJ, Challagundla P, Jorgensen JL. Hematogones with light chain restriction: A potential diagnostic pitfall when using flow cytometry analysis to assess bone marrow specimens. Leuk Res 2021; 111:106704. [PMID: 34735934 DOI: 10.1016/j.leukres.2021.106704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/30/2021] [Accepted: 09/02/2021] [Indexed: 12/20/2022]
Abstract
Light-chain restricted hematogones (LCR HGs) detected by flow cytometry analysis can mimic bone marrow involvement by B-cell lymphoma. This phenomenon can present a diagnostic pitfall and negatively impact patient management, as misinterpretation may upgrade disease stage. In this study, we characterized the immunophenotype of LCR HGs with an aim to differentiate them from B-cell lymphoma. We analyzed 24 patients with LCR HGs, 12 (50 %) were kappa light chain restricted and 12 (50 %) were lambda light chain restricted. LCR HGs account for 51 % (range, 1.5%-99%) of B cells, and 0.5 % (range, 0.1%-3.7%) of total cells. In 15 patients in whom multiple specimens were analyzed, 10 (67 %) showed persistent LCR HGs in more than 1 specimen, and the duration of the light chain restriction ranged from 4 months to 2 years. Among 24 patients, 4 (16.6 %) cases were concurrently involved by B-cell lymphoma/myeloma in addition to LCR HGs. With the exception of light chain restriction, LCR HGs showed a similar immunophenotype as normal HGs and had a distinct location on the CD45/Side Scatter (SSC) plot. They were also consistently positive for CD10, CD19, CD38 (bright), CD43, and CD200. CD20 expression showed a spectrum from dim/negative to positive.
Collapse
Affiliation(s)
- Siba El Hussein
- University of Rochester Medical Center, Department of Pathology, Rochester, New York, USA; The University of Texas MD Anderson Cancer Center, Department of Hematopathology, Houston, Texas, USA.
| | - Wei Wang
- The University of Texas MD Anderson Cancer Center, Department of Hematopathology, Houston, Texas, USA
| | - Sa A Wang
- The University of Texas MD Anderson Cancer Center, Department of Hematopathology, Houston, Texas, USA
| | - Hong Fang
- The University of Texas MD Anderson Cancer Center, Department of Hematopathology, Houston, Texas, USA
| | - Sofia Garces
- The University of Texas MD Anderson Cancer Center, Department of Hematopathology, Houston, Texas, USA
| | - Mehrnoosh Tashakori
- The University of Minnesota, Department of Pathology, Minneapolis, Minnesota, USA
| | - Kirill A Lyapichev
- University of Texas Medical Branch, Department of Pathology, Galveston, Texas, USA
| | - Lianqun Qiu
- The University of Texas MD Anderson Cancer Center, Department of Hematopathology, Houston, Texas, USA
| | - Xiaoqiong Wang
- The University of Texas MD Anderson Cancer Center, Department of Hematopathology, Houston, Texas, USA
| | - Sanam Loghavi
- The University of Texas MD Anderson Cancer Center, Department of Hematopathology, Houston, Texas, USA
| | - L Jeffrey Medeiros
- The University of Texas MD Anderson Cancer Center, Department of Hematopathology, Houston, Texas, USA
| | - Pramoda Challagundla
- Takeda Pharmaceutical International Co., OTAU Precision and Translation Medicine, Cambridge, Massachusetts, USA
| | - Jeffrey L Jorgensen
- The University of Texas MD Anderson Cancer Center, Department of Hematopathology, Houston, Texas, USA.
| |
Collapse
|
12
|
Noori S, Verkleij CPM, Zajec M, Langerhorst P, Bosman PWC, de Rijke YB, Zweegman S, VanDuijn M, Luider T, van de Donk NWCJ, Jacobs JFM. Monitoring the M-protein of multiple myeloma patients treated with a combination of monoclonal antibodies: the laboratory solution to eliminate interference. Clin Chem Lab Med 2021; 59:1963-1971. [PMID: 34392637 DOI: 10.1515/cclm-2021-0399] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/08/2021] [Indexed: 01/01/2023]
Abstract
OBJECTIVES The therapeutic monoclonal antibody (t-mAb) daratumumab, used to treat multiple myeloma (MM) patients, interferes with routine, electrophoretic based M-protein diagnostics. Electrophoretic response assessment becomes increasingly difficult when multiple t-mAbs are combined for use in a single patient. This is the first study to address the analytical challenges of M-protein monitoring when multiple t-mAbs are combined. METHODS In this proof-of-principle study we evaluate two different methods to monitor M-protein responses in three MM patients, who receive both daratumumab and nivolumab. The double hydrashift assay aims to resolve t-mAb interference on immunofixation. The MS-MRD (mass spectrometry minimal residual disease) assay measures clonotypic peptides to quantitate both M-protein and t-mAb concentrations. RESULTS After exposure to daratumumab and nivolumab, both t-mAbs become visible on immunofixation electrophoresis (IFE) as two IgG-kappa bands that migrate close to each other at the cathodal end of the γ-region. In case the M-protein co-migrates with these t-mAbs, the observed interference was completely abolished with the double IFE hydrashift assay. In all three patients the MS-MRD assay was also able to distinguish the M-protein from the t-mAbs. Additional advantage of the MS-MRD assay is that this multiplex assay is more sensitive and allows quantitative M-protein-, daratumumab- and nivolumab-monitoring. CONCLUSIONS Daratumumab and nivolumab interfere with electrophoretic M-protein diagnostics. However, the M-protein can be distinguished from both t-mAbs by use of a double hydrashift assay. The MS-MRD assay provides an alternative method that allows sensitive and simultaneous quantitative monitoring of both the M-protein and t-mAbs.
Collapse
Affiliation(s)
- Somayya Noori
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Christie P M Verkleij
- Department of Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marina Zajec
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Pieter Langerhorst
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Patricia W C Bosman
- Department of Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Yolanda B de Rijke
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Martijn VanDuijn
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Theo Luider
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Niels W C J van de Donk
- Department of Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Joannes F M Jacobs
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
13
|
Kirchhoff DC, Murata K, Thoren KL. Use of a Daratumumab-Specific Immunofixation Assay to Assess Possible Immunotherapy Interference at a Major Cancer Center: Our Experience and Recommendations. J Appl Lab Med 2021; 6:1476-1483. [PMID: 34293131 DOI: 10.1093/jalm/jfab055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 05/03/2021] [Indexed: 11/12/2022]
Abstract
BACKGROUND The incorporation of monoclonal antibodies, such as daratumumab, into multiple myeloma treatment regimens has led to the issue of false-positive interference in both serum protein electrophoresis (SPEP) and immunofixation (IF). The Hydrashift assay removes daratumumab interference from IF, allowing for correct interpretation. Here, we retrospectively examined the use of the Hydrashift assay at a large cancer center and provide guidelines on its most appropriate use. METHODS 38 patients with distinct daratumumab peaks on their SPEP were selected and were used to quantify the daratumumab peak on SPEP using the Sebia Phoresis software. A retrospective review of all Hydrashift assays ordered at our institution from July 2018 to March 2020 was performed. Data collected included patient clone type, IF migration patterns, and Hydrashift result. Serial quantification of SPEP results was performed as the corresponding IF transitioned from a true positive to a false positive. RESULTS Daratumumab adds a maximum magnitude of 0.20 g/dL on SPEP. Serial SPEP quantification showed IF transitioned from true positive to false positive when M-spikes ranged from 0.09 g/dL to 0.11 g/dL. Over 20 months, our laboratory performed 280 Hydrashift assays on 96 patients, 43/96 of whom had comigrating daratumumab/IgG-K IF bands. CONCLUSIONS The Hydrashift assay is typically unnecessary in patients with large M-spikes, >0.25 g/dL, regardless of clone type. When patient history is available, we recommend the Hydrashift assay be used in patients with comigrating daratumumab/IgG-K bands with M-spikes of <0.25 g/dL.
Collapse
Affiliation(s)
- Daniel C Kirchhoff
- Clinical Chemistry Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kazunori Murata
- Clinical Chemistry Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Katie L Thoren
- Clinical Chemistry Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
14
|
Hashmi H, Husnain M, Khan A, Usmani SZ. CD38-Directed Therapies for Management of Multiple Myeloma. Immunotargets Ther 2021; 10:201-211. [PMID: 34235096 PMCID: PMC8254545 DOI: 10.2147/itt.s259122] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/12/2021] [Indexed: 12/18/2022] Open
Abstract
The survival outcomes for multiple myeloma have improved several-fold in the past two decades, primarily due to the introduction of therapies with novel mechanisms of action including immunomodulatory agents, proteasome inhibitors, stem cell transplant and monoclonal antibodies in the schema of therapy. Antibody-based therapies targeting the surface marker CD38, namely daratumumab and isatuximab, have emerged as being highly effective as single agents as well as in combination regimens for both newly diagnosed and relapsed settings. Herein, the authors summarize the most recent data with both the current and emerging CD38-directed therapies in multiple myeloma.
Collapse
Affiliation(s)
- Hamza Hashmi
- Department of Medicine, Division of Hematology/Oncology, Medical University of South Carolina, Charleston, SC, USA
| | - Muhammad Husnain
- Department of Medicine, Division of Hematology/Oncology, University of Arizona, Tucson, AZ, USA
| | - Ali Khan
- Division of Plasma Cell Disorders, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA
| | - Saad Z Usmani
- Division of Plasma Cell Disorders, Levine Cancer Institute/Atrium Health, Charlotte, NC, USA
| |
Collapse
|
15
|
Mellors PW, Kohlhagen MC, Dasari S, Willrich MAV, Gertz MA, Kumar SK, Lacy MQ, Murray DL, Dispenzieri A. Belantamab mafodotin detection by MASS-FIX and immunofixation. Clin Chem Lab Med 2021; 59:e430-e433. [PMID: 34114387 DOI: 10.1515/cclm-2021-0326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/28/2021] [Indexed: 11/15/2022]
Affiliation(s)
| | - Mindy C 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 A V Willrich
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Morie A Gertz
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Shaji K Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Martha Q Lacy
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - David L Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Angela Dispenzieri
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
16
|
Jacobs JFM, Turner KA, Graziani MS, Frinack JL, Ettore MW, Tate JR, Booth RA, McCudden CR, Keren DF, Delgado JC, Zemtsovskaja G, Fullinfaw RO, Caldini A, de Malmanche T, Katakouzinos K, Burke M, Palladini G, Altinier S, Zaninotto M, Righetti G, Melki MT, Bell S, Willrich MAV. An international multi-center serum protein electrophoresis accuracy and M-protein isotyping study. Part II: limit of detection and follow-up of patients with small M-proteins. Clin Chem Lab Med 2021; 58:547-559. [PMID: 31940285 DOI: 10.1515/cclm-2019-1105] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 12/17/2019] [Indexed: 11/15/2022]
Abstract
Background Electrophoretic methods to detect, characterize and quantify M-proteins play an important role in the management of patients with monoclonal gammopathies (MGs). Significant uncertainty in the quantification and limit of detection (LOD) is documented when M-proteins are <10 g/L. Using spiked sera, we aimed to assess the variability in intact M-protein quantification and LOD across 16 laboratories. Methods Sera with normal, hypo- or hyper-gammaglobulinemia were spiked with daratumumab or elotuzumab, with concentrations from 0.125 to 10 g/L (n = 62) along with a beta-migrating sample (n = 9). Laboratories blindly analyzed samples according to their serum protein electrophoresis (SPEP)/isotyping standard operating procedures. LOD and intra-laboratory percent coefficient of variation (%CV) were calculated and further specified with regard to the method (gel/capillary electrophoresis [CZE]), gating strategy (perpendicular drop [PD]/tangent skimming [TS]), isotyping (immunofixation/immunosubtraction [ISUB]) and manufacturer (Helena/Sebia). Results All M-proteins ≥1 g/L were detected by SPEP. With isotyping the LOD was moderately more sensitive than with SPEP. The intensity of polyclonal background had the biggest negative impact on LOD. Independent of the method used, the intra-laboratory imprecision of M-protein quantification was small (mean CV = 5.0%). Low M-protein concentration and high polyclonal background had the strongest negative impact on intra-laboratory precision. All laboratories were able to follow trend of M-protein concentrations down to 1 g/L. Conclusions In this study, we describe a large variation in the reported LOD for both SPEP and isotyping; overall LOD is most affected by the polyclonal immunoglobulin background. Satisfactory intra-laboratory precision was demonstrated. This indicates that the quantification of small M-proteins to monitor patients over time is appropriate, when subsequent testing is performed within the same laboratory.
Collapse
Affiliation(s)
- Joannes F M Jacobs
- Laboratory Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands
| | - Katherine A Turner
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Jody L Frinack
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Michael W Ettore
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Jillian R Tate
- Department of Chemical Pathology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Ronald A Booth
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Ottawa, ON, Canada
| | - Christopher R McCudden
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Ottawa, ON, Canada
| | - David F Keren
- Department of Pathology, The University of Michigan, Ann Arbor, MI, USA
| | - Julio C Delgado
- ARUP Laboratories, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Galina Zemtsovskaja
- Clinical Chemistry Laboratory, North Estonia Medical Centre, Tallinn, Estonia
| | - Robert O Fullinfaw
- Department of Chemical Pathology, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Anna Caldini
- General Laboratory, Careggi University Hospital, Florence, Italy
| | - Theo de Malmanche
- NSW Health Pathology, Immunology Department, John Hunter Hospital, New Lambton Heights NSW, Australia
| | - Katina Katakouzinos
- Immunopathology Department, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Matthew Burke
- Department of Chemical Pathology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Giovanni Palladini
- Amyloidosis Research and Treatment Center, Foundation IRCCS Policlinico San Matteo, and Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Sara Altinier
- Laboratory Medicine of the University Hospital of Padova, Padova, Italy
| | - Martina Zaninotto
- Laboratory Medicine of the University Hospital of Padova, Padova, Italy
| | | | | | | | - Maria Alice Vieira Willrich
- Division of Clinical Biochemistry and Immunology, Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
17
|
Hosoya H, Sidana S. Antibody-Based Treatment Approaches in Multiple Myeloma. Curr Hematol Malig Rep 2021; 16:183-191. [PMID: 33730360 PMCID: PMC8715952 DOI: 10.1007/s11899-021-00624-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2021] [Indexed: 01/25/2023]
Abstract
PURPOSE OF REVIEW The field of multiple myeloma treatment has entered a new era with antibody-based approaches in clinical practice. In this review, we focus on the clinical approaches of utilizing antibody-based modality, specifically monoclonal antibodies, antibody-drug conjugates, and bispecific T-cell antibodies in the treatment of multiple myeloma. RECENT FINDINGS Three monoclonal antibodies (daratumumab, isatuximab, elotuzumab) and one anti-BCMA (B-cell maturation antigen) antibody-drug conjugate (belantamab mafodotin) have been approved by the FDA in the last 5 years for the treatment of multiple myeloma. There are many ongoing clinical trials using novel targets and constructs, including bispecific antibodies against BCMA, GPRC5D, and FCRH5. In addition to exploring efficacy, there are ongoing efforts to overcome the resistance to therapy. Antibody-based therapy has improved the outcomes of patients with multiple myeloma and has been incorporated in the standard of care. We expect to see novel targets and constructs that can achieve a deeper and more durable response while minimizing toxicity, as well as better strategies for toxicity management for existing agents. We also expect that antibody-based strategies will be used in earlier lines of therapy in the future.
Collapse
Affiliation(s)
- Hitomi Hosoya
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
- Stanford Cancer Institute, Stanford, CA, USA
| | - Surbhi Sidana
- Stanford Cancer Institute, Stanford, CA, USA.
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University, 300 Pasteur Drive, Room H0101c, Stanford, CA, USA.
| |
Collapse
|
18
|
Radocha J, van de Donk NWCJ, Weisel K. Monoclonal Antibodies and Antibody Drug Conjugates in Multiple Myeloma. Cancers (Basel) 2021; 13:1571. [PMID: 33805481 PMCID: PMC8037134 DOI: 10.3390/cancers13071571] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/22/2021] [Accepted: 03/27/2021] [Indexed: 12/24/2022] Open
Abstract
Multiple myeloma is the second most common hematologic malignancy. Current treatment strategies are mainly based on immunomodulatory drugs, proteasome inhibitors or combination of both. Novel agents added to these backbone treatments represent a promising strategy in treatment of newly diagnosed as well as relapsed and refractory multiple myeloma patients. In this respect, the incorporation of monoclonal antibodies into standard-of-care regimens markedly improved prognosis of myeloma patients during the last years. More specifically, monoclonal anti-CD38 antibodies, daratumumab and isatuximab, have been implemented into treatment strategies from first-line treatment to refractory disease. In addition, the monoclonal anti-SLAM-F7 antibody elotuzumab in combination with immunomodulatory drugs has improved the clinical outcomes of patients with relapsed/refractory disease. Belantamab mafodotin is the first approved antibody drug conjugate directed against B cell maturation antigen and is currently used as a monotherapy for patients with advanced disease. This review focuses on clinical efficacy and safety of monoclonal antibodies as well as antibody drug conjugates in multiple myeloma.
Collapse
Affiliation(s)
- Jakub Radocha
- 4th Department of Internal Medicine-Hematology, Faculty of Medicine in Hradec Králové, University Hospital Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic
| | - Niels W. C. J. van de Donk
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Katja Weisel
- II Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany;
| |
Collapse
|
19
|
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
|
20
|
Immunotherapy with Antibodies in Multiple Myeloma: Monoclonals, Bispecifics, and Immunoconjugates. HEMATO 2021. [DOI: 10.3390/hemato2010007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In the 2010s, immunotherapy revolutionized the treatment landscape of multiple myeloma. CD38-targeting antibodies were initially applied as monotherapy in end-stage patients, but are now also approved by EMA/FDA in combination with standards-of-care in newly diagnosed disease or in patients with early relapse. The approved SLAMF7-targeting antibody can also be successfully combined with lenalidomide or pomalidomide in relapsed/refractory myeloma. Although this has resulted in improved clinical outcomes, there remains a high unmet need in patients who become refractory to immunomodulatory drugs, proteasome inhibitors and CD38-targeting antibodies. Several new antibody formats, such as antibody–drug conjugates (e.g., belantamab mafodotin, which was approved in 2020 and targets BCMA) and T cell redirecting bispecific antibodies (e.g., teclistamab, talquetamab, cevostamab, AMG-420, and CC-93269) are active in these triple-class refractory patients. Based on their promising efficacy, it is expected that these new antibody formats will also be combined with other agents in earlier disease settings.
Collapse
|
21
|
Chen S, Kiguchi T, Nagata Y, Tamai Y, Ikeda T, Kajiya R, Ono T, Sugiyama D, Nishikawa H, Akatsuka Y. A simple method to distinguish residual elotuzumab from monoclonal paraprotein in immunofixation assays for multiple myeloma patients. Int J Hematol 2021; 113:473-479. [PMID: 33507526 DOI: 10.1007/s12185-021-03088-9] [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: 06/26/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 11/24/2022]
Abstract
Negative immunofixation electrophoresis (IFE) of serum and/or urine is a diagnostic marker for determining a complete response (CR) after immunotherapy for multiple myeloma (MM). However, residual therapeutic antibodies such as elotuzumab (IgG-κ), can compromise IFE evaluation when the affected immunoglobulins belong to the same IgG-κ subclass. We thus sought to develop a simple and rapid method to treat patient serum before IFE to distinguish the residual elotuzumab. Serum samples from patients receiving elotuzumab were treated with a predetermined amount of soluble signaling lymphocyte activation molecule F7 (SLAMF7) protein and then subjected to conventional IFE testing. We tested our method in samples from 12 patients. The IgG-κ band in IFE disappeared or shifted after elotuzumab treatment in four patients with no bone marrow minimal residual disease and normalized free light chain, whereas seven patients with any sign of residual MM showed a remaining IgG-κ band after treatment. One-hour incubation of samples with 6-9 molar excess soluble SLAMF7 before IFE was sufficient to distinguish residual elotuzumab in 11 of 12 samples. This simple method does not require special reagents, can be performed in most clinical laboratories, and enables differentiation between patients with a CR and those requiring further treatment.
Collapse
Affiliation(s)
- Shurui Chen
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Toru Kiguchi
- Department of Hematology, Chugoku Central Hospital, Fukuyama, Japan
| | - Yasuyuki Nagata
- Division of Hematology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yotaro Tamai
- Division of Hematology, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Takeshi Ikeda
- Division of Hematology and Stem Cell Transplantation, Shizuoka Cancer Center, Shizuoka, Japan
| | - Ryoko Kajiya
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Takaaki Ono
- Division of Hematology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Daisuke Sugiyama
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Hiroyoshi Nishikawa
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Yoshiki Akatsuka
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.
| |
Collapse
|
22
|
Kleinot W, Aguilera N, Courville EL. Daratumumab Interference in Flow Cytometry Producing a False Kappa Light Chain Restriction in Plasma Cells. Lab Med 2020; 52:403-409. [PMID: 33345283 DOI: 10.1093/labmed/lmaa107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
False kappa light chain restriction on hematogones (normal B-lineage precursors) has been described in patients on the therapeutic anti-CD38 monoclonal antibody daratumumab. In this article, we present a novel case report of pseudo-kappa light chain restriction on lambda-restricted neoplastic plasma cells in a patient with progressive plasma cell myeloma while on daratumumab. Flow cytometric technologists and pathologists need to be aware of this potential diagnostic pitfall.
Collapse
Affiliation(s)
- William Kleinot
- Department of Pathology, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Nadine Aguilera
- Department of Pathology, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Elizabeth L Courville
- Department of Pathology, University of Virginia Health System, Charlottesville, Virginia, USA
| |
Collapse
|
23
|
Korst CLBM, van de Donk NWCJ. Should all newly diagnosed MM patients receive CD38 antibody-based treatment? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2020; 2020:259-263. [PMID: 33275735 PMCID: PMC7727592 DOI: 10.1182/hematology.2020000161] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
CD38 antibodies were first evaluated in extensively pretreated patients with multiple myeloma (MM). Currently, there are 3 CD38 antibody-based regimens approved for the treatment of both transplant-eligible (daratumumab plus bortezomib-thalidomide-dexamethasone [D-VTd]) and transplant-ineligible (daratumumab plus lenalidomide-dexamethasone [D-Rd] or daratumumab plus bortezomib-melphalan-prednisone [D-VMP]) patients with newly diagnosed MM (NDMM). The phase 3 studies that evaluated these regimens uniformly showed that the addition of daratumumab to backbone regimens improved the depth of response, which translated into improved progression-free survival and also overall survival in 2 of the studies. Importantly, elderly patients age 75 years or older benefit from these regimens, indicating that these regimens have an acceptable safety profile. Although the number of patients with high-risk cytogenetics was relatively small, these patients also experienced benefit from the addition of daratumumab to standard-of-care regimens, but poor risk conferred by the cytogenetic aberrations is not completely abrogated. Altogether, daratumumab-based regimens have high anti-MM activity and a favorable toxicity profile and therefore represent new standards of care for patients with NDMM.
Collapse
Affiliation(s)
- Charlotte L B M Korst
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Niels W C J van de Donk
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Zajec M, Langerhorst P, VanDuijn MM, Gloerich J, Russcher H, van Gool AJ, Luider TM, Joosten I, de Rijke YB, Jacobs JFM. Mass Spectrometry for Identification, Monitoring, and Minimal Residual Disease Detection of M-Proteins. Clin Chem 2020; 66:421-433. [PMID: 32031591 DOI: 10.1093/clinchem/hvz041] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/13/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Monoclonal gammopathies (MGs) are plasma cell disorders defined by the clonal expansion of plasma cells, resulting in the characteristic excretion of a monoclonal immunoglobulin (M-protein). M-protein detection and quantification are integral parts of the diagnosis and monitoring of MGs. Novel treatment modalities impose new challenges on the traditional electrophoretic and immunochemical methods that are routinely used for M-protein diagnostics, such as interferences from therapeutic monoclonal antibodies and the need for increased analytical sensitivity to measure minimal residual disease. CONTENT Mass spectrometry (MS) is ideally suited to accurate mass measurements or targeted measurement of unique clonotypic peptide fragments. Based on these features, MS-based methods allow for the analytically sensitive measurement of the patient-specific M-protein. SUMMARY This review provides a comprehensive overview of the MS methods that have been developed recently to detect, characterize, and quantify M-proteins. The advantages and disadvantages of using these techniques in clinical practice and the impact they will have on the management of patients with MGs are discussed.
Collapse
Affiliation(s)
- M Zajec
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.,Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - P Langerhorst
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M M VanDuijn
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - J Gloerich
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - H Russcher
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - A J van Gool
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - T M Luider
- Department of Neurology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - I Joosten
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Y B de Rijke
- Department of Clinical Chemistry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - J F M Jacobs
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
25
|
Jullien M, Touzeau C, Moreau P. Monoclonal antibodies as an addition to current myeloma therapy strategies. Expert Rev Anticancer Ther 2020; 21:33-43. [PMID: 33052750 DOI: 10.1080/14737140.2021.1837627] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Immunotherapy has emerged as a major class in the therapeutic arsenal of multiple myeloma. Cell-based immunotherapy (CAR T-cells) and monoclonal antibody-based immunotherapy (naked monoclonal antibodies, antibody-drug conjugates, and bispecific T-cell engagers) are the two cornerstones of this novel approach for myeloma patients. Among numerous targets evaluated in the previous decade; CD38, SLAMF7, and, more recently, BCMA stand as the most promising. AREAS COVERED This review presents and discusses the currently available data regarding monoclonal antibodies in the treatment of multiple myeloma. EXPERT OPINION Anti-CD38-naked monoclonal antibodies have become a standard-of-care in multiple myeloma, greatly improving the depth and duration of response when combined with conventional therapy. Elotuzumab is approved in the relapse setting in combination with pomalidomide and maybe an interesting option in patients whose disease became refractory to anti-CD38 monoclonal antibodies. Anti-BCMA drug conjugates and bispecific T-cell engager antibodies are promising new molecules in the multiple myeloma armamentarium.
Collapse
Affiliation(s)
- Maxime Jullien
- Clinical Hematology, Nantes University Hospital , Nantes, France
| | - Cyrille Touzeau
- Clinical Hematology, Nantes University Hospital , Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université De Nantes , Nantes, France.,Site De Recherche Intégrée Sur Le Cancer (SIRIC) « ILIAD » , Nantes, France
| | - Philippe Moreau
- Clinical Hematology, Nantes University Hospital , Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université De Nantes , Nantes, France.,Site De Recherche Intégrée Sur Le Cancer (SIRIC) « ILIAD » , Nantes, France
| |
Collapse
|
26
|
Novel Approaches to Improve Myeloma Cell Killing by Monoclonal Antibodies. J Clin Med 2020; 9:jcm9092864. [PMID: 32899714 PMCID: PMC7564331 DOI: 10.3390/jcm9092864] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022] Open
Abstract
The monoclonal antibodies (mAbs) have significantly changed the treatment of multiple myeloma (MM) patients. However, despite their introduction, MM remains an incurable disease. The mAbs currently used for MM treatment were developed with different mechanisms of action able to target antigens, such as cluster of differentiation 38 (CD38) and SLAM family member 7 (SLAMF7) expressed by both, MM cells and the immune microenvironment cells. In this review, we focused on the mechanisms of action of the main mAbs approved for the therapy of MM, and on the possible novel approaches to improve MM cell killing by mAbs. Actually, the combination of anti-CD38 or anti-SLAMF7 mAbs with the immunomodulatory drugs significantly improved the clinical effect in MM patients. On the other hand, pre-clinical evidence indicates that different approaches may increase the efficacy of mAbs. The use of trans-retinoic acid, the cyclophosphamide or the combination of anti-CD47 and anti-CD137 mAbs have given the rationale to design these types of combinations therapies in MM patients in the future. In conclusion, a better understanding of the mechanism of action of the mAbs will allow us to develop novel therapeutic approaches to improve their response rate and to overcome their resistance in MM patients.
Collapse
|
27
|
Cerebrospinal Fluid Penetrance of Daratumumab in Leptomeningeal Multiple Myeloma. Hemasphere 2020; 4:e413. [PMID: 32904033 PMCID: PMC7448363 DOI: 10.1097/hs9.0000000000000413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/06/2020] [Indexed: 11/26/2022] Open
|
28
|
Horowitz GL. Monoclonal Proteins: Then and Now. J Appl Lab Med 2020. [DOI: 10.1093/jalm/jfaa036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Gary L Horowitz
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA
| |
Collapse
|
29
|
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: 26] [Impact Index Per Article: 6.5] [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
|
30
|
Complete Depletion of Daratumumab Interference in Serum Samples from Plasma Cell Myeloma Patients Improves the Detection of Endogenous M-Proteins in a Preliminary Study. Diagnostics (Basel) 2020; 10:diagnostics10040219. [PMID: 32295157 PMCID: PMC7235820 DOI: 10.3390/diagnostics10040219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/11/2020] [Accepted: 04/08/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Therapeutic humanized IgG1 kappa monoclonal antibody (t-mAb), daratumumab (DARA) is a Food and Drug Administration approved drug for the treatment of relapsed/refractory plasma cell myeloma (PCM). DARA appears on serum protein electrophoresis (SPEP) and on serum immunofixation (sIFE) as an IgG kappa monoclonal immunoglobulin protein (M-protein), complicating the assessment of the patients' response to therapy. A more ominous threat to patient safety can occur with the misinterpretation of the presence of a small t-mAb spike as being the residual product of the patient's neoplastic clone, presented either as oligoclonality or new clonality, which could result in incorrect interpretation of failure to achieve remission. METHODS In this report, we describe a novel and cost-effective technique based on biotinylated recombinant CD38 and streptavidin-coated magnetic beads to capture and remove residual DARA present in PCM patient serum samples. The treated samples are then run like regular samples on SPEP and sIFE. We validated this simple technique in DARA-spiked PCM samples and patient samples on DARA treatment. RESULTS Our simple capture technique completely extracted DARA in all of the tested serum specimens and allowed the assessment of residual M-protein without DARA interference. The results were reproducible and highly specific for DARA, and did not have any impact on endogenous M-protein migration and quantification by SPEP and sIFE. The cost of this technique is much lower and it can be performed in-house with a very short turnaround time compared to the currently available alternative methods. There is a great need for such reflex technologies to avoid interpretation errors. CONCLUSIONS This method is an effective way to eliminate DARA interference in SPEP and sIFE, and can be easily implemented in any clinical laboratory without any patent restriction. This simple technique can be adopted for other t-mAbs using their respective ligands and will help to reduce additional doses of toxic treatment and further testing in patients on t-mAbs with a false positive M-protein spike.
Collapse
|
31
|
Klener P, Etrych T, Klener P. Biological Therapy of Hematologic Malignancies: Toward a Chemotherapy- free Era. Curr Med Chem 2019; 26:1002-1018. [PMID: 28990505 DOI: 10.2174/0929867324666171006144725] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 09/07/2017] [Accepted: 09/15/2017] [Indexed: 12/20/2022]
Abstract
Less than 70 years ago, the vast majority of hematologic malignancies were untreatable diseases with fatal prognoses. The development of modern chemotherapy agents, which had begun after the Second World War, was markedly accelerated by the discovery of the structure of DNA and its role in cancer biology and tumor cell division. The path travelled from the first temporary remissions observed in children with acute lymphoblastic leukemia treated with single-agent antimetabolites until the first cures achieved by multi-agent chemotherapy regimens was incredibly short. Despite great successes, however, conventional genotoxic cytostatics suffered from an inherently narrow therapeutic index and extensive toxicity, which in many instances limited their clinical utilization. In the last decade of the 20th century, increasing knowledge on the biology of certain malignancies resulted in the conception and development of first molecularly targeted agents designed to inhibit specific druggable molecules involved in the survival of cancer cells. Advances in technology and genetic engineering enabled the production of structurally complex anticancer macromolecules called biologicals, including therapeutic monoclonal antibodies, antibody-drug conjugates and antibody fragments. The development of drug delivery systems (DDSs), in which conventional drugs were attached to various types of carriers including nanoparticles, liposomes or biodegradable polymers, represented an alternative approach to the development of new anticancer agents. Despite the fact that the antitumor activity of drugs attached to DDSs was not fundamentally different, the improved pharmacokinetic profiles, decreased toxic side effects and significantly increased therapeutic indexes resulted in their enhanced antitumor efficacy compared to conventional (unbound) drugs. Approval of the first immune checkpoint inhibitor for the treatment of cancer in 2011 initiated the era of cancer immunotherapy. Checkpoint inhibitors, bispecific T-cell engagers, adoptive T-cell approaches and cancer vaccines have joined the platform so far, represented mainly by recombinant cytokines, therapeutic monoclonal antibodies and immunomodulatory agents. In specific clinical indications, conventional drugs have already been supplanted by multi-agent, chemotherapy-free regimens comprising diverse immunotherapy and/or targeted agents. The very distinct mechanisms of the anticancer activity of new immunotherapy approaches not only call for novel response criteria, but might also change fundamental treatment paradigms of certain types of hematologic malignancies in the near future.
Collapse
Affiliation(s)
- Pavel Klener
- First Medical Department- Dept. of Hematology, First Faculty of Medicine and General University Hospital, Charles University, Czech Republic.,Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Czech Republic
| | - Tomas Etrych
- Department of biomedical polymers, Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, 162 06 Prague, Czech Republic
| | - Pavel Klener
- First Medical Department- Dept. of Hematology, First Faculty of Medicine and General University Hospital, Charles University, Czech Republic
| |
Collapse
|
32
|
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
|
33
|
Sidiqi MH, Gertz M. Daratumumab for the treatment of AL amyloidosis. Leuk Lymphoma 2019; 60:295-301. [PMID: 30033840 PMCID: PMC6342668 DOI: 10.1080/10428194.2018.1485914] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/25/2018] [Accepted: 05/29/2018] [Indexed: 12/19/2022]
Abstract
Autologous stem cell transplantation (ASCT) has been used as treatment for immunoglobulin light-chain (AL) amyloidosis for over two decades with improving outcomes; however, the majority of patients are not candidates for this therapy at diagnosis. Novel agents such as immunomodulatory drugs, proteasome inhibitors, and immunotherapy with monoclonal antibodies targeting CD38 have been adopted from the multiple myeloma spheres with encouraging results. Herein, we discuss the role of daratumumab, a monoclonal antibody to CD38, in the treatment of AL amyloidosis. We focus on its mechanism of action, tolerability, and the current published data on its use in AL amyloidosis. Early data from phase I and phase II studies show that daratumumab is tolerated well in this population and induces rapid and deep responses. Phase III trials are currently accruing and we envision daratumumab becoming a key component in the treatment of AL amyloidosis in the future.
Collapse
Affiliation(s)
- M Hasib Sidiqi
- Division of Hematology, Department of Internal Medicine, Mayo Clinic Rochester
| | - Morie Gertz
- Division of Hematology, Department of Internal Medicine, Mayo Clinic Rochester
| |
Collapse
|
34
|
Chen PP, Tormey CA, Eisenbarth SC, Torres R, Richardson SS, Rinder HM, Smith BR, Siddon AJ. False-Positive Light Chain Clonal Restriction by Flow Cytometry in Patients Treated With Alemtuzumab: Potential Pitfalls for the Misdiagnosis of B-Cell Neoplasms. Am J Clin Pathol 2019; 151:154-163. [PMID: 30307483 DOI: 10.1093/ajcp/aqy129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Objectives To increase awareness of potential diagnostic test interference associated with alemtuzumab, which is a therapeutic immunoglobulin G1 κ monoclonal antibody used in hematologic malignancies, autoimmune diseases, and transplant-related disorders. Methods Bone marrow and blood from patients with T-cell prolymphocytic leukemia treated with alemtuzumab were evaluated by flow cytometry. Healthy donor blood was analyzed with or without in vitro treatment with alemtuzumab for comparison. Results Immunophenotypic analysis of bone marrow collected 4 weeks after alemtuzumab treatment demonstrated artifactual surface κ light chain restriction in CD19+ B cells and CD3+ T cells. Similar findings were observed in blood from another patient in a specimen collected 3 days after alemtuzumab treatment. These findings were recapitulated in healthy donor blood incubated with alemtuzumab. Conclusions Alemtuzumab can produce direct interference during flow cytometry analysis, resulting in false-positive evidence of light chain clonality. Clinicians and laboratorians should be cognizant of this risk to avoid misdiagnosis of B-cell neoplasms.
Collapse
Affiliation(s)
- Peter P Chen
- Department of Pathology, Yale School of Medicine, New Haven, CT
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT
| | - Christopher A Tormey
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT
- Pathology and Laboratory Medicine Service, VA Connecticut Healthcare System, West Haven
| | | | - Richard Torres
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT
| | - Susan S Richardson
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT
| | - Henry M Rinder
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT
| | - Brian R Smith
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT
| | - Alexa J Siddon
- Department of Pathology, Yale School of Medicine, New Haven, CT
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT
- Pathology and Laboratory Medicine Service, VA Connecticut Healthcare System, West Haven
| |
Collapse
|
35
|
Croizier C, Bailly S. [Daratumumab for multiple myeloma]. Bull Cancer 2018; 105:985-991. [PMID: 30430991 DOI: 10.1016/j.bulcan.2018.09.003] [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: 06/13/2018] [Revised: 08/28/2018] [Accepted: 09/03/2018] [Indexed: 11/18/2022]
Abstract
Daratumumab is a monoclonal antibody that targets CD38. It has an anti-tumor action on the myeloma cell and an immunomodulatory action. For relapsing and/or refractory myeloma patients, daratumumab is effective both alone and in combination and significantly improves progression-free survival. Daratumumab has been approved in 2016 in France for treatment of relapses or refractory multiple myeloma.
Collapse
Affiliation(s)
- Carolyne Croizier
- CHU de Clermont-Ferrand, service thérapie cellulaire et hématologie clinique, 1, rue Lucie-Aubrac, 63100 Clermont-Ferrand, France.
| | - Sébastien Bailly
- CHU de Clermont-Ferrand, service thérapie cellulaire et hématologie clinique, 1, rue Lucie-Aubrac, 63100 Clermont-Ferrand, France
| |
Collapse
|
36
|
Caers J, Garderet L, Kortüm KM, O'Dwyer ME, van de Donk NWCJ, Binder M, Dold SM, Gay F, Corre J, Beguin Y, Ludwig H, Larocca A, Driessen C, Dimopoulos MA, Boccadoro M, Gramatzki M, Zweegman S, Einsele H, Cavo M, Goldschmidt H, Sonneveld P, Delforge M, Auner HW, Terpos E, Engelhardt M. European Myeloma Network recommendations on tools for the diagnosis and monitoring of multiple myeloma: what to use and when. Haematologica 2018; 103:1772-1784. [PMID: 30171031 PMCID: PMC6278986 DOI: 10.3324/haematol.2018.189159] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 08/27/2018] [Indexed: 01/04/2023] Open
Abstract
The diagnosis of multiple myeloma can be challenging, even for experienced physicians, and requires close collaboration between numerous disciplines (orthopedics, radiology, nuclear medicine, radiation therapy, hematology and oncology) before the final diagnosis of myeloma is made. The definition of multiple myeloma is based on the presence of clinical, biochemical, histopathological, and radiological markers of disease. Specific tests are needed both at presentation and during follow-up in order to reach the correct diagnosis and characterize the disease precisely. These tests can also serve prognostic purposes and are useful for follow-up of myeloma patients. Molecular analyses remain pivotal for defining high-risk myeloma and are used in updated patient stratifications, while minimal residual disease assessment via flow cytometry, molecular techniques and radiological approaches provides additional prognostic information on patients' long-term outcome. This pivotal information will guide our future treatment decisions in forthcoming clinical trials. The European Myeloma Network group updated their guidelines on different diagnostic recommendations, which should be of value to enable appropriate use of the recommendations both at diagnosis and during follow-up.
Collapse
Affiliation(s)
- Jo Caers
- Department of Hematology, University Hospital of Liege, Belgium .,Laboratory of Hematology, GIGA-I3, University of Liège, Belgium
| | | | - K Martin Kortüm
- Department of Internal Medicine II, University Hospital of Wuerzburg, Germany
| | - Michael E O'Dwyer
- Department of Hematology, National University of Ireland Galway, Ireland
| | | | - Mascha Binder
- Department of Internal Medicine II, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra Maria Dold
- Department of Medicine I, Hematology, Oncology & Stem Cell Transplantation, Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Francesca Gay
- Department of Hematology-Oncology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Jill Corre
- Unit for Genomics in Myeloma, Institut Universitaire du Cancer - Oncopole, Toulouse, France
| | - Yves Beguin
- Department of Hematology, University Hospital of Liege, Belgium.,Laboratory of Hematology, GIGA-I3, University of Liège, Belgium
| | - Heinz Ludwig
- Wilhelminen Cancer Research Institute, Vienna, Austria
| | - Alessandra Larocca
- Department of Hematology-Oncology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Christoph Driessen
- Department of Oncology and Hematology, Cantonal Hospital St. Gallen, Switzerland
| | | | - Mario Boccadoro
- Department of Hematology-Oncology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Martin Gramatzki
- Division of Stem Cell Transplantation and Immunotherapy, University of Kiel, Germany
| | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Wuerzburg, Germany
| | - Michele Cavo
- Seragnoli 'Institute of Hematology, Bologna University School of Medicine, Italy
| | - Hartmut Goldschmidt
- Department of Hematology, Rheumatology and Oncology, University Hospital Heidelberg, Germany.,National Center for Tumor Diseases, Heidelberg Medical University, Germany
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Michel Delforge
- Department of Hematology, University Hospital Leuven, Belgium
| | - Holger W Auner
- Centre for Haematology, Hammersmith Hospital, Imperial College London, UK
| | - Evangelos Terpos
- School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Monika Engelhardt
- Department of Medicine I, Hematology, Oncology & Stem Cell Transplantation, Medical Center, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
37
|
A universal solution for eliminating false positives in myeloma due to therapeutic monoclonal antibody interference. Blood 2018; 132:670-672. [DOI: 10.1182/blood-2018-05-848986] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
38
|
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: 26] [Impact Index Per Article: 4.3] [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
|
39
|
Ludwig H, Delforge M, Facon T, Einsele H, Gay F, Moreau P, Avet-Loiseau H, Boccadoro M, Hajek R, Mohty M, Cavo M, Dimopoulos MA, San-Miguel JF, Terpos E, Zweegman S, Garderet L, Mateos MV, Cook G, Leleu X, Goldschmidt H, Jackson G, Kaiser M, Weisel K, van de Donk NWCJ, Waage A, Beksac M, Mellqvist UH, Engelhardt M, Caers J, Driessen C, Bladé J, Sonneveld P. Prevention and management of adverse events of novel agents in multiple myeloma: a consensus of the European Myeloma Network. Leukemia 2018; 32:1542-1560. [PMID: 29720735 PMCID: PMC6035147 DOI: 10.1038/s41375-018-0040-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/23/2017] [Accepted: 11/30/2017] [Indexed: 01/03/2023]
Abstract
During the last few years, several new drugs have been introduced for treatment of patients with multiple myeloma, which have significantly improved the treatment outcome. All of these novel substances differ at least in part in their mode of action from similar drugs of the same drug class, or are representatives of new drug classes, and as such present with very specific side effect profiles. In this review, we summarize these adverse events, provide information on their prevention, and give practical guidance for monitoring of patients and for management of adverse events.
Collapse
Affiliation(s)
- Heinz Ludwig
- Wilhelminen Cancer Research Institute, Vienna, Austria.
| | - Michel Delforge
- Department of Hematology, University Hospital Leuven, Catholic University Leuven, Leuven, Belgium
| | - Thierry Facon
- Department of Hematology, Lille University Hospital, Lille, France
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Francesca Gay
- Myeloma Unit, Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Philippe Moreau
- Department of Hematology, University Hospital, University of Nantes, Nantes, France
| | - Hervé Avet-Loiseau
- Centre de Recherches en Cancerologie de Toulouse CRCT, Institut National de la Sante et de la Recherche Medicale, Université Toulouse, Toulouse, France
| | - Mario Boccadoro
- Division of Hematology, Citta della Salute e della Scienza, University of Torino, Torino, Italy
| | - Roman Hajek
- Faculty of Medicine, University Hospital Ostrava, University of Ostrava, Ostrava, Czech Republic
| | - Mohamad Mohty
- Department of Haematology, Saint Antoine Hospital, University Pierre and Marie Curie, and INSERM UMRs 938, Paris, France
| | - Michele Cavo
- A 'Seràgnoli' Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Jesús F San-Miguel
- Department of Hematology, Clinica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Laurent Garderet
- Department of Haematology, Saint Antoine Hospital, University Pierre and Marie Curie, and INSERM UMRs 938, Paris, France
| | - María-Victoria Mateos
- Hospital Universitario de Salamanca, Instituto Biosanitario de Salamanca (IBSAL), Salamanca, Spain
| | - Gordon Cook
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Xavier Leleu
- Service d'Hématologie et Thérapie Cellulaire, PRC, and Inserm CIC1402, Hospital de la Miléterie, Poitiers, France
| | - Hartmut Goldschmidt
- National Center for Tumor Diseases, Heidelberg Medical University, Heidelberg, Germany
| | - Graham Jackson
- Department of Hematology, Newcastle University, Newcastle, UK
| | - Martin Kaiser
- Myeloma Group, The Institute of Cancer Research ICR, London, UK
| | - Katja Weisel
- Department of Hematology and Oncology, University of Tuebingen, Tuebingen, Germany
| | | | - Anders Waage
- Department of Hematology, St. Olavs Hospital, and IKOM, NTNU, Trondheim, Norway
| | - Meral Beksac
- Department of Medicine, Ankara University, Ankara, Turkey
| | - Ulf H Mellqvist
- Department of Hematology, Sahlgrenska Hospital, Gothenburg, Sweden
| | - Monika Engelhardt
- Department of Hematology and Oncology, University of Freiburg Medical Center, Freiburg, Germany
| | - Jo Caers
- Department of Hematology, University Hospital of Liège, Liège, Belgium
| | - Christoph Driessen
- Department of Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Joan Bladé
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic de Barcelona, Spain
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| |
Collapse
|
40
|
Plesner T, Krejcik J. Daratumumab for the Treatment of Multiple Myeloma. Front Immunol 2018; 9:1228. [PMID: 29915586 PMCID: PMC5994592 DOI: 10.3389/fimmu.2018.01228] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/16/2018] [Indexed: 11/13/2022] Open
Abstract
This mini-review will summarize the present state of development of the CD38 antibody daratumumab for the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Torben Plesner
- Institute of Regional Health Science, University of Southern Denmark, Vejle Hospital, Vejle, Denmark
| | - Jakub Krejcik
- Department of Hematology, Vejle Hospital, Vejle, Denmark
| |
Collapse
|
41
|
Thoren KL, Pianko MJ, Maakaroun Y, Landgren CO, Ramanathan LV. Distinguishing Drug from Disease by Use of the Hydrashift 2/4 Daratumumab Assay. J Appl Lab Med 2018; 3:857-863. [PMID: 31639760 DOI: 10.1373/jalm.2018.026476] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/30/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Daratumumab, a monoclonal antibody used to treat relapsed or refractory multiple myeloma, can interfere with protein electrophoresis and immunofixation assays. False-positive immunofixation results due to daratumumab can cause uncertainty regarding the status of a patient's disease and lead to potential misclassification of their response to therapy. The Hydrashift 2/4 Daratumumab assay (Sebia) was recently cleared by the Food and Drug Administration for resolving daratumumab interference on immunofixation. Here, we evaluate the performance of the Hydrashift assay in multiple myeloma patients receiving treatment with daratumumab-based regimens. METHODS Waste serum samples from multiple myeloma patients (n = 40) receiving daratumumab were analyzed by standard immunofixation and the Hydrashift assay. Results from these tests were compared and were evaluated along with pretreatment serum protein electrophoresis and immunofixation results, if available. RESULTS The Hydrashift assay shifted the migration of daratumumab in patient samples. In 27 cases, the patient's M protein was distinguishable from daratumumab by standard immunofixation. In these cases, the Hydrashift assay confirmed that the IgGκ band was daratumumab and helped identify the presence of treatment-related oligoclonal bands. There were 11 instances in which the patient's IgGκ M protein comigrated with daratumumab. In all 11 cases, the Hydrashift assay confirmed the presence of residual M protein. Finally, in 2 patients whose pretreatment immunofixation results were not available, the Hydrashift assay confirmed that the IgGκ band visible on immunofixation was due to daratumumab alone. CONCLUSIONS The Hydrashift 2/4 Daratumumab assay is a useful tool to clarify the source of an IgGκ band on immunofixation and allow a patient's M protein to be viewed without interference.
Collapse
Affiliation(s)
- Katie L Thoren
- Clinical Chemistry Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY;
| | - Matthew J Pianko
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - C Ola Landgren
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lakshmi V Ramanathan
- Clinical Chemistry Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
42
|
Bride KL, Vincent TL, Im SY, Aplenc R, Barrett DM, Carroll WL, Carson R, Dai Y, Devidas M, Dunsmore KP, Fuller T, Glisovic-Aplenc T, Horton TM, Hunger SP, Loh ML, Maude SL, Raetz EA, Winter SS, Grupp SA, Hermiston ML, Wood BL, Teachey DT. Preclinical efficacy of daratumumab in T-cell acute lymphoblastic leukemia. Blood 2018; 131:995-999. [PMID: 29305553 PMCID: PMC5833263 DOI: 10.1182/blood-2017-07-794214] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/11/2017] [Indexed: 12/27/2022] Open
Abstract
As a consequence of acquired or intrinsic disease resistance, the prognosis for patients with relapsed or refractory T-cell acute lymphoblastic leukemia (T-ALL) is dismal. Novel, less toxic drugs are clearly needed. One of the most promising emerging therapeutic strategies for cancer treatment is targeted immunotherapy. Immune therapies have improved outcomes for patients with other hematologic malignancies including B-cell ALL; however no immune therapy has been successfully developed for T-ALL. We hypothesize targeting CD38 will be effective against T-ALL. We demonstrate that blasts from patients with T-ALL have robust surface CD38 surface expression and that this expression remains stable after exposure to multiagent chemotherapy. CD38 is expressed at very low levels on normal lymphoid and myeloid cells and on a few tissues of nonhematopoietic origin, suggesting that CD38 may be an ideal target. Daratumumab is a human immunoglobulin G1κ monoclonal antibody that binds CD38, and has been demonstrated to be safe and effective in patients with refractory multiple myeloma. We tested daratumumab in a large panel of T-ALL patient-derived xenografts (PDX) and found striking efficacy in 14 of 15 different PDX. These data suggest that daratumumab is a promising novel therapy for pediatric T-ALL patients.
Collapse
Affiliation(s)
- Karen L Bride
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Tiffaney L Vincent
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Soo-Yeon Im
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Richard Aplenc
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David M Barrett
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - William L Carroll
- Laura and Isaac Perlmutter Cancer Center at NYU Langone, New York University, New York, NY
| | | | - Yunfeng Dai
- Department of Biostatistics, University of Florida, Gainesville, FL
| | | | | | - Tori Fuller
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Tina Glisovic-Aplenc
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Terzah M Horton
- Baylor College of Medicine Dan L. Duncan Comprehensive Cancer Center, Houston, TX
| | - Stephen P Hunger
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Mignon L Loh
- Division of Hematology/Oncology, University of California San Francisco Benioff Children's Hospital, San Francisco, CA
| | - Shannon L Maude
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Elizabeth A Raetz
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Stuart S Winter
- Children's Minnesota Cancer and Blood Disorders, Minneapolis, MN; and
| | - Stephan A Grupp
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michelle L Hermiston
- Division of Hematology/Oncology, University of California San Francisco Benioff Children's Hospital, San Francisco, CA
| | | | - David T Teachey
- Division of Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
43
|
Frerichs KA, Nagy NA, Lindenbergh PL, Bosman P, Marin Soto J, Broekmans M, Groen RWJ, Themeli M, Nieuwenhuis L, Stege C, Nijhof IS, Mutis T, Zweegman S, Lokhorst HM, van de Donk NWCJ. CD38-targeting antibodies in multiple myeloma: mechanisms of action and clinical experience. Expert Rev Clin Immunol 2018; 14:197-206. [PMID: 29465271 DOI: 10.1080/1744666x.2018.1443809] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Multiple myeloma (MM) is generally an incurable hematological malignancy with heterogeneous overall survival rates ranging from a few months to more than 10 years. Survival is especially poor for patients who developed disease that is refractory to immunomodulatory drugs and proteasome inhibitors. Areas covered: This review will discuss the importance of CD38-targeting antibodies for the treatment of MM patients to improve their outcome. Expert commentary: Intense immuno-oncological laboratory research has resulted in the development of functionally active monoclonal antibodies against cell surface markers present on MM cells. In this respect, CD38-targeting antibodies such as daratumumab, MOR202, and isatuximab, have high single agent activity in heavily pretreated MM patients by virtue of their pleiotropic mechanisms of action including Fc-dependent effector mechanisms and immunomodulatory activities. Importantly, CD38-targeting antibodies are well tolerated, with infusion reactions as most frequent adverse event. Altogether, this makes them attractive combination partners with other anti-MM agents. Daratumumab is already approved as monotherapy and in combination with lenalidomide-dexamethasone as well as bortezomib-dexamethasone in pretreated MM patients. Furthermore, results from studies evaluating CD38-targeting antibodies in newly diagnosed MM patients are also promising, indicating that CD38-targeting antibodies will be broadly used in MM, resulting in further improvements in survival.
Collapse
Affiliation(s)
- Kristine A Frerichs
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Noemi Anna Nagy
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Pieter L Lindenbergh
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Patty Bosman
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Jhon Marin Soto
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Marloes Broekmans
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Richard W J Groen
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Maria Themeli
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Louise Nieuwenhuis
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Claudia Stege
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Inger S Nijhof
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Tuna Mutis
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Sonja Zweegman
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | - Henk M Lokhorst
- a Department of Hematology , VU University Medical Center , Amsterdam , The Netherlands
| | | |
Collapse
|
44
|
Zajec M, Jacobs JFM, Groenen PJTA, de Kat Angelino CM, Stingl C, Luider TM, De Rijke YB, VanDuijn MM. Development of a Targeted Mass-Spectrometry Serum Assay To Quantify M-Protein in the Presence of Therapeutic Monoclonal Antibodies. J Proteome Res 2018; 17:1326-1333. [DOI: 10.1021/acs.jproteome.7b00890] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Marina Zajec
- Department
of Neurology, Erasmus MC University Medical Center, Rotterdam 3015 GE, The Netherlands
- Department
of Clinical Chemistry, Erasmus MC University Medical Center, Rotterdam 3015 GE, The Netherlands
| | - Joannes F. M. Jacobs
- Department
of Laboratory Medicine, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands
| | | | - Corrie M. de Kat Angelino
- Department
of Laboratory Medicine, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands
| | - Christoph Stingl
- Department
of Neurology, Erasmus MC University Medical Center, Rotterdam 3015 GE, The Netherlands
| | - Theo M. Luider
- Department
of Neurology, Erasmus MC University Medical Center, Rotterdam 3015 GE, The Netherlands
| | - Yolanda B. De Rijke
- Department
of Clinical Chemistry, Erasmus MC University Medical Center, Rotterdam 3015 GE, The Netherlands
| | - Martijn M. VanDuijn
- Department
of Neurology, Erasmus MC University Medical Center, Rotterdam 3015 GE, The Netherlands
| |
Collapse
|
45
|
Kumar SK, Anderson KC. Immune Therapies in Multiple Myeloma. Clin Cancer Res 2018; 22:5453-5460. [PMID: 28151713 DOI: 10.1158/1078-0432.ccr-16-0868] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/20/2016] [Accepted: 09/22/2016] [Indexed: 11/16/2022]
Abstract
Treatment paradigms have changed rapidly for multiple myeloma, and immune therapies have taken center stage. Advances in therapies for myeloma have led to a dramatic improvement in the survival of patients with this incurable malignancy. The immune system is significantly impaired in patients with myeloma as a result of the disease leading to suppression of normal plasma cells as well the negative effects on cellular immunity. Given this scenario, immune approaches have not been successful until recently. Monoclonal antibodies directed against CD38 (daratumumab) and SLAMF7 (elotuzumab) are already in the clinic, and several other antibodies directed against different plasma cell antigens are under evaluation. Although immune checkpoint inhibition with PD-1 inhibitors had no clinical efficacy when the inhibitors were used as single agents, it has led to some dramatic results when the inhibitors are combined with immunomodulatory drugs such as lenalidomide and pomalidomide. Vaccination strategies have shown in vivo immune responses but no clear clinical efficacy. Newer approaches to vaccination with multiple antigens, used in combinations with immunomodulatory drugs and in the setting of minimal residual disease, have all increased possibility of this approach succeeding. Ex vivo effector cell expansion also appears to be promising and is in clinical trials. Finally, a chimeric antigen receptor T-cell approach appears to have some promise based on isolated reports of success and remains an area of intense investigation. Immune-based approaches can potentially augment or even supplant some of the current approaches and, given the low toxicity profile, may hold great potential in the early treatment of precursor-stage diseases. Clin Cancer Res; 22(22); 5453-60. ©2016 AACR SEE ALL ARTICLES IN THIS CCR FOCUS SECTION, "MULTIPLE MYELOMA MULTIPLYING THERAPIES".
Collapse
Affiliation(s)
- Shaji K Kumar
- Division of Hematology, Mayo Clinic, Rochester, Minnesota.
| | | |
Collapse
|
46
|
McCudden CR, Jacobs JF, Keren D, Caillon H, Dejoie T, Andersen K. Recognition and management of common, rare, and novel serum protein electrophoresis and immunofixation interferences. Clin Biochem 2018; 51:72-79. [DOI: 10.1016/j.clinbiochem.2017.08.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 08/21/2017] [Accepted: 08/21/2017] [Indexed: 12/24/2022]
|
47
|
Genzen JR, Murray DL, Abel G, Meng QH, Baltaro RJ, Rhoads DD, Delgado JC, Souers RJ, Bashleben C, Keren DF, Ansari MQ. Screening and Diagnosis of Monoclonal Gammopathies: An International Survey of Laboratory Practice. Arch Pathol Lab Med 2017; 142:507-515. [DOI: 10.5858/arpa.2017-0128-cp] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context.—
Serum tests used for the screening and diagnosis of monoclonal gammopathies include serum protein electrophoresis (SPE; agarose gel or capillary zone), immunofixation (IFE) and immunosubtraction capillary electrophoresis, serum free light chains, quantitative immunoglobulins, and heavy/light–chain combinations. Urine protein electrophoresis and urine IFE may also be used to identify Bence-Jones proteinuria.
Objective.—
To assess current laboratory practice for monoclonal gammopathy testing.
Design.—
In April 2016, a voluntary questionnaire was distributed to 923 laboratories participating in a protein electrophoresis proficiency testing survey.
Results.—
Seven hundred seventy-four laboratories from 38 countries and regions completed the questionnaire (83.9% response rate; 774 of 923). The majority of participants (68.6%; 520 of 758) used agarose gel electrophoresis as their SPE method, whereas 31.4% (238 of 758) used capillary zone electrophoresis. The most common test approaches used in screening were SPE with reflex to IFE/immunosubtraction capillary electrophoresis (39.3%; 299 of 760); SPE only (19.1%; 145 of 760); SPE and IFE or immunosubtraction capillary electrophoresis (13.9%; 106 of 760); and SPE with IFE, serum free light chain, and quantitative immunoglobulins (11.8%; 90 of 760). Only 39.8% (305 of 767) of laboratories offered panel testing for ordering convenience. Although SPE was used by most laboratories in diagnosing new cases of myeloma, when laboratories reported the primary test used to follow patients with monoclonal gammopathy, only 55.7% (403 of 724) chose SPE, with the next most common selections being IFE (18.9%; 137 of 724), serum free light chain (11.7%; 85 of 724), and immunosubtraction capillary electrophoresis (2.1%; 15 of 724).
Conclusions.—
Ordering and testing practices for the screening and diagnosis of monoclonal gammopathy vary widely across laboratories. Improving utilization management and report content, as well as recognition and development of laboratory-directed testing guidelines, may serve to enhance the clinical value of testing.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Mohammad Q. Ansari
- From the Department of Pathology, University of Utah, Salt Lake City, and ARUP Laboratories, Salt Lake City, Utah (Drs Genzen and Delgado); the Department of Laboratory Medicine, Mayo Clinic, Rochester, Minnesota (Dr Murray); the Department of Laboratory Medicine, Lahey Hospital & Medical Center, Burlington, Massachusetts (Dr Abel); the Department of Laboratory Medicine, University of Texas MD An
| |
Collapse
|
48
|
Cho SF, Lin L, Xing L, Yu T, Wen K, Anderson KC, Tai YT. Monoclonal Antibody: A New Treatment Strategy against Multiple Myeloma. Antibodies (Basel) 2017; 6:antib6040018. [PMID: 31548533 PMCID: PMC6698817 DOI: 10.3390/antib6040018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 12/22/2022] Open
Abstract
2015 was a groundbreaking year for the multiple myeloma community partly due to the breakthrough approval of the first two monoclonal antibodies in the treatment for patients with relapsed and refractory disease. Despite early disappointments, monoclonal antibodies targeting CD38 (daratumumab) and signaling lymphocytic activation molecule F7 (SLAMF7) (elotuzumab) have become available for patients with multiple myeloma in the same year. Specifically, phase 3 clinical trials of combination therapies incorporating daratumumab or elotuzumab indicate both efficacy and a very favorable toxicity profile. These therapeutic monoclonal antibodies for multiple myeloma can kill target cells via antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity, and antibody-dependent phagocytosis, as well as by direct blockade of signaling cascades. In addition, their immunomodulatory effects may simultaneously inhibit the immunosuppressive bone marrow microenvironment and restore the key function of immune effector cells. In this review, we focus on monoclonal antibodies that have shown clinical efficacy or promising preclinical anti-multiple myeloma activities that warrant further clinical development. We summarize mechanisms that account for the in vitro and in vivo anti-myeloma effects of these monoclonal antibodies, as well as relevant preclinical and clinical results. Monoclonal antibody-based immunotherapies have already and will continue to transform the treatment landscape in multiple myeloma.
Collapse
Affiliation(s)
- Shih-Feng Cho
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Liang Lin
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Lijie Xing
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, No. 324, Jingwu Road, Jinan 250021, China.
| | - Tengteng Yu
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Kenneth Wen
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
49
|
CD38 antibodies in multiple myeloma: back to the future. Blood 2017; 131:13-29. [PMID: 29118010 DOI: 10.1182/blood-2017-06-740944] [Citation(s) in RCA: 304] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/17/2017] [Indexed: 11/20/2022] Open
Abstract
CD38 is highly and uniformly expressed on multiple myeloma (MM) cells, and at relatively low levels on normal lymphoid and myeloid cells, and in some tissues of nonhematopoietic origin. CD38 is a transmembrane glycoprotein with ectoenzymatic activity, and also functions as a receptor and adhesion molecule. Altogether, this has triggered the development of several CD38 antibodies including daratumumab (fully human), isatuximab (chimeric), and MOR202 (fully human). CD38 antibodies have pleiotropic mechanisms of action including Fc-dependent immune-effector mechanisms, direct apoptotic activity, and immunomodulatory effects by the elimination of CD38+ immune-suppressor cells. CD38-targeting antibodies are generally well tolerated and induce partial response or better in ∼30% of heavily pretreated MM patients as monotherapy. Based on their distinct mechanisms of action, favorable toxicity profile, and single-agent activity, CD38 antibodies are attractive partners in combination regimens. Indeed, deep responses and prolonged progression-free survival can be achieved in relapsed/refractory MM patients when CD38 antibodies are combined with immunomodulatory agents or proteasome inhibitors. Infusion-related reactions, which typically occur during the first infusion, are the most frequent adverse events. Attention should also be paid to the interference of CD38 antibodies with certain laboratory assays, which may complicate response evaluation and blood compatibility testing. Several studies are currently examining the role of CD38-based therapies in newly diagnosed and high-risk smoldering MM. Furthermore, CD38 antibodies are currently also under investigation in other hematologic malignancies, including acute lymphoblastic leukemia, natural killer/T-cell lymphoma, and acute myeloid leukemia, as well as in solid tumors.
Collapse
|
50
|
Zhang Z, Hu W, Li L, Ding H, Li H. Therapeutic monoclonal antibodies and clinical laboratory tests: When, why, and what is expected? J Clin Lab Anal 2017; 32. [PMID: 28810082 DOI: 10.1002/jcla.22307] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/22/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND We herein provide an overview of the clinical laboratory tests that should be performed before, during and after using therapeutic monoclonal antibodies (mAbs) and the clinical laboratory tests that may be affected by mAbs. METHODS The labels of FDA-approved therapeutic mAbs were downloaded from DailyMed (the official website for drug labels) and were used as the sources of data for this review. RESULTS It was found that most of the labels provided information relevant to the clinical laboratory tests, including the tests needed before mAbs treatment to check the patients' background status and to identify potentially sensitive patients, the tests needed during or after the treatment to evaluate the patients' response, and the mAbs that may lead to false positive or negative results for clinical laboratory tests. CONCLUSIONS The present findings will be of interest to physicians, laboratory scientists, those involved in drug development and surveillance and individuals making health care policy.
Collapse
Affiliation(s)
- Zhanhu Zhang
- Institute of Medical Genetics and Reproductive Medicine, Nantong Maternity and Child Health Hospital, Nantong, Jiangsu, China
| | - Wenjie Hu
- Department of Clinical Laboratory Medicine, Nantong Maternal and Child Health Hospital, Nantong, Jiangsu, China
| | - Linlin Li
- Department of Clinical Laboratory Medicine, Nantong Maternal and Child Health Hospital, Nantong, Jiangsu, China
| | | | - Haibo Li
- Department of Clinical Laboratory Medicine, Nantong Maternal and Child Health Hospital, Nantong, Jiangsu, China
| |
Collapse
|