1
|
Hosoya H, Carleton M, Tanaka K, Sworder B, Syal S, Sahaf B, Maltos AM, Silva O, Stehr H, Hovanky V, Duran G, Zhang T, Liedtke M, Arai S, Iberri D, Miklos D, Khodadoust MS, Sidana S, Kurtz DM. Deciphering response dynamics and treatment resistance from circulating tumor DNA after CAR T-cells in multiple myeloma. Nat Commun 2025; 16:1824. [PMID: 39979252 PMCID: PMC11842827 DOI: 10.1038/s41467-025-56486-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 01/15/2025] [Indexed: 02/22/2025] Open
Abstract
Despite advances in treatments, multiple myeloma (MM) remains an incurable cancer where relapse is common. We developed a circulating tumor DNA (ctDNA) approach in order to characterize tumor genomics, monitor treatment response, and detect early relapse in MM. By sequencing 412 specimens from 64 patients with newly diagnosed or relapsed/refractory disease, we demonstrate the correlation between ctDNA and key clinical biomarkers, as well as patient outcomes. We further extend our approach to simultaneously track CAR-specific cell-free DNA (CAR-cfDNA) in patients undergoing anti-BCMA CAR T-cell (BCMA-CAR) therapy. We demonstrate that ctDNA levels following BCMA-CAR inversely correlate with relative time to progression (TTP), and that measurable residual disease (MRD) quantified by peripheral blood ctDNA (ctDNA-MRD) was concordant with clinical bone marrow MRD. Finally, we show that ctDNA-MRD can anticipate clinical relapse and identify the emergence of genomically-defined therapy-resistant clones. These findings suggest multiple clinical uses of ctDNA for MM in molecular characterization and disease surveillance.
Collapse
Affiliation(s)
- Hitomi Hosoya
- Division of Blood and Marrow Transplant and Cell Therapy, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Mia Carleton
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Kailee Tanaka
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Brian Sworder
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine CA, USA
| | - Shriya Syal
- Center for Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Bita Sahaf
- Center for Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Alisha M Maltos
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Oscar Silva
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Henning Stehr
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Vanna Hovanky
- Division of Blood and Marrow Transplant and Cell Therapy, Department of Medicine, Stanford University, Stanford, CA, USA
| | - George Duran
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Tian Zhang
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Michaela Liedtke
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Sally Arai
- Division of Blood and Marrow Transplant and Cell Therapy, Department of Medicine, Stanford University, Stanford, CA, USA
| | - David Iberri
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - David Miklos
- Division of Blood and Marrow Transplant and Cell Therapy, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Michael S Khodadoust
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Surbhi Sidana
- Division of Blood and Marrow Transplant and Cell Therapy, Department of Medicine, Stanford University, Stanford, CA, USA.
| | - David M Kurtz
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
2
|
Giles HV, Karunanithi K. Performance Characteristics and Limitations of the Available Assays for the Detection and Quantitation of Monoclonal Free Light Chains and New Emerging Methodologies. Antibodies (Basel) 2024; 13:19. [PMID: 38534209 PMCID: PMC10967543 DOI: 10.3390/antib13010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Light chain measurements form an essential component of the testing strategy for the detection and monitoring of patients with suspected and/or proven plasma cell disorders. Urine-based electrophoretic assays remain at the centre of the international guidelines for response assessment but the supplementary role of serum-free light chain (FLC) assays in response assessment and the detection of disease progression due to their increased sensitivity has been increasingly recognised since their introduction in 2001. Serum FLC assays have also been shown to be prognostic across the spectrum of plasma cell disorders and are now incorporated into risk stratification scores for patients with monoclonal gammopathy of undetermined significance (MGUS), smouldering multiple myeloma, and light chain amyloidosis (AL amyloidosis), as well as being incorporated into the criteria for defining symptomatic multiple myeloma. There are now multiple different commercially available serum FLC assays available with differing performance characteristics, which are discussed in this review, along with the implications of these for patient monitoring. Finally, newer methodologies for the identification and characterisation of monoclonal FLC, including modifications to electrophoretic techniques, mass spectrometry-based assays and Amylite, are also described along with the relevant published data available regarding the performance of each assay.
Collapse
Affiliation(s)
- Hannah V. Giles
- Department of Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2SY, UK
- Instute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Kamaraj Karunanithi
- Department of Clinical Haematology, University Hospitals North Midlands NHS Trust, Royal Stoke Hospital, Newcastle Road, Stoke-on-Trent ST4 6QG, UK;
- School of Medicine, Keele University, Keele, Newcastle-under-Lyme ST5 5BG, UK
| |
Collapse
|
3
|
Westhrin M, Blazevski J, Textor A, Abdollahi P, Gopalakrishnan RP, Ngo LT, Hofgaard PO, Heinzelbecker J, Bobic S, Fossum E, Spång HCL, Braathen R, Bogen B. Id-neoantigen vaccine induces therapeutic CD8 + T cells against multiple myeloma: H chain-loss escapees cause FLC MM. J Immunother Cancer 2023; 11:e006944. [PMID: 37607769 PMCID: PMC10445383 DOI: 10.1136/jitc-2023-006944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) cancers originate from plasma cells that have passed through the germinal center reaction where somatic hypermutation of Ig V regions takes place. Myeloma protein V regions often express many mutations and are thus a rich source of neoantigens (traditionally called idiotopes (Id)). Therefore, these are highly tumor-specific and excellent targets for immunotherapy. METHODS We have developed a DNA Id vaccine which as translated protein targets conventional dendritic cells (cDC) for CCL3-mediated delivery of myeloma protein V regions in a single-chain fragment variable (scFv) format. Vaccine efficacy was studied in the mouse MM model, mineral oil-induced plasmacytoma 315.BM. RESULTS The Id vaccine protected mice against a challenge with MM cells. Moreover, the vaccine had a therapeutic effect. However, in some of the vaccinated mice, MM cells not producing H chains escaped rejection, resulting in free light chain (FLC) MM. Depletion of CD8+ T cells abrogated vaccine efficacy, and protection was observed to be dependent on cDC1s, using Batf3-/- mice. Modifications of scFv in the vaccine demonstrated that CD8+ T cells were specific for two mutated VH sequences. CONCLUSIONS VH neoantigen-specific CD8+ T cells elicited by CCL3-containing Id vaccines had a therapeutic effect against MM in a mouse model. MM cells could escape rejection by losing expression of the H chain, thus giving rise to FLC MM.
Collapse
Affiliation(s)
- Marita Westhrin
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jana Blazevski
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ana Textor
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Pegah Abdollahi
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | | | - Linda Thuy Ngo
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Peter Olaf Hofgaard
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Julia Heinzelbecker
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sonja Bobic
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Even Fossum
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | | | - Ranveig Braathen
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Bjarne Bogen
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
4
|
Jia X, Liu X, Yang W. Predictive Value Analysis of Serum Ig A, Ig G, and TNF- α in Recurrence of Multiple Myeloma. DISEASE MARKERS 2022; 2022:2095696. [PMID: 36277989 PMCID: PMC9581636 DOI: 10.1155/2022/2095696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/23/2022] [Accepted: 09/30/2022] [Indexed: 09/07/2024]
Abstract
Objective The study is aimed at analyzing the predictive value of serum Ig A, Ig G, and TNF-α in the recurrence of multiple myeloma (MM). Methods 136 patients with MM treated in our hospital from January 2010 to January 2017 were followed up for 5 years. Finally, 100 patients who met the inclusion and exclusion criteria and had the complete follow-up visit were selected as the study subjects, with the recurrence of MM as endpoint event, and the observation was taken until the occurrence of endpoint event in patients or the termination of this study. They were divided into the recurrence group (RG) and the nonrecurrence group (NRG) according to whether the endpoint event occurred. The venous blood of patients was collected at the first diagnosis and subsequent visit (at the time of recurrence or termination of the study) to measure the Ig A and Ig G using a full automatic special protein analyzer and the TNF-α level by enzyme-linked immunosorbent assay. The data obtained in this study were analyzed by univariate analysis to choose the factors with difference in statistical significance to draw the ROC curve, and the areas under the curve (AUC) were recorded to analyze the potential mechanism of Ig A, Ig G, and TNF-α in predicting the recurrence of MM. Results After follow-up visit, there were 62 patients with recurrence (62.0%) and 38 patients without recurrence (38.0%), with no obvious difference in gender, age, body weight, and immune classification between the two groups (P > 0.05). Compared with the NRG, the levels of soluble interleukin-2 receptor (sIL-2R) and β 2-microglobulin (β 2-MG) in the RG at the first diagnosis were distinctly higher (P < 0.001); the levels of Ig A, Ig G, and TNF-α in the RG at the first diagnosis were visibly higher (P < 0.05); and the levels of Ig A, Ig G, and TNF-α in the RG at the subsequent visit were clearly higher (P < 0.05). There was a correlation between Ig G, Ig A, and TNF-α and β 2-MG at the first diagnosis and the subsequent visit (P < 0.05); there was a correlation between Ig G and TNF-α, and sIL-2R at the first diagnosis and the subsequent visit (P < 0.05); and there was a correlation between Ig A and sIL-2R at the subsequent visit (P < 0.05). The AUC of Ig G, Ig A, and TNF-α in predicting the MM at the first diagnosis were 0.772, 0.776, and 0.778, respectively. Conclusion The serum Ig A, Ig G, and TNF-α had a predictive value in the recurrence of MM, and TNF-α was correlated with sIL-2R and β 2-MG, with the highest AUC and the best predictive value.
Collapse
Affiliation(s)
- Xinyan Jia
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiangxin Liu
- Department of Hematology, Ji'an Hospital of Shanghai East Hospital, Ji'an, 343000 Jiangxi, China
| | - Wenzhong Yang
- Department of Hematology, Shanghai Punan Hospital of Pudong New District, Shanghai 200125, China
| |
Collapse
|
5
|
Bujarski S, Sutanto C, Spektor TM, To J, Swift RA, Green T, Eades BR, Emamy-Sadr M, Souther E, Berenson JR. Use of serum B-cell maturation antigen levels to predict outcomes for myeloma patients treated with ruxolitinib, lenalidomide and methylprednisolone. Hematol Oncol 2022; 40:243-248. [PMID: 34982491 DOI: 10.1002/hon.2961] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 01/14/2023]
Abstract
Previous retrospective studies have shown that serum B-cell maturation antigen (sBCMA) levels predict outcomes among patients with multiple myeloma (MM) undergoing new treatments. Specifically, baseline levels and changes during treatment of this protein predict both progression free survival (PFS) and overall survival. However, prospective studies are lacking evaluating sBCMA for determining outcomes among MM patients undergoing new treatments. Thus, we evaluated whether its baseline levels and changes during treatment in the amount of this serum marker predict outcomes among 38 relapsed/refractory MM patients treated with ruxolitinib, lenalidomide and methylprednisolone in a phase 1 trial. Patients with baseline sBCMA levels in the lowest three quartiles had longer PFS (median PFS 136 vs. 28 days; p < 0.0001). This was also shown for patients with baseline levels below the median (median PFS 140 vs. 77 days; p = 0.0225). PFS was shorter for patients whose sBCMA levels increased ≥25% through their first cycle (median PFS: 50 vs. 134 days, p = 0.0022), second cycle (median PFS: 50 vs. 141 days, p = 0.0273), and during the first three cycles of study treatment (median PFS: 50 vs. 220 days, p < 0.0001). No patient whose sBCMA increased ≥25% during cycle 1 responded whereas the majority (58%) of patients whose level increased <25% responded. This is the first prospective study to determine whether sBCMA levels predict outcomes for MM patients undergoing a non-BCMA directed treatment regimen and demonstrates that baseline levels and its changes during treatment predict PFS and the likelihood of responding to their treatment. These results add to the growing literature suggesting that this serum marker will be useful for determining outcomes for patients undergoing treatment for MM.
Collapse
Affiliation(s)
- Sean Bujarski
- James R. Berenson, MD, Inc., West Hollywood, California, USA
| | | | | | - Jennifer To
- Oncotherapeutics, West Hollywood, California, USA
| | - Regina A Swift
- James R. Berenson, MD, Inc., West Hollywood, California, USA
| | - Tracy Green
- James R. Berenson, MD, Inc., West Hollywood, California, USA
| | | | | | | | - James R Berenson
- James R. Berenson, MD, Inc., West Hollywood, California, USA.,OncoTracker, West Hollywood, California, USA.,Oncotherapeutics, West Hollywood, California, USA.,Institute for Myeloma and Bone Cancer Research, West Hollywood, California, USA
| |
Collapse
|
6
|
Markovic U, Romano A, Bellofiore C, Condorelli A, Garibaldi B, Bulla A, Duminuco A, Del Fabro V, Di Raimondo F, Conticello C. Role of Serum Free Light Chain Assay in Relapsed/Refractory Multiple Myeloma. A Real-Life Unicentric Retrospective Study. Cancers (Basel) 2021; 13:cancers13236017. [PMID: 34885127 PMCID: PMC8656731 DOI: 10.3390/cancers13236017] [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: 11/09/2021] [Revised: 11/24/2021] [Accepted: 11/27/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND In the era of novel drugs a growing number of multiple myeloma (MM) patients are treated until disease progression. Serum free light chain (sFLC) assay is recommended for disease monitoring in oligo-secretory and micromolecular MM. METHODS In this real-life survey, a total of 130 relapsed/refractory MM patients treated at our center with at least three lines were investigated as a retrospective cohort. RESULTS The median age at diagnosis was 64 years and more than half of patients were male. A total of 24 patients (18%) had oligo-secretory/micromolecular disease at diagnosis. More than 20% of 106 normo-secretory patients had oligo-secretory/micromolecular escape. In order to evaluate potential role of sFLC assay before ("pre") and after ("post") every treatment line, involved serum free light chain values (iFLC) less than 138 mg/mL and serum free light chain ratios (FLCr) <25 were identified by using ROC curve analysis. The analysis of the entire cohort throughout four treatment lines demonstrated a statistically significant negative impact on progression-free survival (PFS) for both involved pre-sFLC and its ratio (respectively p = 0.0086 and p = 0.0065). Furthermore, both post-iFLC and post-FLCr greater than the pre-established values had a negative impact on PFS of the study cohort; respectively, p = 0.014 and p = 0.0079. Odds ratio analysis evidenced that patients with both involved post-sFLC greater than 138 mg/mL and post-FLCr above 25 at disease relapse had a higher probability of having clinical relapse (respectively p = 0.026 and p = 0.006). CONCLUSIONS Alterations of sFLC values, namely iFLC and FLCr, both prior to treatment initiation and in the course of therapy at every treatment line, could be of aid in relapse evaluation and treatment outcome. We therefore suggest close periodical monitoring of sFLC assay, independently from secretory status.
Collapse
Affiliation(s)
- Uros Markovic
- Division of Hematology, Azienda Policlinico-OVE, University of Catania, 95124 Catania, Italy; (A.R.); (C.B.); (A.C.); (B.G.); (A.B.); (A.D.); (V.D.F.); (F.D.R.); (C.C.)
- Oncohematology and BMT Unit, Mediterranean Institute of Oncology, 95029 Viagrande, Italy
- Correspondence:
| | - Alessandra Romano
- Division of Hematology, Azienda Policlinico-OVE, University of Catania, 95124 Catania, Italy; (A.R.); (C.B.); (A.C.); (B.G.); (A.B.); (A.D.); (V.D.F.); (F.D.R.); (C.C.)
- Postgraduate School of Hematology, University of Catania, 95124 Catania, Italy
| | - Claudia Bellofiore
- Division of Hematology, Azienda Policlinico-OVE, University of Catania, 95124 Catania, Italy; (A.R.); (C.B.); (A.C.); (B.G.); (A.B.); (A.D.); (V.D.F.); (F.D.R.); (C.C.)
- Postgraduate School of Hematology, University of Catania, 95124 Catania, Italy
| | - Annalisa Condorelli
- Division of Hematology, Azienda Policlinico-OVE, University of Catania, 95124 Catania, Italy; (A.R.); (C.B.); (A.C.); (B.G.); (A.B.); (A.D.); (V.D.F.); (F.D.R.); (C.C.)
- Postgraduate School of Hematology, University of Catania, 95124 Catania, Italy
| | - Bruno Garibaldi
- Division of Hematology, Azienda Policlinico-OVE, University of Catania, 95124 Catania, Italy; (A.R.); (C.B.); (A.C.); (B.G.); (A.B.); (A.D.); (V.D.F.); (F.D.R.); (C.C.)
- Postgraduate School of Hematology, University of Catania, 95124 Catania, Italy
| | - Anna Bulla
- Division of Hematology, Azienda Policlinico-OVE, University of Catania, 95124 Catania, Italy; (A.R.); (C.B.); (A.C.); (B.G.); (A.B.); (A.D.); (V.D.F.); (F.D.R.); (C.C.)
| | - Andrea Duminuco
- Division of Hematology, Azienda Policlinico-OVE, University of Catania, 95124 Catania, Italy; (A.R.); (C.B.); (A.C.); (B.G.); (A.B.); (A.D.); (V.D.F.); (F.D.R.); (C.C.)
- Postgraduate School of Hematology, University of Catania, 95124 Catania, Italy
| | - Vittorio Del Fabro
- Division of Hematology, Azienda Policlinico-OVE, University of Catania, 95124 Catania, Italy; (A.R.); (C.B.); (A.C.); (B.G.); (A.B.); (A.D.); (V.D.F.); (F.D.R.); (C.C.)
| | - Francesco Di Raimondo
- Division of Hematology, Azienda Policlinico-OVE, University of Catania, 95124 Catania, Italy; (A.R.); (C.B.); (A.C.); (B.G.); (A.B.); (A.D.); (V.D.F.); (F.D.R.); (C.C.)
- Postgraduate School of Hematology, University of Catania, 95124 Catania, Italy
| | - Concetta Conticello
- Division of Hematology, Azienda Policlinico-OVE, University of Catania, 95124 Catania, Italy; (A.R.); (C.B.); (A.C.); (B.G.); (A.B.); (A.D.); (V.D.F.); (F.D.R.); (C.C.)
| |
Collapse
|
7
|
Klein EM, Tichy D, Salwender HJ, Mai EK, Duerig J, Weisel KC, Benner A, Bertsch U, Akhavanpoor M, Besemer B, Munder M, Lindemann HW, Hose D, Seckinger A, Luntz S, Jauch A, Elmaagacli A, Fuhrmann S, Brossart P, Goerner M, Bernhard H, Raab MS, Blau IW, Haenel M, Scheid C, Goldschmidt H, on behalf of the German-Speaking Myeloma Multicenter Group (GMMG). Prognostic Impact of Serum Free Light Chain Ratio Normalization in Patients with Multiple Myeloma Treated within the GMMG-MM5 Trial. Cancers (Basel) 2021; 13:cancers13194856. [PMID: 34638344 PMCID: PMC8507729 DOI: 10.3390/cancers13194856] [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: 08/31/2021] [Revised: 09/19/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary For multiple myeloma (MM) patients with measurable disease, there is no recommendation to monitor serum free light chains during therapy. However, this could provide important information in terms of prognosis. We investigated the prognostic impact of serum free light chain ratio (FLCr) normalization in 590 patients with secretory MM during first-line treatment within the German-Speaking Myeloma Multicenter Group MM5 trial. We are able to show that there is an increasing percentage of patients who achieve FLCr normalization during therapy. Importantly, we demonstrate that FLCr normalization at any time before the start of maintenance is significantly associated with prolonged progression-free and overall survival in multivariable time-dependent Cox regression analyses. This suggests that FLCr normalization during therapy is an important and simple way to assess prognostic factor in MM and supports the serial measurement of serum free light chains during therapy, even in patients with secretory MM. Abstract We investigated the prognostic impact of time-dependent serum free light chain ratio (FLCr) normalization in 590 patients with secretory multiple myeloma (MM) during first-line treatment within the German-Speaking Myeloma Multicenter Group MM5 trial. Serum free light chains (sFLC) were assessed by the Freelite test at baseline, after induction, mobilization, autologous blood stem cell transplantation, consolidation and every three months during maintenance or follow up within two years after the start of maintenance. The proportion of patients with a normal or normalized FLCr increased from 3.6% at baseline to 23.2% after induction and 64.7% after consolidation. The achievement of FLCr normalization at any one time before the start of maintenance was associated with significantly prolonged progression-free survival (PFS) (p < 0.01, hazard ratio (HR) = 0.61, 95% confidence interval (95% CI) = 0.47–0.79) and overall survival (OS) (p = 0.02, HR = 0.67, 95% CI = 0.48–0.93) in multivariable time-dependent Cox regression analyses. Furthermore, reaching immune reconstitution, defined as the normalization of uninvolved immunoglobulins, before maintenance was associated with superior PFS (p = 0.04, HR = 0.77, 95% CI = 0.60–0.99) and OS (p = 0.01, HR = 0.59, 95% CI = 0.41–0.86). We conclude that FLCr normalization during therapy is an important favorable prognostic factor in MM. Therefore, we recommend serial measurements of sFLC during therapy until achieving FLCr normalization, even in patients with secretory MM.
Collapse
Affiliation(s)
- Eva-Maria Klein
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, 69120 Heidelberg, Germany; (E.K.M.); (U.B.); (M.A.); (D.H.); (A.S.); (M.S.R.); (H.G.)
- Department of Internal Medicine 5, Klinikum Nuremberg, Paracelsus Medical University, 90419 Nuremberg, Germany
- Correspondence: ; Tel.: +49-911-398-114957
| | - Diana Tichy
- Division of Biostatistics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (D.T.); (A.B.)
| | - Hans J. Salwender
- Asklepios Tumorzentrum Hamburg, AK Altona and AK St. Georg, 22763 Hamburg, Germany;
| | - Elias K. Mai
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, 69120 Heidelberg, Germany; (E.K.M.); (U.B.); (M.A.); (D.H.); (A.S.); (M.S.R.); (H.G.)
| | - Jan Duerig
- Department of Hematology, University Clinic Essen, 45147 Essen, Germany;
| | - Katja C. Weisel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (D.T.); (A.B.)
| | - Uta Bertsch
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, 69120 Heidelberg, Germany; (E.K.M.); (U.B.); (M.A.); (D.H.); (A.S.); (M.S.R.); (H.G.)
- National Center for Tumor Diseases, 69120 Heidelberg, Germany
| | - Mabast Akhavanpoor
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, 69120 Heidelberg, Germany; (E.K.M.); (U.B.); (M.A.); (D.H.); (A.S.); (M.S.R.); (H.G.)
| | - Britta Besemer
- Department of Hematology, Oncology and Immunology, University Hospital Tübingen, 72076 Tübingen, Germany;
| | - Markus Munder
- Department of Internal Medicine III, University Medical Center Mainz, 55131 Mainz, Germany;
| | - Hans-Walter Lindemann
- Department of Hematology and Oncology, Katholisches Krankenhaus Hagen, 58097 Hagen, Germany;
| | - Dirk Hose
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, 69120 Heidelberg, Germany; (E.K.M.); (U.B.); (M.A.); (D.H.); (A.S.); (M.S.R.); (H.G.)
| | - Anja Seckinger
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, 69120 Heidelberg, Germany; (E.K.M.); (U.B.); (M.A.); (D.H.); (A.S.); (M.S.R.); (H.G.)
| | - Steffen Luntz
- Coordination Centre for Clinical Trials (KKS) Heidelberg, 69120 Heidelberg, Germany;
| | - Anna Jauch
- Institute of Human Genetics, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Ahmet Elmaagacli
- Department of Hematology and Oncology, Asklepios Hospital Hamburg St. Georg, 20099 Hamburg, Germany;
| | - Stephan Fuhrmann
- Department of Hematology and Oncology, Helios Hospital Berlin Buch, 13125 Berlin, Germany;
| | - Peter Brossart
- Department of Internal Medicine, Oncology, Hematology, Immuno-Oncology and Rheumatology/Clinical Immunology, University Hospital Bonn, 53127 Bonn, Germany;
| | - Martin Goerner
- Department of Hematology, Oncology and Palliative Care, Klinikum Bielefeld, 33604 Bielefeld, Germany;
| | - Helga Bernhard
- Internal Medicine V, Klinikum Darmstadt, 64283 Darmstadt, Germany;
| | - Marc S. Raab
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, 69120 Heidelberg, Germany; (E.K.M.); (U.B.); (M.A.); (D.H.); (A.S.); (M.S.R.); (H.G.)
| | - Igor W. Blau
- Medical Clinic, Charité University Medicine Berlin, 13353 Berlin, Germany;
| | - Mathias Haenel
- Department of Internal Medicine III, Klinikum Chemnitz, 09116 Chemnitz, Germany;
| | - Christof Scheid
- Department of Internal Medicine I, University Hospital Cologne, 50937 Cologne, Germany;
| | - Hartmut Goldschmidt
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, 69120 Heidelberg, Germany; (E.K.M.); (U.B.); (M.A.); (D.H.); (A.S.); (M.S.R.); (H.G.)
- National Center for Tumor Diseases, 69120 Heidelberg, Germany
| | | |
Collapse
|
8
|
Clonal Evolution of Multiple Myeloma-Clinical and Diagnostic Implications. Diagnostics (Basel) 2021; 11:diagnostics11091534. [PMID: 34573876 PMCID: PMC8469181 DOI: 10.3390/diagnostics11091534] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/22/2022] Open
Abstract
Plasma cell dyscrasias are a heterogeneous group of diseases characterized by the expansion of bone marrow plasma cells. Malignant transformation of plasma cells depends on the continuity of events resulting in a sequence of well-defined disease stages, from monoclonal gammopathy of undetermined significance (MGUS) through smoldering myeloma (SMM) to symptomatic multiple myeloma (MM). Evolution of a pre-malignant cell into a malignant cell, as well as further tumor progression, dissemination, and relapse, require development of multiple driver lesions conferring selective advantage of the dominant clone and allowing subsequent evolution under selective pressure of microenvironment and treatment. This process of natural selection facilitates tumor plasticity leading to the formation of genetically complex and heterogenous tumors that are notoriously difficult to treat. Better understanding of the mechanisms underlying tumor evolution in MM and identification of lesions driving the evolution from the premalignant clone is therefore a key to development of effective treatment and long-term disease control. Here, we review recent advances in clonal evolution patterns and genomic landscape dynamics of MM, focusing on their clinical implications.
Collapse
|
9
|
Prognostic impact of posttransplant FDG PET/CT scan in multiple myeloma. Blood Adv 2021; 5:2753-2759. [PMID: 34242392 DOI: 10.1182/bloodadvances.2020004131] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/22/2021] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma (MM) is a heterogeneous disease that may be evaluated by a broad array of imaging and laboratory techniques to measure disease activity and predict prognosis. Fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) scanning has been shown to be predictive of patient outcomes throughout the disease course. We sought to corroborate these findings by examining the prognostic impact of PET/CT scanning in the posttransplant setting. We retrospectively analyzed PET/CT scans in 229 MM patients receiving an autologous stem cell transplant (ASCT) near day 100, and correlated these findings with time to progression(TTP) and overall survival (OS) to assess the impact of day 100 PET/CT scan findings as an independent prognostic factor. The median OS for the entire cohort was 61.5 months (95% confidence interval [CI], 49-75) and the median TTP was 18.5 months (95% CI, 15.4-21.8). Among patients with abnormal day 100 PET findings (PET+), median TTP was 12.4 months vs 24 months among those with normal PET findings (PET-) (P < .0001). The median OS in the PET+ group was 46 months compared with 99 months in the PET- group (P < .0001). We conclude that an abnormal PET/CT scan near day 100 post-ASCT is predictive of shorter TTP and OS, with prognostic significance retained after adjusting for disease response and other prognostic variables in MM.
Collapse
|
10
|
Bujarski S, Udd K, Soof C, Chen H, Spektor TM, Safaie T, Li M, Stern J, Wang C, Xu N, Emamy-Sadr M, Swift R, Rahbari A, Patil S, Souther E, Regidor B, Sutanto C, Berenson JR. Baseline and Changes in Serum B-Cell Maturation Antigen Levels Rapidly Indicate Changes in Clinical Status Among Patients with Relapsed/Refractory Multiple Myeloma Starting New Therapy. Target Oncol 2021; 16:503-515. [PMID: 34097243 DOI: 10.1007/s11523-021-00821-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND B-cell maturation antigen (BCMA) is expressed on malignant plasma cells from patients with multiple myeloma (MM). These patients have higher levels of serum (s)BCMA than healthy subjects, and levels correlate with disease status. The half-life of sBCMA is only 24-36 h, and levels are independent of renal function. OBJECTIVE We determined whether baseline sBCMA values, a ≥ 25% increase, and a ≥ 50% decrease during treatment predicted progression-free survival (PFS) and overall survival (OS) among 81 patients with relapsed/refractory MM (RRMM) starting new treatments. METHODS Serum was obtained on day 22 of each patient's 28-day cycle of new therapy. Kaplan-Meier survival analysis and log-rank comparison tests were used to determine the effect of baseline sBCMA. The effect of percentage change in sBCMA was investigated using time-dependent Cox proportional hazard models. RESULTS Patients with baseline sBCMA levels above the median had a shorter PFS (p = 0.0077), and those in the highest quartile had a shorter PFS (p = 0.0012) and OS (p = 0.0022). A ≥ 25% increase at week 4, week 8, and anytime through week 12 predicted a shorter PFS (p = 0.0011, p = 0.0005, and p < 0.0001, respectively). A ≥ 50% decrease at week 4, week 8, and anytime through week 12 predicted a longer PFS (p = 0.0045, p = 0.029, p = 0.0055, respectively). A ≥ 25% increase in sBCMA occurred before progression according to International Myeloma Working Group criteria in 67.5% of patients. CONCLUSIONS Our results indicate the potential for the use of sBCMA as a new biomarker for monitoring patients with RRMM.
Collapse
Affiliation(s)
- Sean Bujarski
- Institute for Myeloma and Bone Cancer Research, 9201 W. Sunset Blvd., Ste. 300, West Hollywood, CA, 90069, USA
- James R Berenson MD, Inc., West Hollywood, CA, USA
| | - Kyle Udd
- Institute for Myeloma and Bone Cancer Research, 9201 W. Sunset Blvd., Ste. 300, West Hollywood, CA, 90069, USA
- James R Berenson MD, Inc., West Hollywood, CA, USA
| | | | - Haiming Chen
- Institute for Myeloma and Bone Cancer Research, 9201 W. Sunset Blvd., Ste. 300, West Hollywood, CA, 90069, USA
| | | | | | - Mingjie Li
- Institute for Myeloma and Bone Cancer Research, 9201 W. Sunset Blvd., Ste. 300, West Hollywood, CA, 90069, USA
| | - Joshua Stern
- Institute for Myeloma and Bone Cancer Research, 9201 W. Sunset Blvd., Ste. 300, West Hollywood, CA, 90069, USA
| | - Cathy Wang
- Institute for Myeloma and Bone Cancer Research, 9201 W. Sunset Blvd., Ste. 300, West Hollywood, CA, 90069, USA
| | - Ning Xu
- Institute for Myeloma and Bone Cancer Research, 9201 W. Sunset Blvd., Ste. 300, West Hollywood, CA, 90069, USA
| | | | - Regina Swift
- James R Berenson MD, Inc., West Hollywood, CA, USA
| | - Ashkon Rahbari
- Institute for Myeloma and Bone Cancer Research, 9201 W. Sunset Blvd., Ste. 300, West Hollywood, CA, 90069, USA
| | - Saurabh Patil
- Institute for Myeloma and Bone Cancer Research, 9201 W. Sunset Blvd., Ste. 300, West Hollywood, CA, 90069, USA
| | | | | | | | - James R Berenson
- Institute for Myeloma and Bone Cancer Research, 9201 W. Sunset Blvd., Ste. 300, West Hollywood, CA, 90069, USA.
- James R Berenson MD, Inc., West Hollywood, CA, USA.
- ONCOtherapuetics, West Hollywood, CA, USA.
- ONCOtracker, West Hollywood, CA, USA.
| |
Collapse
|
11
|
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: 7.6] [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
|
12
|
Markovic U, Leotta V, Tibullo D, Giubbolini R, Romano A, Del Fabro V, Parrinello NL, Cannizzaro MT, Di Raimondo F, Conticello C. Serum free light chains and multiple myeloma: Is it time to extend their application? Clin Case Rep 2020; 8:617-624. [PMID: 32274022 PMCID: PMC7141730 DOI: 10.1002/ccr3.2636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/14/2019] [Accepted: 12/01/2019] [Indexed: 02/06/2023] Open
Abstract
In nonsecretory, oligo-secretory, and light chain multiple myeloma patients, serial sFLC evaluation could precede biochemical and clinical disease progression, even in extramedullary relapse, thus initiating early treatment with novel anti-MM agents.
Collapse
Affiliation(s)
- Uros Markovic
- UOC di Ematologia con Trapianto di Midollo OsseoAOU "Policlinico‐Vittorio Emanuele”CataniaItaly
| | - Valerio Leotta
- UOC di Ematologia con Trapianto di Midollo OsseoAOU "Policlinico‐Vittorio Emanuele”CataniaItaly
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaCataniaItaly
| | - Rachele Giubbolini
- UOC di Ematologia con Trapianto di Midollo OsseoAOU "Policlinico‐Vittorio Emanuele”CataniaItaly
- Division of Hematology Largo del PozzoAOU PoliclinicoModenaItaly
| | - Alessandra Romano
- UOC di Ematologia con Trapianto di Midollo OsseoAOU "Policlinico‐Vittorio Emanuele”CataniaItaly
| | - Vittorio Del Fabro
- UOC di Ematologia con Trapianto di Midollo OsseoAOU "Policlinico‐Vittorio Emanuele”CataniaItaly
| | | | | | - Francesco Di Raimondo
- UOC di Ematologia con Trapianto di Midollo OsseoAOU "Policlinico‐Vittorio Emanuele”CataniaItaly
| | - Concetta Conticello
- UOC di Ematologia con Trapianto di Midollo OsseoAOU "Policlinico‐Vittorio Emanuele”CataniaItaly
| |
Collapse
|