Early peripheral blood and bone marrow MRD as prognostic markers in quadruplet-treated multiple myeloma without transplant

IF 14.6 2区 医学 Q1 HEMATOLOGY
HemaSphere Pub Date : 2025-08-19 DOI:10.1002/hem3.70186
Benjamin A. Derman, Jennifer Cooperrider, Anna Pula, Heidi Simmons, Tadeusz Kubicki, Jeffrey Zonder, Aimaz Afrough, David Grinblatt, Jacalyn Rosenblatt, Larry D. Anderson Jr., Andrew Kin, David Avigan, Sunil Narula, Shayan Rayani, Ken Jiang, Ajay Major, Theodore Karrison, Allison Jacob, Andrzej J. Jakubowiak
{"title":"Early peripheral blood and bone marrow MRD as prognostic markers in quadruplet-treated multiple myeloma without transplant","authors":"Benjamin A. Derman,&nbsp;Jennifer Cooperrider,&nbsp;Anna Pula,&nbsp;Heidi Simmons,&nbsp;Tadeusz Kubicki,&nbsp;Jeffrey Zonder,&nbsp;Aimaz Afrough,&nbsp;David Grinblatt,&nbsp;Jacalyn Rosenblatt,&nbsp;Larry D. Anderson Jr.,&nbsp;Andrew Kin,&nbsp;David Avigan,&nbsp;Sunil Narula,&nbsp;Shayan Rayani,&nbsp;Ken Jiang,&nbsp;Ajay Major,&nbsp;Theodore Karrison,&nbsp;Allison Jacob,&nbsp;Andrzej J. Jakubowiak","doi":"10.1002/hem3.70186","DOIUrl":null,"url":null,"abstract":"<p>Assessing measurable residual disease (MRD) in the peripheral blood (PB) of patients with multiple myeloma (MM) could mitigate issues of patchy and/or extramedullary disease leading to false negative MRD tests based on bone marrow (BM)-only assessments and reduce patient discomfort associated with serial BM sampling. Early disease response assessment using serum protein electrophoresis and immunofixation may not be able to account for interference with therapeutic monoclonal antibodies such as daratumumab (Dara) and elotuzumab (Elo), nor do they account for the extended half-lives of immunoglobulins due to recycling. Identification of a PB assay that better reflects a patient's response status compared to the conventional International Myeloma Working Group (IMWG) response criteria<span><sup>1</sup></span> could enable a more informative response-adapted treatment approach as part of decision-making regarding consolidative strategies following induction. PB MRD by flow cytometry has previously been found to be an early prognostic marker among patients receiving triplet induction and was shown to be prognostic when utilized much later in the maintenance setting.<span><sup>2, 3</sup></span></p><p>In this analysis, we investigated the potential role and prognostic significance of early MRD status after four cycles of carfilzomib-based quadruplet induction therapy by next-generation sequencing (NGS) in both the BM and PB as well as by conventional IMWG response criteria.</p><p>This was a post hoc analysis from two multicenter Phase II studies investigating the efficacy and safety of elotuzumab, carfilzomib, lenalidomide, and dexamethasone (Elo-KRd) (NCT02969837) and Dara-KRd (NCT 03500445) in patients with newly diagnosed MM without transplant intent.<span><sup>4, 5</sup></span></p><p>In both studies, a landmark analysis was performed after Cycle 4 (C4), which included MRD evaluation by clonoSEQ (Adaptive Biotechnologies, Seattle) NGS (limit of detection 6.8 × 10<sup>−7</sup> with input of 20 μg DNA) in BM and PB samples. MRD assessment by NGS was performed on PB or BM mononuclear cells for patients with available suitable material and with an identified dominant clonotype. MRD &lt; 10<sup>−6</sup> in the BM was considered MRD negative, or MRD &lt; 10<sup>−5</sup> if insufficient input to determine 10<sup>−6</sup> status. PB mononuclear cells were evaluated for MRD using the same assay; any quantifiable signal was considered positive in PB. Conventional response was assessed per IMWG response criteria.<span><sup>6</sup></span></p><p>This landmark modified intent-to-treat (ITT) analysis focused on patients who remained on treatment at the end of C4 and had samples available for analysis (Supporting Information S1: Figure S1). Paired IMWG response and MRD status in the PB and BM were assessed for agreement and prognostic significance at the end of C4. Differences between groups were examined using the chi-square or Fisher's exact test. Agreement between BM and PB MRD status was assessed by Cohen's kappa coefficient. The Kaplan–Meier method was used to assess progression-free survival (PFS) and overall survival (OS) from the start of protocol therapy. Hazard ratios (HRs) and corresponding 95% confidence intervals (CIs) were estimated with the use of Cox proportional hazards regression models, and where applicable, the Bonferroni method was applied to adjust for multiplicity testing. The data cutoff date for this analysis was July 20, 2024.</p><p>Among 62 patients with IMWG response status and either a C4 BM or PB MRD result, 53 had a BM MRD result and 57 had a PB MRD result (Supporting Information S1: Figure S1 and Table S1). In the landmark analysis group with a median follow-up of 50 months, there were 17 PFS events, including 8 deaths (7 from progression). There were no differences in PFS (log-rank P = 0.90) or OS (P = 0.31) between those receiving Elo-KRd and Dara-KRd.</p><p>Of the 62 patients evaluable at C4, 19 (31%) achieved at least a complete response (CR). CR status at C4 was not associated with PFS (HR 0.47, 95% CI 0.13–1.63, P = 0.24) nor OS (HR 0.79, 95% 0.16–3.93, P = 0.78) using the landmark method (Supporting Information S1: Figure S2); the same was true for best IMWG response. Among the 20 patients with IgG kappa paraprotein at baseline and in a VGPR at C4, no patient had an M-protein &gt; 0.1 g/L by mass spectrometry (the limit of detection for serum immunofixation), suggesting that interference from therapeutic monoclonal antibodies did not play a significant role in downgrading responses.</p><p>BM MRD samples by NGS at C4 were available in 53 patients, of whom 18 (34%) were MRD negative using the best available threshold (11 at 10<sup>−6</sup> and 7 at 10<sup>−5</sup>). Using the landmark method, BM MRD negativity at C4 was associated with superior PFS (HR 0.11, 95% CI 0.015–0.85; P = 0.034; Figure 1A) and OS (HR not estimable, log-rank P = 0.033; Figure 1B).</p><p>PB MRD samples by NGS at C4 were available in 57 patients, of whom 42 (74%) were MRD negative. The median input of cellular equivalents of PB mononuclear cells was 1.9 million cells; 49/57 samples reached 10<sup>−6</sup> sensitivity, and 8 reached 10<sup>−5</sup> sensitivity.</p><p>Using the landmark method, PB MRD positivity at C4 was associated with inferior PFS (HR 5.27, 95% CI 1.94–14.33, P = 0.001; Figure 1C) and OS (HR 7.74, 95% CI 1.49–40.14, P = 0.015; Figure 1D); this association remained significant in bivariate analyses that accounted for ISS and the presence of ≥1 HRCA (Supporting Information S1: Table S2). The 4-year PFS for patients with C4 PB MRD negativity was 83% (95% CI: 66%–92%) versus 40% (95% CI: 14%–65%) for C4 PB MRD positivity (P = 0.0053).</p><p>There were 57 patients with paired IMWG response and PB MRD status (Supporting Information S1: Table S3). Of the 15 patients with PB MRD positivity at C4, 4 (27%) patients were in ≥CR; 28/42 (67%) patients with PB MRD negativity at C4 were in a PR or VGPR. When assessing the combined prognostic impact of IMWG response and PB MRD status, we found a significant association with PFS (log-rank P = 0.0054) but not with OS (log-rank P = 0.13), mainly driven by worse outcomes among patients who were PB positive and &lt;CR (Figure 2A,B).</p><p>In comparing paired BM and PB MRD status (<i>n</i> = 48), there was only 52% agreement (Cohen's kappa 0.085, 95% CI 0.021–0.356). In total, 10/11 (91%) patients with PB MRD positivity at C4 also had BM MRD positivity; conversely, 22/37 (59%) patients with BM MRD positivity at C4 had PB MRD negativity (Supporting Information S1: Table S4 and Figures S3 and S4).</p><p>All 10 matched BM MRD-positive/PB MRD-positive samples at the C4 timepoint had higher concentrations of aberrant cells in the BM; BM MRD concentrations were a median log difference of 1.68 (range 0.07–3.52) higher than PB MRD concentrations.</p><p>When assessing for the combined prognostic impact of PB and BM MRD status, we found a significant association with PFS (log-rank P = 0.0012) and OS (log-rank P &lt; 0.001), mainly driven by worse outcomes in the PB/BM MRD-positive group (<i>n</i> = 10) compared to PB/BM MRD negative (<i>n</i> = 15) or only one positive (<i>n</i> = 23) (Figure 2C,D).</p><p>This is the first study to assess the prognostic impact of PB and BM MRD assays early during extended quadruplet regimens for MM in the absence of ASCT. Together, PB and BM MRD assessments at C4 offer two prognostic insights. C4 PB MRD positivity by clonoSEQ identified patients at high risk for early relapse and was more predictive of outcomes than conventional IMWG response criteria, while early attainment of BM MRD &lt; 10<sup>−6</sup> marked a trajectory of sustained MRD negativity and prolonged PFS and OS. These complementary insights underscore the value of incorporating both compartments in early MRD assessment.</p><p>Importantly, the results did not change in an intention-to-treat analysis that imputed missing C4 MRD data as positive. Reported MRD negativity rates should be interpreted in the context of a landmark analysis, which excluded patients who experienced early progression or death.</p><p>PB paraprotein-based assays, including serum protein electrophoresis, immunofixation, and even mass spectrometry, are limited in assessing early response due to delayed paraprotein clearance. In contrast, post-induction MRD testing in PB and BM using NGS provides a more immediate and highly prognostic assessment of disease burden, outperforming IMWG criteria. Moreover, PB MRD may also be valuable in contexts such as oligosecretory disease, extramedullary involvement, or post-chimeric antigen receptor (CAR) T-cell therapy as a less invasive alternative to serial BM biopsies or extensive imaging.</p><p>In summary, we demonstrate that both PB MRD and BM MRD status at C4, assessed by clonoSEQ, were independently prognostic for PFS and OS in frontline therapy for MM without transplant intent (Supporting Information S2: Visual Summary). In contrast, conventional IMWG response at the same timepoint lacked prognostic significance. These findings highlight the potential utility of early PB and BM MRD status to inform risk-adapted treatment strategies. BM MRD negativity after 4–6 cycles of induction—achieved in 23%–27% after four cycles of Dara-KRd or Isa-KRd<span><sup>7, 8</sup></span>; and in 50% after 6 cycles of Isa-KRd<span><sup>9</sup></span>—may justify deferring frontline ASCT in selected patients. This approach is supported by the MIDAS trial, where patients achieving MRD &lt; 10<sup>−5</sup> after induction had comparable rates of MRD &lt; 10<sup>−6</sup> following either ASCT or additional Isa-KRd.<span><sup>9</sup></span> Conversely, PB MRD positivity at C4 flags a high-risk group that may benefit from treatment intensification, including ASCT or even CAR T-cell therapy. While later MRD assessments by BM and/or emerging PB modalities such as mass spectrometry may guide therapy de-escalation in patients with deep responses, early MRD assessment in both PB and BM provides a valuable opportunity to identify patients in need of a more aggressive approach. Future trials should integrate PB MRD into response-adapted strategies to personalize treatment intensity and improve outcomes in MM.</p><p><b>Benjamin A. Derman</b>: Conceptualization; investigation; writing—original draft; methodology; writing—review and editing; formal analysis; data curation; supervision; project administration. <b>Jennifer Cooperrider</b>: Writing—review and editing; investigation; project administration. <b>Anna Pula</b>: Writing—review and editing; project administration; formal analysis; data curation; investigation. <b>Heidi Simmons</b>: Writing—review and editing; resources; methodology; investigation; conceptualization. <b>Tadeusz Kubicki</b>: Writing—review and editing; formal analysis; project administration; data curation; resources; investigation. <b>Jeffrey Zonder</b>: Writing—review and editing; investigation; resources; project administration. <b>Aimaz Afrough</b>: Writing—review and editing; investigation; project administration; resources. <b>David Grinblatt</b>: Writing—review and editing; investigation; project administration; resources. <b>Jacalyn Rosenblatt</b>: Resources; project administration; writing—review and editing; investigation. <b>Andrew Kin</b>: Resources; project administration; writing—review and editing; investigation. <b>David Avigan</b>: Investigation; writing—review and editing; project administration; resources. <b>Sunil Narula</b>: Resources; project administration; writing—review and editing; investigation. <b>Shayan Rayani</b>: Investigation; writing—review and editing; project administration; resources. <b>Ken Jiang</b>: Resources; data curation; investigation; methodology; writing—review and editing. <b>Ajay Major</b>: Writing—review and editing; investigation; resources; project administration. <b>Theodore Karrison</b>: Resources; project administration; writing—review and editing; investigation; conceptualization; methodology. <b>Allison Jacob</b>: Conceptualization; methodology; writing—review and editing; investigation; project administration; resources. <b>Andrzej J. Jakubowiak</b>: Conceptualization; investigation; writing—review and editing; resources; project administration; supervision.</p><p>B.A.D. declares consultancy for Janssen, Sanofi, COTA, and Canopy; independent reviewer of a clinical trial for BMS; research funding from Amgen and GSK. J.C., Ta.Ku., A.K., S.N., S.R., K.J., A.M., and Th.Ka declare no competing financial interests. A.P. declares honoraria from Janssen, Roche, and Amgen; research funding from Sanofi and GSK. H.S. and A.J. are employees of Adaptive Biotechnologies. J.Z. declares employment by BMS; advisory role for Takeda, Sanofi, and Regeneron, and research funding from BMS and Janssen. A.A. declares advisory roles for Karyopharm, BMS, Sanofi, Janssen, and Pfizer; research funding from AbbVie, Adaptive Biotechnologies, K36 Therapeutics, Janssen, and Regeneron. D.G. declares consultancy for BMS. J.R. declares advisory role for Janssen; data safety monitoring committee for Karyopharm; research funding from BMS and Sanofi. L.D.A. declares honoraria from GSK, Amgen, Janssen, Bristol Myers Squibb/Celgene, Prothena, AbbVie, BeiGene, Cellectar, and Sanofi; advisory roles for Janssen, Amgen, GSK, Bristol Myers Squibb/Celgene, AbbVie, Prothena, BeiGene, Cellectar, and Sanofi; research funding from Celgene, Janssen, and BMS. D.A. declares advisory roles for BMS, Takeda, Chugai, Kite Pharma, and Janssen. A.J.J. declares advisory roles for AbbVie, Amgen, BMS, Celgene, GSK, Gracell, Janssen, Karyopharm, and Sanofi.</p><p>The studies contained herein were approved by the Institutional Review Boards of each participating institution. All procedures involving human participants were conducted in accordance with the Declaration of Helsinki. Written informed consent was obtained from all participants.</p><p>This research received no funding.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 8","pages":""},"PeriodicalIF":14.6000,"publicationDate":"2025-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70186","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"HemaSphere","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/hem3.70186","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"HEMATOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Assessing measurable residual disease (MRD) in the peripheral blood (PB) of patients with multiple myeloma (MM) could mitigate issues of patchy and/or extramedullary disease leading to false negative MRD tests based on bone marrow (BM)-only assessments and reduce patient discomfort associated with serial BM sampling. Early disease response assessment using serum protein electrophoresis and immunofixation may not be able to account for interference with therapeutic monoclonal antibodies such as daratumumab (Dara) and elotuzumab (Elo), nor do they account for the extended half-lives of immunoglobulins due to recycling. Identification of a PB assay that better reflects a patient's response status compared to the conventional International Myeloma Working Group (IMWG) response criteria1 could enable a more informative response-adapted treatment approach as part of decision-making regarding consolidative strategies following induction. PB MRD by flow cytometry has previously been found to be an early prognostic marker among patients receiving triplet induction and was shown to be prognostic when utilized much later in the maintenance setting.2, 3

In this analysis, we investigated the potential role and prognostic significance of early MRD status after four cycles of carfilzomib-based quadruplet induction therapy by next-generation sequencing (NGS) in both the BM and PB as well as by conventional IMWG response criteria.

This was a post hoc analysis from two multicenter Phase II studies investigating the efficacy and safety of elotuzumab, carfilzomib, lenalidomide, and dexamethasone (Elo-KRd) (NCT02969837) and Dara-KRd (NCT 03500445) in patients with newly diagnosed MM without transplant intent.4, 5

In both studies, a landmark analysis was performed after Cycle 4 (C4), which included MRD evaluation by clonoSEQ (Adaptive Biotechnologies, Seattle) NGS (limit of detection 6.8 × 10−7 with input of 20 μg DNA) in BM and PB samples. MRD assessment by NGS was performed on PB or BM mononuclear cells for patients with available suitable material and with an identified dominant clonotype. MRD < 10−6 in the BM was considered MRD negative, or MRD < 10−5 if insufficient input to determine 10−6 status. PB mononuclear cells were evaluated for MRD using the same assay; any quantifiable signal was considered positive in PB. Conventional response was assessed per IMWG response criteria.6

This landmark modified intent-to-treat (ITT) analysis focused on patients who remained on treatment at the end of C4 and had samples available for analysis (Supporting Information S1: Figure S1). Paired IMWG response and MRD status in the PB and BM were assessed for agreement and prognostic significance at the end of C4. Differences between groups were examined using the chi-square or Fisher's exact test. Agreement between BM and PB MRD status was assessed by Cohen's kappa coefficient. The Kaplan–Meier method was used to assess progression-free survival (PFS) and overall survival (OS) from the start of protocol therapy. Hazard ratios (HRs) and corresponding 95% confidence intervals (CIs) were estimated with the use of Cox proportional hazards regression models, and where applicable, the Bonferroni method was applied to adjust for multiplicity testing. The data cutoff date for this analysis was July 20, 2024.

Among 62 patients with IMWG response status and either a C4 BM or PB MRD result, 53 had a BM MRD result and 57 had a PB MRD result (Supporting Information S1: Figure S1 and Table S1). In the landmark analysis group with a median follow-up of 50 months, there were 17 PFS events, including 8 deaths (7 from progression). There were no differences in PFS (log-rank P = 0.90) or OS (P = 0.31) between those receiving Elo-KRd and Dara-KRd.

Of the 62 patients evaluable at C4, 19 (31%) achieved at least a complete response (CR). CR status at C4 was not associated with PFS (HR 0.47, 95% CI 0.13–1.63, P = 0.24) nor OS (HR 0.79, 95% 0.16–3.93, P = 0.78) using the landmark method (Supporting Information S1: Figure S2); the same was true for best IMWG response. Among the 20 patients with IgG kappa paraprotein at baseline and in a VGPR at C4, no patient had an M-protein > 0.1 g/L by mass spectrometry (the limit of detection for serum immunofixation), suggesting that interference from therapeutic monoclonal antibodies did not play a significant role in downgrading responses.

BM MRD samples by NGS at C4 were available in 53 patients, of whom 18 (34%) were MRD negative using the best available threshold (11 at 10−6 and 7 at 10−5). Using the landmark method, BM MRD negativity at C4 was associated with superior PFS (HR 0.11, 95% CI 0.015–0.85; P = 0.034; Figure 1A) and OS (HR not estimable, log-rank P = 0.033; Figure 1B).

PB MRD samples by NGS at C4 were available in 57 patients, of whom 42 (74%) were MRD negative. The median input of cellular equivalents of PB mononuclear cells was 1.9 million cells; 49/57 samples reached 10−6 sensitivity, and 8 reached 10−5 sensitivity.

Using the landmark method, PB MRD positivity at C4 was associated with inferior PFS (HR 5.27, 95% CI 1.94–14.33, P = 0.001; Figure 1C) and OS (HR 7.74, 95% CI 1.49–40.14, P = 0.015; Figure 1D); this association remained significant in bivariate analyses that accounted for ISS and the presence of ≥1 HRCA (Supporting Information S1: Table S2). The 4-year PFS for patients with C4 PB MRD negativity was 83% (95% CI: 66%–92%) versus 40% (95% CI: 14%–65%) for C4 PB MRD positivity (P = 0.0053).

There were 57 patients with paired IMWG response and PB MRD status (Supporting Information S1: Table S3). Of the 15 patients with PB MRD positivity at C4, 4 (27%) patients were in ≥CR; 28/42 (67%) patients with PB MRD negativity at C4 were in a PR or VGPR. When assessing the combined prognostic impact of IMWG response and PB MRD status, we found a significant association with PFS (log-rank P = 0.0054) but not with OS (log-rank P = 0.13), mainly driven by worse outcomes among patients who were PB positive and <CR (Figure 2A,B).

In comparing paired BM and PB MRD status (n = 48), there was only 52% agreement (Cohen's kappa 0.085, 95% CI 0.021–0.356). In total, 10/11 (91%) patients with PB MRD positivity at C4 also had BM MRD positivity; conversely, 22/37 (59%) patients with BM MRD positivity at C4 had PB MRD negativity (Supporting Information S1: Table S4 and Figures S3 and S4).

All 10 matched BM MRD-positive/PB MRD-positive samples at the C4 timepoint had higher concentrations of aberrant cells in the BM; BM MRD concentrations were a median log difference of 1.68 (range 0.07–3.52) higher than PB MRD concentrations.

When assessing for the combined prognostic impact of PB and BM MRD status, we found a significant association with PFS (log-rank P = 0.0012) and OS (log-rank P < 0.001), mainly driven by worse outcomes in the PB/BM MRD-positive group (n = 10) compared to PB/BM MRD negative (n = 15) or only one positive (n = 23) (Figure 2C,D).

This is the first study to assess the prognostic impact of PB and BM MRD assays early during extended quadruplet regimens for MM in the absence of ASCT. Together, PB and BM MRD assessments at C4 offer two prognostic insights. C4 PB MRD positivity by clonoSEQ identified patients at high risk for early relapse and was more predictive of outcomes than conventional IMWG response criteria, while early attainment of BM MRD < 10−6 marked a trajectory of sustained MRD negativity and prolonged PFS and OS. These complementary insights underscore the value of incorporating both compartments in early MRD assessment.

Importantly, the results did not change in an intention-to-treat analysis that imputed missing C4 MRD data as positive. Reported MRD negativity rates should be interpreted in the context of a landmark analysis, which excluded patients who experienced early progression or death.

PB paraprotein-based assays, including serum protein electrophoresis, immunofixation, and even mass spectrometry, are limited in assessing early response due to delayed paraprotein clearance. In contrast, post-induction MRD testing in PB and BM using NGS provides a more immediate and highly prognostic assessment of disease burden, outperforming IMWG criteria. Moreover, PB MRD may also be valuable in contexts such as oligosecretory disease, extramedullary involvement, or post-chimeric antigen receptor (CAR) T-cell therapy as a less invasive alternative to serial BM biopsies or extensive imaging.

In summary, we demonstrate that both PB MRD and BM MRD status at C4, assessed by clonoSEQ, were independently prognostic for PFS and OS in frontline therapy for MM without transplant intent (Supporting Information S2: Visual Summary). In contrast, conventional IMWG response at the same timepoint lacked prognostic significance. These findings highlight the potential utility of early PB and BM MRD status to inform risk-adapted treatment strategies. BM MRD negativity after 4–6 cycles of induction—achieved in 23%–27% after four cycles of Dara-KRd or Isa-KRd7, 8; and in 50% after 6 cycles of Isa-KRd9—may justify deferring frontline ASCT in selected patients. This approach is supported by the MIDAS trial, where patients achieving MRD < 10−5 after induction had comparable rates of MRD < 10−6 following either ASCT or additional Isa-KRd.9 Conversely, PB MRD positivity at C4 flags a high-risk group that may benefit from treatment intensification, including ASCT or even CAR T-cell therapy. While later MRD assessments by BM and/or emerging PB modalities such as mass spectrometry may guide therapy de-escalation in patients with deep responses, early MRD assessment in both PB and BM provides a valuable opportunity to identify patients in need of a more aggressive approach. Future trials should integrate PB MRD into response-adapted strategies to personalize treatment intensity and improve outcomes in MM.

Benjamin A. Derman: Conceptualization; investigation; writing—original draft; methodology; writing—review and editing; formal analysis; data curation; supervision; project administration. Jennifer Cooperrider: Writing—review and editing; investigation; project administration. Anna Pula: Writing—review and editing; project administration; formal analysis; data curation; investigation. Heidi Simmons: Writing—review and editing; resources; methodology; investigation; conceptualization. Tadeusz Kubicki: Writing—review and editing; formal analysis; project administration; data curation; resources; investigation. Jeffrey Zonder: Writing—review and editing; investigation; resources; project administration. Aimaz Afrough: Writing—review and editing; investigation; project administration; resources. David Grinblatt: Writing—review and editing; investigation; project administration; resources. Jacalyn Rosenblatt: Resources; project administration; writing—review and editing; investigation. Andrew Kin: Resources; project administration; writing—review and editing; investigation. David Avigan: Investigation; writing—review and editing; project administration; resources. Sunil Narula: Resources; project administration; writing—review and editing; investigation. Shayan Rayani: Investigation; writing—review and editing; project administration; resources. Ken Jiang: Resources; data curation; investigation; methodology; writing—review and editing. Ajay Major: Writing—review and editing; investigation; resources; project administration. Theodore Karrison: Resources; project administration; writing—review and editing; investigation; conceptualization; methodology. Allison Jacob: Conceptualization; methodology; writing—review and editing; investigation; project administration; resources. Andrzej J. Jakubowiak: Conceptualization; investigation; writing—review and editing; resources; project administration; supervision.

B.A.D. declares consultancy for Janssen, Sanofi, COTA, and Canopy; independent reviewer of a clinical trial for BMS; research funding from Amgen and GSK. J.C., Ta.Ku., A.K., S.N., S.R., K.J., A.M., and Th.Ka declare no competing financial interests. A.P. declares honoraria from Janssen, Roche, and Amgen; research funding from Sanofi and GSK. H.S. and A.J. are employees of Adaptive Biotechnologies. J.Z. declares employment by BMS; advisory role for Takeda, Sanofi, and Regeneron, and research funding from BMS and Janssen. A.A. declares advisory roles for Karyopharm, BMS, Sanofi, Janssen, and Pfizer; research funding from AbbVie, Adaptive Biotechnologies, K36 Therapeutics, Janssen, and Regeneron. D.G. declares consultancy for BMS. J.R. declares advisory role for Janssen; data safety monitoring committee for Karyopharm; research funding from BMS and Sanofi. L.D.A. declares honoraria from GSK, Amgen, Janssen, Bristol Myers Squibb/Celgene, Prothena, AbbVie, BeiGene, Cellectar, and Sanofi; advisory roles for Janssen, Amgen, GSK, Bristol Myers Squibb/Celgene, AbbVie, Prothena, BeiGene, Cellectar, and Sanofi; research funding from Celgene, Janssen, and BMS. D.A. declares advisory roles for BMS, Takeda, Chugai, Kite Pharma, and Janssen. A.J.J. declares advisory roles for AbbVie, Amgen, BMS, Celgene, GSK, Gracell, Janssen, Karyopharm, and Sanofi.

The studies contained herein were approved by the Institutional Review Boards of each participating institution. All procedures involving human participants were conducted in accordance with the Declaration of Helsinki. Written informed consent was obtained from all participants.

This research received no funding.

Abstract Image

早期外周血和骨髓MRD作为未移植四肾治疗多发性骨髓瘤的预后指标
评估多发性骨髓瘤(MM)患者外周血(PB)中可测量的残留疾病(MRD)可以减轻斑块性和/或髓外疾病导致基于骨髓(BM)评估的MRD检测假阴性的问题,并减少患者与连续BM取样相关的不适。使用血清蛋白电泳和免疫固定进行早期疾病反应评估可能无法解释治疗性单克隆抗体(如达拉单抗(Dara)和埃妥珠单抗(Elo))的干扰,也无法解释由于循环使用而延长的免疫球蛋白半衰期。与传统的国际骨髓瘤工作组(IMWG)反应标准相比,确定一种能更好地反映患者反应状态的PB测定方法,可以使更有信息的反应适应治疗方法成为诱导后巩固策略决策的一部分。流式细胞术的PB MRD先前已被发现是接受三胞胎诱导的患者的早期预后标志物,并且在维持环境中较晚使用时显示出预后。2,3在这项分析中,我们通过下一代测序(NGS)在BM和PB中以及通过传统的IMWG反应标准研究了卡非zomib四联体诱导治疗四个周期后早期MRD状态的潜在作用和预后意义。这是一项来自两项多中心II期研究的事后分析,该研究调查了elotuzumab、carfilzomib、来那度胺和地塞米松(eloo - krd) (NCT02969837)和Dara-KRd (nct03500445)在新诊断的无移植意图的MM患者中的疗效和安全性。4,5在这两项研究中,在周期4 (C4)后进行了里程碑式的分析,包括克隆seq (Adaptive Biotechnologies, Seattle) NGS(检测限6.8 × 10−7,输入20 μg DNA)对BM和PB样品进行MRD评估。通过NGS对有合适材料和确定显性克隆型的患者的PB或BM单个核细胞进行MRD评估。BM的MRD &lt; 10−6被认为是MRD阴性,如果输入不足,则MRD &lt; 10−5无法确定10−6状态。使用相同的方法评估PB单个核细胞的MRD;任何可量化的信号都被认为是PB阳性。根据IMWG反应标准评估常规反应。6这项具有里程碑意义的修改意向治疗(ITT)分析侧重于在C4结束时仍在接受治疗并有样本可用于分析的患者(支持信息S1:图S1)。在C4结束时,评估PB和BM的配对IMWG反应和MRD状态的一致性和预后意义。组间差异采用卡方检验或费雪精确检验。用Cohen’s kappa系数评价BM和PB MRD状态的一致性。Kaplan-Meier方法用于评估方案治疗开始时的无进展生存期(PFS)和总生存期(OS)。使用Cox比例风险回归模型估计风险比(hr)和相应的95%置信区间(ci),并在适用的情况下使用Bonferroni方法进行多重检验调整。该分析的数据截止日期为2024年7月20日。在62例IMWG反应状态和C4 BM或PB MRD结果的患者中,53例为BM MRD结果,57例为PB MRD结果(支持信息S1:图S1和表S1)。在中位随访50个月的里程碑分析组中,有17例PFS事件,包括8例死亡(7例因进展)。Elo-KRd组和Dara-KRd组的PFS (log-rank P = 0.90)和OS (P = 0.31)差异无统计学意义。在62例可评估C4的患者中,19例(31%)达到至少完全缓解(CR)。采用地标性方法,C4处CR状态与PFS (HR 0.47, 95% CI 0.13-1.63, P = 0.24)和OS (HR 0.79, 95% 0.16-3.93, P = 0.78)无关(支持信息S1:图S2);对于IMWG的最佳响应也是如此。在20例基线和C4 VGPR检测IgG kappa副蛋白的患者中,通过质谱分析(血清免疫固定的检测限),没有患者检测到m蛋白&gt; 0.1 g/L,这表明治疗性单克隆抗体的干扰在降低应答中没有显著作用。53例患者在C4处获得了NGS的BM MRD样本,其中18例(34%)使用最佳可用阈值(10 - 6时11例,10 - 5时7例)为MRD阴性。使用地标法,C4部位BM MRD阴性与较好的PFS (HR 0.11, 95% CI 0.015-0.85; P = 0.034;图1A)和OS (HR不可估计,log-rank P = 0.033;图1B)相关。57例患者可获得C4处NGS的PB MRD样本,其中42例(74%)为MRD阴性。PB单个核细胞的细胞当量中位输入为1。 900万个细胞;49/57个样品灵敏度达到10−6,8个样品灵敏度达到10−5。使用地标法,C4处PB MRD阳性与较差的PFS (HR 5.27, 95% CI 1.94-14.33, P = 0.001;图1C)和OS (HR 7.74, 95% CI 1.49-40.14, P = 0.015;图1D)相关;在考虑ISS和HRCA≥1的双变量分析中,这种关联仍然显著(支持信息S1:表S2)。C4 PB MRD阴性患者的4年PFS为83% (95% CI: 66%-92%), C4 PB MRD阳性患者的4年PFS为40% (95% CI: 14%-65%) (P = 0.0053)。57例患者同时出现IMWG反应和PB MRD状态(支持信息S1:表S3)。15例C4期PB MRD阳性患者中,4例(27%)CR≥;28/42 (67%) C4部位PB MRD阴性的患者处于PR或VGPR。在评估IMWG反应和PB MRD状态对预后的综合影响时,我们发现与PFS (log-rank P = 0.0054)有显著相关性,但与OS无关(log-rank P = 0.13),这主要是由于PB阳性和CR患者的预后较差(图2A,B)。在比较配对的BM和PB MRD状态(n = 48)时,只有52%的一致性(Cohen’s kappa 0.085, 95% CI 0.021-0.356)。总的来说,10/11 (91%)C4处PB MRD阳性的患者同时也有BM MRD阳性;相反,22/37 (59%)C4部位BM MRD阳性的患者PB MRD阴性(支持信息S1:表S4和图S3和S4)。在C4时间点,所有10个匹配的BM mrd阳性/PB mrd阳性样本的BM中都有较高浓度的异常细胞;BM MRD浓度的中位对数差比PB MRD浓度高1.68(范围0.07-3.52)。当评估PB和BM MRD状态对预后的综合影响时,我们发现与PFS (log-rank P = 0.0012)和OS (log-rank P &lt; 0.001)有显著的关联,主要是由于PB/BM MRD阳性组(n = 10)的预后比PB/BM MRD阴性组(n = 15)或只有一个阳性组(n = 23)更差(图2C,D)。这是第一个评估在没有ASCT的情况下,在延长的四联体治疗方案中早期PB和BM MRD检测对MM预后影响的研究。总之,C4的PB和BM MRD评估提供了两种预后见解。clonoSEQ检测C4 PB MRD阳性可识别早期复发高风险患者,并且比常规IMWG反应标准更能预测预后,而早期达到BM MRD &lt; 10−6标志着持续MRD阴性和延长PFS和OS的发展趋势。这些互补的见解强调了在早期MRD评估中合并这两个部分的价值。重要的是,在将缺失的C4 MRD数据归为阳性的意向治疗分析中,结果没有改变。报告的MRD阴性率应在里程碑式分析的背景下进行解释,该分析排除了经历早期进展或死亡的患者。基于PB副蛋白的检测,包括血清蛋白电泳,免疫固定,甚至质谱,在评估由于副蛋白清除延迟的早期反应方面是有限的。相比之下,使用NGS对PB和BM进行诱导后MRD检测提供了更直接和高度预后的疾病负担评估,优于IMWG标准。此外,PB MRD在诸如少分泌性疾病、髓外受累性疾病或后嵌合抗原受体(CAR) t细胞治疗等情况下也有价值,可作为一系列BM活检或广泛成像的侵入性较小的替代方法。总之,我们证明C4的PB MRD和BM MRD状态,通过clonoSEQ评估,是无移植意图的MM一线治疗中PFS和OS的独立预后(支持信息S2:视觉总结)。相比之下,同一时间点的常规IMWG反应缺乏预后意义。这些发现强调了早期PB和BM MRD状态的潜在效用,可以为适应风险的治疗策略提供信息。4-6次诱导后BM MRD呈阴性,Dara-KRd或isa - krd4次诱导后达到23%-27% 7,8;在6个周期的isa - krd9治疗后,50%的患者可能有理由推迟一线ASCT治疗。该方法得到了MIDAS试验的支持,在该试验中,诱导后达到MRD &lt 10−5的患者与ASCT或额外的isa - krd的MRD &lt 10−6的比例相当相反,C4的PB MRD阳性标志着高危人群可能受益于强化治疗,包括ASCT甚至CAR - t细胞治疗。虽然BM和/或新兴的PB模式(如质谱)的后期MRD评估可以指导深度反应患者的治疗降级,但PB和BM的早期MRD评估为识别需要更积极治疗的患者提供了宝贵的机会。未来的试验应将PB MRD纳入适应反应的策略中,以个性化治疗强度并改善mm的预后。 德曼:概念化;调查;原创作品草案;方法;写作——审阅和编辑;正式的分析;数据管理;监督;项目管理。Jennifer Cooperrider:写作、评论和编辑;调查;项目管理。安娜·普拉:写作、评论和编辑;项目管理;正式的分析;数据管理;调查。海蒂·西蒙斯:写作、评论和编辑;资源;方法;调查;概念化。塔德乌什·库比基:写作、评论和编辑;正式的分析;项目管理;数据管理;资源;调查。Jeffrey Zonder:写作、评论和编辑;调查;资源;项目管理。Aimaz Afrough:写作-审查和编辑;调查;项目管理;资源。大卫·格林布拉特:写作、评论和编辑;调查;项目管理;资源。Jacalyn Rosenblatt:资源;项目管理;写作——审阅和编辑;调查。Andrew Kin:资源;项目管理;写作——审阅和编辑;调查。大卫·阿维根:调查;写作——审阅和编辑;项目管理;资源。Sunil Narula:资源;项目管理;写作——审阅和编辑;调查。Shayan Rayani:调查;写作——审阅和编辑;项目管理;资源。姜健:资源;数据管理;调查;方法;写作-审查和编辑。主修专业:文审编辑;调查;资源;项目管理。西奥多·卡里森:资源;项目管理;写作——审阅和编辑;调查;概念化;方法。Allison Jacob:概念化;方法;写作——审阅和编辑;调查;项目管理;资源。Andrzej J. Jakubowiak:概念化;调查;写作——审阅和编辑;资源;项目管理;supervision.B.A.D。杨森、赛诺菲、COTA和Canopy的申报顾问;BMS临床试验的独立审稿人;安进和葛兰素史克的研究经费。李鸿源Ta.Ku。, a.k., s.n., s.r., k.j., a.m.,和。我声明没有竞争的经济利益。美联社宣布从杨森、罗氏和安进获得酬金;赛诺菲和葛兰素史克的研究经费。h。s。和a。j。是适应性生物技术公司的雇员。J.Z.通过BMS申报就业;为武田、赛诺菲和Regeneron提供咨询服务,并获得BMS和杨森的研究资助。A.A.宣布担任Karyopharm、BMS、赛诺菲、杨森和辉瑞的顾问;AbbVie、Adaptive Biotechnologies、K36 Therapeutics、Janssen和Regeneron的研究资金。D.G.宣布为BMS提供咨询服务。J.R.宣布担任杨森公司的顾问;核制药数据安全监测委员会;BMS和赛诺菲的研究经费。L.D.A.宣布来自GSK、安进、杨森、百时美施贵宝/新基、Prothena、艾伯维、百济神州、Cellectar和赛诺菲的酬金;担任杨森、安进、葛兰素史克、百时美施贵宝/新基、艾伯维、Prothena、百济神州、Cellectar和赛诺菲的顾问;Celgene, Janssen和BMS的研究经费。D.A.宣布为BMS、武田、Chugai、Kite Pharma和Janssen担任顾问。A.J.J.宣布担任艾伯维、安进、BMS、Celgene、GSK、Gracell、杨森、Karyopharm和赛诺菲的顾问。本文所载的研究是由每个参与机构的机构审查委员会批准的。所有涉及人类参与者的程序都是按照《赫尔辛基宣言》进行的。所有参与者均获得书面知情同意。这项研究没有得到资助。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
HemaSphere
HemaSphere Medicine-Hematology
CiteScore
6.10
自引率
4.50%
发文量
2776
审稿时长
7 weeks
期刊介绍: HemaSphere, as a publication, is dedicated to disseminating the outcomes of profoundly pertinent basic, translational, and clinical research endeavors within the field of hematology. The journal actively seeks robust studies that unveil novel discoveries with significant ramifications for hematology. In addition to original research, HemaSphere features review articles and guideline articles that furnish lucid synopses and discussions of emerging developments, along with recommendations for patient care. Positioned as the foremost resource in hematology, HemaSphere augments its offerings with specialized sections like HemaTopics and HemaPolicy. These segments engender insightful dialogues covering a spectrum of hematology-related topics, including digestible summaries of pivotal articles, updates on new therapies, deliberations on European policy matters, and other noteworthy news items within the field. Steering the course of HemaSphere are Editor in Chief Jan Cools and Deputy Editor in Chief Claire Harrison, alongside the guidance of an esteemed Editorial Board comprising international luminaries in both research and clinical realms, each representing diverse areas of hematologic expertise.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信