Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Genomic and immune signatures predict clinical outcome in newly diagnosed multiple myeloma treated with immunotherapy regimens

Abstract

Despite improving outcomes, 40% of patients with newly diagnosed multiple myeloma treated with regimens containing daratumumab, a CD38-targeted monoclonal antibody, progress prematurely. By integrating tumor whole-genome and microenvironment single-cell RNA sequencing from upfront phase 2 trials using carfilzomib, lenalidomide and dexamethasone with daratumumab (NCT03290950), we show how distinct genomic drivers including high APOBEC mutational activity, IKZF3 and RPL5 deletions and 8q gain affect clinical outcomes. Furthermore, evaluation of paired bone marrow profiles, taken before and after eight cycles of carfilzomib, lenalidomide and dexamethasone with daratumumab, shows that numbers of natural killer cells before treatment, high T cell receptor diversity before treatment, the disappearance of sustained immune activation (that is, B cells and T cells) and monocyte expansion over time are all predictive of sustained minimal residual disease negativity. Overall, this study provides strong evidence of a complex interplay between tumor cells and the immune microenvironment that is predictive of clinical outcome and depth of treatment response in patients with newly diagnosed multiple myeloma treated with highly effective combinations containing anti-CD38 antibodies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Clinical impact of mutational signatures on PFS.
Fig. 2: Impact of recurrent copy number variants on PFS.
Fig. 3: Impact of recurrent large copy number variants and SVs on PFS.
Fig. 4: Landscape of the immune environment according to timepoint and response group.
Fig. 5: Analysis of NK subpopulations.
Fig. 6: Analysis of monocyte subpopulations.
Fig. 7: Impact of immune microenvironment on depth of response.

Similar content being viewed by others

Data availability

WGS and scRNA-seq data have been uploaded to EGA: EGAD00001011132. The Kydar scRNA-seq data are publicly available at the National Center for Biotechnology Information’s Gene Expression Omnibus (accession no. GSE161195). The CoMMpass data were downloaded from the MMRF researcher gateway portal (https://research.themmrf.org). CoMMpass raw data are accessible in dbgap: phs000748.v1.p1. Further information on research design is available in the Nature Research Reporting Summary linked to this article. Source data are provided with this paper.

Code availability

All the code used for the genomic and single-cell analyses has been uploaded to https://github.com/UM-Myeloma-Genomics and https://github.com/Eileen-Boyle/CITE-DKRD.

References

  1. Rajkumar, S. V. Multiple myeloma: 2020 update on diagnosis, risk-stratification and management. Am. J. Hematol. 95, 548–567 (2020).

    CAS  PubMed  Google Scholar 

  2. Shah, U. A. & Mailankody, S. Emerging immunotherapies in multiple myeloma. BMJ 370, m3176 (2020).

    PubMed  Google Scholar 

  3. Costa, L. J. et al. Daratumumab, carfilzomib, lenalidomide, and dexamethasone with minimal residual disease response-adapted therapy in newly diagnosed multiple myeloma. J. Clin. Oncol. 40, 2901–2912 (2022).

  4. Derman, B. A. et al. Elotuzumab and weekly carfilzomib, lenalidomide, and dexamethasone in patients with newly diagnosed multiple myeloma without transplant intent: a phase 2 measurable residual disease-adapted study. JAMA Oncol. 8,1278–1286 (2022).

  5. Diamond, B. et al. Dynamics of minimal residual disease in patients with multiple myeloma on continuous lenalidomide maintenance: a single-arm, single-centre, phase 2 trial. Lancet Haematol. 8, e422–e432 (2021).

    CAS  PubMed  Google Scholar 

  6. Kumar, S. et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 17, e328–e346 (2016).

    PubMed  Google Scholar 

  7. Facon, T. et al. Daratumumab plus lenalidomide and dexamethasone for untreated myeloma. N. Engl. J. Med. 380, 2104–2115 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Landgren, O. et al. Safety and effectiveness of weekly carfilzomib, lenalidomide, dexamethasone, and daratumumab combination therapy for patients with newly diagnosed multiple myeloma: the MANHATTAN nonrandomized clinical trial. JAMA Oncol. 7, 862–868 (2021).

    PubMed  PubMed Central  Google Scholar 

  9. Moreau, P. et al. Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study. Lancet 394, 29–38 (2019).

    CAS  PubMed  Google Scholar 

  10. San-Miguel, J. et al. Sustained minimal residual disease negativity in newly diagnosed multiple myeloma and the impact of daratumumab in MAIA and ALCYONE. Blood 139, 492–501 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Voorhees, P. M. et al. Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: the GRIFFIN trial. Blood 136, 936–945 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Kortum, K. M. et al. Targeted sequencing of refractory myeloma reveals a high incidence of mutations in CRBN and Ras pathway genes. Blood 128, 1226–1233 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Misund, K. et al. Clonal evolution after treatment pressure in multiple myeloma: heterogenous genomic aberrations and transcriptomic convergence. Leukemia 36, 1887–1897 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Rasche, L. et al. The spatio-temporal evolution of multiple myeloma from baseline to relapse-refractory states. Nat. Commun. 13, 4517 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Maura, F., Rustad, E. H., Boyle, E. M. & Morgan, G. J. Reconstructing the evolutionary history of multiple myeloma. Best Pract. Res. Clin. Haematol. 33, 101145 (2020).

    PubMed  PubMed Central  Google Scholar 

  16. Rustad, E. H. et al. Revealing the impact of structural variants in multiple myeloma. Blood Cancer Discov. 1, 258–273 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Bailur, J. K. et al. Early alterations in stem-like/resident T cells, innate and myeloid cells in the bone marrow in preneoplastic gammopathy. JCI Insight 5, e127807 (2019).

    PubMed  Google Scholar 

  18. Dhodapkar, K. M. et al. Changes in bone marrow tumor and immune cells correlate with durability of remissions following BCMA CAR T therapy in myeloma. Blood Cancer Discov. 3, 490–501 (2022).

  19. Zavidij, O. et al. Single-cell RNA sequencing reveals compromised immune microenvironment in precursor stages of multiple myeloma. Nat Cancer 1, 493–506 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Chattopadhyay, P. K., Gierahn, T. M., Roederer, M. & Love, J. C. Single-cell technologies for monitoring immune systems. Nat. Immunol. 15, 128–135 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Richardson, P. G. et al. Triplet therapy, transplantation, and maintenance until progression in myeloma. N. Engl. J. Med. 387, 132–147 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Maura, F. et al. A practical guide for mutational signature analysis in hematological malignancies. Nat. Commun. 10, 2969 (2019).

    PubMed  PubMed Central  Google Scholar 

  23. Rustad, E. H. et al. Timing the initiation of multiple myeloma. Nat. Commun. 11, 1917 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Maura, F. et al. Biological and prognostic impact of APOBEC-induced mutations in the spectrum of plasma cell dyscrasias and multiple myeloma cell lines. Leukemia 32,1044–1048 (2018).

  25. Walker, B. A. et al. APOBEC family mutational signatures are associated with poor prognosis translocations in multiple myeloma. Nat. Commun. 6, 6997 (2015).

    CAS  PubMed  Google Scholar 

  26. Maura, F. et al. Genomic landscape and chronological reconstruction of driver events in multiple myeloma. Nat. Commun. 10, 3835 (2019).

    PubMed  PubMed Central  Google Scholar 

  27. Walker, B. A. et al. Identification of novel mutational drivers reveals oncogene dependencies in multiple myeloma. Blood 132, 587–597 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Cohen, Y. C. et al. Identification of resistance pathways and therapeutic targets in relapsed multiple myeloma patients through single-cell sequencing. Nat. Med. 27, 491–503 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Leung-Hagesteijn, C. et al. Xbp1s-negative tumor B cells and pre-plasmablasts mediate therapeutic proteasome inhibitor resistance in multiple myeloma. Cancer Cell 24, 289–304 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Kronke, J. et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 343, 301–305 (2014).

    PubMed  Google Scholar 

  31. Gooding, S. et al. Multiple cereblon genetic changes are associated with acquired resistance to lenalidomide or pomalidomide in multiple myeloma. Blood 137, 232–237 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Ziccheddu, B. et al. Functional impact of genomic complexity on the transcriptome of multiple myeloma. Clin. Cancer Res. 27, 6479–6490 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Jain, M. D. et al. Tumor interferon signaling and suppressive myeloid cells are associated with CAR T-cell failure in large B-cell lymphoma. Blood 137, 2621–2633 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Casneuf, T. et al. Effects of daratumumab on natural killer cells and impact on clinical outcomes in relapsed or refractory multiple myeloma. Blood Adv. 1, 2105–2114 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Monaco, G. et al. RNA-seq signatures normalized by mRNA abundance allow absolute deconvolution of human immune cell types. Cell Rep. 26, 1627–1640.e1627 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Friedrich, M. J. et al. The pre-existing T cell landscape determines the response to bispecific T cell engagers in multiple myeloma patients. Cancer Cell 41, 711–725.e6 (2023).

  37. Dwivedi, A. K., Mallawaarachchi, I. & Alvarado, L. A. Analysis of small sample size studies using nonparametric bootstrap test with pooled resampling method. Stat. Med. 36, 2187–2205 (2017).

    PubMed  Google Scholar 

  38. Hofman, I. J. F. et al. RPL5 on 1p22.1 is recurrently deleted in multiple myeloma and its expression is linked to bortezomib response. Leukemia 31, 1706–1714 (2017).

    CAS  PubMed  Google Scholar 

  39. Walker, B. A. et al. A high-risk, double-hit, group of newly diagnosed myeloma identified by genomic analysis. Leukemia 33, 159–170 (2019).

    CAS  PubMed  Google Scholar 

  40. Gambella, M. et al. High XBP1 expression is a marker of better outcome in multiple myeloma patients treated with bortezomib. Haematologica 99, e14–e16 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Cohen, Y. C. et al. Identification of resistance pathways and therapeutic targets in relapsed multiple myeloma patients through single-cell sequencing. Nat. Med. 7, 491–503 (2021).

  42. Alizadeh, D. et al. IFNγ is critical for CAR T cell-mediated myeloid activation and induction of endogenous immunity. Cancer Discov. 11, 2248–2265 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Coffey, D. G. et al. Normalization of the immune microenvironment during lenalidomide maintenance is associated with sustained MRD negativity in patients with multiple myeloma. Blood 138, 329–329 (2021).

    Google Scholar 

  44. Bolzoni, M. et al. IL21R expressing CD14+CD16+ monocytes expand in multiple myeloma patients leading to increased osteoclasts. Haematologica 102, 773–784 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Viola, D. et al. Daratumumab induces mechanisms of immune activation through CD38+ NK cell targeting. Leukemia 35, 189–200 (2021).

    CAS  PubMed  Google Scholar 

  46. Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature 578, 94–101 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Rustad, E. H. et al. mmsig: a fitting approach to accurately identify somatic mutational signatures in hematological malignancies. Commun. Biol. 4, 424 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Stoeckius, M. et al. Cell hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 19, 224 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Yang, S. et al. Decontamination of ambient RNA in single-cell RNA-seq with DecontX. Genome Biol. 21, 57 (2020).

    PubMed  PubMed Central  Google Scholar 

  51. Spellerberg, I. F. & Fedor, P. J. A tribute to Claude Shannon (1916–2001) and a plea for more rigorous use of species richness, species diversity and the ‘Shannon–Wiener’Index. Glob. Ecol. Biogeogr. 12, 177–179 (2003).

  52. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Korotkevich, G. et al. Fast gene set enrichment analysis.Preprint at https://www.biorxiv.org/content/10.1101/060012v3

Download references

Acknowledgements

This work was supported by the Myeloma Solutions Fund (MSF), Paula and Rodger Riney Multiple Myeloma Research Program Fund, the Multiple Myeloma Research Foundation (MMRF), the Perelman Family Foundation, the Memorial Sloan Kettering Cancer Center National Cancer Institute (NCI) Core Grant (P30 CA 008748) and the Sylvester Comprehensive Cancer Center NCI Core Grant (P30 CA 240139). F.M. is supported by the American Society of Hematology, Leukemia & Lymphoma Society (LLS) and International Myeloma Society. K.M. is supported by the American Society of Hematology, the MMRF and the International Myeloma Society. G.J.M. received grant support through a Translational Research Program award from the Leukemia & Lymphoma Society (6020-20) and Myeloma Solution Fund. O.L. received grant support from the Paula and Rodger Riney Multiple Myeloma Research Program Fund, Myeloma Solutions Fund, the MMRF, the Perelman Family Foundation and the Sylvester Comprehensive Cancer Center NCI Core Grant (P30 CA 240139). Cell sorting and flow cytometry technologies were provided by NYU Langone’s Cytometry and Cell Sorting Laboratory, which is supported in part by grant P30CA016087 from the NCI. Single-cell sequencing was performed in NYU’s Genome Technology Center shared resource, partially supported by the Cancer Center Support Grant P30CA016087 at the Laura and Isaac Perlmutter Cancer Center. We particularly thank P. Meyn, Y. Zhang, C. Marier and E. Guzman. Cartoon figures were designed using BioRender.com. U.A.S. is supported by the American Society of Hematology Scholar Award, International Myeloma Society Career Development Award and NCI MSK Paul Calabresi Career Development Award for Clinical Oncology K12 CA184746.

Author information

Authors and Affiliations

Authors

Contributions

F.M., O.L. and G.J.M. designed and supervised the study, collected and analyzed the data, and contributed to the writing of the paper. E.M.B. designed the single-cell experiment, collected and analyzed the data, and contributed to the writing of the paper. D.C. and K.M. collected and analyzed the data and contributed to the writing of the paper. D.G., B.D., H.G., P.B., B.Z., A.C. and M.C. analyzed the data. Y.W., A.S., J.E.H., D.K., H.H., E.G., S.M., M.R., Q.G., U.S., C.T., M.H., M.S., G.S., H.L., D.J.C., S.G., Y.Z., A.A., A.D., A.M.L., F.E.D., S.U. and N.K. collected the data.

Corresponding authors

Correspondence to Francesco Maura, Gareth J. Morgan or Ola Landgren.

Ethics declarations

Competing interests

O.L. has received research funding from the National Institutes of Health (NIH), NCI, U.S. Food and Drug Administration, MMRF, International Myeloma Foundation, Leukemia and Lymphoma Society, Paula and Rodger Riney Myeloma Foundation, Tow Foundation, Myeloma Solutions Fund, Perelman Family Foundation, Rising Tide Foundation, Amgen, Celgene, Janssen, Takeda, Glenmark, Pfizer, Seattle Genetics and Karyopharm; has received honoraria from and/or served on advisory boards for Adaptive, Amgen, Binding Site, BMS, Celgene, Cellectis, Glenmark, Janssen, Juno and Pfizer; and serves on independent data monitoring committees for clinical trials lead by Takeda, Merck, Janssen and Theradex. G.J.M. has received funding from the NIH, NCI, MMRF, Leukemia and Lymphoma Society, Perelman Family Foundation, Amgen, Celgene, Janssen and Takeda; has received honoraria from and/or served on advisory boards for Adaptive, Amgen, BMS, Celgene and Janssen; and serves on independent data monitoring committees for clinical trials lead by Takeda, Karyopharm and Sanofi. M.S. has received clinical trial research support to the institution from Angiocrine Bioscience, Inc., Omeros Corporation and Amgen, Inc.; has provided consultancy services to Omeros Corporation, Angiocrine Bioscience, Inc. (past) and McKinsey & Company (past); has served on an ad hoc advisory board for Kite – A Gilead Company (past); and has received honoraria for CME activity from i3Health (past), Medscape, LLC. (past) and CancerNetwork. G.S. has received research funding to the institution from Janssen, Amgen, BMS, and Beyond Spring. DSMB for Arcellx. S.G. receives research funding from Miltenyi Biotec, Takeda Pharmaceutical Co., Celgene Corp., Amgen Inc., Sanofi, Johnson and Johnson, Inc. and Actinium Pharmaceuticals, Inc. and is on advisory boards for Kite Pharmaceuticals, Inc., Celgene Corp., Sanofi, Novartis, Johnson and Johnson, Inc., Amgen Inc., Takeda Pharmaceutical Co., Jazz Pharmaceuticals, Inc. and Actinium Pharmaceuticals, Inc. U.A.S. reports research funding support from Celgene/BMS and Janssen to the institution and nonfinancial research support; and personal fees from ACCC, MashUp MD, Janssen Biotech, Sanofi, BMS, MJH LifeSciences, Intellisphere, Phillips Gilmore Oncology Communications, i3 Health and RedMedEd outside the submitted work. S.U. has received research funding from Abbvie, Amgen, Array Biopharma, BMS, Celgene, Gilead, GSK, Janssen, Merck, Pharmacyclics, Sanofi, Seattle Genetics, SkylineDX and Takeda; and has had advisory and/or consulting roles with Abbvie, Amgen, BMS, Celgene, EdoPharma, Genentech, Gilead, GSK, Janssen, K36 Therapeutics, Moderna, Novartis, Oncopeptides, Sanofi, Seattle Genetics, SecuraBio, SkylineDX, Takeda and TeneoBio. A.D. has served as a consultant for Incyte, EUSA Pharma and Loxo and receives research support from Roche and Takeda. B.D. has received honoraria from and served on advisory boards for Janssen and Sanofi. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks Lawrence Boise, Jérôme Moreaux and Xuan Zhang for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Clinical features and outcomes.

Clinical impact of sex (a, Female n = 31, Male n = 18), Race (b, African Americans n = 4, White n = 38), age (c,  < 65 years n = 37; >65 years n = 12), Isotype (d, IgG n = 30; IgG 30; Light chain 9), and ISS (e, ISS1 n = 22, ISS2 n = 24, ISS3 n = 3). PFS: progression free survival; ISS: international scoring system.

Extended Data Fig. 2 Focal and large copy number aberrations associated with poor prognosis and non-sustained MRD-negativity.

a) Copy number cumulative plot for XBP1 deletions across the CoMMpass trial (n = 752) and the (D)KRd study (n = 60). B) Correlation between XBP1 and CD38 expression using CoMMpass newly diagnosed MM patients with available RNAseq data (n = 591). R2 and p-value were estimated using linear regression (lm R package). c-f) Copy number cumulative plot for CCSER1 deletion, large gains on 18q24, 17q22 and 8q. In a) and c-d) vertical black lines represent the GISTIC peak positions. g-h) Clinical impact of large gains on 4q (g) and 17q (h). In g-h) dark green always reflect the wild type (WT), while the dark yellow reflect the mutated cases.

Extended Data Fig. 3 Landscape of the immune environment according to time point and response group.

Projected cells on reference UMAP by time-point and response. Each UMAP has a comparable number of cells to the normal bone marrow reference. T1=time-point 1, T2=timepoint 2, n=number of cells, ref_UMAP= reference UMAP, WNN_UMAP= Weighted Nearest Neighbor Analysis derived UMAP, Mono= monocytes, NK=natural killer, DC=dendritic-cells. Visual differences can be appreciated such as the significant reduction in B and T-cell at T2 in patients who achieve MRD-negativity. n=represent number of cells.

Extended Data Fig. 4 Impact of number of cell to the cell type annotation.

Random sampling of a maximum of 500, 1000, 1500, 2000, 2500, 3000, and 3500 cells per sample (samples, n = 37). Two-sided p-values were estimated using Kruskal-Wallis test. Boxplots represent quartiles centered around the median.

Extended Data Fig. 5 Changes in cellular population across time and response.

Boxplot showing the differences in cellular populations between T1 (baseline) and T2 (post-therapy) in patients that achieve or fail to achieve sustained MRD-negativity. N1=sustained MRD-negativity group at T1 (n = 9), N2=sustained MRD-negativity group at T2 (n = 10), P1=no sustained MRD-negativity group at T1 (n = 8), P2=no sustained MRD-negativity group at T2 (n = 10). Two-sided p-values were estimated using Kruskal-Wallis test. Boxplots represent quartiles centered around the median. a. Memory B-cells b. Naïve B-cells. c. B1 Progenitor cells d. B2 progenitor cells e. Dendritic cell Progenitor. f. megakaryocytic progenitor g. lympho-myeloid progenitor. h. red blood cell progenitor, i. granulocyte-monocyte progenitor j. hematopoietic stem-cell. k. gamma/delta T-cell l. mucosae-associated T-cells m. Regulatory T-cells, n. CD4 effector 1, o. CD4 effector 2, p. CD8 memory 1, q. CD8 memory 2, r. CD8 naïve, s. CD4 memory. t. CD4 naïve u CD4/CD8 ratio, v. classical dendritic cells 2, w. Plasmacytoid dendritic cells.

Extended Data Fig. 6 Heatmap representation of the NK clusters.

Three NK clusters were identified: #0, #1, #2. T1=timepoint 1, T2=timepoint 2.

Extended Data Fig. 7 Shannon diversity and number of T-cell clones per sample identified.

a-b) Number of clones(a) and Shannon diversity (b) index across different groups and time pointes. N1=sustained MRD-negativity group at T1 (n = 9), N2=sustained MRD-negativity group at T2 (n = 10), P1=no sustained MRD-negativity group at T1 (n = 8), P2=no sustained MRD-negativity group at T2 (n = 10). Two-sided p-values were estimated using Kruskal-Wallis test. Boxplots represent quartiles centered around the median.

Extended Data Fig. 8 Monocytes and dendritic cell clustering highlight phenotypic changes.

a) Heatmap representation of the monocyte and dendritic cells. b) Differential expression between P1 and N1 monocytes. c) Pathway enrichment between P1 and N1 monocytes.

Extended Data Fig. 9 Pairwise comparison of overall populations (L1) and subpopulations (L2) of samples with both timepoints available.

P-values were estimated using Wilcoxon-paired test.

Extended Data Fig. 10 Identification of response associated immune-signatures.

Partition Around Medoids (PAM) bi-plot representation. Blue-lines indicate loading vectors (NK = NK cells, Mono=Monocytes, T = T-cells), Confidence ellipse. T1=first timepoint, T2=second timepoint).

Supplementary information

Supplementary Information

Flow sorting gating strategy.

Reporting Summary

Supplementary Table 1

Supplementary Tables 1–15.

Source data

Source Data

Raw data for main figures and extended data figures.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Maura, F., Boyle, E.M., Coffey, D. et al. Genomic and immune signatures predict clinical outcome in newly diagnosed multiple myeloma treated with immunotherapy regimens. Nat Cancer 4, 1660–1674 (2023). https://doi.org/10.1038/s43018-023-00657-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-023-00657-1

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer