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.

  • Resource
  • Published:

The multiple myeloma microenvironment is defined by an inflammatory stromal cell landscape

Abstract

Progression and persistence of malignancies are influenced by the local tumor microenvironment, and future eradication of currently incurable tumors will, in part, hinge on our understanding of malignant cell biology in the context of their nourishing surroundings. Here, we generated paired single-cell transcriptomic datasets of tumor cells and the bone marrow immune and stromal microenvironment in multiple myeloma. These analyses identified myeloma-specific inflammatory mesenchymal stromal cells, which spatially colocalized with tumor cells and immune cells and transcribed genes involved in tumor survival and immune modulation. Inflammatory stromal cell signatures were driven by stimulation with proinflammatory cytokines, and analyses of immune cell subsets suggested interferon-responsive effector T cell and CD8+ stem cell memory T cell populations as potential sources of stromal cell–activating cytokines. Tracking stromal inflammation in individuals over time revealed that successful antitumor induction therapy is unable to revert bone marrow inflammation, predicting a role for mesenchymal stromal cells in disease persistence.

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: Identification of myeloma-specific inflammatory MSCs.
Fig. 2: Distinct localization of CD44+ iMSCs in bone marrow.
Fig. 3: Potential activators of iMSCs.
Fig. 4: Activation recapitulates stromal inflammation.
Fig. 5: The myeloma immune microenvironment at single-cell resolution.
Fig. 6: Predicted interactions of iMSCs with myeloma cells.
Fig. 7: Predicted interactions in the myeloma microenvironment.
Fig. 8: Bone marrow inflammation persists after successful antitumor therapy.

Similar content being viewed by others

Data availability

The single-cell RNA sequencing datasets of the immune and non-hematopoietic bone marrow microenvironment and plasma cells of newly diagnosed individuals with MM and individuals without myeloma are available on ArrayExpress, no. E-MTAB-9139. The bulk RNA-sequencing datasets of the non-hematopoietic bone marrow microenvironment of individuals with MM at diagnosis and after induction treatment are available on ArrayExpress, no. E-MTAB-9285. Files summarizing receptor–ligand interactions as computed by CellphoneDB between the non-hematopoietic, CD38, CD38+SDC1 and CD38+SDC1+ datasets are available through GitHub (https://github.com/MyelomaRotterdam/Microenvironment). Source data are provided with this paper.

Code availability

The code generated during this study to analyze the single-cell datasets is available through GitHub at https://github.com/MyelomaRotterdam/Microenvironment.

References

  1. Scrivo, R., Vasile, M., Bartosiewicz, I. & Valesini, G. Inflammation as ‘common soil’ of the multifactorial diseases. Autoimmun. Rev. 10, 369–374 (2011).

    Article  PubMed  Google Scholar 

  2. Lomas, O. C., Tahri, S. & Ghobrial, I. M. The microenvironment in myeloma. Curr. Opin. Oncol. 32, 170–175 (2020).

    Article  PubMed  Google Scholar 

  3. Monteran, L. & Erez, N. The dark side of fibroblasts: cancer-associated fibroblasts as mediators of immunosuppression in the tumor microenvironment. Front. Immunol. 10, 1835 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Baryawno, N. et al. A cellular taxonomy of the bone marrow stroma in homeostasis and leukemia. Cell 177, 1915–1932 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Helbling, P. M. et al. Global transcriptomic profiling of the bone marrow stromal microenvironment during postnatal development, aging, and inflammation. Cell Rep. 29, 3313–3330 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Cassese, G. et al. Plasma cell survival is mediated by synergistic effects of cytokines and adhesion-dependent signals. J. Immunol. 171, 1684–1690 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Corre, J. et al. Bone marrow mesenchymal stem cells are abnormal in multiple myeloma. Leukemia 21, 1079–1088 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ghobrial, I. M., Detappe, A., Anderson, K. C. & Steensma, D. P. The bone-marrow niche in MDS and MGUS: implications for AML and MM. Nat. Rev. Clin. Oncol. 15, 219–233 (2018).

    Article  PubMed  Google Scholar 

  9. Das, R. et al. Microenvironment-dependent growth of preneoplastic and malignant plasma cells in humanized mice. Nat. Med. 22, 1351–1357 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  11. 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).

    Article  Google Scholar 

  12. Görgün, G. T. et al. Tumor-promoting immune-suppressive myeloid-derived suppressor cells in the multiple myeloma microenvironment in humans. Blood 121, 2975–2987 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Favaloro, J. et al. Myeloid derived suppressor cells are numerically, functionally and phenotypically different in patients with multiple myeloma. Leuk. Lymphoma 55, 2893–2900 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Nooka, A. K. et al. Daratumumab in multiple myeloma. Cancer 125, 2364–2382 (2019).

    Article  PubMed  Google Scholar 

  15. 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).

    Article  CAS  PubMed  Google Scholar 

  16. Kumar, S. K. et al. Multiple myeloma. Nat. Rev. Dis. Prim. 3, 17046 (2017).

    Article  PubMed  Google Scholar 

  17. Barrionuevo, N., Gatica, S., Olivares, P., Cabello-Verrugio, C. & Simon, F. Endothelial cells exhibit two waves of P-selectin surface aggregation under endotoxic and oxidative conditions. Protein J. 38, 667–674 (2019).

    Article  CAS  PubMed  Google Scholar 

  18. Barut, B. A. et al. Role of interleukin 6 in the growth of myeloma-derived cell lines. Leuk. Res. 16, 951–959 (1992).

    Article  CAS  PubMed  Google Scholar 

  19. Nishimoto, N. et al. Oncostatin M, leukemia inhibitory factor, and interleukin 6 induce the proliferation of human plasmacytoma cells via the common signal transducer, gp130. J. Exp. Med. 179, 1343–1347 (1994).

    Article  CAS  PubMed  Google Scholar 

  20. Alfaro, C. et al. Tumor-produced interleukin-8 attracts human myeloid-derived suppressor cells and elicits extrusion of neutrophil extracellular traps (NETs). Clin. Cancer Res. 22, 3924–3936 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Shi, H. et al. Chemokine (C-X-C motif) ligand 1 and CXCL2 produced by tumor promote the generation of monocytic myeloid-derived suppressor cells. Cancer Sci. 109, 3826–3839 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Schaeuble, K. et al. Attenuation of chronic antiviral T-cell responses through constitutive COX2-dependent prostanoid synthesis by lymph node fibroblasts. PLoS Biol. 17, e3000072 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Molejon, M. I. et al. Deciphering the cellular source of tumor relapse identifies CD44 as a major therapeutic target in pancreatic adenocarcinoma. Oncotarget 6, 7408–7423 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Yang, C. et al. Inducible formation of leader cells driven by CD44 switching gives rise to collective invasion and metastases in luminal breast carcinomas. Oncogene 38, 7113–7132 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Filippi, I. et al. Different adaptive responses to hypoxia in normal and multiple myeloma endothelial cells. Cell. Physiol. Biochem. 46, 203–212 (2018).

    Article  CAS  PubMed  Google Scholar 

  26. Kang, H. S. et al. CD44 plays a critical role in regulating diet-induced adipose inflammation, hepatic steatosis, and insulin resistance. PLoS ONE 8, e58417 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Szklarczyk, D. et al. STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47, D607–D613 (2019).

    Article  CAS  PubMed  Google Scholar 

  28. Hideshima, T., Chauhan, D., Schlossman, R., Richardson, P. & Anderson, K. C. The role of tumor necrosis factor α in the pathophysiology of human multiple myeloma: therapeutic applications. Oncogene 20, 4519–4527 (2001).

    Article  CAS  PubMed  Google Scholar 

  29. Morris, E. V. et al. Myeloma cells down-regulate adiponectin in bone marrow adipocytes via TNF-α. J. Bone Miner. Res. 35, 942–955 (2020).

    Article  CAS  PubMed  Google Scholar 

  30. Gars, M. L. et al. CD38 contributes to human natural killer cell responses through a role in immune synapse formation. Preprint at bioRxiv, https://doi.org/10.1101/349084 (2019).

  31. Efremova, M., Vento-Tormo, M., Teichmann, S. A. & Vento-Tormo, R. CellPhoneDB: inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes. Nat. Protoc. 15, 1484–1506 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Loberg, R. D. et al. CCL2 is a potent regulator of prostate cancer cell migration and proliferation. Neoplasia 8, 578–586 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Chen, X. et al. CCL2/CCR2 regulates the tumor microenvironment in HER-2/neu-driven mammary carcinomas in mice. PLoS ONE 11, e0165595 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Moll, G. et al. Mesenchymal stromal cells engage complement and complement receptor bearing innate effector cells to modulate immune responses. PLoS ONE 6, e21703 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Perretti, M. & D’Acquisto, F. Annexin A1 and glucocorticoids as effectors of the resolution of inflammation. Nat. Rev. Immunol. 9, 62–70 (2009).

    Article  CAS  PubMed  Google Scholar 

  36. Diakos, C. I., Charles, K. A., McMillan, D. C. & Clarke, S. J. Cancer-related inflammation and treatment effectiveness. Lancet Oncol. 15, e493–e503 (2014).

    Article  PubMed  Google Scholar 

  37. Rubartelli, A., Bajetto, A., Allavena, G., Cozzolino, F. & Sitia, R. Post-translational regulation of interleukin 1β secretion. Cytokine 5, 117–124 (1993).

    Article  CAS  PubMed  Google Scholar 

  38. Erez, N., Truitt, M., Olson, P. & Hanahan, D. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-κB-dependent manner. Cancer Cell 17, 135–147 (2010).

    Article  CAS  PubMed  Google Scholar 

  39. Tong, D. et al. The roles of the COX2/PGE2/EP axis in therapeutic resistance. Cancer Metastasis Rev. 37, 355–368 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Li, A., Chen, P., Leng, Y. & Kang, J. Histone deacetylase 6 regulates the immunosuppressive properties of cancer-associated fibroblasts in breast cancer through the STAT3–COX2-dependent pathway. Oncogene 37, 5952–5966 (2018).

    Article  CAS  PubMed  Google Scholar 

  41. Santi, A., Kugeratski, F. G. & Zanivan, S. Cancer associated fibroblasts: the architects of stroma remodeling. Proteomics 18, e1700167 (2018).

    Article  PubMed  CAS  Google Scholar 

  42. Miyai, Y., Esaki, N., Takahashi, M. & Enomoto, A. Cancer-associated fibroblasts that restrain cancer progression: hypotheses and perspectives. Cancer Sci. 111, 1047–1057 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Liu, T. et al. Cancer-associated fibroblasts: an emerging target of anti-cancer immunotherapy. J. Hematol. Oncol. 12, 86 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhang, H. et al. CXCL2/MIF–CXCR2 signaling promotes the recruitment of myeloid-derived suppressor cells and is correlated with prognosis in bladder cancer. Oncogene 36, 2095–2104 (2017).

    Article  CAS  PubMed  Google Scholar 

  45. Tobin, R. P. et al. IL-6 and IL-8 are linked with myeloid-derived suppressor cell accumulation and correlate with poor clinical outcomes in melanoma patients. Front. Oncol. 9, 1223 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Kumar, H., Kawai, T. & Akira, S. Pathogen recognition by the innate immune system. Int. Rev. Immunol. 30, 16–34 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. Amarante-Mendes, G. P. et al. Pattern recognition receptors and the host cell death molecular machinery. Front. Immunol. 9, 2379 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Hope, C. et al. Immunoregulatory roles of versican proteolysis in the myeloma microenvironment. Blood 128, 680–685 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Vulpis, E. et al. Genotoxic stress modulates the release of exosomes from multiple myeloma cells capable of activating NK cell cytokine production: role of HSP70/TLR2/NF-kB axis. Oncoimmunology 6, e1279372 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Ping, Z. et al. Activation of NF-κB driven inflammatory programs in mesenchymal elements attenuates hematopoiesis in low-risk myelodysplastic syndromes. Leukemia 33, 536–541 (2019).

    Article  CAS  PubMed  Google Scholar 

  51. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Waltman, L. & van Eck, N. J. A smart local moving algorithm for large-scale modularity-based community detection. Eur. Phys. J. B 86, 471 (2013).

    Article  CAS  Google Scholar 

  54. McInnes, L., Healy, J., Saul, N. & Grossberger, L. UMAP: uniform manifold approximation and projection. J. Open Source Softw. 3, 861 (2018).

    Article  Google Scholar 

  55. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  56. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Patro, R., Duggal, G., Love, M. I., Irizarry, R. A. & Kingsford, C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Soneson, C., Love, M. I. & Robinson, M. D. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res 4, 1521 (2015).

    Article  PubMed  CAS  Google Scholar 

  60. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Stephens, M. False discovery rates: a new deal. Biostatistics 18, 275–294 (2016).

    PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of Myeloma Research Rotterdam and the Department of Hematology for critical discussions and reading of the manuscript, and the participants, families and nurses for their contributions to this study. We thank N. Leimkühler (Department of Hematology, Erasmus MC, the Netherlands) for help with the collection of healthy bone marrow biopsies and E. Bindels (Department of Hematology, Erasmus MC, the Netherlands) for help with sequencing. This work was supported by grants from the European Myeloma Network (EMN, grant HO95-EMN02-FB01102018 to P.S.) and ZonMw (grant 95103008 to P.S.). C.K. is supported by the Jose Carreras Leukaemia Foundation (grant DJCLS 17R/2018), the Deutsche Krebshilfe (grant 70112392), Deutsche Forschungsgemeinschaft (grant KH331/2–3) and the intramural funding of the faculty of Medicine at University Hospital of Muenster (grant Kha2/002/20). This project received funding from the European Union’s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant agreement no. 707404 (to Z.K.). The opinions expressed in this document reflect only the authors’ views. The European Commission is not responsible for any use that may be made of the information it contains.

Author information

Authors and Affiliations

Authors

Contributions

T.C. and P.S. conceptualized the study. T.C. and M.D.J. were responsible for the study methodology. M.D.J., Z.K., N.P. and S.T. were responsible for study investigation. M.D.J., Z.K., N.P., S.T., R.H., M.S. and T.C. analyzed the data. D.H.O.B., P.V.D.W, P.K.B., C.K., J.V., P.M., M.V.D. and A.B. provided resources for the study. M.D.J., R.H. and M.S. curated the data. M.D.J. contributed to data visualization. M.D.J. and T.C. wrote the original draft, and all authors reviewed and edited the final manuscript. P.S. and Z.K. acquired funding. T.C., A.B. and P.S. supervised the study, and T.C. and P.S. were responsible for project administration.

Corresponding authors

Correspondence to Pieter Sonneveld or Tom Cupedo.

Ethics declarations

Competing interests

J.V. is an employee of Janssen; A.B. consults for Celgene, Janssen, Amgen and Takeda; P.S. consults for and receives research support from Celgene, Janssen, Karyopharm and SkylineDx; P.M. is on the advisory boards of and receives honoraria from Janssen, BMS/Celgene, Amgen and Abbvie. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks Nicola Giuliani, Shannon Turley and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Related to Figs. 1, 4 and 6.

(a) FACS gating strategy for total non-hematopoietic cells or bone marrow mesenchymal stromal cells from CD45-depleted bone marrow mononuclear cells (MNCs) from aspirates for bulk RNA sequencing and single-cell RNA sequencing experiments (b) FACS gating strategy of major immune cell populations in bone marrow aspirates for qPCR. (c) FACS gating strategy of CD38- and CD38+ hematopoietic cells from non-depleted bone marrow aspirate MNCs for single-cell RNA sequencing experiments.

Extended Data Fig. 2 Related to Fig. 1.

(a) Bar graph depicting relative abundance of non-hematopoietic cells of total bone marrow aspirates. Data points represent patients (myeloma, n = 40; treated, n = 16; control, n = 8) (b) Transcription of FN1 and DCN in non-hematopoietic cells (c) Violin plots depicting transcription of collagen genes in the 8 clusters of non-hematopoietic cells (d) Transcription of PDGFRA, PDGFRB and ACTA2 in non-hematopoietic cells (e) Transcription of CD36 and THBD in non-hematopoietic cells (f) Transcription of SP7 in non-hematopoietic cells (g) Circle diagrams depicting cluster fractions of total in control- and myeloma datasets (h) Bar plot depicting fraction of cells per cluster per patient (left Y-axis); line graph depicting total number of sequenced non-hematopoietic cells per patient (right Y-axis) (i) Bar graph depicting patient-specific iMSC-fraction of total mesenchymal stromal cells, segregated by R-ISS stage. Datapoints represent patients (R-ISS stage I, n = 4; II, n = 7; III, n = 2) (j) Spearman (two-tailed) correlation of iMSC-fraction of total mesenchymal stromal cells with percentage of CD38+CD138+ plasma cells in aspirates taken at diagnosis. Datapoints represent patients (k) Heatmap of top 50 differentially expressed genes up in MSC1/2 compared to MSC3/4/5 (left) or down in MSC1/2 compared to MSC3/4/5 (right) as described in Supplementary Table 5. The data in (a) and (i) are presented as mean ± standard error of the mean (SEM). Significance was calculated using Mann-Whitney U (two-tailed). NS; p > 0.05.

Source data

Extended Data Fig. 3 Related to Figs. 1 and 3.

(a) UMAP of the integrated dataset of 389 cells from freshly aspired bone marrow of 1 newly diagnosed myeloma patient showing 7 clusters identified by integrated analysis, accompanied by UMAPs showing transcription of CXCL12 (MSCs) and CD36 (endothelial cells). Colors represent clusters (b) Transcription of inflammatory genes in fresh non-hematopoietic cells shown in (a) (c) Spearman (two-tailed) correlation of iMSC-fraction of total mesenchymal stromal cells with inflammatory protein levels in bone marrow plasma. Datapoints represent patients (d) Monocle 3 pseudotime analysis of mesenchymal stromal cells in controls. ‘S’ represents arbitrary selected starting point of trajectory calculations.

Extended Data Fig. 4 Related to Figs. 2 and 4.

(a) Flow cytometric gating strategy to determine CD44 protein expression on CD271+ MSCs (b) FACS gating strategy of CD56dim, CD56bright and CD56brightCD27+ cells for qPCR (c) Flow cytometric gating strategy of CD38+CD3+CD8+CD45RA+CD27+FAS+ stem cell memory T cells, CD38+CD3-CD56bright NK cells and CD38+CD3-CD56brightCD27+ NK cells in bone marrow MNCs. Same sample as used for FACS in (b).

Extended Data Fig. 5 Related to Fig. 4.

(a) Transcription of NCR1, FCGR3A, IL4 and RORC in the CD38- dataset (b) Transcription of interferon-response genes IFIT1, IFIT2 and IFIT3 in the CD38- dataset (c) Transcription of FCGR3A in the CD38+ dataset (d) Transcription of CD27 in the CD38+ dataset (e) Transcription of GZMK and GZMB in NK cell subsets of the myeloma bone marrow (n = 5) as determined by qPCR. Data are presented as mean ± standard error of the mean (SEM). Significance was calculated using Mann-Whitney U (two-tailed). **; p ≤0.01 (f) Original UMAPs of CD38- dataset split by patient. CBM: non-myeloma control; PH: patient with hyperdiploid tumor; PT: patient with tumor harboring translocations (g) Original UMAPs of CD38+ dataset split by patient. CBM: non-myeloma control; PH: patient with hyperdiploid tumor; PT: patient with tumor harboring translocations.

Source data

Extended Data Fig. 6 Related to Fig. 4.

(a) UMAP of 32,412 CD38+SDC1+ myeloma cells of 13 myeloma patients showing 7 clusters identified by integrated analysis. Colors represent clusters (b) Transcription of marker genes in the CD38+SDC1+ myeloma cell dataset (c) Original UMAPs of CD38+SDC1+ myeloma cell dataset split by patient. PH: patient with hyperdiploid tumor; PT: patient with tumor harboring translocations (d) Transcription of TNF and IL1B in the CD38+SDC1+ myeloma cell dataset.

Extended Data Fig. 7 Related to Fig. 6.

(a) Presence of canonical immune subset transcripts in bulk RNA sequencing datasets of purified MSC from newly-diagnosed myeloma (blue, n=14) and non-myeloma control patients (green, n=4) (b) Transcription of stromal cell subset markers comparing myeloma (blue) to non-myeloma (green) (c) Transcription of genes encoding extracellular matrix (ECM) proteins comparing myeloma (blue) to non-myeloma (green) (d) Transcription of canonical immune subset transcripts in bulk RNA sequencing datasets of purified MSC comparing post-induction treatment (blue, n = 9) to diagnosis (green, n = 9) (e) Transcription of stromal cell subset markers comparing post-induction treatment (blue) to diagnosis (green) (f) Transcription of genes encoding ECM proteins comparing post-induction treatment (blue) to diagnosis (green) (g) Heatmap of top differentially expressed genes up or down in treated patients compared to newly diagnosed samples as described in Supplementary Table 13. The data in (a), (b), (c), (d), (e) and (f) are presented as mean ± standard error of the mean (SEM). Significance was calculated using the Wald test (two-tailed), followed by Benjamini Hochberg correction. NS; p >0.05, *; p ≤0.05, **; p ≤0.01. TPM; transcripts per million.

Source data

Extended Data Fig. 8 Hypothetical model.

Myeloma cells present in the bone marrow either activate stromal cells directly or trigger local inflammation, leading to the production of inflammatory mediators. Activation of local MSCs leads to formation of two subsets of iMSC (MSC2 and MSC1). iMSCs start to transcribe genes that might support MM cell survival and attract and modulate immune subsets, including myeloid cells.

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

de Jong, M.M.E., Kellermayer, Z., Papazian, N. et al. The multiple myeloma microenvironment is defined by an inflammatory stromal cell landscape. Nat Immunol 22, 769–780 (2021). https://doi.org/10.1038/s41590-021-00931-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00931-3

This article is cited by

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