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.

  • Technical Report
  • Published:

Microenvironment-dependent growth of preneoplastic and malignant plasma cells in humanized mice

Abstract

Most human cancers, including myeloma, are preceded by a precursor state. There is an unmet need for in vivo models to study the interaction of human preneoplastic cells in the bone marrow microenvironment with non-malignant cells. Here, we genetically humanized mice to permit the growth of primary human preneoplastic and malignant plasma cells together with non-malignant cells in vivo. Growth was largely restricted to the bone marrow, mirroring the pattern in patients with myeloma. Xenografts captured the genomic complexity of parental tumors and revealed additional somatic changes. Moreover, xenografts from patients with preneoplastic gammopathy showed progressive growth, suggesting that the clinical stability of these lesions may in part be due to growth controls extrinsic to tumor cells. These data demonstrate a new approach to investigate the entire spectrum of human plasma cell neoplasia and illustrate the utility of humanized models for understanding the functional diversity of human tumors.

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

Figure 1: Engraftment and phenotype of human plasma cell tumors in MIS(KI)TRG6 mice.
Figure 2: Engraftment of different tumor cellular compartments, the spectrum of non-malignant cells that engrafted and the potential for serial transplantation.
Figure 3: Pattern of tumor cell growth from a spectrum of clonal plasma cell tumors and preneoplastic lesions.
Figure 4: Comparison of the MIS(KI)TRG6 and SCID-hu models for the growth of preneoplastic gammopathies.
Figure 5: Genomic analysis of tumor cells engrafted in MIS(KI)TRG6 mice.
Figure 6: Analysis of somatic non-synonymous variants (SNVs).

Similar content being viewed by others

References

  1. Palumbo, A. & Anderson, K. Multiple myeloma. N. Engl. J. Med. 364, 1046–1060 (2011).

    Article  CAS  Google Scholar 

  2. Schüler, J., Ewerth, D., Waldschmidt, J., Wäsch, R. & Engelhardt, M. Preclinical models of multiple myeloma: a critical appraisal. Expert Opin. Biol. Ther. 13 (Suppl. 1), S111–S123 (2013).

    Article  Google Scholar 

  3. Yaccoby, S., Barlogie, B. & Epstein, J. Primary myeloma cells growing in SCID-hu mice: a model for studying the biology and treatment of myeloma and its manifestations. Blood 92, 2908–2913 (1998).

    Article  CAS  Google Scholar 

  4. Calimeri, T. et al. A unique three-dimensional SCID-polymeric scaffold (SCID-synth-hu) model for in vivo expansion of human primary multiple myeloma cells. Leukemia 25, 707–711 (2011).

    Article  CAS  Google Scholar 

  5. Rongvaux, A. et al. Human hemato-lymphoid system mice: current use and future potential for medicine. Annu. Rev. Immunol. 31, 635–674 (2013).

    Article  CAS  Google Scholar 

  6. Rongvaux, A. et al. Development and function of human innate immune cells in a humanized mouse model. Nat. Biotechnol. 32, 364–372 (2014).

    Article  CAS  Google Scholar 

  7. Deng, K. et al. Broad CTL response is required to clear latent HIV-1 due to dominance of escape mutations. Nature 517, 381–385 (2015).

    Article  CAS  Google Scholar 

  8. Kishimoto, T. Interleukin-6: from basic science to medicine—40 years in immunology. Annu. Rev. Immunol. 23, 1–21 (2005).

    Article  CAS  Google Scholar 

  9. Kishimoto, T., Akira, S. & Taga, T. Interleukin-6 and its receptor: a paradigm for cytokines. Science 258, 593–597 (1992).

    Article  CAS  Google Scholar 

  10. Tassone, P. et al. A clinically relevant SCID-hu in vivo model of human multiple myeloma. Blood 106, 713–716 (2005).

    Article  CAS  Google Scholar 

  11. Matsui, W. et al. Characterization of clonogenic multiple myeloma cells. Blood 103, 2332–2336 (2004).

    Article  CAS  Google Scholar 

  12. Hosen, N. et al. CD138-negative clonogenic cells are plasma cells but not B cells in some multiple myeloma patients. Leukemia 26, 2135–2141 (2012).

    Article  CAS  Google Scholar 

  13. Kim, D., Park, C.Y., Medeiros, B.C. & Weissman, I.L. CD19CD45low/− CD38high/CD138+ plasma cells enrich for human tumorigenic myeloma cells. Leukemia 26, 2530–2537 (2012).

    Article  CAS  Google Scholar 

  14. Kukreja, A. et al. Enhancement of clonogenicity of human multiple myeloma by dendritic cells. J. Exp. Med. 203, 1859–1865 (2006).

    Article  CAS  Google Scholar 

  15. Klco, J.M. et al. Functional heterogeneity of genetically defined subclones in acute myeloid leukemia. Cancer Cell 25, 379–392 (2014).

    Article  CAS  Google Scholar 

  16. Goyama, S., Wunderlich, M. & Mulloy, J.C. Xenograft models for normal and malignant stem cells. Blood 125, 2630–2640 (2015).

    Article  CAS  Google Scholar 

  17. Zhao, S. et al. Serial exome analysis of disease progression in premalignant gammopathies. Leukemia 28, 1548–1552 (2014).

    Article  CAS  Google Scholar 

  18. Hanamura, I. et al. Frequent gain of chromosome band 1q21 in plasma-cell dyscrasias detected by fluorescence in situ hybridization: incidence increases from MGUS to relapsed myeloma and is related to prognosis and disease progression following tandem stem-cell transplantation. Blood 108, 1724–1732 (2006).

    Article  CAS  Google Scholar 

  19. Nair, S. et al. Clonal immunoglobulin against lysolipids in the origin of myeloma. N. Engl. J. Med. 374, 555–561 (2016).

    Article  CAS  Google Scholar 

  20. Koduru, S. et al. Dendritic cell-mediated activation-induced cytidine deaminase (AID)-dependent induction of genomic instability in human myeloma. Blood 119, 2302–2309 (2012).

    Article  CAS  Google Scholar 

  21. Dhodapkar, M.V. & Dhodapkar, K.M. Immune modulation in hematologic malignancies. Semin. Oncol. 42, 617–625 (2015).

    Article  CAS  Google Scholar 

  22. Melchor, L. et al. Single-cell genetic analysis reveals the composition of initiating clones and phylogenetic patterns of branching and parallel evolution in myeloma. Leukemia 28, 1705–1715 (2014).

    Article  CAS  Google Scholar 

  23. Ghobrial, I.M. Myeloma as a model for the process of metastasis: implications for therapy. Blood 120, 20–30 (2012).

    Article  CAS  Google Scholar 

  24. Walker, B.A. et al. Intraclonal heterogeneity is a critical early event in the development of myeloma and precedes the development of clinical symptoms. Leukemia 28, 384–390 (2014).

    Article  Google Scholar 

  25. López-Corral, L. et al. The progression from MGUS to smoldering myeloma and eventually to multiple myeloma involves a clonal expansion of genetically abnormal plasma cells. Clin. Cancer Res. 17, 1692–1700 (2011).

    Article  Google Scholar 

  26. Dhodapkar, M.V. et al. Clinical, genomic, and imaging predictors of myeloma progression from asymptomatic monoclonal gammopathies (SWOG S0120). Blood 123, 78–85 (2014).

    Article  CAS  Google Scholar 

  27. Dhodapkar, M.V. et al. A reversible defect in natural killer T cell function characterizes the progression of premalignant to malignant multiple myeloma. J. Exp. Med. 197, 1667–1676 (2003).

    Article  CAS  Google Scholar 

  28. Dhodapkar, M.V., Krasovsky, J., Osman, K. & Geller, M.D. Vigorous premalignancy-specific effector T cell response in the bone marrow of patients with monoclonal gammopathy. J. Exp. Med. 198, 1753–1757 (2003).

    Article  CAS  Google Scholar 

  29. Spisek, R. et al. Frequent and specific immunity to the embryonal stem cell-associated antigen SOX2 in patients with monoclonal gammopathy. J. Exp. Med. 204, 831–840 (2007).

    Article  CAS  Google Scholar 

  30. Dhodapkar, M.V. et al. Prospective analysis of antigen-specific immunity, stem-cell antigens, and immune checkpoints in monoclonal gammopathy. Blood 126, 2475–2478 (2015).

    Article  CAS  Google Scholar 

  31. Lawson, M.A. et al. Osteoclasts control reactivation of dormant myeloma cells by remodelling the endosteal niche. Nat. Commun. 6, 8983 (2015).

    Article  CAS  Google Scholar 

  32. Sehgal, K. et al. Clinical and pharmacodynamic analysis of pomalidomide dosing strategies in myeloma: impact of immune activation and cereblon targets. Blood 125, 4042–4051 (2015).

    Article  CAS  Google Scholar 

  33. Zhao, S. et al. Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proc. Natl. Acad. Sci. USA 110, 2916–2921 (2013).

    Article  CAS  Google Scholar 

  34. Choi, M. et al. K+ channel mutations in adrenal aldosterone-producing adenomas and hereditary hypertension. Science 331, 768–772 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by funds from the US National Institutes of Health (CA156689, CA106802, CA197603) and the clinical research priority program of the University of Zurich. The authors thank F. Giráldez-López, I. Tikhonova, S. Mane and M. Youngblood for assistance with sequencing analyses and C. Weibel, J. Alderman and C. Foster (all at Yale School of Medicine) for assistance with mouse colonies.

Author information

Authors and Affiliations

Authors

Contributions

M.V.D. and R.A.F. conceived and supervised the overall project, designed experiments and analyzed results. R.D., T.S. and R.V. designed and performed experiments and analyzed data. S.K., A.H., S.H., M.H.K., C.B., L.Z. and A.B. performed experiments and analyzed data. E.E. and M.G.M. designed experiments and analyzed data. M.V.D., R.D., T.S., R.V., M.G.M. and R.A.F. wrote the manuscript.

Corresponding authors

Correspondence to Richard A Flavell or Madhav V Dhodapkar.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text, Figures and Tables

Supplementary Figures 1–6 and Supplementary Tables 1–3 (PDF 29135 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Das, R., Strowig, T., Verma, R. et al. Microenvironment-dependent growth of preneoplastic and malignant plasma cells in humanized mice. Nat Med 22, 1351–1357 (2016). https://doi.org/10.1038/nm.4202

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4202

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