Skip to main content
Top
Published in: International Journal of Hematology 3/2013

01-03-2013 | Progress in Hematology

Multiple myeloma-initiating cells

Author: Naoki Hosen

Published in: International Journal of Hematology | Issue 3/2013

Login to get access

Abstract

Multiple myeloma (MM) is characterized by the clonal expansion of malignant plasma cells. As in other cancers, MM plasma cells are thought to be derived from MM-initiating cells, although these remain unidentified. MM patients harbor phenotypic CD19+ B cells expressing the immunoglobulin gene sequence and the idiotype unique to the individual myeloma clone. Some previous studies have reported that CD19+ clonotypic B cells can serve as MM-initiating cells. However, we and another group have recently showed that CD19+ B cells from many MM patients do not reconstitute MM disease upon transplantation into NOD/SCID IL2Rγc−/− mice. In the SCID-rab and SCID-hu models, which enable engraftment of human MM in vivo, CD19CD38++ plasma cells engrafted and rapidly propagated MM, while engraftment of CD19+ B cells was not detected. Both CD138 and CD138+ plasma cells have the potential to propagate MM clones in vivo in the absence of CD19+ B cells. Distinct from acute myeloid leukemia-initiating cells, which are derived from undifferentiated stem or progenitor cells, MM-initiating cells are derived from plasma cells, which are terminally differentiated cells. An improved understanding of how the bone marrow microenvironment supports MM-initiating plasma cells, which can initiate MM disease in the SCID-hu (or rab) model, is thus now essential.
Literature
3.
go back to reference Kapoor P, Rajkumar SV. Update on risk stratification and treatment of newly diagnosed multiple myeloma. Int J Hematol. 2011;94(4):310–20.PubMedCrossRef Kapoor P, Rajkumar SV. Update on risk stratification and treatment of newly diagnosed multiple myeloma. Int J Hematol. 2011;94(4):310–20.PubMedCrossRef
4.
go back to reference Bakkus MH, Heirman C, Van Riet I, Van Camp B, Thielemans K. Evidence that multiple myeloma Ig heavy chain VDJ genes contain somatic mutations but show no intraclonal variation. Blood. 1992;80:2326–35.PubMed Bakkus MH, Heirman C, Van Riet I, Van Camp B, Thielemans K. Evidence that multiple myeloma Ig heavy chain VDJ genes contain somatic mutations but show no intraclonal variation. Blood. 1992;80:2326–35.PubMed
5.
go back to reference Vescio RA, Cao J, Hong CH, et al. Myeloma Ig heavy chain V region sequences reveal prior antigenic selection and marked somatic mutation but no intraclonal diversity. J Immunol. 1995;155:2487–97.PubMed Vescio RA, Cao J, Hong CH, et al. Myeloma Ig heavy chain V region sequences reveal prior antigenic selection and marked somatic mutation but no intraclonal diversity. J Immunol. 1995;155:2487–97.PubMed
6.
go back to reference Sahota SS, Leo R, Hamblin TJ, Stevenson FK. Myeloma VL and VH gene sequences reveal a complementary imprint of antigen selection in tumor cells. Blood. 1997;89:219–26.PubMed Sahota SS, Leo R, Hamblin TJ, Stevenson FK. Myeloma VL and VH gene sequences reveal a complementary imprint of antigen selection in tumor cells. Blood. 1997;89:219–26.PubMed
7.
go back to reference Pilarski LM, Jensen GS. Monoclonal circulating B cells in multiple myeloma. A continuously differentiating, possibly invasive, population as defined by expression of CD45 isoforms and adhesion molecules. Hematol Oncol Clin North Am. 1992;6:297–322.PubMed Pilarski LM, Jensen GS. Monoclonal circulating B cells in multiple myeloma. A continuously differentiating, possibly invasive, population as defined by expression of CD45 isoforms and adhesion molecules. Hematol Oncol Clin North Am. 1992;6:297–322.PubMed
8.
go back to reference Bergsagel PL, Smith AM, Szczepek A, Mant MJ, Belch AR, Pilarski LM. In multiple myeloma, clonotypic B lymphocytes are detectable among CD19+ peripheral blood cells expressing CD38, CD56, and monotypic Ig light chain. Blood. 1995;85:436–47.PubMed Bergsagel PL, Smith AM, Szczepek A, Mant MJ, Belch AR, Pilarski LM. In multiple myeloma, clonotypic B lymphocytes are detectable among CD19+ peripheral blood cells expressing CD38, CD56, and monotypic Ig light chain. Blood. 1995;85:436–47.PubMed
9.
go back to reference Chen BJ, Epstein J. Circulating clonal lymphocytes in myeloma constitute a minor subpopulation of B cells. Blood. 1996;87:1972–6.PubMed Chen BJ, Epstein J. Circulating clonal lymphocytes in myeloma constitute a minor subpopulation of B cells. Blood. 1996;87:1972–6.PubMed
10.
go back to reference Rasmussen T, Jensen L, Johnsen HE. The CD19 compartment in myeloma includes a population of clonal cells persistent after high-dose treatment. Leuk Lymphoma. 2002;43:1075–7.PubMedCrossRef Rasmussen T, Jensen L, Johnsen HE. The CD19 compartment in myeloma includes a population of clonal cells persistent after high-dose treatment. Leuk Lymphoma. 2002;43:1075–7.PubMedCrossRef
11.
go back to reference Pilarski LM, Seeberger K, Coupland RW, et al. Leukemic B cells clonally identical to myeloma plasma cells are myelomagenic in NOD/SCID mice. Exp Hematol. 2002;30:221–8.PubMedCrossRef Pilarski LM, Seeberger K, Coupland RW, et al. Leukemic B cells clonally identical to myeloma plasma cells are myelomagenic in NOD/SCID mice. Exp Hematol. 2002;30:221–8.PubMedCrossRef
12.
go back to reference Pilarski LM, Hipperson G, Seeberger K, Pruski E, Coupland RW, Belch AR. Myeloma progenitors in the blood of patients with aggressive or minimal disease: engraftment and self-renewal of primary human myeloma in the bone marrow of NOD SCID mice. Blood. 2000;95:1056–65.PubMed Pilarski LM, Hipperson G, Seeberger K, Pruski E, Coupland RW, Belch AR. Myeloma progenitors in the blood of patients with aggressive or minimal disease: engraftment and self-renewal of primary human myeloma in the bone marrow of NOD SCID mice. Blood. 2000;95:1056–65.PubMed
13.
go back to reference Matsui W, Huff CA, Wang Q, et al. Characterization of clonogenic multiple myeloma cells. Blood. 2004;103:2332–6.PubMedCrossRef Matsui W, Huff CA, Wang Q, et al. Characterization of clonogenic multiple myeloma cells. Blood. 2004;103:2332–6.PubMedCrossRef
14.
go back to reference Matsui W, Wang Q, Barber JP, et al. Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance. Cancer Res. 2008;68:190–7.PubMedCrossRef Matsui W, Wang Q, Barber JP, et al. Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance. Cancer Res. 2008;68:190–7.PubMedCrossRef
15.
go back to reference Kapoor P, Greipp PT, Morice WG, Rajkumar SV, Witzig TE, Greipp PR. Anti-CD20 monoclonal antibody therapy in multiple myeloma. Br J Haematol. 2008;141:135–48.PubMedCrossRef Kapoor P, Greipp PT, Morice WG, Rajkumar SV, Witzig TE, Greipp PR. Anti-CD20 monoclonal antibody therapy in multiple myeloma. Br J Haematol. 2008;141:135–48.PubMedCrossRef
16.
go back to reference Hosen N, Matsuoka Y, Kishida S, et al. CD138-negative clonogenic cells are plasma cells but not B cells in some multiple myeloma patients. Leukemia. 2012;26:2135–41.PubMedCrossRef Hosen N, Matsuoka Y, Kishida S, et al. CD138-negative clonogenic cells are plasma cells but not B cells in some multiple myeloma patients. Leukemia. 2012;26:2135–41.PubMedCrossRef
17.
go back to reference Kim D, Park CY, Medeiros BC, Weissman IL. CD19(−)CD45(low/−)CD38(high)/CD138(+) plasma cells enrich for human tumorigenic myeloma cells. Leukemia. 2012;26(12):2530–7.PubMedCrossRef Kim D, Park CY, Medeiros BC, Weissman IL. CD19(−)CD45(low/−)CD38(high)/CD138(+) plasma cells enrich for human tumorigenic myeloma cells. Leukemia. 2012;26(12):2530–7.PubMedCrossRef
18.
go back to reference 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. 1998;92:2908–13.PubMed 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. 1998;92:2908–13.PubMed
19.
go back to reference Yata K, Yaccoby S. The SCID-rab model: a novel in vivo system for primary human myeloma demonstrating growth of CD138-expressing malignant cells. Leukemia. 2004;18:1891–7.PubMedCrossRef Yata K, Yaccoby S. The SCID-rab model: a novel in vivo system for primary human myeloma demonstrating growth of CD138-expressing malignant cells. Leukemia. 2004;18:1891–7.PubMedCrossRef
20.
go back to reference Kawano Y, Fujiwara S, Wada N, et al. Multiple myeloma cells expressing low levels of CD138 have an immature phenotype and reduced sensitivity to lenalidomide. Int J Oncol. 2012;41(3):876–84.PubMed Kawano Y, Fujiwara S, Wada N, et al. Multiple myeloma cells expressing low levels of CD138 have an immature phenotype and reduced sensitivity to lenalidomide. Int J Oncol. 2012;41(3):876–84.PubMed
21.
go back to reference Fuhler GM, Baanstra M, Chesik D, et al. Bone marrow stromal cell interaction reduces syndecan-1 expression and induces kinomic changes in myeloma cells. Exp Cell Res. 2010;316:1816–28.PubMedCrossRef Fuhler GM, Baanstra M, Chesik D, et al. Bone marrow stromal cell interaction reduces syndecan-1 expression and induces kinomic changes in myeloma cells. Exp Cell Res. 2010;316:1816–28.PubMedCrossRef
22.
go back to reference Jakubikova J, Adamia S, Kost-Alimova M, et al. Lenalidomide targets clonogenic side population in multiple myeloma: pathophysiologic and clinical implications. Blood. 2011;117:4409–19.PubMedCrossRef Jakubikova J, Adamia S, Kost-Alimova M, et al. Lenalidomide targets clonogenic side population in multiple myeloma: pathophysiologic and clinical implications. Blood. 2011;117:4409–19.PubMedCrossRef
23.
go back to reference Hideshima T, Mitsiades C, Tonon G, Richardson PG, Anderson KC. Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer. 2007;7(8):585–98.PubMedCrossRef Hideshima T, Mitsiades C, Tonon G, Richardson PG, Anderson KC. Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer. 2007;7(8):585–98.PubMedCrossRef
24.
go back to reference Abe M. Targeting the interplay between myeloma cells and the bone marrow microenvironment in myeloma. Int J Hematol. 2011;94(4):334–43.PubMedCrossRef Abe M. Targeting the interplay between myeloma cells and the bone marrow microenvironment in myeloma. Int J Hematol. 2011;94(4):334–43.PubMedCrossRef
25.
go back to reference Iriuchishima H, Takubo K, Miyakawa Y, et al. Neovascular niche for human myeloma cells in immunodeficient mouse bone. PLoS One. 2012;7(2):e30557.PubMedCrossRef Iriuchishima H, Takubo K, Miyakawa Y, et al. Neovascular niche for human myeloma cells in immunodeficient mouse bone. PLoS One. 2012;7(2):e30557.PubMedCrossRef
26.
go back to reference Arai F, Yoshihara H, Hosokawa K, et al. Niche regulation of hematopoietic stem cells in the endosteum. Ann N Y Acad Sci. 2009;1176:36–46.PubMedCrossRef Arai F, Yoshihara H, Hosokawa K, et al. Niche regulation of hematopoietic stem cells in the endosteum. Ann N Y Acad Sci. 2009;1176:36–46.PubMedCrossRef
27.
go back to reference Kikushige Y, Ishikawa F, Miyamoto T, et al. Self-renewing hematopoietic stem cell is the primary target in pathogenesis of human chronic lymphocytic leukemia. Cancer Cell. 2011;20(2):246–59.PubMedCrossRef Kikushige Y, Ishikawa F, Miyamoto T, et al. Self-renewing hematopoietic stem cell is the primary target in pathogenesis of human chronic lymphocytic leukemia. Cancer Cell. 2011;20(2):246–59.PubMedCrossRef
28.
go back to reference Peacock CD, Wang Q, Gesell GS, et al. Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma. Proc Natl Acad Sci USA. 2007;104:4048–53.PubMedCrossRef Peacock CD, Wang Q, Gesell GS, et al. Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma. Proc Natl Acad Sci USA. 2007;104:4048–53.PubMedCrossRef
29.
go back to reference Nefedova Y, Sullivan DM, Bolick SC, Dalton WS, Gabrilovich DI. Inhibition of Notch signaling induces apoptosis of myeloma cells and enhances sensitivity to chemotherapy. Blood. 2008;111:2220–9.PubMedCrossRef Nefedova Y, Sullivan DM, Bolick SC, Dalton WS, Gabrilovich DI. Inhibition of Notch signaling induces apoptosis of myeloma cells and enhances sensitivity to chemotherapy. Blood. 2008;111:2220–9.PubMedCrossRef
30.
go back to reference Hideshima T, Anderson KC. Novel therapies in MM: from the aspect of preclinical studies. Int J Hematol. 2011;94(4):344–54.PubMedCrossRef Hideshima T, Anderson KC. Novel therapies in MM: from the aspect of preclinical studies. Int J Hematol. 2011;94(4):344–54.PubMedCrossRef
31.
go back to reference Benson DM Jr, Byrd JC. CS1-directed monoclonal antibody therapy for multiple myeloma. J Clin Oncol. 2012;30(16):2013–5.PubMedCrossRef Benson DM Jr, Byrd JC. CS1-directed monoclonal antibody therapy for multiple myeloma. J Clin Oncol. 2012;30(16):2013–5.PubMedCrossRef
32.
go back to reference de Weers M, Tai YT, van der Veer MS, et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J Immunol. 2011;186(3):1840–8.PubMedCrossRef de Weers M, Tai YT, van der Veer MS, et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J Immunol. 2011;186(3):1840–8.PubMedCrossRef
33.
go back to reference Hosen N, Ichihara H, Mugitani A, et al. CD48 as a novel molecular target for antibody therapy in multiple myeloma. Br J Haematol. 2011;156:213–24.PubMedCrossRef Hosen N, Ichihara H, Mugitani A, et al. CD48 as a novel molecular target for antibody therapy in multiple myeloma. Br J Haematol. 2011;156:213–24.PubMedCrossRef
Metadata
Title
Multiple myeloma-initiating cells
Author
Naoki Hosen
Publication date
01-03-2013
Publisher
Springer Japan
Published in
International Journal of Hematology / Issue 3/2013
Print ISSN: 0925-5710
Electronic ISSN: 1865-3774
DOI
https://doi.org/10.1007/s12185-013-1293-0

Other articles of this Issue 3/2013

International Journal of Hematology 3/2013 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine