Skip to main content
American Journal of Blood Research logoLink to American Journal of Blood Research
. 2011 Sep 8;1(2):135–145.

Progress in myeloma stem cells

Richard Dela Cruz 1, Guido Tricot 1, Maurizio Zangari 1, Fenghuang Zhan 1
PMCID: PMC3301427  PMID: 22432075

Abstract

Multiple myeloma (MM) is the second most common hematologic malignancy in the United States and affects about 4 in 100,000 Americans. Even though much progress has been made in MM therapy, MM remains an incurable disease for the vast majority of patients. The existence of MM stem cell is considered one of the major causes of MM drug-resistance, leading to relapse. This highlights the importance and urgency of developing approaches to target MM stem cells. However, very little is known about the molecular characteristics of the MM stem cells, which makes it difficult to target MM stem cells therapeutically. Evidence of the existence of a myeloma stem cell has been provided by Matsui et al. showing that the CD138- and CD20+ fraction, which is a minor population of the MM cells, has a greater clonogenic potential and has the phenotype of a memory B-cell (CD19+, CD27+). In this review, we report recent progress of cell surface markers in cancer stem cells, especially in myeloma and the molecular mechanisms related to drug resistance and myeloma disease progression.

Keywords: Cancer stem cell, multiple myeloma, drug resistance, and cell signaling

Introduction

Multiple myeloma (MM) is a hematologic cancer subtype characterized by uncontrolled proliferation of plasma cells in the bone marrow. The exact cause of MM is unknown and MM patients generally have no symptoms until the disease is already in an advanced state. In 2010, the NCI estimates that there were 20,180 new cases of MM and 10,650 deaths directly attributed to this cancer. Research into the molecular, cytogenetic, gene expression, and signaling pathway signature of MM (as reviewed and discussed in [1-4]) has significantly improved our understanding of how MM develops and progresses, and has led to advances in MM therapies and increased median survival rates. Unfortunately, this disease eventually relapses and then becomes incurable. One theory, based on the cancer stem cell (CSC) model, is that current MM treatments kill the bulk of tumor cells but not the self-renewing MM cancer stem cells (MMSCs) subpopulation. The latter type of cells is believed to be the most malignant and drug resistant but also a rare sub-population of MM cells with the ability for self-renewal, division, and maintenance of its undif-ferentiated state not unlike normal hematopoiets stem cells (HSCs). Although the stem cell phenotype of acute myeloid leukemia has long been identified and is generally accepted, the definitive MMSC phenotype / markers are still being deliberated in the MM research community and there are currently several viewpoints on MM cells with stemness characteristics. Targeting of MMSCs is clinically relevant since insight into the characteristics of this subpopulation, if it exists, could result in novel therapeutics that can make MM a more manageable and even curable disease.

Normal and cancer stem cells in the hematopoietic system

The existence of HSCs is an accepted concept along with their important use clinically (as reviewed in [5, 6]). HSCs are typically isolated based on their surface markers as first shown by the Weissman group for both mouse and human stem cells [7, 8], and it has now been 10 years since it was demonstrated that a single bone marrow-derived cell can recapitulate blood cell lineages in irradiated mice [9]. Gene expression profiling (GEP) of highly purified HSCs versus non-self-renewing multipotent hematopoietic progenitors helped identify the SLAM family of cell surface markers, and has further improved surface marker-based fluorescence activated cell sorting (FACS) to almost 100% purity with about half of the purified fraction possessing full HSC activity [10]. The relatively successful isolation of semi-pure HSC has been translated into clinical use (as reviewed in [5, 6]). HSCs are critical in stem cell transplantation (SCT) after myeloablative therapy to treat various diseases including leukemia and multiple myeloma [11]. With the exception of myeloma, SCT are mainly performed upon relapse, which could reflect the resistance of CSCs to traditional cancer therapies (the relevance of CSCs to BMT is discussed in [12]). Although the idea of the clonal origin of malignancies and the still utilized clonogenesis assay for CSCs have been around for more than 40 years now [13-15], definitive evidence for hematopoietic CSCs and its hierarchal organization was only first presented by Bonnet and Dick in 1997 based on their seminal observation that only the CD34+/CD38- leukemia cell subpopulation is able to induce leukemia in a SCID mouse model [16]. The unique MMSC signature, unfortunately, is still unresolved. While the markers for different CSC types differ from one another, there is congruence in their functional properties including ability for self-renewal, tumorigenesis, and drug-resistance. Thus, these stemness characteristics are the basis for trying to functionally identify the definitive MMSC signature.

Multiple myeloma stem cells

HSCs are different from CSCs in that the former's activity and function are tightly regulated whereas they are dysregulated in CSCs, which eventually lead to cancer cell overpopulation (where the CSC remains a minor fraction). In MM, this minor population has functionally been shown to have increased potential of self renewal. The cancer stem cell model is based on the concept that most cancers just like the normal hematopoiesis, are hierarchically organized [17]. This implies that CSCs give not only to daughter cancer cells that are more differentiated, but also to another CSC (asymmetrical division). Based on this model, MMSCs then should possess a surface marker signature more similar to plasma cell precursors. Indeed drug-resistant clonotypic cells from myeloma patients with stemness characteristics have been reported to have memory B cell-like surface marker profile [18-21] although MM cells with stemness characteristics not possessing this signature have also been reported [22, 23].

HSC and CSC research share a common problem that these rare cells being very difficult to identify, isolate, and characterize. The possible existence of MMSCs was first postulated based on experiments using MM cell lines and patient MM samples indicating that there is a small population of MM cells with the ability for unlimited self renewal [24, 25]. Second, the capacity for self-renewal of primary MM samples in vitro, using the sphere-forming assay [26], as well as in vivo, by serial transplantability in NOD/SCID mice [27] was also observed. Finally, the fact that most MM patients after traditional treatment ultimately relapse also points to the existence of MMSC.

The identification of SC markers in combination with FACS has exponentially increased the efficiency of HSC and CSC identification and isolation. This has led to progress in trying to identify the unique MMSC surface marker signature.

  1. The SCID-hu in vivo mouse model and FACS were used by Yaccoby et al. to show that only primary CD38++/CD45- plasma cells proliferate successfully within the engrafted fetal bone microenvironment although it did not invade the murine host bone [22] and CD38++ is not a plasma cell precursor marker but is a plasma cell marker. However, the purity of the population is never 100% and it is possible that the disease was propagated by the few CD 138-cells.

  2. Drug resistant MM cells in a novel 3D model were CD20+, although expression of CD20 is heterogenous in bulk myeloma cells and is only detected in a minority of patients [28].

  3. Cells with the CD138-/CD19+/CD20+/CD27+ phenotype which is memory B cell-like, are serially transplantable. This is consistent with the theory that CSC can arise from hierarchally more primitive than malignant plasma cells [18, 19, 29].

  4. The CD138- MM phenotype has also been associated with increased apoptosis of these cells [30]. It is now generally accepted that apoptotic myeloma cells lose CD138 marker on its membrane.

  5. Yaccoby showed that a co-culture of differentiated MM cells with OCs reprogrammed the differentiated MM cells to immature CD138low MM cells that were drug resistant [31].

  6. Pilarski et al. showed CD34+/CD45low clonotypic myeloma cells xenograft in NOD/SCID mice [32]. A subpopulation of cycling CD34+/CD138+/ B7-/H1+ mature plasma cells from primary MM patients was shown to be malignant and could be a possible source of mature MM plasma cell de-differentiation to MMSC [33]. This intriguing possibility was also raised by results recently published by the Weinberg group showing that neoplastic mammary epithelial cells spontaneously convert to a stem-cell like state [34].

  7. Aside from surface markers, FACS has also been used to sort MMSC subpopulation based on functional stem cell markers including ALDH expression [35] and efflux pump ABCG2 activation in a side population (SP) by Hoechst 33342 staining [36].

  8. Jakubikova et al. recently showed that various MM cell lines contain SPs that are inhibited by lenalidomide and that this SP has no correlation with CD138- [23] but Matsui et al., in an earlier publication, showed that ALDH+ and SP are enriched for CD138- MMSC [18].

MMSC stem cell markers agreeable with the hierarchal model of CSC theory, along with the stem cell markers of various types of malignancies, are shown in Table 1 [37-39]. The MM sub-populations with stemness features above were tested in vivo using SCID mouse models that permit proliferation initiated by clonogenic myeloma cells (except for [28]). The robust in vivo NOD/SCID model was also used to verify most of the cancer stem cell markers listed in Table 1 [37]. A related model, the SCID-hu, which involves implantation of fetal bone and injection of myeloma cells in this “pseudo-synergistic” humanized environment, was used by Yaccoby et al. to identify the clonogenic potential of the CD38++/CD45- population [22]. One criticism against the SCID models is that the clonogenic growth of the transplantable subpopulations could be dependent on the recipient's weakened immune system. A recent study seems to support this hypothesis. The use of a more severely immunocompromised model, the IL2rγ null NOD/SCID mice, led to at least a 250-fold increased tumor formation in vivo compared to NOD/SCID mice [40, 41]. The MM-specific 5T33 or 5T2 murine mouse model will be useful in confirming the validity of currently published MMSC markers in identifying MMSC since the clonogenicity of the injected murine myeloma cells will be dependent on, and synergistic to, the recipient bone marrow microenvironment (as further discussed in [42-44]).

TABLE 1.

Cancer stem cell markers

Cancer CD19 CD20 CD24 CD27 CD34 CD38 CD44 CD133 CD138 CD166 ALDH1 ESA other
breast - / low + + + + B1, Lin-
brain +
colon + + +
head and neck +
leukemia (ALL) + - t(12;21), Lin-
leukemia (AML) + - t(9;22), Lin-
lung + +
melanoma + + CD271, Hoescht 33342 SP
pancreatic + + +
prostate + + + B1
ovarian Hoescht 33342 SP
multiple myeloma + + + - + Hoescht 33342 SP

Table modified from [37-39]. B1 = integrin B1 chain; ALDH = aldehyde dehydrogenase; ESA = epidermal surface antigen

There is currently a major controversy regarding the MMSC signature, although this issue is not unique to myeloma [21]. Methodological differences in stem cell purification and source undoubtedly perturb the primary stem cell niche [45] to different extents depending on the protocol employed and could contribute to the identification of various MM subpopulations with stemness characteristics. To definitively confirm self-renewal of the proposed MMSC populations (compared to differentiated but highly proliferative cells) above, in vitro assays need to be done at the single cell level together with the appropriate in vivo model.

MMSC in MM treatment and relapse

Intensive research on the molecular mechanisms of MM disease and progression has led the discovery of a new generation of therapeutics against MM. The thalidomide derivative, lenalidomide, has recently been shown to be effective against relapsed and refractory MM and has been experimentally shown to target the MM SP [23]. The 26S proteasome inhibitor, bortezomib, significantly delays MM progression compared to dexamethasone [46] by inducing apoptosis and inactivation of upregulated NFκB [47-49] although MM cells with constitutively activated NFκB expression are resistant to bortezomib [50]. Synergistic effects have been observed when these drugs are used in combinatorial therapy [51, 52]. Even with more available treatment options and a better understanding of the molecular underpinnings of MM, MM remains a largely incurable disease. In general, current systemic cancer therapies fail to cure advanced tumors, and MM is no exception. Based on CSC theory, MM relapse can be explained by the persistence of cancer stem cells after chemotherapy.

Targeting MMSC

Beside the distinctive properties of self-renewal and proliferation, cancer stem cells are thought to be much more resistant to chemo- and radiotherapy and are a logical cause of drug resistance [17,53,54]. MM is no exception as most anti-MM drugs prolong survival but do not cure the disease. The SCID-hu model is used to test anti-MM drugs in vivo [55-57] but not specifically against the MMSC subpopulation. The CD138-/CD19+/CD20+/CD27+ MM stem cells have been shown to be resistant to dexamethasone, lenalidomide, cyclophosphamide and bortezomib [18] . However, in other studies, the MM SP stem cells were shown to be susceptible to lenalidomide treatment [23]. These results indicate that while lenalidomide can kill potential MMSCs, it may still be ineffective against other possible MMSC subpopulations. One possible solution is to target the self-renewal pathway specific for CSCs. These include the Hedgehog (HH), Wnt, and Notch pathways which are typically active in cancer stem cells [29, 17, 58-63]. One study in particular showed that the HH pathway through SMO upregulation allows for clonal expansion of MM CD138- cells in vitro, while inhibition of CD138- proliferation was observed by using the HH pathway inhibitor cyclopamine [29]. Numerous CSC-targeted therapeutics , specifically inhibiting the HH / Wnt/ Notch self-renewal pathways, are now currently in clinical trials including two specifically for MM patients after autologous stem cell transplantation (Table 2).

Table 2.

Potential cancer stem cell self-renewal pathway inhibitors

Drug Company / Institute Pathway Indication Combination drug Phase
LGK974 Novartis Pharmaceuticals Wnt melanoma, breast neoplasms, lobular carcinoma I
PRI-724 Prism Biolab Corp Wnt advanced solid tumors I
Resvaratrol Univ of California, Irvine Wnt colon cancer I / II
Diclofenac + Vitamin D3 Maastricht Univ Medical Center Wnt + HH basal cell carcinoma III
RO4929097 Cancer Institute of New Jersey / NCI Notch multiple myeloma and plasma cell neoplasm after autologous stem cell transplant melphalan II
RO4929097 M.D. Anderson Cancer Center / NCI Notch lung cancer after front-line chemotherapy II
RO4929097 Albert Einstein College of Medicine of Yeshiva Univ / NCI Notch cutaneous melanoma II
RO4929097 Children's Oncology Group / NCI Notch brain and central nervous system tumors leukemia, lymphoma, unspecified childhood solid tumor dexamethasone for all indications I / II
PF-04449913 Pfizer HH solid tumors, unspecified hematological malignancies I
PF-04449913 Pfizer HH chronic myeloid leukemia dasatinib, bosutinib I
LEQ506 Novartis Pharmaceuticals HH advanced solid tumors, recurrent or refractory medulloblastoma I
IPI-926 Infinity Pharmaceuticals HH metastatic pancreatic cancer, recurrent head and neck cancer conventional chondrosarcoma gemcitabine, cetuximab I / II I II
GDC-0449 Roche Pharma AG Genentech HH various indications including MM** various interventions** I / II

A major reason of failure in cancer treatment is the existence of a drug resistant sub-clone, which may represent cancer stem cells at diagnosis and may expand during treatment. A comprehensive analysis of the genetic and molecular mechanisms of drug resistances in cancers remains elusive. We were the first to describe that the presence of chromosomal abnormalities detected by conventional metaphase cytogenetic analysis, as the most important predictor for outcome in myeloma [64]. In particular, complete or partial deletion of chromosome 13, present in 17% of newly diagnosed myeloma patients, was associated with poor prognosis [65]. This was surprising since such a deletion by fluorescence in-situ hybridization (FISH) analysis is associated with a good prognosis in chronic lymphocytic leukemia (CLL) [66]. Using FISH analysis, many of the partners involved in translocations with 14q32 (heavy chain gene locus) have been identified, some of which could not be detected with conventional cytogenetics, because of their telomeric location [67]. These include t(4;14) involving FGFR3, t(14;16), involving c-MAF and t(14;20), involving MAF-B. We now know that these translocations have a poor prognosis and are very often associated with a deletion of chromosome 13. Therefore, the poor prognosis of chromosome 13 by conventional cytogenetics is most likely not directly related to loss of cytogenetic material of this chromosome, but to the frequent association with these poor-prognosis translocations defined by FISH [68]. These translocations are much more frequently seen in hypodiploid myelomas, which are known to have a poor prognosis [69]. Large studies using FISH analysis also identified that a deletion of 17p13 (p53 gene locus) was a poor prognostic indicator. In contrast, hyperdiploidy or translocations involving Cyclin D1, t(11;14) or Cyclin D3, t(6;14) have a good prognosis. Cyclin D abnormalities are an early event in the development of myeloma [70]. Although cytogenetic abnormalities are more frequently (almost in 100% of patients) found with FISH analysis, the predictive value of metaphase abnormalities, found in one third of patients has proven to be superior to that of FISH [71], because they represent the best surrogate marker for bone marrow stroma dependency of the myeloma cells. Typically, if myeloma cells are removed from their micro-environmental support system and transferred to a hostile environment in the test tube, they rapidly apoptose. The mitoses observed in such patients by conventional cytogenetics are derived from the remaining normal hematopoietic cells. If on the other hand, myeloma cells have become stroma-independent, which is a feature of advanced and aggressive myeloma, they will not undergo apoptosis after removal from the bone marrow stroma, and will be able to survive and grow in the test tube and thereby will give rise to abnormal metaphase cytogenetics [72].

Global gene expression profiling (GEP) has emerged as a powerful tool for classifying disease subtypes, for developing robust prognostic models and identifying new drug-resistance targets in cancers [73-76]. Using GEP analysis, we have established transcriptomes by comparing multiple myeloma (MM), monoclonal gammopathy of undetermined significance (MGUS), and normal plasma cells [77, 78]. We have elucidated the role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions [79, 80]; and identified molecular and genetic myeloma subgroups [3, 81]. We have also defined a 70-high-risk gene model through the GEP analysis of more than 550 newly diagnosed MM patients [4]. Through GEP analysis, our lab determined that CKS1B gene, which is in the 1q21 chromosome amplification hotspot in MM and is overexpressed in relapsed patients, is associated with increased drug resistance and is a viable anti-myeloma target [82-84]. Using fluorescence in situ hybridization (FISH) we investigated amplification of chromosome band 1q21 (Amp1q21) in 479 untreated MM patients and 22 MM cell lines. The frequency of Amp1q21 was 43% in newly diagnosed MM, but was present in 21 of 22 (95%) in MM cell lines. We also correlated microarray data with FISH results in plasma cells from 250 patients. This analysis revealed that the percentage of cells with amplification of the CKS1B locus was a significant predictor of CKS1B gene expression level (P < 0.0001). Newly diagnosed MM patients with Amp1q21 had an inferior 5-year overall survival compared with those lacking Amp1q21 (P < 0.001). Also, patients, who had the highest expression of CKS1B (top 13% of 351 patients) at baseline and at relapse, had the worst 5-year overall survival when receiving salvage therapies (P = 0.0025). We further demonstrated that CKS1B plays a crucial role in MM cell growth and survival and for the first time provides direct evidence for the crucial role of CKS1B in myeloma multidrug-resistance [84]. We identified STAT3 and MEK/ERK/BCL2 as CKS1B-downstream signaling pathways; and thereby provided targets for the development of new therapeutic approaches for CKS1B overexpressing myeloma and other malignancies [84].

GEP was also utilized in MMSC CD138- and CD138+ cells isolated from ARP.1, CAG, H929, JJN3, KMS28PE, OCI-MY5, OPM2, U266 and XG1 cell lines and we identified 645 genes that were significantly differentially expressed in CD138- cells compared to CD138+ cells (SAM, FDR<1%), with 598 up-regulated and 47 down-regulated. Our preliminary data based on analysis of 9 MM cell lines show that RARα is the top one up-regulated gene in CD138- MM stem cells. RARα has two major isoforms, RARα1 and RARα2. Real-time PCR detected significantly higher expression of RARα2 but not RARα1 in CD138- MM stem cells compared to CD138+ tumor cells. While RARα1 was ubiquitously expressed in MM cells, RARα2 was present in only 30% of newly diagnosed patients [85]. Patients with RARα2 expression had a significantly shorter overall-survival, also on multivariate analysis. RARα2-knockdown in RARα2+ MM cells induced cell growth arrest and apoptosis [85]. Wnt signaling is activated in MM stem cells and forced-RARα2-expression in MM cells induced Wnt activation These results suggest that RARα2-mediated Wnt signal activation may be critical for MM stem cell survival and MM progression. Both RARα1 and RARα2 are specific receptors for all-trans retinoic acid (ATRA). Interestingly, we found that ATRA selectively killed MM stem cells while sparing CD138+ tumor cells. Forced expression of RARα2 in RA-Rα2-deficient MM tumor cells restored sensitivity to ATRA. Finally, ATRA was shown to down-regulate Wnt signaling (β catenin and other downstream targets).

We also compared GEP samples at baseline, in remission, and at relapse from patients enrolled on our 25009 protocol, and defined 56 genes associated with drug resistance (persistence after transplantation) or rapid disease relapse (< 2 years after the second transplant). The chromosomal instability signatures (CINs) were identified as the most differentially expressed genes, with low expression at diagnosis and high expression after chemotherapy (at a stage with minimal residual disease) and at relapse. Our functional assays indicate that increased CIN signature promotes MMSC self-renewal and induces drug resistance by up-regulating ABCG2 efflux and the Wnt pathway (submitted manuscript). We are currently working on verifying these novel MMSC-specific targets with the goal of using these findings in future MM therapies.

However, even if a potential drug candidate is found that specifically targets the self-renewal pathways, there must also be a more selective targeting of MMSC compared with normal HSC. Better targeted combinatorial therapy using the self-renewal pathway inhibitor plus either traditional chemotherapeutic drugs (as in Table 2) or a selective inhibitor of MMSCs and not HSCs needs to be developed. For example, a potential target, cell surface receptor CS1, is highly expressed in the bulk of the myeloma cells but not HSC, has been targeted successfully with a monoclonal antibody that blocks CS1 activity and inhibits MM cell bone adhesion [86, 87]. Antibodies raised against CD44, an acute myeloid leukemia SC marker, successfully eliminated CSCs in vivo in a NOD-SCID mouse model [88]. Similarly, in MM, CD20 antibody was demonstrated to inhibit CD138- clonogenic recovery in a complement-dependent manner [18] although CD20 is only expressed in a minority of MM patients and response to CD20 antibody treatment is variable as tabulated by Kapoor et al. [89].

Future directions

What is a cancer stem cell? Ideally the answer should only depend on the type of cancer, but currently in MM the CSC phenotype is still poorly defined as is the concept of CSC itself. Even if the MMSC signature is found, there are still many questions to answer: Can MMSCs be expanded in in vitro cultures? How do MMSCs interact with the bulk tumor? What is its niche? Are the MMSC always the same cells or is there plasticity as has been shown in HSC?

The physical isolation of the MMSC intrinsically disturbs the CSC niche and could contribute to the differences in phenotype being observed but it is still of vital importance to identify this (these?) subpopulation(s) if a cure for MM is to ever to be found. Single cell in vitro assays can be used to compare the absolute clonogenicity of the various potential MMSC populations. A distinct advantage of studying MM is that the HSC system is well established and the blood cancer leukemia stem cell system serves as a guide for elucidating the MMSC system. If a definitive MMSC signature / marker attains universal acceptance, microarray profiling could be used to associate this signature / marker with a prognosis for MM patient survival. The Morrison group used GEP to find the leukemia SC marker SLAM [10]. Similarly, our group has performed GEP between new and relapsed patients, and between CD138- and CD138+ MM cell lines and have found promising targets that seem to be involved in drug resistance and self-renewal [84, 87, 90]. Targeting of MMSC will also have a different therapeutic profile than traditional drugs, and attacking the bulk tumor, the stem cell niche, and the microenvironment needs to be considered especially if the more differentiated bulk tumor might have the capacity to revert to the stem cell-like phenotype [31, 34].

Acknowledgments

This work was supported by National Cancer Institute R01CA115399 (GT), R01CA152105 (FZ), R21CA143887 (FZ), leukemia lymphoma society (FZ), Multiple Myeloma Research Foundation (FZ 2008 & 2010), and institutional start -up funds from the School of Medicine and Huntsman Cancer institute of the University of Utah (FZ).

References

  • 1.Chng W, Glebov O, Bergsagel P, Kuehl W. Genetic events in the pathogenesis of multiple myeloma. Best Pract Res Clin Haematol. 2007;20:571–596. doi: 10.1016/j.beha.2007.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kuehl WM, Bergsagel PL. Multiple myeloma: evolving genetic events and host interactions. Nat Rev Cancer. 2002;2:175–187. doi: 10.1038/nrc746. [DOI] [PubMed] [Google Scholar]
  • 3.Zhan F, Huang Y, Colla S, Stewart JP, Hanamura I, Gupta S, Epstein J, Yaccoby S, Sawyer J, Burington B, Anaissie E, Hollmig K, Pineda-Roman M, Tricot G, van Rhee F, Walker R, Zangari M, Crowley J, Barlogie B, Shaughnessy JD., Jr The molecular classification of multiple myeloma. Blood. 2006;108:2020–2028. doi: 10.1182/blood-2005-11-013458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Shaughnessy JD, Jr, Zhan F, Burington BE, Huang Y, Colla S, Hanamura I, Stewart JP, Kordsmeier B, Randolph C, Williams DR, Xiao Y, Xu H, Epstein J, Anaissie E, Krishna SG, Cottler-Fox M, Hollmig K, Mohiuddin A, Pineda-Roman M, Tricot G, van Rhee F, Sawyer J, Alsayed Y, Walker R, Zangari M, Crowley J, Barlogie B. A validated gene expression model of high-risk multiple myeloma is defined by deregulated expression of genes mapping to chromosome 1. Blood. 2007;109:2276–2284. doi: 10.1182/blood-2006-07-038430. [DOI] [PubMed] [Google Scholar]
  • 5.Kirschstein RL, Skirboll LR, editors. Bethesda: National Institutes of Health; 2001. Stem cells: scientific progress and future research directions. [Google Scholar]
  • 6. Regenerative Medicine. Department of Health and Human Services. August 2006. </info/scireport/2006report.htm>.
  • 7.Baum CM, Weissman IL, Tsukamoto AS, Buckle AM, Peault B. Isolation of a candidate human hematopoietic stem-cell population. Proc Natl Acad Sci U S A. 1992;89:2804–8. doi: 10.1073/pnas.89.7.2804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Spangrude GJ, Heimfeld S, Weissman IL. Purification and characterization of mouse hematopoietic stem cells. Science. 1988;241:58–62. doi: 10.1126/science.2898810. [DOI] [PubMed] [Google Scholar]
  • 9.Krause DS, Theise ND, Collector MI, Henegariu O, Hwang S, Gardner R, Neutzel S, Sharkis SJ. Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell. 2001;105:369–377. doi: 10.1016/s0092-8674(01)00328-2. [DOI] [PubMed] [Google Scholar]
  • 10.Kiel MJ, Yilmaz ÖH, Iwashita T, Yilmaz OH, Terhorst C, Morrison SJ. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 2005;121:1109–1121. doi: 10.1016/j.cell.2005.05.026. [DOI] [PubMed] [Google Scholar]
  • 11.Gojo I, Meisenberg B, Guo C, Fassas A, Murthy A, Fenton R, Takebe N, Heyman M, Philips G, Cottler-Fox M. Autologous stem cell transplantation followed by consolidation chemotherapy for patients with multiple myeloma. Bone marrow transplant. 2005;37:65–72. doi: 10.1038/sj.bmt.1705192. [DOI] [PubMed] [Google Scholar]
  • 12.Lin T, Jones R, Matsui W. Cancer stem cells: relevance to SCT. Bone marrow transplant. 2009;43:517–523. doi: 10.1038/bmt.2009.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bruce W, Van Der Gaag H. A quantitative assay for the number of murine lymphoma cells capable of proliferation in vivo. Nature. 1963;199:79–80. doi: 10.1038/199079a0. [DOI] [PubMed] [Google Scholar]
  • 14.Park C, Bergsagel D, McCulloch E. Mouse myeloma tumor stem cells: a primary cell culture assay. J Natl Cancer Inst. 1971;46:411–22. [PubMed] [Google Scholar]
  • 15.Hamburger AW, Salmon SE. Primary bioassay of human tumor stem cells. Science. 1977;197:461–3. doi: 10.1126/science.560061. [DOI] [PubMed] [Google Scholar]
  • 16.Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3:730–737. doi: 10.1038/nm0797-730. [DOI] [PubMed] [Google Scholar]
  • 17.Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–111. doi: 10.1038/35102167. [DOI] [PubMed] [Google Scholar]
  • 18.Matsui W, Wang Q, Barber JP, Brennan S, Smith BD, Borrello I, McNiece I, Lin L, Ambinder RF, Peacock C. Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance. Cancer Res. 2008;68:190–7. doi: 10.1158/0008-5472.CAN-07-3096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Matsui W, Huff CA, Wang Q, Malehorn MT, Barber J, Tanhehco Y, Smith BD, Civin CI, Jones RJ. Characterization of clonogenic multiple myeloma cells. Blood. 2004;103:2332–6. doi: 10.1182/blood-2003-09-3064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ghosh N, Matsui W. Cancer stem cells in multiple myeloma. Cancer lett. 2009;277:1–7. doi: 10.1016/j.canlet.2008.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Brennan SK, Matsui W. Cancer stem cells: controversies in multiple myeloma. J Mol Med. 2009;87:1079–1085. doi: 10.1007/s00109-009-0531-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Yaccoby S, Epstein J. The proliferative potential of myeloma plasma cells manifest in the SCID-hu host. Blood. 1999;94:3576–3582. [PubMed] [Google Scholar]
  • 23.Jakubikova J, Adamia S, Kost-Alimova M, Klippel S, Cervi D, Daley JF, Cholujova D, Kong SY, Leiba M, Blotta S. Lenalidomide targets clonogenic side population in multiple myeloma: pathophysiologic and clinical implications. Blood. 2011;117:4409–19. doi: 10.1182/blood-2010-02-267344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bergsagel D, Valeriote F. Growth characteristics of a mouse plasma cell tumor. Cancer Res. 1968;28:2187–96. [PubMed] [Google Scholar]
  • 25.Drewinko B, Alexanian R, Boyer H, Barlogie B, Rubinow S. The growth fraction of human myeloma cells. Blood. 1981;57:333–8. [PubMed] [Google Scholar]
  • 26.Hamburger A, Salmon SE. Primary bioassay of human myeloma stem cells. J Clin Invest. 1977;60:846–54. doi: 10.1172/JCI108839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.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] [Google Scholar]
  • 28.Kirshner J, Thulien KJ, Martin LD, Debes Marun C, Reiman T, Belch AR, Pilarski LM. A unique three-dimensional model for evaluating the impact of therapy on multiple myeloma. Blood. 2008;112:2935–45. doi: 10.1182/blood-2008-02-142430. [DOI] [PubMed] [Google Scholar]
  • 29.Peacock CD, Wang Q, Gesell GS, Corcoran-Schwartz IM, Jones E, Kim J, Devereux WL, Rhodes JT, Huff CA, Beachy PA. Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma. Proc Natl Acad Sci U SA. 2007;104:4048–53. doi: 10.1073/pnas.0611682104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Khotskaya YB, Dai Y, Ritchie JP, MacLeod V, Yang Y, Zinn K, Sanderson RD. Syndecan-1 is required for robust growth, vascularization, and metastasis of myeloma tumors in vivo. J Biol Chem. 2009;284:26085–95. doi: 10.1074/jbc.M109.018473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Yaccoby S. The phenotypic plasticity of myeloma plasma cells as expressed by dedifferentiation into an immature, resilient, and apoptosis-resistant phenotype. Clin Cancer Res. 2005;11:7599–606. doi: 10.1158/1078-0432.CCR-05-0523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Pilarski LM, Belch AR. Clonotypic myeloma cells able to xenograft myeloma to nonobese diabetic severe combined immunodeficient mice copurify with CD34+ hematopoietic progenitors. Clin Cancer Res. 2002;8:3198–204. [PubMed] [Google Scholar]
  • 33.Kuranda K, Berthon C, Dupont C, Wolowiec D, Leleu X, Polakowska R, Jouy N, Quesnel B. A subpopulation of malignant CD34+ CD138+ B7-H1+ plasma cells is present in multiple myeloma patients. Exp Hematol. 2010;38:124–131. doi: 10.1016/j.exphem.2009.11.008. e124. [DOI] [PubMed] [Google Scholar]
  • 34.Chaffer CL, Brueckmann I, Scheel C, Kaestli AJ, Wiggins PA, Rodrigues LO, Brooks M, Reinhardt F, Su Y, Polyak K. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc Natl Acad Sci U S A. 2011;108:7950–5. doi: 10.1073/pnas.1102454108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Jones R, Barber J, Vala M, Collector M, Kaufmann S, Ludeman S, Colvin O, Hilton J. Assessment of aldehyde dehydrogenase in viable cells. Blood. 1995;85:2742–6. [PubMed] [Google Scholar]
  • 36.Goodell MA, Brose K, Paradis G, Conner AS, Mulligan RC. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med. 1996;183:1797–806. doi: 10.1084/jem.183.4.1797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Sengupta A, Cancelas JA. Cancer stem cells: A stride towards cancer cure? J Cell Physiol. 2010;225:7–14. doi: 10.1002/jcp.22213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Boman BM, Wicha MS. Cancer stem cells: a step toward the cure. J Clin Oncol. 2008;26:2795–9. doi: 10.1200/JCO.2008.17.7436. [DOI] [PubMed] [Google Scholar]
  • 39.Nirmalanandhan VS, Sittampalam GS. Stem cells in drug discovery, tissue engineering, and regenerative medicine: emerging opportunities and challenges. J Biomol Screen. 2009;14:755–68. doi: 10.1177/1087057109336591. [DOI] [PubMed] [Google Scholar]
  • 40.Cheng L, Ramesh AV, Flesken-Nikitin A, Choi J, Nikitin AY. Mouse models for cancer stem cell research. Toxicol Pathol. 2010;38:62–71. doi: 10.1177/0192623309354109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Morrison SJ. Efficient tumour formation by single human melanoma cells. Nature. 2008;456:593–598. doi: 10.1038/nature07567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Roodman GD. Myeloma bone disease (current clinical oncology) Humana Pr Inc. 2010 [Google Scholar]
  • 43.Brown RD, Ho PJ. Multiple myeloma: methods and protocols. Humana Pr Inc. 2005 [Google Scholar]
  • 44.Anderson KC, Ghobrial IM. Multiple myeloma: translational and emerging therapies. Informa Healthcare. 2007 [Google Scholar]
  • 45.Hope K, Bhatia M. Clonal interrogation of stem cells. Nat Methods. 2011;8:S36–S40. doi: 10.1038/nmeth.1590. [DOI] [PubMed] [Google Scholar]
  • 46.Richardson PG, Sonneveld P, Schuster MW, Irwin D, Stadtmauer EA, Facon T, Harousseau JL, Ben-Yehuda D, Lonial S, Goldschmidt H. Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Engl J Med. 2005;352:2487–2498. doi: 10.1056/NEJMoa043445. [DOI] [PubMed] [Google Scholar]
  • 47.Hideshima T, Mitsiades C, Akiyama M, Hayashi T, Chauhan D, Richardson P, Schlossman R, Podar K, Munshi NC, Mitsiades N. Molecular mechanisms mediating antimyeloma activity of proteasome inhibitor PS-341. Blood. 2003;101:1530–4. doi: 10.1182/blood-2002-08-2543. [DOI] [PubMed] [Google Scholar]
  • 48.Hideshima T, Richardson P, Chauhan D, Palombella VJ, Elliott PJ, Adams J, Anderson KC. The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells. Cancer Res. 2001;61:3071–6. [PubMed] [Google Scholar]
  • 49.Annunziata CM, Davis RE, Demchenko Y, Bellamy W, Gabrea A, Zhan F, Lenz G, Hanamura I, Wright G, Xiao W. Frequent Engagement of the Classical and Alternative NF-[kappa] B Pathways by Diverse Genetic Abnormalities in Multiple Myeloma. Cancer cell. 2007;12:115–130. doi: 10.1016/j.ccr.2007.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Markovina S, Callander NS, O'Connor SL, Kim J, Werndli JE, Raschko M, Leith CP, Kahl BS, Kim KM, Miyamoto S. Bortezomib-resistant nuclear factor- B activity in multiple myeloma cells. Mol Cancer Res. 2008;6:1356–64. doi: 10.1158/1541-7786.MCR-08-0108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, Spicka I, Petrucci MT, Palumbo A, Samoilova OS. Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med. 2008;359:906–917. doi: 10.1056/NEJMoa0801479. [DOI] [PubMed] [Google Scholar]
  • 52.Richardson PG, Weller E, Lonial S, Jakubowiak AJ, Jagannath S, Raje NS, Avigan DE, Xie W, Ghobrial IM, Schlossman RL, Mazumder A, Munshi NC, Vesole DH, Joyce R, Kaufman JL, Doss D, Warren DL, Lunde LE, Kaster S, Delaney C, Hideshima T, Mitsiades CS, Knight R, Esseltine DL, Anderson KC. Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma. Blood. 2010;116:679–686. doi: 10.1182/blood-2010-02-268862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Jordan CT, Guzman ML, Noble M. Cancer stem cells. N Engl J Med. 2006;355:1253–1261. doi: 10.1056/NEJMra061808. [DOI] [PubMed] [Google Scholar]
  • 54.Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444:756–760. doi: 10.1038/nature05236. [DOI] [PubMed] [Google Scholar]
  • 55.Yaccoby S, Johnson CL, Mahaffey SC, Wezeman MJ, Barlogie B, Epstein J. Antimyeloma efficacy of thalidomide in the SCID-hu model. Blood. 2002;100:4162–8. doi: 10.1182/blood-2002-03-0939. [DOI] [PubMed] [Google Scholar]
  • 56.Tassone P, Neri P, Burger R, Savino R, Shammas M, Catley L, Podar K, Chauhan D, Masciari S, Gozzini A. Combination therapy with in-terleukin-6 receptor superantagonist Sant7 and dexamethasone induces antitumor effects in a novel SCID-hu In vivo model of human multiple myeloma. Clin Cancer Res. 2005;11:4251–8. doi: 10.1158/1078-0432.CCR-04-2611. [DOI] [PubMed] [Google Scholar]
  • 57.Ikeda H, Hideshima T, Fulciniti M, Perrone G, Miura N, Yasui H, Okawa Y, Kiziltepe T, Santo L, Vallet S. PI3K/p110 is a novel therapeutic target in multiple myeloma. Blood. 2010;116:1460–8. doi: 10.1182/blood-2009-06-222943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Taipale J, Beachy PA. The Hedgehog and Wnt signalling pathways in cancer. Nature. 2001;411:349–354. doi: 10.1038/35077219. [DOI] [PubMed] [Google Scholar]
  • 59.He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B, Kinzler KW. Identification of c-MYC as a target of the APC pathway. Science. 1998;281:1509–1512. doi: 10.1126/science.281.5382.1509. [DOI] [PubMed] [Google Scholar]
  • 60.Shtutman M, Zhurinsky J, Simcha I, Albanese C, D'Amico M, Pestell R, Ben-Ze'ev A. The cyclin D1 gene is a target of the beta-catenin/LEF -1 pathway. Proc Natl Acad Sci U S A. 1999;96:5522–5527. doi: 10.1073/pnas.96.10.5522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Tetsu O, McCormick F. Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature. 1999;398:422–426. doi: 10.1038/18884. [DOI] [PubMed] [Google Scholar]
  • 62.Wielenga VJ, Smits R, Korinek V, Smit L, Kielman M, Fodde R, Clevers H, Pals ST. Expression of CD44 in Apc and Tcf mutant mice implies regulation by the WNT pathway. Am J Pathol. 1999;154:515–523. doi: 10.1016/S0002-9440(10)65297-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Schwartz DR, Wu R, Kardia SL, Levin AM, Huang CC, Shedden KA, Kuick R, Misek DE, Hanash SM, Taylor JM, Reed H, Hendrix N, Zhai Y, Fearon ER, Cho KR. Novel candidate targets of beta-catenin/T-cell factor signaling identified by gene expression profiling of ovarian endometrioid adenocarcinomas. Cancer Res. 2003;63:2913–2922. [PubMed] [Google Scholar]
  • 64.Tricot G, Sawyer JR, Jagannath S, Desikan KR, Siegel D, Naucke S, Mattox S, Bracy D, Munshi N, Barlogie B. Unique role of cytogenetics in the prognosis of patients with myeloma receiving high-dose therapy and autotransplants. J Clin Oncol. 1997;15:2659–2666. doi: 10.1200/JCO.1997.15.7.2659. [DOI] [PubMed] [Google Scholar]
  • 65.Tricot G, Barlogie B, Jagannath S, Bracy D, Mattox S, Vesole DH, Naucke S, Sawyer JR. Poor prognosis in multiple myeloma is associated only with partial or complete deletions of chromosome 13 or abnormalities involving 11q and not with other karyotype abnormalities. Blood. 1995;86:4250–4256. [PubMed] [Google Scholar]
  • 66.Dohner H, Stilgenbauer S, Benner A, Leupolt E, Krober A, Bullinger L, Dohner K, Bentz M, Lichter P. Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med. 2000;343:1910–1916. doi: 10.1056/NEJM200012283432602. [DOI] [PubMed] [Google Scholar]
  • 67.Avet-Loiseau H, Facon T, Grosbois B, Magrangeas F, Rapp MJ, Harousseau JL, Minvielle S, Bataille R. Oncogenesis of multiple myeloma: 14q32 and 13q chromosomal abnormalities are not randomly distributed, but correlate with natural history, immunological features, and clinical presentation. Blood. 2002;99:2185–2191. doi: 10.1182/blood.v99.6.2185. [DOI] [PubMed] [Google Scholar]
  • 68.Moreau P, Facon T, Leleu X, Morineau N, Huyghe P, Harousseau JL, Bataille R, Avet-Loiseau H. Recurrent 14q32 translocations determine the prognosis of multiple myeloma, especially in patients receiving intensive chemotherapy. Blood. 2002;100:1579–1583. doi: 10.1182/blood-2002-03-0749. [DOI] [PubMed] [Google Scholar]
  • 69.Smadja NV, Leroux D, Soulier J, Dumont S, Arnould C, Taviaux S, Taillemite JL, Bastard C. Further cytogenetic characterization of multiple myeloma confirms that 14q32 translocations are a very rare event in hyperdiploid cases. Genes Chromosomes Cancer. 2003;38:234–239. doi: 10.1002/gcc.10275. [DOI] [PubMed] [Google Scholar]
  • 70.Fonseca R, Bergsagel PL, Drach J, Shaughnessy J, Gutierrez N, Stewart AK, Morgan G, Van Ness B, Chesi M, Minvielle S, Neri A, Barlogie B, Kuehl WM, Liebisch P, Davies F, Chen-Kiang S, Durie BG, Carrasco R, Sezer O, Reiman T, Pilarski L, Avet-Loiseau H. International Myeloma Working Group molecular classification of multiple myeloma: spotlight review. Leukemia. 2009;23:2210–2221. doi: 10.1038/leu.2009.174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Chiecchio L, Protheroe RK, Ibrahim AH, Cheung KL, Rudduck C, Dagrada GP, Cabanas ED, Parker T, Nightingale M, Wechalekar A, Orchard KH, Harrison CJ, Cross NC, Morgan GJ, Ross FM. Deletion of chromosome 13 detected by conventional cytogenetics is a critical prognostic factor in myeloma. Leukemia. 2006;20:1610–1617. doi: 10.1038/sj.leu.2404304. [DOI] [PubMed] [Google Scholar]
  • 72.Zhan F, Sawyer J, Tricot G. The role of cytogenetics in myeloma. Leukemia. 2006;20:1484–1486. doi: 10.1038/sj.leu.2404330. [DOI] [PubMed] [Google Scholar]
  • 73.Valk PJ, Verhaak RG, Beijen MA, Erpelinck CA, Barjesteh van Waalwijk van Doorn-Khosrovani S, Boer JM, Beverloo HB, Moorhouse MJ, van der Spek PJ, Lowenberg B, Delwel R. Prognostically useful gene-expression profiles in acute myeloid leukemia. N Engl J Med. 2004;350:1617–1628. doi: 10.1056/NEJMoa040465. [DOI] [PubMed] [Google Scholar]
  • 74.Shipp MA, Ross KN, Tamayo P, Weng AP, Kutok JL, Aguiar RC, Gaasenbeek M, Angelo M, Reich M, Pinkus GS, Ray TS, Koval MA, Last KW, Norton A, Lister TA, Mesirov J, Neuberg DS, Lander ES, Aster JC, Golub TR. Diffuse large B-cell lymphoma outcome prediction by gene-expression profiling and supervised machine learning. Nat Med. 2002;8:68–74. doi: 10.1038/nm0102-68. [DOI] [PubMed] [Google Scholar]
  • 75.Yeoh EJ, Ross ME, Shurtleff SA, Williams WK, Patel D, Mahfouz R, Behm FG, Raimondi SC, Relling MV, Patel A, Cheng C, Campana D, Wilkins D, Zhou X, Li J, Liu H, Pui CH, Evans WE, Naeve C, Wong L, Downing JR. Classification, subtype discovery, and prediction of outcome in pediatric acute lymphoblastic leukemia by gene expression profiling. Cancer Cell. 2002;1:133–143. doi: 10.1016/s1535-6108(02)00032-6. [DOI] [PubMed] [Google Scholar]
  • 76.van't Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, Peterse HL, van der Kooy K, Marton MJ, Witteveen AT, Schreiber GJ, Kerkhoven RM, Roberts C, Linsley PS, Bernards R, Friend SH. Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002;415:530–536. doi: 10.1038/415530a. [DOI] [PubMed] [Google Scholar]
  • 77.Zhan F, Hardin J, Kordsmeier B, Bumm K, Zheng M, Tian E, Sanderson R, Yang Y, Wilson C, Zangari M, Anaissie E, Morris C, Muwalla F, van Rhee F, Fassas A, Crowley J, Tricot G, Barlogie B, Shaughnessy J., Jr Global gene expression profiling of multiple myeloma, monoclonal gammopathy of undetermined significance, and normal bone marrow plasma cells. Blood. 2002;99:1745–1757. doi: 10.1182/blood.v99.5.1745. [DOI] [PubMed] [Google Scholar]
  • 78.Zhan F, Barlogie B, Arzoumanian V, Huang Y, Williams DR, Hollmig K, Pineda-Roman M, Tricot G, van Rhee F, Zangari M, Dhodapkar M, Shaughnessy JD., Jr Gene-expression signature of benign monoclonal gammopathy evident in multiple myeloma is linked to good prognosis. Blood. 2007;109:1692–1700. doi: 10.1182/blood-2006-07-037077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Tian E, Zhan F, Walker R, Rasmussen E, Ma Y, Barlogie B, Shaughnessy JD., Jr The role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. N Engl J Med. 2003;349:2483–2494. doi: 10.1056/NEJMoa030847. [DOI] [PubMed] [Google Scholar]
  • 80.Colla S, Zhan F, Xiong W, Wu X, Xu H, Stephens O, Yaccoby S, Epstein J, Barlogie B, Shaughnessy JD., Jr The oxidative stress response regulates DKK1 expression through the JNK signaling cascade in multiple myeloma plasma cells. Blood. 2007;109:4470–4477. doi: 10.1182/blood-2006-11-056747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Bergsagel PL, Kuehl WM, Zhan F, Sawyer J, Barlogie B, Shaughnessy J., Jr Cyclin D dys-regulation: an early and unifying pathogenic event in multiple myeloma. Blood. 2005;106:296–303. doi: 10.1182/blood-2005-01-0034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Hanamura I, Stewart JP, Huang Y, Zhan F, Santra M, Sawyer JR, Hollmig K, Zangarri M, Pineda-Roman M, Van Rhee F. 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. 2006;108:1724–32. doi: 10.1182/blood-2006-03-009910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Zhan F, Colla S, Wu X, Chen B, Stewart JP, Kuehl WM, Barlogie B, Shaughnessy JD. CKS1B, overexpressed in aggressive disease, regulates multiple myeloma growth and survival through SKP2-and p27Kip1-dependent and-independent mechanisms. Blood. 2007;109:4995–5001. doi: 10.1182/blood-2006-07-038703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Shi L, Wang S, Zangari M, Xu H, Cao TM, Xu C, Wu Y, Xiao F, Liu Y, Yang Y, Salama M, Li G, Tricot G, Zhan F. Over-expression of CKS1B activates both MEK/ERK and JAK/STAT3 signaling pathways and promotes myeloma cell drug-resistance. Oncotarget. 2010;1:22–33. doi: 10.18632/oncotarget.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wang S, Tricot G, Shi L, Xiong W, Zeng Z, Xu H, Zangari M, Barlogie B, Shaughnessy JD, Jr, Zhan F. RARalpha2 expression is associated with disease progression and plays a crucial role in efficacy of ATRA treatment in myeloma. Blood. 2009;114:600–607. doi: 10.1182/blood-2008-12-194126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Hsi ED, Steinle R, Balasa B, Szmania S, Draksharapu A, Shum BP, Huseni M, Powers D, Nanisetti A, Zhang Y. CS1, a potential new therapeutic antibody target for the treatment of multiple myeloma. Clin Cancer Res. 2008;14:2775–84. doi: 10.1158/1078-0432.CCR-07-4246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Tai YT, Dillon M, Song W, Leiba M, Li XF, Burger P, Lee AI, Podar K, Hideshima T, Rice AG. Anti-CS1 humanized monoclonal antibody Hu-Luc63 inhibits myeloma cell adhesion and induces antibody-dependent cellular cytotoxicity in the bone marrow milieu. Blood. 2008;112:1329–37. doi: 10.1182/blood-2007-08-107292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med. 2006;12:1167–1174. doi: 10.1038/nm1483. [DOI] [PubMed] [Google Scholar]
  • 89.Kapoor P, Greipp PT, Morice WG, Rajkumar SV, Witzig TE, Greipp PR. Anti CD20 monoclonal antibody therapy in multiple myeloma. British J Haematol. 2008;141:135–148. doi: 10.1111/j.1365-2141.2008.07024.x. [DOI] [PubMed] [Google Scholar]
  • 90.Anguiano A, Tuchman SA, Acharya C, Salter K, Gasparetto C, Zhan F, Dhodapkar M, Nevins J, Barlogie B, Shaughnessy JD. Gene expression profiles of tumor biology provide a novel approach to prognosis and may guide the selection of therapeutic targets in multiple myeloma. J Clin Oncol. 2009;27:4197–203. doi: 10.1200/JCO.2008.19.1916. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Blood Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES