Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2013

Open Access 01.12.2013 | Review

Fact or fiction - identifying the elusive multiple myeloma stem cell

verfasst von: Joshua Kellner, Bei Liu, Yubin Kang, Zihai Li

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2013

Abstract

Multiple Myeloma (MM) is a debilitating disease of proliferating and malignant plasma cells that is currently incurable. The ability of monoclonal recurrence of disease suggests it might arise from a stem cell-like population capable of self-renewal. The difficulty to isolate the cancer stem-like cell in MM has introduced confusion toward this hypothesis. However, recent evidence has suggested that MM originates from the B cell lineage with memory-B cell like features, allowing for self-renewal of the progenitor-like status and differentiation to a monoclonal plasma cell population. Furthermore, this tumor-initiating cell uses signaling pathways and microenvironment similar to the hematopoietic stem cell, though hijacking these mechanisms to create and favor a more tumorigenic environment. The bone marrow niche allows for pertinent evasion, either through avoiding immunosurveillance or through direct interaction with the stroma, inducing quiescence and thus drug resistance. Understanding the interaction of the MM stem cell to the microenvironment and the mechanisms utilized by various stem cell-like populations to allow persistence and therapy-resistance can enable for better targeting of this cell population and potential eradication of the disease.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1756-8722-6-91) contains supplementary material, which is available to authorized users.

Competing interest

The authors declare that they have no competing interest.

Authors’ contributions

All authors wrote, read and approved the final manuscript.

Background

Stem cells are classified as cells that are pluripotent and can propagate the cells of a specific lineage while also maintaining self-renewal. Recent evidence has suggested that cancer has exploited this unique machinery and contains a stem-like population that maintains and propagates disease. The current paradigm regarding the cancer stem cell (CSC) is that the tumor either arises from a normal stem cell or inherently contains a “tumor” stem cell that drives tumor formation. However, it is debatable if these paradigms can apply to all cancers or if they are unique to several specific cancer types. Two initial studies laid claim to the hypothesis of CSCs. Bergsagel et al. characterized a low frequency population with tumorigenicity in a plasma cell tumor model of inflammation which led to studies where murine myeloma cells, isolated from ascites, could form in-vitro colonies at a ratio of only 1 in 10,000 to 1 in 100 cells [1, 2]. Various solid tumors, such as lung and ovarian cancer, also exhibited a high degree of tumor-initiating heterogeneity with only a small subset of the tumor population exhibiting clonogenic potential [3]. The development of an in vitro assay to study human myeloma clonogenic cells furthered the work delving into the hypothesis of a progenitor cancer cell [4]. However, it was the work from Dick and colleagues who identified a CSC from a specific subset of acute myelogenous leukemia (AML) cells that had demonstrated the clonogenic activity of a particular isolated population with confidence [5]. This minute population, demonstrating a varied frequency of about 0.2% in some patients, was capable of transfer disease into immunodeficient NOD/SCID mice. These studies suggest the presence of a CSC but it is difficult to determine whether they are generated from a mutational hit on normal stem cells or from a specific primitive tumor stem cell. The ability to isolate primitive hematopoietic stem cells (HSC) and our understanding of the stem-like mechanisms of HSCs has enabled better understanding of CSCs in leukemias but has proven to be more difficult in myeloma.

Pathology of disease

MM is an incurable blood malignancy characterized by extensive proliferation of plasma cells (PC) and displaying an incidence of about 20,000 annually in the United States [6]. The tumorigenic PCs secrete monoclonal immunoglobulin and induce a wide range of pathology including lytic bone disease, hypercalcemia, immunodeficiency, anemia and kidney and bone marrow (BM) dysfunction [7]. Nearly all MM patients derive from asymptomatic monoclonal gammopathy of undetermined significance (MGUS). Patients could present as smoldering MM phase that then progresses to advanced symptomatic phases of MM, which include an active, relapsing and refractory periods [8]. Various treatments for MM have been developed including corticosteroids, DNA alkylating agents, immune-modifying drugs, proteasome inhibitors and hematopoietic stem cell transplantation (SCT). Over the last decade, the overall survival of MM patients has improved from a median of 3–4 years to currently at 5–7 years, largely due to the use of several highly active chemoagents and the incorporation of autologous HSC transplantation. However, almost all MM patients will relapse [9]. This high relapse rate in MM patients has suggested the possibility of a CSC that can drive disease progression.

Evidence for the multiple myeloma stem cell

Background

The supposition of a multiple myeloma stem cell (MMSC) has been made for a few decades but identification of the exact cell or population has been difficult to accomplish. Biologically, B cells are derived from the common lymphoid progenitor cell and driven through pro-B to pre-B cell subsets by activation of transcription factors and subsequent expression of the μ chain immunoglobulin and rearrangement of the heavy chain. Development then moves to secondary lymphoid organs (i.e. spleen, lymph nodes) where exposure to antigens induces generation of germinal centers, somatic hypermutation at the Ig locus and proliferation to create clonal-specific memory and short-term and long-term antibody-secreting plasma cells (PCs) that can respond to subsequent antigen exposures. Memory and long-term PCs reside in the BM where they receive support from the BM stroma for survival and activation. The specific cell population, within the B cell/PC lineage, that contains the supposed MM CSC, however, is still unknown.

Cellular identification

The key cellular component of MM, the monoclonal (M) protein-secreting plasma cell, is a terminally differentiated cell type that arises from the B cell lineage. The pathological highlights of MM suggest, however, that malignancy is incurred in B cells and not in the plasma cell population. Early studies demonstrated that in MGUS and MM patients, a fraction of B cells, branded as clonotypic B cells, were present at differentiated states; though this population exhibited heterogeneity [10, 11]. Additionally, Bergsagel et al. identified these clonotypic cells in assorted numbers among patients, with steady levels observed during treatment but significantly higher levels during relapse states [12]. These circulating B cells also had chromosomal aberrations and Ig rearrangements particular to a certain idiotype seen in the malignant PC [1315]. These cells also could differentiate into antibody secreting plasma cells suggesting that the progenitor population of myeloma is contained in the B cell fraction and has progenitor-like characteristics [16]. Further studies of these B cells identified somatic hypermutation in the VDJ region of the genome with deficiency of intraclonal variation suggesting a post-germinal center B cell [10, 17]. Phenotypic studies of these circulating clonotypic B cells demonstrated that they resembled memory B cells, a post-germinal center, pre-plasma cell generated to establish long-term immunity [18]. This property along with the ability to self-renew suggested that this phenotype may be the population containing the malignant myeloma stem cell but the research by Rasmussen et al. [18] has been the only reported suggestion of memory B cells as the myeloma CSC population.

Biological characteristics

Biological activity of the proposed MMSCs has been variable due to the plasticity of the surface markers and the assay used to determine clonogenic activity. This has led to some confusion regarding the surface marker phenotype of the MMSC population (summarized in Table 1). The first model to understand the biology of myeloma stem cells was done by directly injecting myeloma cells from the BM of patients into a subcutaneously implanted human fetal bone chip in SCID mice (named as SCID-hu) [19]. These mice developed clinical characteristics of MM, such as hypercalcemia and circulating M protein. In a later study, it was reported that cells from reconstituted SCID-hu mice were able to engraft secondary SCID recipients, which validates the transferable phenotype of a stem cell population [20]. However, the engrafted population was primarily a CD38++CD45- surface phenotype and no CD19+ B cell was capable of growth [20]. A secondary study has also identified the CD19-CD45-CD38 + CD138+ population as being the tumorigenic stem-like population for MM in another mouse model [21].
Table 1
List of cell surface markers utilized to identify proposed multiple myeloma stem cell
Phenotype
References
CD19 + CD38-CD27+
Rasmussen, T et al., Leukemia and Lymphoma 2004 [18]
CD19 + CD138-
Pilarski, LM et al., Blood 2000 [22]
Pilarski, LM et al., Exp Hematology 2002 [23]
Matsui, W et al., Blood 2004 [24]
CD19 + CD138-CD27+
Matsui, W et al., Cancer Research 2008 [25]
CD138-ALDH+
Reghunathan, R et al., Oncotarget 2013 [26]
Matsui, W et al., Cancer Research 2008 [25]
CD19 + CD34-Lchain(λ) + ALDH+
Boucher, K et al., Clinical Cancer Research 2012 [55]
CD38 + CD45-
Yaccoby, S et al., Blood 1999 [20]
CD19-CD45-CD38 + CD138+
Kim, D et al., Leukemia 2012 [21]
In stark contrast though, Pilarski et al. isolated clonotypic circulating B cells from a progressed MM patient, with the ability to engraft immunodeficient mice and demonstrating clinical phenotypes of lytic bone disease and the presence of circulating M-protein [22]. Furthermore, this leukemic B cell had Ig rearrangements identical to the CD138+ plasma cell, suggesting the involvement of the CD19+ B cell as the progenitor population [14]. The ability to transfer into secondary recipients was not performed, however. Nonetheless, this study was subsequently followed to better ascertain the surface marker phenotype of the myeloma CSC. CD19+ cells lacking the plasma cell marker, CD138/syndecan1, were able to give rise to the tumor population in NOD/SCID mice generating both CD19+ and CD138+ myeloma cells [23]. This further suggested that the myelomagenic population was contained in the B cell lineage but not in the plasma cell pool.
Another study confirmed the lack of CD138 expression in the MMSC phenotype, as primary myeloma BM samples expressing CD138+ were unable to engraft NOD/SCID mice, with the engraftment potential contained solely in the CD138- cell population, incurring plasma cell proliferation and inducing production of M protein in vivo[24]. In vitro colony forming assays further demonstrated the clonogenic capacity of CD138- and not CD138+ cells validating the in vivo transplantation studies. Additional studies to identify the cell surface subset of myeloma CSCs found that the cells resembled a memory B subset in that the clonogenic population expressed CD19 + CD27 + CD138 [25]. This population, obtained from peripheral blood of myeloma patients, engrafted NOD/SCID animals and was transferrable to secondary recipients as the CD19+ cells from the BM of the primary mice engrafted. Additionally, the potential validity of CD138- MMSC and “stemness” has been described by Reghunathan et al. using human MM cell lines to demonstrate the CD138- MMSC neoplasticity [26].
Though research suggests conflicting subsets in identification of the MMSC, the issue may be simply due to the source of the cells, the isolation procedure of these cells or the in vivo or in vitro assay used to determine potential clonogenicity and progenitor status. Collectively, however, the MMSC population appears to reside in the B cell lineage but not in the plasma cell pool.

Signaling pathways

CSCs utilize many of the pathways that regulate and maintain normal stem cells, adapting the ability to self-renew to maintain the malignancy. A feature identified from embryonic stem cells (ESC) to HSCs is the use of pathways established in many developmental mechanisms, including Hedgehog (Hh), Wnt and Notch pathways. Early reports demonstrated the role of these pathways in a number of cancers establishing the manipulation of self-renewal mechanisms by malignant cells to continue disease progression [2730].
Hedgehog signaling was the first to be implicated in the maintenance of MM CSCs demonstrating overexpression in the pathway both from human myeloma cell lines and primary human myeloma samples [31]. Biologically, Hedgehog is involved in stem cell maintenance of ESCs and utilizes a ligand-receptor mechanism of Hh to Patched 1 (Ptch1) to the receptor Smoothened to induce activation of the pathway [32]. Cyclopamine, which targets and inhibits Smoothened, was found to induce apoptosis in MM cells [31]. The use of cyclopamine in specifically treating cancer stem cells was demonstrated in lung and prostate cancer studies, inducing apoptosis and inhibiting growth of the malignant cells [33, 34]. One clinical trial utilizing a small molecule inhibitor of the Hedgehog pathway demonstrated significant responses in over 50% of advanced metastatic basal cell carcinoma patients [35]. Another clinical use of this molecule was performed in a medulloblastoma patient with some success [36]. However, the clinical use of Hedgehog inhibitors in treating MM has not been reported.
The Wnt pathway utilizes 19 conserved glycoproteins that bind to the transmembrane receptor Frizzled, activating canonical β-catenin signaling and noncanonical pathways to induce proliferation and activation [37]. In fact, genetic manipulation of normal Wnt signaling affects the development and function of multiple organs [38]. Aberrant activation of the Wnt pathway promotes proliferation of both MM cell lines and primary patient samples [39]. This activation is induced by intracellular mechanisms and through crosstalk with the BM microenvironment [4043]. Small molecule inhibitors of the Wnt pathway have disrupted the maintenance of MM cells both in vitro and in vivo providing the possibility of developing Wnt-targeted inhibitors for clinical treatment of MM [44, 45] gp96 is a molecular chaperone in the endoplasmic reticulum and it is regulated by the unfolded protein response pathway [46]. It was shown recently that gp96 is a critical chaperone for Wnt co-receptor LRP6 [47]. Targeted inhibition of gp96 genetically and pharmacologically has shown to be an effective strategy against multiple myeloma through inhibition of Wnt-LRP6-surivivin pathway [48].
The Notch signaling pathway is involved in various cellular events from proliferation, differentiation and apoptosis to cell maintenance and survival [49]. The expression of Notch in stem-like populations in various cancers promotes survival of the CSC and progression of disease [5054]. Activation of Notch in MM promotes proliferation and induces enhanced development of the disease [5557]. One study investigated the expression of Notch on BM clonotypic B cells from MM patients and found high expression of Notch on these cells indicating an involvement of Notch signaling in maintaining the MMSC [58]. Inhibitors of Notch signaling have successfully prevented localization and migration of MM cells to the BM and induced apoptosis of these cells but this has not been attempted in clinical settings [5961].
These studies underlie the fact that CSCs, and more specifically, MMSCs, utilize mechanisms similar to normal stem cells to survive, maintain disease and increase progression of disease. Identifying the specific MMSC population that maintains MM would be advantageous in developing a therapy unique to targeting these cells.

Recurrence and disease progression

Current therapeutics

Treatment of MM has largely been established through utilization of chemotherapy. Weder in 1950 published the first successful treatment of MM through the use of urethane, an ethyl carbamate that had little success in treating other hematological malignancies such as leukemia [62]. The use of melphalan and cyclophosphamide and the corticosteroids prednisone and dexamethasone enhanced the treatment of MM and the subsequent combination of these drugs has improved the treatment response rate and the disease progression-free survival [6366]. However, these treatments still did not produce greater overall survival and long-term remission, requiring the development of better therapies and treatments [67]. Immunomodulatory agents, thalidomide and lenalidomide, both have had promising results in improving time to remission and survival in newly diagnosed and relapsed MM [68, 69]. Proteasome inhibitors such as bortezomib and carfilzomib are effective in the treatment of multiple myeloma. Autologous hematopoietic stem cell transplantation has also been used as therapy in healthy, fit patient population. Induction chemotherapy combining 2–3 drugs with immunomudulatory agent, proteasome inhibitor and corticosteroid, followed by autologous SCT and post-transplant maintenance therapy has become a gold standard of therapy for myeloma. This approach has significantly improved the response rate and patients’ overall outcome. Other novel agents have also emerged for the treatment of multiple myeloma including HSP90 inhibitors [70], Bruton’s tyrosine kinase (BTK) inhibitors [71], and novel immunomodulating agents [72, 73]. Unfortunately these therapies have been unable to provide complete eradication of disease. Nearly all MM patients will eventually relapse and become resistant to currently available chemoagents. The exact cause for the relapse remains to be defined and may vary between individual patients. One of the possibilities for the relapse is the persistence of a CSC in myeloma [9]. Many mechanisms involving CSC dormancy have been proposed including evasion of anti-tumor immunity by the tumor cell, the ability of the immune system to control residual tumor cells and the lack of an optimal microenvironment for growth [74].

Microenvironment and drug resistance

The capacity for the CSC to remain dormant at length requires interaction with a specialized niche or microenvironment for optimal support. It is unknown whether MMSC resides in BM or in the post-germinal center of secondary lymphoid organs. It was postulated that similar to B cells and plasma cells, MMSCs are maintained at specific niches in the BM for a long-term survival [75, 76]. The BM microenvironment is characterized by extracellular matrix (ECM) components, including collagens, fibronectin and laminin and cellular parts including hematopoietic cells, BM stromal cells, BM endothelial cells, osteoblasts and osteoclasts [7678] (See Figure 1). Extracellular signaling and cell-cell interactions maintain the homeostatic environment and contribute to MM pathogenesis and maintenance. The various BM stromal cells secrete factors including interleukin 6 (IL6), RANK ligand (RANKL), insulin-like growth factor 1 (IGF1), tumor necrosis factor alpha (TNFα), vascular endothelial growth factor (VEGF), B cell activating factor (BAFF) and stromal cell-derived factor 1 alpha (SDF1) which are required for normal cell function and exacerbate MM disease progression [7983]. A paracrine loop is stimulated in that MM adhesion to the BM niche induces a response by stromal cells to secrete TNFα and VEGF which upregulate IL6 secretion [8486].
The bone cellular components, osteoblasts and osteoclasts, are involved in MM. Osteoblasts secrete IL6 which induces bone lysis along with MM cell proliferation [87, 88]. They also secrete osteoprotegerin (OPG), which prevents TRAIL-mediated death in MM cells [89]. Osteoclasts are involved in bone destruction and seen to be over proliferative in patients [90]. The cells are activated by RANKL, IL-3 and IL-6, all of which are secreted heavily by BM stromal cells [91]. RANK is expressed on osteoclast progenitor cells, inducing their differentiation to osteoclasts and further exacerbating bone lytic damage [92].
Another important mechanism in cell-niche interaction of normal and cancer cells are the family of integrins [93]. Very late antigen 4 (VLA-4), which is composed of α4 and β1, and VLA-5, which includes α5 and β1, are both heavily involved in hematopoietic cell and plasma cell adhesion [94, 95]. Interestingly, there is a downregulation of VLA-5 on progressing malignant PCs and an upregulation of VLA-4 [94]. Binding of VLA-4 to the niche induces NFκB activation in MM cells and induces a cell adhesion-mediated drug resistance (CAM-DR) [95]. Interestingly, the overexpression of VLA-4 is found dramatically in resistant plasma cells. CAM-DR, initially identified in MM, has been attributed to other stromal-microenvironment diseases including glioblastoma and acute myelogenous leukemia [96, 97]. The SDF-1–CXCR4 signaling pathway also plays a critical role in MM cell maintenance. CXCR4 knockdown prevented MM cell line migration to SDF-1 demonstrating the need for this receptor for proper localization [98, 99].
The microenvironment of the BM critically maintains various different cells, such as HSCs and memory B cells, for long-term functional responses. Interestingly, many of the signaling molecules involved in maintaining self-renewal of HSCs are involved in propagation of MM indicating that MMSCs may hijack these pathways to maintain long-term survival and maintenance (Figure 1).

Immune system and tumor dormancy

Though the mechanisms contributing to CSC dormancy may be collective, the immune system plays an important role in either inducing suppressive function or being inhibited by tumor cells. Tumor dormancy has been recognized in AML, CML and breast cancer, where circulating or residual tumor cells are detected though complete remission by clinical standards has been demonstrated [100102]. A classic model of studying dormant CSCs has been with the DA1-3B Bcr-Abl murine model [103]. Although leukemic cell vaccination by gene transfer (CD80, CD154 and GM-CSF) can induce a tumor dormancy phenomenon compatible with long-term survival, residue disease does persist in a large number of mice [104]. Over time, the lack of sensitivity provided an evasion mechanism inducing a more progressive disease. Further studies have demonstrated that the resistant tumor cells had altered the immune surveillance mechanisms, changing the response by the host immune system and providing protection to targeted cell death [105]. In these tumor studies, blocking CTLA4 enhanced CTL-mediated death of tumor cells, a reason for the development of anti-CTLA4 treatment in the clinic [106]. Specifically, this phenomenon of immune dormancy and CSCs has been demonstrated in colon carcinoma and melanoma [107]. With regard to patients with MM, evidence of immune suppression is abundantly clear from frequent infections to the development of secondary malignancies. MM CSCs may evade the immune system via altering the homeostasis of the various cells in the immune compartments.

T cells

Early reports have shown that decreases in CD4 and CD8 T cells negatively correlate with survival in MM and loss of T cell function was demonstrated in MM patients indicating an important role for the immune system in MM [108, 109]. Of recent note, there have been studies implicating the role of T regulatory (Treg) cells and Th17 cells in MM pathogenesis (Figure 2). The role of Tregs in MM has been conflicting as evidence has pointed to both increased and decreased numbers of Tregs in MM and MGUS patients [109, 110]. Though differences in identification of Tregs may contribute to the contradictory results, another study correlated an increased number of Tregs with advanced pathology and disease progression providing a link between Treg numbers and disease status [111]. A study into Th17 cells in the BM of MM patients found there was a significant increase in numbers compared to the peripheral blood (PB), a result not seen in MGUS patients [112]. Elevated levels of Th17 cells can induce myeloma cell proliferation in vitro and in vivo and the increased levels of IL6 and TGFβ, typically observed in MM patients, can contribute to the differentiation and proliferation of Th17 cells [113]. These data suggest that T cells may contribute to the high frequency of relapse in MM patients, inhibiting anti-tumor immunity and inducing inflammation and proliferation of cells.

MDSCs and NKs

Myeloid-derived suppressor cells (MDSC) are an immature myeloid population naturally suppressing natural killer (NK) cells, natural killer T cells (NKT) and anti-tumor activity induced by T cells. Suppression is induced by secretion of ROS, COX-2, nitric oxide, IL6 and IL10, among others. A recent report showed that MDSC populations in BM and peripheral blood of MM patients was dramatically increased compared to control donors, correlating with disease progression [114]. Conversely, NK cells, which have cytotoxic activity on tumor cells, have demonstrated different levels of activity correlating with MM disease stage [115]. In early stages of disease, MM has higher expression of NKG2D ligand such as MHC class I related chain protein A (MICA), inducing NKG2D-triggered cell lysis. However, resistance to NK lytic mechanisms is incurred upon more progressive disease with less surface MICA. To further validate the role of NK cells, another report demonstrated that levels of NKT cells were significantly decreased in advanced stage MM versus early MM, MGUS or control patients [116]. This suggests that the cyto-lytic compartment of the immune system is attenuated as disease becomes more progressive providing further tumor dormancy.

Conclusion

Many studies have looked into the hypothesis of the MMSC with regard to disease relapse of MM, however with little success. The most significant problem is the difficulty to definitely isolate the MMSC to study the unique biology of this cell type. Until this occurs, researchers are left trying to understand the progression of myeloma without knowledge of the start. Though the complexities of specific translocations and mutations in the genetic structure of B cells contribute to the tumorigenic nature of the MMSC, the cell must reside in a hospitable niche, to sustain long-term survival. Understanding the interaction of the MMSC with the surrounding BM microenvironment will enable us to ascertain the required elements for MMSC maintenance and avoidance from therapies. Furthermore, the evasion from immunosurveillance needs to be better studied to comprehend potential ways of targeting the cell. Interestingly, there are many similarities between the HSC and MMSC concerning extracellular and intracellular receptors and signaling. The understanding we have on the complexities required for self-renewal and maintenance of the HSC in the BM could be applied to the MMSC to potentially identify and eradicate the cancer stem cell from the BM environment. Utilizing human cell products and transferring them into mice does not allow for efficient studying of the host microenvironment, particularly related to the roles of immune system in either controlling or later promoting MM. Studies delving into isolating potential tumorigenic populations in syngeneic but humanized mouse models would provide valuable information to study the role of myeloma disease progression, and put into rest the elusive question of the existence of MMSC.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver ( https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​ ) applies to the data made available in this article, unless otherwise stated.

Competing interest

The authors declare that they have no competing interest.

Authors’ contributions

All authors wrote, read and approved the final manuscript.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.
Literatur
1.
Zurück zum Zitat Bergsagel DE, Valeriote FA: Growth characteristics of a mouse plasma cell tumor. Cancer Res. 1968, 28 (11): 2187-2196.PubMed Bergsagel DE, Valeriote FA: Growth characteristics of a mouse plasma cell tumor. Cancer Res. 1968, 28 (11): 2187-2196.PubMed
2.
Zurück zum Zitat Park CH, Bergsagel DE, McCulloch EA: Mouse myeloma tumor stem cells: a primary cell culture assay. J Natl Cancer Inst. 1971, 46 (2): 411-422.PubMed Park CH, Bergsagel DE, McCulloch EA: Mouse myeloma tumor stem cells: a primary cell culture assay. J Natl Cancer Inst. 1971, 46 (2): 411-422.PubMed
3.
Zurück zum Zitat Hamburger AW, Salmon SE: Primary bioassay of human tumor stem cells. Science. 1977, 197 (4302): 461-463.PubMedCrossRef Hamburger AW, Salmon SE: Primary bioassay of human tumor stem cells. Science. 1977, 197 (4302): 461-463.PubMedCrossRef
5.
Zurück zum Zitat Bonnet D, Dick JE: Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997, 3 (7): 730-737.PubMedCrossRef Bonnet D, Dick JE: Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997, 3 (7): 730-737.PubMedCrossRef
6.
Zurück zum Zitat Siegel R, Naishadham D, Jemal A: Cancer statistics, 2012. CA Cancer J Clin. 2012, 62 (1): 10-29.PubMedCrossRef Siegel R, Naishadham D, Jemal A: Cancer statistics, 2012. CA Cancer J Clin. 2012, 62 (1): 10-29.PubMedCrossRef
7.
8.
Zurück zum Zitat Boyd KD, et al: Understanding the molecular biology of myeloma and its therapeutic implications. Expert Rev Hematol. 2012, 5 (6): 603-617.PubMedCrossRef Boyd KD, et al: Understanding the molecular biology of myeloma and its therapeutic implications. Expert Rev Hematol. 2012, 5 (6): 603-617.PubMedCrossRef
9.
Zurück zum Zitat Mahindra A, et al: Latest advances and current challenges in the treatment of multiple myeloma. Nat Rev Clin Oncol. 2012, 9 (3): 135-143.PubMedCrossRef Mahindra A, et al: Latest advances and current challenges in the treatment of multiple myeloma. Nat Rev Clin Oncol. 2012, 9 (3): 135-143.PubMedCrossRef
10.
Zurück zum Zitat Bakkus MH, et al: Evidence that multiple myeloma Ig heavy chain VDJ genes contain somatic mutations but show no intraclonal variation. Blood. 1992, 80 (9): 2326-2335.PubMed Bakkus MH, et al: Evidence that multiple myeloma Ig heavy chain VDJ genes contain somatic mutations but show no intraclonal variation. Blood. 1992, 80 (9): 2326-2335.PubMed
11.
Zurück zum Zitat Bakkus MH, et al: Evidence that the clonogenic cell in multiple myeloma originates from a pre-switched but somatically mutated B cell. Br J Haematol. 1994, 87 (1): 68-74.PubMedCrossRef Bakkus MH, et al: Evidence that the clonogenic cell in multiple myeloma originates from a pre-switched but somatically mutated B cell. Br J Haematol. 1994, 87 (1): 68-74.PubMedCrossRef
12.
Zurück zum Zitat Bergsagel PL, et al: In multiple myeloma, clonotypic B lymphocytes are detectable among CD19+ peripheral blood cells expressing CD38, CD56, and monotypic Ig light chain. Blood. 1995, 85 (2): 436-447.PubMed Bergsagel PL, et al: In multiple myeloma, clonotypic B lymphocytes are detectable among CD19+ peripheral blood cells expressing CD38, CD56, and monotypic Ig light chain. Blood. 1995, 85 (2): 436-447.PubMed
13.
Zurück zum Zitat Billadeau D, et al: The bone marrow of multiple myeloma patients contains B cell populations at different stages of differentiation that are clonally related to the malignant plasma cell. J Exp Med. 1993, 178 (3): 1023-1031.PubMedCrossRef Billadeau D, et al: The bone marrow of multiple myeloma patients contains B cell populations at different stages of differentiation that are clonally related to the malignant plasma cell. J Exp Med. 1993, 178 (3): 1023-1031.PubMedCrossRef
14.
Zurück zum Zitat Pilarski LM, et al: In multiple myeloma, circulating hyperdiploid B cells have clonotypic immunoglobulin heavy chain rearrangements and may mediate spread of disease. Clin Cancer Res. 2000, 6 (2): 585-596.PubMed Pilarski LM, et al: In multiple myeloma, circulating hyperdiploid B cells have clonotypic immunoglobulin heavy chain rearrangements and may mediate spread of disease. Clin Cancer Res. 2000, 6 (2): 585-596.PubMed
15.
Zurück zum Zitat Szczepek AJ, et al: A high frequency of circulating B cells share clonotypic Ig heavy-chain VDJ rearrangements with autologous bone marrow plasma cells in multiple myeloma, as measured by single-cell and in situ reverse transcriptase-polymerase chain reaction. Blood. 1998, 92 (8): 2844-2855.PubMed Szczepek AJ, et al: A high frequency of circulating B cells share clonotypic Ig heavy-chain VDJ rearrangements with autologous bone marrow plasma cells in multiple myeloma, as measured by single-cell and in situ reverse transcriptase-polymerase chain reaction. Blood. 1998, 92 (8): 2844-2855.PubMed
16.
Zurück zum Zitat Bergui L, et al: Interleukin 3 and interleukin 6 synergistically promote the proliferation and differentiation of malignant plasma cell precursors in multiple myeloma. J Exp Med. 1989, 170 (2): 613-618.PubMedCrossRef Bergui L, et al: Interleukin 3 and interleukin 6 synergistically promote the proliferation and differentiation of malignant plasma cell precursors in multiple myeloma. J Exp Med. 1989, 170 (2): 613-618.PubMedCrossRef
17.
Zurück zum Zitat Vescio RA, 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 (5): 2487-2497.PubMed Vescio RA, 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 (5): 2487-2497.PubMed
18.
Zurück zum Zitat Rasmussen T, et al: In multiple myeloma clonotypic CD38- /CD19+ / CD27+ memory B cells recirculate through bone marrow, peripheral blood and lymph nodes. Leuk Lymphoma. 2004, 45 (7): 1413-1417.PubMedCrossRef Rasmussen T, et al: In multiple myeloma clonotypic CD38- /CD19+ / CD27+ memory B cells recirculate through bone marrow, peripheral blood and lymph nodes. Leuk Lymphoma. 2004, 45 (7): 1413-1417.PubMedCrossRef
19.
Zurück zum Zitat 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 (8): 2908-2913.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 (8): 2908-2913.PubMed
20.
Zurück zum Zitat Yaccoby S, Epstein J: The proliferative potential of myeloma plasma cells manifest in the SCID-hu host. Blood. 1999, 94 (10): 3576-3582.PubMed Yaccoby S, Epstein J: The proliferative potential of myeloma plasma cells manifest in the SCID-hu host. Blood. 1999, 94 (10): 3576-3582.PubMed
21.
Zurück zum Zitat Kim D, et al: CD19-CD45 low/- CD38 high/CD138+ plasma cells enrich for human tumorigenic myeloma cells. Leukemia. 2012, 26 (12): 2530-2537.PubMedCrossRef Kim D, et al: CD19-CD45 low/- CD38 high/CD138+ plasma cells enrich for human tumorigenic myeloma cells. Leukemia. 2012, 26 (12): 2530-2537.PubMedCrossRef
22.
Zurück zum Zitat Pilarski LM, et al: 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 (3): 1056-1065.PubMed Pilarski LM, et al: 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 (3): 1056-1065.PubMed
23.
Zurück zum Zitat Pilarski LM, et al: Leukemic B cells clonally identical to myeloma plasma cells are myelomagenic in NOD/SCID mice. Exp Hematol. 2002, 30 (3): 221-228.PubMedCrossRef Pilarski LM, et al: Leukemic B cells clonally identical to myeloma plasma cells are myelomagenic in NOD/SCID mice. Exp Hematol. 2002, 30 (3): 221-228.PubMedCrossRef
25.
26.
Zurück zum Zitat Reghunathan R, et al: Clonogenic multiple myeloma cells have shared stemness signature assocuated with patient survival. Oncotarget. 2013, 4 (8): 1230-1240.PubMedCentralPubMedCrossRef Reghunathan R, et al: Clonogenic multiple myeloma cells have shared stemness signature assocuated with patient survival. Oncotarget. 2013, 4 (8): 1230-1240.PubMedCentralPubMedCrossRef
27.
Zurück zum Zitat Taipale J, Beachy PA: The Hedgehog and Wnt signalling pathways in cancer. Nature. 2001, 411 (6835): 349-354.PubMedCrossRef Taipale J, Beachy PA: The Hedgehog and Wnt signalling pathways in cancer. Nature. 2001, 411 (6835): 349-354.PubMedCrossRef
28.
Zurück zum Zitat Ruizi Altaba AP, Sanchez N, Dahmane : Gli and hedgehog in cancer: tumours, embryos and stem cells. Nat Rev Cancer. 2002, 2 (5): 361-372.CrossRef Ruizi Altaba AP, Sanchez N, Dahmane : Gli and hedgehog in cancer: tumours, embryos and stem cells. Nat Rev Cancer. 2002, 2 (5): 361-372.CrossRef
30.
Zurück zum Zitat Takebe N, et al: Targeting cancer stem cells by inhibiting Wnt, Notch, and Hedgehog pathways. Nat Rev Clin Oncol. 2011, 8 (2): 97-106.PubMedCrossRef Takebe N, et al: Targeting cancer stem cells by inhibiting Wnt, Notch, and Hedgehog pathways. Nat Rev Clin Oncol. 2011, 8 (2): 97-106.PubMedCrossRef
31.
Zurück zum Zitat Peacock CD, et al: Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma. Proc Natl Acad Sci U S A. 2007, 104 (10): 4048-4053.PubMedCentralPubMedCrossRef Peacock CD, et al: Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma. Proc Natl Acad Sci U S A. 2007, 104 (10): 4048-4053.PubMedCentralPubMedCrossRef
32.
Zurück zum Zitat Ingham PW, McMahon AP: Hedgehog signaling in animal development: paradigms and principles. Genes Dev. 2001, 15 (23): 3059-3087.PubMedCrossRef Ingham PW, McMahon AP: Hedgehog signaling in animal development: paradigms and principles. Genes Dev. 2001, 15 (23): 3059-3087.PubMedCrossRef
33.
Zurück zum Zitat Vestergaard J, et al: Hedgehog signaling in small-cell lung cancer: frequent in vivo but a rare event in vitro. Lung Cancer. 2006, 52 (3): 281-290.PubMedCrossRef Vestergaard J, et al: Hedgehog signaling in small-cell lung cancer: frequent in vivo but a rare event in vitro. Lung Cancer. 2006, 52 (3): 281-290.PubMedCrossRef
34.
Zurück zum Zitat Mimeault M, et al: Cytotoxic effects induced by docetaxel, gefitinib, and cyclopamine on side population and nonside population cell fractions from human invasive prostate cancer cells. Mol Cancer Ther. 2010, 9 (3): 617-630.PubMedCentralPubMedCrossRef Mimeault M, et al: Cytotoxic effects induced by docetaxel, gefitinib, and cyclopamine on side population and nonside population cell fractions from human invasive prostate cancer cells. Mol Cancer Ther. 2010, 9 (3): 617-630.PubMedCentralPubMedCrossRef
35.
Zurück zum Zitat Von Hoff DD, et al: Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. N Engl J Med. 2009, 361 (12): 1164-1172.PubMedCrossRef Von Hoff DD, et al: Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. N Engl J Med. 2009, 361 (12): 1164-1172.PubMedCrossRef
36.
Zurück zum Zitat Rudin CM, et al: Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. N Engl J Med. 2009, 361 (12): 1173-1178.PubMedCrossRef Rudin CM, et al: Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. N Engl J Med. 2009, 361 (12): 1173-1178.PubMedCrossRef
37.
Zurück zum Zitat Angers S, Moon RT: Proximal events in Wnt signal transduction. Nat Rev Mol Cell Biol. 2009, 10 (7): 468-477.PubMedCrossRef Angers S, Moon RT: Proximal events in Wnt signal transduction. Nat Rev Mol Cell Biol. 2009, 10 (7): 468-477.PubMedCrossRef
38.
Zurück zum Zitat Grigoryan T, et al: Deciphering the function of canonical Wnt signals in development and disease: conditional loss- and gain-of-function mutations of beta-catenin in mice. Genes Dev. 2008, 22 (17): 2308-2341.PubMedCentralPubMedCrossRef Grigoryan T, et al: Deciphering the function of canonical Wnt signals in development and disease: conditional loss- and gain-of-function mutations of beta-catenin in mice. Genes Dev. 2008, 22 (17): 2308-2341.PubMedCentralPubMedCrossRef
39.
Zurück zum Zitat Derksen PW, et al: Illegitimate WNT signaling promotes proliferation of multiple myeloma cells. Proc Natl Acad Sci U S A. 2004, 101 (16): 6122-6127.PubMedCentralPubMedCrossRef Derksen PW, et al: Illegitimate WNT signaling promotes proliferation of multiple myeloma cells. Proc Natl Acad Sci U S A. 2004, 101 (16): 6122-6127.PubMedCentralPubMedCrossRef
40.
Zurück zum Zitat Bueno C, Lopes LF, Menendez P: Bone marrow stromal cell-derived Wnt signals as a potential underlying mechanism for cyclin D1 deregulation in multiple myeloma lacking t(11;14)(q13;q32). Blood Cells Mol Dis. 2007, 39 (3): 366-368.PubMedCrossRef Bueno C, Lopes LF, Menendez P: Bone marrow stromal cell-derived Wnt signals as a potential underlying mechanism for cyclin D1 deregulation in multiple myeloma lacking t(11;14)(q13;q32). Blood Cells Mol Dis. 2007, 39 (3): 366-368.PubMedCrossRef
41.
Zurück zum Zitat Chim CS, et al: Epigenetic dysregulation of Wnt signaling pathway in multiple myeloma. Leukemia. 2007, 21 (12): 2527-2536.PubMedCrossRef Chim CS, et al: Epigenetic dysregulation of Wnt signaling pathway in multiple myeloma. Leukemia. 2007, 21 (12): 2527-2536.PubMedCrossRef
42.
Zurück zum Zitat Dutta-Simmons J, et al: Aurora kinase A is a target of Wnt/beta-catenin involved in multiple myeloma disease progression. Blood. 2009, 114 (13): 2699-2708.PubMedCrossRef Dutta-Simmons J, et al: Aurora kinase A is a target of Wnt/beta-catenin involved in multiple myeloma disease progression. Blood. 2009, 114 (13): 2699-2708.PubMedCrossRef
43.
44.
Zurück zum Zitat Yao H, et al: AV-65, a novel Wnt/beta-catenin signal inhibitor, successfully suppresses progression of multiple myeloma in a mouse model. Blood Cancer J. 2011, 1 (11): e43-PubMedCentralPubMedCrossRef Yao H, et al: AV-65, a novel Wnt/beta-catenin signal inhibitor, successfully suppresses progression of multiple myeloma in a mouse model. Blood Cancer J. 2011, 1 (11): e43-PubMedCentralPubMedCrossRef
45.
Zurück zum Zitat Narayanan BA, et al: Antagonistic effect of small-molecule inhibitors of Wnt/beta-catenin in multiple myeloma. Anticancer Res. 2012, 32 (11): 4697-4707.PubMedCentralPubMed Narayanan BA, et al: Antagonistic effect of small-molecule inhibitors of Wnt/beta-catenin in multiple myeloma. Anticancer Res. 2012, 32 (11): 4697-4707.PubMedCentralPubMed
47.
Zurück zum Zitat Liu B, et al: Essential roles of grp94 in gut homeostasis via chaperoning canonical Wnt pathway. Proc Natl Acad Sci U S A. 2013, 110 (17): 6877-6882.PubMedCentralPubMedCrossRef Liu B, et al: Essential roles of grp94 in gut homeostasis via chaperoning canonical Wnt pathway. Proc Natl Acad Sci U S A. 2013, 110 (17): 6877-6882.PubMedCentralPubMedCrossRef
48.
Zurück zum Zitat Hua Y, et al: Molecular chaperone gp96 is a novel therapeutic target of multiple myeloma. Clin Cancer Res. 2013, 19 (22): 6242-6251.PubMedCrossRef Hua Y, et al: Molecular chaperone gp96 is a novel therapeutic target of multiple myeloma. Clin Cancer Res. 2013, 19 (22): 6242-6251.PubMedCrossRef
49.
Zurück zum Zitat Artavanis-Tsakonas S, Rand MD, Lake RJ: Notch signaling: cell fate control and signal integration in development. Science. 1999, 284 (5415): 770-776.PubMedCrossRef Artavanis-Tsakonas S, Rand MD, Lake RJ: Notch signaling: cell fate control and signal integration in development. Science. 1999, 284 (5415): 770-776.PubMedCrossRef
50.
Zurück zum Zitat Hassan KA, et al: Notch pathway activity identifies cells with cancer stem cell-like properties and correlates with worse survival in lung adenocarcinoma. Clin Cancer Res. 2013, 19 (8): 1972-1980.PubMedCentralPubMedCrossRef Hassan KA, et al: Notch pathway activity identifies cells with cancer stem cell-like properties and correlates with worse survival in lung adenocarcinoma. Clin Cancer Res. 2013, 19 (8): 1972-1980.PubMedCentralPubMedCrossRef
51.
Zurück zum Zitat Won HY, et al: Loss of Mel-18 enhances breast cancer stem cell activity and tumorigenicity through activating Notch signaling mediated by the Wnt/TCF pathway. FASEB J. 2012, 26 (12): 5002-5013.PubMedCrossRef Won HY, et al: Loss of Mel-18 enhances breast cancer stem cell activity and tumorigenicity through activating Notch signaling mediated by the Wnt/TCF pathway. FASEB J. 2012, 26 (12): 5002-5013.PubMedCrossRef
52.
Zurück zum Zitat Bao B, et al: Notch-1 induces epithelial-mesenchymal transition consistent with cancer stem cell phenotype in pancreatic cancer cells. Cancer Lett. 2011, 307 (1): 26-36.PubMedCentralPubMedCrossRef Bao B, et al: Notch-1 induces epithelial-mesenchymal transition consistent with cancer stem cell phenotype in pancreatic cancer cells. Cancer Lett. 2011, 307 (1): 26-36.PubMedCentralPubMedCrossRef
53.
Zurück zum Zitat Garcia Campelo MR, et al: Stem cell and lung cancer development: blaming the Wnt, Hh and Notch signalling pathway. Clin Transl Oncol. 2011, 13 (2): 77-83.PubMedCrossRef Garcia Campelo MR, et al: Stem cell and lung cancer development: blaming the Wnt, Hh and Notch signalling pathway. Clin Transl Oncol. 2011, 13 (2): 77-83.PubMedCrossRef
54.
Zurück zum Zitat Hovinga KE, et al: Inhibition of notch signaling in glioblastoma targets cancer stem cells via an endothelial cell intermediate. Stem Cells. 2010, 28 (6): 1019-1029.PubMedCrossRef Hovinga KE, et al: Inhibition of notch signaling in glioblastoma targets cancer stem cells via an endothelial cell intermediate. Stem Cells. 2010, 28 (6): 1019-1029.PubMedCrossRef
55.
Zurück zum Zitat Chiron D, et al: Critical role of the NOTCH ligand JAG2 in self-renewal of myeloma cells. Blood Cells Mol Dis. 2012, 48 (4): 247-253.PubMedCrossRef Chiron D, et al: Critical role of the NOTCH ligand JAG2 in self-renewal of myeloma cells. Blood Cells Mol Dis. 2012, 48 (4): 247-253.PubMedCrossRef
56.
Zurück zum Zitat Xu D, et al: Dll1/Notch activation accelerates multiple myeloma disease development by promoting CD138+ MM-cell proliferation. Leukemia. 2012, 26 (6): 1402-1405.PubMedCrossRef Xu D, et al: Dll1/Notch activation accelerates multiple myeloma disease development by promoting CD138+ MM-cell proliferation. Leukemia. 2012, 26 (6): 1402-1405.PubMedCrossRef
57.
Zurück zum Zitat Jundt F, et al: Jagged1-induced Notch signaling drives proliferation of multiple myeloma cells. Blood. 2004, 103 (9): 3511-3515.PubMedCrossRef Jundt F, et al: Jagged1-induced Notch signaling drives proliferation of multiple myeloma cells. Blood. 2004, 103 (9): 3511-3515.PubMedCrossRef
59.
Zurück zum Zitat Nefedova Y, et al: Inhibition of Notch signaling induces apoptosis of myeloma cells and enhances sensitivity to chemotherapy. Blood. 2008, 111 (4): 2220-2229.PubMedCrossRef Nefedova Y, et al: Inhibition of Notch signaling induces apoptosis of myeloma cells and enhances sensitivity to chemotherapy. Blood. 2008, 111 (4): 2220-2229.PubMedCrossRef
60.
Zurück zum Zitat Schwarzer R, et al: Notch inhibition blocks multiple myeloma cell-induced osteoclast activation. Leukemia. 2008, 22 (12): 2273-2277.PubMedCrossRef Schwarzer R, et al: Notch inhibition blocks multiple myeloma cell-induced osteoclast activation. Leukemia. 2008, 22 (12): 2273-2277.PubMedCrossRef
61.
Zurück zum Zitat Mirandola L, et al: Anti-Notch treatment prevents multiple myeloma cells localization to the bone marrow via the chemokine system CXCR4/SDF-1. Leukemia. 2013, 27 (7): 1558-1566.PubMedCrossRef Mirandola L, et al: Anti-Notch treatment prevents multiple myeloma cells localization to the bone marrow via the chemokine system CXCR4/SDF-1. Leukemia. 2013, 27 (7): 1558-1566.PubMedCrossRef
63.
Zurück zum Zitat Hoogstraten B, et al: Melphalan in multiple myeloma. Blood. 1967, 30 (1): 74-83.PubMed Hoogstraten B, et al: Melphalan in multiple myeloma. Blood. 1967, 30 (1): 74-83.PubMed
64.
Zurück zum Zitat Korst DR, et al: Multiple myeloma. Ii. Analysis of cyclophosphamide therapy in 165 patients. JAMA. 1964, 189: 758-762.PubMedCrossRef Korst DR, et al: Multiple myeloma. Ii. Analysis of cyclophosphamide therapy in 165 patients. JAMA. 1964, 189: 758-762.PubMedCrossRef
65.
Zurück zum Zitat Alexanian R, et al: Treatment for multiple myeloma. Combination chemother with different melphalan dose regimens. JAMA. 1969, 208 (9): 1680-1685.PubMedCrossRef Alexanian R, et al: Treatment for multiple myeloma. Combination chemother with different melphalan dose regimens. JAMA. 1969, 208 (9): 1680-1685.PubMedCrossRef
66.
Zurück zum Zitat Cooper MR, et al: Single, sequential, and multiple alkylating agent therapy for multiple myeloma: a CALGB Study. J Clin Oncol. 1986, 4 (9): 1331-1339.PubMed Cooper MR, et al: Single, sequential, and multiple alkylating agent therapy for multiple myeloma: a CALGB Study. J Clin Oncol. 1986, 4 (9): 1331-1339.PubMed
67.
Zurück zum Zitat Blade J, et al: Increased conventional chemotherapy does not improve survival in multiple myeloma: long-term results of two PETHEMA trials including 914 patients. Hematol J. 2001, 2 (4): 272-278.PubMedCrossRef Blade J, et al: Increased conventional chemotherapy does not improve survival in multiple myeloma: long-term results of two PETHEMA trials including 914 patients. Hematol J. 2001, 2 (4): 272-278.PubMedCrossRef
68.
Zurück zum Zitat Singhal S, et al: Antitumor activity of thalidomide in refractory multiple myeloma. N Engl J Med. 1999, 341 (21): 1565-1571.PubMedCrossRef Singhal S, et al: Antitumor activity of thalidomide in refractory multiple myeloma. N Engl J Med. 1999, 341 (21): 1565-1571.PubMedCrossRef
69.
Zurück zum Zitat Dimopoulos M, et al: Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N Engl J Med. 2007, 357 (21): 2123-2132.PubMedCrossRef Dimopoulos M, et al: Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N Engl J Med. 2007, 357 (21): 2123-2132.PubMedCrossRef
70.
Zurück zum Zitat Usmani SZ, et al: The anti-myeloma activity of a novel purine scaffold HSP90 inhibitor PU-H71 is via inhibition of both HSP90A and HSP90B1. J Hematol Oncol. 2010, 3 (1): 40-PubMedCentralPubMedCrossRef Usmani SZ, et al: The anti-myeloma activity of a novel purine scaffold HSP90 inhibitor PU-H71 is via inhibition of both HSP90A and HSP90B1. J Hematol Oncol. 2010, 3 (1): 40-PubMedCentralPubMedCrossRef
72.
Zurück zum Zitat Saini N, Mahindra A: Novel immunomodulatory compounds in multiple myeloma. Expert Opin Investig Drugs. 2013, 22 (2): 207-215.PubMedCrossRef Saini N, Mahindra A: Novel immunomodulatory compounds in multiple myeloma. Expert Opin Investig Drugs. 2013, 22 (2): 207-215.PubMedCrossRef
74.
Zurück zum Zitat Clevers H: The cancer stem cell: premises, promises and challenges. Nat Med. 2011, 17 (3): 313-319.PubMedCrossRef Clevers H: The cancer stem cell: premises, promises and challenges. Nat Med. 2011, 17 (3): 313-319.PubMedCrossRef
75.
Zurück zum Zitat Moser K, et al: Stromal niches, plasma cell differentiation and survival. Curr Opin Immunol. 2006, 18 (3): 265-270.PubMedCrossRef Moser K, et al: Stromal niches, plasma cell differentiation and survival. Curr Opin Immunol. 2006, 18 (3): 265-270.PubMedCrossRef
76.
Zurück zum Zitat Tokoyoda K, et al: Organization of immunological memory by bone marrow stroma. Nat Rev Immunol. 2010, 10 (3): 193-200.PubMedCrossRef Tokoyoda K, et al: Organization of immunological memory by bone marrow stroma. Nat Rev Immunol. 2010, 10 (3): 193-200.PubMedCrossRef
77.
Zurück zum Zitat Anjos-Afonso F, Bonnet D: Flexible and dynamic organization of bone marrow stromal compartment. Br J Haematol. 2007, 139 (3): 373-384.PubMedCrossRef Anjos-Afonso F, Bonnet D: Flexible and dynamic organization of bone marrow stromal compartment. Br J Haematol. 2007, 139 (3): 373-384.PubMedCrossRef
78.
Zurück zum Zitat Nilsson SK, et al: Immunofluorescence characterization of key extracellular matrix proteins in murine bone marrow in situ. J Histochem Cytochem. 1998, 46 (3): 371-377.PubMedCrossRef Nilsson SK, et al: Immunofluorescence characterization of key extracellular matrix proteins in murine bone marrow in situ. J Histochem Cytochem. 1998, 46 (3): 371-377.PubMedCrossRef
79.
Zurück zum Zitat Vanderkerken K, et al: Insulin-like growth factor-1 acts as a chemoattractant factor for 5 T2 multiple myeloma cells. Blood. 1999, 93 (1): 235-241.PubMed Vanderkerken K, et al: Insulin-like growth factor-1 acts as a chemoattractant factor for 5 T2 multiple myeloma cells. Blood. 1999, 93 (1): 235-241.PubMed
80.
Zurück zum Zitat Podar K, et al: Vascular endothelial growth factor triggers signaling cascades mediating multiple myeloma cell growth and migration. Blood. 2001, 98 (2): 428-435.PubMedCrossRef Podar K, et al: Vascular endothelial growth factor triggers signaling cascades mediating multiple myeloma cell growth and migration. Blood. 2001, 98 (2): 428-435.PubMedCrossRef
81.
Zurück zum Zitat Mitsiades CS, et al: Inhibition of the insulin-like growth factor receptor-1 tyrosine kinase activity as a therapeutic strategy for multiple myeloma, other hematologic malignancies, and solid tumors. Cancer Cell. 2004, 5 (3): 221-230.PubMedCrossRef Mitsiades CS, et al: Inhibition of the insulin-like growth factor receptor-1 tyrosine kinase activity as a therapeutic strategy for multiple myeloma, other hematologic malignancies, and solid tumors. Cancer Cell. 2004, 5 (3): 221-230.PubMedCrossRef
82.
Zurück zum Zitat Chauhan D, et al: Multiple myeloma cell adhesion-induced interleukin-6 expression in bone marrow stromal cells involves activation of NF-kappa B. Blood. 1996, 87 (3): 1104-1112.PubMed Chauhan D, et al: Multiple myeloma cell adhesion-induced interleukin-6 expression in bone marrow stromal cells involves activation of NF-kappa B. Blood. 1996, 87 (3): 1104-1112.PubMed
83.
Zurück zum Zitat Moreaux J, et al: BAFF and APRIL protect myeloma cells from apoptosis induced by interleukin 6 deprivation and dexamethasone. Blood. 2004, 103 (8): 3148-3157.PubMedCentralPubMedCrossRef Moreaux J, et al: BAFF and APRIL protect myeloma cells from apoptosis induced by interleukin 6 deprivation and dexamethasone. Blood. 2004, 103 (8): 3148-3157.PubMedCentralPubMedCrossRef
84.
Zurück zum Zitat Dankbar B, et al: Vascular endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma. Blood. 2000, 95 (8): 2630-2636.PubMed Dankbar B, et al: Vascular endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma. Blood. 2000, 95 (8): 2630-2636.PubMed
85.
Zurück zum Zitat Hideshima T, et al: The role of tumor necrosis factor alpha in the pathophysiology of human multiple myeloma: therapeutic applications. Oncogene. 2001, 20 (33): 4519-4527.PubMedCrossRef Hideshima T, et al: The role of tumor necrosis factor alpha in the pathophysiology of human multiple myeloma: therapeutic applications. Oncogene. 2001, 20 (33): 4519-4527.PubMedCrossRef
86.
Zurück zum Zitat Gupta D, et al: Adherence of multiple myeloma cells to bone marrow stromal cells upregulates vascular endothelial growth factor secretion: therapeutic applications. Leukemia. 2001, 15 (12): 1950-1961.PubMedCrossRef Gupta D, et al: Adherence of multiple myeloma cells to bone marrow stromal cells upregulates vascular endothelial growth factor secretion: therapeutic applications. Leukemia. 2001, 15 (12): 1950-1961.PubMedCrossRef
87.
88.
Zurück zum Zitat Barille S, et al: Myeloma cells upregulate interleukin-6 secretion in osteoblastic cells through cell-to-cell contact but downregulate osteocalcin. Blood. 1995, 86 (8): 3151-3159.PubMed Barille S, et al: Myeloma cells upregulate interleukin-6 secretion in osteoblastic cells through cell-to-cell contact but downregulate osteocalcin. Blood. 1995, 86 (8): 3151-3159.PubMed
89.
Zurück zum Zitat Shipman CM, Croucher PI: Osteoprotegerin is a soluble decoy receptor for tumor necrosis factor-related apoptosis-inducing ligand/Apo2 ligand and can function as a paracrine survival factor for human myeloma cells. Cancer Res. 2003, 63 (5): 912-916.PubMed Shipman CM, Croucher PI: Osteoprotegerin is a soluble decoy receptor for tumor necrosis factor-related apoptosis-inducing ligand/Apo2 ligand and can function as a paracrine survival factor for human myeloma cells. Cancer Res. 2003, 63 (5): 912-916.PubMed
90.
Zurück zum Zitat Bataille R, et al: Mechanisms of bone destruction in multiple myeloma: the importance of an unbalanced process in determining the severity of lytic bone disease. J Clin Oncol. 1989, 7 (12): 1909-1914.PubMed Bataille R, et al: Mechanisms of bone destruction in multiple myeloma: the importance of an unbalanced process in determining the severity of lytic bone disease. J Clin Oncol. 1989, 7 (12): 1909-1914.PubMed
91.
92.
Zurück zum Zitat Ehrlich LA, Roodman GD: The role of immune cells and inflammatory cytokines in Paget's disease and multiple myeloma. Immunol Rev. 2005, 208: 252-266.PubMedCrossRef Ehrlich LA, Roodman GD: The role of immune cells and inflammatory cytokines in Paget's disease and multiple myeloma. Immunol Rev. 2005, 208: 252-266.PubMedCrossRef
93.
94.
Zurück zum Zitat Pellat-Deceunynck C, et al: Adhesion molecules on human myeloma cells: significant changes in expression related to malignancy, tumor spreading, and immortalization. Cancer Res. 1995, 55 (16): 3647-3653.PubMed Pellat-Deceunynck C, et al: Adhesion molecules on human myeloma cells: significant changes in expression related to malignancy, tumor spreading, and immortalization. Cancer Res. 1995, 55 (16): 3647-3653.PubMed
95.
Zurück zum Zitat Damiano JS, et al: Cell adhesion mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human myeloma cell lines. Blood. 1999, 93 (5): 1658-1667.PubMed Damiano JS, et al: Cell adhesion mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human myeloma cell lines. Blood. 1999, 93 (5): 1658-1667.PubMed
96.
Zurück zum Zitat Westhoff MA, et al: Identification of a novel switch in the dominant forms of cell adhesion-mediated drug resistance in glioblastoma cells. Oncogene. 2008, 27 (39): 5169-5181.PubMedCrossRef Westhoff MA, et al: Identification of a novel switch in the dominant forms of cell adhesion-mediated drug resistance in glioblastoma cells. Oncogene. 2008, 27 (39): 5169-5181.PubMedCrossRef
97.
Zurück zum Zitat Matsunaga T, et al: Combination therapy of an anticancer drug with the FNIII14 peptide of fibronectin effectively overcomes cell adhesion-mediated drug resistance of acute myelogenous leukemia. Leukemia. 2008, 22 (2): 353-360.PubMedCrossRef Matsunaga T, et al: Combination therapy of an anticancer drug with the FNIII14 peptide of fibronectin effectively overcomes cell adhesion-mediated drug resistance of acute myelogenous leukemia. Leukemia. 2008, 22 (2): 353-360.PubMedCrossRef
98.
Zurück zum Zitat Alsayed Y, et al: Mechanisms of regulation of CXCR4/SDF-1 (CXCL12)-dependent migration and homing in multiple myeloma. Blood. 2007, 109 (7): 2708-2717.PubMedCentralPubMed Alsayed Y, et al: Mechanisms of regulation of CXCR4/SDF-1 (CXCL12)-dependent migration and homing in multiple myeloma. Blood. 2007, 109 (7): 2708-2717.PubMedCentralPubMed
99.
Zurück zum Zitat Lapidot T: Mechanism of human stem cell migration and repopulation of NOD/SCID and B2mnull NOD/SCID mice. The role of SDF-1/CXCR4 interactions. Ann N Y Acad Sci. 2001, 938: 83-95.PubMedCrossRef Lapidot T: Mechanism of human stem cell migration and repopulation of NOD/SCID and B2mnull NOD/SCID mice. The role of SDF-1/CXCR4 interactions. Ann N Y Acad Sci. 2001, 938: 83-95.PubMedCrossRef
100.
Zurück zum Zitat Miyamoto T, Weissman IL, Akashi K: AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation. Proc Natl Acad Sci U S A. 2000, 97 (13): 7521-7526.PubMedCentralPubMedCrossRef Miyamoto T, Weissman IL, Akashi K: AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation. Proc Natl Acad Sci U S A. 2000, 97 (13): 7521-7526.PubMedCentralPubMedCrossRef
101.
Zurück zum Zitat Meng S, et al: Circulating tumor cells in patients with breast cancer dormancy. Clin Cancer Res. 2004, 10 (24): 8152-8162.PubMedCrossRef Meng S, et al: Circulating tumor cells in patients with breast cancer dormancy. Clin Cancer Res. 2004, 10 (24): 8152-8162.PubMedCrossRef
103.
104.
Zurück zum Zitat Vereecque R, et al: Gene transfer of GM-CSF, CD80 and CD154 cDNA enhances survival in a murine model of acute leukemia with persistence of a minimal residual disease. Gene Ther. 2000, 7 (15): 1312-1316.PubMedCrossRef Vereecque R, et al: Gene transfer of GM-CSF, CD80 and CD154 cDNA enhances survival in a murine model of acute leukemia with persistence of a minimal residual disease. Gene Ther. 2000, 7 (15): 1312-1316.PubMedCrossRef
105.
Zurück zum Zitat Saudemont A, Quesnel B: In a model of tumor dormancy, long-term persistent leukemic cells have increased B7-H1 and B7.1 expression and resist CTL-mediated lysis. Blood. 2004, 104 (7): 2124-2133.PubMedCrossRef Saudemont A, Quesnel B: In a model of tumor dormancy, long-term persistent leukemic cells have increased B7-H1 and B7.1 expression and resist CTL-mediated lysis. Blood. 2004, 104 (7): 2124-2133.PubMedCrossRef
107.
Zurück zum Zitat Tirapu I, et al: Low surface expression of B7-1 (CD80) is an immunoescape mechanism of colon carcinoma. Cancer Res. 2006, 66 (4): 2442-2450.PubMedCrossRef Tirapu I, et al: Low surface expression of B7-1 (CD80) is an immunoescape mechanism of colon carcinoma. Cancer Res. 2006, 66 (4): 2442-2450.PubMedCrossRef
108.
Zurück zum Zitat Pratt G, Goodyear O, Moss P: Immunodeficiency and immunotherapy in multiple myeloma. Br J Haematol. 2007, 138 (5): 563-579.PubMedCrossRef Pratt G, Goodyear O, Moss P: Immunodeficiency and immunotherapy in multiple myeloma. Br J Haematol. 2007, 138 (5): 563-579.PubMedCrossRef
110.
Zurück zum Zitat Beyer M, et al: In vivo peripheral expansion of naive CD4 + CD25high FoxP3+ regulatory T cells in patients with multiple myeloma. Blood. 2006, 107 (10): 3940-3949.PubMedCrossRef Beyer M, et al: In vivo peripheral expansion of naive CD4 + CD25high FoxP3+ regulatory T cells in patients with multiple myeloma. Blood. 2006, 107 (10): 3940-3949.PubMedCrossRef
111.
Zurück zum Zitat Muthu Raja KR, et al: Increased T regulatory cells are associated with adverse clinical features and predict progression in multiple myeloma. PLoS One. 2012, 7 (10): e47077-PubMedCentralPubMedCrossRef Muthu Raja KR, et al: Increased T regulatory cells are associated with adverse clinical features and predict progression in multiple myeloma. PLoS One. 2012, 7 (10): e47077-PubMedCentralPubMedCrossRef
112.
Zurück zum Zitat Dhodapkar KM, et al: Dendritic cells mediate the induction of polyfunctional human IL17-producing cells (Th17-1 cells) enriched in the bone marrow of patients with myeloma. Blood. 2008, 112 (7): 2878-2885.PubMedCentralPubMedCrossRef Dhodapkar KM, et al: Dendritic cells mediate the induction of polyfunctional human IL17-producing cells (Th17-1 cells) enriched in the bone marrow of patients with myeloma. Blood. 2008, 112 (7): 2878-2885.PubMedCentralPubMedCrossRef
113.
Zurück zum Zitat Prabhala RH, et al: Elevated IL-17 produced by TH17 cells promotes myeloma cell growth and inhibits immune function in multiple myeloma. Blood. 2010, 115 (26): 5385-5392.PubMedCentralPubMedCrossRef Prabhala RH, et al: Elevated IL-17 produced by TH17 cells promotes myeloma cell growth and inhibits immune function in multiple myeloma. Blood. 2010, 115 (26): 5385-5392.PubMedCentralPubMedCrossRef
114.
Zurück zum Zitat Gorgun GT, et al: Tumor-promoting immune-suppressive myeloid-derived suppressor cells in the multiple myeloma microenvironment in humans. Blood. 2013, 121 (15): 2975-2987.PubMedCentralPubMedCrossRef Gorgun GT, et al: Tumor-promoting immune-suppressive myeloid-derived suppressor cells in the multiple myeloma microenvironment in humans. Blood. 2013, 121 (15): 2975-2987.PubMedCentralPubMedCrossRef
115.
Zurück zum Zitat Carbone E, et al: HLA class I, NKG2D, and natural cytotoxicity receptors regulate multiple myeloma cell recognition by natural killer cells. Blood. 2005, 105 (1): 251-258.PubMedCrossRef Carbone E, et al: HLA class I, NKG2D, and natural cytotoxicity receptors regulate multiple myeloma cell recognition by natural killer cells. Blood. 2005, 105 (1): 251-258.PubMedCrossRef
116.
Zurück zum Zitat Dhodapkar MV, et al: A reversible defect in natural killer T cell function characterizes the progression of premalignant to malignant multiple myeloma. J Exp Med. 2003, 197 (12): 1667-1676.PubMedCentralPubMedCrossRef Dhodapkar MV, et al: A reversible defect in natural killer T cell function characterizes the progression of premalignant to malignant multiple myeloma. J Exp Med. 2003, 197 (12): 1667-1676.PubMedCentralPubMedCrossRef
Metadaten
Titel
Fact or fiction - identifying the elusive multiple myeloma stem cell
verfasst von
Joshua Kellner
Bei Liu
Yubin Kang
Zihai Li
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2013
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/1756-8722-6-91

Weitere Artikel der Ausgabe 1/2013

Journal of Hematology & Oncology 1/2013 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.