Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Mar 1.
Published in final edited form as: Curr Opin Oncol. 2012 Mar;24(2):170–175. doi: 10.1097/CCO.0b013e32834ec015

Cancer Stem Cells - Relevance to Clinical Transplantation

Gabriel Ghiaur 1, Jonathan M Gerber 1, William Matsui 2, Richard J Jones 1,2
PMCID: PMC3312799  NIHMSID: NIHMS348493  PMID: 22314617

Abstract

Purpose of Review

Despite blood or marrow transplantation (BMT) being arguably the most active modality against hematologic malignancies, relapses remain the major reason for failure. Many cancers have now been shown to harbor cells that are phenotypically and biologically similar to normal cells with self-renewal capacity; these so-called cancer stem cells (CSCs) typically constitute only a small fraction of the total tumor burden, but are hypothesized to be responsible for relapse after conventional-dose therapy. Here, we review whether CSCs may have relevance to BMT.

Recent Findings

CSCs appear to be relatively resistant to standard anticancer therapies in vitro. The often dramatic responses induced by chemotherapy in most hematologic malignancies are likely a consequence of their impressive activity against the bulk tumor cells. Although the clinical importance of CSCs remains unproven, new evidence suggests that the limited durability of many of these responses reflect resistant CSCs. It is possible that CSCs are also relatively resistant to both high-dose myeloablative conditioning and allogeneic graft-versus-tumor effects. Data on the ability of most hematologic CSCs to circulate even early in the natural history of a malignancy, also raises concerns about contamination of autografts contributing to relapse.

Summary

Emerging data for the first time suggest CSCs may be responsible for relapse, even after BMT. However, BMT may be a particularly compelling setting to test CSC-targeting strategies, because it provides the most effective clinical debulking of hematologic malignancies and because CSC-targeting strategies may also enhance allogeneic antitumor immunity.

Keywords: Cancer stem cells, transplantation, leukemia, lymphoma, multiple myeloma

Introduction

Carl Nordling first hypothesized the multi-hit mutation theory of carcinogenesis in 1953 [1]. In this model, further refined by Ashley [2], Knudson [3], and Nowell [4], inherited mutations and/or environmental carcinogens lead to the development of pre-malignant cells. Such cells further accumulate mutations until one cell reaches a critical genetic or epigenetic state that confers a growth and/or survival advantage over its normal counterparts. Over time, if it can evade the immune system, this abnormal cancer-initiating cell would give rise to a malignant tumor.

Such a cancer-initiating cell would need to survive long enough to accumulate the 3 - 7 genetic mutations Ashley [2] postulated were necessary to generate cancer. Moreover, this cell must already manifest proliferative capacity or, alternatively, develop it anew as a consequence of the genetic mutation(s). Nowell hypothesized their inherent longevity and extensive proliferative capacity make tissue stem cells ideal candidate cancer-initiating cells [4]. In contrast, most terminally differentiated cells are short-lived and have only a limited number of divisions remaining in their differentiation program. Accordingly, such cells could only acquire the multiple genetic mutations required for malignant tumor growth if the essential mutations occurred simultaneously or in rapid succession (e.g., as in the generation of induced pluripotent stem cells). However, longevity and extensive proliferative capacity are not traits restricted to classical tissue stem cells. To some degree, hematopoietic progenitors generated by hematopoietic stem cells (HSCs) retain these properties. Moreover, memory lymphocytes are long-lived and self-renewing in order to maintain life-long immunity.

Most tissue-specific stem cells and other normal self-renewing cells, such as memory lymphocytes, generate differentiated progeny. Tumors derived from such transformed cells might therefore be expected to consist of a heterogeneous population of cells that includes the differentiated progeny of the original cell, mimicking to an extent the hierarchical structure of the normal tissue of origin. Accordingly, putative cancer-initiating cells possessing self-renewal and at least some differentiation potential, two of the defining features of normal stem cells, naturally came to be called cancer stem cells (CSCs). Although the CSC model would explain why only a minority of cells from most malignancies are clonogenic in vitro and in vivo, it is also conceptually possible that the low clonogenicity is the result of all cells within a cancer retaining the capacity to proliferate, but only at a low rate. Which of these two scenarios accounts for the low clonogenicity of most cancers, has been debated for years. The first clinical evidence supporting the CSC concept was published more than 40 years ago, when Fialkow et al demonstrated clonal hematopoiesis involving both erythroid and myeloid lineages in patients with chronic myeloid leukemia (CML) [5].

Identifying and characterizing hematologic malignancy CSCs

The stem cell origin of CML was confirmed nearly 20 years ago when several groups, using characteristics known to define normal HSCs, identified and isolated CML cells capable of expansion ex vivo [6]. Dick and colleagues extended these observations, showing cells with a HSC phenotype isolated from CML patients would generate leukemia in vivo in NOD/SCID mice [7]. The expression patterns of CML stem cells have also been shown to closely resemble those of normal HSCs [8*]. Thus, there is now universal agreement that the cancer-initiating event in CML, the Philadelphia (Ph) chromosome, occurs in an early hematopoietic cell if not the HSC itself.

Acute myeloid leukemia (AML) was the first cancer in which malignant cells with the ability to recapitulate the disease in NOD/SCID mice were identified [7,9,10]. These NOD/SCID engrafting leukemia cells also possessed self-renewal capacity and shared many phenotypic features with HSCs. Although leukemia stem cells (LSCs) from AML are the best characterized CSCs, the exact surface phenotype of LSCs continues to be a subject of debate [11**], perhaps because of the heterogeneity of AML.

The first modern use of the term cancer or tumor stem cells was probably by Bergsagel and Valeriote [12], who found that only a minority of mouse multiple myeloma cells were capable of clonogenic growth. Subsequent studies by Salmon and Hamburger confirmed these findings with clinical myeloma specimens, revealing a cloning efficiency ranging from approximately 1:1000-1:100,000 cells [13]. Insufficient tools existed at the time to distinguish whether this low clonogenic potential was the result of proliferative capacity exclusively restricted to a small subset of cancer cells or by all cancer cells retaining the capacity to proliferate but only at a low rate. Work from our laboratory found that the cancer-initiating cells in myeloma are found within the phenotypic memory B-lymphocyte population, with the CD138+ plasma cells representing terminally differentiated progeny of the malignant myeloma B cells. These myeloma CD138neg CSCs expressed CD19, CD20, and the memory B cell marker CD27, as well as high levels of the stem cell marker aldehyde dehydrogenase (ALDH) [14,15].

Recent data suggest the CSC concept may also apply to lymphomas. Hodgkin and Reed-Sternberg (HRS) cells, the hallmark of classical Hodgkin lymphoma (HL), belong to the B lymphoid lineage. However, they are unlike any normal cells of that lineage, and their limited proliferative potential belies the clinical aggressiveness of the disease. More than twenty years ago, Newcom et al identified a small population of phenotypic B cells that appeared to be responsible for the propagation of HRS cells within a HL cell line [16]. Our group recently confirmed these findings in several HL cell lines, and showed these cells also expressed ALDH and the memory B cell marker CD27 [17]. Moreover, clonotypic memory B cells with high levels of ALDH could be isolated from the peripheral blood of most newly-diagnosed HL patients, regardless of stage; importantly, these B cells and the patients’ HRS cells exhibited identical clonal immunoglobulin gene rearrangements [17]. Using a similar isolation strategy, clonotypic CD19+CD5+ALDHhigh B cells were identified in human mantle cell lymphoma (MCL) cell lines, as well as in patients with newly-diagnosed MCL.[18*]. These MCL cells were also found to be relatively quiescent and resistant to many classical chemotherapeutic agents used to treat this lymphoma.

The Paradox of Response and Survival: The Dandelion Phenomenon

Therapeutic advances over the past three decades now allow most hematologic malignancy patients to achieve major clinical responses. Although the responses can clearly decrease side effects and improve quality of life, most patients still eventually relapse and die of their disease. Moreover, there are numerous examples in which complete clinical responses do not produce improved survivals. Indolent lymphoma patients who achieved complete remissions with conventional-dose therapies in the pre-rituximab era did not experience a survival advantage over similar patients treated with a “watch and wait” approach [19]. In multiple myeloma, neither the magnitude nor the kinetics of clinical response impacted survival in some studies [20]. Even the most intensive therapy for myeloma, BMT [21,22], provided no overall survival advantage in the national intergroup trial [23] or in two recent meta-analyses [24,25].

The CSC concept would explain not only the low clonogenic capacity of most malignancies, but also why complete treatment responses translate into cures in only a minority of cancer patients: initial responses in cancer represent therapeutic effectiveness against the bulk cancer cells, while rarer resistant CSCs could be responsible for relapse. Putative CSCs have in fact been reported to be relatively resistant to standard anticancer therapies. Myeloma CSCs have been shown to be resistant to most clinically active agents (e.g., dexamethasone, lenalidomide, bortezomib), at least in part by co-opting normal stem cells’ intrinsic defense mechanisms such as quiescence, efflux pumps, and detoxifying enzymes [15]. Treatments that eliminate tumor bulk but spare the CSCs could be considered analogous to mowing dandelions; although this will eliminate the visible portion of the weeds, the unseen roots also need to be addressed to prevent regrowth [26,27]. Equally problematic would be treatments that are specific for CSCs. Such a treatment effect is akin to attacking just the root of the dandelion; although this has no immediately discernible effect on the weed, over time the weed should eventually wither and die if its root has been eliminated [26,27].

Controversy

Although cells meeting the definition for CSCs have now been described in many malignancies, there remains healthy skepticism about their true biologic significance [28,29]. Even in leukemia where the CSC concept is perhaps best established, a paucity of data indicates that LSCs are in fact responsible for disease resistance or relapse. This has led some to question whether CSCs may be nothing more than laboratory curiosities, simply reflecting the limitations of NOD/SCID mice for assessing tumorigenic potential [30]. However, despite being considered the gold standard assay for CSCs by many in the field, there is no reason to assume that growth in immunosuppressed mice is a relevant assay for clinically-significant CSCs. Actually, the most clinically relevant cancer cells are not necessarily those that engraft immunodeficient mice, but rather those responsible for relapse. Accordingly, even if every cell in a cancer possessed tumorigenic potential, the more clinically relevant question is probably whether any cell can also be responsible for relapse or only a discreet cell subset. Emerging data in several malignancies now suggest that a discreet subset of phenotypic CSCs may in fact be responsible for relapse.

Clinical Relevance

If CSCs are indeed more resistant to therapy than the bulk tumor cells and thus responsible for relapse, minimal residual disease (MRD) after treatment should be enriched for these cells. Furthermore, the presence of CSCs after therapy should predict recurrence. Indeed, residual breast tumor cell populations persisting after conventional treatment have recently been found to be enriched for breast cells with a CSC phenotype [31]. Similarly, patients with deletion 5q myelodysplastic syndrome (MDS) continue to harbor phenotypically distinct MDS stem cells (CD34+CD38lowCD90+), even in complete clinical and cytogenetic remission; these cells appear resistant to lenalidomide treatment and thus may account for disease relapse [32**]. Our group also showed that there was a strong and significant association between myeloma CSC numbers and progression-free survival in patients after treatment with high-dose chemotherapy and rituximab [33]. Our recent data also suggest that MRD in AML has a stem cell phenotype [34], and the presence or absence of AML CSCs after therapy correlates with progression-free survival. These data, perhaps for the first time, provide evidence of clinical relevance for CSC’s.

CSCs and BMT

Autologous BMT as rescue for dose intensification of standard cytotoxic agents is in general usage for three diseases: multiple myeloma, diffuse large cell lymphoma, and Hodgkin lymphoma. However, autologous BMT is unable to cure multiple myeloma, and its effectiveness in relapsed diffuse large cell lymphoma has significantly lessened in the rituximab era [35**]. These data suggest that even very high doses of chemotherapy may be unable to kill some CSCs [15]. Moreover, we found that CSCs circulate in the blood at surprisingly high levels, even in early stage Hodgkin lymphoma and multiple myeloma [15,17], suggesting that contaminating CSCs within the autograft might also contribute to relapses. It is possible that the ability to circulate is an inherent property of CSCs, co-opted from their normal counterparts that migrate for the purposes of maintaining tissue homeostasis and repair.

Relapse remains the major cause for failure of allogeneic BMT as well. Even the potent allogeneic graft-versus-tumor effect appears to provide limited antitumor benefit for many hematologic malignancies; e.g., despite high complete remission rates, relapse rates remain high after alloBMT for multiple myeloma [36] and advanced myelodysplastic syndromes (MDS) [37]. It is likely that those cancer cells remaining after the BMT conditioning regimen are resistant to immunologic, and well as genotoxic, killing. Although there is limited work on mechanisms of resistance to immunologic killing, the same anti-apoptotic signals responsible for cytotoxic resistance may also produce resistance to the induction of immunologic apoptosis. Moreover, LSCs like HSCs upregulate the “don’t eat me signal” CD47, presumably as a mechanism to avoid immune-mediated killing [38].

Although some CSCs may be able to evade high-dose cytotoxic therapy and immunologic graft-versus-tumor effects, allogeneic BMT may be a particularly compelling setting to study novel CSC-targeting approaches. Because BMT provides the most effective clinical debulking of hematologic malignancies, any added antitumor approach should be most effective. Further, the effectiveness of any novel CSC-directed treatment will also be most readily assessed in the setting of optimal clinical debulking. It also appears that many anticancer therapies may interact with the new allogeneic immune system to produce an enhanced antitumor effect. One mechanism of such synergy could be through enhancement of tumor cell recognition by the new immune system [39-41].

Conclusion

Stem cell pathways not only appear to be responsible for CSC resistance to many anticancer agents [15], but they may also lead to the development of novel therapies active across many malignancies. Several signaling pathways that are important for the generation and maintenance of normal stem cells during embryonic development, (e.g, Notch, Wnt, and Hedgehog) [42] and/or postnatally (e.g., telomerase [43]) also appear to be important for the growth of many cancers. In fact, prospective targets shared with normal stem cells may have particularly strong anticancer potential since their conserved expression implies a critical function retained by the CSCs. Preliminary data suggest that inhibition of these pathways, even when they are not mutated or over-expressed, may produce potent antitumor activity across a range of malignancies, possibly because of the key roles these pathways play in stem cell maintenance and growth [44*, 45].

While toxicity is an obvious concern for targets shared with normal stem cells, there are several potential differences between normal stem cells and CSCs that may provide a therapeutic ratio for shared targets. Normal stem cells have normal cell cycle checkpoints that are likely to protect them from cellular damage or crisis. The stage of differentiation at which cancers arise may also provide selectivity for approaches targeting CSCs. Although many cancers may arise from normal cells with stem cell properties, these cells may not be the most primitive tissue stem cells. Accordingly, if a therapy equally eliminated both myeloma CSCs and their normal counterparts, memory B cells, the existence of more primitive normal HSCs should replenish the normal B cell pool.

Differences in the interplay of telomere length and telomerase is another example how a stem cell pathway may provide a therapeutic ratio between CSCs and their normal counterparts [44*]. Normal stem cells require telomerase to prevent telomere shortening, leading to replicative senescence. However, even in the absence of telomerase, normal stem cells can maintain replicative capacity for some period of time because of their relatively long telomeres. Accordingly, telomerase knockout mice show a phenotype only after 4-6 generations [46]. In addition, the major cause of death in dyskeratosis congenita, a congenital disease that results from loss of function mutations in telomerase components, is bone marrow failure but this usually does not manifest until the second or third decade of life [47]. In contrast, uninterrupted telomerase activity may be absolutely required for the maintenance and growth of most malignancies, in order to stabilize the short telomeres that appear to characterize CSCs [48]. In fact, crossing telomerase knock-out mice with INK4a-/- [49] or APCmin [50] mice predisposed to cancer, significantly lowered the development of cancers in these mice. Thus, the differential in telomere length between normal (long) and cancer (short) stem cells could render telomerase inhibition selectively toxic to cancer.

Although the biologic and clinical relevance of CSCs remains controversial, emerging data are beginning to suggest that resistant CSCs are often responsible for relapse. The BMT setting may provide the optimal platform for testing post-transplant CSC-targeting. BMT not only provides arguably the most effective clinical debulking of hematologic malignancies, but the allogeneic immune system may also potentiate CSC-directed therapies. The use of CSC-targeted therapy in the MRD state after BMT potentially offers a novel approach to improving tumor control.

Bullet Points.

  1. Most cancers have been found to have a rare population of cells with stem cell characteristics, but the clinical significance of these cells has been unclear.

  2. The cancer stem cell concept, if true, would explain why complete treatment responses translate into cures in only a minority of cancer patients.

  3. Emerging evidence in several cancers suggest for the first time that cancer stem cells are responsible for relapse, even after blood and marrow transplantation.

  4. BMT may be a particularly compelling setting to test cancer stem cell-targeting strategies, because it provides the most effective clinical debulking of hematologic malignancies and because such strategies may also enhance allogeneic antitumor immunity.

Acknowledgments

Funding: NIH grants P01 CA15396, P01 CA70790 R01CA127574, and R01 CA150142. Gabriel Ghiaur is supported by National Research Service Award from NHLBI, 5T32HL007525. William Matsui is a Clinical Scholar of the Leukemia and Lymphoma Society.

References

  • 1.Nordling CO. A new theory on cancer-inducing mechanism. Br J Cancer. 1953;7:68–72. doi: 10.1038/bjc.1953.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ashley DJ. The two “hit” and multiple “hit” theories of carcinogenesis. Br J Cancer. 1969;23:313–328. doi: 10.1038/bjc.1969.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Knudson AG., Jr Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci U S A. 1971;68:820–823. doi: 10.1073/pnas.68.4.820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Nowell PC. The clonal evolution of tumor cell populations. Science. 1976;194:23–28. doi: 10.1126/science.959840. [DOI] [PubMed] [Google Scholar]
  • 5.Fialkow PJ, Gartler SM, Yoshida A. Clonal origin of chronic myelocytic leukemia in man. Proc Natl Acad Sci U S A. 1967;58:1468–1471. doi: 10.1073/pnas.58.4.1468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bedi A, Zehnbauer BA, Collector MI, et al. BCR-ABL gene rearrangement and expression of primitive hematopoietic progenitors in chronic myeloid leukemia. Blood. 1993;81:2898–2902. [PubMed] [Google Scholar]
  • 7.Sirard C, Lapidot T, Vormoor J, et al. Normal and leukemic SCID-repopulating cells (SRC) coexist in the bone marrow and peripheral blood from CML patients in chronic phase, whereas leukemic SRC are detected in blast crisis. Blood. 1996;87:1539–1548. [PubMed] [Google Scholar]
  • 8*.Gerber JM, Qin L, Kowalski J, et al. Characterization of chronic myeloid leukemia stem cells. Am J Hematol. 2011;86:31–37. doi: 10.1002/ajh.21915. Data suggest that many leukemia-specific antigens being studied are not specific for CML stem cells, but are expressed by normal stem cells and progenitors. WT1 and PRAME may show some leukemia-specificity, however. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.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]
  • 10.Lapidot T, Sirard C, Vormoor J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367:645–648. doi: 10.1038/367645a0. [DOI] [PubMed] [Google Scholar]
  • 11**.Sarry JE, Murphy K, Perry R, et al. Human acute myelogenous leukemia stem cells are rare and heterogeneous when assayed in NOD/SCID/IL2Rgammac-deficient mice. J Clin Invest. 2011;121:384–395. doi: 10.1172/JCI41495. Data demonstrating that leukemia cells capable of initiating leukemia in NOD/SCID mice are phenotypically heterogeneous. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bergsagel DE, Valeriote FA. Growth characteristics of a mouse plasma cell tumor. Cancer Res. 1968;28:2187–2196. [PubMed] [Google Scholar]
  • 13.Hamburger A, Salmon SE. Primary bioassay of human myeloma stem cells. J Clin Invest. 1977;60:846–854. doi: 10.1172/JCI108839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Matsui WH, Huff CA, Wang Q, et al. Characterization of clonogenic multiple myeloma cells. Blood. 2004;103:2332–2336. doi: 10.1182/blood-2003-09-3064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Matsui W, Wang Q, Barber JP, et al. Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance. Cancer Res. 2008;68:190–197. doi: 10.1158/0008-5472.CAN-07-3096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Newcom SR, Kadin ME, Phillips C. L-428 Reed-Sternberg cells and mononuclear Hodgkin’s cells arise from a single cloned mononuclear cell. Int J Cell Cloning. 1988;6:417–431. doi: 10.1002/stem.5530060606. [DOI] [PubMed] [Google Scholar]
  • 17.Jones RJ, Gocke CD, Kasamon YL, et al. Circulating clonotypic B cells in classical Hodgkin’s lymphoma. Blood. 2009;113:5920–5926. doi: 10.1182/blood-2008-11-189688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18*.Brennan SK, Meade B, Wang Q, et al. Mantle cell lymphoma activation enhances bortezomib sensitivity. Blood. 2010;116:4185–4191. doi: 10.1182/blood-2010-02-268375. First evidence for a putative CSC population within mantle cell lymphoma. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Horning SJ. Natural history of and therapy for the indolent non-Hodgkin’s lymphomas. Semin Oncol. 1993;20:75–88. [PubMed] [Google Scholar]
  • 20.Durie BG, Jacobson J, Barlogie B, Crowley J. Magnitude of response with myeloma frontline therapy does not predict outcome: importance of time to progression in southwest oncology group chemotherapy trials. J Clin Oncol. 2004;22:1857–1863. doi: 10.1200/JCO.2004.05.111. [DOI] [PubMed] [Google Scholar]
  • 21.Attal M, Harousseau JL, Stoppa AM, et al. A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Francais du Myelome. N Engl J Med. 1996;335:91–97. doi: 10.1056/NEJM199607113350204. see comments. [DOI] [PubMed] [Google Scholar]
  • 22.Child JA, Morgan GJ, Davies FE, et al. High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. N Engl J Med. 2003;348:1875–1883. doi: 10.1056/NEJMoa022340. [DOI] [PubMed] [Google Scholar]
  • 23.Barlogie B, Kyle RA, Anderson KC, et al. Standard chemotherapy compared with high-dose chemoradiotherapy for multiple myeloma: final results of phase III US Intergroup Trial S9321. J Clin Oncol. 2006;24:929–936. doi: 10.1200/JCO.2005.04.5807. [DOI] [PubMed] [Google Scholar]
  • 24.Levy V, Katsahian S, Fermand JP, et al. A meta-analysis on data from 575 patients with multiple myeloma randomly assigned to either high-dose therapy or conventional therapy. Medicine (Baltimore) 2005;84:250–259. doi: 10.1097/01.md.0000173272.71949.a1. [DOI] [PubMed] [Google Scholar]
  • 25.Koreth J, Cutler CS, Djulbegovic B, et al. High-dose therapy with single autologous transplantation versus chemotherapy for newly diagnosed multiple myeloma: A systematic review and meta-analysis of randomized controlled trials. Biol Blood Marrow Transplant. 2007;13:183–196. doi: 10.1016/j.bbmt.2006.09.010. [DOI] [PubMed] [Google Scholar]
  • 26.Jones RJ, Matsui WH, Smith BD. Cancer stem cells: are we missing the target? J Natl Cancer Inst. 2004;96:583–585. doi: 10.1093/jnci/djh095. [DOI] [PubMed] [Google Scholar]
  • 27.Huff CA, Matsui W, Smith BD, Jones RJ. The paradox of response and survival in cancer therapeutics. Blood. 2006;107:431–434. doi: 10.1182/blood-2005-06-2517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kern SE, Shibata D. The fuzzy math of solid tumor stem cells: a perspective. Cancer Res. 2007;67:8985–8988. doi: 10.1158/0008-5472.CAN-07-1971. [DOI] [PubMed] [Google Scholar]
  • 29.Kelly PN, Dakic A, Adams JM, et al. Tumor growth need not be driven by rare cancer stem cells. Science. 2007;317:337. doi: 10.1126/science.1142596. [DOI] [PubMed] [Google Scholar]
  • 30.Quintana E, Shackleton M, Sabel MS, et al. Efficient tumour formation by single human melanoma cells. Nature. 2008;456:593–598. doi: 10.1038/nature07567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Creighton CJ, Li X, Landis M, et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc Natl Acad Sci U S A. 2009;106:13820–13825. doi: 10.1073/pnas.0905718106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32**.Tehranchi R, Woll PS, Anderson K, et al. Persistent malignant stem cells in del(5q) myelodysplasia in remission. N Engl J Med. 2010;363:1025–1037. doi: 10.1056/NEJMoa0912228. One of first descriptions of a clinical significance for putative CSCs. [DOI] [PubMed] [Google Scholar]
  • 33.Huff CA, Wang Q, Rogers K, et al. Correlation of clonogenic cancer stem cell growth with clinical outcomes in multiple myeloma (MM) patients undergoing treatment with high dose cyclophosphamide and rituximab. Proc AACR. 2008 Late Breaking Abstract:LB:87. [Google Scholar]
  • 34.Gerber JM, Smith BD, Ngwang B, et al. The Clinical Relevance of Acute Myeloid Leukemia Stem Cells. Blood. 2011;118:240a. [Google Scholar]
  • 35**.Gisselbrecht C, Glass B, Mounier N, et al. Salvage regimens with autologous transplantation for relapsed large B-cell lymphoma in the rituximab era. J Clin Oncol. 2010;28:4184–4190. doi: 10.1200/JCO.2010.28.1618. Although autologous transplantation has been considered the treatment of choice for relapsed diffuse large cell lymphoma, this large clinical trial showed that it has become much less effective in the rituximab era. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Gahrton G, Svensson H, Cavo M, et al. Progress in allogenic bone marrow and peripheral blood stem cell transplantation for multiple myeloma: a comparison between transplants performed 1983--93 and 1994--8 at European Group for Blood and Marrow Transplantation centres. Br J Haematol. 2001;113:209–216. doi: 10.1046/j.1365-2141.2001.02726.x. [DOI] [PubMed] [Google Scholar]
  • 37.Sierra J, Perez WS, Rozman C, et al. Bone marrow transplantation from HLA-identical siblings as treatment for myelodysplasia. Blood. 2002;100:1997–2004. [PubMed] [Google Scholar]
  • 38.Jaiswal S, Jamieson CH, Pang WW, et al. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell. 2009;138:271–285. doi: 10.1016/j.cell.2009.05.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Jabbour E, Giralt S, Kantarjian H, et al. Low-dose azacitidine after allogeneic stem cell transplantation for acute leukemia. Cancer. 2009;115:1899–1905. doi: 10.1002/cncr.24198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Bolanos-Meade J, Smith BD, Gore SD, et al. 5-Azacytidine as Salvage Treatment in Relapsed Myeloid Tumors after Allogeneic Bone Marrow Transplantation. Biol Blood Marrow Transplant. 2011;17:754–8. doi: 10.1016/j.bbmt.2010.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Metzelder S, Wang Y, Wollmer E, et al. Compassionate use of sorafenib in FLT3-ITD-positive acute myeloid leukemia: sustained regression before and after allogeneic stem cell transplantation. Blood. 2009;113:6567–6571. doi: 10.1182/blood-2009-03-208298. [DOI] [PubMed] [Google Scholar]
  • 42.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]
  • 43.Harrington L. Does the reservoir for self-renewal stem from the ends? Oncogene. 2004;23:7283–7289. doi: 10.1038/sj.onc.1207948. [DOI] [PubMed] [Google Scholar]
  • 44*.Brennan SK, Wang Q, Tressler R, et al. Telomerase inhibition targets clonogenic multiple myeloma cells through telomere length-dependent and independent mechanisms. PLoS One. 2010;5:e12487. doi: 10.1371/journal.pone.0012487. Data showing inhibition of telomerase has activity against CSCs. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Peacock CD, Wang Q, Gesell GS, et al. Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma. Proc Natl Acad Sci U S A. 2007;104:4048–4053. doi: 10.1073/pnas.0611682104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hao LY, Armanios M, Strong MA, et al. Short telomeres, even in the presence of telomerase, limit tissue renewal capacity. Cell. 2005;123:1121–1131. doi: 10.1016/j.cell.2005.11.020. [DOI] [PubMed] [Google Scholar]
  • 47.Dokal I, Vulliamy T. Dyskeratosis congenita: its link to telomerase and aplastic anaemia. Blood Rev. 2003;17:217–225. doi: 10.1016/s0268-960x(03)00020-1. [DOI] [PubMed] [Google Scholar]
  • 48.Ju Z, Rudolph KL. Telomeres and telomerase in cancer stem cells. Eur J Cancer. 2006;42:1197–1203. doi: 10.1016/j.ejca.2006.01.040. [DOI] [PubMed] [Google Scholar]
  • 49.Greenberg RA, Chin L, Femino A, et al. Short dysfunctional telomeres impair tumorigenesis in the INK4a(delta2/3) cancer-prone mouse. Cell. 1999;97:515–525. doi: 10.1016/s0092-8674(00)80761-8. [DOI] [PubMed] [Google Scholar]
  • 50.Rudolph KL, Millard M, Bosenberg MW, DePinho RA. Telomere dysfunction and evolution of intestinal carcinoma in mice and humans. Nat Genet. 2001;28:155–159. doi: 10.1038/88871. [DOI] [PubMed] [Google Scholar]

RESOURCES